1
|
Schöneberg T. Modulating vertebrate physiology by genomic fine-tuning of GPCR functions. Physiol Rev 2025; 105:383-439. [PMID: 39052017 DOI: 10.1152/physrev.00017.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role as membrane receptors, facilitating the communication of eukaryotic species with their environment and regulating cellular and organ interactions. Consequently, GPCRs hold immense potential in contributing to adaptation to ecological niches and responding to environmental shifts. Comparative analyses of vertebrate genomes reveal patterns of GPCR gene loss, expansion, and signatures of selection. Integrating these genomic data with insights from functional analyses of gene variants enables the interpretation of genotype-phenotype correlations. This review underscores the involvement of GPCRs in adaptive processes, presenting numerous examples of how alterations in GPCR functionality influence vertebrate physiology or, conversely, how environmental changes impact GPCR functions. The findings demonstrate that modifications in GPCR function contribute to adapting to aquatic, arid, and nocturnal habitats, influencing camouflage strategies, and specializing in particular dietary preferences. Furthermore, the adaptability of GPCR functions provides an effective mechanism in facilitating past, recent, or ongoing adaptations in animal domestication and human evolution and should be considered in therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| |
Collapse
|
2
|
Chen YM, Huang J, Fan H, Li WY, Shi TS, Zhao J, Wang CN, Chen WJ, Zhu BL, Qian JJ, Guan W, Jiang B. QRFP and GPR103 in the paraventricular nucleus play a role in chronic stress-induced depressive-like symptomatology by enhancing the hypothalamic-pituitary-adrenal axis. Neuropharmacology 2024; 262:110198. [PMID: 39442911 DOI: 10.1016/j.neuropharm.2024.110198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/04/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis during chronic stress is essential for depression neurobiology. As the latest member of the RFamide peptide family in mammals, pyroglutamylated RFamide peptide (QRFP) is closely implicated in neuroendocrine maintenance by activating G-protein-coupled receptor 103 (GPR103). We hypothesized that QRFP and GPR103 might contribute to chronic stress-induced depression by promoting corticotropin-releasing hormone (CRH) release from neurons in the paraventricular nucleus (PVN), and various methods were employed in this study, with male C57BL/6J mice adopted as the experimental subjects. Chronic stress induced not only depression-like behaviors but also significant enhancement in QRFP and GPR103 in the PVN. Genetic overexpression of QRFP/GPR103 and stereotactic infusion of QRFP-26/QRFP-43 peptide in the PVN all mimicked chronic stress that induced various depression-like phenotypes in naïve mice, and this was mediated by promoting CRH biosynthesis and HPA activity. In contrast, genetic knockdown of QRFP/GPR103 in the PVN produced notable antidepressant-like effects in mice exposed to chronic stress. Furthermore, genetic knockout of QRFP also protected against chronic stress in mice. In addition, both the C-terminal biological region of QRFP and the downstream PKA/PKC-CREB signaling coupled to GPR103 stimulation underlie the role of QRFP and GPR103 in depression. Collectively, QRFP and GPR103 in PVN neurons could be viable targets for novel antidepressants.
Collapse
Affiliation(s)
- Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, Henan, China
| | - Wei-Yu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Tian-Shun Shi
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jie Zhao
- Department of Pharmacy, The Sixth People's Hospital of Nantong, Nantong, 226011 Jiangsu, China
| | - Cheng-Niu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Wei-Jia Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Bao-Lun Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jun-Jie Qian
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
3
|
Iwama A, Kise R, Akasaka H, Sano FK, Oshima HS, Inoue A, Shihoya W, Nureki O. Structure and dynamics of the pyroglutamylated RF-amide peptide QRFP receptor GPR103. Nat Commun 2024; 15:4769. [PMID: 38897996 PMCID: PMC11187126 DOI: 10.1038/s41467-024-49030-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Pyroglutamylated RF-amide peptide (QRFP) is a peptide hormone with a C-terminal RF-amide motif. QRFP selectively activates a class A G-protein-coupled receptor (GPCR) GPR103 to exert various physiological functions such as energy metabolism and appetite regulation. Here, we report the cryo-electron microscopy structure of the QRFP26-GPR103-Gq complex at 3.19 Å resolution. QRFP26 adopts an extended structure bearing no secondary structure, with its N-terminal and C-terminal sides recognized by extracellular and transmembrane domains of GPR103 respectively. This movement, reminiscent of class B1 GPCRs except for orientation and structure of the ligand, is critical for the high-affinity binding and receptor specificity of QRFP26. Mutagenesis experiments validate the functional importance of the binding mode of QRFP26 by GPR103. Structural comparisons with closely related receptors, including RY-amide peptide-recognizing GPCRs, revealed conserved and diversified peptide recognition mechanisms, providing profound insights into the biological significance of RF-amide peptides. Collectively, this study not only advances our understanding of GPCR-ligand interactions, but also paves the way for the development of novel therapeutics targeting metabolic and appetite disorders and emergency medical care.
Collapse
Affiliation(s)
- Aika Iwama
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Hiroaki Akasaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Fumiya K Sano
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Hidetaka S Oshima
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Wataru Shihoya
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
4
|
Devère M, Takhlidjt S, Prévost G, Chartrel N, Leprince J, Picot M. The 26RFa (QRFP)/GPR103 Neuropeptidergic System: A Key Regulator of Energy and Glucose Metabolism. Neuroendocrinology 2024:1-17. [PMID: 38599200 DOI: 10.1159/000538629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Obesity and type 2 diabetes are strongly associated pathologies, currently considered as a worldwide epidemic problem. Understanding the mechanisms that drive the development of these diseases would enable to develop new therapeutic strategies for their prevention and treatment. Particularly, the role of the brain in energy and glucose homeostasis has been studied for 2 decades. In specific, the hypothalamus contains well-identified neural networks that regulate appetite and potentially also glucose homeostasis. A new concept has thus emerged, suggesting that obesity and diabetes could be due to a dysfunction of the same, still poorly understood, neural networks. SUMMARY The neuropeptide 26RFa (also termed QRFP) belongs to the family of RFamide regulatory peptides and has been identified as the endogenous ligand of the human G protein-coupled receptor GPR103 (QRFPR). The primary structure of 26RFa is strongly conserved during vertebrate evolution, suggesting its crucial roles in the control of vital functions. Indeed, the 26RFa/GPR103 peptidergic system is reported to be involved in the control of various neuroendocrine functions, notably the control of energy metabolism in which it plays an important role, both centrally and peripherally, since 26RFa regulates feeding behavior, thermogenesis and lipogenesis. Moreover, 26RFa is reported to control glucose homeostasis both peripherally, where it acts as an incretin, and centrally, where the 26RFa/GPR103 system relays insulin signaling in the brain to control glucose metabolism. KEY MESSAGES This review gives a comprehensive overview of the role of the 26RFa/GPR103 system as a key player in the control of energy and glucose metabolism. In a pathophysiological context, this neuropeptidergic system represents a prime therapeutic target whose mechanisms are highly relevant to decipher.
Collapse
Affiliation(s)
- Mélodie Devère
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| | - Saloua Takhlidjt
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| | - Gaëtan Prévost
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Rouen Normandie, Inserm, Normandie University, NorDiC UMR 1239, CHU Rouen, Rouen, France
| | - Nicolas Chartrel
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| | - Jérôme Leprince
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
- University Rouen Normandie, Normandie University, INSERM US 51, CNRS UAR 2026, HeRacLeS, Rouen, France
| | - Marie Picot
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| |
Collapse
|
5
|
Martin CT, Primeaux SD. The hypothalamic neuropeptide, QRFP, regulates high fat diet intake in female Long-Evans rats following ovariectomy. Peptides 2023; 162:170960. [PMID: 36690209 PMCID: PMC9992330 DOI: 10.1016/j.peptides.2023.170960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/12/2022] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Obesity rates in women continue to increase throughout the lifespan and obesity-related comorbidities are prevalent in women in estrogen deficiency. The hypothalamic neuropeptide, QRFP, is an orexigenic peptide that increases the intake of high fat diet (HFD) in female rats and is overexpressed following ovariectomy (OVX). Therefore, the goal of the current series of experiments was to elucidate the effect of QRFP on HFD intake following OVX and determine if QRFP-26 administration in ovariectomized females altered expression of prepro-neuropeptide Y (NPY), agouti-related peptide (AgRP) and proopiomelanocortin (POMC) mRNA in the mediobasal hypothalamus (MBH) and prepro-orexin in the lateral hypothalamus (LH). The intake of HFD was measured following acute administration of QRFP-26 prior to or following estradiol benzoate (EB) treatment in ovariectomized females. When administered prior to EB treatment, QRFP-26 increased HFD intake. EB treatment attenuated the effects of QRFP-26 on HFD intake. Sub-chronic, continuous administration of QRFP-26 increased HFD intake and weight gain following OVX. Subchronic, continuous administration of QRFP siRNA into the 3rd ventricle via osmotic pump decreased prepro-QRFP mRNA levels in the MBH by ∼75%, decreased HFD intake and decreased weight gain following OVX. QRFP-26administration did not alter the expression of prepro-NPY, AgRP or POMC mRNA in the MBH, but decreased prepro-orexin mRNA in the LH of ovariectomized females. Overall, results from these studies support the orexigenic neuropeptide, QRFP, as an important mediator of the ingestion of highly palatable foods and subsequent weight gain in females during estrogen deficiency.
Collapse
Affiliation(s)
- Cade T Martin
- Department of Physiology, LSU Health Sciences Center-NO, New Orleans, LA 70112, USA
| | - Stefany D Primeaux
- Department of Physiology, LSU Health Sciences Center-NO, New Orleans, LA 70112, USA; Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
6
|
Perucatti A, Iannuzzi A, Armezzani A, Palmarini M, Iannuzzi L. Comparative Fluorescence In Situ Hybridization (FISH) Mapping of Twenty-Three Endogenous Jaagsiekte Sheep Retrovirus (enJSRVs) in Sheep ( Ovis aries) and River Buffalo ( Bubalus bubalis) Chromosomes. Animals (Basel) 2022; 12:ani12202834. [PMID: 36290220 PMCID: PMC9597706 DOI: 10.3390/ani12202834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Endogenous retroviruses (ERVs) are the remnants of ancient infections of host germline cells, thus representing key tools to study host and viral evolution. Homologous ERV sequences often map at the same genomic locus of different species, indicating that retroviral integration occurred in the genomes of the common ancestors of those species. The genome of domestic sheep (Ovis aries) harbors at least twenty-seven copies of ERVs related to the exogenous and pathogenic Jaagsiekte sheep retrovirus (JSRVs), thus referred to as enJSRVs. Some of these loci are unequally distributed between breeds and individuals of the host species due to polymorphic insertions, thereby representing invaluable tools to trace the evolutionary dynamics of virus populations within their hosts. In this study, we extend the cytogenetic physical maps of sheep and river buffalo by performing fluorescent in situ hybridization (FISH) mapping of twenty-three genetically characterized enJSRVs. Additionally, we report the first comparative FISH mapping of enJSRVs in domestic sheep (2n = 54) and river buffalo (Bubalus bubalis, 2n = 50). Finally, we demonstrate that enJSRV loci are conserved in the homologous chromosomes and chromosome bands of both species. Altogether, our results support the hypothesis that enJSRVs were present in the genomes of both species before they differentiated within the Bovidae family.
Collapse
Affiliation(s)
- Angela Perucatti
- National Research Council (CNR), Institute of Animal Production System on Mediterranean Environment (ISPAAM), Piazzale E. Fermi, 1, 80055 Portici, Italy
| | - Alessandra Iannuzzi
- National Research Council (CNR), Institute of Animal Production System on Mediterranean Environment (ISPAAM), Piazzale E. Fermi, 1, 80055 Portici, Italy
- Correspondence: ; Tel.: +39-32-8961-7073
| | - Alessia Armezzani
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61-1QH, UK
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61-1QH, UK
| | - Leopoldo Iannuzzi
- National Research Council (CNR), Institute of Animal Production System on Mediterranean Environment (ISPAAM), Piazzale E. Fermi, 1, 80055 Portici, Italy
| |
Collapse
|
7
|
Li H, Lou R, Xu X, Xu C, Yu Y, Xu Y, Hu L, Xiang Y, Lin X, Tang S. The variations in human orphan G protein-coupled receptor QRFPR affect PI3K-AKT-mTOR signaling. J Clin Lab Anal 2021; 35:e23822. [PMID: 34018631 DOI: 10.1002/jcla.23822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND QRFPR is a recently identified member of the G protein-coupled receptor and is an orphan receptor for 26Rfa, which plays important role in the regulation of many physiological functions. METHODS Here, we employed whole exome sequencing (WES) to examine the patients with intellectual disability (ID) and difficulty in feeding. We performed SIFT and PolyPhen2 predictions for the variants. The structure model was built from scratch by I-TASSER. Here, results derived from a number of cell-based functional assays, including shRNA experiment, intracellular Ca2+ measurement, the expression of PI3 K-AKT-mTOR, and phosphorylation. The functional effect of QRFPR variants on PI3K-AKT-mTOR signaling was evaluated in vitro transfection experiments. RESULT Here, we identified two QRFPR variants at c.202 T>C (p.Y68H) and c.1111C>T (p.R371W) in 2 unrelated individuals. Structural analysis revealed that p.Y68H and p.R371W variants may affect the side chain structure of adjacent amino acids causing reduced binding of QRFPR to 26Rfa. The results show that QRFPR stimulated by 26Rfa leading to the transient rise of intracellular Ca2+ . The QRFPR variations p.Y68H and p.R371 W can reduce the mobilization of intracellular Ca2+ . The phosphorylation levels of the PI3K, Akt, and mTOR were significantly up- or downregulated by QRFPR overexpression or silencing, respectively. The QRFPR variations inhibited PI3K-AKT-mTOR signaling, resulting in downregulation of p-mTOR. CONCLUSIONS Our findings suggest that QRFPR acts as important role in neurodevelopment, and the effects of QRFPR are likely to be mediated by the Ca2+ -dependent PI3K-AKT-mTOR pathways. Importantly, these findings provide a foundation for future elucidation of GPCR-mediated signaling and the physiological implications.
Collapse
Affiliation(s)
- Huanzheng Li
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China.,Human Aging Research Institute, Nanchang University, Nanchang, China
| | - Ran Lou
- Department of Acupuncture, Wenzhou Central Hospital, Wenzhou, China
| | - Xueqin Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chenyang Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yuan Yu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yunzhi Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Lin Hu
- Department of Blood Transfusion, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanbao Xiang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuan Lin
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Shaohua Tang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Ma Q, Cao Z, Li H, Wang W, Tian Y, Yan L, Liao Y, Chen X, Chen Y, Shi Y, Tang S, Zhou N. Two naturally occurring mutations of human GPR103 define distinct G protein selection bias. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119046. [PMID: 33872671 DOI: 10.1016/j.bbamcr.2021.119046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/18/2022]
Abstract
The neuropeptide 26RFa plays important roles in the regulation of many physiological functions. 26RFa has been recognized as an endogenous ligand for receptor GPR103. In the present study, we demonstrate that GPR103 dually couples to Gαq and Gαi/o proteins. However, two naturally occurring missense mutations were identified from a young male patient. In the first, Y68H, induction of Ca2+ mobilization was noted without detection of ERK1/2 activation. In the second, R371W, the potential to activate ERK1/2 signaling was retained but with failure to evoke Ca2+ mobilization. Further analysis provides evidence that Gαq, L-type Ca2+ channel and PKCβI and βII are involved in the Y68H-mediated signaling pathway, whereas Gαi/o, Gβγ, and PKCζ are implicated in the R371W-induced signaling. Our results demonstrate that two point mutations, Y68H and R371W, affect the equilibrium between the different receptor conformations, leading to alteration of G protein-coupling preferences. Importantly, these findings provide a foundation for future elucidation of GPCR-mediated biased signaling and the physiological implications of their bias.
Collapse
Affiliation(s)
- Qiang Ma
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, MOE Frontier Center of Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zheng Cao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huanzheng Li
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China
| | - Weiwei Wang
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yanan Tian
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lili Yan
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yuan Liao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiangnan Chen
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China
| | - Yu Chen
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Shi
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shaohua Tang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 32500, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
9
|
Wang W, Tian Y, Shi X, Ma Q, Xu Y, Yang G, Yi W, Shi Y, Zhou N. N-glycosylation of the human neuropeptide QRFP receptor (QRFPR) is essential for ligand binding and receptor activation. J Neurochem 2021; 158:138-152. [PMID: 33655503 DOI: 10.1111/jnc.15337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 11/29/2022]
Abstract
The newly identified pyroglutamylated RFamide peptide (QRFP) signaling system has been shown to be implicated in regulating a variety of physiological processes. G-protein-coupled receptors (GPCRs) are preferentially N-glycosylated on extracellular domains. The human QRFP receptor QRFPR (GPR103) possesses three N-glycosylation consensus sites, two located on the N-terminal domain (N5 and N19) and one on the first extracellular loop (ECL1) (N106); however, to date, their role in QRFPR expression and signaling has not been established. Here, we combined mutants with glutamine substitution of the critical asparagines of the consensus sites with glycosidase PNGase F and N-glycosylation inhibitor tunicamycin to study the effect of N-glycosylation in the regulation of QRFPR cell surface expression and signaling. Western blot analysis performed with site-directed mutagenesis revealed that two asparagines at N19 in the N-terminus and N106 in ECL1, but not N5 in the N-terminus, served as sites for N-glycosylation. Treatment with PNGase F and tunicamycin resulted in a reduction in both two-protein species, ~43 kDa and ~85 kDa in size, by 2-4 kDa. Analysis with confocal microscopy and quantitative ELISA showed that N-glycosylation of QRFPR is not essentially required for targeting the cell membrane. However, further binding assay and functional assays demonstrated that removal of N-glycosylation sequons or treatment with tunicamycin led to significant impairments in the interaction of receptor with QRFP26 and downstream signaling. Thus, our findings suggest that for the human QRFP receptor (QRFPR), N-glycosylation is not important for cell surface expression but is a pre-requisite for ligand binding and receptor activation.
Collapse
Affiliation(s)
- Weiwei Wang
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanan Tian
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoliu Shi
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiang Ma
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yue Xu
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Gangjie Yang
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wen Yi
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying Shi
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Szenajch J, Szabelska-Beręsewicz A, Świercz A, Zyprych-Walczak J, Siatkowski I, Góralski M, Synowiec A, Handschuh L. Transcriptome Remodeling in Gradual Development of Inverse Resistance between Paclitaxel and Cisplatin in Ovarian Cancer Cells. Int J Mol Sci 2020; 21:E9218. [PMID: 33287223 PMCID: PMC7730278 DOI: 10.3390/ijms21239218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Resistance to anti-cancer drugs is the main challenge in oncology. In pre-clinical studies, established cancer cell lines are primary tools in deciphering molecular mechanisms of this phenomenon. In this study, we proposed a new, transcriptome-focused approach, utilizing a model of isogenic cancer cell lines with gradually changing resistance. We analyzed trends in gene expression in the aim to find out a scaffold of resistance development process. The ovarian cancer cell line A2780 was treated with stepwise increased concentrations of paclitaxel (PTX) to generate a series of drug resistant sublines. To monitor transcriptome changes we submitted them to mRNA-sequencing, followed by the identification of differentially expressed genes (DEGs), principal component analysis (PCA), and hierarchical clustering. Functional interactions of proteins, encoded by DEGs, were analyzed by building protein-protein interaction (PPI) networks. We obtained human ovarian cancer cell lines with gradually developed resistance to PTX and collateral sensitivity to cisplatin (CDDP) (inverse resistance). In their transcriptomes, we identified two groups of DEGs: (1) With fluctuations in expression in the course of resistance acquiring; and (2) with a consistently changed expression at each stage of resistance development, constituting a scaffold of the process. In the scaffold PPI network, the cell cycle regulator-polo-like kinase 2 (PLK2); proteins belonging to the tumor necrosis factor (TNF) ligand and receptor family, as well as to the ephrin receptor family were found, and moreover, proteins linked to osteo- and chondrogenesis and the nervous system development. Our cellular model of drug resistance allowed for keeping track of trends in gene expression and studying this phenomenon as a process of evolution, reflected by global transcriptome remodeling. This approach enabled us to explore novel candidate genes and surmise that abrogation of the osteomimic phenotype in ovarian cancer cells might occur during the development of inverse resistance between PTX and CDDP.
Collapse
Affiliation(s)
- Jolanta Szenajch
- Laboratory for Molecular Oncology and Innovative Therapies, Military Institute of Medicine, 04-141 Warsaw, Poland;
| | - Alicja Szabelska-Beręsewicz
- Department of Mathematical and Statistical Methods, Poznań University of Life Sciences, 60-637 Poznań, Poland; (A.S.-B.); (J.Z.-W.); (I.S.)
| | - Aleksandra Świercz
- Laboratory of Genomics, Institute of Bioorganic Chemistry, Polish Academy of Science, 61-704 Poznań, Poland; (A.Ś.); (M.G.); (L.H.)
- Institute of Computing Science, Poznan University of Technology, 60-965 Poznań, Poland
| | - Joanna Zyprych-Walczak
- Department of Mathematical and Statistical Methods, Poznań University of Life Sciences, 60-637 Poznań, Poland; (A.S.-B.); (J.Z.-W.); (I.S.)
| | - Idzi Siatkowski
- Department of Mathematical and Statistical Methods, Poznań University of Life Sciences, 60-637 Poznań, Poland; (A.S.-B.); (J.Z.-W.); (I.S.)
| | - Michał Góralski
- Laboratory of Genomics, Institute of Bioorganic Chemistry, Polish Academy of Science, 61-704 Poznań, Poland; (A.Ś.); (M.G.); (L.H.)
| | - Agnieszka Synowiec
- Laboratory for Molecular Oncology and Innovative Therapies, Military Institute of Medicine, 04-141 Warsaw, Poland;
| | - Luiza Handschuh
- Laboratory of Genomics, Institute of Bioorganic Chemistry, Polish Academy of Science, 61-704 Poznań, Poland; (A.Ś.); (M.G.); (L.H.)
| |
Collapse
|
11
|
Wang W, Jiang C, Xu Y, Ma Q, Yang J, Shi Y, Zhou N. Functional characterization of neuropeptide 26RFa receptors GPR103A and GPR103B in zebrafish, Danio rerio. Cell Signal 2020; 73:109677. [DOI: 10.1016/j.cellsig.2020.109677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/10/2020] [Accepted: 05/23/2020] [Indexed: 11/25/2022]
|
12
|
Patel SK, Singh SK. Pyroglutamylated RFamide peptide 43: A putative modulator of testicular steroidogenesis. Andrology 2020; 8:1815-1823. [PMID: 32652859 DOI: 10.1111/andr.12864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/04/2020] [Accepted: 07/06/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND We have recently shown that QRFP and its receptor are predominantly expressed in germ cells, Sertoli cells and Leydig cells in mice testes. OBJECTIVE The present study investigated the role of QRFP in testicular steroidogenesis in mice. MATERIALS AND METHODS Both ex vivo and in vivo experiments were performed. For ex vivo, testicular tissues were cultured with 0, 10, 100 and 1000 nM QRFP, with or without hCG, for 6, 12 and 24 h, and media were used for testosterone assay. The hCG-stimulated testicular tissues were used for immunoblot of SF1, StAR, CYP11A1, 3β- and 17β-HSD. For in vivo, mice received bilateral intratesticular injection of saline or 0.3, 1 and 3nmol QRFP and were killed at 6, 12 and 24 h post-injection. Testosterone in serum was measured at above durations, while qRT-PCR of HMG-CoA synthase 1 and SR-B1 and immunoblot of steroidogenesis-related markers were performed at 24 h post-injection. RESULTS Testosterone production under basal and hCG-stimulated conditions increased in a time-dependent manner, and QRFP supplementation to testicular culture caused an increase and a decrease in hormone production. The effect of QRFP on testosterone production under hCG-stimulated culture or in vivo conditions at 6 and 24h was similar. At 6h, testosterone production increased at 10 and 100 nM and also at 0.3 and 1nmol QRFP, while it decreased at 1000 nM and 3 nmol doses. At 24 h, testosterone level decreased at lower concentrations (10 nM and 0.3 nmol) and thereafter increased at middle (100nM and 1nmol) and higher (1000 nM and 3 nmol) concentrations under both hCG-stimulated culture and in vivo. DISCUSSION AND CONCLUSION QRFP induced production of testosterone by modulating steroidogenic machinery at optimal doses and durations. Further, findings of in vivo study indicate that QRFP besides directly regulating testicular steroidogenesis may also have modulated other factors which act together in a holistic manner to control steroidogenesis.
Collapse
Affiliation(s)
- Shishir K Patel
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shio K Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
13
|
Wang T, Cao Z, Shen Z, Yang J, Chen X, Yang Z, Xu K, Xiang X, Yu Q, Song Y, Wang W, Tian Y, Sun L, Zhang L, Guo S, Zhou N. Existence and functions of a kisspeptin neuropeptide signaling system in a non-chordate deuterostome species. eLife 2020; 9:53370. [PMID: 32513385 PMCID: PMC7282810 DOI: 10.7554/elife.53370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
The kisspeptin system is a central modulator of the hypothalamic-pituitary-gonadal axis in vertebrates. Its existence outside the vertebrate lineage remains largely unknown. Here, we report the identification and characterization of the kisspeptin system in the sea cucumber Apostichopus japonicus. The gene encoding the kisspeptin precursor generates two mature neuropeptides, AjKiss1a and AjKiss1b. The receptors for these neuropeptides, AjKissR1 and AjKissR2, are strongly activated by synthetic A. japonicus and vertebrate kisspeptins, triggering a rapid intracellular mobilization of Ca2+, followed by receptor internalization. AjKissR1 and AjKissR2 share similar intracellular signaling pathways via Gαq/PLC/PKC/MAPK cascade, when activated by C-terminal decapeptide. The A. japonicus kisspeptin system functions in multiple tissues that are closely related to seasonal reproduction and metabolism. Overall, our findings uncover for the first time the existence and function of the kisspeptin system in a non-chordate species and provide new evidence to support the ancient origin of intracellular signaling and physiological functions that are mediated by this molecular system.
Collapse
Affiliation(s)
- Tianming Wang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China.,Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Zheng Cao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, China
| | - Zhangfei Shen
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, China
| | - Jingwen Yang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China.,Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Xu Chen
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China
| | - Zhen Yang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China
| | - Ke Xu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China
| | - Xiaowei Xiang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China
| | - Qiuhan Yu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China
| | - Yimin Song
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science College, Zhejiang Ocean University, Zhoushan, China
| | - Weiwei Wang
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, China
| | - Yanan Tian
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, China
| | - Lina Sun
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Libin Zhang
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Su Guo
- Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Dufour S, Quérat B, Tostivint H, Pasqualini C, Vaudry H, Rousseau K. Origin and Evolution of the Neuroendocrine Control of Reproduction in Vertebrates, With Special Focus on Genome and Gene Duplications. Physiol Rev 2019; 100:869-943. [PMID: 31625459 DOI: 10.1152/physrev.00009.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In humans, as in the other mammals, the neuroendocrine control of reproduction is ensured by the brain-pituitary gonadotropic axis. Multiple internal and environmental cues are integrated via brain neuronal networks, ultimately leading to the modulation of the activity of gonadotropin-releasing hormone (GnRH) neurons. The decapeptide GnRH is released into the hypothalamic-hypophysial portal blood system and stimulates the production of pituitary glycoprotein hormones, the two gonadotropins luteinizing hormone and follicle-stimulating hormone. A novel actor, the neuropeptide kisspeptin, acting upstream of GnRH, has attracted increasing attention in recent years. Other neuropeptides, such as gonadotropin-inhibiting hormone/RF-amide related peptide, and other members of the RF-amide peptide superfamily, as well as various nonpeptidic neuromediators such as dopamine and serotonin also provide a large panel of stimulatory or inhibitory regulators. This paper addresses the origin and evolution of the vertebrate gonadotropic axis. Brain-pituitary neuroendocrine axes are typical of vertebrates, the pituitary gland, mediator and amplifier of brain control on peripheral organs, being a vertebrate innovation. The paper reviews, from molecular and functional perspectives, the evolution across vertebrate radiation of some key actors of the vertebrate neuroendocrine control of reproduction and traces back their origin along the vertebrate lineage and in other metazoa before the emergence of vertebrates. A focus is given on how gene duplications, resulting from either local events or from whole genome duplication events, and followed by paralogous gene loss or conservation, might have shaped the evolutionary scenarios of current families of key actors of the gonadotropic axis.
Collapse
Affiliation(s)
- Sylvie Dufour
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Bruno Quérat
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Hervé Tostivint
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Catherine Pasqualini
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Hubert Vaudry
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Karine Rousseau
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| |
Collapse
|
15
|
Molecular codes and in vitro generation of hypocretin and melanin concentrating hormone neurons. Proc Natl Acad Sci U S A 2019; 116:17061-17070. [PMID: 31375626 PMCID: PMC6708384 DOI: 10.1073/pnas.1902148116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypocretin/orexin (HCRT) and melanin concentrating hormone (MCH) neuropeptides are exclusively produced by the lateral hypothalamus and play important roles in sleep, metabolism, reward, and motivation. Loss of HCRT (ligands or receptors) causes the sleep disorder narcolepsy with cataplexy in humans and in animal models. How these neuropeptides are produced and involved in diverse functions remain unknown. Here, we developed methods to sort and purify HCRT and MCH neurons from the mouse late embryonic hypothalamus. RNA sequencing revealed key factors of fate determination for HCRT (Peg3, Ahr1, Six6, Nr2f2, and Prrx1) and MCH (Lmx1, Gbx2, and Peg3) neurons. Loss of Peg3 in mice significantly reduces HCRT and MCH cell numbers, while knock-down of a Peg3 ortholog in zebrafish completely abolishes their expression, resulting in a 2-fold increase in sleep amount. We also found that loss of HCRT neurons in Hcrt-ataxin-3 mice results in a specific 50% decrease in another orexigenic neuropeptide, QRFP, that might explain the metabolic syndrome in narcolepsy. The transcriptome results were used to develop protocols for the production of HCRT and MCH neurons from induced pluripotent stem cells and ascorbic acid was found necessary for HCRT and BMP7 for MCH cell differentiation. Our results provide a platform to understand the development and expression of HCRT and MCH and their multiple functions in health and disease.
Collapse
|
16
|
Ukena K. Avian and murine neurosecretory protein GL participates in the regulation of feeding and energy metabolism. Gen Comp Endocrinol 2018; 260:164-170. [PMID: 28951261 DOI: 10.1016/j.ygcen.2017.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/15/2017] [Accepted: 09/21/2017] [Indexed: 01/14/2023]
Abstract
Probing previously unknown neuropeptides and/or peptide hormones is essential for our understanding of the regulation of energy homeostasis in the brain. We recently performed a cDNA subtractive screening of the chicken hypothalamus, which contained one of the feeding and energy metabolic centers. We found a gene encoding a novel protein of 182 amino acid residues, including one putative small secretory protein of 80 amino acid residues. The C-terminal amino acids of the small protein were Gly-Leu-NH2, and as a result, the small protein was termed neurosecretory protein GL (NPGL). Subcutaneous and intracerebroventricular infusions of NPGL increased body mass gain in chicks, suggesting a central role for this protein in regulating growth and energy homeostasis. A database search revealed that the Npgl gene is conserved in vertebrates, including mice and rats. This review summarizes the advances in the characterization, localization, and biological action of NPGL, in birds and rodents.
Collapse
Affiliation(s)
- Kazuyoshi Ukena
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan.
| |
Collapse
|
17
|
Elphick MR, Mirabeau O, Larhammar D. Evolution of neuropeptide signalling systems. ACTA ACUST UNITED AC 2018; 221:221/3/jeb151092. [PMID: 29440283 PMCID: PMC5818035 DOI: 10.1242/jeb.151092] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuropeptides are a diverse class of neuronal signalling molecules that regulate physiological processes and behaviour in animals. However, determining the relationships and evolutionary origins of the heterogeneous assemblage of neuropeptides identified in a range of phyla has presented a huge challenge for comparative physiologists. Here, we review revolutionary insights into the evolution of neuropeptide signalling that have been obtained recently through comparative analysis of genome/transcriptome sequence data and by ‘deorphanisation’ of neuropeptide receptors. The evolutionary origins of at least 30 neuropeptide signalling systems have been traced to the common ancestor of protostomes and deuterostomes. Furthermore, two rounds of genome duplication gave rise to an expanded repertoire of neuropeptide signalling systems in the vertebrate lineage, enabling neofunctionalisation and/or subfunctionalisation, but with lineage-specific gene loss and/or additional gene or genome duplications generating complex patterns in the phylogenetic distribution of paralogous neuropeptide signalling systems. We are entering a new era in neuropeptide research where it has become feasible to compare the physiological roles of orthologous and paralogous neuropeptides in a wide range of phyla. Moreover, the ambitious mission to reconstruct the evolution of neuropeptide function in the animal kingdom now represents a tangible challenge for the future. Summary: A review of the revolutionary advances in our knowledge of the evolution of neuropeptide signalling systems that have been enabled by comparative genomics and neuropeptide receptor deorphanisation.
Collapse
Affiliation(s)
- Maurice R Elphick
- School of Biological & Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Olivier Mirabeau
- Genetics and Biology of Cancers Unit, Institut Curie, INSERM U830, Paris Sciences et Lettres Research University, Paris 75005, France
| | - Dan Larhammar
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, 75124 Uppsala, Sweden
| |
Collapse
|
18
|
Alavi MS, Shamsizadeh A, Azhdari-Zarmehri H, Roohbakhsh A. Orphan G protein-coupled receptors: The role in CNS disorders. Biomed Pharmacother 2017; 98:222-232. [PMID: 29268243 DOI: 10.1016/j.biopha.2017.12.056] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
There are various types of receptors in the central nervous system (CNS). G protein-coupled receptors (GPCRs) have the highest expression with a wide range of physiological functions. A newer sub group of these receptors namely orphan GPCRs have been discovered. GPR3, GPR6, GPR17, GPR26, GPR37, GPR39, GPR40, GPR50, GPR52, GPR54, GPR55, GPR85, GPR88, GPR103, and GPR139 are the selected orphan GPCRs for this article. Their roles in the central nervous system have not been understood well so far. However, recent studies show that they may have very important functions in the CNS. Hence, in the present study, we reviewed most recent findings regarding the physiological roles of the selected orphan GPCRs in the CNS. After a brief presentation of each receptor, considering the results from genetic and pharmacological manipulation of the receptors, their roles in the pathophysiology of different diseases and disorders including anxiety, depression, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, and substance abuse will be discussed. At present, our knowledge regarding the role of GPCRs in the brain is very limited. However, previous limited studies show that orphan GPCRs have an important place in psychopharmacology and these receptors are potential new targets for the treatment of major CNS diseases.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shamsizadeh
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hassan Azhdari-Zarmehri
- Department of Basic Medical Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Leprince J, Bagnol D, Bureau R, Fukusumi S, Granata R, Hinuma S, Larhammar D, Primeaux S, Sopkova-de Oliveiras Santos J, Tsutsui K, Ukena K, Vaudry H. The Arg-Phe-amide peptide 26RFa/glutamine RF-amide peptide and its receptor: IUPHAR Review 24. Br J Pharmacol 2017; 174:3573-3607. [PMID: 28613414 DOI: 10.1111/bph.13907] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/30/2017] [Accepted: 06/05/2017] [Indexed: 12/21/2022] Open
Abstract
The RFamide neuropeptide 26RFa was first isolated from the brain of the European green frog on the basis of cross-reactivity with antibodies raised against bovine neuropeptide FF (NPFF). 26RFa and its N-terminally extended form glutamine RF-amide peptide (QRFP) have been identified as cognate ligands of the former orphan receptor GPR103, now renamed glutamine RF-amide peptide receptor (QRFP receptor). The 26RFa/QRFP precursor has been characterized in various mammalian and non-mammalian species. In the brain of mammals, including humans, 26RFa/QRFP mRNA is almost exclusively expressed in hypothalamic nuclei. The 26RFa/QRFP transcript is also present in various organs especially in endocrine glands. While humans express only one QRFP receptor, two isoforms are present in rodents. The QRFP receptor genes are widely expressed in the CNS and in peripheral tissues, notably in bone, heart, kidney, pancreas and testis. Structure-activity relationship studies have led to the identification of low MW peptidergic agonists and antagonists of QRFP receptor. Concurrently, several selective non-peptidic antagonists have been designed from high-throughput screening hit optimization. Consistent with the widespread distribution of QRFP receptor mRNA and 26RFa binding sites, 26RFa/QRFP exerts a large range of biological activities, notably in the control of energy homeostasis, bone formation and nociception that are mediated by QRFP receptor or NPFF2. The present report reviews the current knowledge concerning the 26RFa/QRFP-QRFP receptor system and discusses the potential use of selective QRFP receptor ligands for therapeutic applications.
Collapse
Affiliation(s)
- Jérôme Leprince
- INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandy University, Rouen, France
| | - Didier Bagnol
- CNS Drug Discovery, Arena Pharmaceuticals Inc., San Diego, CA, USA
| | - Ronan Bureau
- Normandy Centre for Studies and Research on Medicines (CERMN), Normandy University, Caen, France
| | - Shoji Fukusumi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology, Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Shuji Hinuma
- Department of Food and Nutrition, Faculty of Human Life Science, Senri Kinran University, Suita-City, Osaka, Japan
| | - Dan Larhammar
- Department of Neuroscience, Unit of Pharmacology, Uppsala University, Uppsala, Sweden
| | - Stefany Primeaux
- Department of Physiology, Joint Diabetes, Endocrinology & Metabolism Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, Center for Medical Life Science, Tokyo, Japan
| | - Kazuyoshi Ukena
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | - Hubert Vaudry
- INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandy University, Rouen, France
| |
Collapse
|
20
|
Gesmundo I, Villanova T, Banfi D, Gamba G, Granata R. Role of Melatonin, Galanin, and RFamide Neuropeptides QRFP26 and QRFP43 in the Neuroendocrine Control of Pancreatic β-Cell Function. Front Endocrinol (Lausanne) 2017; 8:143. [PMID: 28729853 PMCID: PMC5499649 DOI: 10.3389/fendo.2017.00143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/06/2017] [Indexed: 12/12/2022] Open
Abstract
Glucose homeostasis is finely regulated by a number of hormones and peptides released mainly from the brain, gastrointestinal tract, and muscle, regulating pancreatic secretion through cellular receptors and their signal transduction cascades. The endocrine function of the pancreas is controlled by islets within the exocrine pancreatic tissue that release hormones like insulin, glucagon, somatostatin, pancreatic polypeptide, and ghrelin. Moreover, both exocrine and endocrine pancreatic functions are regulated by a variety of hormonal and neural mechanisms, such as ghrelin, glucagon-like peptide, glucose-dependent insulinotropic polypeptide, or the inhibitory peptide somatostatin. In this review, we describe the role of neurohormones that have been less characterized compared to others, on the regulation of insulin secretion. In particular, we will focus on melatonin, galanin, and RFamide neuropeptides QRFP26 and QRFP43, which display either insulinotropic or insulinostatic effects. In fact, in addition to other hormones, amino acids, cytokines, and a variety of proteins, brain-derived hormones are now considered as key regulators of glucose homeostasis, representing potential therapeutic targets for the treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Iacopo Gesmundo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Tania Villanova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dana Banfi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giacomo Gamba
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Riccarda Granata
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
- *Correspondence: Riccarda Granata,
| |
Collapse
|
21
|
Hu CK, Southey BR, Romanova EV, Maruska KP, Sweedler JV, Fernald RD. Identification of prohormones and pituitary neuropeptides in the African cichlid, Astatotilapia burtoni. BMC Genomics 2016; 17:660. [PMID: 27543050 PMCID: PMC4992253 DOI: 10.1186/s12864-016-2914-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/06/2016] [Indexed: 12/14/2022] Open
Abstract
Background Cichlid fishes have evolved remarkably diverse reproductive, social, and feeding behaviors. Cell-to-cell signaling molecules, notably neuropeptides and peptide hormones, are known to regulate these behaviors across vertebrates. This class of signaling molecules derives from prohormone genes that have undergone multiple duplications and losses in fishes. Whether and how subfunctionalization, neofunctionalization, or losses of neuropeptides and peptide hormones have contributed to fish behavioral diversity is largely unknown. Information on fish prohormones has been limited and is complicated by the whole genome duplication of the teleost ancestor. We combined bioinformatics, mass spectrometry-enabled peptidomics, and molecular techniques to identify the suite of neuropeptide prohormones and pituitary peptide products in Astatotilapia burtoni, a well-studied member of the diverse African cichlid clade. Results Utilizing the A. burtoni genome, we identified 148 prohormone genes, with 21 identified as a single copy and 39 with at least 2 duplicated copies. Retention of prohormone duplicates was therefore 41 %, which is markedly above previous reports for the genome-wide average in teleosts. Beyond the expected whole genome duplication, differences between cichlids and mammals can be attributed to gene loss in tetrapods and additional duplication after divergence. Mass spectrometric analysis of the pituitary identified 620 unique peptide sequences that were matched to 120 unique proteins. Finally, we used in situ hybridization to localize the expression of galanin, a prohormone with exceptional sequence divergence in cichlids, as well as the expression of a proopiomelanocortin, prohormone that has undergone an additional duplication in some bony fish lineages. Conclusion We characterized the A. burtoni prohormone complement. Two thirds of prohormone families contain duplications either from the teleost whole genome duplication or a more recent duplication. Our bioinformatic and mass spectrometric findings provide information on a major vertebrate clade that will further our understanding of the functional ramifications of these prohormone losses, duplications, and sequence changes across vertebrate evolution. In the context of the cichlid radiation, these findings will also facilitate the exploration of neuropeptide and peptide hormone function in behavioral diversity both within A. burtoni and across cichlid and other fish species. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2914-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caroline K Hu
- Department of Biology, Stanford University, Stanford, CA, 94305, USA.,Present address: Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Karen P Maruska
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Russell D Fernald
- Department of Biology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
22
|
Palotai M, Telegdy G. Anxiolytic effect of the GPR103 receptor agonist peptide P550 (homolog of neuropeptide 26RFa) in mice. Involvement of neurotransmitters. Peptides 2016; 82:20-25. [PMID: 27224020 DOI: 10.1016/j.peptides.2016.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
The GPR103 receptor is a G protein-coupled receptor, which plays a role in several physiological functions. However, the role of the GPR103 receptor in anxiety has not been clarified. The first aim of our study was to elucidate the involvement of the GPR103 receptor in anxious behavior. Mice were treated with peptide P550, which is the mouse homolog of neuropeptide 26RFa and has similar activity for the GPR103 receptor as neuropeptide 26RFa. The anxious behavior was investigated using an elevated plus-maze paradigm. The second aim of our study was to investigate the underlying neurotransmissions. Accordingly, mice were pretreated with a nonselective muscarinic acetylcholine receptor antagonist, atropine, a γ-aminobutyric acid subunit A (GABAA) receptor antagonist, bicuculline, a non-selective 5-HT2 serotonergic receptor antagonist, cyproheptadine, a mixed 5-HT1/5-HT2 serotonergic receptor antagonist, methysergide, a D2, D3, D4 dopamine receptor antagonist, haloperidol, a nonselective α-adrenergic receptor antagonist, phenoxybenzamine and a nonselective β-adrenergic receptor antagonist, propranolol. Our results demonstrated that peptide P550 reduces anxious behavior in elevated plus maze test in mice. Our study shows also that GABAA-ergic, α- and β-adrenergic transmissions are all involved in this action, whereas 5-HT1 and 5-HT2 serotonergic, muscarinic cholinergic and D2, D3, D4 dopaminergic mechanisms may not be implicated.
Collapse
Affiliation(s)
- Miklos Palotai
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary.
| | - Gyula Telegdy
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, Hungary; Neuroscience Research Group of the Hungarian Academy of Sciences, Szeged, Hungary
| |
Collapse
|
23
|
Schreiber AL, Arceneaux KP, Malbrue RA, Mouton AJ, Chen CS, Bench EM, Braymer HD, Primeaux SD. The effects of high fat diet and estradiol on hypothalamic prepro-QRFP mRNA expression in female rats. Neuropeptides 2016; 58:103-9. [PMID: 26823127 PMCID: PMC4960001 DOI: 10.1016/j.npep.2016.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/22/2015] [Accepted: 01/14/2016] [Indexed: 12/11/2022]
Abstract
Estradiol (E2) is a potent regulator of feeding behavior, body weight and adiposity in females. The hypothalamic neuropeptide, QRFP, is an orexigenic peptide that increases the consumption of high fat diet (HFD) in intact female rats. Therefore, the goal of the current series of studies was to elucidate the effects of E2 on the expression of hypothalamic QRFP and its receptors, QRFP-r1 and QRFP-r2, in female rats fed a HFD. Alterations in prepro-QRFP, QRFP-r1, and QRFP-r2 expression across the estrous cycle, following ovariectomy (OVX) and following estradiol benzoate (EB) treatment were assessed in the ventral medial nucleus of the hypothalamus/arcuate nucleus (VMH/ARC) and the lateral hypothalamus. In intact females, consumption of HFD increased prepro-QRFP and QRFP-r1 mRNA levels in the VMH/ARC during diestrus, a phase associated with increased food intake and low levels of E2. To assess the effects of diminished endogenous E2, rats were ovariectomized. HFD consumption and OVX increased prepro-QRFP mRNA in the VMH/ARC. Ovariectomized rats consuming HFD expressed the highest levels of QRFP. In the third experiment, all rats received EB replacement every 4days following OVX to examine the effects of E2 on QRFP expression. Prepro-QRFP, QRFP-r1 and QRFP-r2 mRNA were assessed prior to and following EB administration. EB replacement significantly reduced prepro-QRFP mRNA expression in the VMH/ARC. Overall these studies support a role for E2 in the regulation of prepro-QRFP mRNA in the VMH/ARC and suggest that E2's effects on food intake may be via a direct effect on the orexigenic peptide, QRFP.
Collapse
Affiliation(s)
- Allyson L Schreiber
- Department of Physiology, 1901 Perdido Street, Louisiana State University Health Science Center-New Orleans, New Orleans, LA 70112, USA
| | - Kenneth P Arceneaux
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Raphael A Malbrue
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Alan J Mouton
- Department of Physiology, 1901 Perdido Street, Louisiana State University Health Science Center-New Orleans, New Orleans, LA 70112, USA
| | - Christina S Chen
- Joint Diabetes, Endocrinology & Metabolism Program, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Elias M Bench
- Joint Diabetes, Endocrinology & Metabolism Program, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - H Douglas Braymer
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Stefany D Primeaux
- Department of Physiology, 1901 Perdido Street, Louisiana State University Health Science Center-New Orleans, New Orleans, LA 70112, USA; Joint Diabetes, Endocrinology & Metabolism Program, 6400 Perkins Road, Baton Rouge, LA 70808, USA; Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
24
|
Abstract
UNLABELLED The hypothalamus plays an important role in regulating sleep, but few hypothalamic sleep-promoting signaling pathways have been identified. Here we demonstrate a role for the neuropeptide QRFP (also known as P518 and 26RFa) and its receptors in regulating sleep in zebrafish, a diurnal vertebrate. We show that QRFP is expressed in ∼10 hypothalamic neurons in zebrafish larvae, which project to the hypothalamus, hindbrain, and spinal cord, including regions that express the two zebrafish QRFP receptor paralogs. We find that the overexpression of QRFP inhibits locomotor activity during the day, whereas mutation of qrfp or its receptors results in increased locomotor activity and decreased sleep during the day. Despite the restriction of these phenotypes to the day, the circadian clock does not regulate qrfp expression, and entrained circadian rhythms are not required for QRFP-induced rest. Instead, we find that QRFP overexpression decreases locomotor activity largely in a light-specific manner. Our results suggest that QRFP signaling plays an important role in promoting sleep and may underlie some aspects of hypothalamic sleep control. SIGNIFICANCE STATEMENT The hypothalamus is thought to play a key role in regulating sleep in vertebrate animals, but few sleep-promoting signaling pathways that function in the hypothalamus have been identified. Here we use the zebrafish, a diurnal vertebrate, to functionally and anatomically characterize the neuropeptide QRFP. We show that QRFP is exclusively expressed in a small number of neurons in the larval zebrafish hypothalamus that project widely in the brain. We also show that QRFP overexpression reduces locomotor activity, whereas animals that lack QRFP signaling are more active and sleep less. These results suggest that QRFP signaling participates in the hypothalamic regulation of sleep.
Collapse
|
25
|
Osugi T, Son YL, Ubuka T, Satake H, Tsutsui K. RFamide peptides in agnathans and basal chordates. Gen Comp Endocrinol 2016; 227:94-100. [PMID: 26130238 DOI: 10.1016/j.ygcen.2015.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/12/2022]
Abstract
Since a peptide with a C-terminal Arg-Phe-NH2 (RFamide peptide) was first identified in the ganglia of the venus clam in 1977, RFamide peptides have been found in the nervous system of both invertebrates and vertebrates. In vertebrates, the RFamide peptide family includes gonadotropin-inhibitory hormone (GnIH), neuropeptide FF (NPFF), prolactin-releasing peptide (PrRP), pyroglutamylated RFamide peptide/26RFamide peptide (QRFP/26RFa), and kisspeptins (kiss1 and kiss2). They are involved in important functions such as the release of hormones, regulation of sexual or social behavior, pain transmission, reproduction, and feeding. In contrast to tetrapods and jawed fish, the information available on RFamide peptides in agnathans and basal chordates is limited, thus preventing further insights into the evolution of RFamide peptides in vertebrates. In this review, we focus on the previous research and recent advances in the studies on RFamide peptides in agnathans and basal chordates. In agnathans, the genes encoding GnIH, NPFF, and PrRP precursors and the mature peptides have been identified in lamprey (Petromyzon marinus) and hagfish (Paramyxine atami). Putative kiss1 and kiss2 genes have also been found in the genome database of lamprey. In basal chordates, namely, in amphioxus (Branchiostoma japonicum), a common ancestral form of GnIH and NPFF genes and their mature peptides, as well as the ortholog of the QRFP gene have been identified. The studies revealed that the number of orthologs of vertebrate RFamide peptides present in agnathans and basal chordates is greater than expected, suggesting that the vertebrate RFamide peptides might have emerged and expanded at an early stage of chordate evolution.
Collapse
Affiliation(s)
- Tomohiro Osugi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 1-1-1 Wakayamadai, Shimamoto, Mishima, Osaka 618-8503, Japan.
| | - You Lee Son
- Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Tokyo 162-8480, Japan
| | - Takayoshi Ubuka
- Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Tokyo 162-8480, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 1-1-1 Wakayamadai, Shimamoto, Mishima, Osaka 618-8503, Japan
| | - Kazuyoshi Tsutsui
- Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Tokyo 162-8480, Japan
| |
Collapse
|
26
|
Davies J, Chen J, Pink R, Carter D, Saunders N, Sotiriadis G, Bai B, Pan Y, Howlett D, Payne A, Randeva H, Karteris E. Orexin receptors exert a neuroprotective effect in Alzheimer's disease (AD) via heterodimerization with GPR103. Sci Rep 2015; 5:12584. [PMID: 26223541 PMCID: PMC4519789 DOI: 10.1038/srep12584] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/29/2015] [Indexed: 12/22/2022] Open
Abstract
Orexins are neuropeptides that regulate the sleep-wake cycle and feeding behaviour. QRFP is a newly discovered neuropeptide which exerts similar orexigenic activity, thus playing an important role in energy homeostasis and regulation of appetite. The exact expression and signalling characteristics and physiological actions of QRFP and its receptor GPR103 are poorly understood. Alzheimer's disease (AD) patients experience increased nocturnal activity, excessive daytime sleepiness, and weight loss. We hypothesised therefore that orexins and QRFP might be implicated in the pathophysiology of AD. We report that the down-regulation of hippocampal orexin receptors (OXRs) and GPR103 particularly in the cornu ammonis (CA) subfield from AD patients suffering from early onset familial AD (EOFAD) and late onset familial AD (LOAD). Using an in vitro model we demonstrate that this downregulation is due to to Aβ-plaque formation and tau hyper-phosphorylation. Transcriptomics revealed a neuroprotective role for both orexins and QRFP. Finally we provide conclusive evidence using BRET and FRET that OXRs and GPR103 form functional hetero-dimers to exert their effects involving activation of ERK1/2. Pharmacological intervention directed at the orexigenic system may prove to be an attractive avenue towards the discovery of novel therapeutics for diseases such as AD and improving neuroprotective signalling pathways.
Collapse
Affiliation(s)
- Julie Davies
- Biosciences, College of Health and Life Sciences, Brunel University, Uxbridge, UB8 3PH, UK
| | - Jing Chen
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, P.R. China
| | - Ryan Pink
- Department of Biological and Medical Sciences, Oxford Brookes University, UK
| | - David Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, UK
| | - Nigel Saunders
- Centre for Systems and Synthetic Biology, Brunel University, Uxbridge UB83PH, UK
| | - Georgios Sotiriadis
- Biosciences, College of Health and Life Sciences, Brunel University, Uxbridge, UB8 3PH, UK
| | - Bo Bai
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, P.R. China
| | - Yanyou Pan
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, P.R. China
| | - David Howlett
- Wolfson Centre for Age Related Diseases, King’s College London, London, SE11UL, UK
| | - Annette Payne
- Department of Computer Science, College of Engineering, Design and Physical Sciences, Brunel University, Uxbridge UB8 3PH, UK
| | - Harpal Randeva
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Emmanouil Karteris
- Biosciences, College of Health and Life Sciences, Brunel University, Uxbridge, UB8 3PH, UK
| |
Collapse
|
27
|
Larhammar D, Xu B, Bergqvist CA. Unexpected multiplicity of QRFP receptors in early vertebrate evolution. Front Neurosci 2014; 8:337. [PMID: 25386115 PMCID: PMC4208404 DOI: 10.3389/fnins.2014.00337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/06/2014] [Indexed: 12/04/2022] Open
Abstract
The neuropeptide QRFP, also called 26RFa, and its G protein-coupled receptor GPR103 have been identified in all vertebrates investigated. In mammals, this peptide-receptor pair has been found to have several effects including stimulation of appetite. Recently, we reported that a QRFP peptide is present in amphioxus, Branchiostoma floridae, and we also identified a QRFP receptor (QRFPR) that mediates a functional response to sub-nanomolar concentrations of the amphioxus peptide as well as short and long human QRFP (Xu et al., submitted). Because the ancestral vertebrate underwent two tetraploidizations, it might be expected that duplicates of the QRFP gene and its receptor gene may exist. Indeed, we report here the identification of multiple vertebrate QRFPR genes. Three QRFPR genes are present in the coelacanth Latimeria chalumnae, representing an early diverging sarcopterygian lineage. Three QRFPR genes are present in the basal actinopterygian fish, the spotted gar. Phylogenetic and chromosomal analyses show that only two of these receptor genes are orthologous between the two species, thus demonstrating a total of four distinct vertebrate genes. Three of the QRFPR genes resulted from the early vertebrate tetraploidizations and were copied along with syntenic neuropeptide Y receptor genes. The fourth QRFPR gene may be an even older and distinct lineage. Because mammals and birds have only a single QRFPR gene, this means that three genes have been lost in these lineages, and at least one of these was lost independently in mammals and birds because it is still present in a turtle. In conclusion, these results show that the QRFP system gained considerable complexity in the early stages of vertebrate evolution and still maintains much of this in some lineages, and that it has been secondarily reduced in mammals.
Collapse
Affiliation(s)
- Dan Larhammar
- Unit of Pharmacology, Science for Life Laboratory, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Bo Xu
- Unit of Pharmacology, Science for Life Laboratory, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Christina A Bergqvist
- Unit of Pharmacology, Science for Life Laboratory, Department of Neuroscience, Uppsala University Uppsala, Sweden
| |
Collapse
|
28
|
Affiliation(s)
- Hubert Vaudry
- INSERM U982International Associated Laboratory Samuel de Champlain, PRIMACEN, IRIB, University of Rouen, 76821 Mont-Saint-Aignan, France
| |
Collapse
|
29
|
Elphick MR, Mirabeau O. The Evolution and Variety of RFamide-Type Neuropeptides: Insights from Deuterostomian Invertebrates. Front Endocrinol (Lausanne) 2014; 5:93. [PMID: 24994999 PMCID: PMC4062910 DOI: 10.3389/fendo.2014.00093] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/04/2014] [Indexed: 11/30/2022] Open
Abstract
Five families of neuropeptides that have a C-terminal RFamide motif have been identified in vertebrates: (1) gonadotropin-inhibitory hormone (GnIH), (2) neuropeptide FF (NPFF), (3) pyroglutamylated RFamide peptide (QRFP), (4) prolactin-releasing peptide (PrRP), and (5) Kisspeptin. Experimental demonstration of neuropeptide-receptor pairings combined with comprehensive analysis of genomic and/or transcriptomic sequence data indicate that, with the exception of the deuterostomian PrRP system, the evolutionary origins of these neuropeptides can be traced back to the common ancestor of bilaterians. Here, we review the occurrence of homologs of vertebrate RFamide-type neuropeptides and their receptors in deuterostomian invertebrates - urochordates, cephalochordates, hemichordates, and echinoderms. Extending analysis of the occurrence of the RFamide motif in other bilaterian neuropeptide families reveals RFamide-type peptides that have acquired modified C-terminal characteristics in the vertebrate lineage (e.g., NPY/NPF), neuropeptide families where the RFamide motif is unique to protostomian members (e.g., CCK/sulfakinins), and RFamide-type peptides that have been lost in the vertebrate lineage (e.g., luqins). Furthermore, the RFamide motif is also a feature of neuropeptide families with a more restricted phylogenetic distribution (e.g., the prototypical FMRFamide-related neuropeptides in protostomes). Thus, the RFamide motif is both an ancient and a convergent feature of neuropeptides, with conservation, acquisition, or loss of this motif occurring in different branches of the animal kingdom.
Collapse
Affiliation(s)
- Maurice R. Elphick
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
- *Correspondence: Maurice R. Elphick, School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK e-mail:
| | - Olivier Mirabeau
- Institut Curie, Cancer Genetics Unit, Inserm U830, Paris, France
| |
Collapse
|
30
|
Yun S, Kim DK, Furlong M, Hwang JI, Vaudry H, Seong JY. Does Kisspeptin Belong to the Proposed RF-Amide Peptide Family? Front Endocrinol (Lausanne) 2014; 5:134. [PMID: 25165463 PMCID: PMC4131245 DOI: 10.3389/fendo.2014.00134] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/30/2014] [Indexed: 11/13/2022] Open
Abstract
Kisspeptin (KISS) plays a key role in regulating reproduction by binding to its receptor, GPR54. Because of the Arg-Phe (RF) sequence at its carboxyl terminus, KISS has been proposed to be a member of the RF-amide peptide family consisting of neuropeptide FF (NPFF), neuropeptide VF (NPVF), pyroglutamylated RF-amide peptide (QRFP), and prolactin-releasing hormone (PRLH). Evolutionary relationships of protein families can be determined through phylogenetic analysis. However, phylogenetic analysis among related peptide families often fails to provide sufficient information because only short mature peptide sequences from full preprohormone sequences are conserved. Considering the concept of the coevolution of peptide ligands and their cognate receptors, evolutionary relationships among related receptor families provide clues to explore relationships between their peptides. Although receptors for NPFF, NPVF, and QRFP are phylogenetically clustered together, receptors for PRLH and KISS are on different branches of the phylogenetic tree. In particular, KISS has been proposed to be a member of the KISS/galanin/spexin family based on synteny analysis and the phylogenetic relationship between their receptors. This article discusses the evolutionary history of the receptors for the proposed RF-amide peptide family and proposes that, from an evolutionary aspect, KISS has emerged from an ancestor, which is distinct from those of the other RF-amide peptides, and so should be classed separately.
Collapse
Affiliation(s)
- Seongsik Yun
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Dong-Kyu Kim
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Michael Furlong
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Jong-Ik Hwang
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Hubert Vaudry
- INSERM U982, University of Rouen, Mont-Saint-Aignan, France
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, Seoul, South Korea
- *Correspondence: Jae Young Seong, Graduate School of Medicine, Korea University, Seoul 136-705, South Korea e-mail:
| |
Collapse
|
31
|
Sandvik GK, Hodne K, Haug TM, Okubo K, Weltzien FA. RFamide Peptides in Early Vertebrate Development. Front Endocrinol (Lausanne) 2014; 5:203. [PMID: 25538682 PMCID: PMC4255600 DOI: 10.3389/fendo.2014.00203] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/16/2014] [Indexed: 12/17/2022] Open
Abstract
RFamides (RFa) are neuropeptides involved in many different physiological processes in vertebrates, such as reproductive behavior, pubertal activation of the reproductive endocrine axis, control of feeding behavior, and pain modulation. As research has focused mostly on their role in adult vertebrates, the possible roles of these peptides during development are poorly understood. However, the few studies that exist show that RFa are expressed early in development in different vertebrate classes, perhaps mostly associated with the central nervous system. Interestingly, the related peptide family of FMRFa has been shown to be important for brain development in invertebrates. In a teleost, the Japanese medaka, knockdown of genes in the Kiss system indicates that Kiss ligands and receptors are vital for brain development, but few other functional studies exist. Here, we review the literature of RFa in early vertebrate development, including the possible functional roles these peptides may play.
Collapse
Affiliation(s)
- Guro Katrine Sandvik
- Department of Basic Sciences and Aquatic medicine, Norwegian University of Life Sciences , Oslo , Norway
| | - Kjetil Hodne
- Institute for Experimental Medical Research, Oslo University Hospital , Oslo , Norway
| | | | - Kataaki Okubo
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Bunkyo , Japan
| | - Finn-Arne Weltzien
- Department of Basic Sciences and Aquatic medicine, Norwegian University of Life Sciences , Oslo , Norway
| |
Collapse
|