1
|
Wu Z, Qiu C, Liu Y, Yan X, Li Q, Jiang S, Xu J, Pan X, Ye F, Zhang Z, Ning P, Zhang B, Xu L, Cheng B, Xiang X, Qian C, Du Y, Chen G. Structural insights into prolactin-releasing peptide receptor signaling and G-protein coupling selectivity. Cell Rep 2025; 44:115337. [PMID: 39977266 DOI: 10.1016/j.celrep.2025.115337] [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: 11/04/2024] [Revised: 12/21/2024] [Accepted: 01/29/2025] [Indexed: 02/22/2025] Open
Abstract
Prolactin-releasing peptide receptor (PrRPR), a notable member of the class A peptide-GPCR (G-protein-coupled receptor) family, regulates diverse physiology functions upon activation by PrRP. Herein, we reveal that PrRPR could engage with not only the Gq/11 pathway but also the Gi/o pathway. We further resolve the structures of the PrRPR-Gq and PrRPR-Gi complexes using cryoelectron microscopy (cryo-EM), with PrRP31 as the endogenous ligand. These high-resolution structures enhance our understanding of PrRPR-ligand interactions, aiding the development of targeted drugs aiming at this crucial peptide-receptor system. Comparing these structures with counterparts of other RF-amide peptide receptors accentuates the crucial function of the RF-amide motif in activating receptors and sheds light on the universal mechanism for RF-amide motif detection by RF-amide receptors. Furthermore, structural and functional analysis indicates that conformational alterations in the intracellular loops (ICLs), along with the "wavy hook" of Gα, may explain the selective coupling of G proteins in PrRPR signaling.
Collapse
Affiliation(s)
- Zhangsong Wu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China; The Huanan Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Chen Qiu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yiming Liu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Xiaoyi Yan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Qiaohui Li
- Biological Science Research Center, Academy for Advanced Interdisciplinary Studies, Southwest University, Chongqing, China
| | - Shirui Jiang
- The Huanan Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Jun Xu
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Xin Pan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Fang Ye
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Zhiyi Zhang
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Peiruo Ning
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Binghao Zhang
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Lezhi Xu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Bangning Cheng
- Department of Reagent Research and Development, Shenzhen YHLO Biotech Co., Ltd., Shenzhen, Guangdong, China
| | - Xufu Xiang
- Department of Reagent Research and Development, Shenzhen YHLO Biotech Co., Ltd., Shenzhen, Guangdong, China
| | - Chungen Qian
- Department of Reagent Research and Development, Shenzhen YHLO Biotech Co., Ltd., Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| |
Collapse
|
2
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
3
|
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: 2.0] [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
|
4
|
Holt MK, Rinaman L. The role of nucleus of the solitary tract glucagon-like peptide-1 and prolactin-releasing peptide neurons in stress: anatomy, physiology and cellular interactions. Br J Pharmacol 2021; 179:642-658. [PMID: 34050926 PMCID: PMC8820208 DOI: 10.1111/bph.15576] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroendocrine, behavioural and autonomic responses to stressful stimuli are orchestrated by complex neural circuits. The caudal nucleus of the solitary tract (cNTS) in the dorsomedial hindbrain is uniquely positioned to integrate signals of both interoceptive and psychogenic stress. Within the cNTS, glucagon‐like peptide‐1 (GLP‐1) and prolactin‐releasing peptide (PrRP) neurons play crucial roles in organising neural responses to a broad range of stressors. In this review we discuss the anatomical and functional overlap between PrRP and GLP‐1 neurons. We outline their co‐activation in response to stressful stimuli and their importance as mediators of behavioural and physiological stress responses. Finally, we review evidence that PrRP neurons are downstream of GLP‐1 neurons and outline unexplored areas of the research field. Based on the current state‐of‐knowledge, PrRP and GLP‐1 neurons may be compelling targets in the treatment of stress‐related disorders.
Collapse
Affiliation(s)
- Marie K Holt
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
5
|
Constantin S, Pizano K, Matson K, Shan Y, Reynolds D, Wray S. An Inhibitory Circuit From Brainstem to GnRH Neurons in Male Mice: A New Role for the RFRP Receptor. Endocrinology 2021; 162:6132086. [PMID: 33564881 PMCID: PMC8016070 DOI: 10.1210/endocr/bqab030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/31/2022]
Abstract
RFamide-related peptides (RFRPs, mammalian orthologs of gonadotropin-inhibitory hormone) convey circadian, seasonal, and social cues to the reproductive system. They regulate gonadotropin secretion by modulating gonadotropin-releasing hormone (GnRH) neurons via the RFRP receptor. Mice lacking this receptor are fertile but exhibit abnormal gonadotropin responses during metabolic challenges, such as acute fasting, when the normal drop in gonadotropin levels is delayed. Although it is known that these food intake signals to the reproductive circuit originate in the nucleus tractus solitarius (NTS) in the brainstem, the phenotype of the neurons conveying the signal remains unknown. Given that neuropeptide FF (NPFF), another RFamide peptide, resides in the NTS and can bind to the RFRP receptor, we hypothesized that NPFF may regulate GnRH neurons. To address this question, we used a combination of techniques: cell-attached electrophysiology on GnRH-driven green fluorescent protein-tagged neurons in acute brain slices; calcium imaging on cultured GnRH neurons; and immunostaining on adult brain tissue. We found (1) NPFF inhibits GnRH neuron excitability via the RFRP receptor and its canonical signaling pathway (Gi/o protein and G protein-coupled inwardly rectifying potassium channels), (2) NPFF-like fibers in the vicinity of GnRH neurons coexpress neuropeptide Y, (3) the majority of NPFF-like cell bodies in the NTS also coexpress neuropeptide Y, and (4) acute fasting increased NPFF-like immunoreactivity in the NTS. Together these data indicate that NPFF neurons within the NTS inhibit GnRH neurons, and thus reproduction, during fasting but prior to the energy deficit.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Katherine Pizano
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Kaya Matson
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Yufei Shan
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Daniel Reynolds
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
- Correspondence: Dr. Susan Wray, Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive MSC 3703, Building 35, Room 3A1012, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Li H, Chen JA, Ding QZ, Lu GY, Wu N, Su RB, Li F, Li J. Behavioral sensitization induced by methamphetamine causes differential alterations in gene expression and histone acetylation of the prefrontal cortex in rats. BMC Neurosci 2021; 22:24. [PMID: 33823794 PMCID: PMC8022387 DOI: 10.1186/s12868-021-00616-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 02/09/2021] [Indexed: 01/01/2023] Open
Abstract
Background Methamphetamine (METH) is one of the most widely abused illicit substances worldwide; unfortunately, its addiction mechanism remains unclear. Based on accumulating evidence, changes in gene expression and chromatin modifications might be related to the persistent effects of METH on the brain. In the present study, we took advantage of METH-induced behavioral sensitization as an animal model that reflects some aspects of drug addiction and examined the changes in gene expression and histone acetylation in the prefrontal cortex (PFC) of adult rats. Methods We conducted mRNA microarray and chromatin immunoprecipitation (ChIP) coupled to DNA microarray (ChIP-chip) analyses to screen and identify changes in transcript levels and histone acetylation patterns. Functional enrichment analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, were performed to analyze the differentially expressed genes. We then further identified alterations in ANP32A (acidic leucine-rich nuclear phosphoprotein-32A) and POU3F2 (POU domain, class 3, transcription factor 2) using qPCR and ChIP-PCR assays. Results In the rat model of METH-induced behavioral sensitization, METH challenge caused 275 differentially expressed genes and a number of hyperacetylated genes (821 genes with H3 acetylation and 10 genes with H4 acetylation). Based on mRNA microarray and GO and KEGG enrichment analyses, 24 genes may be involved in METH-induced behavioral sensitization, and 7 genes were confirmed using qPCR. We further examined the alterations in the levels of the ANP32A and POU3F2 transcripts and histone acetylation at different periods of METH-induced behavioral sensitization. H4 hyperacetylation contributed to the increased levels of ANP32A mRNA and H3/H4 hyperacetylation contributed to the increased levels of POU3F2 mRNA induced by METH challenge-induced behavioral sensitization, but not by acute METH exposure. Conclusions The present results revealed alterations in transcription and histone acetylation in the rat PFC by METH exposure and provided evidence that modifications of histone acetylation contributed to the alterations in gene expression caused by METH-induced behavioral sensitization.
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China
| | - Jing-An Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China
| | - Qian-Zhi Ding
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Guan-Yi Lu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China
| | - Ning Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China
| | - Rui-Bin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China
| | - Fei Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China. .,Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, 100850, Beijing, China.
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, People's Republic of China. .,Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, 100850, Beijing, China.
| |
Collapse
|
7
|
Paciaroni NG, Norwood VM, Ratnayake R, Luesch H, Huigens RW. Yohimbine as a Starting Point to Access Diverse Natural Product-Like Agents with Re-programmed Activities against Cancer-Relevant GPCR Targets. Bioorg Med Chem 2020; 28:115546. [PMID: 32616180 DOI: 10.1016/j.bmc.2020.115546] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 11/29/2022]
Abstract
G protein-coupled receptors (GPCRs) constitute the largest protein superfamily in the human genome. GPCRs play key roles in mediating a wide variety of physiological events including proliferation and cancer metastasis. Given the major roles that GPCRs play in mediating cancer growth, they present promising targets for small molecule therapeutics. One of the principal goals of our lab is to identify complex natural products (NPs) suitable for ring distortion, or the dramatic altering of the inherently complex architectures of NPs, to rapidly generate an array of compounds with diverse molecular skeletal systems. The overarching goal of our ring distortion approach is to re-program the biological activity of select natural products and identify new compounds of importance to the treatment of disease, such as cancer. Described herein are the results from biological screens of diverse small molecules derived from the indole alkaloid yohimbine against a panel of GPCRs involved in various diseases. Several analogues displayed highly differential antagonistic activities across the GPCRs tested. We highlight the re-programmed profile of one analogue, Y7g, which exhibited selective antagonistic activities against AVPR2 (IC50 = 459 nM) and OXTR (IC50 = 1.16 µM). The activity profile of Y7g could correlate its HIF-dependent anti-cancer activity to its GPCR antagonism since these receptors are known to be upregulated in hypoxic cellular environments. Our findings demonstrate that the ring distortion of yohimbine can lead to the identification of new compounds capable of interacting with distinct cancer-relevant targets.
Collapse
Affiliation(s)
- Nicholas G Paciaroni
- University of Florida, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Verrill M Norwood
- University of Florida, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Ranjala Ratnayake
- University of Florida, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Hendrik Luesch
- University of Florida, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, 1345 Center Dr., Gainesville, FL 32610, United States.
| | - Robert W Huigens
- University of Florida, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, 1345 Center Dr., Gainesville, FL 32610, United States.
| |
Collapse
|
8
|
Prolactin-Releasing Peptide: Physiological and Pharmacological Properties. Int J Mol Sci 2019; 20:ijms20215297. [PMID: 31653061 PMCID: PMC6862262 DOI: 10.3390/ijms20215297] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/18/2022] Open
Abstract
Prolactin-releasing peptide (PrRP) belongs to the large RF-amide neuropeptide family with a conserved Arg-Phe-amide motif at the C-terminus. PrRP plays a main role in the regulation of food intake and energy expenditure. This review focuses not only on the physiological functions of PrRP, but also on its pharmacological properties and the actions of its G-protein coupled receptor, GPR10. Special attention is paid to structure-activity relationship studies on PrRP and its analogs as well as to their effect on different physiological functions, mainly their anorexigenic and neuroprotective features and the regulation of the cardiovascular system, pain, and stress. Additionally, the therapeutic potential of this peptide and its analogs is explored.
Collapse
|
9
|
Davis XS, Grill H. The hindbrain is a site of energy balance action for prolactin-releasing peptide: feeding and thermic effects from GPR10 stimulation of the nucleus tractus solitarius/area postrema. Psychopharmacology (Berl) 2018; 235:2287-2301. [PMID: 29796829 PMCID: PMC8019516 DOI: 10.1007/s00213-018-4925-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/07/2018] [Indexed: 01/24/2023]
Abstract
PURPOSE Prolactin-releasing peptide (PrRP) is a neuropeptide that suppresses food intake and increases body temperature when delivered to the forebrain ventricularly or parenchymally. However, PrRP's receptor GPR10 is widely distributed throughout the brain with particularly high levels found in the dorsomedial hindbrain. Thus, we hypothesized that hindbrain-directed PrRP administration would affect energy balance and motivated feeding behavior. METHODS To address this hypothesis, a range of behavioral and physiologic variables were measured in Sprague-Dawley rats that received PrRP delivered to the fourth ventricle (4V) or the nucleus of the solitary tract (NTS) at the level of the area postrema (AP). RESULTS 4V PrRP delivery decreased chow intake and body weight, in part, through decreasing meal size in ad libitum maintained rats tested at dark onset. PrRP inhibited feeding when delivered to the nucleus tractus solitarius (NTS), but not to more ventral hindbrain structures. In addition, 4V as well as direct NTS administration of PrRP increased core temperature. By contrast, 4V PrRP did not reduce ad libitum intake of highly palatable food or the motivation to work for or seek palatable foods. CONCLUSIONS The dorsomedial hindbrain and NTS/AP, in particular, are sites of action in PrRP/GPR10-mediated control of chow intake, core temperature, and body weight.
Collapse
Affiliation(s)
- X. S. Davis
- Department of Psychology, University of Pennsylvania, 433 S. University Avenue, Rm. 327, Philadelphia, PA 19104, USA
| | - H.J. Grill
- Department of Psychology, University of Pennsylvania, 433 S. University Avenue, Rm. 327, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Csabafi K, Bagosi Z, Dobó É, Szakács J, Telegdy G, Szabó G. Kisspeptin modulates pain sensitivity of CFLP mice. Peptides 2018; 105:21-27. [PMID: 29709623 DOI: 10.1016/j.peptides.2018.04.018] [Citation(s) in RCA: 2] [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: 01/24/2018] [Revised: 04/10/2018] [Accepted: 04/27/2018] [Indexed: 01/10/2023]
Abstract
Kisspeptin, a hypothalamic neuropeptide, is a member of the RF-amide family, which have been known to modify pain sensitivity in rodents. The aim of the present study was to investigate the effect of kisspeptin-13 (KP-13), an endogenous derivative of kisspeptin, on nociception in adult male and female CFLP mice and the possible interaction of KP-13 with morphine on nociception. Mice were injected with different doses of KP-13, 30, 60 and 120 min after of which the nociceptive sensitivity were assessed via the tail-flick test. To investigate the receptor involved in the mediation a kisspeptin receptor antagonist (KP-234) pretreatment was applied before KP-13 administration. Furthermore, we investigated the effect of KP-13 on the acute antinociceptive effect of morphine, on acute morphine tolerance and on naloxone-precipitated withdrawal. Last, the Von Frey test was used in order to assess KP-13's effect on mechanical nociception. Our results showed that KP-13 decreased the nociceptive threshold of both males and females independent of sex, which was prevented by KP-234. Furthermore, KP-13 treatment depressed the acute antinociceptive effect of morphine and attenuated the development of morphine tolerance. KP-13 also induced a mechanical hypersensitivity. These data underlie kisspeptin's hyperalgesic action and argues for the role of kisspeptin receptor 1 in the mediation of its action. Furthermore, our results suggest that central KP-13 administration can modify the acute effects of morphine.
Collapse
Affiliation(s)
- Krisztina Csabafi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, 6701, Szeged, Hungary.
| | - Zsolt Bagosi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, 6701, Szeged, Hungary
| | - Éva Dobó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, 6701, Szeged, Hungary
| | - Júlia Szakács
- Department of Pathophysiology, University of Szeged, P.O. Box 427, 6701, Szeged, Hungary
| | - Gyula Telegdy
- Department of Pathophysiology, University of Szeged, P.O. Box 427, 6701, Szeged, Hungary; Neuroscience Research Group of the Hungarian Academy of Sciences, P.O. Box 521, 6701, Szeged, Hungary
| | - Gyula Szabó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, 6701, Szeged, Hungary
| |
Collapse
|
11
|
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: 4.9] [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
|
12
|
Identification of G Protein-Coupled Receptors (GPCRs) in Primary Cilia and Their Possible Involvement in Body Weight Control. PLoS One 2015; 10:e0128422. [PMID: 26053317 PMCID: PMC4459993 DOI: 10.1371/journal.pone.0128422] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/27/2015] [Indexed: 11/30/2022] Open
Abstract
Primary cilia are sensory organelles that harbor various receptors such as G protein-coupled receptors (GPCRs). We analyzed subcellular localization of 138 non-odorant GPCRs. We transfected GPCR expression vectors into NIH3T3 cells, induced ciliogenesis by serum starvation, and observed subcellular localization of GPCRs by immunofluorescent staining. We found that several GPCRs whose ligands are involved in feeding behavior, including prolactin-releasing hormone receptor (PRLHR), neuropeptide FF receptor 1 (NPFFR1), and neuromedin U receptor 1 (NMUR1), localized to the primary cilia. In addition, we found that a short form of dopamine receptor D2 (DRD2S) is efficiently transported to the primary cilia, while a long form of dopamine receptor D2 (DRD2L) is rarely transported to the primary cilia. Using an anti-Prlhr antibody, we found that Prlhr localized to the cilia on the surface of the third ventricle in the vicinity of the hypothalamic periventricular nucleus. We generated the Npy2r-Cre transgenic mouse line in which Cre-recombinase is expressed under the control of the promoter of Npy2r encoding a ciliary GPCR. By mating Npy2r-Cre mice with Ift80 flox mice, we generated Ift80 conditional knockout (CKO) mice in which Npy2r-positive cilia were diminished in number. We found that Ift80 CKO mice exhibited a body weight increase. Our results suggest that Npy2r-positive cilia are important for body weight control.
Collapse
|
13
|
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: 3.1] [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
|
14
|
Ceremuga TE, Shellabarger P, Persson T, Fanning M, Galey P, Robinson D, Bertsch S, Ceremuga GA, Bentley M. Effects of tetrahydropalmatine on post-traumatic stress disorder-induced changes in rat brain gene expression. J Integr Neurosci 2013; 12:513-28. [DOI: 10.1142/s0219635213500313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
15
|
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: 6.8] [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
|
16
|
Loss of the repressor REST in uterine fibroids promotes aberrant G protein-coupled receptor 10 expression and activates mammalian target of rapamycin pathway. Proc Natl Acad Sci U S A 2013; 110:2187-92. [PMID: 23284171 DOI: 10.1073/pnas.1215759110] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Uterine fibroids (leiomyomas) are the most common tumors of the female reproductive tract, occurring in up to 77% of reproductive-aged women, yet molecular pathogenesis remains poorly understood. A role for atypically activated mammalian target of rapamycin (mTOR) pathway in the pathogenesis of uterine fibroids has been suggested in several studies. We identified that G protein-coupled receptor 10 [GPR10, a putative signaling protein upstream of the phosphoinositide 3-kinase-protein kinase B/AKT-mammalian target of rapamycin (PI3K/AKT-mTOR) pathway] is aberrantly expressed in uterine fibroids. The activation of GPR10 by its cognate ligand, prolactin releasing peptide, promotes PI3K-AKT-mTOR pathways and cell proliferation specifically in cultured primary leiomyoma cells. Additionally, we report that RE1 suppressing transcription factor/neuron-restrictive silencing factor (REST/NRSF), a known tumor suppressor, transcriptionally represses GPR10 in the normal myometrium, and that the loss of REST in fibroids permits GPR10 expression. Importantly, mice overexpressing human GPR10 in the myometrium develop myometrial hyperplasia with excessive extracellular matrix deposition, a hallmark of uterine fibroids. We demonstrate previously unrecognized roles for GPR10 and its upstream regulator REST in the pathogenesis of uterine fibroids. Importantly, we report a unique genetically modified mouse model for a gene that is misexpressed in uterine fibroids.
Collapse
|
17
|
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: 1.0] [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
|
18
|
Li M, Zhou L, Ma G, Dong S. Analgesic properties of chimeric peptide based on morphiceptin and PFRTic-amide. ACTA ACUST UNITED AC 2012; 179:23-8. [DOI: 10.1016/j.regpep.2012.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 06/29/2012] [Accepted: 08/27/2012] [Indexed: 11/29/2022]
|
19
|
Rőszer T, Bánfalvi G. FMRFamide-related peptides: anti-opiate transmitters acting in apoptosis. Peptides 2012; 34:177-85. [PMID: 21524675 DOI: 10.1016/j.peptides.2011.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 04/04/2011] [Accepted: 04/07/2011] [Indexed: 01/10/2023]
Abstract
Members of the FMRFamide-related peptide (FaRP) family are neurotransmitters, hormone-like substances and tumor suppressor peptides. In mammals, FaRPs are considered as anti-opiate peptides due to their ability to inhibit opioid signaling. Some FaRPs are asserted to attenuate opiate tolerance. A recently developed chimeric FaRP (Met-enkephalin-FMRFa) mimics the analgesic effects of opiates without the development of opiate-dependence, displaying a future therapeutical potential in pain reduction. In this review we support the notion, that opiates and representative members of the FaRP family show overlapping effects on apoptosis. Binding of FaRPs to opioid receptors or to their own receptors (G-protein linked membrane receptors and acid-sensing ion channels) evokes or suppresses cell death, in a cell- and receptor-type manner. With the dramatically increasing incidence of opiate abuse and addiction, understanding of opioid-induced cell death, and in this context FaRPs will deserve growing attention.
Collapse
Affiliation(s)
- Tamás Rőszer
- Department of Microbial Biotechnology & Cell Biology, University of Debrecen, Debrecen, Hungary.
| | | |
Collapse
|
20
|
Prolactin-releasing peptide enhances synaptic transmission in rat thalamus. Neuroscience 2011; 172:1-11. [DOI: 10.1016/j.neuroscience.2010.10.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 10/14/2010] [Accepted: 10/29/2010] [Indexed: 11/21/2022]
|
21
|
Moulédous L, Mollereau C, Zajac JM. Opioid-modulating properties of the neuropeptide FF system. Biofactors 2010; 36:423-9. [PMID: 20803521 DOI: 10.1002/biof.116] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 06/30/2010] [Indexed: 12/21/2022]
Abstract
Opioid receptors are involved in the control of pain perception in the central nervous system together with endogenous neuropeptides, termed opioid-modulating peptides, participating in a homeostatic system. Neuropeptide FF (NPFF) and related peptides possess anti-opioid properties, the cellular mechanisms of which are still unclear. The purpose of this review is to detail the phenomenon of cross-talk taking place between opioid and NPFF systems at the in vivo pharmacological level and to propose cellular and molecular models of functioning. A better knowledge of the mechanisms underlying opioid-modulating properties of NPFF has potential therapeutic interest for the control of opioid functions, notably for alleviating pain and/or for the treatment of opioid abuse.
Collapse
Affiliation(s)
- Lionel Moulédous
- Institut de Pharmacologie et de Biologie Structurale, CNRS UMR5089, Université de Toulouse, 205 route de Narbonne, Toulouse CEDEX 04, France
| | | | | |
Collapse
|
22
|
Mochiduki A, Takeda T, Kaga S, Inoue K. Stress response of prolactin-releasing peptide knockout mice as to glucocorticoid secretion. J Neuroendocrinol 2010; 22:576-84. [PMID: 20298457 DOI: 10.1111/j.1365-2826.2010.01993.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Prolactin-releasing peptide (PrRP) is known to have functions in prolactin secretion, stress responses, cardiovascular regulation and food intake suppression. In addition, PrRP-knockout (KO) male mice show obesity from the age of 22 weeks and increase their food intake. The plasma concentrations of insulin, leptin, cholesterol and triglyceride are also increased in obese PrRP-KO mice. Fatty liver, hypertrophied white adipose tissue, decreased uncoupling protein 1 mRNA expression in brown adipose tissue and glucose intolerance were observed in obese PrRP-KO mice. As we reported previously, PrRP stimulates corticotrophin-releasing factor and regulates the hypothalamic-pituitary-adrenal axis. Therefore, it is speculated that PrRP regulates both food intake and metabolism as a stress responses. In the present study, we compared blood glucose and plasma glucocorticoid concentrations in PrRP-KO mice, and found that PrRP-KO mice showed higher concentrations of blood glucose and corticosterone compared to wild-type mice after restraint stress. By contrast, there were no difference in c-Fos expression in the paraventricular hypothalamic nucleus and plasma adrenocorticotrophic hormone concentrations between the two groups. These results suggest that the different stress responses as to glucocorticoid secretion may be induced by different responses of the adrenal glands between wild-type and PrRP-KO mice. Thus, we conclude that PrRP-KO mice become obese as a result of increased food intake, a change in metabolism, and abnormal stress responses as to glucose concentration and glucocorticoid secretion.
Collapse
Affiliation(s)
- A Mochiduki
- Department of Regulatory Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | | | | | | |
Collapse
|
23
|
Onaka T, Takayanagi Y, Leng G. Metabolic and stress-related roles of prolactin-releasing peptide. Trends Endocrinol Metab 2010; 21:287-93. [PMID: 20122847 DOI: 10.1016/j.tem.2010.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 12/23/2009] [Accepted: 01/07/2010] [Indexed: 12/26/2022]
Abstract
In the modern world, improvements in human health can be offset by unhealthy lifestyle factors, including the deleterious consequences of stress and obesity. For energy homeostasis, humoral factors and neural afferents from the gastrointestinal tract, in combination with long-term nutritional signals, communicate information to the brain to regulate energy intake and expenditure. Energy homeostasis and stress interact with each other, and stress affects both food intake and energy expenditure. Prolactin-releasing peptide, synthesized in discrete neuronal populations in the hypothalamus and brainstem, plays an important role in integrating these responses. This review describes how prolactin-releasing peptide neurons receive information concerning both internal metabolic states and environmental conditions, and play a key role in energy homeostasis and stress responses.
Collapse
Affiliation(s)
- Tatsushi Onaka
- Department of Physiology, Jichi Medical University, Shimotsuke-shi, Tochigi-ken 329-0498, Japan
| | | | | |
Collapse
|
24
|
Anand KJS, Willson DF, Berger J, Harrison R, Meert KL, Zimmerman J, Carcillo J, Newth CJL, Prodhan P, Dean JM, Nicholson C, Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network. Tolerance and withdrawal from prolonged opioid use in critically ill children. Pediatrics 2010; 125:e1208-25. [PMID: 20403936 PMCID: PMC3275643 DOI: 10.1542/peds.2009-0489] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE After prolonged opioid exposure, children develop opioid-induced hyperalgesia, tolerance, and withdrawal. Strategies for prevention and management should be based on the mechanisms of opioid tolerance and withdrawal. PATIENTS AND METHODS Relevant manuscripts published in the English language were searched in Medline by using search terms "opioid," "opiate," "sedation," "analgesia," "child," "infant-newborn," "tolerance," "dependency," "withdrawal," "analgesic," "receptor," and "individual opioid drugs." Clinical and preclinical studies were reviewed for data synthesis. RESULTS Mechanisms of opioid-induced hyperalgesia and tolerance suggest important drug- and patient-related risk factors that lead to tolerance and withdrawal. Opioid tolerance occurs earlier in the younger age groups, develops commonly during critical illness, and results more frequently from prolonged intravenous infusions of short-acting opioids. Treatment options include slowly tapering opioid doses, switching to longer-acting opioids, or specifically treating the symptoms of opioid withdrawal. Novel therapies may also include blocking the mechanisms of opioid tolerance, which would enhance the safety and effectiveness of opioid analgesia. CONCLUSIONS Opioid tolerance and withdrawal occur frequently in critically ill children. Novel insights into opioid receptor physiology and cellular biochemical changes will inform scientific approaches for the use of opioid analgesia and the prevention of opioid tolerance and withdrawal.
Collapse
Affiliation(s)
- Kanwaljeet J S Anand
- Department of Pediatrics, Le Bonheur Children's Hospital and University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Prolactin-releasing peptide regulates cardiac contractility. ACTA ACUST UNITED AC 2010; 159:9-13. [DOI: 10.1016/j.regpep.2009.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 07/07/2009] [Accepted: 07/10/2009] [Indexed: 11/24/2022]
|
26
|
Uchida K, Kobayashi D, Das G, Onaka T, Inoue K, Itoi K. Participation of the prolactin-releasing peptide-containing neurones in caudal medulla in conveying haemorrhagic stress-induced signals to the paraventricular nucleus of the hypothalamus. J Neuroendocrinol 2010; 22:33-42. [PMID: 19912474 DOI: 10.1111/j.1365-2826.2009.01935.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The prolactin-releasing peptide (PrRP) has been proposed to be a co-transmitter or modulator of noradrenaline (NA) because it colocalises with NA in the A1 (in the ventrolateral reticular formation) and A2 (in the nucleus of the solitary tract; NTS) cell groups in the caudal medulla. The baroreceptor signals, originating from the great vessels, are transmitted primarily to the NTS, and then part of the signals is conveyed to the hypothalamic neuroendocrine neurones via the ascending NA neurones. The hypotensive haemorrhagic paradigm was employed to examine whether the PrRP-containing neurones in the caudal medulla participate in conveying signals to the hypothalamic neuroendocrine neurones. Among the caudal medullary A1 or A2 neurones, the majority of the PrRP-immunoreactive (-ir) neurones became c-Fos-ir at 2 h after hypotensive haemorrhage. Hypothalamic corticotrophin-releasing hormone-ir neurones and vasopressin-ir neurones became c-Fos positive in parallel with the activation of medullary PrRP-ir neurones. After delivery of retrograde tracer fluorogold (FG) to the paraventricular nucleus of the hypothalamus (PVN), part of the PrRP/FG double-labelled neurones in the A1 and A2 became c-Fos-ir after haemorrhage, demonstrating that PrRP-ir neurones participate in conveying the haemorrhagic stress-induced signals from the medulla to the PVN. PrRP and/or NA were microinjected directly to the PVN of conscious rats, and they presented a synergistic action on arginine vasopressin release, whereas an additive action was observed for adrenocorticotrophin release. These results suggest that the PrRP-containing NA neurones in the caudal medulla may relay the haemorrhagic stress-induced medullary inputs to the hypothalamic neuroendocrine neurones.
Collapse
Affiliation(s)
- K Uchida
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Aramaki-aza Aoba, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Effect of KEPI (Ppp1r14c) deletion on morphine analgesia and tolerance in mice of different genetic backgrounds: when a knockout is near a relevant quantitative trait locus. Neuroscience 2009; 165:882-95. [PMID: 19819304 DOI: 10.1016/j.neuroscience.2009.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 09/23/2009] [Accepted: 10/03/2009] [Indexed: 01/01/2023]
Abstract
We previously identified KEPI as a morphine-regulated gene using subtractive hybridization and differential display PCR. Upon phosphorylation by protein kinase C, KEPI becomes a powerful inhibitor of protein phosphatase 1. To gain insights into KEPI functions, we created KEPI knockout (KO) mice on mixed 129S6xC57BL/6 genetic backgrounds. KEPI maps onto mouse chromosome 10 close to the locus that contains the mu-opioid receptor (Oprm1) and provides a major quantitative trait locus for morphine effects. Analysis of single nucleotide polymorphisms in and near the Oprm1 locus identified a doubly-recombinant mouse with C57BL/6 markers within 1 Mb on either side of the KEPI deletion. This strategy minimized the amount of 129S6 DNA surrounding the transgene and documented the C57BL/6 origin of the Oprm1 gene in this founder and its offspring. Recombinant KEPIKO mice displayed (a) normal analgesic responses and normal locomotion after initial morphine treatments, (b) accelerated development of tolerance to analgesic effects of morphine, (c) elevated activity of protein phosphatase 1 in thalamus, (d) attenuated morphine reward as assessed by conditioned place preference. These data support roles for KEPI action in adaptive responses to repeated administration of morphine that include analgesic tolerance and drug reward.
Collapse
|
28
|
Sun B, Mochiduki A, Nakamura K, Yokoyama K, Adachi S, Fujiwara K, Matsumoto H, Inoue K. Blockade of PrRP attenuates MPTP-induced toxicity in mice. Peptides 2009; 30:1267-75. [PMID: 19540425 DOI: 10.1016/j.peptides.2009.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 03/17/2009] [Accepted: 03/17/2009] [Indexed: 11/20/2022]
Abstract
Prolactin-releasing peptide (PrRP) was isolated as an endogenous ligand of the orphan G-protein coupled receptor hGR3. PrRP has been shown to be involved in the regulation of food intake, stress responses, prolactin secretion and release, blood pressure, and the opioid system. Here we report that PrRP and its receptor, GPR10, were found in the mouse substantia nigra pars compacta (SNpc), the main location of dopaminergic (DA) neurons of the nigrostriatal system. We generated PrRP knockout (KO) mice, and then treated PrRP KO mice and their wild type (WT) littermates with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a neuron toxin that selectively damages DA neurons in the SNpc. We found that PrRP KO mice were resistant to MPTP-induced lesions of the nigrostriatal system. These effects were further confirmed by the intracerebroventricular injection of P2L-1C, a monoclonal antibody against PrRP into mice. Taken together, our data established a critical role of PrRP in MPTP intoxication in mice.
Collapse
Affiliation(s)
- Binggui Sun
- Department of Regulation Biology, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama 338-8570, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Takayanagi Y, Matsumoto H, Nakata M, Mera T, Fukusumi S, Hinuma S, Ueta Y, Yada T, Leng G, Onaka T. Endogenous prolactin-releasing peptide regulates food intake in rodents. J Clin Invest 2008; 118:4014-24. [PMID: 19033670 PMCID: PMC2575834 DOI: 10.1172/jci34682] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Accepted: 09/17/2008] [Indexed: 02/03/2023] Open
Abstract
Food intake is regulated by a network of signals that emanate from the gut and the brainstem. The peripheral satiety signal cholecystokinin is released from the gut following food intake and acts on fibers of the vagus nerve, which project to the brainstem and activate neurons that modulate both gastrointestinal function and appetite. In this study, we found that neurons in the nucleus tractus solitarii of the brainstem that express prolactin-releasing peptide (PrRP) are activated rapidly by food ingestion. To further examine the role of this peptide in the control of food intake and energy metabolism, we generated PrRP-deficient mice and found that they displayed late-onset obesity and adiposity, phenotypes that reflected an increase in meal size, hyperphagia, and attenuated responses to the anorexigenic signals cholecystokinin and leptin. Hypothalamic expression of 6 other appetite-regulating peptides remained unchanged in the PrRP-deficient mice. Blockade of endogenous PrRP signaling in WT rats by central injection of PrRP-specific mAb resulted in an increase in food intake, as reflected by an increase in meal size. These data suggest that PrRP relays satiety signals within the brain and that selective disturbance of this system can result in obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Hirokazu Matsumoto
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Masanori Nakata
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Takashi Mera
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Shoji Fukusumi
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Shuji Hinuma
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yoichi Ueta
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Toshihiko Yada
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Gareth Leng
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Frontier Research Laboratories I, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Tsukuba, Ibaraki, Japan.
Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan.
Department of Physiology, University of Occupational and Environmental Health, Kitakyushu, Japan.
Research Information & Alliances Strategic Research Planning Department, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Juso-honmachi, Yodogawaku, Osaka, Japan.
Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
30
|
Abstract
Prolactin-releasing peptide (PrRP) was initially isolated from the bovine hypothalamus as an activating component that stimulated arachidonic acid release from cells stably expressing the orphan G protein-coupled receptor hGR3 (Hinuma et al. 1998) [also known as GPR10 (Marchese et al. 1995), or UHR-1 for the rat orthologue (Welch et al. 1995)]. Initially touted as a prolactin-releasing factor (therefore aptly named prolactin-releasing peptide), the perspective on the function of this peptide in the organism has been greatly expanded. Over 120 papers have been published on this subject since its initial discovery in 1998. Herein I review the state of knowledge of the PrRP system, its putative function in the organism, and implications for therapy.
Collapse
|
31
|
Candidate gene polymorphisms predicting individual sensitivity to opioids. Naunyn Schmiedebergs Arch Pharmacol 2007; 377:269-81. [DOI: 10.1007/s00210-007-0205-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 10/18/2007] [Indexed: 11/26/2022]
|
32
|
Tzschentke TM. Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol 2007; 12:227-462. [PMID: 17678505 DOI: 10.1111/j.1369-1600.2007.00070.x] [Citation(s) in RCA: 1032] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conditioned place preference (CPP) continues to be one of the most popular models to study the motivational effects of drugs and non-drug treatments in experimental animals. This is obvious from a steady year-to-year increase in the number of publications reporting the use this model. Since the compilation of the preceding review in 1998, more than 1000 new studies using place conditioning have been published, and the aim of the present review is to provide an overview of these recent publications. There are a number of trends and developments that are obvious in the literature of the last decade. First, as more and more knockout and transgenic animals become available, place conditioning is increasingly used to assess the motivational effects of drugs or non-drug rewards in genetically modified animals. Second, there is a still small but growing literature on the use of place conditioning to study the motivational aspects of pain, a field of pre-clinical research that has so far received little attention, because of the lack of appropriate animal models. Third, place conditioning continues to be widely used to study tolerance and sensitization to the rewarding effects of drugs induced by pre-treatment regimens. Fourth, extinction/reinstatement procedures in place conditioning are becoming increasingly popular. This interesting approach is thought to model certain aspects of relapse to addictive behavior and has previously almost exclusively been studied in drug self-administration paradigms. It has now also become established in the place conditioning literature and provides an additional and technically easy approach to this important phenomenon. The enormous number of studies to be covered in this review prevented in-depth discussion of many methodological, pharmacological or neurobiological aspects; to a large extent, the presentation of data had to be limited to a short and condensed summary of the most relevant findings.
Collapse
Affiliation(s)
- Thomas M Tzschentke
- Grünenthal GmbH, Preclinical Research and Development, Department of Pharmacology, Aachen, Germany.
| |
Collapse
|
33
|
Watanabe A, Okuno S, Okano M, Jordan S, Aihara K, Watanabe TK, Yamasaki Y, Kitagawa H, Sugawara K, Kato S. Altered emotional behaviors in the diabetes mellitus OLETF type 1 congenic rat. Brain Res 2007; 1178:114-24. [PMID: 17916333 DOI: 10.1016/j.brainres.2007.07.075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Revised: 07/26/2007] [Accepted: 07/28/2007] [Indexed: 11/28/2022]
Abstract
GPR10 is a G-protein-coupled receptor expressed in thalamic and hypothalamic brain regions, including the reticular thalamic nucleus (RTN) and periventricular nucleus (Pev), and the endogenous ligand for this receptor, prolactin-releasing peptide (PrRP), has demonstrated regulatory effects on the stress response. We produced a congenic rat by introducing the Dmo1 allele from the OLETF rat which encodes the amino acid sequences of GPR10 with a truncated NH2-terminus, into the Brown-Norway background. Using receptor autoradiography, we determined a lack of specific [125I]PrRP binding in the RTN and Pev of these mutant rats compared to the control rats. Furthermore, intracerebroventricular injection of PrRP did not induce a significant increase of c-fos-like immunoreactivity in the paraventricular nucleus of the mutant rats compared to the control rats. The mutant rats also displayed a less anxious-like phenotype in three behavioral-based models of anxiety-like behavior (open field, elevated plus maze and defensive withdrawal test). These data show the mutant congenic rat, of which GPR10 neither binds nor responds to PrRP, expresses less anxious-like phenotypes. On the basis of these observations, the GPR10 might be a novel target for the developing new drugs against anxiety and/or other stress-related diseases.
Collapse
Affiliation(s)
- Akihito Watanabe
- Department of Molecular Neurobiology, Graduate School of Medicine, Kanazawa University, Kanazawa, 920-8640, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
This paper is the 28th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning over a quarter-century of research. It summarizes papers published during 2005 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity, neurophysiology and transmitter release (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY 11367, USA.
| | | |
Collapse
|
35
|
Bradshaw HB, Rimmerman N, Krey JF, Walker JM. Sex and hormonal cycle differences in rat brain levels of pain-related cannabimimetic lipid mediators. Am J Physiol Regul Integr Comp Physiol 2006; 291:R349-58. [PMID: 16556899 DOI: 10.1152/ajpregu.00933.2005] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One important function of endocannabinoids and related lipid mediators in mammalian central nervous system is modulation of pain. Evidence obtained during the last decade shows that altered levels of these compounds in the brain accompany decreases in pain sensitivity. Such changes, if sexually dimorphic, could account for sex differences in pain and differences that occur during different phases of the hormonal cycle in females. To examine this possibility, we measured the levels of the pain-modulatory lipids anandamide, 2-arachidonoyl glycerol, N-arachidonoyl glycine, N-arachidonoyl gamma amino butyric acid, and N-arachidonoyl dopamine in seven different brain areas (pituitary, hypothalamus, thalamus, striatum, midbrain, hippocampus, and cerebellum) in male rats, and in female rats at five different points in the estrous cycle. The cerebellum did not demonstrate a change in endocannabinoid production across the estrous cycle, whereas all other areas tested showed significant differences in at least one of the compounds measured. These changes in levels occurred predominantly within the 36-h time period surrounding ovulation and behavioral estrus. Differences between males and females were measured as either estrous cycle-independent (all estrous cycles combined) or cycle-dependent (comparisons of males to each estrous cycle). In cycle-independent analyses, small sex differences were observed in the pituitary, hypothalamus, cerebellum, and striatum, whereas no differences were observed in the thalamus, midbrain, and hippocampus. In cycle-dependent analyses, the hypothalamus and pituitary showed largest sex differences followed by the striatum, midbrain, and hippocampus, whereas no sex differences were measured in thalamus and cerebellum. These data provide a basis for investigations into how differences in sex and hormonal status play a role in mechanisms regulating endocannabinoid production and pain.
Collapse
Affiliation(s)
- Heather B Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, 1101 East 10th St., Bloomington, IN 47405, USA
| | | | | | | |
Collapse
|
36
|
Mera T, Fujihara H, Kawasaki M, Hashimoto H, Saito T, Shibata M, Saito J, Oka T, Tsuji S, Onaka T, Ueta Y. Prolactin-releasing peptide is a potent mediator of stress responses in the brain through the hypothalamic paraventricular nucleus. Neuroscience 2006; 141:1069-1086. [PMID: 16730416 DOI: 10.1016/j.neuroscience.2006.04.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 04/04/2006] [Accepted: 04/05/2006] [Indexed: 12/13/2022]
Abstract
The effects of i.c.v. administration of prolactin-releasing peptide on neurons in the paraventricular nucleus of rats and plasma corticosterone levels were examined by measuring changes in Fos-like immunoreactivity, c-fos mRNA using in situ hybridization histochemistry, and plasma corticosterone using a specific radioimmunoassay. Approximately 80% of corticotropin-releasing hormone immunoreactive cells exhibited Fos-like immunoreactivity in the parvocellular division of the paraventricular nucleus 90 min after i.c.v. administration of prolactin-releasing peptide. The greatest induction of the c-fos mRNA expression in the paraventricular nucleus was observed 30 min after administration of prolactin-releasing peptide, and occurred in a dose-related manner. Plasma corticosterone levels were also significantly increased 30 min after administration of prolactin-releasing peptide. Next, the effects of restraint stress, nociceptive stimulus and acute inflammatory stress on the expression of the prolactin-releasing peptide mRNA in the dorsomedial hypothalamic nucleus, nucleus of the solitary tract and ventrolateral medulla were examined using in situ hybridization histochemistry for prolactin-releasing peptide mRNA. Restraint stress and acute inflammatory stress upregulated the prolactin-releasing peptide mRNA expression in the nucleus of the solitary tract and ventrolateral medulla. Nociceptive stimulus upregulated the prolactin-releasing peptide mRNA expression in the ventrolateral medulla. Finally, we observed that pretreatment (i.c.v. administration) with an anti-prolactin-releasing peptide antibody significantly attenuated nociceptive stimulus-induced c-fos mRNA expression in the paraventricular nucleus. These results suggest that prolactin-releasing peptide is a potent and important mediator of the stress response in the brain through the hypothalamic paraventricular nucleus.
Collapse
Affiliation(s)
- T Mera
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - H Fujihara
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - M Kawasaki
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - H Hashimoto
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - T Saito
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - M Shibata
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - J Saito
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - T Oka
- Department of Neurology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - S Tsuji
- Department of Neurology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - T Onaka
- Department of Physiology, Jichi Medical School, Tochigi 329-0498, Japan
| | - Y Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| |
Collapse
|