1
|
Xu JF, Liu L, Liu Y, Lu KX, Zhang J, Zhu YJ, Fang F, Dou YN. Spinal Nmur2-positive Neurons Play a Crucial Role in Mechanical Itch. THE JOURNAL OF PAIN 2024; 25:104504. [PMID: 38442838 DOI: 10.1016/j.jpain.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
The dorsal spinal cord is crucial for the transmission and modulation of multiple somatosensory modalities, such as itch, pain, and touch. Despite being essential for the well-being and survival of an individual, itch and pain, in their chronic forms, have increasingly been recognized as clinical problems. Although considerable progress has been made in our understanding of the neurochemical processing of nociceptive and chemical itch sensations, the neural substrate that is crucial for mechanical itch processing is still unclear. Here, using genetic and functional manipulation, we identified a population of spinal neurons expressing neuromedin U receptor 2 (Nmur2+) as critical elements for mechanical itch. We found that spinal Nmur2+ neurons are predominantly excitatory neurons, and are enriched in the superficial laminae of the dorsal horn. Pharmacogenetic activation of cervical spinal Nmur2+ neurons evoked scratching behavior. Conversely, the ablation of these neurons using a caspase-3-based method decreased von Frey filament-induced scratching behavior without affecting responses to other somatosensory modalities. Similarly, suppressing the excitability of cervical spinal Nmur2+ neurons via the overexpression of functional Kir2.1 potassium channels reduced scratching in response to innocuous mechanical stimuli, but not to pruritogen application. At the lumbar level, pharmacogenetic activation of these neurons evoked licking and lifting behaviors. However, ablating these neurons did not affect the behavior associated with acute pain. Thus, these results revealed the crucial role of spinal Nmur2+ neurons in mechanical itch. Our study provides important insights into the neural basis of mechanical itch, paving the way for developing novel therapies for chronic itch. PERSPECTIVE: Excitatory Nmur2+ neurons in the superficial dorsal spinal cord are essential for mechanical but not chemical itch information processing. These spinal Nmur2+ neurons represent a potential cellular target for future therapeutic interventions against chronic itch. Spinal and supraspinal Nmur2+ neurons may play different roles in pain signal processing.
Collapse
Affiliation(s)
- Jun-Feng Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lian Liu
- Department of Endocrinology and Metabolic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Lingang Laboratory, Shanghai, China
| | - Ke-Xing Lu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jun Zhang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan-Jing Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Fang Fang
- Department of Endocrinology and Metabolic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Nong Dou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
2
|
Botticelli L, Micioni Di Bonaventura E, Del Bello F, Giorgioni G, Piergentili A, Quaglia W, Bonifazi A, Cifani C, Micioni Di Bonaventura MV. The neuromedin U system: Pharmacological implications for the treatment of obesity and binge eating behavior. Pharmacol Res 2023; 195:106875. [PMID: 37517560 DOI: 10.1016/j.phrs.2023.106875] [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: 06/20/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/01/2023]
Abstract
Neuromedin U (NMU) is a bioactive peptide produced in the gut and in the brain, with a role in multiple physiological processes. NMU acts by binding and activating two G protein coupled receptors (GPCR), the NMU receptor 1 (NMU-R1), which is predominantly expressed in the periphery, and the NMU receptor 2 (NMU-R2), mainly expressed in the central nervous system (CNS). In the brain, NMU and NMU-R2 are consistently present in the hypothalamus, commonly recognized as the main "feeding center". Considering its distribution pattern, NMU revealed to be an important neuropeptide involved in the regulation of food intake, with a powerful anorexigenic ability. This has been observed through direct administration of NMU and by studies using genetically modified animals, which revealed an obesity phenotype when the NMU gene is deleted. Thus, the development of NMU analogs or NMU-R2 agonists might represent a promising pharmacological strategy to treat obese individuals. Furthermore, NMU has been demonstrated to influence the non-homeostatic aspect of food intake, playing a potential role in binge eating behavior. This review aims to discuss and summarize the current literature linking the NMU system with obesity and binge eating behavior, focusing on the influence of NMU on food intake and the neuronal mechanisms underlying its anti-obesity properties. Pharmacological strategies to improve the pharmacokinetic profile of NMU will also be reported.
Collapse
Affiliation(s)
- Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, Camerino 62032, Italy
| | | | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, Camerino 62032, Italy.
| | | |
Collapse
|
3
|
Ligand recognition and activation of neuromedin U receptor 2. Nat Commun 2022; 13:7955. [PMID: 36575163 PMCID: PMC9794833 DOI: 10.1038/s41467-022-34814-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/08/2022] [Indexed: 12/29/2022] Open
Abstract
Neuromedin U receptor 2 (NMU2), an emerging attractive target for treating obesity, has shown the capability in reducing food intake and regulating energy metabolism when activated. However, drug development of NMU2 was deferred partially due to the lack of structural information. Here, we present the cryo-electron microscopy (cryo-EM) structure of NMU2 bound to the endogenous agonist NmU-25 and Gi1 at 3.3 Å resolution. Combined with functional and computational data, the structure reveals the key factors that govern the recognition and selectivity of peptide agonist as well as non-peptide antagonist, providing the structural basis for design of novel and highly selective drugs targeting NMU2. In addition, a 25-degree rotation of Gi protein in reference to NMU2 is also observed compared in other structures of class A GPCR-Gi complexes, suggesting heterogeneity in the processes of G protein-coupled receptors (GPCRs) activation and G protein coupling.
Collapse
|
4
|
Porcu A, Nilsson A, Booreddy S, Barnes SA, Welsh DK, Dulcis D. Seasonal changes in day length induce multisynaptic neurotransmitter switching to regulate hypothalamic network activity and behavior. SCIENCE ADVANCES 2022; 8:eabn9867. [PMID: 36054362 PMCID: PMC10848959 DOI: 10.1126/sciadv.abn9867] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/19/2022] [Indexed: 05/18/2023]
Abstract
Seasonal changes in day length (photoperiod) affect numerous physiological functions. The suprachiasmatic nucleus (SCN)-paraventricular nucleus (PVN) axis plays a key role in processing photoperiod-related information. Seasonal variations in SCN and PVN neurotransmitter expression have been observed in humans and animal models. However, the molecular mechanisms by which the SCN-PVN network responds to altered photoperiod is unknown. Here, we show in mice that neuromedin S (NMS) and vasoactive intestinal polypeptide (VIP) neurons in the SCN display photoperiod-induced neurotransmitter plasticity. In vivo recording of calcium dynamics revealed that NMS neurons alter PVN network activity in response to winter-like photoperiod. Chronic manipulation of NMS neurons is sufficient to induce neurotransmitter switching in PVN neurons and affects locomotor activity. Our findings reveal previously unidentified molecular adaptations of the SCN-PVN network in response to seasonality and the role for NMS neurons in adjusting hypothalamic function to day length via a coordinated multisynaptic neurotransmitter switching affecting behavior.
Collapse
Affiliation(s)
- Alessandra Porcu
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Anna Nilsson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Sathwik Booreddy
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - David K. Welsh
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Davide Dulcis
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
5
|
Piwowarczyk-Nowak A, Pałasz A, Suszka-Świtek A, Della Vecchia A, Grajoszek A, Krzystanek M, Worthington JJ. Escitalopram alters local expression of noncanonical stress-related neuropeptides in the rat brain via NPS receptor signaling. Pharmacol Rep 2022; 74:637-653. [PMID: 35653031 DOI: 10.1007/s43440-022-00374-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Neuropeptide S (NPS) is a multifunctional regulatory factor that exhibits a potent anxiolytic activity in animal models. However, there are no reports dealing with the potential molecular relationships between the anxiolytic activity of selective serotonin reuptake inhibitors (SSRIs) and NPS signaling, especially in the context of novel stress-related neuropeptides action. The present work therefore focused on gene expression of novel stress neuropeptides in the rat brain after acute treatment with escitalopram and in combination with neuropeptide S receptor (NPSR) blockade. METHODS Studies were carried out on adult, male Sprague-Dawley rats that were divided into five groups: animals injected with saline (control) and experimental rats treated with escitalopram (at single dose 10 mg/kg daily), escitalopram and SHA-68, a selective NPSR antagonist (at a single dose of 40 mg/kg), SHA-68 alone and corresponding vehicle (solvent SHA-68) control. To measure anxiety-like behavior and locomotor activity the open field test was performed. All individuals were killed under anaesthesia and the whole brain was excised. Total mRNA was isolated from homogenized samples of the amygdala, hippocampus, hypothalamus, thalamus, cerebellum, and brainstem. Real-time PCR was used for estimation of related NPS, NPSR, neuromedin U (NMU), NMU receptor 2 (NMUR2) and nesfatin-1 precursor nucleobindin-2 (NUCB2) gene expression. RESULTS Acute escitalopram administration affects the local expression of the examined neuropeptides mRNA in a varied manner depending on brain location. An increase in NPSR and NUCB2 mRNA expression in the hypothalamus and brainstem was abolished by SHA-68 coadministration, while NMU mRNA expression was upregulated after NPSR blockade in the hippocampus and cerebellum. CONCLUSIONS The pharmacological effects of escitalopram may be connected with local NPSR-related alterations in NPS/NMU/NMUR2 and nesfatin-1 gene expression at the level of selected rat brain regions. A novel alternative mode of SSRI action can be therefore cautiously proposed.
Collapse
Affiliation(s)
- Aneta Piwowarczyk-Nowak
- Department of Anatomy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland.
| | - Aleksandra Suszka-Świtek
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Alessandra Della Vecchia
- Section of Psychiatry, Department of Clinical and Experimental Medicine, University of Pisa, 67, Via Roma, 56100, Pisa, Italy
| | - Aniela Grajoszek
- Department for Experimental Medicine, Medical University of Silesia, ul. Medyków 4, 40-752, Katowice, Poland
| | - Marek Krzystanek
- Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences in Katowice, Clinic of Psychiatric Rehabilitation, Medical University of Silesia, ul. Ziolowa 45/47, 40-635, Katowice, Poland
| | - John J Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YQ, UK
| |
Collapse
|
6
|
Piwowarczyk-Nowak A, Pałasz A, Suszka-Świtek A, Błaszczyk I, Bogus K, Łasut-Szyszka B, Krzystanek M, Worthington JJ. Effect of Escitalopram on the Number of DCX-Positive Cells and NMUR2 Receptor Expression in the Rat Hippocampus under the Condition of NPSR Receptor Blockade. Pharmaceuticals (Basel) 2022; 15:631. [PMID: 35631458 PMCID: PMC9143903 DOI: 10.3390/ph15050631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Neuropeptide S (NPS) is a multifunctional regulatory factor that exhibits a potent anxiolytic activity in animal models. However, there are no reports dealing with the potential molecular interactions between the activity of selective serotonin reuptake inhibitors (SSRIs) and NPS signaling, especially in the context of adult neurogenesis and the expression of noncanonical stress-related neuropeptides such as neuromedin U (NMU). The present work therefore focused on immunoexpression of neuromedin U receptor 2 (NMUR2) and doublecortin (DCX) in the rat hippocampus after acute treatment with escitalopram and in combination with selective neuropeptide S receptor (NPSR) blockade. METHODS Studies were carried out on adult, male Sprague-Dawley rats that were divided into five groups: animals injected with saline (control) and experimental individuals treated with escitalopram (at single dose 10 mg/kg daily), escitalopram + SHA-68, a selective NPSR antagonist (at single dose 40 mg/kg), SHA-68 alone, and corresponding vehicle control. All animals were sacrificed under halothane anaesthesia. The whole hippocampi were quickly excised, fixed, and finally sliced for general qualitative immunohistochemical assessment of the NPSR and NMUR2 expression. The number of immature neurons was enumerated using immunofluorescent detection of doublecortin (DCX) expression within the subgranular zone (SGZ). RESULTS Acute escitalopram administration affects the number of DCX and NMUR2-expressing cells in the adult rat hippocampus. A decreased number of DCX-expressing neuroblasts after treatment with escitalopram was augmented by SHA-68 coadministration. CONCLUSIONS Early pharmacological effects of escitalopram may be at least partly connected with local NPSR-related alterations of neuroblast maturation in the rat hippocampus. Escitalopram may affect neuropeptide and DCX-expression starting even from the first dose. Adult neurogenesis may be regulated via paracrine neuropeptide S and NMU-related signaling.
Collapse
Affiliation(s)
- Aneta Piwowarczyk-Nowak
- Department of Anatomy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland;
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Aleksandra Suszka-Świtek
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Iwona Błaszczyk
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Katarzyna Bogus
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752 Katowice, Poland; (A.S.-Ś.); (I.B.); (K.B.)
| | - Barbara Łasut-Szyszka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland;
| | - Marek Krzystanek
- Clinic of Psychiatric Rehabilitation, Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Ziolowa 45/47, 40-635 Katowice, Poland;
| | - John J. Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK;
| |
Collapse
|
7
|
Wang H, Chen W, Dong Z, Xing G, Cui W, Yao L, Zou WJ, Robinson HL, Bian Y, Liu Z, Zhao K, Luo B, Gao N, Zhang H, Ren X, Yu Z, Meixiong J, Xiong WC, Mei L. A novel spinal neuron connection for heat sensation. Neuron 2022; 110:2315-2333.e6. [PMID: 35561677 DOI: 10.1016/j.neuron.2022.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022]
Abstract
Heat perception enables acute avoidance responses to prevent tissue damage and maintain body thermal homeostasis. Unlike other modalities, how heat signals are processed in the spinal cord remains unclear. By single-cell gene profiling, we identified ErbB4, a transmembrane tyrosine kinase, as a novel marker of heat-sensitive spinal neurons in mice. Ablating spinal ErbB4+ neurons attenuates heat sensation. These neurons receive monosynaptic inputs from TRPV1+ nociceptors and form excitatory synapses onto target neurons. Activation of ErbB4+ neurons enhances the heat response, while inhibition reduces the heat response. We showed that heat sensation is regulated by NRG1, an activator of ErbB4, and it involves dynamic activity of the tyrosine kinase that promotes glutamatergic transmission. Evidence indicates that the NRG1-ErbB4 signaling is also engaged in hypersensitivity of pathological pain. Together, these results identify a spinal neuron connection consisting of ErbB4+ neurons for heat sensation and reveal a regulatory mechanism by the NRG1-ErbB4 signaling.
Collapse
Affiliation(s)
- Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wenbing Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wanpeng Cui
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Lingling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wen-Jun Zou
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Heath L Robinson
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Yaoyao Bian
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zhipeng Liu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Kai Zhao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Nannan Gao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - James Meixiong
- Solomon H. Snyder Department of Neuroscience and Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
8
|
Hsiao YT, Manikowski KJ, Snyder S, Griffin N, Orr AL, Hulsey EQ, Born-Evers G, Zukosky T, Squire ME, Hum JM, Metzger CE, Allen MR, Lowery JW. NMUR1 in the NMU-Mediated Regulation of Bone Remodeling. Life (Basel) 2021; 11:life11101028. [PMID: 34685399 PMCID: PMC8538501 DOI: 10.3390/life11101028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022] Open
Abstract
Neuromedin-U (NMU) is an evolutionarily conserved peptide that regulates varying physiologic effects including blood pressure, stress and allergic responses, metabolic and feeding behavior, pain perception, and neuroendocrine functions. Recently, several lines of investigation implicate NMU in regulating bone remodeling. For instance, global loss of NMU expression in male and female mice leads to high bone mass due to elevated bone formation rate with no alteration in bone resorption rate or observable defect in skeletal patterning. Additionally, NMU treatment regulates the activity of osteoblasts in vitro. The downstream pathway utilized by NMU to carry out these effects is unknown as NMU signals via two G-protein-coupled receptors (GPCRs), NMU receptor 1 (NMUR1), and NMU receptor 2 (NMUR2), and both are expressed in the postnatal skeleton. Here, we sought to address this open question and build a better understanding of the downstream pathway utilized by NMU. Our approach involved the knockdown of Nmur1 in MC3T3-E1 cells in vitro and a global knockout of Nmur1 in vivo. We detail specific cell signaling events (e.g., mTOR phosphorylation) that are deficient in the absence of NMUR1 expression yet trabecular bone volume in femora and tibiae of 12-week-old male Nmur1 knockout mice are unchanged, compared to controls. These results suggest that NMUR1 is required for NMU-dependent signaling in MC3T3-E1 cells, but it is not required for the NMU-mediated effects on bone remodeling in vivo. Future studies examining the role of NMUR2 are required to determine the downstream pathway utilized by NMU to regulate bone remodeling in vivo.
Collapse
Affiliation(s)
- Yu-Tin Hsiao
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Kelli J. Manikowski
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Samantha Snyder
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Nicole Griffin
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Ashley L. Orr
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Elizabeth Q. Hulsey
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Gabriella Born-Evers
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Tara Zukosky
- Department of Biology, The University of Scranton, Scranton, PA 18503, USA; (T.Z.); (M.E.S.)
| | - Maria E. Squire
- Department of Biology, The University of Scranton, Scranton, PA 18503, USA; (T.Z.); (M.E.S.)
| | - Julia M. Hum
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
| | - Corinne E. Metzger
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (C.E.M.); (M.R.A.)
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew R. Allen
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (C.E.M.); (M.R.A.)
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jonathan W. Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46022, USA; (Y.-T.H.); (K.J.M.); (S.S.); (N.G.); (A.L.O.); (E.Q.H.); (G.B.-E.); (J.M.H.)
- Bone and Muscle Research Group, Marian University, Indianapolis, IN 46022, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Fax: +1-317-955-6621
| |
Collapse
|
9
|
Neuromedin U, a Key Molecule in Metabolic Disorders. Int J Mol Sci 2021; 22:ijms22084238. [PMID: 33921859 PMCID: PMC8074168 DOI: 10.3390/ijms22084238] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is now a public health concern. The leading cause of obesity is an energy imbalance between ingested and expended calories. The mechanisms of feeding behavior and energy metabolism are regulated by a complex of various kinds of molecules, including anorexigenic and orexigenic neuropeptides. One of these neuropeptides, neuromedin U (NMU), was isolated in the 1980s, and its specific receptors, NMUR1 and NMUR2, were defined in 2000. A series of subsequent studies has revealed many of the physiological roles of the NMU system, including in feeding behavior, energy expenditure, stress responses, circadian rhythmicity, and inflammation. Particularly over the past decades, many reports have indicated that the NMU system plays an essential and direct role in regulating body weight, feeding behavior, energy metabolism, and insulin secretion, which are tightly linked to obesity pathophysiology. Furthermore, another ligand of NMU receptors, NMS (neuromedin S), was identified in 2005. NMS has physiological functions similar to those of NMU. This review summarizes recent observations of the NMU system in relation to the pathophysiology of obesity in both the central nervous systems and the peripheral tissues.
Collapse
|
10
|
Malendowicz LK, Rucinski M. Neuromedins NMU and NMS: An Updated Overview of Their Functions. Front Endocrinol (Lausanne) 2021; 12:713961. [PMID: 34276571 PMCID: PMC8283259 DOI: 10.3389/fendo.2021.713961] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/21/2021] [Indexed: 11/19/2022] Open
Abstract
More than 35 years have passed since the identification of neuromedin U (NMU). Dozens of publications have been devoted to its physiological role in the organism, which have provided insight into its occurrence in the body, its synthesis and mechanism of action at the cellular level. Two G protein-coupled receptors (GPCRs) have been identified, with NMUR1 distributed mainly peripherally and NMUR2 predominantly centrally. Recognition of the role of NMU in the control of energy homeostasis of the body has greatly increased interest in this neuromedin. In 2005 a second, structurally related peptide, neuromedin S (NMS) was identified. The expression of NMS is more restricted, it is predominantly found in the central nervous system. In recent years, further peptides related to NMU and NMS have been identified. These are neuromedin U precursor related peptide (NURP) and neuromedin S precursor related peptide (NSRP), which also exert biological effects without acting via NMUR1, or NMUR2. This observation suggests the presence of another, as yet unrecognized receptor. Another unresolved issue within the NMU/NMS system is the differences in the effects of various NMU isoforms on diverse cell lines. It seems that development of highly specific NMUR1 and NMUR2 receptor antagonists would allow for a more detailed understanding of the mechanisms of action of NMU/NMS and related peptides in the body. They could form the basis for attempts to use such compounds in the treatment of disorders, for example, metabolic disorders, circadian rhythm, stress, etc.
Collapse
|
11
|
Ye Y, Liang Z, Xue L. Neuromedin U: potential roles in immunity and inflammation. Immunology 2021; 162:17-29. [PMID: 32888314 PMCID: PMC7730025 DOI: 10.1111/imm.13257] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 02/05/2023] Open
Abstract
Since the discovery of neuromedin U (NmU) from porcine spinal cord in 1985, this neuropeptide has been subsequently identified in many other species with multiple physiological and pathophysiological roles detected, ranging from smooth muscle contraction, feeding, energy balance to tumorigenesis. Intriguingly, NmU is also emerging to play pro-inflammatory roles involving immune cell activation and cytokine release in a neuron-dependent or neuron-independent manner. The NmU-mediated inflammatory responses have already been observed in worm infection, sepsis, autoimmune arthritis and allergic animal models. In this review, we focus on the roles of NmU in immunity and inflammation by highlighting the interactions between NmU and immune cells, summarizing the signalling mechanism involved in their reactions and discussing its potential contributions to inflammatory diseases.
Collapse
Affiliation(s)
- Yuan Ye
- The Respiratory Medicine UnitOxford NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
- Department of Respiratory and Critical Care MedicineWest China School of Medicine and West China HospitalSichuan UniversityChengduChina
| | - Zongan Liang
- Department of Respiratory and Critical Care MedicineWest China School of Medicine and West China HospitalSichuan UniversityChengduChina
| | - Luzheng Xue
- The Respiratory Medicine UnitOxford NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
| |
Collapse
|
12
|
Yvone GM, Chavez-Martinez CL, Nguyen AR, Wang DJ, Phelps PE. Reelin dorsal horn neurons co-express Lmx1b and are mispositioned in disabled-1 mutant mice. Eur J Neurosci 2020; 52:3322-3338. [PMID: 32492253 PMCID: PMC9451954 DOI: 10.1111/ejn.14847] [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: 08/27/2019] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 11/30/2022]
Abstract
Mice missing either Reelin or Disabled-1 (Dab1) exhibit dorsal horn neuronal positioning errors and display heat hypersensitivity and mechanical insensitivity. Reelin binds its receptors, apolipoprotein E receptor 2 and very low-density lipoprotein receptor, leading to the recruitment and phosphorylation of Dab1 and activation of downstream pathways that regulate neuronal migration. Previously, we reported that 70% of Dab1 laminae I-II neurons co-expressed LIM-homeobox transcription factor 1-beta (Lmx1b). Here, we asked whether Reelin-expressing dorsal horn neurons co-express Lmx1b, are mispositioned in dab1 mutants, and contribute to nociceptive abnormalities. About 90% of Reelin-labeled neurons are Lmx1b-positive in laminae I-II, confirming that most Reelin and Dab1 neurons are glutamatergic. We determined that Reelin-Lmx1b and Dab1-Lmx1b dorsal horn neurons are separate populations, and together, comprise 37% of Lmx1b-positive cells within and above the Isolectin B4 (IB4) layer in wild-type mice. Compared to wild-type mice, dab1 mutants have a reduced area of laminae I-II outer (above the IB4 layer), more Reelin-Lmx1b neurons within the IB4 layer, and fewer Reelin-Lmx1b neurons within the lateral reticulated area of lamina V and lateral spinal nucleus. Interestingly, both Reelin- and Dab1-labeled dorsal horn neurons sustain similar positioning errors in mutant mice. After noxious thermal and mechanical stimulation, Reelin, Lmx1b, and Reelin-Lmx1b neurons expressed Fos in laminae I-II and the lateral reticulated area in wild-type mice and, therefore, participate in nociceptive circuits. Together, our data suggest that disruption of the Reelin-signaling pathway results in neuroanatomical abnormalities that contribute to the nociceptive changes that characterize these mutant mice.
Collapse
Affiliation(s)
- Griselda M Yvone
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | | | - Amanda R Nguyen
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Deborah J Wang
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Patricia E Phelps
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| |
Collapse
|
13
|
Bell AM, Gutierrez-Mecinas M, Stevenson A, Casas-Benito A, Wildner H, West SJ, Watanabe M, Todd AJ. Expression of green fluorescent protein defines a specific population of lamina II excitatory interneurons in the GRP::eGFP mouse. Sci Rep 2020; 10:13176. [PMID: 32764601 PMCID: PMC7411045 DOI: 10.1038/s41598-020-69711-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/15/2020] [Indexed: 01/27/2023] Open
Abstract
Dorsal horn excitatory interneurons that express gastrin-releasing peptide (GRP) are part of the circuit for pruritogen-evoked itch. They have been extensively studied in a transgenic line in which enhanced green fluorescent protein (eGFP) is expressed under control of the Grp gene. The GRP-eGFP cells are separate from several other neurochemically-defined excitatory interneuron populations, and correspond to a class previously defined as transient central cells. However, mRNA for GRP is widely distributed among excitatory interneurons in superficial dorsal horn. Here we show that although Grp mRNA is present in several transcriptomically-defined populations, eGFP is restricted to a discrete subset of cells in the GRP::eGFP mouse, some of which express the neuromedin receptor 2 and likely belong to a cluster defined as Glut8. We show that these cells receive much of their excitatory synaptic input from MrgA3/MrgD-expressing nociceptive/pruritoceptive afferents and C-low threshold mechanoreceptors. Although the cells were not innervated by pruritoceptors expressing brain natriuretic peptide (BNP) most of them contained mRNA for NPR1, the receptor for BNP. In contrast, these cells received only ~ 10% of their excitatory input from other interneurons. These findings demonstrate that the GRP-eGFP cells constitute a discrete population of excitatory interneurons with a characteristic pattern of synaptic input.
Collapse
Affiliation(s)
- Andrew M Bell
- Spinal Cord Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK.
| | - Maria Gutierrez-Mecinas
- Spinal Cord Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK
| | - Anna Stevenson
- Spinal Cord Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK
| | - Adrian Casas-Benito
- Spinal Cord Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland.,Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Steven J West
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, 060-8638, Japan
| | - Andrew J Todd
- Spinal Cord Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK.
| |
Collapse
|
14
|
De Prins A, Allaoui W, Medrano M, Van Eeckhaut A, Ballet S, Smolders I, De Bundel D. Effects of neuromedin U-8 on stress responsiveness and hypothalamus-pituitary-adrenal axis activity in male C57BL/6J mice. Horm Behav 2020; 121:104666. [PMID: 31899262 DOI: 10.1016/j.yhbeh.2019.104666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/29/2019] [Accepted: 12/24/2019] [Indexed: 01/31/2023]
Abstract
Neuromedin U (NMU) is a highly conserved neuropeptide that has been implicated in the stress response. To better understand how it influences various aspects of the stress response, we studied the effects of intracerebroventricular NMU-8 administration on stress-related behavior and activity of the hypothalamus-pituitary-adrenal (HPA) axis in male C57BL/6J mice. We investigated these NMU-8 effects when mice remained in their home cage and when they were challenged by exposure to forced swim stress. NMU-8 administration resulted in increased grooming behavior in mice that remained in their home cage and in a significant increase in c-Fos immunoreactivity in the paraventricular hypothalamus (PVH) and arcuate nucleus (ARC). Surprisingly, NMU-8 administration significantly decreased plasma corticosterone concentrations. Furthermore, NMU-8 administration increased immobility in the forced swim test in both naïve mice and mice that were previously exposed to swim stress. The effect of NMU-8 on c-Fos immunoreactivity in the PVH was dependent on previous exposure to swim stress given that we observed no significant changes in mice exposed for the first time to swim stress. In contrast, in the ARC we observed a significant increase in c-Fos immunoreactivity regardless of previous stress exposure. Interestingly, NMU-8 administration also significantly decreased plasma corticosterone concentrations in mice that were exposed to single forced swim stress, while this effect was no longer observed when mice were exposed to forced swim stress for a second time. Taken together, our data indicate that NMU-8 regulates stress responsiveness and suggests that its effects depend on previous stress exposure.
Collapse
Affiliation(s)
- An De Prins
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Wissal Allaoui
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Mireia Medrano
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ann Van Eeckhaut
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
15
|
Hsiao YT, Jestes KJ, Jackson KL, Zukosky T, Squire ME, Hum JM, Lowery JW. Neuromedin U (NMU) regulates osteoblast differentiation and activity. Biochem Biophys Res Commun 2020; 524:890-894. [PMID: 32057362 DOI: 10.1016/j.bbrc.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/02/2020] [Indexed: 12/12/2022]
Abstract
Osteoporosis is a disease of low bone mass that places individuals at enhanced risk for fracture, disability, and death. Osteoporosis rates are expected to rise significantly in the coming decades yet there are limited pharmacological treatment options, particularly for long-term management of this chronic condition. The drug development pipeline is relatively bereft of new strategies, causing an urgent and unmet need for developing new strategies and targets for treating osteoporosis. Here, we examine a lesser-studied bone remodeling pathway, Neuromedin U (NMU), which is expressed in the bone microenvironment along with its cognate receptors NMU receptor 1 (NMUR1) and 2 (NMUR2). We independently corroborate a prior report that global loss of NMU expression leads to high bone mass and test the hypothesis that NMU negatively regulates osteoblast differentiation. Consistent with this, in vitro studies reveal NMU represses osteoblastic differentiation of osteogenic precursors but, in contrast, promotes osteoblastic marker expression, proliferation and activity of osteoblast-like cells. Phospho-profiling arrays were used to detail differential signaling outcomes that may underlie the opposite responses of these cell types. Collectively, our findings indicate that NMU exerts cell-type-specific responses to regulate osteoblast differentiation and activity.
Collapse
Affiliation(s)
- Yu-Tin Hsiao
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, USA; Bone & Mineral Research Group, Marian University, Indianapolis, IN, USA
| | - Kelli J Jestes
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, USA; Bone & Mineral Research Group, Marian University, Indianapolis, IN, USA
| | - Krista L Jackson
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, USA; Bone & Mineral Research Group, Marian University, Indianapolis, IN, USA
| | - Tara Zukosky
- Department of Biology, The University of Scranton, Scranton, PA, USA
| | - Maria E Squire
- Department of Biology, The University of Scranton, Scranton, PA, USA
| | - Julia M Hum
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, USA; Bone & Mineral Research Group, Marian University, Indianapolis, IN, USA
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, USA; Bone & Mineral Research Group, Marian University, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
16
|
Vestlund J, Kalafateli AL, Studer E, Westberg L, Jerlhag E. Neuromedin U induces self-grooming in socially-stimulated mice. Neuropharmacology 2019; 162:107818. [PMID: 31647973 DOI: 10.1016/j.neuropharm.2019.107818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/17/2019] [Accepted: 10/18/2019] [Indexed: 11/19/2022]
Abstract
Emerging evidence suggest that appetite-regulating peptides modulate social behaviors. We here investigate whether the anorexigenic peptide neuromedin U (NMU) modulates sexual behavior in male mice. However, instead of modulating sexual behaviors, NMU administered into the third ventricle increased self-grooming behavior. In addition, NMU-treatment increased self-grooming behavior when exposed to other mice or olfactory social-cues, but not when exposed to non-social environments. As the neuropeptide oxytocin is released during social investigation and exogenous oxytocin induces self-grooming, its role in NMU-induced self-grooming behavior was investigated. In line with our hypothesis, the oxytocin receptor antagonist inhibited NMU-induced self-grooming behavior in mice exposed to olfactory social-cues. Moreover, dopamine in the mesocorticolimbic system is known to be a key regulator of self-grooming behavior. In line with this, we proved that infusion of NMU into nucleus accumbens increased self-grooming behavior in mice confronted with an olfactory social-cue and that this behavior was inhibited by antagonism of dopamine D2, but not D1/D5, receptors. Moreover repeated NMU treatment enhanced ex vivo dopamine levels and decreased the expression of dopamine D2 receptors in nucleus accumbens in socially housed mice. On the other hand, the olfactory stimuli-dependent NMU-induced self-grooming was not affected by a corticotrophin-releasing hormone antagonist, and NMU-treatment did not influence repetitive behaviors in the marble burying test. In conclusion, our results suggest that NMU treatment and, social cues - potentially triggering oxytocin release - together induce excessive grooming behavior in male mice. The mesolimbic dopamine system, including accumbal dopamine D2 receptors, was identified as a crucial downstream mechanism.
Collapse
Affiliation(s)
- Jesper Vestlund
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Aimilia Lydia Kalafateli
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Studer
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lars Westberg
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
17
|
The YAP1-NMU Axis Is Associated with Pancreatic Cancer Progression and Poor Outcome: Identification of a Novel Diagnostic Biomarker and Therapeutic Target. Cancers (Basel) 2019; 11:cancers11101477. [PMID: 31575084 PMCID: PMC6826421 DOI: 10.3390/cancers11101477] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/11/2019] [Accepted: 09/27/2019] [Indexed: 12/31/2022] Open
Abstract
Yes-associated protein (YAP)-1 is highly upregulated in pancreatic cancer and associated with tumor progression. However, little is known about the role of YAP1 and related genes in pancreatic cancer. Here, we identified target genes regulated by YAP1 and explored their role in pancreatic cancer progression and the related clinical implications. Analysis of different pancreatic cancer databases showed that Neuromedin U (NMU) expression was positively correlated with YAP1 expression in the tumor group. The Cancer Genome Atlas data indicated that high YAP1 and NMU expression levels were associated with poor mean and overall survival. YAP1 overexpression induced NMU expression and transcription and promoted cell motility in vitro and tumor metastasis in vivo via upregulation of epithelial-mesenchymal transition (EMT), whereas specific inhibition of NMU in cells stably expressing YAP1 had the opposite effect in vitro and in vivo. To define this functional association, we identified a transcriptional enhanced associate domain (TEAD) binding site in the NMU promoter and demonstrated that YAP1-TEAD binding upstream of the NMU gene regulated its transcription. These results indicate that the identified positive correlation between YAP1 and NMU is a potential novel drug target and biomarker in metastatic pancreatic cancer.
Collapse
|
18
|
Exome sequencing in a familial form of anorexia nervosa supports multigenic etiology. J Neural Transm (Vienna) 2019; 126:1505-1511. [DOI: 10.1007/s00702-019-02056-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
|
19
|
Peng S, Lu Y, Li P, Liu P, Shi X, Liu C, Zhang Y, Liu S, Wang J. The short interference RNA (siRNA) targeting NMUR2 relieves nociception in a bone cancer pain model of rat through PKC-ERK and PI3K-AKT pathways. Biochem Biophys Res Commun 2019; 512:616-622. [PMID: 30914203 DOI: 10.1016/j.bbrc.2019.03.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/12/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND/AIM Bone cancer pain (BCP) causes troubles and burdens to patients globally. Increasing evidence proved that neuromedin U receptor 2 (NMUR2) was involved in pains. Our study was performed to investigate the role of NMUR2 on BCP and the underlying mechanism. METHODS The rats were raised and BCP rat model was established by injection with Walker 256 cells. The RNA and protein expression levels of NMUR2 in rat neurons-dorsal spinal cord cells, RNdsc cells were detected by qRT-PCR and western blot. The administration with NMUR2 was via intrathecal injection with siRNA to silence NMUR2. The tolerance of rat to pain was measured by mechanical allodynia test and presented by paw withdrawal threshold (PWT) value. The effects on protein kinase C (PKC)/extracellular regulated protein kinases (ERK) and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signal pathways were examined by western blot. RESULTS The expression of NMUR2 in both mRNA and protein levels was upregulated in BCP rat model. In addition, siRNA injection significantly decreased the expression of NMUR2 on the 3rd, 7th and 14th day. BCP group revealed lower PWT value compared with control while NMUR2 silence increased the PWT value compared with negative control. The phosphorylation of PKC, ERK, PI3K and AKT was increased in BCP model while was decreased by si-NMUR2. PKC/ERK and PI3K/AKT inhibitor administration increased the PWT value compared with BCP group. CONCLUSION si-NMUR2 alleviates BCP via inactivation of PKC/ERK and PI3K/AKT signal pathways.
Collapse
Affiliation(s)
- Sheng Peng
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| | - Yingjun Lu
- Department of Anesthesiology, Shanghai Songjiang District Central Hospital, No.748 Zhongshan Middle Road, Shanghai, 201600, China.
| | - Pengyi Li
- Department of Anesthesiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, No.42 Baiziting, Nanjing, 210009, China
| | - Peirong Liu
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| | - Xiaowei Shi
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| | - Chunliang Liu
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| | - Yu Zhang
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| | - Shasha Liu
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| | - Jing Wang
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Shanghai, 200137, China
| |
Collapse
|
20
|
Jarry AC, Merah N, Cisse F, Cayetanot F, Fiamma MN, Willemetz A, Gueddouri D, Barka B, Valet P, Guilmeau S, Bado A, Le Beyec J, Bodineau L, Le Gall M. Neuromedin U is a gut peptide that alters oral glucose tolerance by delaying gastric emptying via direct contraction of the pylorus and vagal-dependent mechanisms. FASEB J 2019; 33:5377-5388. [PMID: 30753087 DOI: 10.1096/fj.201801718r] [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] [Indexed: 12/12/2022]
Abstract
The gut-brain peptide neuromedin U (NMU) decreases food intake and body weight and improves glucose tolerance. Here, we characterized NMU as an enteropeptide and determined how it impacts glucose excursion. NMU was expressed predominantly in the proximal small intestine, and its secretion was triggered by ingestion of a mixed meal. Although a single peripheral injection of NMU in C57BL/6NRj mice prevented the rise of glycemia upon an oral but not an intraperitoneal load of glucose, it unexpectedly prevented insulin secretion, only slightly improved peripheral insulin sensitivity, and barely reduced intestinal glucose absorption. Interestingly, peripheral administration of NMU abrogated gastric emptying. NMU receptors 1 and 2 were detected in pyloric muscles and NMU was able to directly induce pyloric contraction in a dose-dependent manner ex vivo in isometric chambers. Using a modified glucose tolerance test, we demonstrate that improvement of oral glucose tolerance by NMU was essentially, if not exclusively, because of its impact on gastric emptying. Part of this effect was abolished in vagotomized (VagoX) mice, suggesting implication of the vagus tone. Accordingly, peripheral injection of NMU was associated with increased number of c-FOS-positive neurons in the nucleus of the solitary tract, which was partly prevented in VagoX mice. Finally, NMU kept its ability to improve oral glucose tolerance in obese and diabetic murine models. Together, these data demonstrate that NMU is an enteropeptide that prevents gastric emptying directly by triggering pylorus contraction and indirectly through vagal afferent neurons. This blockade consequently reduces intestinal nutrient absorption and thereby results in an apparent improved tolerance to oral glucose challenge.-Jarry, A.-C., Merah, N., Cisse, F., Cayetanot, F., Fiamma, M.-N., Willemetz, A., Gueddouri, D., Barka, B., Valet, P., Guilmeau, S., Bado, A., Le Beyec, J., Bodineau, L., Le Gall, M. Neuromedin U is a gut peptide that alters oral glucose tolerance by delaying gastric emptying via direct contraction of the pylorus and vagal-dependent mechanisms.
Collapse
Affiliation(s)
- Anne-Charlotte Jarry
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France
| | - Nadir Merah
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France
| | - Fanta Cisse
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France
| | - Florence Cayetanot
- Sorbonne Université, INSERM, Unité Mixte de Recherche (UMR) S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Marie-Noëlle Fiamma
- Sorbonne Université, INSERM, Unité Mixte de Recherche (UMR) S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Alexandra Willemetz
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France
| | - Dalale Gueddouri
- INSERM, Unité 1016, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8104, Institut Cochin, Université Paris Descartes, Paris, France
| | - Besma Barka
- Sorbonne Université, INSERM, Unité Mixte de Recherche (UMR) S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Philippe Valet
- INSERM, Unité 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Sandra Guilmeau
- INSERM, Unité 1016, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8104, Institut Cochin, Université Paris Descartes, Paris, France
| | - André Bado
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France
| | - Johanne Le Beyec
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France.,Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Department of Endocrine and Oncological Biochemistry, Paris, France
| | - Laurence Bodineau
- Sorbonne Université, INSERM, Unité Mixte de Recherche (UMR) S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Maude Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1149, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Paris, France
| |
Collapse
|
21
|
Chang X, Pellegrino R, Garifallou J, March M, Snyder J, Mentch F, Li J, Hou C, Liu Y, Sleiman PMA, Hakonarson H. Common variants at 5q33.1 predispose to migraine in African-American children. J Med Genet 2018; 55:831-836. [PMID: 30266756 DOI: 10.1136/jmedgenet-2018-105359] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/16/2018] [Accepted: 08/29/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified multiple susceptibility loci for migraine in European adults. However, no large-scale genetic studies have been performed in children or African Americans with migraine. METHODS We conducted a GWAS of 380 African-American children and 2129 ancestry-matched controls to identify variants associated with migraine. We then attempted to replicate our primary analysis in an independent cohort of 233 African-American patients and 4038 non-migraine control subjects. RESULTS The results of this study indicate that common variants at 5q33.1 associated with migraine risk in African-American children (rs72793414, p=1.94×10-9). The association was validated in an independent study (p=3.87×10-3) for an overall meta-analysis p value of 3.81×10-10. eQTL (Expression quantitative trait loci) analysis of the Genotype-Tissue Expression data also shows the genotypes of rs72793414 were strongly correlated with the mRNA expression levels of NMUR2 at 5q33.1. NMUR2 encodes a G protein-coupled receptor of neuromedin-U (NMU). NMU, a highly conserved neuropeptide, participates in diverse physiological processes of the central nervous system. CONCLUSIONS This study provides new insights into the genetic basis of childhood migraine and allow for precision therapeutic development strategies targeting migraine patients of African-American ancestry.
Collapse
Affiliation(s)
- Xiao Chang
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Renata Pellegrino
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - James Garifallou
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michael March
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - James Snyder
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Frank Mentch
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jin Li
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Cuiping Hou
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Yichuan Liu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Patrick M A Sleiman
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Wan Y, Zhang J, Fang C, Chen J, Li J, Li J, Wu C, Wang Y. Characterization of neuromedin U (NMU), neuromedin S (NMS) and their receptors (NMUR1, NMUR2) in chickens. Peptides 2018; 101:69-81. [PMID: 29288685 DOI: 10.1016/j.peptides.2017.12.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022]
Abstract
Neuromedin U (NMU) and its structurally-related peptide, neuromedin S (NMS), are reported to regulate many physiological processes and their actions are mediated by two NMU receptors (NMUR1, NMUR2) in mammals. However, the information regarding NMU, NMS, and their receptors is limited in birds. In this study, we examined the structure, functionality, and expression of NMS, NMU, NMUR1 and NMUR2 in chickens. The results showed that: 1) chicken (c-) NMU cDNA encodes a 181-amino acid precursor, which may generate two forms of NMU peptide with 9 (cNMU-9) and 25 amino acids (cNMU-25), respectively. 2) Interestingly, two cNMS transcripts encoding two cNMS precursors of different lengths were identified from chicken pituitary, and both cNMS precursors may produce a mature cNMS peptide of 9 amino acids (cNMS-9). 3) cNMU-9, cNMU-25 and cNMS-9 could activate cNMUR1 expressed in HEK293 cells potently, as monitored by three cell-based luciferase reporter systems, indicating that cNMUR1 can act as a receptor common for cNMU and cNMS peptides, whereas cNMUR2 could be potently activated by cNMS-9, but not by cNMU-9/cNMU-25. 4) cNMU and cNMUR1 are widely expressed in chicken tissues with abundant expression noted in the gastrointestinal tract, as detected by quantitative real-time PCR, whereas cNMUR2 expression is mainly restricted to the brain and anterior pituitary, and cNMS is widely expressed in chicken tissues. Collectively, our data helps to elucidate the physiological roles of NMU/NMS peptides in birds and reveal the functional conservation and changes of NMU/NMS-NMUR axis across vertebrates.
Collapse
Affiliation(s)
- Yiping Wan
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China
| | - Jiannan Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China
| | - Chao Fang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China
| | - Junan Chen
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China
| | - Jing Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China
| | - Juan Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China.
| | - Chenlei Wu
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China
| | - Yajun Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China.
| |
Collapse
|
23
|
Nagai H, Kaisho T, Yokoyama K, Asakawa T, Fujita H, Matsumiya K, Noguchi J, Tsuchimori K, Nishizawa N, Kanematsu-Yamaki Y, Dote K, Inooka H, Sakamoto JI, Ohtaki T, Asami T, Takekawa S. Differential effects of selective agonists of neuromedin U1 and U2 receptors in obese and diabetic mice. Br J Pharmacol 2017; 175:359-373. [PMID: 29057457 DOI: 10.1111/bph.14077] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Neuromedin U (NmU) may be a novel target for obesity treatment owing to its anorectic and energy expenditure enhancing effects. Although two receptors, NMU1 and NMU2, are both responsible for the NmU-mediated anti-obesity effects, the receptor agonist with the most appropriate profiles for treating obesity and diabetes in terms of efficacy and safety is as yet unknown. Thus, we developed and evaluated novel NMU1/2 receptor-selective agonists. EXPERIMENTAL APPROACH Efficacy and safety were assessed in mice with diet-induced obesity (DIO) and those with leptin-deficient diabetes (ob/ob) through repeated peripheral administration of selective agonists to NMU1 (NMU-6102) and NMU2 (NMU-2084), along with non-selective NMU1/2 agonists (NMU-0002 and NMU-6014). We also performed immunohistochemistry for c-Fos protein expression in the brain to probe their mechanisms of action. KEY RESULTS Although both non-selective NMU1/2 agonists and the NMU2-selective agonist had high efficacy compared with the NMU1-selective agonist, only the NMU2-selective agonist led to relatively low adverse effects, such as diarrhoea, in DIO mice. However, the non-selective NMU1/2 agonist and the NMU1-selective agonist, but not the NMU2-selective agonist, were effective in diabetic ob/ob mice. Mechanistically, NMU2-selective agonists preferentially activate pro-opiomelanocortin neurons in the hypothalamic arcuate nucleus but not in the paraventricular nucleus. CONCLUSIONS AND IMPLICATIONS These results suggest that an NMU2 receptor-selective agonist may be a well-balanced drug for the treatment of obesity and that an NMU1 receptor-selective agonist may also be beneficial for treating obesity and diabetes once its side effects are minimized.
Collapse
Affiliation(s)
- Hiroaki Nagai
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoko Kaisho
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kotaro Yokoyama
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoko Asakawa
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hisashi Fujita
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kouta Matsumiya
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Jiro Noguchi
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kazue Tsuchimori
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Naoki Nishizawa
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | | | - Katsuko Dote
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroshi Inooka
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | | | - Tetsuya Ohtaki
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Taiji Asami
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shiro Takekawa
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
24
|
Vallöf D, Ulenius L, Egecioglu E, Engel JA, Jerlhag E. Central administration of the anorexigenic peptide neuromedin U decreases alcohol intake and attenuates alcohol-induced reward in rodents. Addict Biol 2017; 22:640-651. [PMID: 26769653 PMCID: PMC6680249 DOI: 10.1111/adb.12355] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/26/2015] [Accepted: 12/01/2015] [Indexed: 01/24/2023]
Abstract
By investigating the neurochemical mechanisms through which alcohol activates the brain reward systems, novel treatment strategies for alcohol use disorder (AUD), a chronic relapsing disease, can be developed. In contrast to the common view of the function of gut-brain peptides, such as neuromedin U (NMU), to regulate food intake and appetite, a novel role in reinforcement mediation has been implied. The anorexigenic effects of NMU are mediated via NMU2 receptors, preferably in the arcuate nucleus and paraventricular nucleus. The expression of NMU2 receptors is also expressed in several reward-related areas in the brain, suggesting a role in reward regulation. The present experiments were therefore set up to investigate the effect of intracerebroventricular administration of NMU on alcohol-mediated behaviors in rodents. We found that central administration of NMU attenuated alcohol-induced locomotor stimulation, accumbal dopamine release and the expression of conditioned place preference in mice. In addition, NMU dose dependently decreased alcohol intake in high, but not in low, alcohol-consuming rats. Central NMU administration did not alter the blood alcohol concentrations nor change the corticosterone levels in rodents. Given that AUD is a major health-care challenge causing an enormous cost to society and novel treatment strategies are warranted, our data suggest that NMU analogues deserve to be evaluated as novel treatment of AUD in humans.
Collapse
Affiliation(s)
- Daniel Vallöf
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Lisa Ulenius
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Emil Egecioglu
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Jörgen A. Engel
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| |
Collapse
|
25
|
McCue DL, Kasper JM, Hommel JD. Regulation of motivation for food by neuromedin U in the paraventricular nucleus and the dorsal raphe nucleus. Int J Obes (Lond) 2017; 41:120-128. [PMID: 27748746 PMCID: PMC5209284 DOI: 10.1038/ijo.2016.178] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 09/02/2016] [Accepted: 09/13/2016] [Indexed: 12/02/2022]
Abstract
BACKGROUND Motivation for high-fat food is thought to contribute to excess caloric intake in obese individuals. A novel regulator of motivation for food may be neuromedin U (NMU), a highly-conserved neuropeptide that influences food intake. Although these effects of NMU have primarily been attributed to signaling in the paraventricular nucleus of the hypothalamus (PVN), NMU has also been found in other brain regions involved in both feeding behavior and motivation. We investigate the effects of NMU on motivation for food and food intake, and identify the brain regions mediating these effects. METHODS The motivational state for a particular reinforcer (e.g., high-fat food) can be assessed using a progressive-ratio schedule of reinforcement under which an increasing number of lever presses are required to obtain subsequent reinforcers. Here, we have used a progressive-ratio operant responding paradigm in combination with an assessment of cumulative food intake to evaluate the effects of NMU administration in rats, and identify the brain regions mediating these effects. RESULTS We found that peripheral administration of NMU decreases operant responding for high-fat food in rats. Evaluation of Fos-like immunoreactivity in response to peripheral NMU indicated the PVN and dorsal raphe nucleus (DRN) as sites of action for NMU. NMU infusion into either region mimics the effects of peripheral NMU on food intake and operant responding for food. NMU-containing projections from the lateral hypothalamus (LH) to the PVN and DRN were identified as an endogenous source of NMU. CONCLUSIONS These results identify the DRN as a site of action for NMU, demonstrate that the LH provides endogenous NMU to the PVN and DRN and implicate NMU signaling in the PVN and DRN as a novel regulator of motivation for high-fat foods.
Collapse
Affiliation(s)
- David L. McCue
- Department of Neuroscience, University of Texas Medical Branch, Galveston, TX 77555-0615
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX 77555-0615
| | - James M. Kasper
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX 77555-0615
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555-0615
| | - Jonathan D. Hommel
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX 77555-0615
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555-0615
| |
Collapse
|
26
|
Schlegel P, Texada MJ, Miroschnikow A, Schoofs A, Hückesfeld S, Peters M, Schneider-Mizell CM, Lacin H, Li F, Fetter RD, Truman JW, Cardona A, Pankratz MJ. Synaptic transmission parallels neuromodulation in a central food-intake circuit. eLife 2016; 5:16799. [PMID: 27845623 PMCID: PMC5182061 DOI: 10.7554/elife.16799] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
NeuromedinU is a potent regulator of food intake and activity in mammals. In Drosophila, neurons producing the homologous neuropeptide hugin regulate feeding and locomotion in a similar manner. Here, we use EM-based reconstruction to generate the entire connectome of hugin-producing neurons in the Drosophila larval CNS. We demonstrate that hugin neurons use synaptic transmission in addition to peptidergic neuromodulation and identify acetylcholine as a key transmitter. Hugin neuropeptide and acetylcholine are both necessary for the regulatory effect on feeding. We further show that subtypes of hugin neurons connect chemosensory to endocrine system by combinations of synaptic and peptide-receptor connections. Targets include endocrine neurons producing DH44, a CRH-like peptide, and insulin-like peptides. Homologs of these peptides are likewise downstream of neuromedinU, revealing striking parallels in flies and mammals. We propose that hugin neurons are part of an ancient physiological control system that has been conserved at functional and molecular level. DOI:http://dx.doi.org/10.7554/eLife.16799.001
Collapse
Affiliation(s)
- Philipp Schlegel
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Michael J Texada
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Anton Miroschnikow
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Andreas Schoofs
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Sebastian Hückesfeld
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | - Marc Peters
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| | | | - Haluk Lacin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Feng Li
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Richard D Fetter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - James W Truman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Albert Cardona
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michael J Pankratz
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
27
|
Chiu CN, Rihel J, Lee DA, Singh C, Mosser EA, Chen S, Sapin V, Pham U, Engle J, Niles BJ, Montz CJ, Chakravarthy S, Zimmerman S, Salehi-Ashtiani K, Vidal M, Schier AF, Prober DA. A Zebrafish Genetic Screen Identifies Neuromedin U as a Regulator of Sleep/Wake States. Neuron 2016; 89:842-56. [PMID: 26889812 DOI: 10.1016/j.neuron.2016.01.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 11/16/2015] [Accepted: 12/24/2015] [Indexed: 12/27/2022]
Abstract
Neuromodulation of arousal states ensures that an animal appropriately responds to its environment and engages in behaviors necessary for survival. However, the molecular and circuit properties underlying neuromodulation of arousal states such as sleep and wakefulness remain unclear. To tackle this challenge in a systematic and unbiased manner, we performed a genetic overexpression screen to identify genes that affect larval zebrafish arousal. We found that the neuropeptide neuromedin U (Nmu) promotes hyperactivity and inhibits sleep in zebrafish larvae, whereas nmu mutant animals are hypoactive. We show that Nmu-induced arousal requires Nmu receptor 2 and signaling via corticotropin releasing hormone (Crh) receptor 1. In contrast to previously proposed models, we find that Nmu does not promote arousal via the hypothalamic-pituitary-adrenal axis, but rather probably acts via brainstem crh-expressing neurons. These results reveal an unexpected functional and anatomical interface between the Nmu system and brainstem arousal systems that represents a novel wake-promoting pathway.
Collapse
Affiliation(s)
- Cindy N Chiu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Daniel A Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Chanpreet Singh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Eric A Mosser
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Shijia Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Viveca Sapin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Uyen Pham
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jae Engle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Brett J Niles
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Christin J Montz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sridhara Chakravarthy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Steven Zimmerman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kourosh Salehi-Ashtiani
- Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Marc Vidal
- Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Division of Sleep Medicine, Harvard University, Cambridge, MA 02138, USA.
| | - David A Prober
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
28
|
Martinez VG, O'Driscoll L. Neuromedin U: a multifunctional neuropeptide with pleiotropic roles. Clin Chem 2015; 61:471-82. [PMID: 25605682 DOI: 10.1373/clinchem.2014.231753] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Neuromedin U (NmU) belongs to the neuromedin family, comprising a series of neuropeptides involved in the gut-brain axis and including neuromedins B and C (bombesin-like), K (neurokinin B), L (neurokinin A or neurotensin), N, S, and U. CONTENT Although initially isolated from porcine spinal cord on the basis of their ability to induce uterine smooth muscle contraction, these peptides have now been found to be expressed in several different tissues and have been ascribed numerous functions, from appetite regulation and energy balance control to muscle contraction and tumor progression. NmU has been detected in several species to date, particularly in mammals (pig, rat, rabbit, dog, guinea pig, human), but also in amphibian, avian, and fish species. The NmU sequence is highly conserved across different species, indicating that this peptide is ancient and plays an important biological role. Here, we summarize the main structural and functional characteristics of NmU and describe its many roles, highlighting the jack-of-all-trades nature of this neuropeptide. SUMMARY NmU involvement in key processes has outlined the possibility that this neuropeptide could be a novel target for the treatment of obesity and cancer, among other disorders. Although the potential for NmU as a therapeutic target is obvious, the multiple functions of this molecule should be taken into account when designing an approach to targeting NmU and/or its receptors.
Collapse
Affiliation(s)
- Vanesa G Martinez
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
29
|
Do DN, Strathe AB, Ostersen T, Jensen J, Mark T, Kadarmideen HN. Genome-wide association study reveals genetic architecture of eating behavior in pigs and its implications for humans obesity by comparative mapping. PLoS One 2013; 8:e71509. [PMID: 23977060 PMCID: PMC3747221 DOI: 10.1371/journal.pone.0071509] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/01/2013] [Indexed: 01/07/2023] Open
Abstract
This study was aimed at identifying genomic regions controlling feeding behavior in Danish Duroc boars and its potential implications for eating behavior in humans. Data regarding individual daily feed intake (DFI), total daily time spent in feeder (TPD), number of daily visits to feeder (NVD), average duration of each visit (TPV), mean feed intake per visit (FPV) and mean feed intake rate (FR) were available for 1130 boars. All boars were genotyped using the Illumina Porcine SNP60 BeadChip. The association analyses were performed using the GenABEL package in the R program. Sixteen SNPs were found to have moderate genome-wide significance (p<5E-05) and 76 SNPs had suggestive (p<5E-04) association with feeding behavior traits. MSI2 gene on chromosome (SSC) 14 was very strongly associated with NVD. Thirty-six SNPs were located in genome regions where QTLs have previously been reported for behavior and/or feed intake traits in pigs. The regions: 64–65 Mb on SSC 1, 124–130 Mb on SSC 8, 63–68 Mb on SSC 11, 32–39 Mb and 59–60 Mb on SSC 12 harbored several signifcant SNPs. Synapse genes (GABRR2, PPP1R9B, SYT1, GABRR1, CADPS2, DLGAP2 and GOPC), dephosphorylation genes (PPM1E, DAPP1, PTPN18, PTPRZ1, PTPN4, MTMR4 and RNGTT) and positive regulation of peptide secretion genes (GHRH, NNAT and TCF7L2) were highly significantly associated with feeding behavior traits. This is the first GWAS to identify genetic variants and biological mechanisms for eating behavior in pigs and these results are important for genetic improvement of pig feed efficiency. We have also conducted pig-human comparative gene mapping to reveal key genomic regions and/or genes on the human genome that may influence eating behavior in human beings and consequently affect the development of obesity and metabolic syndrome. This is the first translational genomics study of its kind to report potential candidate genes for eating behavior in humans.
Collapse
Affiliation(s)
- Duy Ngoc Do
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | | | | | | | | |
Collapse
|
30
|
Rahman AA, Shahid IZ, Pilowsky PM. Neuromedin U causes biphasic cardiovascular effects and impairs baroreflex function in rostral ventrolateral medulla of spontaneously hypertensive rat. Peptides 2013; 44:15-24. [PMID: 23538213 DOI: 10.1016/j.peptides.2013.03.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/15/2013] [Accepted: 03/15/2013] [Indexed: 02/07/2023]
Abstract
Neuromedin U (NMU) causes biphasic cardiovascular and sympathetic responses and attenuates adaptive reflexes in the rostral ventrolateral medulla (RVLM) and spinal cord in normotensive animal. However, the role of NMU in the pathogenesis of hypertension is unknown. The effect of NMU on baseline cardiorespiratory variables in the RVLM and spinal cord were investigated in urethane-anaesthetized, vagotomized and artificially ventilated male spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY). Experiments were also conducted to determine the effects of NMU on somatosympathetic and baroreceptor reflexes in the RVLM of SHR and WKY. NMU injected into the RVLM and spinal cord elicited biphasic response, a brief pressor and sympathoexcitatory response followed by a prolonged depressor and sympathoinhibitory response in both hypertensive and normotensive rat models. The pressor, sympathoexcitatory and sympathoinhibitory responses evoked by NMU were exaggerated in SHR. Phrenic nerve amplitude was also increased following intrathecal or microinjection of NMU into the RVLM of both strains. NMU injection into the RVLM attenuated the somatosympathetic reflex in both SHR and WKY. Baroreflex sensitivity was impaired in SHR at baseline and further impaired following NMU injection into the RVLM. NMU did not affect baroreflex activity in WKY. The present study provides functional evidence that NMU can have an important effect on the cardiovascular and reflex responses that are integrated in the RVLM and spinal cord. A role for NMU in the development and maintenance of essential hypertension remains to be determined.
Collapse
Affiliation(s)
- Ahmed A Rahman
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW 2112, Australia
| | | | | |
Collapse
|
31
|
Gilbert AK, Puma C, Xu X, Laird J. Neuromedin U Receptor 2 does not play a role in the development of neuropathic pain following nerve injury in mice. Eur J Pain 2013; 17:1147-55. [DOI: 10.1002/j.1532-2149.2013.00288.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2012] [Indexed: 11/10/2022]
Affiliation(s)
- A.-K. Gilbert
- AstraZeneca R&D Montreal; CNS&Pain Innovative Medicines Unit; Montreal; Canada
| | - C. Puma
- AstraZeneca R&D Montreal; CNS&Pain Innovative Medicines Unit; Montreal; Canada
| | - X. Xu
- AstraZeneca R&D Mölndal; Mölndal; Sweden
| | | |
Collapse
|
32
|
Panetta R, Meury L, Cao CQ, Puma C, Mennicken F, Cassar PA, Laird J, Groblewski T. Functional genomics of the rat neuromedin U receptor 1 reveals a naturally occurring deleterious allele. Physiol Genomics 2013; 45:89-97. [DOI: 10.1152/physiolgenomics.00070.2012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Neuromedin U (NMU) plays an important role in a number of physiological processes, but the relative contribution of its two known receptors, NMUR1 and NMUR2, is still poorly understood. Here we report the existence of a SNP T1022→A (Val341→Glu) in the third exon of the rat Nmur1 gene that leads to an inactive receptor. This SNP is present within the coding region of the highly conserved NPXXY motif found within all class A type G protein-coupled receptors and translates to an NMUR1 receptor that is not expressed on the cell surface. Genetic analysis of the Nmur1 gene in a population of Sprague-Dawley rats revealed that this strain is highly heterogeneous for the inactivating polymorphism. The loss of functional NMUR1 receptors in Sprague-Dawley rats homozygous for the inactive allele was confirmed by radioligand binding studies on native tissue expressing NMUR1. The physiological relevance of this functional genomics finding was examined in two nociceptive response models. The pronociceptive effects of NMU were abolished in rats lacking functional NMUR1 receptors. The existence of naturally occurring NMUR1-deficient rats provides a novel and powerful tool to investigate the physiological role of NMU and its receptors. Furthermore, it highlights the importance of verifying the NMUR1 single nucleotide polymorphism status for rats used in physiological, pharmacological or toxicological studies conducted with NMUR1 modulators.
Collapse
Affiliation(s)
- Rosemarie Panetta
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| | - Luc Meury
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| | - Chang Qing Cao
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| | - Carole Puma
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| | - Françoise Mennicken
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| | - Paul A. Cassar
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| | - Jennifer Laird
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
- Department of Pharmacology & Experimental Therapeutics and Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Thierry Groblewski
- AstraZeneca Research and Development, CNS & Pain Innovative Medicines Science Unit, Montreal (Ville Saint-Laurent), Quebec, Canada; and
| |
Collapse
|
33
|
Rahman AA, Shahid IZ, Pilowsky PM. Intrathecal neuromedin U induces biphasic effects on sympathetic vasomotor tone, increases respiratory drive and attenuates sympathetic reflexes in rat. Br J Pharmacol 2012; 164:617-31. [PMID: 21488865 DOI: 10.1111/j.1476-5381.2011.01436.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Neuromedin U (NMU) is a brain-gut peptide that plays regulatory roles in feeding, anxiety, smooth muscle contraction, blood flow, pain and adrenocortical function via two receptors, the NMU receptor 1 and NMU receptor 2. NMU has several known functions in the periphery, but its role in central cardiorespiratory regulation remains poorly understood. EXPERIMENTAL APPROACH Experiments were conducted on urethane-anaesthetized, vagotomized and artificially ventilated male Sprague-Dawley rats (n= 42) to determine if NMU modulates sympathetic vasomotor output at the spinal level or modulates baro-, chemo- and somato-sympathetic reflexes. KEY RESULTS Intrathecal (i.t.) injections of NMU (2.5-20 nmol) caused a dose-dependent biphasic response, initially a brief period of hypertension and sympatho-excitation followed by prolonged hypotension and sympatho-inhibition. Peak excitatory as well as inhibitory responses were observed at 20 nmol. NMU (20 nmol) initially increased mean arterial pressure and splanchnic sympathetic nerve activity by 24 mmHg and 27% and then reduced these by 37 mmHg and 47%, respectively. NMU also dose-dependently increased respiratory drive, as indicated by a rise in phrenic nerve amplitude, an increase in neural minute ventilation and a shortening of the inspiratory period. Both sympatho-excitatory peaks of the somato-sympathetic reflex were abolished by i.t. NMU. Pressor, sympatho-excitatory and tachycardiac responses to chemoreceptor activation (100% N₂) were blocked or significantly reduced following i.t. NMU. NMU also reduced barosensitivity. CONCLUSIONS The data demonstrate that NMU, acting in the spinal cord, differentially contributes to the control of sympathetic tone and adaptive sympathetic reflexes.
Collapse
Affiliation(s)
- A A Rahman
- Australian School of Advanced Medicine, Macquarie University, Sydney, Australia
| | | | | |
Collapse
|
34
|
Abstract
G protein-coupled receptors (GPCRs) play important roles in inflammation. Inflammatory cells such as polymorphonuclear leukocytes (PMN), monocytes and macrophages express a large number of GPCRs for classic chemoattractants and chemokines. These receptors are critical to the migration of phagocytes and their accumulation at sites of inflammation, where these cells can exacerbate inflammation but also contribute to its resolution. Besides chemoattractant GPCRs, protease activated receptors (PARs) such as PAR1 are involved in the regulation of vascular endothelial permeability. Prostaglandin receptors play different roles in inflammatory cell activation, and can mediate both proinflammatory and anti-inflammatory functions. Many GPCRs present in inflammatory cells also mediate transcription factor activation, resulting in the synthesis and secretion of inflammatory factors and, in some cases, molecules that suppress inflammation. An understanding of the signaling paradigms of GPCRs in inflammatory cells is likely to facilitate translational research and development of improved anti-inflammatory therapies.
Collapse
|
35
|
Rahman AA, Shahid IZ, Pilowsky PM. Differential Cardiorespiratory and Sympathetic Reflex Responses to Microinjection of Neuromedin U in Rat Rostral Ventrolateral Medulla. J Pharmacol Exp Ther 2012; 341:213-24. [DOI: 10.1124/jpet.111.191254] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
36
|
Malendowicz LK, Ziolkowska A, Rucinski M. Neuromedins U and S involvement in the regulation of the hypothalamo-pituitary-adrenal axis. Front Endocrinol (Lausanne) 2012; 3:156. [PMID: 23227022 PMCID: PMC3514618 DOI: 10.3389/fendo.2012.00156] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/20/2012] [Indexed: 11/26/2022] Open
Abstract
We reviewed neuromedin U (NMU) and neuromedin S (NMS) involvement in the regulation of the hypothalamo-pituitary-adrenal (HPA) axis function. NMU and NMS are structurally related and highly conserved neuropeptides. They exert biological effects via two GPCR receptors designated as NMUR1 and NMUR2 which show differential expression. NMUR1 is expressed predominantly at the periphery, while NMUR2 in the central nervous system. Elements of the NMU/NMS and their receptors network are also expressed in the HPA axis and progress in molecular biology techniques provided new information on their actions within this system. Several lines of evidence suggest that within the HPA axis NMU and NMS act at both hypothalamic and adrenal levels. Moreover, new data suggest that NMU and NMS are involved in central and peripheral control of the stress response.
Collapse
Affiliation(s)
- Ludwik K. Malendowicz
- *Correspondence: Ludwik K. Malendowicz, Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Swięcicki St., 60-781 Poznan, Poland. e-mail:
| | | | | |
Collapse
|
37
|
Nixon JP, Kotz CM, Novak CM, Billington CJ, Teske JA. Neuropeptides controlling energy balance: orexins and neuromedins. Handb Exp Pharmacol 2012:77-109. [PMID: 22249811 DOI: 10.1007/978-3-642-24716-3_4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this chapter, we review the feeding and energy expenditure effects of orexin (also known as hypocretin) and neuromedin. Orexins are multifunctional neuropeptides that affect energy balance by participating in regulation of appetite, arousal, and spontaneous physical activity. Central orexin signaling for all functions originates in the lateral hypothalamus-perifornical area and is likely functionally differentiated based on site of action and on interacting neural influences. The effect of orexin on feeding is likely related to arousal in some ways but is nonetheless a separate neural process that depends on interactions with other feeding-related neuropeptides. In a pattern distinct from other neuropeptides, orexin stimulates both feeding and energy expenditure. Orexin increases in energy expenditure are mainly by increasing spontaneous physical activity, and this energy expenditure effect is more potent than the effect on feeding. Global orexin manipulations, such as in transgenic models, produce energy balance changes consistent with a dominant energy expenditure effect of orexin. Neuromedins are gut-brain peptides that reduce appetite. There are gut sources of neuromedin, but likely the key appetite-related neuromedin-producing neurons are in the hypothalamus and parallel other key anorectic neuropeptide expression in the arcuate to paraventricular hypothalamic projection. As with other hypothalamic feeding-related peptides, hindbrain sites are likely also important sources and targets of neuromedin anorectic action. Neuromedin increases physical activity in addition to reducing appetite, thus producing a consistent negative energy balance effect. Together with the other various neuropeptides, neurotransmitters, neuromodulators, and neurohormones, neuromedin and orexin act in the appetite network to produce changes in food intake and energy expenditure, which ultimately influences the regulation of body weight.
Collapse
Affiliation(s)
- Joshua P Nixon
- Veterans Affairs Medical Center, Research Service (151), Minneapolis, MN, USA
| | | | | | | | | |
Collapse
|
38
|
Peier AM, Desai K, Hubert J, Du X, Yang L, Qian Y, Kosinski JR, Metzger JM, Pocai A, Nawrocki AR, Langdon RB, Marsh DJ. Effects of peripherally administered neuromedin U on energy and glucose homeostasis. Endocrinology 2011; 152:2644-54. [PMID: 21586559 DOI: 10.1210/en.2010-1463] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neuromedin U (NMU) is a highly conserved peptide reported to modulate energy homeostasis. Pharmacological studies have shown that centrally administered NMU inhibits food intake, reduces body weight, and increases energy expenditure. NMU-deficient mice develop obesity, whereas transgenic mice overexpressing NMU become lean and hypophagic. Two high-affinity NMU receptors, NMUR1 and NMUR2, have been identified. NMUR1 is found primarily in the periphery and NMUR2 primarily in the brain, where it mediates the anorectic effects of centrally administered NMU. Given the broad expression pattern of NMU, we evaluated whether peripheral administration of NMU has effects on energy homeostasis. We observed that acute and chronic peripheral administration of NMU in rodents dose-dependently reduced food intake and body weight and that these effects required NMUR1. The anorectic effects of NMU appeared to be partly mediated by vagal afferents. NMU treatment also increased core body temperature and metabolic rate in mice, suggesting that peripheral NMU modulates energy expenditure. Additionally, peripheral administration of NMU significantly improved glucose excursion. Collectively, these data suggest that NMU functions as a peripheral regulator of energy and glucose homeostasis and the development of NMUR1 agonists may be an effective treatment for diabetes and obesity.
Collapse
Affiliation(s)
- Andrea M Peier
- Merck Research Laboratories, Rahway, New Jersey 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Maruyama K, Kaiya H, Miyazato M, Konno N, Wakasugi T, Uchiyama M, Shioda S, Murakami N, Matsuda K. Isolation and characterisation of two cDNAs encoding the neuromedin U receptor from goldfish brain. J Neuroendocrinol 2011; 23:282-91. [PMID: 21182546 DOI: 10.1111/j.1365-2826.2010.02106.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intracerebroventricular administration of neuromedin U (NMU) exerts an anorexigenic effect in a goldfish model. However, little is known about the NMU receptor and its signalling system in fish. In the present study, we isolated and cloned two cDNAs encoding different proteins comprising 429 and 388 amino acid residues from the goldfish brain based on the nucleotide sequences of human NMU receptor 1 (NMU-R1) and receptor 2 (NMU-R2). Hydropathy and phylogenetic analyses suggested that these two proteins were orthologues of NMU-R1 and -R2 of goldfish. We established two human embryonic kidney 293 cell lines stably expressing putative NMU-R1 and -R2, respectively, and showed that NMU induced an increase in intracellular calcium concentration ([Ca(2+)](i)) in these cells. We examined the presence of NMU-R1 and -R2 in the goldfish brain by western blotting analysis using affinity-purified antisera raised against peptide fragments derived from these receptors. NMU-R1-specific and NMU-R2-specific antisera detected a 49-kDa and 45-kDa immunopositive bands, respectively, in the brain extract. The mass of each band corresponded to that of the deduced respective primary structures. Reverse transcriptase-polymerase chain reaction analysis showed that NMU-R1 and -R2 transcripts were detected in several tissues. In particular, both mRNAs were strongly expressed in the goldfish brain. By contrast, NMU-R2 mRNA was also expressed in the gut. These results indicate for the first time that NMU-R orthologues exist in goldfish, and suggest physiological roles of NMU and its receptor system in fish.
Collapse
Affiliation(s)
- K Maruyama
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Budhiraja S, Chugh A. Neuromedin U: physiology, pharmacology and therapeutic potential. Fundam Clin Pharmacol 2009; 23:149-57. [PMID: 19645813 DOI: 10.1111/j.1472-8206.2009.00667.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neuromedin U (NmU), a multifunctional neuropeptide, belongs to a family of neuropeptides, the neuromedins. It is ubiquitously distributed with highest levels found in the gastrointestinal tract and pituitary. The conservation of structural elements of NmU across species, the widespread distribution of NmU and its receptors throughout the body point to a fundamental role in key physiological processes. Two G protein coupled receptors for NmU have been cloned NmU R1 and NmU R2. NmU R1 is expressed pre-dominantly in the periphery especially the gastrointestinal tract whereas NmU R2 is expressed pre-dominantly in the central nervous system. Current evidence suggests a role of NmU in pain, in regulation of feeding and energy homeostasis, stress, cancer, immune mediated inflammatory diseases like asthma, inflammatory diseases, maintaining the biological clock, in the regulation of smooth muscle contraction in the gastrointestinal and genitourinary tract, and in the control of blood flow and blood pressure. With the development of drugs selectively acting on receptors and knockout animal models, exact pathophysiological roles of NmU will become clearer.
Collapse
Affiliation(s)
- S Budhiraja
- Department of Pharmacology, Pt. B. D. Sharma, PGIMS, Rohtak-124001, Haryana, India.
| | | |
Collapse
|
41
|
Egecioglu E, Ploj K, Xu X, Bjursell M, Salomé N, Andersson N, Ohlsson C, Taube M, Hansson C, Bohlooly-Y M, Morgan DGA, Dickson SL. Central NMU signaling in body weight and energy balance regulation: evidence from NMUR2 deletion and chronic central NMU treatment in mice. Am J Physiol Endocrinol Metab 2009; 297:E708-16. [PMID: 19584200 DOI: 10.1152/ajpendo.91022.2008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate the role of the central neuromedin U (NMU) signaling system in body weight and energy balance regulation, we examined the effects of long-term intracerebroventricular (icv) infusion of NMU in C57Bl/6 mice and in mice lacking the gene encoding NMU receptor 2. In diet-induced obese male and female C57BL/6 mice, icv infusion of NMU (8 microg x day(-1) x mouse(-1)) for 7 days decreased body weight and total energy intake compared with vehicle treatment. However, these parameters were unaffected by NMU treatment in lean male and female C57BL/6 mice fed a standard diet. In addition, female (but not male) NMUR2-null mice had increased body weight and body fat mass when fed a high-fat diet but lacked a clear body weight phenotype when fed a standard diet compared with wild-type littermates. Furthermore, female (but not male) NMUR2-null mice fed a high-fat diet were protected from central NMU-induced body weight loss compared with littermate wild-type mice. Thus, we provide the first evidence that long-term central NMU treatment reduces body weight, food intake, and adiposity and that central NMUR2 signaling is required for these effects in female but not male mice.
Collapse
Affiliation(s)
- Emil Egecioglu
- Dept. of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the Univ. of Gothenburg, Medicinaregatan, Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Neuromedin U (NMU) is known to have potent actions on appetite and energy expenditure. Deletion of the NMU gene in mice leads to an obese phenotype, characterized by hyperphagia and decreased energy expenditure. Conversely, transgenic mice that overexpress proNMU exhibit reduced body weight and fat storage. Here, we show that central administration of NMU or the related peptide neuromedin S (NMS) dose-dependently decreases food intake, increases metabolic rate, and leads to significant weight loss in mice. The effects of NMU and NMS on both feeding and metabolism are almost completely lost in mice lacking the putative CNS receptor for NMU and NMS, NMUr2. However, NMUr2 knockout mice do not exhibit overt differences in body weight or energy expenditure compared with wild-type mice, suggesting that the dramatic phenotype of the NMU gene knockout mouse is not due simply to the loss of NMU/NMUr2 signaling. Putative proteolytic cleavage sites indicate that an additional peptide is produced from the NMU precursor protein, which is extremely well conserved between human, mouse, and rat. Here, we demonstrate that this peptide, proNMU(104-136), has a pronounced effect on energy balance in mice. Specifically, central administration of proNMU(104-136) causes a significant but transient ( approximately 4 h) increase in feeding, yet both food intake and body weight are decreased over the following 24 h. proNMU(104-136) administration also significantly increased metabolic rate. These results suggest that proNMU(104-136) is a novel modulator of energy balance and may contribute to the phenotype exhibited by NMU knockout mice.
Collapse
Affiliation(s)
- David A Bechtold
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | |
Collapse
|
43
|
Peier A, Kosinski J, Cox-York K, Qian Y, Desai K, Feng Y, Trivedi P, Hastings N, Marsh DJ. The antiobesity effects of centrally administered neuromedin U and neuromedin S are mediated predominantly by the neuromedin U receptor 2 (NMUR2). Endocrinology 2009; 150:3101-9. [PMID: 19324999 PMCID: PMC2703546 DOI: 10.1210/en.2008-1772] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neuromedin U (NMU) and neuromedin S (NMS) are structurally related neuropeptides that have been reported to modulate energy homeostasis. Pharmacological data have shown that NMU and NMS inhibit food intake when administered centrally and that NMU increases energy expenditure. Additionally, NMU-deficient mice develop obesity, whereas transgenic mice overexpressing NMU are lean and hypophagic. Two high-affinity NMU/NMS receptors, NMUR1 and NMUR2, have been identified. NMUR1 is predominantly expressed in the periphery, whereas NMUR2 is predominantly expressed in the brain, suggesting that the effects of centrally administered NMU and NMS are mediated by NMUR2. To evaluate the role of NMUR2 in the regulation of energy homeostasis, we characterized NMUR2-deficient (Nmur2(-/-)) mice. Nmur2(-/-) mice exhibited a modest resistance to diet-induced obesity that was at least in part due to reduced food intake. Acute central administration of NMU and NMS reduced food intake in wild-type but not in Nmur2(-/-) mice. The effects on activity and core temperature induced by centrally administered NMU were also absent in Nmur2(-/-) mice. Moreover, chronic central administration of NMU and NMS evoked significant reductions in body weight and sustained reductions in food intake in mice. In contrast, Nmur2(-/-) mice were largely resistant to these effects. Collectively, these data demonstrate that the anorectic and weight-reducing actions of centrally administered NMU and NMS are mediated predominantly by NMUR2, suggesting that NMUR2-selective agonists may be useful for the treatment of obesity.
Collapse
Affiliation(s)
- Andrea Peier
- Merck Research Laboratories, Department of Metabolic Disorders, 126 East Lincoln Avenue, RY80L-126, Rahway, New Jersey 07065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mitchell JD, Maguire JJ, Davenport AP. Emerging pharmacology and physiology of neuromedin U and the structurally related peptide neuromedin S. Br J Pharmacol 2009; 158:87-103. [PMID: 19519756 DOI: 10.1111/j.1476-5381.2009.00252.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Neuromedin U (NMU) has been paired with the G-protein-coupled receptors (GPRs) NMU(1) (formerly designated as the orphan GPR66 or FM-3) and NMU(2) (FM-4 or hTGR-1). Recently, a structurally related peptide, neuromedin S (NMS), which shares an amidated C-terminal heptapeptide motif, has been identified in both rat and human, and has been proposed as a second ligand for these receptors. Messenger RNA encoding NMU receptor subtypes shows differential expression: NMU(1) is predominantly expressed in peripheral tissues, particularly the gastrointestinal tract, whereas NMU(2) is abundant within the brain and spinal cord. NMU peptide parallels receptor distribution with highest expression in the gastrointestinal tract and specific structures within the brain, reflecting its major role in the regulation of energy balance. The NMU knockout mouse has an obese phenotype and, in agreement, the Arg165Trp amino acid variant of NMU-25 in humans, which is functionally inactive, co-segregated with childhood-onset obesity. Emerging physiological roles for NMU include vasoconstriction mediated predominantly via NMU(1) with nociception and bone remodelling via NMU(2). The NMU system has also been implicated in the pathogenesis of septic shock and cancers including bladder carcinoma and acute myeloid leukaemia. Intriguingly, NMS is more potent at NMU(2) receptors in vivo where it has similar central actions in suppression of feeding and regulation of circadian rhythms to NMU. Taken together with its vascular actions, NMU may be a functional link between energy balance and the cardiovascular system and may provide a future target for therapies directed against the disorders that comprise metabolic syndrome.
Collapse
Affiliation(s)
- J D Mitchell
- Clinical Pharmacology Unit, University of Cambridge, Level 6 Centre for Clinical Investigation, Cambridge, UK
| | | | | |
Collapse
|
45
|
Liu JJ, Payza K, Huang J, Liu R, Chen T, Coupal M, Laird JMA, Cao CQ, Butterworth J, Lapointe S, Bayrakdarian M, Trivedi S, Bostwick JR. Discovery and Pharmacological Characterization of a Small-Molecule Antagonist at Neuromedin U Receptor NMUR2. J Pharmacol Exp Ther 2009; 330:268-75. [DOI: 10.1124/jpet.109.152967] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
46
|
Abbondanzo SJ, Manfra DJ, Chen SC, Pinzon-Ortiz M, Sun Y, Phillips JE, Laverty M, Vassileva G, Hu W, Yang S, Gustafson EL, Fine JS, Hedrick JA. Nmur1-/- mice are not protected from cutaneous inflammation. Biochem Biophys Res Commun 2008; 378:777-82. [PMID: 19070594 DOI: 10.1016/j.bbrc.2008.11.148] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 11/22/2008] [Indexed: 11/29/2022]
Abstract
Neuromedin U (Nmu) is a neuropeptide expressed primarily in the gastrointestinal tract and central nervous system. Previous reports have identified two G protein-coupled receptors (designated Nmur1 and Nmur2) that bind Nmu. Recent reports suggest that Nmu mediates immune responses involving mast cells, and Nmur1 has been proposed to mediate these responses. In this study, we generated mice with an Nmur1 deletion and then profiled the responses of these mice in a cutaneous inflammation model utilizing complete Freund's adjuvant (CFA). We report here that mice lacking Nmur1 had normal inflammation responses with moderate changes in serum cytokines compared to Nmur1(+/+) littermates. Although differences in IL-6 were observed in mice lacking Nmu peptide, these mice exhibited a normal response to CFA. Our data argues against a major role for Nmur1 in mediating the reported inflammatory functions of NmU.
Collapse
Affiliation(s)
- Susan J Abbondanzo
- Department of Discovery Technology, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Seto K, Sasaki T, Katsunuma K, Kobayashi N, Tanaka K, Tack J. Acotiamide hydrochloride (Z-338), a novel prokinetic agent, restores delayed gastric emptying and feeding inhibition induced by restraint stress in rats. Neurogastroenterol Motil 2008; 20:1051-9. [PMID: 18482254 DOI: 10.1111/j.1365-2982.2008.01135.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Acotiamide hydrochloride (Z-338) is a member of new class prokinetic agents currently being developed for the treatment of functional dyspepsia (FD). DNA microarray analysis showed that acotiamide altered the expressions of stress-related genes such as gamma-aminobutyric acid (GABA) receptors, GABA transporters and neuromedin U (NmU) in the medulla oblongata or hypothalamus after administration of acotiamide. Therefore, effects of acotiamide on stress-related symptoms, delayed gastric emptying and feeding inhibition, in rats were examined. Acotiamide significantly improved both delayed gastric emptying and feeding inhibition in restraint stress-induced model, but did not affect both basal gastric emptying and feeding in intact rats, indicating that acotiamide exerted effects only on gastric emptying and feeding impaired by the stress. On the other hand, mosapride showed significant acceleration of gastric emptying in intact and restraint stress-induced model, and itopride showed no effect on restraint stress-induced delayed gastric emptying. In addition, gene expression of NmU increased by restraint stress was suppressed by administration of acotiamide, while acotiamide had no effect on delayed gastric emptying induced by an intracerebroventricular administration of NmU, suggesting that the suppressive effect of acotiamide on gene expression of NmU might be important to restore delayed gastric emptying or feeding inhibition induced by restraint stress. These findings suggest that acotiamide might play an important role in regulation of stress response. As stress is considered to be a major contributing factor in the development of FD, the observed effects may be relevant for symptom improvement in FD.
Collapse
Affiliation(s)
- K Seto
- Central Research Laboratories, Zeria Pharmaceutical Co Ltd, Saitama, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Mangold C, Ksiazek I, Yun SW, Berger E, Binkert C. Distribution of neuromedin U binding sites in the rat CNS revealed by in vitro receptor autoradiography. Neuropeptides 2008; 42:377-86. [PMID: 18547640 DOI: 10.1016/j.npep.2008.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 04/28/2008] [Accepted: 04/29/2008] [Indexed: 10/22/2022]
Abstract
Neuromedin U (NMU), a neuropeptide implicated in feeding, inflammation, pain control and anxiety-related behaviours, is widely distributed in peripheral organs and the CNS. These effects are thought to be mediated by its receptors NMU(1) and NMU(2). Since its precise sites of interaction in the CNS were to date unknown, we studied the distribution of in vitro binding sites for (125)I-NMU-23 in the rat CNS by receptor autoradiography. High-density specific binding was found in discrete areas of the brain and spinal cord, namely in the limbic system (hippocampal formation, septohippocampal nucleus, indusium griseum, hypothalamus, amygdaloid nuclei), superior colliculus, dorsal raphé, and substantia gelatinosa of the spinal cord. Our findings provide further supportive evidence for a multifunctional role for the peptide in the brain and spinal cord.
Collapse
Affiliation(s)
- C Mangold
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | | | | | | | |
Collapse
|
49
|
Brighton PJ, Wise A, Dass NB, Willars GB. Paradoxical behavior of neuromedin U in isolated smooth muscle cells and intact tissue. J Pharmacol Exp Ther 2008; 325:154-64. [PMID: 18180374 DOI: 10.1124/jpet.107.132803] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuromedin U (NmU) is a neuropeptide showing high levels of structural conservation across different species. Since its discovery in 1985, NmU has been implicated in numerous physiological roles, including smooth muscle contraction, energy homeostasis, stress, intestinal ion transport, pronociception, and circadian rhythm. Two G-protein-coupled receptors have been identified for NmU and cloned from humans, rats, and mice. Recombinantly expressed NmU receptors couple to both Galpha(q/11) and Galpha(i) G-proteins, and NmU binds essentially irreversibly, preventing signaling to repetitive applications of NmU. However, it is unclear whether these properties reflect those of endogenously expressed NmU receptors or how these properties influence the functional consequences of NmU receptor signaling. Here, we have explored the signaling by rat NmU receptors expressed endogenously in cultured rat colonic smooth muscle cells and explore the functional consequence of this signaling by investigating the NmU-mediated contraction of ex vivo rat colonic smooth muscle preparations. We demonstrate that endogenous rat NmU receptors couple to both Galpha(q/11) and Galpha(i) G-proteins. Furthermore, we show complex patterns of Ca(2+) signaling, including oscillations, and provide evidence of essentially irreversible binding of NmU to smooth muscle cells. Challenge of either circular or longitudinal rat isolated colonic smooth muscle preparations with NmU resulted in robust contractions. Stimulation was direct, and paradoxically, repetitive applications of NmU mediated repetitive contractions with no evidence of desensitization, highlighting a major discrepancy in the behavior of NmU in single cells and in intact tissues. The reason for this discrepancy is presently unknown.
Collapse
Affiliation(s)
- Paul J Brighton
- Department of Cell Physiology and Pharmacology, Medical Sciences Building, University of Leicester, University Road, Leicester LE1 9HN, UK
| | | | | | | |
Collapse
|
50
|
Sato S, Hanada R, Kimura A, Abe T, Matsumoto T, Iwasaki M, Inose H, Ida T, Mieda M, Takeuchi Y, Fukumoto S, Fujita T, Kato S, Kangawa K, Kojima M, Shinomiya KI, Takeda S. Central control of bone remodeling by neuromedin U. Nat Med 2007; 13:1234-40. [PMID: 17873881 DOI: 10.1038/nm1640] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Accepted: 08/08/2007] [Indexed: 11/08/2022]
Abstract
Bone remodeling, the function affected in osteoporosis, the most common of bone diseases, comprises two phases: bone formation by matrix-producing osteoblasts and bone resorption by osteoclasts. The demonstration that the anorexigenic hormone leptin inhibits bone formation through a hypothalamic relay suggests that other molecules that affect energy metabolism in the hypothalamus could also modulate bone mass. Neuromedin U (NMU) is an anorexigenic neuropeptide that acts independently of leptin through poorly defined mechanisms. Here we show that Nmu-deficient (Nmu-/-) mice have high bone mass owing to an increase in bone formation; this is more prominent in male mice than female mice. Physiological and cell-based assays indicate that NMU acts in the central nervous system, rather than directly on bone cells, to regulate bone remodeling. Notably, leptin- or sympathetic nervous system-mediated inhibition of bone formation was abolished in Nmu-/- mice, which show an altered bone expression of molecular clock genes (mediators of the inhibition of bone formation by leptin). Moreover, treatment of wild-type mice with a natural agonist for the NMU receptor decreased bone mass. Collectively, these results suggest that NMU may be the first central mediator of leptin-dependent regulation of bone mass identified to date. Given the existence of inhibitors and activators of NMU action, our results may influence the treatment of diseases involving low bone mass, such as osteoporosis.
Collapse
Affiliation(s)
- Shingo Sato
- Department of Orthopaedic Surgery, Graduate School, 21st Century Center of Excellence Program, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8519, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|