Next Article in Journal
Comparative Transcriptome Analysis Revealed the Key Genes Regulating Ascorbic Acid Synthesis in Actinidia
Next Article in Special Issue
A New Perspective on Thyroid Hormones: Crosstalk with Reproductive Hormones in Females
Previous Article in Journal
Genomic and Metabolic Characteristics of the Pathogenicity in Pseudomonas aeruginosa
Previous Article in Special Issue
Changes in Porcine Corpus Luteum Proteome Associated with Development, Maintenance, Regression, and Rescue during Estrous Cycle and Early Pregnancy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel Pituitary Actions of TAC4 Gene Products in Teleost

1
College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
2
State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
*
Authors to whom correspondence should be addressed.
Both Xuetao Shi and Cheng Ye contribute to this work equally.
Int. J. Mol. Sci. 2021, 22(23), 12893; https://doi.org/10.3390/ijms222312893
Submission received: 14 October 2021 / Revised: 24 November 2021 / Accepted: 24 November 2021 / Published: 29 November 2021
(This article belongs to the Special Issue Hormones and Receptors in Sexual Reproduction)

Abstract

:
Tachykinin 4 (TAC4) is the latest member of the tachykinin family involved in several physiological functions in mammals. However, little information is available about TAC4 in teleost. In the present study, we firstly isolated TAC4 and six neurokinin receptors (NKRs) from grass carp brain and pituitary. Sequence analysis showed that grass carp TAC4 could encode two mature peptides (namely hemokinin 1 (HK1) and hemokinin 2 (HK2)), in which HK2 retained the typical FXGLM motif in C-terminal of tachyinin, while HK1 contained a mutant VFGLM motif. The ligand-receptor selectivity showed that HK2 could activate all 6 NKRs but with the highest activity for the neurokinin receptor 2 (NK2R). Interestingly, HK1 displayed a very weak activation for each NKR isoform. In grass carp pituitary cells, HK2 could induce prolactin (PRL), somatolactin α (SLα), urotensin 1 (UTS1), neuromedin-B 1 (NMB1), cocaine- and amphetamine-regulated transcript 2 (CART2) mRNA expression mediated by NK2R and neurokinin receptor 3 (NK3R) via activation cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), phospholipase C (PLC)/inositol 1,4,5-triphosphate (IP3)/protein kinase C (PKC) and calcium2+ (Ca2+)/calmodulin (CaM)/calmodulin kinase-II (CaMK II) cascades. However, the corresponding stimulatory effects triggered by HK1 were found to be notably weaker. Furthermore, based on the structural base for HK1, our data suggested that a phenylalanine (F) to valine (V) substitution in the signature motif of HK1 might have contributed to its weak agonistic actions on NKRs and pituitary genes regulation.

1. Introduction

In mammals, the tachykinin (TAC) family includes three members, namely tachykinin 1 (TAC1), tachykinin 3 (TAC3) (tachykinin 2 (TAC2) in rodents), and TAC4, respectively. Among them, TAC1 gene encodes substance P (SP) and neurokinin A (NKA). SP is known to be involved in the regulation of pain control/injury [1], neurogenicinflammation [2] and obesity [3] with mammals. Compared with mammals, teleost have undergone an additional genome duplication during evolution, which is called fish-specific genome duplication (FSGD) or the 3R hypothesis [4]. So there were two isoforms TAC1 (namely tachykinin 1 isoforms a (TAC1a) and tachykinin 1 isoforms b (TAC1b)) and TAC3 (namely tachykinin 3 isoforms a (TAC3a), and tachykinin 3 isoforms b (TAC3b)), respectively. Similar to mammals, teleost TAC1 also encodes two mature peptides, namely SP and NKA, respectively [5]. In teleost, SP and NKA can trigger the secretion of luteinizing hormone β (LHβ), PRL, and SLα in the pituitary cells of carp, and the transcriptional levels of PRL and SLα increase in parallel. Short-term SP treatment (3 h) induced the release of LHβ, while prolonged induction time (24 h) inhibited LHβ mRNA expression [5].
In mammals, TAC3 encodes neurokinin B (NKB), is known to be involved in the regulation of smooth muscles of the gastrointestinal tract, secretion of intestinal epithelial fluid, vasodilation, and stimulating sperm motility [6,7,8]. Recent studies have found that NKB is a key regulator of mammalian reproductive function, especially controlling the release of gonadotropin-releasing hormone (GnRH) in the hypothalamus [9,10]. In contrast to mammals, TAC3 does not only encode NKB, but also a novel tachykinin peptide in teleost, named neurokinin B-related peptide (NKBRP) [11]. The research on the function of the TAC3 gene products in fish has mainly focused on the regulation of the reproductive process. Previous studies have found that intraperitoneal injection of NKB or NKBRP could both induce LHβ secretion in sexually mature female zebrafish [12]. In addition, our previous studies have shown that both NKB and NKBRP can promote PRL and SLα secretion and mRNA synthesis in grass carp pituitary cells [11].
Compared to TAC1 and TAC3, TAC4 is the last member of the tachykinin family. In 2000, Zhang et al. was first isolated the TAC4 gene from mouse hematopoietic stem cells, and named its gene production as hemokin 1 (HK1) [13]. Subsequently, TAC4 was cloned from rats and humans. Among them, the mouse hemokinin 1 (mHK1) encoded by the rat TAC4 gene has extremely high similarity with rat hemokinin 1 (rHK1), while the human race TAC4 gene encoding product was fairly different [14]. The TAC4 gene can encode six neuropeptides, namely HK1, HK1 (4–11), endokinin A (EKA), endokinin B (EKB), endokinin C (EKC), and endokinin D (EKD), respectively [15,16,17]. In mammals, recent studies have shown that HK1 and EKs were new mediators of pain and inflammation, and also play the crucial role in the hematopoietic system, anti-anxiety, and anti-depression [18,19,20]. However, little information is available about TAC4 in teleost.
In the present study, using grass carp (Ctenopharyngodon idellus) as a model, we try to examine the pituitary actions of TAC4 gene product in teleost. Firstly, the grass carp TAC4 were cloned, which encoded two mature peptides, namely HK-1 and HK-2, respectively. Secondly, six potential NKRs were isolated from grass carp. Then, by using transfection and dual-luciferase detection, we tried to confirm the specific receptor for HK-1 and HK-2, respectively. Thirdly, using grass carp pituitary cells as a model, we try to examine the direct pituitary actions of HK-1 and HK-2 in teleost. Finally, we try to clarify the mechanism of functional differences between HK-1 and HK-2 in teleost.

2. Materials and Methods

2.1. Animals and Chemicals

Two-year-old grass carps with a body weight of 1.7 ± 0.2 kg were purchased from a local market and kept in a well-aerated 250 L aquaria under a 12 h light/12 h dark photoperiod at 28 ± 1 °C for seven days. Grass carps at this stage were prepuberal and the sex character was not obvious, so breed fish with mixed sexes for pituitary cell preparation. All experimental procedures were approved by the Huazhong Agricultural University for Laboratory Animal Care (Ethical Approval No. HBAC20091138; Date: 15 November 2009). Grass carps selected in the experiment were all subjected to complete anesthesia at 0.05% MS222 (Sigma, St. Louis, MO, USA). Grass carp HK2 (QKFQTFVGLM-NH2), HK1 (TPGLQQVFGLM-NH2), and HK1-V7F (TPGLQQFFGLM-NH2) were synthesized by GeneScript Corporation (Piscataway, NJ, USA). The three mature peptides were subpackaged at 1 mM concentration using ultrapure water and stored at −80 °C.

2.2. Molecular Cloning and Tissue Distribution of Grass Carp TAC4 and NKRs

In order to clarify the distribution of TAC4 and its receptors in various tissues of grass carp, total RNA were extracted from each brain subregion, pituitary gland, and peripheral tissues of grass carp. Then the concentration was detected by the Nanodrop 2000, and each tissue took an equal amount of total RNA for reverse transcription to prepare a cDNA template. Primer Premier 6 was used to design specific quantitative primers for these genes (Supplementary Tables S1 and S2), and their transcript level in grass carp brain subregions and peripheral tissues was detected by real-time quantitative PCR (RT-qPCR). In these studies, β-actin was performed as an internal control. Sequence alignment and phylogenetic analysis of TAC4 and NKRs was conducted with ClustalX 2.1 and MEGA 7.0 (http://www.megasoftware.net/). The 3D structures for TAC4 and NKRs were predicted by I-TASSE (https://zhanglab.ccmb.med.umich.edu/I-TASSER/) and SWISS-MODEL (https://swissmodel.expasy.org/), respectively.

2.3. Functional Expression of Grass Carp NKRs in HEK293T Cells

To clarify the receptor selectivity of TAC4 gene products, a transfection experiment was conducted in HEK293T cells, and the fluorescent signal was detected by the firefly luciferase reporter gene detection kit. Firstly, the cDNA sequences of neurokinin receptor 1 isoforms a (NK1Ra), neurokinin receptor 1 isoforms b (NK1Rb), NK2R, neurokinin receptor 3 isoforms a1 (NK3Ra1), neurokinin receptor 3 isoforms a2 (NK3Ra2), and neurokinin receptor 3 isoforms b (NK3Rb) were isolated from grass carp pituitary, and the coding region of the six receptors were then subcloned into the eukaryotic expression vector pcDNA3.1. Secondly, the HEK293T cells were co-transfected with the plasmids of NFAT-Luc (Nuclear factor of activated T cells-luciferase), green fluorescent protein (GFP), and each pcDNA3.1-NKRs by using Lipofectamine 3000 (Thermo Fisher). The transfected HEK293T cells were incubated with various concentration HK-1 (1–10,000 nM), HK-2 (1–10,000 nM), or HK1-V7F (1–10,000 nM) for 24 h, respectively. Finally, the transfected cells were collected to detect the luciferase value according to the instructions of the firefly luciferase reporter gene detection kit.

2.4. Transcriptome Sequence and Bioinformatics

Grass carp pituitary cells were obtained by trypsin digestion [21]. Then, the pituitary cells were seeded into a 24-well cell culture plate at a density of 2.5 × 106 cells/well, and incubated at 28 °C under 5% CO2 in cell culture incubator. The primary culture pituitary cells were treated by HK2 (1 μM) for 24 h. Then, the cells were collected to extract the total RNA by the Trizol method. The high-quality samples were sent to the Majorbio Genome Center (Shanghai, China) for transcriptomic sequencing. Using the negative binomial distribution model, the edgeR software was applied to calculate the differential expression based on the FPKM (fragments per kilobase of exon per million fragments mapped) value of the gene. The screening criteria for differentially expressed genes (DEGs) are FDR (false discovery rate) ≤ 0.05 and FC (fold change) ≥ 1.5. Functional annotation of gene ontology (GO) terms was analyzed by using Goatolls (http://github.com/tanghaibao/GOatolls), and KEGG functional classification were analyzed by using KOBAS (http://kobas.cbi.pku.edu.cn/home.do).

2.5. Real-Time Quantitative PCR (RT-qPCR) Validation

To further confirm the pituitary actions of HKs in grass carp, trypsin digestion method was used to prepare for grass carp primary cultured pituitary cells. In the time-dependent experiment, the pituitary cells were incubated with HK1 (1 µM) or HK2 (1 µM) for 3 h, 12 h, 24 h, and 48 h, respectively. In the dose-dependent experiment, the pituitary cells were incubated with HK1 (0.1–1000 nM) or HK2 (0.1–1000 nM) for 48 h, respectively. After drug treatment, total RNA was extracted from adherent cells by Trizol, and then subjected to reverse transcription using HifairTM III 1st Strand cDNA Synthesis Kit (Yeasen, Shanghai, China). In the present study, β-actin was used as the internal reference gene, and then, the transcript levels of PRL, SLα, CART2, UTS1, and NMB1 were detected by using a ABI 7500 real-time PCR system (see Supplementary Table S2 for primer sequences and PCR condition). In addition, the plasmid DNA containing the gene coding sequence was serially diluted as a standard for data calibration.

2.6. Statistical Analysis

In this study, RT-qPCR was used to detect mRNA expression for TAC4, NKRs, PRL, SLα, CART2, UTS1, SN2, and NMB1. In the ABI7500 software, the dynamic mRNA concentration detection range of the double standard curve is 105, and the correlation coefficient is >0.95. Since the transcript level of β-actin showed no significant changes, the data of PRL, SLα, CART2, UTS1, and NMB1 were converted into average percentages in the control group (% Ctrl). In the transfection experiment, the detected firefly luciferase activity was routinely normalized with the GFP activity expressed in the same well, expressed as the luciferin activity ratio Luc ratio. The data from four parallel experiments was presented as mean ± SEM in the figure. All the data were analyzed by ANOVA, and then subjected to Dunnett’s test in GraphPad Prism 7.0. The significant differences between groups were indicated by p < 0.05.

3. Result

3.1. Sequence Analysis and Tissue Distribution of TAC4 and NKRs

To examine the pituitary action of TAC4 gene products in grass carp, the cDNA sequence of TAC4 was firstly isolated from the brain of grass carp. Sequence analysis showed that grass carp TAC4 encoded two mature peptides, including an 11-a.a HK2 carrying the classical tachykinin signature motif (FXGLM) and a 10-a.a HK1 containing a mutated form of tacykinin consensus motif form (VFGLM) (Figure 1A,C). In addition, similar to the 3D structure of other tachykinin peptids, grass carp HK1 and HK2 were also found to be in the form of a short peptide with a α helix covering the region of the central core to the C-terminus (Figure 1D). Phylogenetic analysis revealed that the newly isolated grass carp TAC4 could be clustered in the clade of fish TAC4 with a close evolutionary relationship with common carp TAC4 (Figure 1E), and TAC4 group displayed a closer evolutionship with the TAC1 cluster compared to TAC3 (Figure 1E). Furthermore, we also isolated six neurokinin receptors from grass carp brain and pituitary, namely NK1Ra (Supplementary Figures S1 and S2), NK1Rb (Supplementary Figures S3 and S4), NK2R (Supplementary Figures S5 and S6), NK3Ra1 (Supplementary Figures S7 and S8), NK3Ra2 (Supplementary Figures S9 and S10) and NK3Rb (Supplementary Figures S11 and S12), respectively. Similar to mammals, the six NKRs in grass carp were all typical G protein- coupled receptor (Supplementary Figure S13). Phylogenetic analysis showed that the 6 NKRs were clustered into three groups, including NK1R, NK2R, and NK3R group, respectively (Figure 1F). Finally, tissue distribution revealed that TAC4 was widely distributed in various brain regions except for the cerebellum, and its transcript level was extremely high in the medulla oblongata and the olfactory bulb (Figure 1B). In addition, six NKRs could all be widely detected in various brain subregions and pituitary, and NK1Rb was more highly detected in the brain compared to other NKRs (Figure 1B).

3.2. Receptor Selectivity of TAC4 Gene Products

To clarify the receptor selectivity of HK1 and HK2 for the six NKRs, HEK-293T cell lines were established to stably express NK1Ra, NK1Rb, NK2R, NK3Rb, NK3Ra1, or NK3Ra2 were established, respectively. The cells were then used for transfection study with NFAT-Luc vectors, which was allowed for functional evaluation of the activation status of Ca2+-dependent pathway. The results showed that the HK2 could activate all 6 NKR isoforms, but HK1 displayed very weak activation for these NKRs (Figure 2). In the case of HK2, it exhibited affinity intensity was NK2R (EC50, 122.9 nM) > NK3Rb (EC50,145.6 nM) > NK1Ra (EC50,76.29 nM) ≈ NK3Ra1 (EC50,1344 nM) ≈ NK1Rb (EC50, 337.8 nM) > NK3Ra2 (EC50, 1232 nM) (Figure 2).

3.3. Transcriptomic Analysis of HK2 in the Pituitary

In order to reveal the pituitary actions of HK2 in lower vertebrates, the pituitary cells of grass carp were incubated with HK2 (1 μM) for 24 h. Then, high-throughput transcriptome technology was used to detect the changes in gene transcription levels in the pituitary after HK2 treatment (Figure 3). Compared with the control group, a total of 1151 differentially expressed genes (DEGs) were found in the HK2 treatment group, including 364 up-regulated DEGs and 787 down-regulated DEGs. The results of GO enrichment analysis showed that the up-regulated DEGs in the molecular function (MF) category were mainly involved in sequence-specific DNA binding, hormone activity, and receptor ligand activity, respectively (Figure 3A). In the biological process (BP) category, the main clusters were the negative regulation of metabolic processes, the G protein-coupled receptor signaling pathway, and the negative regulation of appetite (Figure 3A). Most of the cell components (CC) categories were enriched in kinetochore, plasma membrane, and cell junctions (Figure 3A). Down-regulated DEGs in the molecular function (MF) category were mainly clustered in G protein-coupled receptor activity, calcium ion binding, and steroid hormone receptor activity (Figure 3A). In the biological process (BP) category, they were mainly involved in cell adhesion, the regulation of innate immune response, and hormone-mediated signaling pathways (Figure 3A). In the cell components (CC) category, they were mostly enriched in the plasma membrane, ion channel complexes and transcription factor complexes (Figure 3A). The results of KEGG enrichment analysis showed that the up-regulated DEGs were mainly involved in the synthesis of steroid hormones, the cGMP-PKG signaling pathway, and the Jak-STAT signaling pathway. The down-regulated DEGs were mostly enriched in the cAMP signaling pathway, neuroactive ligand-receptor interactions, and phospholipase D signal pathway, respectively (Figure 3B). Finally, the GO enrichment group involved in reproduction function, growth factor, hormone activity, immune response, and feeding regulation are shown in Figure 4. In addition, the top 20 up-regulated DEGs are shown in Table 1, including CART2, PRL, SLα, UTS1, and NMB1. The top 20 down-regulated DEGs were displayed in Table 2, including somatostatin receptor type 2 isoforms a (SSTR2a), somatostatin receptor type 3 (SSTR3), opioid growth factor receptor-like protein 1 (OGFR), growth hormone secretagogue receptor type 1 (GHSR1), and fibroblast growth factor receptor 2 (FGFR2).

3.4. Regulation of HK1 and HK2 on the Key DEGs Expression in Grass Carp Pituitary Cells

To further confirm the pituitary actions of HK1 and HK2 in grass carp, HK1 (1 µM) or HK2 (1 µM) were used to incubate grass carp pituitary cells for 3 h, 6 h, 24 h, and 48 h, respectively. The results showed that HK2 (1 µM) could significantly induce SLα (Figure 5A), PRL (Figure 5B), CART2 (Figure 6A), NMB1 (Figure 6B), and UTS1 (Figure 6C) mRNA expression in a time-dependent manner. However, HK1 (1 μM) showed no effect on these genes in grass carp pituitary cells (Figure 5 and Figure 6). To further evaluate the dose-dependence of these stimulatory effects on pituitary hormones (SLα and PRL) and feeding peptides (CART2, NMB1 and UTS1), grass carp pituitary cells were exposed to increasing concentrations (0.1–1000 nM) of HK1 or HK2 for 24 h, respectively. In this case, HK2 treatment could consistently induce SLα (Figure 5A), PRL (Figure 5B), CART2 (Figure 6A), NMB1 (Figure 6B), and UTS1 (Figure 6C) mRNA expression in a concentration-related fashion. Similar to our time-course study, increasing levels of HK1 were ineffective in altering transcript expression of the five genes mRNA expression in grass carp pituitary cell even up to 1 µM concentration (Figure 5 and Figure 6).

3.5. Receptor Specificity and Post-Receptor Signal Transduction for HK2-Induced SLα and PRL mRNA Expression in Grass Carp Pituitary Cells

In this experiment, a pharmacological approach was used to clarify the receptor specificity for SLα and PRL regulation by HK2. Pituitary cells were incubated for 24 h with grass carp HK2 (1 μM) with simultaneous treatment of the NK1R antagonist Rolaptiant (10 μM) or NK3R antagonist SB222200 (10 μM), respectively. Similar to the results of the proceeding studies, HK2 could increase the basal transcript level of SLα and PRL. The stimulatory effects on SLα mRNA expression could be totally blocked by co-treatment with the NK3R anatagonist SB222200, but NK1R antagonist could only partially block HK2-induced SLα mRNA expression (Figure 7A). In the parallel experiment, HK2-induced PRL mRNA expression could not be totally abolished by NK1R antagonist Rolaptiant or NK3R antagonist SB222200, respectively (Figure 7A).
To elucidate the signal transduction for PRL and SLα regulation by HK2, the possible involvement of the cAMP-dependent cascade was examined by using the inhibitors for the cAMP pathway. As shown in Figure 7B, co-treatment with AC inhibitor MDL1230A (10 μM) or PKA inhibitor H89 (10 μM) were both effective in blocking the stimulatory effects of HK2 (1 μM) on PRL and SLα mRNA expression. In parallel experiments for testing the functional role of the PLC/IP3/PKC pathway, HK2-induced PRL and SLα mRNA expression could be both totally abolished by simultaneous treatment with the PLC inhibitor U73122 (10 μM), PKC inhibitor GF109203X (10 μM), or IP3 receptor blocker 2-APB (10 μM), respectively (Figure 7C). Furthermore, HK2-induced PRL and SLα mRNA expression were found to be suppressed or totally abolished by co-treatment with the VSCC inhibitor nifedipine (10 μM), CaM antagonist calmidazolium (1 μM) or CaMK-II blocker KN62 (10 μM), respectively (Figure 7D).

3.6. Receptor Specificity and Signal Transduction for HK2-Induced UTS1, NMB1 and CART2 mRNA Expression

In this part, the pharmacological approach was also recruited to clarify the receptor specificity and signal transduction for CART2, UTS1, or NMB1 transcript regulation by HK2. As shown in Figure 8A, the stimulatory effects of HK2 on CART2, UTS1, or NMB1 mRNA expression could be totally abolished by NK3R antagonist SB222200 (10 μM) but not with the NK1R antagonist Rolaptiant (10 μM) (Figure 8A). To shed light on the signal transduction for CART2, UTS1, or NMB1 transcript regulation by HK2, the possible involvement of cAMP-dependent pathway was examined firstly using the co-treatment with various inhibitors for the individual components of cAMP pathway. As shown in Figure 8B, HK2-induced CART2, UTS1, or NMB1 mRNA expression could be abolished by simultaneous incubation with the AC inhibitor MDL12330A (10 μM) or PKA inhibitor H89 (10 μM). In the parallel studies, co-treatment with PLC inhibitor U73122 (10 μM), PKC inhibitor GF109203X (10 μM), or IP3 receptor blocker 2-APB (10 μM) were all effective in blocking the stimulatory effects of HK2 (1 μM) on CART2, UTS1 or NMB1 mRNA expression (Figure 8C). Furthermore, HK2-induced CART2, UTS1, or NMB1 mRNA expression were found to be suppressed/totally abolished by co-treatment with VSCC inhibitor nifedipine (10 μM), CaM antagonist calmidazolium (1 μM) or CaMK-II blocker KN62 (10 μM), respectively (Figure 8D).

3.7. Phe to Val Mutation in FXGLM Motif in UTS1, PRL and SLα Regulation by HK2

As carp HK1 is unique for having a Phe to Val mutation in the “FXGLM” signature motif, the biological relevance of this mutation in pituitary hormone regulation was examined by mutating the Val residue at position 7 of carp HK1 back to Phe as in the case of the typical “FXGLM” motif. As shown in Figure 9A, the dose-response study has shown that the HK1-V7F mutant could significantly enhance the stimulatory effects of HK1 on SLα, PRL, and UTS1 mRNA expression to levels comparable to that induced by HK2. In addition, Phe to Val mutation in the “FXGLM” motif in HK1 on binding affinity to NKR were also examined. In HEK-293T cells with stable expression grass carp each NKR, co-transfection with NFAT-Luc expressing vectors were conducted to allow for biochemical probing of functional activation of NKR. Similar to the results of pituitary hormone genes regulation in carp pituitary cells, carp HK1 with a “VXGLM” motif was found to be a relatively low efficient stimulant for six NKR isoforms (Figure 2). In parallel experiments with the treatment of increasing doses of the Val to Phe mutant HK1-V7F, the stimulatory effects on NFAT-Luc mediated luciferase activity expression was notably enhanced with significant increases in the corresponding ED50 value from >10 μM to the nanomolar dose range (Figure 2).
Furthermore, to verify whether HK1 could function as an endogenous antagonist of NKRs, we examined the antagonist effect of HK1 on HK2-induced pituitary hormone genes expression. As shown in Figure 9B, a 24 h static incubation with a 0.1 µM dose carp HK2 was effective in triggering significant rises in SLα, PRL, and UTS1 mRNA expression in grass carp pituitary cells. Of note, the stimulatory effects of HK1 treatment were found to be much weaker compared to the corresponding stimulation induced by HK2 (Figure 9B). In the same experiment, co-treated with increasing levels of carp HK1 (1 µM and 10 µM doses) could not suppress the stimulatory actions caused by HK2 on SLα, PRL, and UTS1 mRNA expression (Figure 9B).

4. Discussion

In mammals, multiple tachykinin genes with different gene products, including TAC1 encoding SP and NKA, TAC3 (also referred to as TAC2 in rodents) encoding NKB and TAC4 encoding HK1 and EKs, have been widely reported [22]. Compared to mammals, due to the third round of whole-genome duplication, two TAC1 isoforms (TAC1a and TAC1b) [5] and two TAC3 isoforms (TAC3a and TAC3b) [23] have been reported in teleost. However, in the present study, only one TAC4 isoform was isolated in teleost, which might be the result of the non-functionalization by forming pseudogenes or deletion/mutations leading to the loss of redundant genes [24] (Supplement Figure S14). In mammals, TAC4 gene products were various in different species. TAC4 in mice and rats can encode one hemokinin mature peptide with 11 amino acid length [25]. In human, TAC4 encodes for hemokinin-1 (hHK-1), but its sequence is different from its murine counterpart. It is spliced into four alternative transcripts (α, β, γ and δ) that give rise to four different peptides which have been named endokinis A (EKA), B (EKB), C (EKC), and D (EKD), respectively [26,27]. In the present study, in contrast to mammals, TAC4 in teleost could not only encode HK-1, but also another mature tachykinin HK-2, which might be due to a loss of segmentally duplicated gene fragments in TAC4 during tetrapod evolution.
In mammals, the biological action of tachykinins is mediated by three GPCRs, namely NK1R, NK2R, and NK3R, which are stimulated preferentially by SP, NKA, and NKB, respectively [28]. Both in mice and rats, HK-1 has a similar affinity to SP at the NK1R [29,30], while human HK-1 binds to NK1R with a much lower affinity than SP [31]. In addition, high-dose human HK-1 can also bind to NK2R and NK3R [14]. In the present study, due to the third round whole-genome duplication, 6 NKRs were detected in grass carp, namely NK1Ra, NK1Rb, NK2R, NK3Ra1, NK3Ra2, and NK3Rb, respectively. Based on the ligand-receptor selectivity, we found that HK2 could activate all 6 NKR isoforms, but showed the highest affinity for NK2R. Our previous study has revealed that carp NK2R is a multiligand receptor, which could be activated by SP, NKA, NKBa, NKBRPa, NKBRPb with comparable efficacy and potency [5]. These results indicated that teleost hemokinin preferentially stimulated the multiligand receptor NK2R, which suggests that hemokinin in teleost may analog the function of other tachykinins through activation of NK2R.
In mammals, many studies have focused on the actions of hemokinin on immunological regulation and inflammation [32]. However, little information is available about its endocrinology function. In the present study, firstly, we found that six NKRs could be detected in grass carp pituitary, which indicated that HKs play an important role in the pituitary. Then, using grass carp pituitary cells as a model, we found that carp HK2 could significantly induce PRL and SLα mRNA expression mediated by NK2R and NK3R via activation of the cAMP/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK II cascade. Similar results were also reported in our previous TAC3 studies, which indicated that NKB could significantly induce pituitary PRL and SLα synthesis and secretion [17]. In fish models, PRL is involved in a wide range of physiological functions, including reproductive migration [33], gonadal maturation [34], reproductive cycling [35,36], nesting and brooding behaviors [37], and osmoregulation [38]. As the latest member of the growth hormone (GH)/PRL family, somatolactin was a pituitary hormone unique to fish species and has been reported to play a functional role in the regulation of reproduction [39], growth [40] and pigmentation [41]. These results, as a whole, suggested that HK2 was involved in pituitary regulation of PRL and SLα mRNA expression, which presumably may contribute to the reproduction and growth metabolism in teleost.
In addition to the regulation of pituitary PRL and SLα, our present study also found that HK2 could significantly induce pituitary UTS1, CART2, and NMB1 mRNA expression mediated by NK3R via activation of cAMP/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK II signal pathways. Interestingly, our recent study also found that NKB could stimulate UTS1, CART2, and NMB1 mRNA expression in grass carp pituitary cells [42]. Previous studies reported that CART could inhibit feeding in rats [43] and goldfish [44]. Similarly, NMB has also been found to inhibit feeding and gastric peristalsis [45,46]. Fish UTS1 showed a closed structural and biological homology with the corticortropin-releasing hormone (CRH) family. In mammals, CRH could reduce food intake and induce energy expenditure [47]. In teleost, the CRH system could also exert an anorexigenic effect [48]. These results, taken together, suggested that HK2 might be involved in the regulation of feeding in teleost.
In our functional studies with carp pituitary cells, interestingly, the potency and efficacy of the other TAC4 gene product, namely HK1, was found to be ineffective for UTS1, PRL, and SLα mRNA expression. In addition, HK1 could barely activate any NKR isoforms in HEK-293T cells. Sequence analysis showed that HK1 from different fish species was also very unique in having a well-conserved Phe to Val mutation in the signature motif “VXGLM” located in the C-terminal. The functional role of the new signature motif “VXGLM” in HK1 was currently unclear, but an “FXGLL” motif has been previously reported in human EKC and EKD despite the fact that the other gene products of human TAC4, namely EKA and EKB, still have the original “FXGLM” signature sequence [49]. In human patients with TAC3 gene mutations, a Thr for Met mutation in the “FXGLM” motif of NKB was known to cause hypogonadism or even infertility in adulthood [1]. To confirm that the low intrinsic activity of HK1 was caused by the Phe to Val mutation in the C-terminal of the signature motif, the Val to Phe mutant of HK1, namely HK1-V7F, was synthesized and tested for its bioactivity on SLα regulation in carp pituitary cells. In this case, the stimulatory effects of the HK1-V7F with the regeneration of the original “FXGLM” motif on SLα mRNA expression was markedly enhanced to the levels for HK2. These findings, as a whole, indicated that (i) the F residue in the “FXGLM” motif played a crucial role in the bioactivity of tachykinins in the fish model, and (ii) the Phe to Val mutation form in the “VXGLM” motif represented the molecular determinant in HK1 leading to weak agonistic actions on SLα regulation, presumably by interfering the interaction with target receptors. Our findings were also in agreement with the previous studies in mammalian cell models, e.g., COS-7 cells with NK3 receptor [9,12], confirming that the C-terminal “FXGLM” motif was essential for the bioactivity and receptor binding [32,50].
In rats, central administration of SP and EKA/B is known to induce pain-related behaviors, e.g., scratching paw withdrawal and thermal hyperalgesia [26,51], mainly through activation of NK1R expressed in the brain and/or spinal cord [52]. However, these stimulatory effects can be blocked by simultaneous treatment with EKC/EKD, and apparently, the “antagonistic effects” on NK1R activation induced by SP or EKA/B are mediated through the “FXGLL” motif of EKC/EKD [26,53,54]. These previous findings in mammalian models indicated that the Phe to Val mutation found in the “VXGLM” motif of HK1 might also have a functional impact on the receptor interaction and/or biological actions of other tachykinins in fish species. However, our present study found that HK1 could not block HK2-induced UTS1, PRL, and SLα mRNA expression. These results suggested that the V to F substitution in the signature motif of HK1 did not contribute to its “antagonistic effect” for HK2 on NKR binding/activation.
In summary, in order to clarify the pituitary actions of TAC4 gene products in lower vertebrates, the TAC4 and six NKR isoforms were isolated from grass carp. Sequence analysis showed that grass carp TAC4 could encode two mature peptides (namely HK1 and HK2), in which HK2 retained the typical FXGLM motif of tachyinin in the C-terminal, while HK1 contained a mutant VFGLM motif. The ligand-receptor selectivity showed that HK2 could activate all six NKR isoforms but with the highest activity for NK2R. Interestingly, HK1 could display a very weak activation for NK2R and NK3Ra2. Using grass carp pituitary cells as a model, we found that HK2 could significantly induce PRL, SLα, UTS1, NMB1, and CART mRNA expression mediated by NK2R and NK3R coupled with cAMP/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK II cascades (Figure 10). These results indicated that HK2 might be involved in the regulation of reproduction, growth metabolism, and feeding in teleost. However, the potency and efficacy of HK1 was found to be ineffective for the six pituitary genes’ expression. Further studies indicated that the F to V mutation in “VXGLM” motif in HK1 contributed to its weak pituitary actions. Furthermore, we found that HK1 could not contribute to its “antagonistic effect” for HK2 on NKR binding/activation.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijms222312893/s1.

Author Contributions

Data curation, X.S. and C.Y.; formal analysis, L.Z. and X.Q.; funding acquisition, G.H. and Z.Y.; investigation, X.S., C.X. and T.C.; methodology, X.S. and Y.X.; resources, X.S. and C.X.; software, L.Z. and X.S.; supervision, G.H. and Z.Y.; writing—original draft, X.S.; writing—review and editing, G.H. All authors have read and agreed to the published version of the manuscript.

Funding

Funding support was provided by the Fundamental Research Funds for the Central Universities (2662019PY006 to G.F.), National Key R&D Program of China (2018YFD0900205 to Z.Y.), China Postdoctoral Science Foundation (2019M662747 to G.H.), and Hubei Province Postdoctoral Science Foundation (237934 to G.H.).

Institutional Review Board Statement

All experimental procedures were approved by the Huazhong Agricultural University Administrative Panel for Laboratory Animal Care (Ethical Approval No. HBAC20091138; Date: 15 November 2009).

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author.

Acknowledgments

This article is dedicated to Anderson O.L. Wong (University of Hong Kong, China) for his genuine interest in training young scientists in the field of comparative endocrinology.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AC, adenylate cyclase; cAMP, cyclic adenosine monophosphate; TAC, tachykinin; NKR, neurokinin receptor; HK, hemokinin; PKA, protein kinase A; PLC, phospholipase C; IP3, inositol trisphosphate PKC, protein kinase C; CaM, calmodulin; CaMK, calmodulin kinase; GPCR, G protein coupled receptor; SLα, somatolactin α; PRL, prolactin; NMB, neuromedin B; UTS, urotensin; CART, cocaine-and amphetamine-regulated transcript; NFAT, nuclear factor of activated T cells; TMD, transmembrane domain; WGD, whole genome duplication.

References

  1. Lin, C.; Chen, W.N.; Chen, C.J.; Lin, Y.W.; Zimmer, A.; Chen, C.C. An antinoci-ceptive role for substance P in acid-induced chronic muscle pain. Proc. Natl. Acad. Sci. USA 2012, 109, E76–E83. [Google Scholar] [CrossRef] [Green Version]
  2. Ang, S.F.; Moochhala, S.M.; Macary, P.A.; Bhatia, M. Hydrogen sulfide and neurogenic inflammation in polymicrobial sepsis: Involvement of substance P and ERK-NF-κB signaling. PLoS ONE 2017, 6, e24535. [Google Scholar] [CrossRef]
  3. Chitrang, T.; Shan, X.; Loraine, Y.C.; Dhiraj, K.; Jenna, H.; Sarah, A.; Kristy, H.; Henriette, K.; Yeo, G.S.H.; Diego, P.T. Tachykinin-1 in the central nervous system regulates adiposity in rodents. Endocrinology 2015, 156, 1714–1723. [Google Scholar]
  4. Meyer, A.; Schartl, M. Gene and genome duplications in vertebrates: The one-to-f-our (-to-eight in fish) rule and the evolution of novel gene functions. Curr. Opin. Cell Biol. 1999, 11, 699–704. [Google Scholar] [CrossRef] [Green Version]
  5. Hu, G.F.; He, M.; Ko, W.; Wong, A. TAC1 GeneProducts RegulatePituitary Hormone Secretion and Gene Expression in Prepubertal Grass Carp Pituitary Cells. Endocrinology 2017, 158, 1776–1797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Abdelrahman, A.M.; Pang, C. Effect of Substance P on Venous Tone in Conscious Rats. J. Cardiovac. Pharmacol. 2005, 45, 49. [Google Scholar] [CrossRef]
  7. Lecci, A.; Capriati, A.; Altamura, M.; Maggi, C.A. Tachykinins and tachykinin receptors in the gut, with special reference to NK2 receptors in human. Auton. Neurosci. 2006, 126, 232–249. [Google Scholar] [CrossRef] [PubMed]
  8. Bae, Y.K.; Kani, S.; Shimizu, T.; Tanabe, K.; Nojima, H.; Kimura, Y.; Higashijima, S.I.; Hibi, M. Anatomy of zebrafish cerebellum and screen for mutations affecting its development. Dev. Biol. 2009, 330, 406–426. [Google Scholar] [CrossRef] [Green Version]
  9. Topaloglu, A.K.; Reimann, F.; Guclu, M.; Yalin, A.S.; Semple, R.K. Tac3 and tacr3 mutations in familial hypogonadotropic hypogonadism reveal a key role for neurokinin b in the central control of reproduction. Nat. Genet. 2009, 41, 354–358. [Google Scholar] [CrossRef] [Green Version]
  10. Goodman, R.L.; Coolen, L.M.; Lehman, M.N. A role for neurokinin b in pulsatile gnrh secretion in the ewe. Neuroendocrinology 2014, 99, 18. [Google Scholar] [CrossRef]
  11. Hu, G.F.; Lin, C.; He, M.; Wong, A. Neurokinin B and reproductive functions: “KNDy neuron” model in mammals and the emerging story in fish. Gen. Comp. Endocrinol. 2014, 208, 94–108. [Google Scholar] [CrossRef] [Green Version]
  12. Biran, J.; Palevitch, O.; Ben-Dor, S.; Levavi-Sivan, B. Neurokinin Bs and neurokinin B receptors in zebrafish-potential role in controlling fish reproduction. Proc. Natl. Acad. Sci. USA 2012, 109, 10269–10274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhang, Y.; Lu, L.; Furlonger, C.; Wu, G.; Paige, C. Hemokinin is a hematopoieticspecific tachykinin that regulates B lymphopoiesis. Nat. Immunol. 2000, 1, 392–397. [Google Scholar] [CrossRef] [PubMed]
  14. Kurtz, M.M.; Wang, R.; Clements, M.K.; Cascieri, M.A.; Austin, C.P.; Cunningh-am, B.R.; Chicchi, G.G.; Liu, Q. Identification, localization and receptor characterization of novel mammalian substance P-like peptides. Gene 2002, 296, 205–212. [Google Scholar] [CrossRef]
  15. Page, N.M.; Bell, N.J.; Gardiner, S.M.; Manyonda, I.T.; Brayley, K.J.; Strange, P.G.; Lowry, P.J. Characterization of the endokinins: Human tachykinins with cardiovascular activity. Proc. Natl. Acad. Sci. USA 2003, 100, 6245–6250. [Google Scholar] [CrossRef] [Green Version]
  16. Steinhoff, M.S.; Mentzer, B.V.; Geppetti, P.; Pothoulakis, C.; Bunnett, N.W. Tac-hykinins and their receptors: Contributions to physiological control and the mechanisms of disease. Physiol. Rev. 2014, 94, 265–301. [Google Scholar] [CrossRef] [Green Version]
  17. Zieglgänsberger, W. Substance P and pain chronicity. Cell Tissue Res. 2019, 375, 227–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Dai, L.; Perera, D.S.; King, D.W.; Southwell, B.R.; Burcher, E.; Liu, L. Hemoki-nin-1 stimulates prostaglandin E₂ production in human colon through activationof cyclooxygenase-2 and inhibition of 15-hydroxyprostaglandin dehydrogenase. J. Pharmacol. Exp. Ther. 2012, 340, 27–36. [Google Scholar] [CrossRef] [Green Version]
  19. Tsilioni, I.; Russell, J.; Stewart, R.; Gleason, T.; Theoharides, T.C. Neuropeptides CRH, SP, HK-1, and Inflammatory Cytokines IL-6 and TNF Are Increased in Serum of Patients with Fibromyalgia Syndrome, Implicating Mast Cells. J. Pharmacol. Exp. Ther. 2016, 356, 664–672. [Google Scholar] [CrossRef] [Green Version]
  20. Borbély, É.; Helyes, Z. Role of hemokinin-1 in health and disease. Neuropeptides 2017, 64, 9–17. [Google Scholar] [CrossRef]
  21. Wong, A.O.; Ng, S.; Lee, E.K.; Leung, R.C.; Ho, W.K. Somatostatin inhibits (d-Arg6, Pro9-NEt) salmon gonadotropin-releasing hormone and dopamine D1-stim-ulated growth hormone release from perifused pituitary cells of chinese grass carp, Ctenopharyngodon idellus. Gen. Comp. Endocrinol. 1998, 110, 29–45. [Google Scholar] [CrossRef] [PubMed]
  22. Penn, E.F.; Ather, J.N.; Lecci, A.; Candenas, M.L.; Patak, E.; Pinto, F.M.; Maggi, C.A. Tachykinins and tachykinin receptors: A growing family. Life Sci. 2004, 74, 1445–1463. [Google Scholar]
  23. Hu, G.; He, M.; Ko, W.K.W.; Lin, C.; Wong, A.O.L. Novel pituitary actions of TAC3 gene products in fish model: Receptor specificity and signal transduction for prolactin and somatolactin α regulation by neurokinin B (NKB) and NKB-related peptide in carp pituitary cells. Endocrinology 2014, 155, 3582–3596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. He, X. Rapid subfunctionalization accompanied by prolonged and substantial n-eofunctionalization in duplicate gene evolution. Genetics 2005, 169, 1157–1164. [Google Scholar] [CrossRef] [Green Version]
  25. Khorasani, S.; Boroumand, N.; Arab, F.L.; Hashemy, S.I. The immunomodulatory effects of tachykinins and their receptors. J. Cell. Biochem. 2020, 121, 3031–3041. [Google Scholar] [CrossRef]
  26. Yang, Y.; Dong, S. Effects of Endokinin A/B and Endokinin C/D on the modulation of pain in mice. Peptides 2010, 31, 94–100. [Google Scholar] [CrossRef]
  27. Naono, R.; Nakayama, T.; Ikeda, T.; Matsushima, O.; Nishimori, T. Effect of the carboxyl-terminal of endokinins on SP-induced pain-related behavior. Biochem. Biophys. Res. Commun. 2009, 378, 182–185. [Google Scholar] [CrossRef]
  28. Almeida, T.A.; Rojo, J.; Nieto, P.M.; Pinto, F.M.; Hernandez, M.; Martín, J.D.; Candenas, M.L. Tachykinins and tachykinin receptors: Structure and activity relationships. Curr. Med. Chem. 2004, 11, 2045–2081. [Google Scholar] [CrossRef]
  29. Duffy, R.A.; Hedrick, J.A.; Randolph, G.; Morgan, C.A.; Cohen-Williams, M.E.; Vassileva, G.; Lachowicz, J.E.; Laverty, M.; Maguire, M.; Shan, L.S. Centrally administered hemokinin-1 (HK-1), a neurokinin NK1 receptor agonist, produces substance P-like behavioral effects in mice and gerbils. Neuropharmacology 2003, 45, 242–250. [Google Scholar] [CrossRef]
  30. Morteau, O.; Lu, B.; Gerard, C.; Gerard, N.P. Hemokinin 1 is a full agonist at the substance p receptor. Nat. Immunol. 2002, 2, 1088. [Google Scholar] [CrossRef]
  31. Bellucci, F.; Carini, F.; Catalani, C.; Cucchi, P.; Lecci, A.; Meini, S.; Patacchini, R.; Quartara, L.; Ricci, R.; Tramontana, M.; et al. Pharmacological profile of the novel mammalian tachykinin, hemokinin 1. Br. J. Pharmacol. 2002, 135, 266–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Aczél, T.; Kecskés, A.; Kun, J.; Szenthe, K.; Bánáti, F.; Szathmary, S.; Herczeg, R.; Urbán, P.; Gyenesei, A.; Gaszner, B.; et al. Hemokinin-1 Gene Expression Is Upregulated in Trigeminal Ganglia in an Inflammatory Orofacial Pain Model: Potential Role in Peripheral Sensitization. Int. J. Mol. Sci. 2020, 21, 2938. [Google Scholar] [CrossRef] [PubMed]
  33. Onuma, T.A.; Ban, M.; Makino, K.; Katsumata, H.; Hu, W.W.; Ando, H.; Fukuwaka, M.A.; Azumaya, T.; Urano, A. Changes in gene expression for GH/PRL/SL family hormones in the pituitaries of homing chum salmon during ocean migration through upstream migration. Gen. Comp. Endocrinol. 2010, 166, 537–548. [Google Scholar] [CrossRef] [PubMed]
  34. Degani, G.; Yom-Din, S.; Goldberg, D.; Jackson, K. cDNA cloning of blue gour-ami (Trichogaster trichopterus) prolactin and its expression during the gonadal cycles of males and females. J. Endocrinol. Investig. 2010, 33, 7–12. [Google Scholar] [CrossRef] [PubMed]
  35. Tse, M.; Wong, G.; Ping, X.; Cheng, C.; Chan, K.M. Down-regulation of goldfis-h (Carassius auratus) prolactin gene expression by dopamine and thyrotropin releasing hormone. Gen. Comp. Endocrinol. 2008, 155, 729–741. [Google Scholar] [CrossRef] [PubMed]
  36. Noh, G.E.; Rho, S.; Yong, J.C.; Min, B.H. Gene encoding prolactin in cinnamon clownfish Amphiprion melanopus and its expression upon acclimation to low salinities. Aquat. Biosyst. 2013, 9, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Whittington, C.M.; Wilson, A.B. Wilson. The role of prolactin in fish reproduct-ion. Gen. Comp. Endocrinol. 2013, 191, 123–136. [Google Scholar] [CrossRef] [Green Version]
  38. Shu, Y.Q.; Lou, Q.Y.; Dai, Z.R.; Dai, X.Y.; He, J.Y.; Hu, W.; Yin, Z. The basal function of teleost prolactin as a key regulator on ion uptake identified with zebrafish knockout models. Sci. Rep. 2016, 6, 18579. [Google Scholar] [CrossRef]
  39. Benedet, S.; Björnsson, B.T.; Taranger, G.L.; Andersson, E. Cloning of somatolactin alpha, beta forms and the somatolactin receptor in Atlantic salmon: Seasonalexpression profile in pituitary and ovary of maturing female broodstock. Reprod. Biol. Endocrinol. 2008, 6, 42. [Google Scholar] [CrossRef] [Green Version]
  40. Wan, G.H.; Chan, K.M. A study of somatolactin acitons by ectopic expression in transgenic zebrafish larvae. J. Mol. Endocinol. 2010, 45, 301–315. [Google Scholar] [CrossRef] [Green Version]
  41. Bertolesi, G.E.; McFarlane, S. Melanin-concentrating hormone like and somatolactin. Ateleost-specific hypothalamic-hypophyseal axis system linking physiologicaland morphological pigmentation. Pigment Cell Melanoma Res. 2021, 34, 564–574. [Google Scholar]
  42. Xu, S.H.; Zhou, L.L.; Chen, X.Y.; Hu, Q.Y.; Shi, X.T.; Xia, C.H.; Zhang, H.Y.; Jia, J.Y.; Yin, Z.; Hu, G.F. Novel pituitary actions of NKB for anorectic peptides regulation in grass carp. Aquaculture 2021, 531, 735857. [Google Scholar] [CrossRef]
  43. Lambert, R.; Couceyro, R.; Koylu, E.; Ling, N.; Kuhar, M. A role for novel CART peptide fragments in the central control of food intake. Neuropeptides 1997, 31, 620–621. [Google Scholar]
  44. Volkoff, H.; Peter, R.E. Effects of CART peptides on food consumption, feedingand associated behaviors in the goldfish, Carassius-auratus: Actions on neuro-peptide Y and orexin A-induced feeding. Brain Res. 2000, 887, 125–133. [Google Scholar] [CrossRef]
  45. Tachibana, T.; Matsuda, K.; Khan, S.I.; Ueda, H.; Cline, M.A. Feeding and drin-king response following central administrations of bombesin-like peptidesin chi-cks. Comp. Biochem. Physiol. Part A 2010, 156, 394–399. [Google Scholar] [CrossRef]
  46. Sayegh, A.I. The role of bombesin and bombesin-related peptides in the shortt-erm control of food intake. Prog. Mol. Biol. Transl. 2013, 114, 343–370. [Google Scholar]
  47. Kuperman, Y.; Chen, A. Urocortins: Emerging metabolic and energy homeostasi-s perspectives. Trends Endocrinol. Metab. 2008, 19, 122–129. [Google Scholar] [CrossRef]
  48. Bernier, N.J.; Peter, R.E. Appetite-suppressing e ects of urotensin I and cortico- tropin-releasing hormone in goldfish (Carassius auratus). Neuroendocrinology 2001, 73, 248. [Google Scholar] [CrossRef]
  49. Page, N.M. Hemokinins and endokinins. Cell. Mol. Life Sci. 2004, 61, 1652–1663. [Google Scholar] [CrossRef]
  50. Zhou, W.; Li, S.; Liu, Y.; Qi, X.; Chen, H.; Cheng, C.H.K.; Liu, X.; Zhang, Y.; Lin, H. The evolution of tachykinin/tachykinin receptor (TAC/TACR) in vertebrates and molecular identification of the TAC3/TACR3 system in zebrafish (Danio rerio). Mol. Cell. Endocrinol. 2012, 361, 202–212. [Google Scholar] [CrossRef]
  51. Grace, R.C.; Chandrashekar, I.R.; Cowsik, S.M. Solution structure of the tachyk-inin peptide eledoisin. Biophys. J. 2003, 84, 655–664. [Google Scholar] [CrossRef] [Green Version]
  52. Ganjiwale, A.D.; Rao, G.S.; Cowsik, S.M. Molecular modeling of neurokinin B and tachykinin NK3 receptor complex. J. Chem. Inf. Model. 2011, 51, 2932–2938. [Google Scholar] [CrossRef] [PubMed]
  53. Naono, R.; Nakayama, T.; Ikeda, T.; Matsushima, O.; Nishimori, T. Leucine at the carboxyl-terminal of endokinins C and D contributes to elicitation of the antagonistic effect on substance P in rat pain processing. Brain Res. 2007, 1165, 71–80. [Google Scholar] [CrossRef] [PubMed]
  54. Yoshioka, D.; Takebuchi, F.; Nishimori, T.; Naono, R.; Ikeda, T.; Nakayama, T. Intrathecal administration of the common carboxyl-terminal decapeptide in end-okinin A and endokinin B evokes scratching behavior and thermal hyperalgesiain therat. Neurosci. Lett. 2006, 410, 193–197. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Sequence analysis of tachykinin 4 (TAC4) and six neurokinin receptors (NKRs) in grass carp. (A) Sequence analysis of TAC4 preprohormones among different vertebrates. The protein sequence of grass carp TAC4 was aligned with that reported in other vertebrates. The two mature peptides residues in these sequences were labeled in blue or red color, respectively. (B) Transcript level of TAC4 and six NKRs in various brain subregions and pituitary. Total RNA was extracted from various brain areas or pituitary of grass carp, and RT-PCR was performed by using specific primers for grass carp TAC4 and NKRs. (C) Sequences were compared among ten tachykinin peptides in grass carp. (D) Ribbon representation of grass carp HK1 and HK2 structural model. The amino acids with hydrophobic side chains were colored blue, while those with hydrophilic side chains were colored red. (E) The phylogenetic analysis of vertebrate TAC4 was performed using the neighbor-joining method (MEGA 6.0), and the grass carp TAC4 was highlighted in the red box. (F) The phylogenetic analysis of vertebrate six NKRs was performed by using the neighbor-joining method (MEGA 6.0).
Figure 1. Sequence analysis of tachykinin 4 (TAC4) and six neurokinin receptors (NKRs) in grass carp. (A) Sequence analysis of TAC4 preprohormones among different vertebrates. The protein sequence of grass carp TAC4 was aligned with that reported in other vertebrates. The two mature peptides residues in these sequences were labeled in blue or red color, respectively. (B) Transcript level of TAC4 and six NKRs in various brain subregions and pituitary. Total RNA was extracted from various brain areas or pituitary of grass carp, and RT-PCR was performed by using specific primers for grass carp TAC4 and NKRs. (C) Sequences were compared among ten tachykinin peptides in grass carp. (D) Ribbon representation of grass carp HK1 and HK2 structural model. The amino acids with hydrophobic side chains were colored blue, while those with hydrophilic side chains were colored red. (E) The phylogenetic analysis of vertebrate TAC4 was performed using the neighbor-joining method (MEGA 6.0), and the grass carp TAC4 was highlighted in the red box. (F) The phylogenetic analysis of vertebrate six NKRs was performed by using the neighbor-joining method (MEGA 6.0).
Ijms 22 12893 g001
Figure 2. Receptor selectivity of HK1, HK2, and HK1-V7F. The nuclear factor of activated T-cells (NFAT)-Luc reporter together with NK1Ra, NK1Rb, NK2R, NK3Rb, NK3Ra1, or NK3Ra2, were transiently transfected into HEK-293 cells, respectively. The cells were incubated with increasing levels of grass carp HK1, HK2, or HK1-V7F (0.1–1000 nM), respectively. The luciferase activities were normalized against renilla expression in the same sample to adjust for potential variations in transfection efficiency. Each point is determined quadruplicate and is given as a mean ± SEM. The groups denoted by different letters represent significant differences at p-value < 0.05 (ANOVA followed Dunnett’s test).
Figure 2. Receptor selectivity of HK1, HK2, and HK1-V7F. The nuclear factor of activated T-cells (NFAT)-Luc reporter together with NK1Ra, NK1Rb, NK2R, NK3Rb, NK3Ra1, or NK3Ra2, were transiently transfected into HEK-293 cells, respectively. The cells were incubated with increasing levels of grass carp HK1, HK2, or HK1-V7F (0.1–1000 nM), respectively. The luciferase activities were normalized against renilla expression in the same sample to adjust for potential variations in transfection efficiency. Each point is determined quadruplicate and is given as a mean ± SEM. The groups denoted by different letters represent significant differences at p-value < 0.05 (ANOVA followed Dunnett’s test).
Ijms 22 12893 g002
Figure 3. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of HK2-regulated different expressed genes (DEGs) in grass carp pituitary cells. (A) In GO analysis, DEGs were composed of three parts: cell component (CC), biological process (BP), and molecular function (MF). (B) KEGG analysis were used to enrich the top 10 KEGG pathways, including up-regulated and down-regulated pathways, respectively. Up, up-regulated genes; down, down-regulated genes; count, the number of DEGs.
Figure 3. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of HK2-regulated different expressed genes (DEGs) in grass carp pituitary cells. (A) In GO analysis, DEGs were composed of three parts: cell component (CC), biological process (BP), and molecular function (MF). (B) KEGG analysis were used to enrich the top 10 KEGG pathways, including up-regulated and down-regulated pathways, respectively. Up, up-regulated genes; down, down-regulated genes; count, the number of DEGs.
Ijms 22 12893 g003
Figure 4. DEGs were enriched in the reproduction function, feeding regulation, growth factor, immune response, and hormone activity of trashcription. The pink color indicates upregulated DEGs, while the green color indicates downregulated DEGs.
Figure 4. DEGs were enriched in the reproduction function, feeding regulation, growth factor, immune response, and hormone activity of trashcription. The pink color indicates upregulated DEGs, while the green color indicates downregulated DEGs.
Ijms 22 12893 g004
Figure 5. TAC4 gene products (HK1 and HK2) induced PRL (A) and SLα (B) mRNA expression in grass carp pituitary cells. Time course of grass carp HK1 (1 μM) and HK2 (1 μM) on PRL (A) and SLα (B) mRNA expression in grass carp pituitary cells. Dose-dependence of 24-h treatment with increasing levels of HK1 and HK2 (0.1–1000 nM) on PRL (A) and SLα (B) mRNA expression in grass carp pituitary cells. After drug treatment, the total RNA of the cells was extracted by the Trizol method, and the expression of various genes was detected by RT-PCR. Data presented were expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Figure 5. TAC4 gene products (HK1 and HK2) induced PRL (A) and SLα (B) mRNA expression in grass carp pituitary cells. Time course of grass carp HK1 (1 μM) and HK2 (1 μM) on PRL (A) and SLα (B) mRNA expression in grass carp pituitary cells. Dose-dependence of 24-h treatment with increasing levels of HK1 and HK2 (0.1–1000 nM) on PRL (A) and SLα (B) mRNA expression in grass carp pituitary cells. After drug treatment, the total RNA of the cells was extracted by the Trizol method, and the expression of various genes was detected by RT-PCR. Data presented were expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Ijms 22 12893 g005
Figure 6. TAC4 gene products (HK1 and HK2) induced CART2, NMB1, and UTS1 mRNA expression in grass carp pituitary cells. Time course of grass carp HK1 (1 μM) and HK2 (1 μM) on CART2 (A), NMB1 (B), and UTS1 (C) mRNA expression in grass carp pituitary cells. Dose-dependence of 24-h treatment with increasing levels of HK1 or HK2 (0.1–1000 nM) on CART2 (A), NMB1 (B), and UTS1 (C) mRNA expression in grass carp pituitary cells. After drug treatment, the total RNA of the cells was extracted by the Trizol method, and the expression of various genes were detected by RT-PCR. Data presented were expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Figure 6. TAC4 gene products (HK1 and HK2) induced CART2, NMB1, and UTS1 mRNA expression in grass carp pituitary cells. Time course of grass carp HK1 (1 μM) and HK2 (1 μM) on CART2 (A), NMB1 (B), and UTS1 (C) mRNA expression in grass carp pituitary cells. Dose-dependence of 24-h treatment with increasing levels of HK1 or HK2 (0.1–1000 nM) on CART2 (A), NMB1 (B), and UTS1 (C) mRNA expression in grass carp pituitary cells. After drug treatment, the total RNA of the cells was extracted by the Trizol method, and the expression of various genes were detected by RT-PCR. Data presented were expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Ijms 22 12893 g006
Figure 7. The receptor specificity and signal transduction for PRL and SLα regulation by HK2 in grass carp pituitary cells. (A) Effects of NKR subtype-specific antagonists on HK2-induced PRL and SLα mRNA expression. Pituitary cells were challenged for 24 h with grass carp HK2 (1 μM) in the presence or absence of the NK1R antagonist Rolaptiant (10 μM) or NK3R antagonist SB22200 (10 μM), respectively. (B) Effects of 24-h co-treatment with AC inhibitor MDL12330A (10 μM) or PKA inhibitor H89 (10 μM) on HK2-induced PRL and SLα mRNA expression. (C) Effects of 24-h co-treatment with PLC inhibitor U73122, PKC inhibitor GF109203X (10 μM), or IP3 receptor blocker 2-APB (10 μM) on HK2-induced PRL and SLα mRNA expression. (D) Effects of 24-h co-treatment with the VSCC inhibitor nifedipine (10 μM), CaM antagonist calmidazolium (1 μM) or CaMK-II blocker KN62 (10 μM) on HK2-induced PRL and SLα mRNA expression. After drug treatment, the total RNA of the cells was extracted by the Trizol method, and the expression of various genes was detected by RT-PCR. Data presented were expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Figure 7. The receptor specificity and signal transduction for PRL and SLα regulation by HK2 in grass carp pituitary cells. (A) Effects of NKR subtype-specific antagonists on HK2-induced PRL and SLα mRNA expression. Pituitary cells were challenged for 24 h with grass carp HK2 (1 μM) in the presence or absence of the NK1R antagonist Rolaptiant (10 μM) or NK3R antagonist SB22200 (10 μM), respectively. (B) Effects of 24-h co-treatment with AC inhibitor MDL12330A (10 μM) or PKA inhibitor H89 (10 μM) on HK2-induced PRL and SLα mRNA expression. (C) Effects of 24-h co-treatment with PLC inhibitor U73122, PKC inhibitor GF109203X (10 μM), or IP3 receptor blocker 2-APB (10 μM) on HK2-induced PRL and SLα mRNA expression. (D) Effects of 24-h co-treatment with the VSCC inhibitor nifedipine (10 μM), CaM antagonist calmidazolium (1 μM) or CaMK-II blocker KN62 (10 μM) on HK2-induced PRL and SLα mRNA expression. After drug treatment, the total RNA of the cells was extracted by the Trizol method, and the expression of various genes was detected by RT-PCR. Data presented were expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Ijms 22 12893 g007
Figure 8. The receptor specificity and signal transduction for CART2, NMB1, and UTS1 regulation by HK2 in grass carp pituitary cells. (A) The pituitary cells were treated for 24 h with grass carp HK2 (1 μM) in the presence or absence of NK1R antagonist Rolaptiant (10 μM) or NK3R antagonist SB222200 (10 μM). (B) Effects of 24-h co-treatment with an AC inhibitor MDL12330A (10 μM) or PKA inhibitor H89 (10 μM) on HK2-induced CART2, UTS1, or NMB1 mRNA expression. (C) Effects of 24h co-treatment with a PLC inhibitor U73122, PKC inhibitor GF109203X (10 μM), or IP3 receptor blocker 2-APB (10 μM) on HK2-induced CART2, UTS1 or NMB1 mRNA expression. (D) Effects of 24-h co-treatment with the VSCC inhibitor nifedipine (10 μM), CaM antagonist calmidazolium (1 μM), or CaMK-II blocker KN62 (10 μM) on HK2-induced CART2, UTS1 or NMB1 mRNA expression.
Figure 8. The receptor specificity and signal transduction for CART2, NMB1, and UTS1 regulation by HK2 in grass carp pituitary cells. (A) The pituitary cells were treated for 24 h with grass carp HK2 (1 μM) in the presence or absence of NK1R antagonist Rolaptiant (10 μM) or NK3R antagonist SB222200 (10 μM). (B) Effects of 24-h co-treatment with an AC inhibitor MDL12330A (10 μM) or PKA inhibitor H89 (10 μM) on HK2-induced CART2, UTS1, or NMB1 mRNA expression. (C) Effects of 24h co-treatment with a PLC inhibitor U73122, PKC inhibitor GF109203X (10 μM), or IP3 receptor blocker 2-APB (10 μM) on HK2-induced CART2, UTS1 or NMB1 mRNA expression. (D) Effects of 24-h co-treatment with the VSCC inhibitor nifedipine (10 μM), CaM antagonist calmidazolium (1 μM), or CaMK-II blocker KN62 (10 μM) on HK2-induced CART2, UTS1 or NMB1 mRNA expression.
Ijms 22 12893 g008
Figure 9. Phe to Val mutation in FXGLM motif in PRL, SLα, and UTS1 regulation by HK2 in grass carp pituitary cells. (A) Effects of HK1-V7F on PRL, SLα, and UTS1 mRNA expression in grass carp pituitary cells. In this experiment, grass carp pituitary cells were incubated for 24 h with increasing levels of HK1-V7F (0.1–1000 nM). (B) Suppressed effects of HK1 on HK2-induced PRL, SLα, and UTS1 mRNA expression in grass carp pituitary cells. Pituitary cells were challenged for 24 h with grass carp HK2 (0.1 µM) in the presence or absence of HK1 (1 µM) and HK1 (10 µM). In these experiment, Trizol method was used to extract total cell RNA, and RT-PCR was used to detect PRL, SLα, and UTS1 mRNA expression. Data presented are expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Figure 9. Phe to Val mutation in FXGLM motif in PRL, SLα, and UTS1 regulation by HK2 in grass carp pituitary cells. (A) Effects of HK1-V7F on PRL, SLα, and UTS1 mRNA expression in grass carp pituitary cells. In this experiment, grass carp pituitary cells were incubated for 24 h with increasing levels of HK1-V7F (0.1–1000 nM). (B) Suppressed effects of HK1 on HK2-induced PRL, SLα, and UTS1 mRNA expression in grass carp pituitary cells. Pituitary cells were challenged for 24 h with grass carp HK2 (0.1 µM) in the presence or absence of HK1 (1 µM) and HK1 (10 µM). In these experiment, Trizol method was used to extract total cell RNA, and RT-PCR was used to detect PRL, SLα, and UTS1 mRNA expression. Data presented are expressed as mean ± SEM, and the differences between groups were significant at p-value < 0.05 by labeling diverse letters.
Ijms 22 12893 g009
Figure 10. Working model of TAC4 gene products in the pituitary. TAC4 could encode two mature peptides, namely HK1 and HK2. HK2 could stimulate PRL, SLα, UTS1, NMB1, and CART2 mRNA expression mediated by NK2R and NK3R coupled with cAMP/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK II cascades. In contrast, HK1 was found to be ineffective for the six pituitary genes’ expression.
Figure 10. Working model of TAC4 gene products in the pituitary. TAC4 could encode two mature peptides, namely HK1 and HK2. HK2 could stimulate PRL, SLα, UTS1, NMB1, and CART2 mRNA expression mediated by NK2R and NK3R coupled with cAMP/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK II cascades. In contrast, HK1 was found to be ineffective for the six pituitary genes’ expression.
Ijms 22 12893 g010
Table 1. The up-regulated genes induced by HK2 in grass carp pituitary cells.
Table 1. The up-regulated genes induced by HK2 in grass carp pituitary cells.
GeneDescriptionFCGo-Biological Process
CART2Cocaine and amphetamine-regulated transcript 2 precursors27.23Negative regulation of appetite
CART5Cocaine and amphetamine-regulated transcript 5 precursors1.23Negative regulation of appetite
CKBBrain creatine kinase b1.63Creatine Kinase Activity
CRFB2Cytokine receptor family member b2 precursor1.77Interferon receptor activity
GHITMGrowth hormone-induced transmembrane protein1.21The components of the membrane
GPR186G protein-coupled receptor 1861.83Signal sensor activity
IGFBP1Insulin-like growth factor-binding protein 11.59Cell growth regulation
MPR63Mitochondrial ribosomal protein 631.35Mitochondrial Ribosome
NLRP12Proteins containing NACHT, LRR, and PYD domains 122.73ATP binding
NMB1Neurotonin B11.50Neuropeptide signaling pathway
NPAS4Neuron contains PAS domain protein 42.69DNA binding
NSFbN-ethylmaleimide sensitive factor b1.94ATP binding
PRL1Prolactin 11.71Hormonal activity
RBM24RNA binding protein 242.92Nucleotide binding
SDC2Heparan sulfate proteoglycan precursor1.62Gastrointestinal development
SERPINE2Glial cell-derived neuron precursor1.33Extracellular space
SID4Secreted immunoglobulin domain 4 precursor3.89Same protein binding
SLαSomatostatin alpha subunit1.81Hormonal activity
SN2Secretagogue 2 precursor1.78Secretory granules
UTS1Urotensin 17.31Hormonal activity
FC: fold change; FDR: false discovery rate.
Table 2. The down-regulated genes induced by HK2 in grass carp pituitary cells.
Table 2. The down-regulated genes induced by HK2 in grass carp pituitary cells.
GeneDescriptionFCGo-Biological Process
AIG1Androgen inducible protein 10.77The components of the membrane
CALCRLbCalcitonin gene-related peptide-like receptor b0.75Calcitonin receptor activity
DHRS13Dehydrogenase/reductase SDR family member 130.43Oxidoreductase activity
FGF14Fibroblast Growth Factor 140.80Growth factor activity
FGFR2Fibroblast Growth Factor Receptor 20.46Fibroblast growth factor binding
GHSR1Growth hormone secretagogue receptor type 10.14Signal sensor activity
GnIHR3Gonadotropin inhibits hormone receptor 30.68Receptor activity
GPER1G protein-coupled estrogen receptor 10.81G protein-coupled receptor activity
GRB2aGrowth factor receptor-binding protein 2a0.77Binding activity
IGF1RaInsulin-like growth factor type 1 receptor a0.68Insulin-like growth factor binding
INO1Inositol-3-phosphate synthase 10.78Inositol-3-phosphate synthase activity
MESDLDLR molecular chaperone MESD0.69Wnt signaling pathway
NRP2bNeurocin 2b0.71Metal ion binding
OGFRopioid growth factor receptor-like protein 10.36Receptor activity
OLFM2Olfactory protein 20.66Neural c cell development
S100A10S100 Calbindin A10b0.60Calcium ion binding
SAASerum amyloid A precursor0.72Response to bacteria
SSTR2aSomatostatin receptor 20.54Somatostatin receptor activity
SSTR3Somatostatin Receptor Type 30.57Somatostatin receptor activity
ZMYND11Zinc finger MYND domain-containing protein 110.49Zinc ion binding
FC: fold change; FDR: false discovery rate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shi, X.; Ye, C.; Qin, X.; Zhou, L.; Xia, C.; Cai, T.; Xie, Y.; Yin, Z.; Hu, G. Novel Pituitary Actions of TAC4 Gene Products in Teleost. Int. J. Mol. Sci. 2021, 22, 12893. https://doi.org/10.3390/ijms222312893

AMA Style

Shi X, Ye C, Qin X, Zhou L, Xia C, Cai T, Xie Y, Yin Z, Hu G. Novel Pituitary Actions of TAC4 Gene Products in Teleost. International Journal of Molecular Sciences. 2021; 22(23):12893. https://doi.org/10.3390/ijms222312893

Chicago/Turabian Style

Shi, Xuetao, Cheng Ye, Xiangfeng Qin, Lingling Zhou, Chuanhui Xia, Tianyi Cai, Yunyi Xie, Zhan Yin, and Guangfu Hu. 2021. "Novel Pituitary Actions of TAC4 Gene Products in Teleost" International Journal of Molecular Sciences 22, no. 23: 12893. https://doi.org/10.3390/ijms222312893

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop