Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.118
Filter
1.
Biochem Biophys Res Commun ; 717: 149992, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38714013

ABSTRACT

Insects have about 50 neuropeptide genes and about 70 genes, coding for neuropeptide G protein-coupled receptors (GPCRs). An important, but small family of evolutionarily related insect neuropeptides consists of adipokinetic hormone (AKH), corazonin, and AKH/corazonin-related peptide (ACP). Normally, insects have one specific GPCR for each of these neuropeptides. The tick Ixodes scapularis is not an insect, but belongs to the subphylum Chelicerata, which comprises ticks, scorpions, mites, spiders, and horseshoe crabs. Many of the neuropeptides and neuropeptide GPCRs occurring in insects, also occur in chelicerates, illustrating that insects and chelicerates are evolutionarily closely related. The tick I. scapularis is an ectoparasite and health risk for humans, because it infects its human host with dangerous pathogens during a blood meal. Understanding the biology of ticks will help researchers to prevent tick-borne diseases. By annotating the I. scapularis genome sequence, we previously found that ticks contain as many as five genes, coding for presumed ACP receptors. In the current paper, we cloned these receptors and expressed each of them in Chinese Hamster Ovary (CHO) cells. Each expressed receptor was activated by nanomolar concentrations of ACP, demonstrating that all five receptors were functional ACP receptors. Phylogenetic tree analyses showed that the cloned tick ACP receptors were mostly related to insect ACP receptors and, next, to insect AKH receptors, suggesting that ACP receptor genes and AKH receptor genes originated by gene duplications from a common ancestor. Similar duplications have probably occurred for the ligand genes, during a process of ligand/receptor co-evolution. Interestingly, chelicerates, in contrast to all other arthropods, do not have AKH or AKH receptor genes. Therefore, the ancestor of chelicerates might have lost AKH and AKH receptor genes and functionally replaced them by ACP and ACP receptor genes. For the small family of AKH, ACP, and corazonin receptors and their ligands, gene losses and gene gains occur frequently between the various ecdysozoan clades. Tardigrades, for example, which are well known for their survival in extreme environments, have as many as ten corazonin receptor genes and six corazonin peptide genes, while insects only have one of each, or none.


Subject(s)
Insect Hormones , Ixodes , Neuropeptides , Oligopeptides , Pyrrolidonecarboxylic Acid , Receptors, G-Protein-Coupled , Animals , Neuropeptides/metabolism , Neuropeptides/genetics , Insect Hormones/metabolism , Insect Hormones/genetics , Ixodes/metabolism , Ixodes/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Oligopeptides/metabolism , Oligopeptides/genetics , Oligopeptides/chemistry , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Phylogeny , Amino Acid Sequence , Cricetulus , CHO Cells , Insect Proteins/genetics , Insect Proteins/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics
2.
Biomolecules ; 14(4)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38672450

ABSTRACT

Motilin is a gastrointestinal hormone that is mainly produced in the duodenum of mammals, and it is responsible for regulating appetite. However, the role and expression of motilin are poorly understood during starvation and the weaning stage, which is of great importance in the seeding cultivation of fish. In this study, the sequences of Yangtze sturgeon (Acipenser dabryanus Motilin (AdMotilin)) motilin receptor (AdMotilinR) were cloned and characterized. The results of tissue expression showed that by contrast with mammals, AdMotilin mRNA was richly expressed in the brain, whereas AdMotilinR was highly expressed in the stomach, duodenum, and brain. Weaning from a natural diet of T. Limnodrilus to commercial feed significantly promoted the expression of AdMotilin in the brain during the period from day 1 to day 10, and after re-feeding with T. Limnodrilus the change in expression of AdMotilin was partially reversed. Similarly, it was revealed that fasting increased the expression of AdMotilin in the brain (3 h, 6 h) and duodenum (3 h), and the expression of AdMotilinR in the brain (1 h) in a time-dependent manner. Furthermore, it was observed that peripheral injection of motilin-NH2 increased food intake and the filling index of the digestive tract in the Yangtze sturgeon, which was accompanied by the changes of AdMotilinR and appetite factors expression in the brain (POMC, CART, AGRP, NPY and CCK) and stomach (CCK). These results indicate that motilin acts as an indicator of nutritional status, and also serves as a novel orexigenic factor that stimulates food intake in Acipenser dabryanus. This study lays a strong foundation for the application of motilin as a biomarker in the estimation of hunger in juvenile Acipenser dabryanu during the weaning phase, and enhances the understanding of the role of motilin as a novel regulator of feeding in fish.


Subject(s)
Feeding Behavior , Fishes , Motilin , Animals , Brain/metabolism , Fish Proteins/metabolism , Fishes/metabolism , Fishes/genetics , Fishes/physiology , Motilin/genetics , Motilin/metabolism , Motilin/pharmacology , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics
3.
J Toxicol Sci ; 49(4): 163-174, 2024.
Article in English | MEDLINE | ID: mdl-38556353

ABSTRACT

Mas-related G-protein-coupled receptor X2 (MRGPRX2), expressed on mast cells, is associated with drug-induced pseudo-allergic reactions. Although it is well known that there are differences of sensitivity between species in the pseudo-allergic reactions, no platform for evaluating a human risk of the pseudo-allergic reactions observed in nonclinical studies has been established. Valemetostat tosylate, developed as an anti-cancer drug, induced histamine release in a nonclinical study with dogs. The purpose of the current study was to identify the mechanism and assess the human risk of valemetostat-tosylate-induced histamine release using dog and human MRGPRX2-expressing cells. In an experiment with human or dog MRGPRX2-expressing cells, valemetostat tosylate caused activation of human and dog MRGPRX2. Importantly, the EC50 for dog MRGPRX2 was consistent with the Cmax value at which histamine release was observed in dogs. Furthermore, the EC50 for human MRGPRX2 was ca. 27-fold higher than that for dog MRGPRX2, indicating a species difference in histamine-releasing activity. In a clinical trial, histamine release was not observed in patients receiving valemetostat tosylate. In conclusion, an in vitro assay using human and animal MRGPRX2-expressing cells would be an effective platform to investigate the mechanism and predict the human risk of histamine release observed in nonclinical studies.


Subject(s)
Anaphylaxis , Histamine Release , Humans , Animals , Dogs , Anaphylaxis/chemically induced , Receptors, G-Protein-Coupled/genetics , Mast Cells , Nerve Tissue Proteins/genetics , Receptors, Neuropeptide/genetics
4.
Biomed Pharmacother ; 174: 116471, 2024 May.
Article in English | MEDLINE | ID: mdl-38547764

ABSTRACT

The mast cell receptor Mrgprb2, a mouse orthologue of human Mrgprx2, is known as an inflammatory receptor and its elevated expression is associated with various diseases such as ulcerative colitis. We aimed to elucidate the role of Mrgprb2/x2 and the effect of its ligands on a chemically induced murine colitis model. We showed that in Mrgprb2-/- mice, there is a differential regulation of cytokine releases in the blood plasma and severe colonic damages after DSS treatment. Unexpectedly, we demonstrated that known Mrgprb2/x2 agonists (peptide P17, P17 analogues and CST-14) and antagonist (GE1111) similarly increased the survival rate of WT mice subjected to 4% DSS-induced colitis, ameliorated the colonic damages of 2.5% DSS-induced colitis, restored major protein mRNA expression involved in colon integrity, reduced CD68+ and F4/80+ immune cell infiltration and restored cytokine levels. Collectively, our findings highlight the eminent role of Mrpgrb2/x2 in conferring a beneficial effect in the colitis model, and this significance is demonstrated by the heightened severity of colitis with altered cytokine releases and inflammatory immune cell infiltration observed in the Mrgprb2 knockout mice. Elevated expression of Mrgprb2 in WT colitis murine models may represent the organism's adaptive protective mechanism since Mrgprb2 knockout results in severe colitis. On the other hand, both agonist and antagonist of Mrgprb2 analogously mitigated the severity of colitis in DSS-induced colitis model by altering Mrgprb2 expression, immune cell infiltration and inflammatory cytokine releases.


Subject(s)
Colitis , Cytokines , Dextran Sulfate , Mice, Inbred C57BL , Mice, Knockout , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Mice , Cytokines/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Colon/pathology , Colon/drug effects , Colon/metabolism , Male , Disease Models, Animal , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics
5.
Eur J Pharmacol ; 969: 176457, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38395375

ABSTRACT

Neuropeptide FF (NPFF) plays a critical role in various physiological processes through the activation of neuropeptide FF receptor 1 and 2 (NPFFR1 and NPFFR2). Numerous evidence has indicated that NPFF exhibits opposite opioid-modulating effects on opioid-induced analgesia after supraspinal and spinal administrations, while the detailed role of NPFFR1 and NPFFR2 remains unclear. In this study, we employed pharmacological and genetic inhibition of NPFFR to investigate the modulating roles of central NPFFR1 and NPFFR2 in opioid-induced analgesia and hyperalgesia, using a male mouse model of acute fentanyl-induced analgesia and secondary hyperalgesia. Our findings revealed that intrathecal (i.t.) injection of the nonselective NPFFR antagonist RF9 significantly enhanced fentanyl-induced analgesia, whereas intracerebroventricular (i.c.v.) injection did not show the same effect. Moreover, NPFFR2 deficient (npffr2-/-) mice exhibited stronger analgesic responses to fentanyl compared to wild type (WT) or NPFFR1 knockout (npffr1-/-) mice. Intrathecal injection of RF9 in npffr1-/- mice also significantly enhanced fentanyl-induced analgesia. These results indicate a crucial role of spinal NPFFR2 in the enhancement of opioid analgesia. Contrastingly, hyperalgesia induced by fentanyl was markedly reversed in npffr1-/- mice but remained unaffected in npffr2-/- mice. Similarly, i.c.v. injection of the selective NPFFR1 antagonist RF3286 effectively prevented fentanyl-induced hyperalgesia in WT or npffr2-/- mice. Notably, co-administration of i.c.v. RF3286 and i.t. RF9 augmented fentanyl-induced analgesia while reducing hyperalgesia. Collectively, these findings highlight the modulating effects of blocking spinal NPFFR2 and supraspinal NPFFR1 on fentanyl-induced analgesia and hyperalgesia, respectively, which shed a light on understanding the pharmacological function of NPFF system in future studies.


Subject(s)
Analgesia , Hyperalgesia , Mice , Male , Animals , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Fentanyl/pharmacology , Analgesics, Opioid/pharmacology , Pain , Receptors, Neuropeptide/genetics
6.
Gene ; 907: 148283, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38354915

ABSTRACT

BACKGROUND: Isolated growth hormone deficiency (IGHD) is a rare genetically heterogeneous disorder caused primarily by mutations in GH1 and GH releasing hormone receptor (GHRHR). The aim of this study was to identify the molecular etiology of a Chinese boy with IGHD. METHODS: Whole-exome sequencing, sanger sequencing and bioinformatic analysis were performed to screen for candidate mutations. The impacts of candidate mutation on gene expression, intracellular localization and protein function were further evaluated by in vitro assays. RESULTS: A novel heterozygous frameshift mutation in the GHRH gene (c.91dupC, p.R31Pfs*98) was identified in a Chinese boy clinically diagnosed as having IGHD. The mutation was absent in multiple public databases, and considered as deleterious using in silico prediction, conservative analysis and three-dimensional homology modeling. Furthermore, mRNA and protein expression levels of mutant GHRH were significantly increased than wild-type GHRH (p < 0.05). Moreover, mutant GHRH showed an aberrant accumulation within the cytoplasm, and obviously reduced ability to stimulate GH secretion and cAMP accumulation in human GHRHR-expressing pituitary GH3 cells compared to wild-type GHRH (p < 0.05). CONCLUSION: Our study discovered the first loss-of function mutation of GHRH in a Chinese boy with IGHD and provided new insights on IGHD pathogenesis caused by GHRH haploinsufficiency.


Subject(s)
Dwarfism, Pituitary , Growth Hormone-Releasing Hormone , Human Growth Hormone , Humans , Male , China , Dwarfism, Pituitary/genetics , Frameshift Mutation , Growth Hormone , Human Growth Hormone/genetics , Mutation , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Growth Hormone-Releasing Hormone/genetics , East Asian People/genetics
7.
Skin Res Technol ; 30(2): e13588, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38284237

ABSTRACT

BACKGROUND: Prurigo nodularis (PN) is a chronic inflammatory skin disorder that is characterized by extremely itchy nodules. Proadrenomedullin N-terminal 20 (PAMP) activates mast cell degranulation via Mas-related G protein-coupled receptor X2 (MRGPRX2), which is associated with pruritus in allergic contact dermatitis. However, the mechanisms underlying the action of PAMP and MRGPRX2 in PN remain unclear. OBJECTIVE: To determine the role of PAMP-induced mast cell activation via MRGPRX2 (mouse homologous Mrgprb2) in PN. METHODS: The expression of PAMP and the number of MRGPRX2-expressing mast cells in the skin biopsies of patients with PN, atopic dermatitis (AD), and healthy participants were analyzed using immunohistochemistry and immunofluorescence, respectively. The biphasic response of PAMP9-20 mediated by Mrgprb2 in mouse peritoneal mast cells (PMC) was validated in vitro using qRT-PCR, ELISA, flow cytometry, and siRNA techniques. RESULTS: PAMP expression and the number of MRGPRX2+ mast cells in lesional PN skin, but not in AD, were elevated compared to healthy skin. PAMP9-20 mediates the immediate and delayed phase responses of PMC, such as degranulation, histamine and ß-hexosaminidase release, and secretion of inflammatory factors such as CCL2, TNF-α, and GM-CSF. These effects were inhibited when Mrgprb2 expression was silenced. Silencing Mrgprb2 did not affect the biphasic response of PMC that was induced by IgE-FcεRI activation. CONCLUSIONS: The results show that PAMP mediates mouse mast cell activation via Mrgprb2, which may be involved in the pathogenesis of PN. The PAMP/ Mrgprb2 pathway, independent of classical IgE signaling, could be developed as a candidate drug target for treating PN.


Subject(s)
Dermatitis, Atopic , Prurigo , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Adrenomedullin/metabolism , Dermatitis, Atopic/pathology , Immunoglobulin E/metabolism , Mast Cells/metabolism , Mast Cells/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prurigo/metabolism , Prurigo/pathology , Pruritus , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Skin/metabolism
8.
Steroids ; 202: 109349, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38072091

ABSTRACT

RFRP-3 is a functional ortholog of avian GnIH and regulates reproductive activities in the gonads of animals. However, the role of RFRP-3 in the function of ovarian granulosa cells in mice remains unclear. First, we detected the expression of the RFRP-3 receptor (GPR147) in the ovarian granulosa cells of mice. Second, the effect of RFRP-3 treatment on estradiol and progesterone secretions from granulosa cells was tested by ELISA. Meanwhile, the expression of genes and proteins regulating steroid hormone synthesis was respectively examined by qPCR and western blot. Furthermore, the effect of RFRP-3 treatment on the apoptosis of granulosa cells was analyzed. The results revealed that the GPR147 protein (a RFRP-3 receptor) was expressed in the ovarian granulosa cells of mice. Low and medium doses RFRP-3 treatment significantly reduced progesterone secretion in the granulosa cells (P < 0.05), while RFRP-3 suppressed p450scc, 3ß-HSD, StAR, and FSHR expression in a non-dose-dependent manner. Moreover, RFRP-3 treatment might induce the apoptosis of granulosa cells. Additionally, low doses RFRP-3 significantly reduced p-ERK1/2 protein expression (P < 0.05) in the ovarian granulosa cells. We here, for the first time, confirmed that GPR147 was expressed in the ovarian granulosa cells of mice. Our findings suggested that and RFRP-3 regulates the granulosa cell function through the ERK signaling pathway, which will lay the foundation for uncovering molecular mechanisms by which RFRP-3 regulates follicle development in future.


Subject(s)
Neuropeptides , Progesterone , Receptors, Neuropeptide , Female , Mice , Animals , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Progesterone/pharmacology , Granulosa Cells , Apoptosis
9.
Insect Sci ; 31(2): 417-434, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37464946

ABSTRACT

Mythimna separata is a notorious phytophagous pest which poses serious threats to cereal crops owing to the gluttony of the larvae. Because short neuropeptide F (sNPF) and its receptor sNPFR are involved in a diversity of physiological functions, especially in functions related to feeding in insects, it is a molecular target for pest control. Herein, an sNPF and 2 sNPFRs were identified and cloned from M. separata. Bioinformatics analysis revealed that the sNPF and its receptors had a highly conserved RLRFamide C-terminus and 7 transmembrane domains, respectively. The sNPF and its receptor genes were distributed across larval periods and tissues, but 2 receptors had distinct expression patterns. The starvation-induced assay elucidated that sNPF and sNPFR expression levels were downregulated under food deprivation and recovered with subsequent re-feeding. RNA interference knockdown of sNPF, sNPFR1, and sNPFR2 by injection of double-stranded RNA into larvae not only suppressed food consumption and increased body size and weight, but also led to decrease of glycogen and total lipid contents, and increase of trehalose compared with double-stranded green fluorescent protein injection. Furthermore, molecular docking was performed on the interaction mode between sNPFR protein and its ligand sNPF based on the 3-dimensional models constructed by AlphaFold; the results indicated that both receptors were presumably activated by the mature peptide sNPF-2. These results revealed that sNPF signaling played a considerably vital role in the feeding regulation of M. separata and represents a potential control target for this pest.


Subject(s)
Neuropeptides , Receptors, Neuropeptide , Animals , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Larva/genetics , Larva/metabolism , Molecular Docking Simulation , Neuropeptides/genetics , Neuropeptides/metabolism
10.
Front Immunol ; 14: 1197821, 2023.
Article in English | MEDLINE | ID: mdl-38022672

ABSTRACT

Background: Mycosis fungoides (MF) is an indolent T-cell lymphoma that mainly affects the skin and presents with itch in more than half of the patients. Recently, the expression of Mas-related G protein-coupled receptor X2 (MRGPRX2), a receptor of mast cell (MC) responsible for the IgE-independent non-histaminergic itch, has been shown in lesional skin of patients with pruritic skin diseases, including chronic urticaria, prurigo, and mastocytosis. As of yet, limited knowledge exists regarding the MRGPRX2 expression in the skin of patients with MF. Objectives: To investigate the number of MRGPRX2-expressing (MRGPRX2+) cells in the skin of patients with MF and its correlation with clinical and laboratory characteristics of the disease. Methods: MRGPRX2 was analyzed in lesional and non-lesional skin of MF patients and healthy skin tissues by immunohistochemistry. Co-localization of MRGPRX2 with the MC marker tryptase was assessed by immunofluorescence. Public single-cell RNAseq data was reanalyzed to identify the MRGPRX2 expression on the distinct cell types. Results: In lesional skin of MF patients, MRGPRX2+ cell number was higher than in non-lesional skin and healthy control skin (mean:15.12 vs. 6.84 vs. 5.51 cells/mm2, p=0.04), and correlated with MC numbers (r=0.73, p=0.02). MC was the primary cell type expressing MRGPRX2 in MF patients. The ratio of MRGPRX2+ MCs to MRGPRX2+ cells in lesional and non-lesional skin correlated with the severity of disease (r=0.71, p=0.02 and r=0.67, p=0.03, respectively). Conclusions: Our findings point to the role of MRGPRX2 and MC in the pathogenesis of MF that should be investigated in further studies.


Subject(s)
Mycosis Fungoides , Skin Neoplasms , Humans , Mycosis Fungoides/pathology , Skin/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Skin Neoplasms/pathology , Pruritus/metabolism , Cell Count , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
11.
Vitam Horm ; 123: 1-26, 2023.
Article in English | MEDLINE | ID: mdl-37717982

ABSTRACT

The hypothalamic peptide growth hormone-releasing hormone (GHRH) stimulates the secretion of growth hormone (GH) from the pituitary through binding and activation of the pituitary type of GHRH receptor (GHRH-R), which belongs to the family of G protein-coupled receptors with seven potential membrane-spanning domains. Splice variants of GHRH-Rs (SV) in human tumors and other extra pituitary tissues were identified and their cDNA was sequenced. Among the SVs, splice variant 1 (SV1) possesses the greatest similarity to the full-length GHRH-R and remains functional by eliciting cAMP signaling and mitogenic activity upon GHRH stimulation. A large body of work have evaluated potential clinical applications of agonists and antagonists of GHRH in diverse fields, including endocrinology, oncology, cardiology, diabetes, obesity, metabolic dysfunctions, Alzheimer's disease, ophthalmology, wound healing and other applications. In this chapter, we briefly review the expression and potential function of GHRH-Rs and their SVs in various tissues and also elucidate and summarize the activation, molecular mechanism and signalization pathways of these receptors. Therapeutic applications of GHRH analogs are also discussed.


Subject(s)
Receptors, Neuropeptide , Signal Transduction , Humans , Growth Hormone , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics
12.
Fish Physiol Biochem ; 49(5): 983-1003, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37670169

ABSTRACT

The neuropeptide B/W signaling system is composed of neuropeptide B (NPB), neuropeptide W (NPW), and two cognate receptors, NPBWR1 and NPBWR2, which are involved in diverse physiological processes, including the central regulation of neuroendocrine axes in vertebrates. The components of this signaling system are not well conserved during vertebrate evolution, implicating its functional diversity. The present study characterized the ricefield eel neuropeptide B/W system, generated a specific antiserum against the neuropeptide B/W receptor, and examined the potential roles of the system in the regulation of adenohypophysial functions. The ricefield eel genome contains npba, npbb, and npbwr2b but lacks the npw, npbwr1, and npbwr2a genes. The loss of npw and npbwr1 probably occurred at the base of ray-finned fish radiation and that of npbwr2a species specifically in ray-finned fish. Npba and npbb genes are produced through whole-genome duplication (WGD) in ray-finned fish. The ricefield eel npba was expressed in the brain and some peripheral tissues, while npbb was predominantly expressed in the brain. The ricefield eel npbwr2b was also expressed in the brain and in some peripheral tissues, such as the pituitary, gonad, heart, and eye. Immunoreactive Npbwr2b was shown to be localized to Lh and Fsh cells but not to Gh or Prl cells in the pituitary of ricefield eels. Npba upregulated the expression of fshb and cga but not lhb mRNA in pituitary fragments of ricefield eels cultured in vitro. The results of the present study suggest that the NPB system of ricefield eels may be involved in the neuroendocrine regulation of reproduction.


Subject(s)
Eels , Neuropeptides , Animals , Eels/genetics , Eels/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Gonadotropins/metabolism , Receptors, Neuropeptide/genetics
13.
Science ; 381(6663): 1189-1196, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37708282

ABSTRACT

Eosinophils are granulocytes that play an essential role in type 2 immunity and regulate multiple homeostatic processes in the small intestine (SI). However, the signals that regulate eosinophil activity in the SI at steady state remain poorly understood. Through transcriptome profiling of eosinophils from various mouse tissues, we found that a subset of SI eosinophils expressed neuromedin U (NMU) receptor 1 (NMUR1). Fate-mapping analyses showed that NMUR1 expression in SI eosinophils was programmed by the local microenvironment and further enhanced by inflammation. Genetic perturbation and eosinophil-organoid coculture experiments revealed that NMU-mediated eosinophil activation promotes goblet cell differentiation. Thus, NMU regulates epithelial cell differentiation and barrier immunity by stimulating NMUR1-expressing eosinophils in the SI, which highlights the importance of neuroimmune-epithelial cross-talk in maintaining tissue homeostasis.


Subject(s)
Eosinophils , Immunity, Mucosal , Intestine, Small , Neuropeptides , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Animals , Mice , Eosinophils/immunology , Intestine, Small/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Coculture Techniques , Organoids
14.
Cells ; 12(10)2023 05 17.
Article in English | MEDLINE | ID: mdl-37408245

ABSTRACT

Insect sex pheromones are volatile chemicals that induce mating behavior between conspecific individuals. In moths, sex pheromone biosynthesis is initiated when pheromone biosynthesis-activating neuropeptide (PBAN) synthesized in the suboesophageal ganglion binds to its receptor on the epithelial cell membrane of the pheromone gland. To investigate the function of PBAN receptor (PBANR), we identified two PBANR isoforms, MviPBANR-B and MviPBANR-C, in the pheromone glands of Maruca vitrata. These two genes belong to G protein-coupled receptors (GPCRs) and have differences in the C-terminus but share a 7-transmembrane region and GPCR family 1 signature. These isoforms were expressed in all developmental stages and adult tissues. MviPBANR-C had the highest expression level in pheromone glands among the examined tissues. Through in vitro heterologous expression in HeLa cell lines, only MviPBANR-C-transfected cells responded to MviPBAN (≥5 µM MviPBAN), inducing Ca2+ influx. Sex pheromone production and mating behavior were investigated using gas chromatography and a bioassay after MviPBANR-C suppression by RNA interference, which resulted in the major sex pheromone component, E10E12-16:Ald, being quantitatively reduced compared to the control, thereby decreasing the mating rate. Our findings indicate that MviPBANR-C is involved in the signal transduction of sex pheromone biosynthesis in M. vitrata and that the C-terminal tail plays an important role in its function.


Subject(s)
Moths , Sex Attractants , Humans , Animals , Sex Attractants/metabolism , HeLa Cells , Amino Acid Sequence , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Moths/genetics , Protein Isoforms/metabolism
15.
Sci Rep ; 13(1): 12302, 2023 07 29.
Article in English | MEDLINE | ID: mdl-37516794

ABSTRACT

Migraine ranks among the most prevalent disorders worldwide, leading to disability and decreased quality of life in patients. Recently, neurogenic inflammation has been recognized as a potential underlying pathology contributing to the migraine pain pathway. Mast cells reside in the meninges and have been implicated in contributing to the pathophysiology of migraine. Here we report for the first time that the mouse Mas-Related G-protein-coupled Receptor B2 (MrgprB2), is expressed on meningeal connective tissue mast cells and contributes to Pituitary Adenylate Cyclase Activating Peptide (PACAP)-induced migraine-like pain behavior. We also found that PACAP was able to dose-dependently lead to enzyme release from human mast cells via activation of MRGPRX2; the human homolog of MrgprB2. Using a transgenic MRGPRX2 mouse, we observed significant increases in PACAP-induced migraine-like pain behavior in MRGPRX2+ mice vs mice lacking the receptor. These results reveal both MrgprB2 and MRGPRX2 as important contributors to neuropeptide-induced migraine pain.


Subject(s)
Migraine Disorders , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Humans , Mice , Mast Cells , Meninges , Mice, Transgenic , Migraine Disorders/chemically induced , Nerve Tissue Proteins/genetics , Pain , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Quality of Life , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics
16.
Toxicol Appl Pharmacol ; 474: 116601, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37321326

ABSTRACT

Two potent and selective KRASG12D inhibitors, ERAS-4693 and ERAS-5024, were generated as possible clinical candidates to treat patients harboring G12D mutations in solid tumors. Both molecules exhibited strong anti-tumor activity in the KRASG12D mutant PDAC xenograft mouse models while ERAS-5024 also showed tumor growth inhibition when administered on an intermittent dosing regimen. Acute dose-limiting toxicity consistent with an allergic reaction was observed for both molecules shortly after administration at doses just above those which demonstrated anti-tumor activity, indicative of a narrow therapeutic index. A series of studies were subsequently conducted to identify a common underlying mechanism for the observed toxicity, including CETSA® (Cellular Thermal Shift Assay) as well as several functional off-target screens. Both ERAS-4693 and ERAS-5024 were identified to agonize MRGPRX2 which has been linked to pseudo-allergic reactions. In vivo toxicologic characterization of both molecules included repeat-dose studies in the rat and dog. Dose-limiting toxicities were observed in both species with ERAS-4693 and ERAS-5024 and plasma exposure levels at the maximum tolerated doses were generally below that which caused strong anti-tumor activity, supporting the initial observation of a narrow therapeutic index. Additional overlapping toxicities included a reduction in reticulocytes and clinical pathological changes suggestive of an inflammatory response. Furthermore, increases in plasma histamine were observed in dogs administered ERAS-5024, supporting the hypothesis that MRGPRX2 agonism may be the cause of the pseudo-allergic reaction. This work highlights the importance of balancing both the safety and efficacy of KRASG12D inhibitors as this class of molecules begins to enter clinical development.


Subject(s)
Hypersensitivity , Pancreatic Neoplasms , Humans , Mice , Rats , Animals , Dogs , Proto-Oncogene Proteins p21(ras)/genetics , Pancreatic Neoplasms/pathology , Mutation , Nerve Tissue Proteins , Receptors, Neuropeptide/genetics , Receptors, G-Protein-Coupled/genetics
17.
Int J Biol Macromol ; 244: 125411, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37327925

ABSTRACT

Neuropeptides and neuropeptide receptors are crucial regulators for the behavior, lifecycle, and physiology of insects and are mainly produced and released from the neurosecretory cells of the central nervous system (CNS). In this study, RNA-seq was employed to investigate the transcriptome profile of the CNS which is composed of the brain and ventral nerve cord (VNC) of Antheraea pernyi. From the data sets, a total of 18 and 42 genes were identified, which respectively encode the neuropeptides and neuropeptide receptors involved in regulating multiple behaviors including feeding, reproductive behavior, circadian locomotor, sleep, and stress response and physiological processes such as nutrient absorption, immunity, ecdysis, diapause, and excretion. Comparison of the patterns of expression of those genes between the brain and VNC showed that most had higher levels of expression in the brain than VNC. Besides, 2760 differently expressed genes (DEGs) (1362 up-regulated and 1398 down-regulated ones between the B and VNC group) were also screened and further analyzed via gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses. The results of this study could provide comprehensive profiles of the neuropeptides and neuropeptide receptors of A. pernyi CNS and lay the foundation for further research into their functions.


Subject(s)
Moths , Neuropeptides , Animals , Transcriptome/genetics , Gene Expression Profiling/methods , Moths/genetics , Neuropeptides/genetics , Receptors, Neuropeptide/genetics , Central Nervous System
18.
J Insect Physiol ; 147: 104524, 2023 06.
Article in English | MEDLINE | ID: mdl-37201579

ABSTRACT

In Europe, the tick Ixodes ricinus is the most important vector of numerous pathogens that are transmitted during blood feeding on their vertebrate hosts. To elucidate mechanisms controlling blood intake and associated transmission of pathogens we identified and described expression of short neuropeptide F (sNPF) and its receptors which are known to regulate feeding in insects. Using in situ hybridization (ISH) and immunohistochemistry (IHC) we stained numerous neurons producing sNPF in the central nervous system (CNS; synganglion), while a few peripheral neurons were detected anteriorly to the synganglion, and on the surface of the hindgut and leg muscles. Apparent sNPF expression was also found in enteroendocrine cells individually scattered in anterior lobes of the midgut. In silico analyses and BLAST search for sNPF receptors revealed two putative G protein-coupled receptors (sNPFR1 and sNPFR2) in the I. ricinus genome. Aequorin-based functional assay in CHO cells showed that both receptors were specific and sensitive to sNPF in nanomolar concentrations. Increased expression levels of these receptors in the gut during blood intake suggest that sNPF signaling may be involved in regulation of feeding and digestion processes of I. ricinus.


Subject(s)
Ixodes , Neuropeptides , Animals , Cricetinae , Ixodes/genetics , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Cricetulus , Neuropeptides/genetics , Neuropeptides/metabolism
19.
Biochem Biophys Res Commun ; 660: 28-34, 2023 06 11.
Article in English | MEDLINE | ID: mdl-37060828

ABSTRACT

G protein-coupled receptors (GPCRs) are a major class of membrane receptors that modulate a wide range of physiological functions. These receptors transmit extracellular signals, including secreted bioactive peptides, to intracellular signaling pathways. The nematode Caenorhabditis elegans has FMRFamide-like peptides, which are one of the most diverse neuropeptide families, some of which modulate larval development through GPCRs. In this study, we identified the GPCR neuropeptide receptor (NPR)-15, which modulates C. elegans larval development. Our molecular genetic analyses indicated the following: 1) NPR-15 mainly functions in ASI neurons, which predominantly regulate larval development, 2) NPR-15 interacts with GPA-4, a C. elegans Gα subunit, and 3) NPR-15, along with GPA-4, modulates larval development by regulating the production and secretion of the transforming growth factor-ß (TGF-ß)-like protein DAF-7. The present study is the first report to demonstrate the importance of a GPCR to the direct regulation of a TGF-ß-like protein.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Peptides/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...