1
|
Istiban MN, De Fruyt N, Kenis S, Beets I. Evolutionary conserved peptide and glycoprotein hormone-like neuroendocrine systems in C. elegans. Mol Cell Endocrinol 2024; 584:112162. [PMID: 38290646 PMCID: PMC11004728 DOI: 10.1016/j.mce.2024.112162] [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: 11/30/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/01/2024]
Abstract
Peptides and protein hormones form the largest group of secreted signals that mediate intercellular communication and are central regulators of physiology and behavior in all animals. Phylogenetic analyses and biochemical identifications of peptide-receptor systems reveal a broad evolutionary conservation of these signaling systems at the molecular level. Substantial progress has been made in recent years on characterizing the physiological and putative ancestral roles of many peptide systems through comparative studies in invertebrate models. Several peptides and protein hormones are not only molecularly conserved but also have conserved roles across animal phyla. Here, we focus on functional insights gained in the nematode Caenorhabditis elegans that, with its compact and well-described nervous system, provides a powerful model to dissect neuroendocrine signaling networks involved in the control of physiology and behavior. We summarize recent discoveries on the evolutionary conservation and knowledge on the functions of peptide and protein hormone systems in C. elegans.
Collapse
Affiliation(s)
- Majdulin Nabil Istiban
- Neural Signaling and Circuit Plasticity, Department of Biology, KU Leuven, 3000, Leuven, Belgium
| | - Nathan De Fruyt
- Neural Signaling and Circuit Plasticity, Department of Biology, KU Leuven, 3000, Leuven, Belgium
| | - Signe Kenis
- Neural Signaling and Circuit Plasticity, Department of Biology, KU Leuven, 3000, Leuven, Belgium
| | - Isabel Beets
- Neural Signaling and Circuit Plasticity, Department of Biology, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
2
|
Chen D, Zhang M, Zhang Q, Wu S, Yu B, Zhang X, Hu X, Zhang S, Yang Z, Kuang J, Xu B, Fang Q. The blockade of neuropeptide FF receptor 1 and 2 differentially contributed to the modulating effects on fentanyl-induced analgesia and hyperalgesia in mice. Eur J Pharmacol 2024; 969:176457. [PMID: 38395375 DOI: 10.1016/j.ejphar.2024.176457] [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: 10/28/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024]
Abstract
Neuropeptide FF (NPFF) plays a critical role in various physiological processes through the activation of neuropeptide FF receptor 1 and 2 (NPFFR1 and NPFFR2). Numerous evidence has indicated that NPFF exhibits opposite opioid-modulating effects on opioid-induced analgesia after supraspinal and spinal administrations, while the detailed role of NPFFR1 and NPFFR2 remains unclear. In this study, we employed pharmacological and genetic inhibition of NPFFR to investigate the modulating roles of central NPFFR1 and NPFFR2 in opioid-induced analgesia and hyperalgesia, using a male mouse model of acute fentanyl-induced analgesia and secondary hyperalgesia. Our findings revealed that intrathecal (i.t.) injection of the nonselective NPFFR antagonist RF9 significantly enhanced fentanyl-induced analgesia, whereas intracerebroventricular (i.c.v.) injection did not show the same effect. Moreover, NPFFR2 deficient (npffr2-/-) mice exhibited stronger analgesic responses to fentanyl compared to wild type (WT) or NPFFR1 knockout (npffr1-/-) mice. Intrathecal injection of RF9 in npffr1-/- mice also significantly enhanced fentanyl-induced analgesia. These results indicate a crucial role of spinal NPFFR2 in the enhancement of opioid analgesia. Contrastingly, hyperalgesia induced by fentanyl was markedly reversed in npffr1-/- mice but remained unaffected in npffr2-/- mice. Similarly, i.c.v. injection of the selective NPFFR1 antagonist RF3286 effectively prevented fentanyl-induced hyperalgesia in WT or npffr2-/- mice. Notably, co-administration of i.c.v. RF3286 and i.t. RF9 augmented fentanyl-induced analgesia while reducing hyperalgesia. Collectively, these findings highlight the modulating effects of blocking spinal NPFFR2 and supraspinal NPFFR1 on fentanyl-induced analgesia and hyperalgesia, respectively, which shed a light on understanding the pharmacological function of NPFF system in future studies.
Collapse
Affiliation(s)
- Dan Chen
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Mengna Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Qinqin Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Shuyuan Wu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Bowen Yu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Xiaodi Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Xuanran Hu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Shichao Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Zhenyun Yang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Junzhe Kuang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Biao Xu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| | - Quan Fang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| |
Collapse
|
3
|
Wang Y, Zuo Z, Shi J, Fang Y, Yin Z, Wang Z, Yang Z, Jia B, Sun Y. Modulatory role of neuropeptide FF system in macrophages. Peptides 2024; 174:171164. [PMID: 38272240 DOI: 10.1016/j.peptides.2024.171164] [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: 11/09/2023] [Revised: 12/29/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
Neuropeptide FF (NPFF) is an octapeptide that regulates various cellular processes, especially pain perception. Recently, there has been a growing interest in understanding the modulation of NPFF in neuroendocrine inflammation. This review aims to provide a thorough overview of the regulation of NPFF in macrophage-mediated biological processes. We delve into the impact of NPFF on macrophage polarization, self-renewal modulation, and the promotion of mitophagy, facilitating the transition from thermogenic fat to fat-storing adipose tissue. Additionally, we explore the NPFF-dependent regulation of the inflammatory response mediated by macrophages, its impact on the differentiation of macrophages, and its capacity to induce alterations in the transcriptome of macrophages. We also address the potential of NPFF as a therapeutic molecule in the field of neuroendocrine inflammation. Overall, our work offers an understanding of the influence of NPFF on macrophage, facilitating the exploration of its pharmacological significance in future studies.
Collapse
Affiliation(s)
- Yaxing Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhuo Zuo
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Jiajia Shi
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yanwei Fang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhongqian Yin
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhe Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhouqi Yang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Bin Jia
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yulong Sun
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
4
|
Strnadová V, Karnošová A, Blechová M, Neprašová B, Holá L, Němcová A, Myšková A, Sýkora D, Železná B, Kuneš J, Maletínská L. Search for lipidized PrRP analogs with strong anorexigenic effect: In vitro and in vivo studies. Neuropeptides 2023; 98:102319. [PMID: 36669365 DOI: 10.1016/j.npep.2022.102319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/18/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023]
Abstract
Prolactin-releasing peptide (PrRP) is an anorexigenic neuropeptide that attenuates food intake and increases energy expenditure. We designed three series of new lipidized PrRP31 analogs of different lengths of fatty acids attached at amino acids 1 or 11 directly or via linkers, part of them acetylated at the N-terminus and/or modified with dichlorophenylalanine (PheCl2) at the C-terminus. We tested their affinity for and activation of signaling pathways relevant to receptors GPR10, NPFF-R2, and NPFF-R1, effect on food intake in fasted or freely fed mice and rats, and stability in rat plasma. We aimed to select a strong dual GPR10/NPFF-R2 agonist whose affinity for NPFF-1 was not enhanced. The selected potent analog was then tested for body weight-lowering potency after chronic administration in mice with diet-induced obesity. PrRP31 analogs lipidized by monocarboxylic fatty acids showed strong dual affinity for both GPR10 and NPFF-R2 and activated MAPK/ERK1/2, Akt and CREB in cells overexpressing GPR10 and NPFF-R2. The selected analog stabilized at N- and C-termini and palmitoylated through the TTDS linker to Lys11 is a powerful dual agonist GPR10/NPFF-R2 at not enhanced affinity for NPFF-R1. It showed strong anti-obesity properties in mice with diet-induced obesity and became a potential compound for further studies.
Collapse
Affiliation(s)
- Veronika Strnadová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic
| | - Alena Karnošová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic; First Faculty of Medicine, Charles University, Prague 121 08, Czech Republic
| | - Miroslava Blechová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic
| | - Barbora Neprašová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic; Institute of Physiology, CAS, Prague 142 00, Czech Republic
| | - Lucie Holá
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic; First Faculty of Medicine, Charles University, Prague 121 08, Czech Republic
| | - Anna Němcová
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic; University of Chemistry and Technology, Prague 166 28, Czech Republic
| | - Aneta Myšková
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic; University of Chemistry and Technology, Prague 166 28, Czech Republic
| | - David Sýkora
- University of Chemistry and Technology, Prague 166 28, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic; Institute of Physiology, CAS, Prague 142 00, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, CAS, Prague 166 10, Czech Republic.
| |
Collapse
|
5
|
Yu H, Wang Y, Gao J, Gao Y, Zhong C, Chen Y. Application of the neuropeptide NPVF to enhance angiogenesis and osteogenesis in bone regeneration. Commun Biol 2023; 6:197. [PMID: 36804475 PMCID: PMC9941492 DOI: 10.1038/s42003-023-04567-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
The brain-bone regulatory system regulates skeletal homeostasis via bioactive neuropeptides, yet the underlying mechanism remains elusive. Here, we report the role of the neuropeptide VF (NPVF, VPNLPQRF-NH2) in enhancing both angiogenesis and osteogenesis in a rat skeletal system and the potential pathways involved. An in vitro study revealed that NPVF not only promotes migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) by activating NPFFR1, which leads to upregulation of miR-181c-3p and downregulation of Argonaute1 (AGO1), but also mediates osteogenic differentiation of bone mesenchymal stem cells (BMSCs) via the Wnt/β-catenin signaling pathway. To improve the stability and bioavailability and thus efficacy of NPVF as a promoter of in vivo bone regeneration, we genetically engineered amyloid-NPVF-fusion proteins and utilized them as self-assembling nanofiber coatings to treat bone defects in a rat calvarial defect model. We found that a porous hydroxyapatite scaffold loaded with the NPVF peptide-fused amyloid coating substantially enhanced angiogenesis and site-specific fresh bone in-growth when implanted in calvarial defects. Taken together, our work uncovered a previously undefined crosstalk between the brain and bone by unveiling the role of NPVF in bone tissue and demonstrated a viable method for promoting bone tissue repairs based upon self-assembling NPVF-containing protein coatings.
Collapse
Affiliation(s)
- Hongping Yu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yanyi Wang
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junjie Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Science, Ningbo, Zhejiang, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Chao Zhong
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
6
|
Palmitoylation of Prolactin-Releasing Peptide Increased Affinity for and Activation of the GPR10, NPFF-R2 and NPFF-R1 Receptors: In Vitro Study. Int J Mol Sci 2021; 22:ijms22168904. [PMID: 34445614 PMCID: PMC8396344 DOI: 10.3390/ijms22168904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
The anorexigenic neuropeptide prolactin-releasing peptide (PrRP) is involved in the regulation of food intake and energy expenditure. Lipidization of PrRP stabilizes the peptide, facilitates central effect after peripheral administration and increases its affinity for its receptor, GPR10, and for the neuropeptide FF (NPFF) receptor NPFF-R2. The two most potent palmitoylated analogs with anorectic effects in mice, palm11-PrRP31 and palm-PrRP31, were studied in vitro to determine their agonist/antagonist properties and mechanism of action on GPR10, NPFF-R2 and other potential off-target receptors related to energy homeostasis. Palmitoylation of both PrRP31 analogs increased the binding properties of PrRP31 to anorexigenic receptors GPR10 and NPFF-R2 and resulted in a high affinity for another NPFF receptor, NPFF-R1. Moreover, in CHO-K1 cells expressing GPR10, NPFF-R2 or NPFF-R1, palm11-PrRP and palm-PrRP significantly increased the phosphorylation of extracellular signal-regulated kinase (ERK), protein kinase B (Akt) and cAMP-responsive element-binding protein (CREB). Palm11-PrRP31, unlike palm-PrRP31, did not activate either c-Jun N-terminal kinase (JNK), p38, c-Jun, c-Fos or CREB pathways in cells expressing NPFF-1R. Palm-PrRP31 also has higher binding affinities for off-target receptors, namely, the ghrelin, opioid (KOR, MOR, DOR and OPR-L1) and neuropeptide Y (Y1, Y2 and Y5) receptors. Palm11-PrRP31 exhibited fewer off-target activities; therefore, it has a higher potential to be used as an anti-obesity drug with anorectic effects.
Collapse
|
7
|
Transcriptomic Changes in Mouse Bone Marrow-Derived Macrophages Exposed to Neuropeptide FF. Genes (Basel) 2021; 12:genes12050705. [PMID: 34065092 PMCID: PMC8151073 DOI: 10.3390/genes12050705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 12/27/2022] Open
Abstract
Neuropeptide FF (NPFF) is a neuropeptide that regulates various biological activities. Currently, the regulation of NPFF on the immune system is an emerging field. However, the influence of NPFF on the transcriptome of primary macrophages has not been fully elucidated. In this study, the effect of NPFF on the transcriptome of mouse bone marrow-derived macrophages (BMDMs) was explored by RNA sequencing, bioinformatics, and molecular simulation. BMDMs were treated with 1 nM NPFF for 18 h, followed by RNA sequencing. Differentially expressed genes (DEGs) were obtained, followed by GO, KEGG, and PPI analysis. A total of eight qPCR-validated DEGs were selected as hub genes. Subsequently, the three-dimensional (3-D) structures of the eight hub proteins were constructed by Modeller and Rosetta. Next, the molecular dynamics (MD)-optimized 3-D structure of hub protein was acquired with Gromacs. Finally, the binding modes between NPFF and hub proteins were studied by Rosetta. A total of 2655 DEGs were obtained (up-regulated 1442 vs. down-regulated 1213), and enrichment analysis showed that NPFF extensively regulates multiple functional pathways mediated by BMDMs. Moreover, the 3-D structure of the hub protein was obtained after MD-optimization. Finally, the docking modes of NPFF-hub proteins were predicted. Besides, NPFFR2 was expressed on the cell membrane of BMDMs, and NPFF 1 nM significantly activated NPFFR2 protein expression. In summary, instead of significantly inhibiting the expression of the immune-related gene transcriptome of RAW 264.7 cells, NPFF simultaneously up-regulated and down-regulated the gene expression profile of a large number of BMDMs, hinting that NPFF may profoundly affect a variety of cellular processes dominated by BMDMs. Our work provides transcriptomics clues for exploring the influence of NPFF on the physiological functions of BMDMs.
Collapse
|
8
|
He C, Wang X, Zhang J, Wang H, Zhao Y, Shah JN, Ma C, Li H, Su W, Zhang Z, Chen S, Zhou L, Dong S. MCRT, a multifunctional ligand of opioid and neuropeptide FF receptors, attenuates neuropathic pain in spared nerve injury model. Basic Clin Pharmacol Toxicol 2021; 128:731-740. [PMID: 33533572 DOI: 10.1111/bcpt.13566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 11/30/2022]
Abstract
Chimeric peptide MCRT (YPFPFRTic-NH2 ) was a multifunctional ligand of opioid and neuropeptide FF (NPFF) receptors and reported to be potentially antalgic in acute tail-flick test. Here, we developed spared nerve injury (SNI) model to explore its efficacy in chronic neuropathic pain. Analgesic tolerance, opioid-induced hyperalgesia and gastrointestinal transit were measured for safety evaluation. Intracerebroventricular (i.c.v.) and intraplantar (i.pl.) injections were conducted as central and peripheral routes, respectively. Results demonstrated that MCRT alleviated neuropathic pain effectively and efficiently, with the ED50 values of 4.93 nmol/kg at the central level and 3.11 nmol/kg at the peripheral level. The antagonist blocking study verified the involvement of mu-, delta-opioid and NPFF receptors in MCRT produced anti-allodynia. Moreover, the separation of analgesia from unwanted effects was preliminarily achieved and that MCRT caused neither analgesic tolerance nor hyperalgesia after chronic i.c.v. administration, nor constipation after i.pl. administration. Notably, the local efficacy of MCRT in SNI mice was about one thousandfold higher than morphine and ten thousandfold higher than pregabalin, indicating a great promise in the future treatment of neuropathic pain.
Collapse
Affiliation(s)
- Chunbo He
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoli Wang
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jing Zhang
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hao Wang
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yaofeng Zhao
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jagat Narayan Shah
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Chan Ma
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hailan Li
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Wenting Su
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Zhe Zhang
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Shasha Chen
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lanxia Zhou
- The Central Laboratory, The First Hospital, Lanzhou University, Lanzhou, China
| | - Shouliang Dong
- Department of Animal and Biomedical Sciences, School of Life Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Sun Y, Kuang Y, Zuo Z, Zhang J, Ma X, Xing X, Liu L, Miao Y, Ren T, Li H, Mei Q. Cellular processes involved in RAW 264.7 macrophages exposed to NPFF: A transcriptional study. Peptides 2021; 136:170469. [PMID: 33309723 DOI: 10.1016/j.peptides.2020.170469] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/25/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022]
Abstract
Neuropeptide FF (NPFF) is a neuropeptide that modulates various physiological processes. The regulatory role of NPFF in the immune and inflammatory response is currently being revealed. However, the effect of NPFF at the transcriptome level in macrophages has not been fully elucidated. Here, the impact of NPFF on gene expression at the transcriptome level of RAW 264.7 cells was investigated by RNA-seq. RAW 264.7 macrophages were treated with NPFF (1 nM) for 18 h, followed by RNA-seq examination. Differentially expressed genes (DEGs) were acquired, followed by GO, KEGG, and PPI analysis. A total of eight qPCR-verified DEGs were obtained. Next, three-dimensional models of the eight hub proteins were constructed by using homology modeling with Modeller (9v23). Finally, molecular dynamics simulation (300 ns) was performed with GROMACS 2018.2 to investigate the structural characteristics of these hub proteins. NPFF had no detectable effect on the morphology of RAW264.7 cells. A total of 211 DEGs were acquired, and an enrichment study demonstrated that the immune response-related pathway was significantly inhibited by NPFF. Moreover, the molecular dynamics optimized-protein models of the hub proteins were obtained. Collectively, NPFF inhibited the expression of immune-related genes in RAW 264.7 cells at the transcriptome level, which suggested a negative relationship between NPFF and this set of immune-related genes in RAW 264.7 macrophages. Therefore, our data may provide direct evidence of the role of NPFF in peripheral or central inflammatory diseases.
Collapse
Affiliation(s)
- Yulong Sun
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China.
| | - Yuanyuan Kuang
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Zhuo Zuo
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Jin Zhang
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xianning West Road, Xi'an, Shaanxi Province, 710049, China
| | - Xiaolong Ma
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Xiaoyu Xing
- School of Humanities, Economics and Laws, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Lingyi Liu
- School of Physical Science and Technology, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Yuchen Miao
- School of Physical Science and Technology, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Tao Ren
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Hui Li
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, South Door Slightly Friendship Road 555, Xi'an, Shaanxi Province, 710054, China
| | - Qibing Mei
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| |
Collapse
|
10
|
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
|
11
|
Zhang R, Xu B, Zhang Q, Chen D, Zhang M, Zhao G, Xu K, Xiao J, Zhu H, Niu J, Li N, Fang Q. Spinal administration of the multi-functional opioid/neuropeptide FF agonist BN-9 produced potent antinociception without development of tolerance and opioid-induced hyperalgesia. Eur J Pharmacol 2020; 880:173169. [PMID: 32416184 DOI: 10.1016/j.ejphar.2020.173169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/24/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Chronic opioids treatment is impeded by the development of analgesic tolerance and opioid-induced hyperalgesia. Recent studies have shown that multi-functional opioid compounds produce analgesic activities with limited side effects. We developed a novel multi-functional peptide targeting opioid and neuropeptide FF receptors named BN-9, which produced potent and non-tolerance forming antinociceptive effect after supraspinal and systemic administrations. In the present study, the analgesic properties and potential side effects of intrathecal BN-9 were investigated in a range of preclinical rodent models. In complete Freund's adjuvant-induced inflammatory pain model, intrathecal BN-9 dose-dependently produced analgesic effect via opioid receptors, and the spinal antinociceptive effect was augmented by the neuropeptide FF receptor antagonist RF9. In contrast, in plantar incision-induced postoperative pain model, BN-9 exhibited potent anti-allodynic effect via opioid receptors and, at least partially, neuropeptide FF receptors. In mouse models of acetic acid-induced visceral pain and formalin pain, BN-9-induced spinal antinociception was mainly mediated by opioid receptors, independent of neuropeptide FF receptors. Furthermore, at the spinal level, chronic treatments with BN-9 did not lead to analgesic tolerance and cross-tolerance to morphine. Moreover, opioid-induced hyperalgesia was observed after repeated administration of morphine, but not BN-9. Taken together, our present study suggests that intrathecal BN-9 produces potent and non-tolerance forming antinociception, and does not cause opioid-induced hyperalgesia. Thus, BN-9 might serve as a promising lead compound in the development of multi-functional opioid analgesics with minimized side effects.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Qinqin 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
| | - Dan Chen
- 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
| | - Mengna 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
| | - Guanghai Zhao
- 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
| | - Kangtai 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
| | - Jian Xiao
- 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
| | - Hanwen Zhu
- 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
| | - Jiandong Niu
- 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.
| | - 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.
| |
Collapse
|
12
|
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
|
13
|
Xu B, Zhang M, Shi X, Zhang R, Chen D, Chen Y, Wang Z, Qiu Y, Zhang T, Xu K, Zhang X, Liedtke W, Wang R, Fang Q. The multifunctional peptide DN-9 produced peripherally acting antinociception in inflammatory and neuropathic pain via μ- and κ-opioid receptors. Br J Pharmacol 2019; 177:93-109. [PMID: 31444977 DOI: 10.1111/bph.14848] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/05/2019] [Accepted: 08/20/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Considerable effort has recently been directed at developing multifunctional opioid drugs to minimize the unwanted side effects of opioid analgesics. We have developed a novel multifunctional opioid agonist, DN-9. Here, we studied the analgesic profiles and related side effects of peripheral DN-9 in various pain models. EXPERIMENTAL APPROACH Antinociceptive effects of DN-9 were assessed in nociceptive, inflammatory, and neuropathic pain. Whole-cell patch-clamp and calcium imaging assays were used to evaluate the inhibitory effects of DN-9 to calcium current and high-K+ -induced intracellular calcium ([Ca2+ ]i ) on dorsal root ganglion (DRG) neurons respectively. Side effects of DN-9 were evaluated in antinociceptive tolerance, abuse, gastrointestinal transit, and rotarod tests. KEY RESULTS DN-9, given subcutaneously, dose-dependently produced antinociception via peripheral opioid receptors in different pain models without sex difference. In addition, DN-9 exhibited more potent ability than morphine to inhibit calcium current and high-K+ -induced [Ca2+ ]i in DRG neurons. Repeated treatment with DN-9 produced equivalent antinociception for 8 days in multiple pain models, and DN-9 also maintained potent analgesia in morphine-tolerant mice. Furthermore, chronic DN-9 administration had no apparent effect on the microglial activation of spinal cord. After subcutaneous injection, DN-9 exhibited less abuse potential than morphine, as was gastroparesis and effects on motor coordination. CONCLUSIONS AND IMPLICATIONS DN-9 produces potent analgesia with minimal side effects, which strengthen the candidacy of peripherally acting opioids with multifunctional agonistic properties to enter human studies to alleviate the current highly problematic misuse of classic opioids on a large scale.
Collapse
Affiliation(s)
- Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Mengna Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xuerui Shi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 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, Lanzhou, China
| | - Dan Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yong Chen
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina
| | - Zilong Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 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, Lanzhou, China
| | - Kangtai Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoyu Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wolfgang Liedtke
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 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, Lanzhou, China
| |
Collapse
|
14
|
Spinal DN-9, a Peptidic Multifunctional Opioid/Neuropeptide FF Agonist Produced Potent Nontolerance Forming Analgesia With Limited Side Effects. THE JOURNAL OF PAIN 2019; 21:477-493. [PMID: 31521796 DOI: 10.1016/j.jpain.2019.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/11/2019] [Accepted: 08/25/2019] [Indexed: 11/20/2022]
Abstract
The development of multitarget opioid drugs has emerged as an attractive therapeutic strategy to eliminate opioid-related side effects. Our previous study developed a series of opioid and neuropeptide FF pharmacophore-containing chimeric peptides, including DN-9 (Tyr-D.Ala-Gly-NMe.Phe-Gly-Pro-Gln-Arg-Phe-NH2), which produced potent nontolerance forming analgesia at the supraspinal level. In the present study, the antinociceptive effects of DN-9 in a series of preclinical pain models and the potential side-effects were investigated at the spinal level in mice. In the tail-flick test, intrathecal injection of DN-9 produced potent analgesia with an ED50 value at 1.33 pmol, and the spinal antinociception of DN-9 was mainly mediated by μ- and κ-opioid receptors. In addition, DN-9-induced spinal antinociception was augmented by the neuropeptide FF receptors antagonist. Furthermore, DN-9 could decrease both the frequency and amplitude of sEPSCs in lamina IIo neurons of the spinal cord, which were mediated by opioid receptors. In contrast to morphine, chronic intrathecal treatments with DN-9 did not induce analgesic tolerance, c-Fos expression or microglial activation. Intrathecal injection of DN-9 showed potent analgesia with antinociceptive ED50 values between .66 and 55.04 pmol in different pain models, including the formalin test, acetic acid-induced writhing test, carrageenan-induced inflammatory pain and neuropathic pain. Moreover, DN-9 did not show side effects in locomotor function and coordination, gastrointestinal transit inhibition, the cardiovascular system, and body temperature regulation at antinociceptive doses. Taken together, the present study showed DN-9 produced effective, nontolerance forming analgesia with reduced side effects at the spinal level. DN-9 might be a promising compound for developing multifunctional opioid analgesics with limited adverse effects. PERSPECTIVE: This article presents the potent and nontolerance forming analgesia effects of DN-9 in a series of preclinical pain models with less opioid related adverse effects at the spinal level in mice. This study also demonstrates that DN-9 has translational potential into an intrathecal analgesic.
Collapse
|
15
|
Discovery of two novel branched peptidomimetics containing endomorphin-2 and RF9 pharmacophores: Synthesis and neuropharmacological evaluation. Bioorg Med Chem 2019; 27:630-643. [DOI: 10.1016/j.bmc.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/25/2018] [Accepted: 01/03/2019] [Indexed: 11/23/2022]
|
16
|
Li N, Han ZL, Xu B, Zhang MN, Zhang T, Shi XR, Zhao WD, Guo YY, Zhang QQ, Fang Q. Systemic administration of the bifunctional opioid/neuropeptide FF receptors agonist BN-9 produced peripheral antinociception in preclinical mouse models of pain. Eur J Pharmacol 2018; 837:53-63. [DOI: 10.1016/j.ejphar.2018.08.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 01/10/2023]
|
17
|
Wang Q, Hong X, Chen H, Yuan L, Zha J. The neuropeptides of Asian freshwater clam (Corbicula fluminea) as new molecular biomarker basing on the responses of organophosphate chemicals exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 160:52-59. [PMID: 29783112 DOI: 10.1016/j.ecoenv.2018.05.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 06/08/2023]
Abstract
In the present study, to discover new biomarker of Asian freshwater clam (Corbicula fluminea) to assess impact of environmental pollutions, cholecystokinin (CCK), conopressin, and Neuropeptide FF (FFamide) in C. fluminea were selected as potent biomarkers. Therefore, their full-length cDNAs were cloned and characterized to investigate the molecular characteristics and expression patterns of neuropeptides in C. fluminea. According to the sequence analysis, CCK, conopressin, and FFamide encoded proteins of 173, 152, and 90 amino acids, respectively. Moreover, the multiple sequence alignment revealed that the bioactive regions of these neuropeptides were well conserved among different invertebrates. In addition, under basal conditions, CCK, conopressin and FFamide mRNA were mainly expressed in the visceral mass, whereas the FFamide mRNA was rarely detected in the foot and mantle. Exposure to 20 and 200 μg/L Tris (2-butoxyethyl) phosphate (TBOEP) and tri-butyl-phosphate (TBP) exposure significantly up-regulated the expression of the CCK and FFamide mRNAs in the visceral mass (p < 0.05), whereas no significant changes in conopressin mRNA levels were observed in response to any treatment. Therefore, CCK and FFamide of C. fluminea neuropeptides are feasible new biomarkers for screening and assessing responses to organophosphate chemicals.
Collapse
Affiliation(s)
- Qi Wang
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Xiangsheng Hong
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Huihui Chen
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
| | - Lilai Yuan
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
| | - Jinmiao Zha
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
18
|
Nourbakhsh F, Atabaki R, Roohbakhsh A. The role of orphan G protein-coupled receptors in the modulation of pain: A review. Life Sci 2018; 212:59-69. [PMID: 30236869 DOI: 10.1016/j.lfs.2018.09.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/04/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022]
Abstract
G protein-coupled receptors (GPCRs) comprise a large number of receptors. Orphan GPCRs are divided into six families. These groups contain orphan receptors for which the endogenous ligands are unclear. They have various physiological effects in the body and have the potential to be used in the treatment of different diseases. Considering their important role in the central and peripheral nervous system, their role in the treatment of pain has been the subject of some recent studies. At present, there are effective therapeutics for the treatment of pain including opioid medications and non-steroidal anti-inflammatory drugs. However, the side effects of these drugs and the risks of tolerance and dependence remain a major problem. In addition, neuropathic pain is a condition that does not respond to currently available analgesic medications well. In the present review article, we aimed to review the most recent findings regarding the role of orphan GPCRs in the treatment of pain. Accordingly, based on the preclinical findings, the role of GPR3, GPR7, GPR8, GPR18, GPR30, GPR35, GPR40, GPR55, GPR74, and GPR147 in the treatment of pain was discussed. The present study highlights the role of orphan GPCRs in the modulation of pain and implies that these receptors are potential new targets for finding better and more efficient therapeutics for the management of pain particularly neuropathic pain.
Collapse
Affiliation(s)
- Fahimeh Nourbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rabi Atabaki
- Rayan Center for Neuroscience & Behavior, Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
|
20
|
Nguyen T, Decker AM, Langston TL, Mathews KM, Siemian JN, Li JX, Harris DL, Runyon SP, Zhang Y. Discovery of Novel Proline-Based Neuropeptide FF Receptor Antagonists. ACS Chem Neurosci 2017; 8:2290-2308. [PMID: 28737888 DOI: 10.1021/acschemneuro.7b00219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The neuropeptide FF (NPFF) system has been implicated in a number of physiological processes including modulating the pharmacological activity of opioid analgesics and several other classes of drugs of abuse. In this study, we report the discovery of a novel proline scaffold with antagonistic activity at the NPFF receptors through a high throughput screening campaign using a functional calcium mobilization assay. Focused structure-activity relationship studies on the initial hit 1 have resulted in several analogs with calcium mobilization potencies in the submicromolar range and modest selectivity for the NPFF1 receptor. Affinities and potencies of these compounds were confirmed in radioligand binding and functional cAMP assays. Two compounds, 16 and 33, had good solubility and blood-brain barrier permeability that fall within the range of CNS permeant candidates without the liability of being a P-glycoprotein substrate. Finally, both compounds reversed fentanyl-induced hyperalgesia in rats when administered intraperitoneally. Together, these results point to the potential of these proline analogs as promising NPFF receptor antagonists.
Collapse
Affiliation(s)
- Thuy Nguyen
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M. Decker
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Tiffany L. Langston
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Kelly M. Mathews
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Justin N. Siemian
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14214, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14214, United States
| | - Danni L. Harris
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Scott P. Runyon
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
21
|
Zhang R, Xu B, Zhang MN, Zhang T, Wang ZL, Zhao G, Zhao GH, Li N, Fang Q, Wang R. Peripheral and central sites of action for anti-allodynic activity induced by the bifunctional opioid/NPFF receptors agonist BN-9 in inflammatory pain model. Eur J Pharmacol 2017; 813:122-129. [DOI: 10.1016/j.ejphar.2017.07.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 07/04/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
|
22
|
Lin YT, Yu YL, Hong WC, Yeh TS, Chen TC, Chen JC. NPFFR2 Activates the HPA Axis and Induces Anxiogenic Effects in Rodents. Int J Mol Sci 2017; 18:ijms18081810. [PMID: 28825666 PMCID: PMC5578197 DOI: 10.3390/ijms18081810] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 01/01/2023] Open
Abstract
Neuropeptide FF (NPFF) belongs to the RFamide family and is known as a morphine-modulating peptide. NPFF regulates various hypothalamic functions through two receptors, NPFFR1 and NPFFR2. The hypothalamic-pituitary-adrenal (HPA) axis participates in physiological stress response by increasing circulating glucocorticoid levels and modulating emotional responses. Other RFamide peptides, including neuropeptide AF, neuropeptide SF and RFamide related peptide also target NPFFR1 or NPFFR2, and have been reported to activate the HPA axis and induce anxiety- or depression-like behaviors. However, little is known about the action of NPFF on HPA axis activity and anxiety-like behaviors, and the role of the individual receptors remains unclear. In this study, NPFFR2 agonists were used to examine the role of NPFFR2 in activating the HPA axis in rodents. Administration of NPFFR2 agonists, dNPA (intracerebroventricular, ICV) and AC-263093 (intraperitoneal, IP), time-dependently (in rats) and dose-dependently (in mice) increased serum corticosteroid levels and the effects were counteracted by the NPFF receptor antagonist, RF9 (ICV), as well as corticotropin-releasing factor (CRF) antagonist, α-helical CRF(9-41) (intravenous, IV). Treatment with NPFFR2 agonist (AC-263093, IP) increased c-Fos protein expression in the hypothalamic paraventricular nucleus and induced an anxiogenic effect, which was evaluated in mice using an elevated plus maze. These findings reveal, for the first time, that the direct action of hypothalamic NPFFR2 stimulates the HPA axis and triggers anxiety-like behaviors.
Collapse
Affiliation(s)
- Ya-Tin Lin
- Graduate Institute of Biomedical Sciences, Department of Physiology and Pharmacology, Chang Gung University, No. 259 Wenhwa 1st Road, Guishan, Taoyuan 333, Taiwan.
| | - Yu-Lian Yu
- Department of Biomedical Sciences, Chang Gung University, Taoyuan 333, Taiwan.
| | - Wei-Chen Hong
- Department of Biomedical Sciences, Chang Gung University, Taoyuan 333, Taiwan.
| | - Ting-Shiuan Yeh
- Department of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
| | - Ting-Chun Chen
- Department of Biomedical Sciences, Chang Gung University, Taoyuan 333, Taiwan.
| | - Jin-Chung Chen
- Graduate Institute of Biomedical Sciences, Department of Physiology and Pharmacology, Chang Gung University, No. 259 Wenhwa 1st Road, Guishan, Taoyuan 333, Taiwan.
- Neuroscience Research Center, Chang Gung Memorial Hospital, No. 5, Fusing St., Guishan, Taoyuan 333, Taiwan.
- Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
23
|
Lin YT, Liu HL, Day YJ, Chang CC, Hsu PH, Chen JC. Activation of NPFFR2 leads to hyperalgesia through the spinal inflammatory mediator CGRP in mice. Exp Neurol 2017; 291:62-73. [DOI: 10.1016/j.expneurol.2017.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/25/2017] [Accepted: 02/01/2017] [Indexed: 01/22/2023]
|
24
|
Wang ZL, Pan JX, Song JJ, Tang HH, Yu HP, Li XH, Li N, Zhang T, Zhang R, Zhang MN, Xu B, Fang Q, Wang R. Structure-Based Optimization of Multifunctional Agonists for Opioid and Neuropeptide FF Receptors with Potent Nontolerance Forming Analgesic Activities. J Med Chem 2016; 59:10198-10208. [PMID: 27798836 DOI: 10.1021/acs.jmedchem.6b01181] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- 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
| | - Jia-Xin Pan
- 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
| | - Jing-Jing Song
- 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
| | - 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
| | - Xu-Hui 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
| | - 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
| | - 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
| | - 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
|
25
|
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
|
26
|
Wang ZL, Li N, Wang P, Tang HH, Han ZL, Song JJ, Li XH, Yu HP, Zhang T, Zhang R, Xu B, Zhang MN, Fang Q, Wang R. Pharmacological characterization of EN-9, a novel chimeric peptide of endomorphin-2 and neuropeptide FF that produces potent antinociceptive activity and limited tolerance. Neuropharmacology 2016; 108:364-72. [DOI: 10.1016/j.neuropharm.2016.03.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/08/2016] [Accepted: 03/08/2016] [Indexed: 01/19/2023]
|
27
|
Roeckel LA, Le Coz GM, Gavériaux-Ruff C, Simonin F. Opioid-induced hyperalgesia: Cellular and molecular mechanisms. Neuroscience 2016; 338:160-182. [PMID: 27346146 DOI: 10.1016/j.neuroscience.2016.06.029] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/10/2016] [Accepted: 06/16/2016] [Indexed: 12/18/2022]
Abstract
Opioids produce strong analgesia but their use is limited by a paradoxical hypersensitivity named opioid-induced hyperalgesia (OIH) that may be associated to analgesic tolerance. In the last decades, a significant number of preclinical studies have investigated the factors that modulate OIH development as well as the cellular and molecular mechanisms underlying OIH. Several factors have been shown to influence OIH including the genetic background and sex differences of experimental animals as well as the opioid regimen. Mu opioid receptor (MOR) variants and interactions of MOR with different proteins were shown important. Furthermore, at the cellular level, both neurons and glia play a major role in OIH development. Several neuronal processes contribute to OIH, like activation of neuroexcitatory mechanisms, long-term potentiation (LTP) and descending pain facilitation. Increased nociception is also mediated by neuroinflammation induced by the activation of microglia and astrocytes. Neurons and glial cells exert synergistic effects, which contribute to OIH. The molecular actors identified include the Toll-like receptor 4 and the anti-opioid systems as well as some other excitatory molecules, receptors, channels, chemokines, pro-inflammatory cytokines or lipids. This review summarizes the intracellular and intercellular pathways involved in OIH and highlights some mechanisms that may be challenged to limit OIH in the future.
Collapse
Affiliation(s)
- Laurie-Anne Roeckel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Glenn-Marie Le Coz
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Claire Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Ecole Supérieure de Biotechnologie de Strasbourg, Université de Strasbourg, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
28
|
Li N, Han ZL, Wang ZL, Xing YH, Sun YL, Li XH, Song JJ, Zhang T, Zhang R, Zhang MN, Xu B, Fang Q, Wang R. BN-9, a chimeric peptide with mixed opioid and neuropeptide FF receptor agonistic properties, produces nontolerance-forming antinociception in mice. Br J Pharmacol 2016; 173:1864-80. [PMID: 27018797 DOI: 10.1111/bph.13489] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE Neuropeptide FF (NPFF) behaves as an endogenous opioid-modulating peptide. In the present study, the opioid and NPFF pharmacophore-containing chimeric peptide BN-9 was synthesized and pharmacologically characterized. EXPERIMENTAL APPROACH Agonist activities of BN-9 at opioid and NPFF receptors were characterized in in vitro cAMP assays. Antinociceptive activities of BN-9 were evaluated in the mouse tail-flick and formalin tests. Furthermore, its side effects were investigated in rotarod, antinociceptive tolerance, reward and gastrointestinal transit tests. KEY RESULTS BN-9 acted as a novel multifunctional agonist at μ, δ, κ, NPFF1 and NPFF2 receptors in cAMP assays. In the tail-flick test, BN-9 produced dose-related antinociception and was approximately equipotent to morphine; this antinociception was blocked by μ and κ receptor antagonists, but not by the δ receptor antagonist. In the formalin test, supraspinal administration of BN-9 produced significant analgesia. Notably, repeated administration of BN-9 produced analgesia without loss of potency over 8 days. In contrast, repeated i.c.v. co-administration of BN-9 with the NPFF receptor antagonist RF9 produced significant antinociceptive tolerance. Furthermore, i.c.v. BN-9 induced conditioned place preference. When given by the same routes, BN-9 had a more than eightfold higher ED50 value for gastrointestinal transit inhibition compared with the ED50 values for antinociception. CONCLUSIONS AND IMPLICATIONS BN-9 produced a robust, nontolerance-forming analgesia with limited inhibition of gastrointestinal transit. As BN-9 is able to activate both opioid and NPFF systems, this provides an interesting approach for the development of novel analgesics with minimal side effects.
Collapse
Affiliation(s)
- Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Zheng-Lan Han
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 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, Lanzhou, China
| | - Yan-Hong Xing
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Long Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xu-Hui Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing-Jing Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 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, Lanzhou, 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, Lanzhou, 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, Lanzhou, 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, Lanzhou, 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, Lanzhou, 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, Lanzhou, China
| |
Collapse
|
29
|
Mudgal A, Kumar K, Mollereau C, Pasha S. NPYFa, A Chimeric Peptide of Met-Enkephalin, and NPFF Induces Tolerance-Free Analgesia. Chem Biol Drug Des 2016; 87:885-94. [DOI: 10.1111/cbdd.12721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/30/2015] [Accepted: 12/30/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Annu Mudgal
- Peptide Synthesis Laboratory; CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB); New Delhi India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IGIB Campus; New Delhi India
| | - Krishan Kumar
- Peptide Synthesis Laboratory; CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB); New Delhi India
- Department of Chemistry; Motilal Nehru College; University of Delhi; Delhi 110021 India
| | - Catherine Mollereau
- Institut de Pharmacologie et Biologie Structurale; Toulouse Cedex France
- Laboratoire Anthropologie Moléculaire et Imagerie de Synthèse; Toulouse Cedex France
| | - Santosh Pasha
- Peptide Synthesis Laboratory; CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB); New Delhi India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IGIB Campus; New Delhi India
| |
Collapse
|
30
|
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
|
31
|
Ding Z, Zajac JM. Cholesterol-rich lipid rafts are involved in neuropeptide FF anti-nociceptin/orphanin FQ effect. J Neurochem 2015; 136:778-790. [PMID: 26617404 DOI: 10.1111/jnc.13450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/21/2015] [Accepted: 11/18/2015] [Indexed: 11/29/2022]
Abstract
The participation of a signaling platform to the anti-nociceptin/orphanin FQ (N/OFQ) effect of neuropeptide FF (NPFF) receptors was investigated in both acutely dissociated neurons and SH-SY5Y human neuroblastoma cells. The NPFF anti-N/OFQ, not anti-μ opioid effect, on the Ca2+ transient triggered by depolarization was reversed by methyl-β-cyclodextrin which depletes cholesterol from cell membranes. While the inactive α-cyclodextrin had no effect. By using [35 S]GTPγS binding assay, a significant 20% decrease in the activity of nociceptin/orphanin FQ peptide receptors induced by the NPFF analog 1DMe was observed in detergent-resistant membranes, but not in total membranes of SH-SY5Y cells. Moreover, siRNA knock-down of G-protein-coupled receptor kinase 2 indicated that G-protein-coupled receptor kinase 2, but not protein kinase C, acted as the mediator in the NPFF anti-N/OFQ process. These data indicate that cholesterol-rich lipid rafts play an important role in the anti-N/OFQ effect of NPFF receptors. We proposed the participation of a signaling platform to the anti-Nociceptin/Orphanin FQ (N/OFQ) effect of Neuropeptide FF (NPFF) receptors both in mouse neurons and SH-SY5Y cells, with GRK2 protein acting as the mediator in this process. These findings should provide a more precise way to understand the anti-opioid effect of NPFF. NOP, Nociceptin/Orphanin FQ peptide.
Collapse
Affiliation(s)
- Zhong Ding
- Institut de Pharmacologie et de Biologie Structurale, CNRS / Université de Toulouse, Toulouse Cedex, France
| | - Jean-Marie Zajac
- Institut de Pharmacologie et de Biologie Structurale, CNRS / Université de Toulouse, Toulouse Cedex, France
| |
Collapse
|
32
|
Mudgal A, Pasha S. Role of opioid receptor heterodimerization in pain modulation and tolerance development. World J Pharmacol 2015; 4:144-159. [DOI: 10.5497/wjp.v4.i1.144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/09/2014] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
Protein to protein interactions leading to homo/heteromerization of receptor is well documented in literature. These interactions leading to dimeric/oligomers formation of receptors are known to modulate their function, particularly in case of G-protein coupled receptors. The opioid receptor heteromers having changed pharmacological properties than the constituent protomers provides preferences for novel drug targets that could lead to potential analgesic activity devoid of tolerance and physical dependence. Heterodimerization of opioid receptors appears to generate novel binding properties with improved specificity and lack of side effects. Further the molecules which can interact simultaneously to both the protomers of the heteromer, or to both the binding sites (orthosteric and allosteric) of a receptor protein could be potential therapeutic molecules. This review highlights the recent advancements in exploring the plausible role of heteromerization of opioid receptors in induction of tolerance free antinociception.
Collapse
|
33
|
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]
|
34
|
Tsutsui K, Ubuka T, Son YL, Bentley GE, Kriegsfeld LJ. Contribution of GnIH Research to the Progress of Reproductive Neuroendocrinology. Front Endocrinol (Lausanne) 2015; 6:179. [PMID: 26635728 PMCID: PMC4655308 DOI: 10.3389/fendo.2015.00179] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/05/2015] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of gonadotropin-releasing hormone (GnRH) in mammals at the beginning of the 1970s, it was generally accepted that GnRH is the only hypothalamic neuropeptide regulating gonadotropin release in mammals and other vertebrates. In 2000, however, gonadotropin-inhibitory hormone (GnIH), a novel hypothalamic neuropeptide that actively inhibits gonadotropin release, was discovered in quail. Numerous studies over the past decade and a half have demonstrated that GnIH serves as a key player regulating reproduction across vertebrates, acting on the brain and pituitary to modulate reproductive physiology and behavior. In the latter case, recent evidence indicates that GnIH can regulate reproductive behavior through changes in neurosteroid, such as neuroestrogen, biosynthesis in the brain. This review summarizes the discovery of GnIH, and the contributions to GnIH research focused on its mode of action, regulation of biosynthesis, and how these findings advance our understanding of reproductive neuroendocrinology.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan
- *Correspondence: Kazuyoshi Tsutsui,
| | - Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan
- Brain Research Institute Monash Sunway of the Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - You Lee Son
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan
| | - George E. Bentley
- Department of Integrative Biology, Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Lance J. Kriegsfeld
- Department of Psychology, Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA, USA
| |
Collapse
|
35
|
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
|
36
|
Ding Z, Zajac JM. Neuropeptide FF receptors exhibit direct and anti-opioid effects on mice dorsal raphe nucleus neurons. Eur J Pharmacol 2014; 740:271-6. [PMID: 25046840 DOI: 10.1016/j.ejphar.2014.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/04/2014] [Accepted: 07/09/2014] [Indexed: 11/25/2022]
Abstract
By using acutely dissociated dorsal raphe nucleus neurons (DRN) from young mice, direct and anti-opioid effects of Neuropeptide FF (NPFF) receptors were measured. The NPFF analog 1 DMe (10 µM) had no effect on resting Ca2+ channels but reduced the magnitude of Ca2+ transients induced by depolarization in 83.3% neurons tested, of which the inhibition rate is 45.4±2.9%. Pertussis toxin treatment reduced to 18.9% the number of responding neurons and attenuated by 47% the response of 1 DMe. In contrast, cholera toxin treatment had no significant effect. Eighteen minute perfusion with 1 DMe at a very low 10 nM concentration, that did not directly inhibit Ca2+ transients triggered by depolarization in every neuron, attenuated by 78% the inhibitory effect of Nociceptin/orphanin FQ (N/OFQ) on Ca2+ transients, but not that of by serotonin. These results demonstrated for the first time that NPFF receptors on mice DRN inhibit Ca2+ transients induced by depolarization via Gi/o protein and also exhibit a specific anti-opioid activity on nociceptin receptors, and that their specific anti-opioid activity is not a direct consequence of their activity on Ca2+ transients.
Collapse
Affiliation(s)
- Zhong Ding
- Institut de Pharmacologie et de Biologie Structurale, CNRS / Université de Toulouse, UMR 5089, 205 route de Narbonne, 31077 Toulouse Cedex, France
| | - Jean-Marie Zajac
- Institut de Pharmacologie et de Biologie Structurale, CNRS / Université de Toulouse, UMR 5089, 205 route de Narbonne, 31077 Toulouse Cedex, France.
| |
Collapse
|
37
|
Carayon K, Moulédous L, Combedazou A, Mazères S, Haanappel E, Salomé L, Mollereau C. Heterologous regulation of Mu-opioid (MOP) receptor mobility in the membrane of SH-SY5Y cells. J Biol Chem 2014; 289:28697-706. [PMID: 25183007 DOI: 10.1074/jbc.m114.588558] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The dynamic organization of G protein-coupled receptors in the plasma membrane is suspected of playing a role in their function. The regulation of the diffusion mode of the mu-opioid (MOP) receptor was previously shown to be agonist-specific. Here we investigate the regulation of MOP receptor diffusion by heterologous activation of other G protein-coupled receptors and characterize the dynamic properties of the MOP receptor within the heterodimer MOP/neuropeptide FF (NPFF2) receptor. The data show that the dynamics and signaling of the MOP receptor in SH-SY5Y cells are modified by the activation of α2-adrenergic and NPFF2 receptors, but not by the activation of receptors not described to interact with the opioid receptor. By combining, for the first time, fluorescence recovery after photobleaching at variable radius experiments with bimolecular fluorescence complementation, we show that the MOP/NPFF2 heterodimer adopts a specific diffusion behavior that corresponds to a mix of the dynamic properties of both MOP and NPFF2 receptors. Altogether, the data suggest that heterologous regulation is accompanied by a specific organization of receptors in the membrane.
Collapse
Affiliation(s)
- Kévin Carayon
- 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
| | - Anne Combedazou
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS/Université de Toulouse, 31077 Toulouse, France
| | - Serge Mazères
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS/Université de Toulouse, 31077 Toulouse, France
| | - Evert Haanappel
- From the Institut de Pharmacologie et Biologie Structurale, UMR5089 CNRS/Université de Toulouse, 31077 Toulouse, France
| | - Laurence Salomé
- 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
| |
Collapse
|
38
|
Wang ZL, Fang Q, Han ZL, Pan JX, Li XH, Li N, Tang HH, Wang P, Zheng T, Chang XM, Wang R. Opposite effects of neuropeptide FF on central antinociception induced by endomorphin-1 and endomorphin-2 in mice. PLoS One 2014; 9:e103773. [PMID: 25090615 PMCID: PMC4121275 DOI: 10.1371/journal.pone.0103773] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 07/02/2014] [Indexed: 11/18/2022] Open
Abstract
Neuropeptide FF (NPFF) is known to be an endogenous opioid-modulating peptide. Nevertheless, very few researches focused on the interaction between NPFF and endogenous opioid peptides. In the present study, we have investigated the effects of NPFF system on the supraspinal antinociceptive effects induced by the endogenous µ-opioid receptor agonists, endomorphin-1 (EM-1) and endomorphin-2 (EM-2). In the mouse tail-flick assay, intracerebroventricular injection of EM-1 induced antinociception via µ-opioid receptor while the antinociception of intracerebroventricular injected EM-2 was mediated by both µ- and κ-opioid receptors. In addition, central administration of NPFF significantly reduced EM-1-induced central antinociception, but enhanced EM-2-induced central antinociception. The results using the selective NPFF1 and NPFF2 receptor agonists indicated that the EM-1-modulating action of NPFF was mainly mediated by NPFF2 receptor, while NPFF potentiated EM-2-induecd antinociception via both NPFF1 and NPFF2 receptors. To further investigate the roles of µ- and κ-opioid systems in the opposite effects of NPFF on central antinociception of endomprphins, the µ- and κ-opioid receptors selective agonists DAMGO and U69593, respectively, were used. Our results showed that NPFF could reduce the central antinociception of DAMGO via NPFF2 receptor and enhance the central antinociception of U69593 via both NPFF1 and NPFF2 receptors. Taken together, our data demonstrate that NPFF exerts opposite effects on central antinociception of endomorphins and provide the first evidence that NPFF potentiate antinociception of EM-2, which might result from the interaction between NPFF and κ-opioid systems.
Collapse
Affiliation(s)
- 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, Lanzhou, 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, Lanzhou, PR China
| | - Zheng-lan Han
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Jia-xin Pan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Xu-hui Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 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, Lanzhou, 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, Lanzhou, PR China
| | - Pei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Ting Zheng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Xue-mei Chang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 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, Lanzhou, PR China
- * E-mail:
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Endogenous mammalian RF-amide peptides, including PrRP, kisspeptin and 26RFa, modulate nociception and morphine analgesia via NPFF receptors. Neuropharmacology 2013; 75:164-71. [PMID: 23911743 DOI: 10.1016/j.neuropharm.2013.07.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 11/23/2022]
Abstract
Mammalian RF-amide peptides are encoded by five different genes and act through five different G protein-coupled receptors. RF-amide-related peptides-1 and -3, neuropeptides AF and FF, Prolactin releasing peptides, Kisspeptins and RFa peptides are currently considered endogenous peptides for NPFF1, NPFF2, GPR10, GPR54 and GPR103 receptors, respectively. However, several studies suggest that the selectivity of these peptides for their receptors is low and indicate that expression patterns for receptors and their corresponding ligands only partially overlap. In this study, we took advantage of the cloning of the five human RF-amide receptors to systematically examine their affinity for and their activation by all human RF-amide peptides. Binding experiments, performed on membranes from CHO cells expressing GPR10, GPR54 and GPR103 receptors, confirmed their high affinity and remarkable selectivity for their cognate ligands. Conversely, NPFF1 and NPFF2 receptors displayed high affinity for all RF-amide peptides. Moreover, GTPγS and cAMP experiments showed that almost all RF-amide peptides efficiently activate NPFF1 and NPFF2 receptors. As NPFF is known to modulate morphine analgesia, we undertook a systematic analysis in mice of the hyperalgesic and anti morphine-induced analgesic effects of a representative set of endogenous RF-amide peptides. All of them induced hyperalgesia and/or prevented morphine analgesia following intracerebroventricular administration. Importantly, these effects were prevented by administration of RF9, a highly selective NPFF1/NPFF2 antagonist. Altogether, our results show that all endogenous RF-amide peptides display pain-modulating properties and point to NPFF receptors as essential players for these effects.
Collapse
|
43
|
Neuropeptide FF analog RF9 is not an antagonist of NPFF receptor and decreases food intake in mice after its central and peripheral administration. Brain Res 2013; 1498:33-40. [DOI: 10.1016/j.brainres.2012.12.037] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 10/09/2012] [Accepted: 12/13/2012] [Indexed: 11/18/2022]
|
44
|
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
|
45
|
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
|
46
|
Mazarguil H, Mollereau C, Czaplicki G, Zajac JM. Study of the N-terminal part of peptidic selective NPFF₂ agonists. Peptides 2012; 37:157-60. [PMID: 22813580 DOI: 10.1016/j.peptides.2012.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/09/2012] [Accepted: 07/09/2012] [Indexed: 10/28/2022]
Abstract
Neuropeptide FF (NPFF) has been shown to act as an endogenous anti-analgesic peptide. In this paper, several peptide analogs of the selective ligand dNP(NMe)AFLFQPQRF-NH(2) modified in the putative address segment, were designed to be selective NPFF(2) receptor probes, synthesized and assayed. One peptide dA(NMe)AAFLFQPQRF-NH(2) displays a very high affinity for NPFF(2) receptors transfected in CHO cells, and a high selectivity versus NPFF(1) receptors. The exact residues carried in the N-terminal part of the ligands are not decisive to obtain a high affinity only the length of the peptide in itself seems important to create selectivity.
Collapse
Affiliation(s)
- Honoré Mazarguil
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | | | | | | |
Collapse
|
47
|
Neuropeptide FF and related peptides attenuates warm-, but not cold-water swim stress-induced analgesia in mice. Behav Brain Res 2012; 233:428-33. [DOI: 10.1016/j.bbr.2012.05.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/18/2012] [Accepted: 05/22/2012] [Indexed: 11/23/2022]
|
48
|
Podwysocka D, Kosson P, Lipkowski AW, Olma A. TAPP analogs containing β3-homo-amino acids: synthesis and receptor binding. J Pept Sci 2012; 18:556-9. [DOI: 10.1002/psc.2433] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/12/2012] [Accepted: 06/19/2012] [Indexed: 01/15/2023]
Affiliation(s)
- D. Podwysocka
- Institute of Organic Chemistry; Lodz University of Technology; Lodz; Poland
| | - P. Kosson
- Mossakowski Medical Research Centre; Polish Academy of Science; Warsaw; Poland
| | - A. W. Lipkowski
- Mossakowski Medical Research Centre; Polish Academy of Science; Warsaw; Poland
| | - A. Olma
- Institute of Organic Chemistry; Lodz University of Technology; Lodz; Poland
| |
Collapse
|
49
|
Mankus JV, McCurdy CR. Nonpeptide ligands of neuropeptide FF: current status and structural insights. Future Med Chem 2012; 4:1085-92. [PMID: 22709252 PMCID: PMC3606921 DOI: 10.4155/fmc.12.67] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Neuropeptide FF (NPFF) was first isolated from the bovine brain in 1985 and is linked with a variety of biological activities. NPFF, which belongs to the RF-amide family of peptides, interacts with two distinct G-protein-coupled receptors, NPFF(1) and NPFF(2). These receptors are distributed throughout the body. The NPFF system was initially described as an anti-opioid system and, while the NPFF system does affect the opioid system, it also has been implicated in pain modulation, changes in arterial blood pressure, feeding behavior and regulation of core body temperature and of monoamine systems. Most of this pharmacology has been realized from the peptide NPFF itself or through peptide analogs. The quest for nonpeptide tools for this receptor system has been limited by low selectivity and poor pharmacokinetic properties. Herein, we summarize the current knowledge from the scientific and patent literature that demonstrates a clear need for future medicinal chemistry efforts.
Collapse
Affiliation(s)
- Jessica V Mankus
- Department of Medicinal Chemistry, 419 Faser Hall, The University of Mississippi, MS 38677, USA
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, 419 Faser Hall, The University of Mississippi, MS 38677, USA
| |
Collapse
|
50
|
Moulédous L, Froment C, Dauvillier S, Burlet-Schiltz O, Zajac JM, Mollereau C. GRK2 protein-mediated transphosphorylation contributes to loss of function of μ-opioid receptors induced by neuropeptide FF (NPFF2) receptors. J Biol Chem 2012; 287:12736-49. [PMID: 22375000 PMCID: PMC3339982 DOI: 10.1074/jbc.m111.314617] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 01/13/2012] [Indexed: 01/25/2023] Open
Abstract
Neuropeptide FF (NPFF) interacts with specific receptors to modulate opioid functions in the central nervous system. On dissociated neurons and neuroblastoma cells (SH-SY5Y) transfected with NPFF receptors, NPFF acts as a functional antagonist of μ-opioid (MOP) receptors by attenuating the opioid-induced inhibition of calcium conductance. In the SH-SY5Y model, MOP and NPFF(2) receptors have been shown to heteromerize. To understand the molecular mechanism involved in the anti-opioid activity of NPFF, we have investigated the phosphorylation status of the MOP receptor using phospho-specific antibody and mass spectrometry. Similarly to direct opioid receptor stimulation, activation of the NPFF(2) receptor by [D-Tyr-1-(NMe)Phe-3]NPFF (1DMe), an analog of NPFF, induced the phosphorylation of Ser-377 of the human MOP receptor. This heterologous phosphorylation was unaffected by inhibition of second messenger-dependent kinases and, contrarily to homologous phosphorylation, was prevented by inactivation of G(i/o) proteins by pertussis toxin. Using siRNA knockdown we could demonstrate that 1DMe-induced Ser-377 cross-phosphorylation and MOP receptor loss of function were mediated by the G protein receptor kinase GRK2. In addition, mass spectrometric analysis revealed that the phosphorylation pattern of MOP receptors was qualitatively similar after treatment with the MOP agonist Tyr-D-Ala-Gly (NMe)-Phe-Gly-ol (DAMGO) or after treatment with the NPFF agonist 1DMe, but the level of multiple phosphorylation was more intense after DAMGO. Finally, NPFF(2) receptor activation was sufficient to recruit β-arrestin2 to the MOP receptor but not to induce its internalization. These data show that NPFF-induced heterologous desensitization of MOP receptor signaling is mediated by GRK2 and could involve transphosphorylation within the heteromeric receptor complex.
Collapse
Affiliation(s)
- Lionel Moulédous
- From the Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Carine Froment
- From the Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Stéphanie Dauvillier
- From the Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Odile Burlet-Schiltz
- From the Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Jean-Marie Zajac
- From the Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Catherine Mollereau
- From the Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| |
Collapse
|