1
|
Wiesner D, Feldengut S, Woelfle S, Boeckers TM, Ludolph AC, Roselli F, Del Tredici K. Neuropeptide FF (NPFF)-positive nerve cells of the human cerebral cortex and white matter in controls, selected neurodegenerative diseases, and schizophrenia. Acta Neuropathol Commun 2024; 12:108. [PMID: 38943180 PMCID: PMC11212262 DOI: 10.1186/s40478-024-01792-1] [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: 02/23/2024] [Accepted: 05/02/2024] [Indexed: 07/01/2024] Open
Abstract
We quantified and determined for the first time the distribution pattern of the neuropeptide NPFF in the human cerebral cortex and subjacent white matter. To do so, we studied n = 9 cases without neurological disorders and n = 22 cases with neurodegenerative diseases, including sporadic amyotrophic lateral sclerosis (ALS, n = 8), Alzheimer's disease (AD, n = 8), Pick's disease (PiD, n = 3), and schizophrenia (n = 3). NPFF-immunopositive cells were located chiefly, but not exclusively, in the superficial white matter and constituted there a subpopulation of white matter interstitial cells (WMIC): Pyramidal-like and multipolar somata predominated in the gyral crowns, whereas bipolar and ovoid somata predominated in the cortex surrounding the sulci. Their sparsely ramified axons were unmyelinated and exhibited NPFF-positive bead-like varicosities. We found significantly fewer NPFF-immunopositive cells in the gray matter of the frontal, cingulate, and superior temporal gyri of both sporadic ALS and late-stage AD patients than in controls, and significantly fewer NPFF-positive cells in the subjacent as well as deep white matter of the frontal gyrus of these patients compared to controls. Notably, the number of NPFF-positive cells was also significantly lower in the hippocampal formation in AD compared to controls. In PiD, NPFF-positive cells were present in significantly lower numbers in the gray and white matter of the cingulate and frontal gyrii in comparison to controls. In schizophrenic patients, lower wNPFF cell counts in the neocortex were significant and global (cingulate, frontal, superior temporal gyrus, medial, and inferior gyri). The precise functions of NPFF-positive cells and their relationship to the superficial corticocortical white matter U-fibers are currently unknown. Here, NPFF immunohistochemistry and expression characterize a previously unrecognized population of cells in the human brain, thereby providing a new entry-point for investigating their physiological and pathophysiological roles.
Collapse
Affiliation(s)
- Diana Wiesner
- Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany
- DZNE, Ulm Site, 89081, Ulm, Germany
| | - Simone Feldengut
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany
| | - Sarah Woelfle
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Tobias M Boeckers
- DZNE, Ulm Site, 89081, Ulm, Germany
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | | | - Francesco Roselli
- Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany.
- DZNE, Ulm Site, 89081, Ulm, Germany.
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany
| |
Collapse
|
2
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [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: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
3
|
Strnadová V, Morgan A, Škrlová M, Haasová E, Bardová K, Myšková A, Sýkora D, Kuneš J, Železná B, Maletínská L. Peripheral administration of lipidized NPAF and NPFF analogs does not influence central food intake regulation but induces anxiety-like behavior. Neuropeptides 2024; 104:102417. [PMID: 38422597 DOI: 10.1016/j.npep.2024.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
RF-amide peptides influence multiple physiological processes, including the regulation of appetite, stress responses, behavior, and reproductive and endocrine functions. In this study, we examined the roles of neuropeptide FF receptors (NPFFR1 and NPFFR2) by generating several lipidized analogs of neuropeptide AF (NPAF) and 1DMe, a stable analog of neuropeptide FF (NPFF). These analogs were administered peripherally for the first time to investigate their effects on food intake and other potential physiological outcomes. Lipidized NPAF and 1DMe analogs exhibited enhanced stability and increased pharmacokinetics. These analogs demonstrated preserved high affinity for NPFFR2 in the nanomolar range, while the binding affinity for NPFFR1 was tens of nanomoles. They activated the ERK and Akt signaling pathways in cells overexpressing the NPFFR1 and NPFFR2 receptors. Acute food intake in fasted mice decreased after the peripheral administration of oct-NPAF or oct-1DMe. However, this effect was not as pronounced as that observed after the injection of palm11-PrRP31, a potent anorexigenic compound used as a comparator that binds to GPR10 and the NPFFR2 receptor with high affinity. Neither oct-1DMe nor oct-NPAF decreased food intake or body weight in mice with diet-induced obesity during long-term treatment. In mice treated with oct-1DMe, we observed decreased activity in the central zone during the open field test and decreased activity in the open arms of the elevated plus maze. Furthermore, we observed a decrease in plasma noradrenaline levels and an increase in plasma corticosterone levels, as well as an increase in Crh expression in the hypothalamus. Moreover, neuronal activity in the hypothalamus was increased after treatment with oct-1DMe. In this study, we report that oct-1DMe did not have any long-term effects on the central regulation of food intake; however, it caused anxiety-like behavior.
Collapse
Affiliation(s)
- Veronika Strnadová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic
| | - Alena Morgan
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic
| | - Magdalena Škrlová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eliška Haasová
- Institute of Physiology, CAS, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | | | - Aneta Myšková
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - David Sýkora
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic; Institute of Physiology, CAS, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, CAS, Prague, Czech Republic.
| |
Collapse
|
4
|
Borkar NA, Ambhore NS, Balraj P, Ramakrishnan YS, Sathish V. Kisspeptin regulates airway hyperresponsiveness and remodeling in a mouse model of asthma. J Pathol 2023; 260:339-352. [PMID: 37171283 PMCID: PMC10759912 DOI: 10.1002/path.6086] [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: 09/06/2022] [Revised: 03/06/2023] [Accepted: 03/31/2023] [Indexed: 05/13/2023]
Abstract
Asthma is a multifactorial disease of origin characterized by airway hyperresponsiveness (AHR) and airway remodeling. Several pieces of evidence from other pathologies suggest that Kisspeptins (Kp) regulate cell proliferation, migration, and invasion, mechanisms that are highly relevant to asthma. Our recent in vitro studies show Kp-10 (active peptide of Kp), via its receptor, KISS1R, inhibits human airway smooth muscle cell proliferation. Here, we hypothesize a crucial role for Kp-10 in regulating AHR and airway remodeling in vivo. Utilizing C57BL/6J mice, we assessed the effect of chronic intranasal Kp-10 exposure on mixed allergen (MA)-induced mouse model of asthma. MA-challenged mice showed significant deterioration of lung function compared to those exposed to vehicle (DPBS); Kp-10 treatment significantly improved the MA-altered lung functions. Mice treated with Kp-10 alone did not show any notable changes in lung functions. MA-exposed mice showed a significant reduction in KISS1R expression as compared to vehicle alone. MA-challenged mice showed significant alterations in immune cell infiltration in the airways and remodeling changes. Proinflammatory cytokines were significantly increased upon MA exposure, an effect abrogated by Kp-10 treatment. Furthermore, biochemical and histological studies showed Kp-10 exposure significantly reduced MA-induced smooth muscle mass and soluble collagen in the lung. Overall, our findings highlight the effect of chronic Kp-10 exposure in regulating MA-induced AHR and remodeling. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | | | | | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| |
Collapse
|
5
|
Wu Z, Jia Q, Liu B, Fang L, Leung PCK, Cheng JC. NPFF stimulates human ovarian cancer cell invasion by upregulating MMP-9 via ERK1/2 signaling. Exp Cell Res 2023; 430:113693. [PMID: 37392963 DOI: 10.1016/j.yexcr.2023.113693] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/17/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Neuropeptide FF (NPFF) belongs to the RFamide peptide family. NPFF regulates a variety of physiological functions by binding to a G protein-coupled receptor, NPFFR2. Epithelial ovarian cancer (EOC) is a leading cause of death among gynecological malignancies. The pathogenesis of EOC can be regulated by many local factors, including neuropeptides, through an autocrine/paracrine manner. However, to date, the expression and/or function of NPFF/NPFFR2 in EOC is undetermined. In this study, we show that the upregulation of NPFFR2 mRNA was associated with poor overall survival in EOC. The TaqMan probe-based RT-qPCR showed that NPFF and NPFFR2 were expressed in three human EOC cells, CaOV3, OVCAR3, and SKOV3. In comparison, NPFF and NPFFR2 expression levels were higher in SKOV3 cells than in CaOV3 or OVCAR3 cells. Treatment of SKOV3 cells with NPFF did not affect cell viability and proliferation but stimulated cell invasion. NPFF treatment upregulates matrix metalloproteinase-9 (MMP-9) expression. Using the siRNA-mediated knockdown approach, we showed that the stimulatory effect of NPFF on MMP-9 expression was mediated by the NPFFR2. Our results also showed that ERK1/2 signaling was activated in SKOV3 cells in response to the NPFF treatment. In addition, blocking the activation of ERK1/2 signaling abolished the NPFF-induced MMP-9 expression and cell invasion. This study provides evidence that NPFF stimulates EOC cell invasion by upregulating MMP-9 expression through the NPFFR2-mediated ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Ze Wu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiongqiong Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Boqun Liu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
6
|
Jiang H, Wang L, Zhu J, Ping Z. NPFFR2 gene compound heterozygous variants associated with preeclampsia identified by whole-exome sequencing. Gene 2023; 854:147108. [PMID: 36535464 DOI: 10.1016/j.gene.2022.147108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Preeclampsia (PE) is an idiopathic disorder of pregnancy. The exact cause of PE remains unknown. Emerging evidence indicates that the cause of PE is linked to genetic factors. Therefore, the aim of this study was to identify the susceptibility genes for PE. METHODS Nine families with severe PE were recruited. The whole-exome sequencing (WES) was performed on each family, and Sanger sequencing was used to identify the potential pathogenic genetic variants. RESULTS After a rigorous bioinformatics analysis, compound heterozygous variants in the NPFFR2 gene, NM_004885.2: c.601A > G, p.Met201Val and c.995C > T, p.Ala332Val were found in the No.4 pedigree. Bioinformatics analysis showed that these sites were highly conserved among several species and were predicted to be pathogenic variants according to multiple online mutational function prediction software packages. Due to the compound heterozygous variants of NPFFR2, more bonds are generated between mutant amino acids and spatial adjacent amino acids, which may lead to more stable active conformation of protein and not easy to be degraded. CONCLUSIONS We demonstrated for the first time that compound heterozygous variants of the NPFFR2 gene might be potentially associated with severe PE, the results of this study provide clinicians and researchers with a better understanding of the molecular mechanisms underlying severe PE in pregnant women.
Collapse
Affiliation(s)
- Huling Jiang
- Department of Prenatal Diagnosis Center, Maternity and Child Health Care Affiliated Hospital, Jiaxing University, Jiaxing 314000, China
| | - Luming Wang
- Department of Prenatal Diagnosis Center, Maternity and Child Health Care Affiliated Hospital, Jiaxing University, Jiaxing 314000, China
| | - Jianjun Zhu
- Department of Prenatal Diagnosis Center, Maternity and Child Health Care Affiliated Hospital, Jiaxing University, Jiaxing 314000, China.
| | - Zepeng Ping
- Department of Prenatal Diagnosis Center, Maternity and Child Health Care Affiliated Hospital, Jiaxing University, Jiaxing 314000, China.
| |
Collapse
|
7
|
Koller J, Herzog H, Zhang L. The distribution of Neuropeptide FF and Neuropeptide VF in central and peripheral tissues and their role in energy homeostasis control. Neuropeptides 2021; 90:102198. [PMID: 34534716 DOI: 10.1016/j.npep.2021.102198] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 11/26/2022]
Abstract
Neuropeptide FF (NPFF) and Neuropeptide VF (NPVF) are part of the extended RFamide peptide family characterized by their common arginine (R) and amidated phenylalanine (F)-motif at the carboxyl terminus. Both peptides signal through their respective high affinity G-protein coupled receptors, NPFFR2 and NPFFR1, but also show binding affinity for the other receptor due to their sequence similarity. NPFF and NPVF are highly conserved throughout evolution and can be found across the whole animal kingdom. Both have been implicated in a variety of biological mechanisms, including nociception, locomotion, reproduction, and response to pain and stress. However, more recently a new major functional role in the control of energy homeostasis has been discovered. In this article we will summarise the current knowledge on the distribution of NPFF, NPVF, and their receptors in central and peripheral tissues, as well as how this relates to the regulation of food intake and energy balance, which will help to better understand their role in these processes and thus might help finding treatments for impaired energy homeostasis disorders, such as obesity or anorexia.
Collapse
Affiliation(s)
- Julia Koller
- Healthy Aging, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Sydney, NSW 2052, Australia
| | - Herbert Herzog
- Healthy Aging, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; School of Medical Sciences, UNSW Sydney, NSW, Australia; Faculty of Medicine, UNSW Sydney, NSW, Australia
| | - Lei Zhang
- Healthy Aging, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Sydney, NSW 2052, Australia.
| |
Collapse
|
8
|
Talmont F, Veneziano R, Dietrich G, Moulédous L, Mollereau C, Zajac JM. Pharmacological insight into the activation of the human neuropeptide FF2 receptor. Peptides 2020; 134:170406. [PMID: 32920044 DOI: 10.1016/j.peptides.2020.170406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/13/2020] [Accepted: 09/08/2020] [Indexed: 01/14/2023]
Abstract
The neuropeptide FF2 (NPFF2) receptor, predominantly expressed in the central nervous system, plays an important role in the modulation of sensory input and opioid analgesia, as well as in locomotion, feeding, intestinal motility, reward, and the control of obesity. The NPFF2 receptor belongs to the RFamide peptide receptor family and to the G protein coupled receptor (GPCR) super family, but contrary to many other class A GPCRs, no 3D structure has been solved. Thus, it is essential to perform mutagenesis to gain information on the fine functioning of the NPFF2 receptor. In this study, we examined the role of aspartic acid (D) from the "D/ERY/F" motif found in the second intracellular loop (ICL2) and the role of the C-terminal end of the receptor in ligand binding and signal transduction. We found that mutation D3.49A does not impair binding capacities but inhibits G protein activation as well as adenylyl cyclase regulation. Truncation of the C terminal part of the receptor has different effects depending on the position of truncation. When truncation was realized downstream of the putative acylation site, ligand binding and signal transduction capabilities were not lost, contrary to total deletion of the C terminus, which totally impairs the activity of the receptor.
Collapse
Affiliation(s)
- Franck Talmont
- CNRS, IPBS (Institut De Pharmacologie Et De Biologie Structurale) 205 Route De Narbonne, 31077 Toulouse, Cedex 4, France; Université Paul Sabatier Toulouse III, F-31300 Toulouse, France.
| | - Remi Veneziano
- CNRS, IPBS (Institut De Pharmacologie Et De Biologie Structurale) 205 Route De Narbonne, 31077 Toulouse, Cedex 4, France; Université Paul Sabatier Toulouse III, F-31300 Toulouse, France
| | - Gilles Dietrich
- INSERM IRSD (Institut De Recherche En Santé Digestive) U1220, CHU Purpan Place Du Docteur Baylac, CS 60039 31024, Toulouse Cedex 3, France
| | - Lionel Moulédous
- CNRS, IPBS (Institut De Pharmacologie Et De Biologie Structurale) 205 Route De Narbonne, 31077 Toulouse, Cedex 4, France; Université Paul Sabatier Toulouse III, F-31300 Toulouse, France
| | - Catherine Mollereau
- CNRS, IPBS (Institut De Pharmacologie Et De Biologie Structurale) 205 Route De Narbonne, 31077 Toulouse, Cedex 4, France; Université Paul Sabatier Toulouse III, F-31300 Toulouse, France
| | - Jean-Marie Zajac
- CNRS, IPBS (Institut De Pharmacologie Et De Biologie Structurale) 205 Route De Narbonne, 31077 Toulouse, Cedex 4, France; Université Paul Sabatier Toulouse III, F-31300 Toulouse, France
| |
Collapse
|
9
|
Nguyen T, Marusich J, Li JX, Zhang Y. Neuropeptide FF and Its Receptors: Therapeutic Applications and Ligand Development. J Med Chem 2020; 63:12387-12402. [PMID: 32673481 DOI: 10.1021/acs.jmedchem.0c00643] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The endogenous neuropeptide FF (NPFF) and its two cognate G protein-coupled receptors, Neuropeptide FF Receptors 1 and 2 (NPFFR1 and NPFFR2), represent a relatively new target system for many therapeutic applications including pain regulation, modulation of opioid side effects, drug reward, anxiety, cardiovascular conditions, and other peripheral effects. Since the cloning of NPFFR1 and NPFFR2 in 2000, significant progress has been made to understand their pharmacological roles and interactions with other receptor systems, notably the opioid receptors. A variety of NPFFR ligands with different mechanisms of action (agonists or antagonists) have been discovered although with limited subtype selectivities. Differential pharmacological effects have been observed for many of these NPFFR ligands, depending on assays/models employed and routes of administration. In this Perspective, we highlight the therapeutic potentials, current knowledge gaps, and latest updates of the development of peptidic and small molecule NPFFR ligands as tool compounds and therapeutic candidates.
Collapse
Affiliation(s)
- Thuy Nguyen
- Center for Drug Discovery, Research Triangle Institute, 3040 East Cornwallis Road, Research Triangle Park, North Carolina 27709, United States
| | - Julie Marusich
- Center for Drug Discovery, Research Triangle Institute, 3040 East Cornwallis Road, Research Triangle Park, North Carolina 27709, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14203, United States
| | - Yanan Zhang
- Center for Drug Discovery, Research Triangle Institute, 3040 East Cornwallis Road, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
10
|
Chen J, Huang S, Zhang J, Li J, Wang Y. Characterization of the neuropeptide FF (NPFF) gene in chickens: evidence for a single bioactive NPAF peptide encoded by the NPFF gene in birds. Domest Anim Endocrinol 2020; 72:106435. [PMID: 32247990 DOI: 10.1016/j.domaniend.2020.106435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 11/09/2019] [Accepted: 01/03/2020] [Indexed: 01/06/2023]
Abstract
The 2 structurally related peptides, neuropeptide FF (NPFF) and neuropeptide AF (NPAF), are encoded by the NPFF gene and have been identified as neuromodulators that regulate nociception and opiate-mediated analgesia via NPFF receptor (NPFFR2) in mammals. However, little is known about these 2 peptides in birds. In this study, we examined the structure, tissue expression profile, and functionality of NPAF and NPFF in chickens. Our results showed that: 1) unlike mammalian NPFF, NPFF from chicken and other avian species is predicted to produce a single bioactive NPAF peptide, whereas the putative avian NPFF peptide likely lacks activity due to the absence of functional RFamide motif at its C-terminus; 2) synthetic chicken (c-) NPAF can potently activate cNPFFR2 (and not cNPFFR1) expressed in HEK293 cells, as monitored by 3 cell-based luciferase reporter systems, indicating that cNPAF is a potent ligand for cNPFFR2, which activation could decrease intracellular cAMP levels and stimulate the MAPK/ERK signaling cascade; interestingly, gonadotropin-inhibitory hormone, a peptide sharing high structural similarity to NPAF, could specifically activate cNPFFR1 (but not cNPFFR2); 3) Quantitative real-time PCR revealed that cNPFF mRNA is widely expressed in chicken tissues with the highest level detected in the hypothalamus, whereas cNPFFR2 is expressed in all tissues examined with the highest level noted in the hypothalamus and anterior pituitary. Taken together, our data reveal that avian NPFF encodes a single bioactive NPAF peptide, which preferentially activates NPFFR2, and provides insights into potential structural and functional changes of NPFF-derived peptides during vertebrate evolution.
Collapse
Affiliation(s)
- J Chen
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - S Huang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - J Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - J Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China.
| | - Y Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
11
|
Zhu H, Peng B, Klausen C, Yi Y, Li Y, Xiong S, von Dadelszen P, Leung PCK. NPFF increases fusogenic proteins syncytin 1 and syncytin 2 via GCM1 in first trimester primary human cytotrophoblast cells. FASEB J 2020; 34:9419-9432. [PMID: 32501590 DOI: 10.1096/fj.201902978r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/25/2020] [Accepted: 05/04/2020] [Indexed: 01/22/2023]
Abstract
Neuropeptide FF (NPFF) is well-known for its roles in the central nervous system. Despite studies demonstrating that NPFF receptor 2 (NPFFR2) mRNA is highest in placenta, nothing is known about NPFF-NPFFR2 functions in placental development. Here, we investigated the effects of NPFF-NPFFR2 on expression of syncytial [human chorionic gonadotropin (hCG) β] and fusogenic [syncytin 1, syncytin 2, and glial cells missing 1 (GCM1)] genes in first trimester primary human cytotrophoblast cells. By analyzing two publicly available microarray data sets, we found that NPFF is consistently expressed throughout gestation whereas NPFFR2 increases in first trimester and is elevated in placenta samples from women with preeclampsia. Immunohistochemistry showed that NPFFR2, syncytin 1/2, and GCM1 each displayed unique patterns of expression among different trophoblast populations in first trimester placenta. Treatment of primary human cytotrophoblast cells with NPFF increased the mRNA and protein levels of hCG β, syncytin 1, syncytin 2, and GCM1; and knockdown of NPFFR2 abolished these effects. Interestingly, GCM1 mediated NPFF-induced upregulation of syncytin 1 and syncytin 2, but not hCG β, in primary human cytotrophoblasts. Our results demonstrate that NPFF acts via NPFFR2 to enhance production of hCG β and promote GCM1-dependent expression of syncytin 1 and 2 in human cytotrophoblasts.
Collapse
Affiliation(s)
- Hua Zhu
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Bo Peng
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yuyin Yi
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yan Li
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Siyuan Xiong
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | | | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Potent effect of KISS1-54 DNA vaccine compared with KISS1-10 DNA vaccine in inhibiting the fertility of female rats. Vaccine 2018; 36:6631-6639. [DOI: 10.1016/j.vaccine.2018.09.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/09/2018] [Accepted: 09/23/2018] [Indexed: 12/13/2022]
|
13
|
Abstract
Chronic, persistent itch is a devastating symptom that causes much suffering. In recent years, there has been great progress made in understanding the molecules, cells, and circuits underlying itch sensation. Once thought to be carried by pain-sensing neurons, itch is now believed to be capable of being transmitted by dedicated sensory labeled lines. Members of the Mas-related G protein-coupled receptor (Mrgpr) family demarcate an itch-specific labeled line in the peripheral nervous system. In the spinal cord, the expression of other proteins identifies additional populations of itch-dedicated sensory neurons. However, as evidence for labeled-line coding has mounted, studies promoting alternative itch-coding strategies have emerged, complicating our understanding of the neural basis of itch. In this review, we cover the molecules, cells, and circuits related to understanding the neural basis of itch, with a focus on the role of Mrgprs in mediating itch sensation.
Collapse
Affiliation(s)
- James Meixiong
- The Solomon H. Snyder Department of Neuroscience and the Center for Sensory Biology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA; ,
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience and the Center for Sensory Biology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA; , .,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
14
|
Fumagalli M, Lecca D, Coppolino GT, Parravicini C, Abbracchio MP. Pharmacological Properties and Biological Functions of the GPR17 Receptor, a Potential Target for Neuro-Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1051:169-192. [PMID: 28828731 DOI: 10.1007/5584_2017_92] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In 2006, cells heterologously expressing the "orphan" receptor GPR17 were shown to acquire responses to both uracil nucleotides and cysteinyl-leukotrienes, two families of signaling molecules accumulating in brain or heart as a result of hypoxic/traumatic injuries. In subsequent years, evidence of GPR17 key role in oligodendrogenesis and myelination has highlighted it as a "model receptor" for new therapies in demyelinating and neurodegenerative diseases. The apparently contrasting evidence in the literature about the role of GPR17 in promoting or inhibiting myelination can be due to its transient expression in the intermediate stages of differentiation, exerting a pro-differentiating function in early oligodendrocyte precursor cells (OPCs), and an inhibitory role in late stage maturing cells. Meanwhile, several papers extended the initial data on GPR17 pharmacology, highlighting a "promiscuous" behavior of this receptor; indeed, GPR17 is able to respond to other emergency signals like oxysterols or the pro-inflammatory cytokine SDF-1, underlying GPR17 ability to adapt its responses to changes of the surrounding extracellular milieu, including damage conditions. Here, we analyze the available literature on GPR17, in an attempt to summarize its emerging biological roles and pharmacological properties.
Collapse
Affiliation(s)
- Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Giusy T Coppolino
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Chiara Parravicini
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
15
|
d'Anglemont de Tassigny X, Jayasena C, Murphy KG, Dhillo WS, Colledge WH. Mechanistic insights into the more potent effect of KP-54 compared to KP-10 in vivo. PLoS One 2017; 12:e0176821. [PMID: 28464043 PMCID: PMC5413024 DOI: 10.1371/journal.pone.0176821] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Kisspeptins regulate the mammalian reproductive axis by stimulating release of gonadotrophin releasing hormone (GnRH). Different length kisspeptins (KP) are found of 54, 14, 13 or 10 amino-acids which share a common C-terminal 10-amino acid sequence. KP-54 and KP-10 have been widely used to stimulate the reproductive axis but data suggest that KP-54 and KP-10 are not equally effective at eliciting reproductive hormone secretion after peripheral delivery. To confirm this, we analysed the effect of systemic administration of KP-54 or KP-10 on luteinizing hormone (LH) secretion into the bloodstream of male mice. Plasma LH measurements 10 min or 2 hours after kisspeptin injection showed that KP-54 can sustain LH release far longer than KP-10, suggesting a differential mode of action of the two peptides. To investigate the mechanism for this, we evaluated the pharmacokinetics of the two peptides in vivo and their potential to cross the blood brain barrier (BBB). We found that KP-54 has a half-life of ~32 min in the bloodstream, while KP-10 has a half-life of ~4 min. To compensate for this difference in half-life, we repeated injections of KP-10 every 10 min over 1 hr but failed to reproduce the sustained rise in LH observed after a single KP-54 injection, suggesting that the failure of KP-10 to sustain LH release may not just be related to peptide clearance. We tested the ability of peripherally administered KP-54 and KP-10 to activate c-FOS in GnRH neurons behind the blood brain barrier (BBB) and found that only KP-54 could do this. These data are consistent with KP-54 being able to cross the BBB and suggest that KP10 may be less able to do so.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Channa Jayasena
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - Kevin G. Murphy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - Waljit S. Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - William H. Colledge
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
16
|
Yu HP, Zhang N, Zhang T, Wang ZL, Li N, Tang HH, Zhang R, Zhang MN, Xu B, Fang Q, Wang R. Activation of NPFF 2 receptor stimulates neurite outgrowth in Neuro 2A cells through activation of ERK signaling pathway. Peptides 2016; 86:24-32. [PMID: 27669639 DOI: 10.1016/j.peptides.2016.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 10/21/2022]
Abstract
Neurite outgrowth is an important process in neural regeneration and plasticity, especially after neural injury, and recent evidence indicates that several Gαi/o protein-coupled receptors play an important role in neurite outgrowth. The neuropeptide (NP)FF system contains two Gαi/o protein-coupled receptors, NPFF1 and NPFF2 receptors, which are mainly distributed in the central nervous system. The aim of the present study was to determine whether the NPFF system is involved in neurite outgrowth in Neuro 2A cells. We showed that Neuro 2A cells endogenously expressed NPFF2 receptor, and the NPFF2 receptor agonist dNPA inhibited cyclic adenosine monophosphate (cAMP) production stimulated by forskolin in Neuro 2A cells. We also demonstrated that NPFF and dNPA dose-dependently induced neurite outgrowth in Neuro 2A cells, which was completely abolished by the NPFF receptor antagonist RF9. Pretreatment with mitogen-activated protein kinase inhibitors PD98059 and U0126 decreased dNPA-induced neurite outgrowth. In addition, dNPA increased phosphorylation of extracellular signal-regulated kinase (ERK) in Neuro 2A cells, which was completely antagonized by pretreatment with U0126. Our results suggest that activation of NPFF2 receptor stimulates neurite outgrowth in Neuro 2A cells through activation of the ERK signaling pathway. Moreover, NPFF2 receptor may be a potential therapeutic target for neural injury and degeneration in the future.
Collapse
Affiliation(s)
- Hong-Ping Yu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Nan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Ting Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Zi-Long Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Hong-Hai Tang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Meng-Na Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| |
Collapse
|
17
|
Kim JS. What's in a Name? Roles of RFamide-Related Peptides Beyond Gonadotrophin Inhibition. J Neuroendocrinol 2016; 28. [PMID: 27369805 DOI: 10.1111/jne.12407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 12/29/2022]
Abstract
RFamide-related peptides (RFRPs) have been heavily implicated in the control of reproductive function subsequent to their discovery more than 16 years ago. However, recent studies using genetic and pharmacological tools have challenged their importance in regulating the hypothalamic-pituitary-gonadal axis. It is generally accepted that RFRPs act as part of a wider RFamide system, which involves two receptors, called the neuropeptide FF receptors (NPFFR1 and R2), and includes the closely-related neuropeptide NPFF. NPFF has been studied ever since the 1980s and many of the functions of NPFF are also shared by RFRPs. The current review questions whether these functions of NPFF are indeed specific to just NPFF alone and presents evidence from both neuroendocrine and pharmacological perspectives. Furthermore, recently emerging new functions of RFRPs are discussed with the overall goal of clarifying the functions of RFRPs beyond the hypothalamic-pituitary-gonadal axis.
Collapse
Affiliation(s)
- J S Kim
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Buffel I, Meurs A, Portelli J, Raedt R, De Herdt V, Poppe L, De Meulenaere V, Wadman W, Bihel F, Schmitt M, Vonck K, Bourguignon JJ, Simonin F, Smolders I, Boon P. The effect of neuropeptide FF in the amygdala kindling model. Acta Neurol Scand 2016; 134:181-8. [PMID: 26503695 DOI: 10.1111/ane.12526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Neuropeptide FF (NPFF) and its receptors (NPFF1 R and NPFF2 R) are differentially distributed throughout the central nervous system. NPFF reduces cortical excitability in rats when administered intracerebroventricularly (i.c.v.), and both NPFF and NPFF1 R antagonists attenuate pilocarpine-induced limbic seizures. In this study, our aim was to determine whether NPFF exerts anticonvulsant or anti-epileptogenic effects in the rat amygdala kindling model for temporal lobe seizures. METHODS Male Wistar rats were implanted with a recording/stimulation electrode in the right amygdala and a cannula in the left lateral ventricle. In a first group of animals, the afterdischarge threshold (ADT) was determined after a single i.c.v. infusion of saline (n = 8) or NPFF (1 nmol/h for 2 h; n = 10). Subsequently, daily infusion of saline (n = 8) or NPFF (1 nmol/h for 2 h; i.c.v.; n = 9) was performed, followed by a kindling stimulus (ADT+200 μA). Afterdischarge duration and seizure severity were evaluated after every kindling stimulus. A second group of rats (n = 7) were fully kindled, and the effect of saline or a high dose of NPFF (10 nmol/h for 2 h, i.c.v.) on ADT and the generalized seizure threshold (GST) was subsequently determined. RESULTS In naive rats, NPFF significantly increased the ADT compared to control (435 ± 72 μA vs 131 ± 23 μA [P < 0.05]). When rats underwent daily stimulations above the ADT, NPFF did not delay or prevent kindling acquisition. Furthermore, a high dose of NPFF did not alter ADT or GST in fully kindled rats. CONCLUSIONS I.c.v. administration of NPFF reduced excitability in the amygdala in naive, but not in fully kindled rats, and had no effect on kindling acquisition.
Collapse
Affiliation(s)
- I. Buffel
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - A. Meurs
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - J. Portelli
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
- Center for Neurosciences; Department of Pharmaceutical Chemistry; Drug Analysis & Drug information; University of Brussels; Brussels Belgium
| | - R. Raedt
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - V. De Herdt
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - L. Poppe
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - V. De Meulenaere
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - W. Wadman
- Swammerdam Institute of Life Sciences; Department of Neurobiology; University of Amsterdam; Amsterdam The Netherlands
| | - F. Bihel
- Therapeuthic Innovation Laboratory; Faculty of Pharmacy; UMR 7200; CNRS; University of Strasbourg; Illkirch Graffenstaden France
| | - M. Schmitt
- Therapeuthic Innovation Laboratory; Faculty of Pharmacy; UMR 7200; CNRS; University of Strasbourg; Illkirch Graffenstaden France
| | - K. Vonck
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| | - J.-J. Bourguignon
- Therapeuthic Innovation Laboratory; Faculty of Pharmacy; UMR 7200; CNRS; University of Strasbourg; Illkirch Graffenstaden France
| | - F. Simonin
- Research Institute of ESBS; CNRS; UMR7242; University of Strasbourg; Illkirch France
| | - I. Smolders
- Center for Neurosciences; Department of Pharmaceutical Chemistry; Drug Analysis & Drug information; University of Brussels; Brussels Belgium
| | - P. Boon
- Laboratory for Clinical and Experimental Neurophysiology; Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent University Hospital; Ghent Belgium
| |
Collapse
|
19
|
Quillet R, Ayachi S, Bihel F, Elhabazi K, Ilien B, Simonin F. RF-amide neuropeptides and their receptors in Mammals: Pharmacological properties, drug development and main physiological functions. Pharmacol Ther 2016; 160:84-132. [PMID: 26896564 DOI: 10.1016/j.pharmthera.2016.02.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RF-amide neuropeptides, with their typical Arg-Phe-NH2 signature at their carboxyl C-termini, belong to a lineage of peptides that spans almost the entire life tree. Throughout evolution, RF-amide peptides and their receptors preserved fundamental roles in reproduction and feeding, both in Vertebrates and Invertebrates. The scope of this review is to summarize the current knowledge on the RF-amide systems in Mammals from historical aspects to therapeutic opportunities. Taking advantage of the most recent findings in the field, special focus will be given on molecular and pharmacological properties of RF-amide peptides and their receptors as well as on their implication in the control of different physiological functions including feeding, reproduction and pain. Recent progress on the development of drugs that target RF-amide receptors will also be addressed.
Collapse
Affiliation(s)
- Raphaëlle Quillet
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Safia Ayachi
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Frédéric Bihel
- Laboratoire Innovation Thérapeutique, UMR 7200 CNRS, Université de Strasbourg, Illkirch, France
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Brigitte Ilien
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
20
|
Buffel I, Meurs A, Portelli J, Raedt R, De Herdt V, Sioncke L, Wadman W, Bihel F, Schmitt M, Vonck K, Bourguignon JJ, Simonin F, Smolders I, Boon P. Neuropeptide FF and prolactin-releasing peptide decrease cortical excitability through activation of NPFF receptors. Epilepsia 2015; 56:489-98. [PMID: 25684325 DOI: 10.1111/epi.12928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Drugs with a novel mechanism of action are needed to reduce the number of people with epilepsy that are refractory to treatment. Increasing attention is paid to neuropeptide systems and several anticonvulsant neuropeptides have already been described, such as galanin, ghrelin, and neuropeptide Y (NPY). Many others, however, have not been investigated for their ability to affect epileptic seizures. In this study, the potential anticonvulsant activities of three members of the RF-amide neuropeptide family, neuropeptide FF (NPFF), prolactin-releasing peptide (PrRP), and kisspeptin (Kp) and other receptor ligands (NPFF1/2 R, GPR10, and GRP54, respectively) were tested in the motor cortex stimulation model. METHODS A train of pulses with increasing intensity (0-10 mA over 150 s, 50 Hz, pulse width 2 msec) was delivered to the motor cortex of rats. The threshold intensity for eliciting a motor response (i.e., motor threshold) was determined through behavioral observation and used as a measure for cortical excitability. The threshold was determined before, during, and after the intracerebroventricular (i.c.v.) administration of various NPFF1/2 R, GPR10, and GPR54 receptor ligands. RESULTS NPFF and PrRP significantly increased the motor threshold by a maximum of 143 ± 27 and 83 ± 13 μA, respectively, for the doses of 1 nmol/h (p < 0.05). The increase of motor threshold by NPFF and PrRP was prevented by pretreatment and co-treatment with the NPFF1/2 R antagonist RF9. Pretreatment with a selective NPFF1 R antagonist also prevented the threshold increase induced by NPFF. Kp did not increase motor threshold. SIGNIFICANCE Intracerebroventricular infusion of NPFF or PrRP decreases cortical excitability in rats through activation of NPFFRs. Furthermore, the NPFF1 R is required for the NPFF-induced decrease in cortical excitability.
Collapse
Affiliation(s)
- Ine Buffel
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology, Department of Neurology, Ghent University Hospital, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Sun Y, Chen X, Chen Z, Ma X, Li D, Shang P, Qian A. Neuropeptide FF attenuates RANKL-induced differentiation of macrophage-like cells into osteoclast-like cells. Arch Oral Biol 2015; 60:282-92. [DOI: 10.1016/j.archoralbio.2014.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/28/2014] [Accepted: 11/08/2014] [Indexed: 01/31/2023]
|
22
|
Herreboudt AM, Kyle VRL, Lawrence J, Doran J, Colledge WH. Kiss1 mutant placentas show normal structure and function in the mouse. Placenta 2014; 36:52-8. [PMID: 25468546 PMCID: PMC4302219 DOI: 10.1016/j.placenta.2014.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/29/2014] [Accepted: 10/31/2014] [Indexed: 01/18/2023]
Abstract
Introduction Kisspeptins, encoded by the Kiss1 gene, are a set of related neuropeptides that are required for activation of the mammalian reproductive axis at puberty and to maintain fertility. In addition, kisspeptin signaling via the G-protein coupled receptor GPR54 (KISS1R) has been suggested to regulate human placental formation and correlations have been found between altered kisspeptin levels in the maternal blood and the development of pre-eclampsia. Methods We have used Kiss1 and Gpr54 mutant mice to investigate the role of kisspeptin signaling in the structure and function of the mouse placenta. Results Expression of Kiss1 and Gpr54 was confirmed in the mouse placenta but no differences in birth weight were found in mice that had been supported by a mutant placenta during fetal development. Stereological measurements found no differences between Kiss1 mutant and wild-type placentas. Measurement of amino-acid and glucose transport across the Kiss1 mutant placentas at E15.5 days did not reveal any functional defects. Discussion These data indicate that mouse placentas can develop a normal structure and function without kisspeptin signaling and can support normal fetal development and growth. The structure and function of Kiss1 and Gpr54 mutant placentas was examined in transgenic mice. Kiss1 and Gpr54 mutant placentas are not associated with fetal loss during gestation or reduced pup weight at birth. Kiss1 mutant placentas show normal structural components. Kiss1 mutant placentas show normal transport of glucose and amino-acids.
Collapse
Affiliation(s)
- A M Herreboudt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - V R L Kyle
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - J Lawrence
- Takeda Cambridge, 418 Science Park, Milton Road, Cambridge CB4 0PZ, United Kingdom
| | - J Doran
- Takeda Cambridge, 418 Science Park, Milton Road, Cambridge CB4 0PZ, United Kingdom
| | - W H Colledge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom.
| |
Collapse
|
23
|
Bray L, Froment C, Pardo P, Candotto C, Burlet-Schiltz O, Zajac JM, Mollereau C, Moulédous L. Identification and functional characterization of the phosphorylation sites of the neuropeptide FF2 receptor. J Biol Chem 2014; 289:33754-66. [PMID: 25326382 DOI: 10.1074/jbc.m114.612614] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The neuropeptide FF2 (NPFF2) receptor belongs to the rhodopsin family of G protein-coupled receptors and mediates the effects of several related RFamide neuropeptides. One of the main pharmacological interests of this system resides in its ability to regulate endogenous opioid systems, making it a potential target to reduce the negative effects of chronic opioid use. Phosphorylation of intracellular residues is the most extensively studied post-translational modification regulating G protein-coupled receptor activity. However, until now, no information concerning NPFF2 receptor phosphorylation is available. In this study, we combined mass spectrometric analysis and site-directed mutagenesis to analyze for the first time the phosphorylation pattern of the NPFF2 receptor and the role of the various phosphorylation sites in receptor signaling, desensitization, and trafficking in a SH-SY5Y model cell line. We identified the major, likely GRK-dependent, phosphorylation cluster responsible for acute desensitization, (412)TNST(415) at the end of the C terminus of the receptor, and additional sites involved in desensitization ((372)TS(373)) and internalization (Ser(395)). We thus demonstrate the key role played by phosphorylation in the regulation of NPFF2 receptor activity and trafficking. Our data also provide additional evidence supporting the concept that desensitization and internalization are partially independent processes relying on distinct phosphorylation patterns.
Collapse
Affiliation(s)
- Lauriane Bray
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Carine Froment
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Pierre Pardo
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Cédric Candotto
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Odile Burlet-Schiltz
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Jean-Marie Zajac
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Catherine Mollereau
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| | - Lionel Moulédous
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS, Université de Toulouse, 31077 Toulouse, France
| |
Collapse
|
24
|
A high-affinity, radioiodinatable neuropeptide FF analogue incorporating a photolabile p-(4-hydroxybenzoyl)phenylalanine. Anal Biochem 2014; 453:50-4. [PMID: 24613258 DOI: 10.1016/j.ab.2014.02.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 11/23/2022]
Abstract
A new radioiodinated photoaffinity compound, [(125)I]YE(Bpa)WSLAAPQRFNH2, derived from a peptide present in the rat neuropeptide FF (NPFF) precursor was synthesized, and its binding characteristics were investigated on a neuroblastoma clone, SH-SY5Y, stably expressing rat NPFF2 receptors tagged with the T7 epitope. The binding of the probe was saturable and revealed a high-affinity interaction (KD=0.24nM) with a single class of binding sites. It was also able to affinity label NPFF2 receptor in a specific and efficient manner given that 38% of the bound radioligand at saturating concentration formed a wash-resistant binding after ultraviolet (UV) irradiation. Photoaffinity labeling with [(125)I]YE(Bpa)WSLAAPQRFamide showed two molecular forms of NPFF2 receptor with apparent molecular weights of 140 and 95kDa in a 2:1 ratio. The comparison of the results between photoaffinity labeling and Western blot analysis suggests that all receptor forms bind the probe irreversibly with the same efficiency. On membranes of mouse olfactory bulb, only the high molecular weight form of NPFF2 receptor is observed. [(125)I]YE(Bpa)WSLAAPQRFamide is an excellent radioiodinated peptidic ligand for direct and selective labeling of NPFF2 receptors in vitro.
Collapse
|
25
|
Ayachi S, Simonin F. Involvement of Mammalian RF-Amide Peptides and Their Receptors in the Modulation of Nociception in Rodents. Front Endocrinol (Lausanne) 2014; 5:158. [PMID: 25324831 PMCID: PMC4183120 DOI: 10.3389/fendo.2014.00158] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/17/2014] [Indexed: 01/04/2023] Open
Abstract
Mammalian RF-amide peptides, which all share a conserved carboxyl-terminal Arg-Phe-NH2 sequence, constitute a family of five groups of neuropeptides that are encoded by five different genes. They act through five G-protein-coupled receptors and each group of peptide binds to and activates mostly one receptor: RF-amide related peptide group binds to NPFFR1, neuropeptide FF group to NPFFR2, pyroglutamylated RF-amide peptide group to QRFPR, prolactin-releasing peptide group to prolactin-releasing peptide receptor, and kisspeptin group to Kiss1R. These peptides and their receptors have been involved in the modulation of several functions including reproduction, feeding, and cardiovascular regulation. Data from the literature now provide emerging evidence that all RF-amide peptides and their receptors are also involved in the modulation of nociception. This review will present the current knowledge on the involvement in rodents of the different mammalian RF-amide peptides and their receptors in the modulation of nociception in basal and chronic pain conditions as well as their modulatory effects on the analgesic effects of opiates.
Collapse
Affiliation(s)
- Safia Ayachi
- UMR 7242 CNRS, Laboratory of Excellence Medalis, Biotechnologie et Signalisation Cellulaire, Université de Strasbourg, Illkirch, France
| | - Frédéric Simonin
- UMR 7242 CNRS, Laboratory of Excellence Medalis, Biotechnologie et Signalisation Cellulaire, Université de Strasbourg, Illkirch, France
- *Correspondence: Frédéric Simonin, UMR 7242 CNRS, Laboratory of Excellence Medalis, Biotechnologie et Signalisation Cellulaire, Université de Strasbourg, 300 Boulevard Sébastien Brant, Illkirch 67412, France e-mail:
| |
Collapse
|
26
|
Sun YL, Zhang XY, Sun T, He N, Li JY, Zhuang Y, Zeng Q, Yu J, Fang Q, Wang R. The anti-inflammatory potential of neuropeptide FF in vitro and in vivo. Peptides 2013; 47:124-32. [PMID: 23856454 DOI: 10.1016/j.peptides.2013.07.003] [Citation(s) in RCA: 20] [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: 05/23/2013] [Revised: 07/03/2013] [Accepted: 07/03/2013] [Indexed: 01/22/2023]
Abstract
Neuropeptide FF (NPFF) has many functions in regulating various biological processes. However, little attention has been focused on the anti-inflammatory effect of this peptide. In the present study, the in vitro anti-inflammatory activity of NPFF in both primary peritoneal macrophages and RAW 264.7 macrophages was investigated. Our data showed that NPFF suppressed the nitric oxide (NO) production of macrophages in the inflammation process. RF9, a reported antagonist of NPFF receptors, completely blocked the NPFF-induced NO suppression, suggesting a NPFF receptors-mediated pathway is mainly involved. Down-regulation of the nitric oxide synthases significantly inhibited the NPFF-induced NO reduction, indicating the involvement of nitric oxide synthases. However, the nitric oxide synthases were not the only route by which NPFF modulated the NO levels of macrophages. Pharmacological antagonists of the NF-κB signal pathway also completely suppressed the NPFF-induced NO decline. Moreover, we also observed that NPFF is capable of blocking the LPS-induced nuclear translocation of p65 in macrophages, implying the involvement of the NF-κB signal pathway. Finally, we observed that NPFF markedly attenuated the carrageenan-induced mouse paw edema, indicating that NPFF is capable of exerting anti-inflammatory potency in vivo. Collectively, our findings reveal the potential role of NPFF in the anti-inflammatory field both in vitro and in vivo, which will be helpful for the further exploitation of NPFF utility therapeutically.
Collapse
Affiliation(s)
- Yu-Long Sun
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology & Psychology, School of Basic Medical Sciences, Lanzhou University, 222 Tian Shui South Road, Lanzhou, Gansu 730000, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Jhamandas JH, Goncharuk V. Role of neuropeptide FF in central cardiovascular and neuroendocrine regulation. Front Endocrinol (Lausanne) 2013; 4:8. [PMID: 23404625 PMCID: PMC3566396 DOI: 10.3389/fendo.2013.00008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 01/22/2013] [Indexed: 11/29/2022] Open
Abstract
Neuropeptide FF (NPFF) is an octapeptide belonging to the RFamide family of peptides that have been implicated in a wide variety of physiological functions in the brain including central cardiovascular and neuroendocrine regulation. The effects of these peptides are mediated via NPFF1 and NPFF2 receptors that are abundantly expressed in the rat and human brain. Herein, we review evidence for the role of NPFF in central regulation of blood pressure particularly within the brainstem and the hypothalamic paraventricular nucleus (PVN). At a cellular level, NPFF demonstrates distinct responses in magnocellular and parvocellular neurons of the PVN, which regulate the secretion of neurohypophyseal hormones and sympathetic outflow, respectively. Finally, the presence of NPFF system in the human brain and its alterations within the hypertensive brain are discussed.
Collapse
Affiliation(s)
- Jack H. Jhamandas
- Division of Neurology, Department of Medicine, Centre for Neuroscience, University of AlbertaEdmonton, AB, Canada
| | - Valeri Goncharuk
- Division of Neurology, Department of Medicine, Centre for Neuroscience, University of AlbertaEdmonton, AB, Canada
- Russian Cardiology Research CenterMoscow, Russia
| |
Collapse
|
28
|
Sun YL, Zhang XY, He N, Sun T, Zhuang Y, Fang Q, Wang KR, Wang R. Neuropeptide FF activates ERK and NF kappa B signal pathways in differentiated SH-SY5Y cells. Peptides 2012; 38:110-7. [PMID: 22981806 DOI: 10.1016/j.peptides.2012.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 08/30/2012] [Accepted: 08/30/2012] [Indexed: 12/27/2022]
Abstract
Neuropeptide FF (NPFF) has been reported to play important roles in regulating diverse biological processes. However, little attention has been focused on the downstream signal transduction pathway of NPFF. Here, we used the differentiated neuroblastoma cell line, dSH-SY5Y, which endogenously expresses hNPFF2 receptor, to investigate the signal transduction downstream of NPFF. In particular we investigated the regulation of the extracellular signal-regulated protein kinase (ERK) and the nuclear factor kappa B (NF-κB) pathways by NPFF in these cells. NPFF rapidly and transiently stimulated ERK. H89, a selective inhibitor of cyclic AMP-dependent protein kinase A (PKA), inhibited the NPFF-activated ERK pathway, indicating the involvement of PKA in the NPFF-induced ERK activation. Down-regulation of nitric oxide synthases also attenuated NPFF-induced ERK activation, suggesting that a nitric oxide synthase-dependent pathway is involved. Moreover, the core upstream components of the NF-κB pathway were also significantly activated in response to NPFF, suggesting that the NF-κB pathway is involved in the signal transduction pathway of NPFF. Collectively, these data demonstrate that nitric oxide synthases are involved in the signal transduction pathway of NPFF, and provide the first evidence for the interaction between NPFF and the NF-κB pathway. These advances in our interpretation of the NPFF pathway mechanism will aid the comprehensive understanding of its function and provide novel molecular insight for further study of the NPFF system.
Collapse
Affiliation(s)
- Yu-long Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
In Vitro Activities of Kissorphin, a Novel Hexapeptide KiSS-1 Derivative, in Neuronal Cells. JOURNAL OF AMINO ACIDS 2012; 2012:691463. [PMID: 22848794 PMCID: PMC3400367 DOI: 10.1155/2012/691463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Accepted: 06/11/2012] [Indexed: 01/14/2023]
Abstract
The primary products of the metastasis-suppressor KiSS-1 gene are the kisspeptin (KP) peptides that stimulate gonadotrophin-releasing-hormone (GnRH) release via GPR-54 receptor activation. Recent studies have suggested that the KP-10 peptide also activates neuropeptide FF (NPFF) receptors. The aim of the current study was to determine the activities of the KiSS-1 derivative kissorphin (KSO), which contains the first six amino acids of the KP-10 peptide, is C-terminally amidated, and shares amino acid similarities with the biologically active NPFF 3–8 sequence. The KSO peptide inhibited forskolin-stimulated cyclic adenosine monophosphate (cAMP) production in ND7/23 neuroblastoma cells via an action that could be inhibited by the NPFF receptor antagonist RF9. Release of GnRH by LA-N-1 neuroblastoma cells was not altered by the KSO peptide. In ND7/23 neuroblastoma cells, the KSO peptide was able to reduce forskolin neuroprotection against H2O2 toxicity. The KSO peptide was also able to prevent prostaglandin E2-induced apoptosis in rat cortical neurons. The NPFF receptor antagonist RF9 could inhibit these actions of the KSO peptide in oxidative stress and apoptosis models. In conclusion, the kissorphin peptide, comprising the amino acid sequence Tyr-Asn-Trp-Asn-Ser-Phe-NH2, has NPFF-like biological activity without showing any GnRH releasing activity and inhibits forskolin-activated cAMP release.
Collapse
|
30
|
Mollereau C, Roumy M, Zajac JM. Neuropeptide FF receptor modulates potassium currents in a dorsal root ganglion cell line. Pharmacol Rep 2012; 63:1061-5. [PMID: 22001995 DOI: 10.1016/s1734-1140(11)70623-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 03/24/2011] [Indexed: 11/26/2022]
Abstract
This study investigated the presence of neuropeptide FF (NPFF) receptors on F-11 cells, a hybridoma derived from rat dorsal root ganglia (DRG) and mouse neuroblastoma. Binding experiments revealed a low density (4 fmol/mg) of high affinity (0.5 nM) [(3)H]-EYF binding sites in these cells. The whole-cell planar patch-clamp technique showed that dNPA, a selective NPFF(2) agonist, increased the voltage-dependent potassium outward currents (about 30 pA/pF) by 21%; this reversible effect on sustained delayed potassium currents is blocked by tetraethylammonium. The similar effects of NPFF and opioid agonists on K(+) currents in this cell line may explain their similar antinociceptive actions at the spinal level.
Collapse
Affiliation(s)
- Catherine Mollereau
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 route de Narbonne, 31077 Toulouse Cedex, France
| | | | | |
Collapse
|
31
|
Shahjahan M, Ikegami T, Osugi T, Ukena K, Doi H, Hattori A, Tsutsui K, Ando H. Synchronised expressions of LPXRFamide peptide and its receptor genes: seasonal, diurnal and circadian changes during spawning period in grass puffer. J Neuroendocrinol 2011; 23:39-51. [PMID: 21083774 DOI: 10.1111/j.1365-2826.2010.02081.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Among the RFamide peptide family, the LPXRFamide peptide (LPXRFa) group regulates the release of various pituitary hormones and, recently, LPXRFa genes were found to be regulated by photoperiod via melatonin. As a first step towards investigating the role of LPXRFa on reproductive function in grass puffer (Takifugu niphobles), which spawns in semilunar cycles, genes encoding LPXRFa and its receptor (LPXRFa-R) were cloned, and seasonal, diurnal and circadian changes in their absolute amounts of mRNAs in the brain and pituitary were examined by quantitative real-time polymerase chain reaction. The grass puffer LPXRFa precursor contains two putative RFamide peptides and one possible RYamide peptide. LPXRFa and LPXRFa-R genes were extensively expressed in the diencephalon and pituitary. The expression levels of both genes were significantly elevated during the spawning periods in both sexes in the brain and pituitary, although they were low in the spawning fish just after releasing eggs and sperm. The treatment of primary pituitary cultures with goldfish LPXRFa increased the amounts of follicle-stimulating hormone β- and luteinising hormone β-subunit mRNAs. In the diencephalon, LPXRFa and LPXRFa-R genes showed synchronised diurnal and circadian variations with one peak at zeitgeber time 3 and circadian time 15, respectively. The correlated expression patterns of LPXRFa and LPXRFa-R genes in the diencephalon and pituitary and the possible stimulatory effects of LPXRFa on gonadotrophin subunit gene expression suggest the functional significance of the LPXRFa and LPXRFa-R system in the regulation of lunar-synchronised spawning of grass puffer.
Collapse
Affiliation(s)
- M Shahjahan
- Laboratory of Advanced Animal and Marine Bioresources, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Moulédous L, Mollereau C, Zajac JM. Opioid-modulating properties of the neuropeptide FF system. Biofactors 2010; 36:423-9. [PMID: 20803521 DOI: 10.1002/biof.116] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 06/30/2010] [Indexed: 12/21/2022]
Abstract
Opioid receptors are involved in the control of pain perception in the central nervous system together with endogenous neuropeptides, termed opioid-modulating peptides, participating in a homeostatic system. Neuropeptide FF (NPFF) and related peptides possess anti-opioid properties, the cellular mechanisms of which are still unclear. The purpose of this review is to detail the phenomenon of cross-talk taking place between opioid and NPFF systems at the in vivo pharmacological level and to propose cellular and molecular models of functioning. A better knowledge of the mechanisms underlying opioid-modulating properties of NPFF has potential therapeutic interest for the control of opioid functions, notably for alleviating pain and/or for the treatment of opioid abuse.
Collapse
Affiliation(s)
- Lionel Moulédous
- Institut de Pharmacologie et de Biologie Structurale, CNRS UMR5089, Université de Toulouse, 205 route de Narbonne, Toulouse CEDEX 04, France
| | | | | |
Collapse
|
33
|
Moulédous L, Barthas F, Zajac JM. Opposite control of body temperature by NPFF1 and NPFF2 receptors in mice. Neuropeptides 2010; 44:453-6. [PMID: 20554321 DOI: 10.1016/j.npep.2010.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 04/23/2010] [Accepted: 05/10/2010] [Indexed: 11/23/2022]
Abstract
Neuropeptide FF (NPFF) is a neurotransmitter known to modulate opioid functions. This study investigates the effects of RF9, a new antagonist of NPFF receptors, on the roles of NPFF1 and NPFF2 receptors in thermoregulation in mice. RF9 (10 nmol) injected into the third ventricle did not modify the body temperature as compared to saline, but it completely antagonized the hypothermic effects of 10 nmol NPVF, a NPFF1 selective agonist, as well as the hyperthermic actions of dNPA (5 nmol), a NPFF2 selective agonist. The use of a specific antagonist demonstrates here that central NPFF1 and NPFF2 receptors control in an opposite manner the body temperature in mice.
Collapse
Affiliation(s)
- Lionel Moulédous
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | | | | |
Collapse
|
34
|
Calleri E, Ceruti S, Cristalli G, Martini C, Temporini C, Parravicini C, Volpini R, Daniele S, Caccialanza G, Lecca D, Lambertucci C, Trincavelli ML, Marucci G, Wainer IW, Ranghino G, Fantucci P, Abbracchio MP, Massolini G. Frontal affinity chromatography-mass spectrometry useful for characterization of new ligands for GPR17 receptor. J Med Chem 2010; 53:3489-501. [PMID: 20394377 DOI: 10.1021/jm901691y] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The application of frontal affinity chromatography-mass spectrometry (FAC-MS), along with molecular modeling studies, to the screening of potential drug candidates toward the recently deorphanized G-protein-coupled receptor (GPCR) GPR17 is shown. GPR17 is dually activated by uracil nucleotides and cysteinyl-leukotrienes, and is expressed in organs typically undergoing ischemic damage (i.e., brain, heart and kidney), thus representing a new pharmacological target for acute and chronic neurodegeneration. GPR17 was entrapped on an immobilized artificial membrane (IAM), and this stationary phase was used to screen a library of nucleotide derivatives by FAC-MS to select high affinity ligands. The chromatographic results have been validated with a reference functional assay ([(35)S]GTPgammaS binding assay). The receptor nucleotide-binding site was studied by setting up a column where a mutated GPR17 receptor (Arg255Ile) has been immobilized. The chromatographic behavior of the tested nucleotide derivatives together with in silico studies have been used to gain insights into the structure requirement of GPR17 ligands.
Collapse
Affiliation(s)
- Enrica Calleri
- Department of Pharmaceutical Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ozawa A, Lindberg I, Roth B, Kroeze WK. Deorphanization of novel peptides and their receptors. AAPS JOURNAL 2010; 12:378-84. [PMID: 20446073 DOI: 10.1208/s12248-010-9198-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 04/14/2010] [Indexed: 12/31/2022]
Abstract
Peptide hormones and neuropeptides play important roles in endocrine and neural signaling, often using G protein-coupled receptor (GPCR)-mediated signaling pathways. However, the rate of novel peptide discovery has slowed dramatically in recent years. Genomic sequencing efforts have yielded a large number of cDNA sequences that potentially encode novel candidate peptide precursors, as well as hundreds of orphan GPCRs with no known cognate ligands. The complexity of peptide signaling is further highlighted by the requirement for specific posttranslational processing steps, and these must be accomplished in vitro prior to testing newly discovered peptide precursor candidates in receptor assays. In this review, we present historic as well as current approaches to peptide discovery and GPCR deorphanization. We conclude that parallel and combinatorial discovery methods are likely to represent the most fruitful avenues for both peptide discovery as well as for matching the remaining GPCRs with their peptide ligands.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore, 20 Penn St. HSFII Rm S251, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
36
|
Lameh J, Bertozzi F, Kelly N, Jacobi PM, Nguyen D, Bajpai A, Gaubert G, Olsson R, Gardell LR. Neuropeptide FF receptors have opposing modulatory effects on nociception. J Pharmacol Exp Ther 2010; 334:244-54. [PMID: 20354177 DOI: 10.1124/jpet.109.164384] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of neuropeptide FF (NPFF) and its analogs in pain modulation is ambiguous. Although NPFF was first characterized as an antiopioid peptide, both antinociceptive and pronociceptive effects have been reported, depending on the route of administration. Currently, two NPFF receptors, termed FF1 and FF2, have been identified and cloned, but their roles in pain modulation remain elusive because of the lack of availability of selective compounds suitable for systemic administration in in vivo models. Ligand-binding studies confirm ubiquitous expression of both subtypes in brain, whereas only FF2 receptors are expressed spinally. This disparity in localization has served as the foundation of the hypothesis that FF1 receptors mediate the pronociceptive actions of NPFF. We have identified novel small molecule NPFF receptor agonists and antagonists with varying degrees of FF2/FF1 functional selectivity. Using these pharmacological tools in vivo has allowed us to define the roles of NPFF receptor subtypes as pertains to the modulation of nociception. We demonstrate that selective FF2 agonism does not modulate acute pain but instead ameliorates inflammatory and neuropathic pains. Treatment with a nonselective FF1/FF2 agonist potentiates allodynia in neuropathic rats and increases sensitivity to noxious thermal and to non-noxious mechanical stimuli in normal rats in an FF1 antagonist-reversible manner. Treatment with FF1 antagonists reversed established mechanical allodynia, indicating the possibility of increased NPFF tone through FF1 receptors. In conclusion, we provide evidence for the opposing roles of NPFF receptors and highlight selective FF2 agonism and/or selective FF1 antagonism as potential targets warranting further investigation.
Collapse
Affiliation(s)
- Jelveh Lameh
- ACADIA Pharmaceuticals, Inc., San Diego, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Moulédous L, Frances B, Zajac JM. Modulation of basal and morphine-induced neuronal activity by a NPFF2 selective agonist measured by c-Fos mapping of the mouse brain. Synapse 2010; 64:672-81. [DOI: 10.1002/syn.20774] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
38
|
Talmont F, Mollereau C, Muller I, Zajac JM. Solubilization and functional reconstitution of human neuropeptide FF2 receptors. Anal Biochem 2010; 398:225-9. [DOI: 10.1016/j.ab.2009.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 11/27/2009] [Accepted: 12/09/2009] [Indexed: 10/20/2022]
|
39
|
Talmont F, Moulédous L, Piedra-Garcia L, Schmitt M, Bihel F, Bourguignon JJ, Zajac JM, Mollereau C. Pharmacological characterization of the mouse NPFF2 receptor. Peptides 2010; 31:215-20. [PMID: 19944730 DOI: 10.1016/j.peptides.2009.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 11/05/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
This study presents the binding and functional properties of the mouse NPFF(2) (mNPFF(2)) receptor, in comparison with its human counterpart (hNPFF(2)). Binding experiments were performed by using the NPFF(2) selective radioligand [(3)H]-EYF in membranes from CHO cells transfected with mouse and human NPFF(2) receptors and compared to membranes from mouse olfactory bulb, the brain region expressing the highest density of NPFF(2) receptors in mouse. mNPFF(2) receptors exhibited a high affinity (Kd=0.2-0.4 nM) for [(3)H]-EYF, comparable to that of hNPFF(2) receptors. Also, the binding selectivity profile of mNPFF(2) receptors was comparable to that of hNPFF(2) receptors, except for three ligands (NPSF, NPVF, RF9) that were about tenfold more potent and active on mouse receptors than on human receptors. In particular, compared to hNPFF(2) receptors, mNPFF(2) receptors were less discriminative towards the proNPFF(B)-derived peptide. This suggests some species-related differences in the binding properties of NPFF(2) receptors that could have repercussion when evaluating the pharmacological properties of drugs in vivo.
Collapse
Affiliation(s)
- Franck Talmont
- CNRS/IPBS (Institut de Pharmacologie et Biologie Structurale), 205 route de Narbonne, 31077 Toulouse Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Gaubert G, Bertozzi F, Kelly NM, Pawlas J, Scully AL, Nash NR, Gardell LR, Lameh J, Olsson R. Discovery of Selective Nonpeptidergic Neuropeptide FF2 Receptor Agonists. J Med Chem 2009; 52:6511-4. [DOI: 10.1021/jm9011998] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gilles Gaubert
- ACADIA Pharmaceuticals AB, Medeon Science Park, S-205 12 Malmö, Sweden
| | - Fabio Bertozzi
- ACADIA Pharmaceuticals AB, Medeon Science Park, S-205 12 Malmö, Sweden
| | - Nicholas M. Kelly
- ACADIA Pharmaceuticals AB, Medeon Science Park, S-205 12 Malmö, Sweden
| | - Jan Pawlas
- ACADIA Pharmaceuticals AB, Medeon Science Park, S-205 12 Malmö, Sweden
| | - Audra L. Scully
- ACADIA Pharmaceuticals Inc., 3911 Sorrento Valley Boulevard, San Diego, California 92121
| | - Norman R. Nash
- ACADIA Pharmaceuticals Inc., 3911 Sorrento Valley Boulevard, San Diego, California 92121
| | - Luis R. Gardell
- ACADIA Pharmaceuticals Inc., 3911 Sorrento Valley Boulevard, San Diego, California 92121
| | - Jelveh Lameh
- ACADIA Pharmaceuticals Inc., 3911 Sorrento Valley Boulevard, San Diego, California 92121
| | - Roger Olsson
- ACADIA Pharmaceuticals AB, Medeon Science Park, S-205 12 Malmö, Sweden
| |
Collapse
|
41
|
Pugliese AM, Trincavelli ML, Lecca D, Coppi E, Fumagalli M, Ferrario S, Failli P, Daniele S, Martini C, Pedata F, Abbracchio MP. Functional characterization of two isoforms of the P2Y-like receptor GPR17: [35S]GTPgammaS binding and electrophysiological studies in 1321N1 cells. Am J Physiol Cell Physiol 2009; 297:C1028-40. [PMID: 19625605 DOI: 10.1152/ajpcell.00658.2008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The previously "orphan" G protein-coupled receptor GPR17 is structurally related to both P2Y nucleotide receptors and to receptors for cysteinyl leukotrienes. Genomic analysis revealed two putative open reading frames encoding for a "short" and a "long" receptor isoform of 339- and 367-amino acids, respectively, with the latter displaying a 28-amino acid longer NH(2) terminus. The short isoform has been recently "deorphanized," revealing dual responses to uracil nucleotides and cysteinyl leukotrienes. No information regarding the ligand specificity, tissue distribution, or pathophysiological roles of the long receptor isoform is available. In the present study, we cloned human long-GPR17, determined its tissue distribution, and characterized its pharmacological specificity in 1321N1 cells by [35S]GTPgammaS binding (which measures the ability of G protein-coupled receptor agonists to increase GTP binding to G proteins) and whole cell patch-clamp recording measuring receptor coupling to K+ channels. [35S]GTPgammaS binding in long-GPR17-expressing 1321N1 cells revealed concentration-dependent responses to uracil nucleotides (UDP-galactose = UDP > UDP-glucose) and cysteinyl leukotrienes (LTC4 > LTD4), which were counteracted by a purinergic (cangrelor) and a cysteinyl leukotriene antagonist (montelukast), respectively. The nonhydrolyzable ATP analog ATPgammaS also acted as an antagonist. GPR17 coupled to Gi and, to a lesser extent, Gq proteins. UDP-glucose and LTD(4) also induced increases in overall outward K+ currents, which were antagonized by the purinergic antagonists MRS2179 and cangrelor and by montelukast. We conclude that the previously uncharacterized long-GPR17 isoform is a functional receptor that is stimulated by both uracil nucleotides and cysteinyl leukotrienes. We also show that the signaling pathway of GPR17 involves the generation of outward K+ currents, an important protective mechanism that, in brain, is specifically aimed at reducing neuronal hyperexcitability and resultant neuronal injury.
Collapse
Affiliation(s)
- Anna Maria Pugliese
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
A new key neurohormone controlling reproduction, gonadotropin-inhibitory hormone (GnIH): Biosynthesis, mode of action and functional significance. Prog Neurobiol 2009; 88:76-88. [PMID: 19428963 DOI: 10.1016/j.pneurobio.2009.02.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 12/22/2008] [Accepted: 02/12/2009] [Indexed: 11/23/2022]
Abstract
Identification of novel neurohormones that play important roles in the regulation of pituitary function is essential for the progress of neurobiology. The decapeptide gonadotropin-releasing hormone (GnRH) is the primary factor responsible for the hypothalamic control of gonadotropin secretion. Gonadal sex steroids and inhibin inhibit gonadotropin secretion via feedback from the gonads, but a neuropeptide inhibitor of gonadotropin secretion was, until recently, unknown in vertebrates. In 2000, a novel hypothalamic dodecapeptide that inhibits gonadotropin release was identified in quail and termed gonadotropin-inhibitory hormone (GnIH). This was the first demonstration of a hypothalamic neuropeptide inhibiting gonadotropin release in any vertebrate. GnIH acts on the pituitary and GnRH neurons in the hypothalamus via a novel G protein-coupled receptor for GnIH to inhibit gonadal development and maintenance by decreasing gonadotropin release and synthesis. GnIH neurons express the melatonin receptor and melatonin stimulates the expression of GnIH. Because GnIH exists and functions in several avian species, GnIH is considered to be a new key neurohormone controlling avian reproduction. From a broader perspective, subsequently the presence of GnIH homologous peptides has been demonstrated in other vertebrates. Mammalian GnIH homologous peptides also act to inhibit reproduction by decreasing gonadotropin release in several mammalian species. Thus, the discovery of GnIH has opened the door to a new research field in reproductive neurobiology. This review summarizes the advances made in our understanding of the biosynthesis, mode of action and functional significance of GnIH, a newly discovered key neurohormone, and its homologous peptides.
Collapse
|
43
|
The recently identified P2Y-like receptor GPR17 is a sensor of brain damage and a new target for brain repair. PLoS One 2008; 3:e3579. [PMID: 18974869 PMCID: PMC2570486 DOI: 10.1371/journal.pone.0003579] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 10/08/2008] [Indexed: 11/25/2022] Open
Abstract
Deciphering the mechanisms regulating the generation of new neurons and new oligodendrocytes, the myelinating cells of the central nervous system, is of paramount importance to address new strategies to replace endogenous damaged cells in the adult brain and foster repair in neurodegenerative diseases. Upon brain injury, the extracellular concentrations of nucleotides and cysteinyl-leukotrienes (cysLTs), two families of endogenous signaling molecules, are markedly increased at the site of damage, suggesting that they may act as “danger signals” to alert responses to tissue damage and start repair. Here we show that, in brain telencephalon, GPR17, a recently deorphanized receptor for both uracil nucleotides and cysLTs (e.g., UDP-glucose and LTD4), is normally present on neurons and on a subset of parenchymal quiescent oligodendrocyte precursor cells. We also show that induction of brain injury using an established focal ischemia model in the rodent induces profound spatiotemporal-dependent changes of GPR17. In the lesioned area, we observed an early and transient up-regulation of GPR17 in neurons expressing the cellular stress marker heat shock protein 70. Magnetic Resonance Imaging in living mice showed that the in vivo pharmacological or biotechnological knock down of GPR17 markedly prevents brain infarct evolution, suggesting GPR17 as a mediator of neuronal death at this early ischemic stage. At later times after ischemia, GPR17 immuno-labeling appeared on microglia/macrophages infiltrating the lesioned area to indicate that GPR17 may also acts as a player in the remodeling of brain circuitries by microglia. At this later stage, parenchymal GPR17+ oligodendrocyte progenitors started proliferating in the peri-injured area, suggesting initiation of remyelination. To confirm a specific role for GPR17 in oligodendrocyte differentiation, the in vitro exposure of cortical pre-oligodendrocytes to the GPR17 endogenous ligands UDP-glucose and LTD4 promoted the expression of myelin basic protein, confirming progression toward mature oligodendrocytes. Thus, GPR17 may act as a “sensor” that is activated upon brain injury on several embryonically distinct cell types, and may play a key role in both inducing neuronal death inside the ischemic core and in orchestrating the local remodeling/repair response. Specifically, we suggest GPR17 as a novel target for therapeutic manipulation to foster repair of demyelinating wounds, the types of lesions that also occur in patients with multiple sclerosis.
Collapse
|
44
|
Cline MA, Bowden CN, Calchary WA, Layne JE. Short-term anorexigenic effects of central neuropeptide VF are associated with hypothalamic changes in chicks. J Neuroendocrinol 2008; 20:971-7. [PMID: 18540998 DOI: 10.1111/j.1365-2826.2008.01749.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The present study was designed to measure food and water intake, changes in hypothalamic chemistry, and other behaviour modifications after central injection of neuropeptide (NP) VF in broiler type chicks. In Experiment 1, chicks responded to central NPVF with a reduction in food intake for up to 90 min post injection. Water intake was unaffected. In Experiment 2, NPVF exerted a less potent and shorter duration of attenuated food intake than did the structurally related NPFF. In Experiment 3, 16.0 nmol NPVF reversed the prolactin-releasing peptide induced orexigenic effect. In Experiment 4, central NPVF treatment was associated with decreased c-Fos immunoreactivity in the lateral hypothalamus, whereas c-Fos immunoreactivity in the dorsomedial nucleus, infundibular nucleus (homologue to the mammalian arcuate nucleus) and ventromedial nucleus was increased. In Experiment 5, behaviours unrelated to ingestion including sit, stand, deep rest and locomotion were affected by central NPVF injection. Some of these behaviours are incompatible with ingestion and may contribute to hypothalamic associated perception of satiety after central NPVF. In conclusion, NVPF is a short-term regulator of appetite and its effects are associated with hypothalamic and behaviour changes in chicks.
Collapse
Affiliation(s)
- M A Cline
- Department of Biology, Radford University, Radford, VA 24142, USA.
| | | | | | | |
Collapse
|
45
|
Lindner D, van Dieck J, Merten N, Mörl K, Günther R, Hofmann HJ, Beck-Sickinger AG. GPC receptors and not ligands decide the binding mode in neuropeptide Y multireceptor/multiligand system. Biochemistry 2008; 47:5905-14. [PMID: 18457425 DOI: 10.1021/bi800181k] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many G protein-coupled receptors belong to families of different receptor subtypes, which are recognized by a variety of distinct ligands. To study such a multireceptor/multiligand system, we investigated the Y-receptor family. This family consists of four G protein-coupled Y receptors in humans (hY 1R, hY 2R, hY 4R, and hY 5R) and is activated by the so-called NPY hormone family, which itself consists of three native peptide ligands named neuropeptide Y (NPY), pancreatic polypeptide (PP), and peptide YY (PYY). The hY 5R shows high affinity for all ligands, although for PP binding, the affinity is slightly decreased. As a rational explanation, we suggest that Tyr (27) is lost as a contact point between PP and the hY 5R in contrast to NPY or PYY. Furthermore, several important residues for ligand binding were identified by the first extensive mutagenesis study of the hY 5R. Using a complementary mutagenesis approach, we were able to discover a novel interaction point between hY 5R and NPY. The interaction between NPY(Arg (25)) and hY 5R(Asp (2.68)) as well as between NPY(Arg (33)) and hY 5R(Asp (6.59)) is maintained in the binding of PYY and PP to hY 5R but different to the PP-hY 4R and NPY-hY 1R contact points. Therefore, we provide evidence that the receptor subtype and not the pre-orientated conformation of the ligand at the membrane decides the binding mode. Furthermore, the first hY 5R model was set up on the basis of the crystal structure of bovine rhodopsin. We can show that most of the residues identified to be critical for ligand binding are located within the now postulated binding pocket.
Collapse
Affiliation(s)
- Diana Lindner
- Leipzig University, Institute of Biochemistry, Brüderstrasse 34, 04103 Leipzig, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Dylag T, Pachuta A, Raoof H, Kotlinska J, Silberring J. A novel cryptic peptide derived from the rat neuropeptide FF precursor reverses antinociception and conditioned place preference induced by morphine. Peptides 2008; 29:473-8. [PMID: 17980934 DOI: 10.1016/j.peptides.2007.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 09/20/2007] [Accepted: 09/21/2007] [Indexed: 12/21/2022]
Abstract
Neuropeptide FF (NPFF) precursors from different species contain at least three known neuropeptides, i.e. FF (FLFQPQRF-NH(2)), AF (AGEGLSSPFWSLAAPQR-NH(2)) and SF (SLAAPQRF-NH(2)). We demonstrate that the rat NPFF precursor contains another bioactive sequence, NAWGPWSKEQLSPQA, spanning between positions 85 and 99. Synthetic NPFF precursor (85-99) (10 and 20 nmol, i.c.v.) blocked the expression of conditioned place preference induced by morphine (5 mg/kg, s.c.). This peptide alone (10 and 20 nmol, i.c.v.) had no influence on the baseline latency of a nociceptive reaction but reversed the antinociceptive activity of morphine (5 mg/kg, s.c.) in the tail-immersion test in rats. These data suggest the existence of a novel bioactive cryptic peptide within an already known NPFF precursor.
Collapse
Affiliation(s)
- Tomasz Dylag
- Faculty of Chemistry and Regional Laboratory, Jagiellonian University, Ingardena 3, Krakow, Poland
| | | | | | | | | |
Collapse
|
47
|
Yang HYT, Tao T, Iadarola MJ. Modulatory role of neuropeptide FF system in nociception and opiate analgesia. Neuropeptides 2008; 42:1-18. [PMID: 17854890 DOI: 10.1016/j.npep.2007.06.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 06/14/2007] [Indexed: 10/22/2022]
Abstract
The tetra-peptide FMRF-NH(2) is a cardioexcitatory peptide in the clam. Using the antibody against this peptide, FMRF-NH(2)-like immunoreactive material was detected in mammalian CNS. Subsequently, mammalian FMRF-NH(2) immunoreactive peptides were isolated from bovine brain and characterized to be FLFQPQRF-NH(2) (NPFF) and AGEGLSSPFWSLAAPQRF-NH(2) (NPAF). The genes encoding NPFF precursor proteins and NPFF receptors 1 and 2 are expressed in all vertebrate species examined to date and are highly conserved. Among many biological roles suggested for the NPFF system, the possible modulatory role of NPFF in nocicetion and opiate analgesia has been most widely investigated. Pharmacologically, NPFF-related peptides were found to exhibit analgesia and also potentiate the analgesic activity of opiates when administered intrathecally but attenuate the opiate induced analgesia when administered intracerebroventricularly. RF-NH(2) peptides including NPFF-related peptides were found to delay the rate of acid sensing ion channels (ASIC) desensitization resulting in enhancing acid gated currents, raising the possibility that NPFF also may have a pain modulatory role through ASIC. The genes for NPFF as well as NPFF-R2, preferred receptor for NPFF, are highly unevenly expressed in the rat CNS with the highest levels localized to the superficial layers of the dorsal spinal cord. These two genes are also present in the dorsal root ganglia (DRG), though at low levels in normal rats. NPFF and NPFF-R2 mRNAs were found to be coordinately up-regulated in spinal cord and DRG of rats with peripheral inflammation. In addition, NPFF-R2 immunoreactivity in the primary afferents was increased by peripheral inflammation. The findings from the early studies on the analgesic and morphine modulating activities suggested a role for NPFF in pain modulation and this possibility is further supported by the distribution of NPFF and its receptor and the regulation of the NPFF system in vivo.
Collapse
Affiliation(s)
- Hsiu-Ying T Yang
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD 20892-4410, USA.
| | | | | |
Collapse
|
48
|
Cline MA, Nandar W, Rogers JO. Central neuropeptide FF reduces feed consumption and affects hypothalamic chemistry in chicks. Neuropeptides 2007; 41:433-9. [PMID: 17936900 DOI: 10.1016/j.npep.2007.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 08/22/2007] [Accepted: 08/23/2007] [Indexed: 10/22/2022]
Abstract
Information on the physiological functions of neuropeptide FF; NPFF, a morphine modulating octapeptide in avians is lacking. Thus, we designed a study to investigate the effects of central NPFF with particular emphasis on appetite-related processes. Cobb-500 chicks were intracerebroventricularly (ICV) injected with 0, 4.16, 8.32 or 16.6nmol NPFF, and feed and water intake were quantified. Feed intake was linearly decreased as NPFF dose increased, and this effect decayed over time and was not significant by 120min post-injection. Water intake was not affected by ICV NPFF. In a second exp, we observed that naloxone completely reversed the NPFF-induced decrease in feed intake. The amount of time a visible marker took to travel through the total length of the alimentary canal linearly increased as NPFF dose increased. We measured neuronal activation in the lateral hypothalamus (LH), paraventricular nucleus (PVN) dorsomedial nucleus (DMN) and ventromedial hypothalamus (VMN) of the hypothalamus, and nucleus dorsomedialis posterior thalami (DMP) of the thalamus. The DMN, DMP, PVN and VMH were all activated by ICV NPFF while the LH was not affected. Finally, we determined that the anorexigenic effect of ICV NPFF is primarily behavior specific, since behaviors unrelated to ingestion were not increased the same duration of time as was consumatory pecking. We conclude that NPFF causes anorexigenic effects in chicks that are primarily behavior specific.
Collapse
Affiliation(s)
- Mark A Cline
- Department of Biology, Radford University, P.O. Box 6931, Radford, VA 24142, USA.
| | | | | |
Collapse
|
49
|
Kotlinska J, Pachuta A, Dylag T, Silberring J. Neuropeptide FF (NPFF) reduces the expression of morphine- but not of ethanol-induced conditioned place preference in rats. Peptides 2007; 28:2235-42. [PMID: 17884254 DOI: 10.1016/j.peptides.2007.08.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 08/10/2007] [Accepted: 08/10/2007] [Indexed: 10/22/2022]
Abstract
Neuropeptide FF (NPFF) has been described as an anti-opioid peptide. It plays a role in opioid antinociception, dependence and tolerance. Previous study has indicated that 1DMe ([D-Tyr(1), (NMe)Phe(3)]NPFF), a stable analog of NPFF, inhibits acquisition of the rewarding effect of morphine but not of ethanol in mice. The rewarding effects of these drugs were measured in the unbiased paradigm of conditioned place preference (CPP). The present study examines the influence of NPFF on the expression of morphine- and ethanol-induced CPP in the biased procedure in rats. Our experiments showed that NPFF, given intracerebroventricularly (i.c.v.) at the doses of 5, 10 and 20 nmol, inhibited the expression of morphine-induced CPP. NPFF gave itself, neither induced place preference nor aversion, although a tendency to aversive effect was seen at the highest dose of 20 nmol. NPFF did not indicate fear behavior in the elevated plus maze test, and did not disturb locomotor activity of rats. However, NPFF was unable to inhibit the expression of ethanol-induced CPP. Probably this effect is due to the fact that ethanol reward is a more complex process and apart from the role of opioids, there are other neurotransmitters also involved in this mechanism. These results suggest that NPFF is involved in the expression of morphine reward. Moreover, our study supports an anti-opioid character of this peptide.
Collapse
Affiliation(s)
- Jolanta Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University School, Staszica 4, 20-081 Lublin, Poland.
| | | | | | | |
Collapse
|
50
|
Lecca D, Abbracchio MP. Deorphanisation of G protein-coupled receptors: A tool to provide new insights in nervous system pathophysiology and new targets for psycho-active drugs. Neurochem Int 2007; 52:339-51. [PMID: 17884255 DOI: 10.1016/j.neuint.2007.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 07/18/2007] [Accepted: 08/06/2007] [Indexed: 11/20/2022]
Abstract
G protein-coupled receptors represent the largest family of membrane receptors translating extracellular into intracellular signals. Endogenous ligands for these receptors range from physical stimuli (e.g., light and odorants) to ions and chemical transmitters, such as "classical" biogenic amines, nucleotides and peptides. Some of these receptors are pathologically altered in neurodegenerative and psychiatric diseases and indeed represent the target for a variety of already marketed psycho-active drugs. With the publication of the human genome, it has become evident that there still are many "orphan" G protein-coupled receptors, i.e., receptors responding to yet-unidentified endogenous ligands. A large amount of these receptors are expressed in nervous tissues, but, apart from their molecular structure, we have no information concerning their physiological roles and alterations in disease states. In this review, we summarise the advancements and pitfalls of the strategies that have been exploited in recent years to "deorphanise" some of these receptors. We also show how, in some cases, this deorphanisation process has resulted in the identification of new potential targets for drug development as well as in the discovery of previously unknown neurotransmitters, including bioactive peptides and substances that had been merely known as metabolic intermediates. We envisage that the deorphanisation of the remaining orphan G protein-coupled receptors will further advance our knowledge of nervous system pathophysiology and unveil additional targets for new therapeutic approaches to human diseases, including psychosis, depression, anxiety, pain and aging-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological Sciences, School of Pharmacy, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | | |
Collapse
|