1
|
Zhang R, Huang D, Gasparini S, Geerling JC. Efferent projections of Nps-expressing neurons in the parabrachial region. J Comp Neurol 2024; 532:e25629. [PMID: 39031887 DOI: 10.1002/cne.25629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 07/22/2024]
Abstract
In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S, NPS) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray, then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known already about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts roles in threat response and circadian behavior.
Collapse
Affiliation(s)
- Richie Zhang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Dake Huang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Silvia Gasparini
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Joel C Geerling
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
2
|
Shirsath KR, Patil VK, Awathale SN, Goyal SN, Nakhate KT. Pathophysiological and therapeutic implications of neuropeptide S system in neurological disorders. Peptides 2024; 175:171167. [PMID: 38325715 DOI: 10.1016/j.peptides.2024.171167] [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: 12/24/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Neuropeptide S (NPS) is a 20 amino acids-containing neuroactive molecule discovered by the reverse pharmacology method. NPS is detected in specific brain regions like the brainstem, amygdala, and hypothalamus, while its receptor (NPSR) is ubiquitously expressed in the central nervous system (CNS). Besides CNS, NPS and NPSR are also expressed in the peripheral nervous system. NPSR is a G-protein coupled receptor that primarily uses Gq and Gs signaling pathways to mediate the actions of NPS. In animal models of Parkinsonism and Alzheimer's disease, NPS exerts neuroprotective effects. NPS suppresses oxidative stress, anxiety, food intake, and pain, and promotes arousal. NPSR facilitates reward, reinforcement, and addiction-related behaviors. Genetic variation and single nucleotide polymorphism in NPSR are associated with depression, schizophrenia, rheumatoid arthritis, and asthma. NPS interacts with several neurotransmitters including glutamate, noradrenaline, serotonin, corticotropin-releasing factor, and gamma-aminobutyric acid. It also modulates the immune system via augmenting pro-inflammatory cytokines and plays an important role in the pathogenesis of rheumatoid arthritis and asthma. In the present review, we discussed the distribution profile of NPS and NPSR, signaling pathways, and their importance in the pathophysiology of various neurological disorders. We have also proposed the areas where further investigations on the NPS system are warranted.
Collapse
Affiliation(s)
- Kamini R Shirsath
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Vaishnavi K Patil
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Sanjay N Awathale
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Sameer N Goyal
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India.
| |
Collapse
|
3
|
Xing L, Zou X, Yin C, Webb JM, Shi G, Ptáček LJ, Fu YH. Diverse roles of pontine NPS-expressing neurons in sleep regulation. Proc Natl Acad Sci U S A 2024; 121:e2320276121. [PMID: 38381789 PMCID: PMC10907243 DOI: 10.1073/pnas.2320276121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Neuropeptide S (NPS) was postulated to be a wake-promoting neuropeptide with unknown mechanism, and a mutation in its receptor (NPSR1) causes the short sleep duration trait in humans. We investigated the role of different NPS+ nuclei in sleep/wake regulation. Loss-of-function and chemogenetic studies revealed that NPS+ neurons in the parabrachial nucleus (PB) are wake-promoting, whereas peri-locus coeruleus (peri-LC) NPS+ neurons are not important for sleep/wake modulation. Further, we found that a NPS+ nucleus in the central gray of the pons (CGPn) strongly promotes sleep. Fiber photometry recordings showed that NPS+ neurons are wake-active in the CGPn and wake/REM-sleep active in the PB and peri-LC. Blocking NPS-NPSR1 signaling or knockdown of Nps supported the function of the NPS-NPSR1 pathway in sleep/wake regulation. Together, these results reveal that NPS and NPS+ neurons play dichotomous roles in sleep/wake regulation at both the molecular and circuit levels.
Collapse
Affiliation(s)
- Lijuan Xing
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - Xianlin Zou
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - Chen Yin
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - John M. Webb
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - Guangsen Shi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan528400, China
| | - Louis J. Ptáček
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94143
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA94143
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA94143
| | - Ying-Hui Fu
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94143
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA94143
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA94143
| |
Collapse
|
4
|
Angelakos CC, Girven KS, Liu Y, Gonzalez OC, Murphy KR, Jennings KJ, Giardino WJ, Zweifel LS, Suko A, Palmiter RD, Clark SD, Krasnow MA, Bruchas MR, de Lecea L. A cluster of neuropeptide S neurons regulates breathing and arousal. Curr Biol 2023; 33:5439-5455.e7. [PMID: 38056461 PMCID: PMC10842921 DOI: 10.1016/j.cub.2023.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/31/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
Neuropeptide S (NPS) is a highly conserved peptide found in all tetrapods that functions in the brain to promote heightened arousal; however, the subpopulations mediating these phenomena remain unknown. We generated mice expressing Cre recombinase from the Nps gene locus (NpsCre) and examined populations of NPS+ neurons in the lateral parabrachial area (LPBA), the peri-locus coeruleus (peri-LC) region of the pons, and the dorsomedial thalamus (DMT). We performed brain-wide mapping of input and output regions of NPS+ clusters and characterized expression patterns of the NPS receptor 1 (NPSR1). While the activity of all three NPS+ subpopulations tracked with vigilance state, only NPS+ neurons of the LPBA exhibited both increased activity prior to wakefulness and decreased activity during REM sleep, similar to the behavioral phenotype observed upon NPSR1 activation. Accordingly, we found that activation of the LPBA but not the peri-LC NPS+ neurons increased wake and reduced REM sleep. Furthermore, given the extended role of the LPBA in respiration and the link between behavioral arousal and breathing rate, we demonstrated that the LPBA but not the peri-LC NPS+ neuronal activation increased respiratory rate. Together, our data suggest that NPS+ neurons of the LPBA represent an unexplored subpopulation regulating breathing, and they are sufficient to recapitulate the sleep/wake phenotypes observed with broad NPS system activation.
Collapse
Affiliation(s)
- Christopher Caleb Angelakos
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Kasey S Girven
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; University of Washington Center for the Neurobiology of Addiction, Pain, and Emotion, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Yin Liu
- Department of Biochemistry, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Oscar C Gonzalez
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Keith R Murphy
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Kim J Jennings
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - William J Giardino
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Azra Suko
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; University of Washington Center for the Neurobiology of Addiction, Pain, and Emotion, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Richard D Palmiter
- Department of Biochemistry, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Mark A Krasnow
- Department of Biochemistry, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; University of Washington Center for the Neurobiology of Addiction, Pain, and Emotion, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
5
|
Grimstvedt JS, Shelton AM, Hoerder‐Suabedissen A, Oliver DK, Berndtsson CH, Blankvoort S, Nair RR, Packer AM, Witter MP, Kentros CG. A multifaceted architectural framework of the mouse claustrum complex. J Comp Neurol 2023; 531:1772-1795. [PMID: 37782702 PMCID: PMC10953385 DOI: 10.1002/cne.25539] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 10/04/2023]
Abstract
Accurate anatomical characterizations are necessary to investigate neural circuitry on a fine scale, but for the rodent claustrum complex (CLCX), this has yet to be fully accomplished. The CLCX is generally considered to comprise two major subdivisions, the claustrum (CL) and the dorsal endopiriform nucleus (DEn), but regional boundaries to these areas are debated. To address this, we conducted a multifaceted analysis of fiber- and cytoarchitecture, genetic marker expression, and connectivity using mice of both sexes, to create a comprehensive guide for identifying and delineating borders to CLCX, including an online reference atlas. Our data indicated four distinct subregions within CLCX, subdividing both CL and DEn into two. Additionally, we conducted brain-wide tracing of inputs to CLCX using a transgenic mouse line. Immunohistochemical staining against myelin basic protein (MBP), parvalbumin (PV), and calbindin (CB) revealed intricate fiber-architectural patterns enabling precise delineations of CLCX and its subregions. Myelinated fibers were abundant dorsally in CL but absent ventrally, whereas PV expressing fibers occupied the entire CL. CB staining revealed a central gap within CL, also visible anterior to the striatum. The Nr2f2, Npsr1, and Cplx3 genes expressed specifically within different subregions of the CLCX, and Rprm helped delineate the CL-insular border. Furthermore, cells in CL projecting to the retrosplenial cortex were located within the myelin sparse area. By combining own experimental data with digitally available datasets of gene expression and input connectivity, we could demonstrate that the proposed delineation scheme allows anchoring of datasets from different origins to a common reference framework.
Collapse
Affiliation(s)
- Joachim S. Grimstvedt
- Kavli Institute for Systems NeuroscienceNTNU Norwegian University of Science and TechnologyTrondheimNorway
| | - Andrew M. Shelton
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
| | | | - David K. Oliver
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
| | - Christin H. Berndtsson
- Kavli Institute for Systems NeuroscienceNTNU Norwegian University of Science and TechnologyTrondheimNorway
| | - Stefan Blankvoort
- Kavli Institute for Systems NeuroscienceNTNU Norwegian University of Science and TechnologyTrondheimNorway
| | - Rajeevkumar R. Nair
- Kavli Institute for Systems NeuroscienceNTNU Norwegian University of Science and TechnologyTrondheimNorway
| | - Adam M. Packer
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
| | - Menno P. Witter
- Kavli Institute for Systems NeuroscienceNTNU Norwegian University of Science and TechnologyTrondheimNorway
| | - Clifford G. Kentros
- Kavli Institute for Systems NeuroscienceNTNU Norwegian University of Science and TechnologyTrondheimNorway
- Institute of NeuroscienceUniversity of OregonEugeneOregonUSA
| |
Collapse
|
6
|
Akçalı İ, Akkan SS, Bülbül M. The regulatory role of central neuropeptide-S in locomotion. Peptides 2023; 170:171110. [PMID: 37832875 DOI: 10.1016/j.peptides.2023.171110] [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: 07/14/2023] [Revised: 09/13/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Central exogenous Neuropeptide-S (NPS) was demonstrated to increase locomotor activity (LMA) in rodent studies. NPS receptor (NPSR) is produced in locomotion-related brain regions including basal ganglia while NPS mediates dopaminergic neurotransmission suggesting that endogenous brain NPS is involved in the regulation of locomotion. Aim of the study was to elucidate whether antagonism of NPSR impairs locomotion and to determine the neurochemical profile of NPSR-expressing cells in basal ganglia network. In the rats received intracerebroventricular injection of selective non-peptide NPSR antagonist ML154 (20 nmol/5 µL) or vehicle, in addition to measurement of catalepsy, motor performance, and motor coordination were evaluated by assessment of LMA and RR test, respectively. The immunoreactivities for NPSR, tyrosine hydroxylase (TH), glutamate decarboxylase 67 (GAD67), and choline acetyltransferase (ChAT) were detected by immunofluorescence in frozen sections. Compared to the control rats, total LMA was significantly declined following ML154 administration. The ML154-injected rats were more prone to fall in rotarod (RR) test, while they exhibited remarkably high catalepsy time. The most robust immunoreactivity for NPSR was detected in globus pallidus externa (GPe), while moderate levels of NPSR expression were observed in substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA), but not in striatum. The NPSR-ir cell bodies were found to express GAD67 in GPe and TH in SNpc and VTA, respectively. NPSR expression was detected in SNpc-projecting pallidal cells. The present findings indicate the regulatory role of central endogenous NPS in the control of locomotion. NPSR may be a potential therapeutic target for the treatment of movement disorders.
Collapse
Affiliation(s)
- İrem Akçalı
- Department of Physiology, Akdeniz University, Faculty of Medicine, Antalya, Turkey
| | - Simla Su Akkan
- Department of Physiology, Akdeniz University, Faculty of Medicine, Antalya, Turkey
| | - Mehmet Bülbül
- Department of Physiology, Akdeniz University, Faculty of Medicine, Antalya, Turkey.
| |
Collapse
|
7
|
Zhang R, Huang D, Gasparini S, Geerling JC. Efferent projections of Nps-expressing neurons in the parabrachial region. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.13.553140. [PMID: 37645772 PMCID: PMC10462015 DOI: 10.1101/2023.08.13.553140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts a role in threat response and circadian behavior.
Collapse
Affiliation(s)
- Richie Zhang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| | - Dake Huang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| | - Silvia Gasparini
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| | - Joel C. Geerling
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| |
Collapse
|
8
|
Xiang G, Liu X, Wang J, Lu S, Yu M, Zhang Y, Sun B, Huang B, Lu XY, Li X, Zhang D. Peroxisome proliferator-activated receptor-α activation facilitates contextual fear extinction and modulates intrinsic excitability of dentate gyrus neurons. Transl Psychiatry 2023; 13:206. [PMID: 37322045 DOI: 10.1038/s41398-023-02496-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 05/06/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023] Open
Abstract
The dentate gyrus (DG) of the hippocampus encodes contextual information associated with fear, and cell activity in the DG is required for acquisition and extinction of contextual fear. However, the underlying molecular mechanisms are not fully understood. Here we show that mice deficient for peroxisome proliferator-activated receptor-α (PPARα) exhibited a slower rate of contextual fear extinction. Furthermore, selective deletion of PPARα in the DG attenuated, while activation of PPARα in the DG by local infusion of aspirin facilitated extinction of contextual fear. The intrinsic excitability of DG granule neurons was reduced by PPARα deficiency but increased by activation of PPARα with aspirin. Using RNA-Seq transcriptome we found that the transcription level of neuropeptide S receptor 1 (Npsr1) was tightly correlated with PPARα activation. Our results provide evidence that PPARα plays an important role in regulating DG neuronal excitability and contextual fear extinction.
Collapse
Affiliation(s)
- Guo Xiang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012, China
| | - Xia Liu
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
| | - Jiangong Wang
- Institute of Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, 256600, China
| | - Shunshun Lu
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
| | - Meng Yu
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
| | - Yuhan Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012, China
| | - Bin Sun
- National Glycoengineering Research Center, Shandong University, Jinan, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012, China
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012, China.
| |
Collapse
|
9
|
Neuropeptide S facilitates extinction of fear via modulation of mesolimbic dopaminergic circuitry. Neuropharmacology 2022; 221:109274. [DOI: 10.1016/j.neuropharm.2022.109274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022]
|
10
|
Huang D, Zhang R, Gasparini S, McDonough MC, Paradee WJ, Geerling JC. Neuropeptide S (NPS) neurons: Parabrachial identity and novel distributions. J Comp Neurol 2022; 530:3157-3178. [PMID: 36036349 PMCID: PMC9588594 DOI: 10.1002/cne.25400] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 01/05/2023]
Abstract
Neuropeptide S (NPS) increases wakefulness. A small number of neurons in the brainstem express Nps. These neurons are located in or near the parabrachial nucleus (PB), but we know very little about their ontogeny, connectivity, and function. To identify Nps-expressing neurons within the molecular framework of the PB region, we used in situ hybridization, immunofluorescence, and Cre-reporter labeling in mice. The primary concentration of Nps-expressing neurons borders the lateral lemniscus at far-rostral levels of the lateral PB. Caudal to this main cluster, Nps-expressing neurons scatter through the PB and form a secondary concentration medial to the locus coeruleus (LC). Most Nps-expressing neurons in the PB region are Atoh1-derived, Foxp2-expressing, and mutually exclusive with neurons expressing Calca or Lmx1b. Among Foxp2-expressing PB neurons, those expressing Nps are distinct from intermingled subsets expressing Cck or Pdyn. Examining Nps Cre-reporter expression throughout the brain identified novel populations of neurons in the nucleus incertus, anterior hypothalamus, and lateral habenula. This information will help focus experimental questions about the connectivity and function of NPS neurons.
Collapse
Affiliation(s)
- Dake Huang
- Department of NeurologyUniversity of IowaIowa CityIowa
| | - Richie Zhang
- Department of NeurologyUniversity of IowaIowa CityIowa
| | | | | | | | | |
Collapse
|
11
|
The association between genetic variability in the NPS/NPSR1 system and chronic stress responses: A gene-environment-(quasi-) experiment. Psychoneuroendocrinology 2022; 144:105883. [PMID: 35914393 DOI: 10.1016/j.psyneuen.2022.105883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022]
Abstract
The neuropeptide S (NPS) and its receptor (NPSR1) have been implicated in stress regulation and stress-related disorders. The present study aimed at investigating the association between overall genetic variability in the NPS/NPSR1 system and psychological and cortisol stress regulation in everyday life. Our study was conceptualized as a gene-environment-(quasi-) experiment, a design that facilitates the detection of true GxE interactions. As environmental variable, we used the preparation for the first state examination for law students. In the prospective and longitudinal LawSTRESS project, students were examined at six sampling points over a 13-months period. While students who prepared for the exam and experienced long-lasting and significant stress, formed the stress group, law students experiencing usual study-related workload were assigned to the control group. As phenotypes we assessed changes over time in the cortisol awakening response (CAR; n = 176), perceived stress levels (n = 401), and anxiety symptoms (n = 397). The CAR was assessed at each sampling point immediately upon awakening and 30 as well as 45 min later. Perceived stress levels in daily life were measured by repeated ambulatory assessments and anxiety symptoms were repeatedly assessed with the anxiety subscale of the Hospital Anxiety and Depression Scale. With gene-set analyses we examined the joint association of 936 NPS/NPSR1 single nucleotide polymorphisms with the phenotypes to overcome well known limitations of candidate gene studies. As previously reported, we found a blunted CAR during the exam as well as significant increases in perceived stress levels and anxiety symptoms until the exam in the stress group, compared to the control group. The gene-set analysis did not confirm associations between genetic variability in the NPS/NPSR1 system and changes in perceived stress levels and anxiety symptoms. Regarding the CAR, we found a significant GxE interaction for the area under the curve with respect to the ground (p = .050) and a trend towards a significant effect for the area under the curve with respect to the increase (p = .054). When the analysis was restricted to the SG, associations for both CAR parameters were significant (ps < .050). This finding suggests that the association between genetic variability in the NPS/NPSR1 system and the CAR becomes visible under the environmental condition 'chronic stress exposure'. We conclude that the present study complements findings from animal models and that it provides novel evidence for a modulatory influence of the NPS/NPSR1 system on cortisol regulation in humans.
Collapse
|
12
|
Li C, Wu XJ, Li W. Neuropeptide S promotes maintenance of newly formed dendritic spines and performance improvement after motor learning in mice. Peptides 2022; 156:170860. [PMID: 35970276 DOI: 10.1016/j.peptides.2022.170860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/18/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Neuropeptide S (NPS), an endogenous neuropeptide consisting of 20 amino acids, selectively binds and activates G protein-coupled receptor named neuropeptide S receptor (NPSR) to regulate a variety of physiological functions. NPS/NPSR system has been shown to play a pivotal role in regulating learning and memory in rodents. However, it remains unclear that how NPS/NPSR system affects neuronal functions and synaptic plasticity after learning. We found that intracerebroventricular (i.c.v.) injection of NPS promoted performance improvement and reduced sleep duration after motor learning, which could be blocked by pre-treatment with intraperitoneal (i.p.) injection of NPSR antagonist SHA 68. Using intravital two-photon imaging, we examined the effect of NPS on the postsynaptic dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex after motor learning. We found that i.c.v. injection of NPS strengthened learning-induce new spines and facilitated their survival over time. Furthermore, i.c.v. injection of NPS increased calcium activity of apical dendrites and dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex during the running period. These findings suggest that activation of NPSR by NPS increases synaptic calcium activity and learning-related synapse maintenance, thereby contributing to performance improvement after motor learning.
Collapse
Affiliation(s)
- Cong Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Xu-Jun Wu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Wei Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| |
Collapse
|
13
|
Gołyszny M, Zieliński M, Paul-Samojedny M, Pałasz A, Obuchowicz E. Chronic treatment with escitalopram and venlafaxine affects the neuropeptide S pathway differently in adult Wistar rats exposed to maternal separation. AIMS Neurosci 2022; 9:395-422. [PMID: 36329901 PMCID: PMC9581731 DOI: 10.3934/neuroscience.2022022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/20/2022] [Accepted: 09/05/2022] [Indexed: 07/05/2024] Open
Abstract
Neuropeptide S (NPS), which is a peptide that is involved in the regulation of the stress response, seems to be relevant to the mechanism of action of antidepressants that have anxiolytic properties. However, to date, there have been no reports regarding the effect of long-term treatment with escitalopram or venlafaxine on the NPS system under stress conditions. This study aimed to investigate the effects of the above-mentioned antidepressants on the NPS system in adult male Wistar rats that were exposed to neonatal maternal separation (MS). Animals were exposed to MS for 360 min. on postnatal days (PNDs) 2-15. MS causes long-lasting behavioral, endocrine and neurochemical consequences that mimic anxiety- and depression-related features. MS and non-stressed rats were given escitalopram or venlafaxine (10mg/kg) IP from PND 69 to 89. The NPS system was analyzed in the brainstem, hypothalamus, amygdala and anterior olfactory nucleus using quantitative RT-PCR and immunohistochemical methods. The NPS system was vulnerable to MS in the brainstem and amygdala. In the brainstem, escitalopram down-regulated NPS and NPS mRNA in the MS rats and induced a tendency to reduce the number of NPS-positive cells in the peri-locus coeruleus. In the MS rats, venlafaxine insignificantly decreased the NPSR mRNA levels in the amygdala and a number of NPSR cells in the basolateral amygdala, and increased the NPS mRNA levels in the hypothalamus. Our data show that the studied antidepressants affect the NPS system differently and preliminarily suggest that the NPS system might partially mediate the pharmacological effects that are induced by these drugs.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Monika Paul-Samojedny
- Department of Medical Genetics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| |
Collapse
|
14
|
Markiewicz-Gospodarek A, Markiewicz R, Dobrowolska B, Rahnama M, Łoza B. Relationship of Neuropeptide S (NPS) with Neurocognitive, Clinical, and Electrophysiological Parameters of Patients during Structured Rehabilitation Therapy for Schizophrenia. J Clin Med 2022; 11:jcm11185266. [PMID: 36142912 PMCID: PMC9506378 DOI: 10.3390/jcm11185266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/29/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction: Neuropeptide S is a biomarker related to various neuropsychiatric and neurocognitive functions. Since the need to improve cognitive functions in schizophrenia is unquestionable, it was valuable to investigate the possible relationships of plasma levels of NPS with neurocognitive, psychopathological and EEG parameters in patients with schizophrenia. Aim: Relationships between the serum NPS level and neurocognitive, clinical, and electrophysiological parameters were investigated in patients diagnosed with schizophrenia who underwent structured rehabilitation therapy. Methods: Thirty-three men diagnosed with schizophrenia were randomized into two groups. The REH group (N16) consisted of patients who underwent structured rehabilitation therapy, the CON group (N17) continued its previous treatment. Additionally, the reference NPS serum results were checked in a group of healthy people (N15). In the study several tests assessing various neurocognitive functions were used: d2 Sustained-Attention Test (d2), Color Trails Test (CTT), Beck Cognitive Insight Scale (BCIS), Acceptance of Illness Scale (AIS), and General Self-Efficacy Scale (GSES). The clinical parameters were measured with Positive and Negative Syndrome Scale (PANSS) and electrophysiological parameters were analyzed with auditory evoked potentials (AEPs) and quantitative electroencephalography (QEEG). The NPS, neurocognitive, clinical, and electrophysiological results of REH and CON groups were recorded at the beginning (T1) and after a period of 3 months (T2). Results: A decreased level of NPS was associated with the improvement in specific complex indices of d2 and BCIS neurocognitive tests, as well as the improvement in the clinical state (PANSS). No correlation was observed between the level of NPS and the results of AEPs and QEEG measurements. Conclusions: A decreased level of NPS is possibly related to the improvement in metacognition and social cognition domains, as well as to clinical improvement during the rehabilitation therapy of patients with schizophrenia.
Collapse
Affiliation(s)
| | - Renata Markiewicz
- Department of Neurology, Neurological and Psychiatric Nursing, Medical University of Lublin, 20-093 Lublin, Poland
| | - Beata Dobrowolska
- Department of Holistic Care and Management in Nursing, Medical University of Lublin, 20-081 Lublin, Poland
| | - Mansur Rahnama
- Department of Oral Surgery, Medical University of Lublin, 20-093 Lublin, Poland
| | - Bartosz Łoza
- Department of Psychiatry, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
15
|
Zheng L, Zhang L. The molecular mechanism of natural short sleep: A path towards understanding why we need to sleep. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Sleep constitutes a third of human life and it is increasingly recognized as important for health. Over the past several decades, numerous genes have been identified to be involved in sleep regulation in animal models, but most of these genes when disturbed impair not only sleep but also health and physiological functions. Human natural short sleepers are individuals with lifelong short sleep and no obvious adverse outcomes associated with the lack of sleep. These traits appear to be heritable, and thus characterization of the genetic basis of natural short sleep provides an opportunity to study not only the genetic mechanism of human sleep but also the relationship between sleep and physiological function. This review focuses on the current understanding of mutations associated with the natural short sleep trait and the mechanisms by which they contribute to this trait.
Collapse
Affiliation(s)
- Liubin Zheng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, Hubei, China
| |
Collapse
|
16
|
A Role for Neuropeptide S in Alcohol and Cocaine Seeking. Pharmaceuticals (Basel) 2022; 15:ph15070800. [PMID: 35890099 PMCID: PMC9317571 DOI: 10.3390/ph15070800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 01/25/2023] Open
Abstract
The neuropeptide S (NPS) is the endogenous ligand of the NPS receptor (NPSR). The NPSR is widely expressed in brain regions that process emotional and affective behavior. NPS possesses a unique physio-pharmacological profile, being anxiolytic and promoting arousal at the same time. Intracerebroventricular NPS decreased alcohol consumption in alcohol-preferring rats with no effect in non-preferring control animals. This outcome is most probably linked to the anxiolytic properties of NPS, since alcohol preference is often associated with high levels of basal anxiety and intense stress-reactivity. In addition, NPSR mRNA was overexpressed during ethanol withdrawal and the anxiolytic-like effects of NPS were increased in rodents with a history of alcohol dependence. In line with these preclinical findings, a polymorphism of the NPSR gene was associated with anxiety traits contributing to alcohol use disorders in humans. NPS also potentiated the reinstatement of cocaine and ethanol seeking induced by drug-paired environmental stimuli and the blockade of NPSR reduced reinstatement of cocaine-seeking. Altogether, the work conducted so far indicates the NPS/NPSR system as a potential target to develop new treatments for alcohol and cocaine abuse. An NPSR agonist would be indicated to help individuals to quit alcohol consumption and to alleviate withdrawal syndrome, while NPSR antagonists would be indicated to prevent relapse to alcohol- and cocaine-seeking behavior.
Collapse
|
17
|
Garau C, Liu X, Calo G, Schulz S, Reinscheid RK. Neuropeptide S Encodes Stimulus Salience in the Paraventricular Thalamus. Neuroscience 2022; 496:83-95. [PMID: 35710064 DOI: 10.1016/j.neuroscience.2022.06.013] [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: 03/13/2022] [Revised: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
Evaluation of stimulus salience is critical for any higher organism, as it allows for prioritizing of vital information, preparation of responses, and formation of valuable memory. The paraventricular nucleus of the thalamus (PVT) has recently been identified as an integrator of stimulus salience but the neurochemical basis and afferent input regarding salience signaling have remained elusive. Here we report that neuropeptide S (NPS) signaling in the PVT is necessary for stimulus salience encoding, including aversive, neutral and reinforcing sensory input. Taking advantage of a striking deficit of both NPS receptor (NPSR1) and NPS precursor knockout mice in fear extinction or novel object memory formation, we demonstrate that intra-PVT injections of NPS can rescue the phenotype in NPS precursor knockout mice by increasing the salience of otherwise low-intensity stimuli, while intra-PVT injections of NPSR1 antagonist in wild type mice partially replicates the knockout phenotype. The PVT appears to provide stimulus salience encoding in a dose- and NPS-dependent manner. PVT NPSR1 neurons recruit the nucleus accumbens shell and structures in the prefrontal cortex and amygdala, which were previously linked to the brain salience network. Overall, these results demonstrate that stimulus salience encoding is critically associated with NPS activity in the PVT.
Collapse
Affiliation(s)
- Celia Garau
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92617, USA
| | - Xiaobin Liu
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92617, USA
| | - Girolamo' Calo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Friedrich-Schiller University, Jena, Germany
| | - Rainer K Reinscheid
- Institute of Pharmacology and Toxicology, Friedrich-Schiller University, Jena, Germany.
| |
Collapse
|
18
|
Relationship of Neuropeptide S with Clinical and Metabolic Parameters of Patients during Rehabilitation Therapy for Schizophrenia. Brain Sci 2022; 12:768. [PMID: 35741653 PMCID: PMC9221542 DOI: 10.3390/brainsci12060768&set/a 869781119+878628306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Neuropeptide S (NPS) is a factor associated with the central regulation of body weight, stress, anxiety, learning, memory consolidation, wakefulness-sleep cycle, and anti-inflammatory and neuroplastic effects. Its stress-reducing, anti-anxiety, arousal without anxiety, and pro-cognitive effects represent an interesting option for the treatment of neuropsychiatric disorders. The purpose of the study was to examine the potential associations of NPS levels in the blood with clinical and metabolic parameters during the rehabilitation therapy of patients with schizophrenia. Thirty-three male subjects diagnosed with schizophrenia were randomly divided into two groups. The rehabilitation group (REH, N16) consisted of patients who were subjected to structured, 3-month intensive rehabilitation therapy, and the control group (CON, N17) consisted of patients who were subjected to a standard support mechanism. Both groups continued their pharmacological treatment as usual. The NPS concentration, as well as clinical and metabolic parameters, were compared in both groups. Additionally, a group of healthy (H) males (N15) was tested for NPS reference scores. To look for the specificity and selectivity of the NPS relationship with clinical results, various factor models of the positive and negative syndrome scale (PANSS) were analyzed, including the original PANSS 2/3 model, its modified four-factor version, the male-specific four-factor model, and two five-factorial models validated in large groups in clinical and multi-ethnic studies. Results and conclusions: (1) Structured rehabilitation therapy, compared to unstructured supportive therapy, significantly reduced the level of schizophrenia disorders defined by various factor models derived from PANSS. (2) The clinical improvement within the 3-month rehabilitation therapy course was correlated with a significant decrease in neuropeptide S (NPS) serum level. (3) The excitement/Hostility (E/H) factor, which included schizophrenic symptoms of the psychotic disorganization, was specific and selective for the reduction in serum NPS, which was stable across all analyzed factor models. (4) The long-term relationship between serum NPS and clinical factors was not accompanied by basic metabolic parameters.
Collapse
|
19
|
Piwowarczyk-Nowak A, Pałasz A, Bogus K, Krzystanek M, Błaszczyk I, Worthington JJ, Grajoszek A. Modulatory effect of long-term treatment with escitalopram and clonazepam on the expression of anxiety-related neuropeptides: neuromedin U, neuropeptide S and their receptors in the rat brain. Mol Biol Rep 2022; 49:9041-9049. [PMID: 35690686 DOI: 10.1007/s11033-022-07578-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/17/2022] [Accepted: 05/06/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Newly identified multifunctional peptidergic modulators of stress responses: neuromedin U (NMU) and neuropeptide S (NPS) are involved in the wide spectrum of brain functions. However, there are no reports dealing with potential molecular relationships between the action of diverse anxiolytic or antidepressant drugs and NMU and NPS signaling in the brain. The present work was therefore focused on local expression of the aforementioned stress-related neuropeptides in the rat brain after long-term treatment with escitalopram and clonazepam. METHODS Studies were carried out on adult, male Sprague-Dawley rats that were divided into 3 groups: animals injected with saline (control) and experimental individuals treated with escitalopram (at single dose 5 mg/kg daily), and clonazepam (at single dose 0.5 mg/kg). All individuals were sacrificed under anaesthesia and the whole brain excised. Total mRNA was isolated from homogenized samples of amygdala, hippocampus, hypothalamus, thalamus, cerebellum and brainstem. Real time-PCR method was used for estimation of related NPS, NPS receptor (NPSR), NMU, NMU and receptor 2 (NMUR2) mRNA expression. The whole brains were also sliced for general immunohistochemical assessment of the neuropeptides expression. RESULTS Chronic administration of clonazepam resulted in an increase of NMU mRNA expression and formation of NMU-expressing fibers in the amygdala, while escitalopram produced a significant decrease in NPSR mRNA level in hypothalamus. Long-term escitalopram administration affects the local expression of examined neuropeptides mRNA in a varied manner depending on the brain structure. CONCLUSIONS Pharmacological effects of escitalopram may be connected with local at least partially NPSR-related alterations in the NPS/NMU/NMUR2 gene expression at the level 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.
| | - Katarzyna Bogus
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, 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 Katowice 40- 635, Katowice, Poland
| | - Iwona Błaszczyk
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - John J Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YQ, Lancaster, UK
| | - Aniela Grajoszek
- Department for Experimental Medicine, Medical University of Silesia, ul. Medyków 4, 40-752, Katowice, Poland
| |
Collapse
|
20
|
Relationship of Neuropeptide S with Clinical and Metabolic Parameters of Patients during Rehabilitation Therapy for Schizophrenia. Brain Sci 2022. [DOI: 10.3390/brainsci12060768
expr 958893762 + 814326274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Neuropeptide S (NPS) is a factor associated with the central regulation of body weight, stress, anxiety, learning, memory consolidation, wakefulness–sleep cycle, and anti-inflammatory and neuroplastic effects. Its stress-reducing, anti-anxiety, arousal without anxiety, and pro-cognitive effects represent an interesting option for the treatment of neuropsychiatric disorders. The purpose of the study was to examine the potential associations of NPS levels in the blood with clinical and metabolic parameters during the rehabilitation therapy of patients with schizophrenia. Thirty-three male subjects diagnosed with schizophrenia were randomly divided into two groups. The rehabilitation group (REH, N16) consisted of patients who were subjected to structured, 3-month intensive rehabilitation therapy, and the control group (CON, N17) consisted of patients who were subjected to a standard support mechanism. Both groups continued their pharmacological treatment as usual. The NPS concentration, as well as clinical and metabolic parameters, were compared in both groups. Additionally, a group of healthy (H) males (N15) was tested for NPS reference scores. To look for the specificity and selectivity of the NPS relationship with clinical results, various factor models of the positive and negative syndrome scale (PANSS) were analyzed, including the original PANSS 2/3 model, its modified four-factor version, the male-specific four-factor model, and two five-factorial models validated in large groups in clinical and multi-ethnic studies. Results and conclusions: (1) Structured rehabilitation therapy, compared to unstructured supportive therapy, significantly reduced the level of schizophrenia disorders defined by various factor models derived from PANSS. (2) The clinical improvement within the 3-month rehabilitation therapy course was correlated with a significant decrease in neuropeptide S (NPS) serum level. (3) The excitement/Hostility (E/H) factor, which included schizophrenic symptoms of the psychotic disorganization, was specific and selective for the reduction in serum NPS, which was stable across all analyzed factor models. (4) The long-term relationship between serum NPS and clinical factors was not accompanied by basic metabolic parameters.
Collapse
|
21
|
Relationship of Neuropeptide S with Clinical and Metabolic Parameters of Patients during Rehabilitation Therapy for Schizophrenia. Brain Sci 2022; 12:brainsci12060768. [PMID: 35741653 PMCID: PMC9221542 DOI: 10.3390/brainsci12060768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 01/09/2023] Open
Abstract
Neuropeptide S (NPS) is a factor associated with the central regulation of body weight, stress, anxiety, learning, memory consolidation, wakefulness–sleep cycle, and anti-inflammatory and neuroplastic effects. Its stress-reducing, anti-anxiety, arousal without anxiety, and pro-cognitive effects represent an interesting option for the treatment of neuropsychiatric disorders. The purpose of the study was to examine the potential associations of NPS levels in the blood with clinical and metabolic parameters during the rehabilitation therapy of patients with schizophrenia. Thirty-three male subjects diagnosed with schizophrenia were randomly divided into two groups. The rehabilitation group (REH, N16) consisted of patients who were subjected to structured, 3-month intensive rehabilitation therapy, and the control group (CON, N17) consisted of patients who were subjected to a standard support mechanism. Both groups continued their pharmacological treatment as usual. The NPS concentration, as well as clinical and metabolic parameters, were compared in both groups. Additionally, a group of healthy (H) males (N15) was tested for NPS reference scores. To look for the specificity and selectivity of the NPS relationship with clinical results, various factor models of the positive and negative syndrome scale (PANSS) were analyzed, including the original PANSS 2/3 model, its modified four-factor version, the male-specific four-factor model, and two five-factorial models validated in large groups in clinical and multi-ethnic studies. Results and conclusions: (1) Structured rehabilitation therapy, compared to unstructured supportive therapy, significantly reduced the level of schizophrenia disorders defined by various factor models derived from PANSS. (2) The clinical improvement within the 3-month rehabilitation therapy course was correlated with a significant decrease in neuropeptide S (NPS) serum level. (3) The excitement/Hostility (E/H) factor, which included schizophrenic symptoms of the psychotic disorganization, was specific and selective for the reduction in serum NPS, which was stable across all analyzed factor models. (4) The long-term relationship between serum NPS and clinical factors was not accompanied by basic metabolic parameters.
Collapse
|
22
|
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
|
23
|
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
|
24
|
Markiewicz-Gospodarek A, Kuszta P, Baj J, Dobrowolska B, Markiewicz R. Can Neuropeptide S Be an Indicator for Assessing Anxiety in Psychiatric Disorders? Front Public Health 2022; 10:872430. [PMID: 35558538 PMCID: PMC9087177 DOI: 10.3389/fpubh.2022.872430] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/29/2022] [Indexed: 12/01/2022] Open
Abstract
Neuropeptide S (NPS) is a neuropeptide primarily produced within three brainstem regions including locus coeruleus, trigeminal nerve nucleus, and lateral parabrachial nucleus. NPS is involved in the central regulation of stress, fear, and cognitive integration. NPS is a mediator of behavior, seeking food, and the proliferation of new adipocytes in the setting of obesity. So far, current research of NPS is only limited to animal models; data regarding its functions in humans is still scarce. Animal studies showed that anxiety and appetite might be suppressed by the action of NPS. The discovery of this neuromodulator peptide is effective considering its strong anxiolytic action, which has the potential to be an interesting therapeutic option in treating neuropsychiatric disorders. In this article, we aimed to analyze the pharmaceutical properties of NPS as well as its influence on several neurophysiological aspects-modulation of behavior, association with obesity, as well as its potential application in rehabilitation and treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | - Piotr Kuszta
- Students Scientific Association at the Department of Human Anatomy, Medical University of Lublin, Lublin, Poland
| | - Jacek Baj
- Department of Human Anatomy, Medical University of Lublin, Lublin, Poland
| | - Beata Dobrowolska
- Department of Holistic Care and Management in Nursing, Medical University of Lublin, Lublin, Poland
| | - Renata Markiewicz
- Department of Neurology, Neurological and Psychiatric Nursing, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
25
|
Bülbül M, Sinen O. The influence of early-life and adulthood stressors on brain neuropeptide-S system. Neuropeptides 2022; 92:102223. [PMID: 34982971 DOI: 10.1016/j.npep.2021.102223] [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: 06/06/2021] [Revised: 12/07/2021] [Accepted: 12/25/2021] [Indexed: 11/18/2022]
Abstract
Central administered neuropeptide-S (NPS) was shown to reduce stress response in rodents. This study aimed to investigate the alterations in NPS system upon chronic exposure to early-life and adulthood stressors. Newborn pups underwent maternal separation (MS) from postnatal day 1 to 14 comprised of daily 3-h separations. In the adulthood, 90-min of restraint stress was loaded to males as an acute stress (AS) model. For chronic homotypic stress (CHS), same stressor was applied for 5 consecutive days. The changes in the expression and the release of NPS were monitored by immunohistochemistry and microdialysis, respectively. Throughout the CHS, heart rate variability (HRV) was analyzed on a daily basis. The immunoreactivity for NPS receptor (NPSR) was detected in basolateral amygdala (BLA) and hypothalamic paraventricular nucleus (PVN) by immunofluorescence staining. The NPS expression in the brainstem was increased upon AS which was more prominent following CHS, whereas these responses were found to be blunted in MS counterparts. Similar to histological data, the stress-induced release of NPS in BLA was attenuated in MS rats. CHS-induced elevations in sympatho-vagal balance were alleviated in control rats; which was not observed in MS rats. The expression of NPSR in BLA and PVN was down-regulated in MS rats. The brain NPS/NPSR system appears to be susceptible to the early-life stressors and the subsequent chronic stress exposure in adulthood which results in altered autonomic outflow.
Collapse
Affiliation(s)
- Mehmet Bülbül
- Department of Physiology, Akdeniz University, Faculty of Medicine, Antalya, Turkey.
| | - Osman Sinen
- Department of Physiology, Akdeniz University, Faculty of Medicine, Antalya, Turkey
| |
Collapse
|
26
|
Shao YF, Wang C, Rao XP, Wang HD, Ren YL, Li J, Dong CY, Xie JF, Yang XW, Xu FQ, Hou YP. Neuropeptide S Attenuates the Alarm Pheromone-Evoked Defensive and Risk Assessment Behaviors Through Activation of Cognate Receptor-Expressing Neurons in the Posterior Medial Amygdala. Front Mol Neurosci 2022; 14:752516. [PMID: 35002616 PMCID: PMC8739225 DOI: 10.3389/fnmol.2021.752516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/09/2021] [Indexed: 11/24/2022] Open
Abstract
Neuropeptide S (NPS) acts by activating its cognate receptor (NPSR). High level expression of NPSR in the posterior medial amygdala suggests that NPS-NPSR system should be involved in regulation of social behaviors induced by social pheromones. The present study was undertaken to investigate the effects of central administration of NPS or with NPSR antagonist on the alarm pheromone (AP)-evoked defensive and risk assessment behaviors in mice. Furthermore, H129-H8, a novel high-brightness anterograde multiple trans-synaptic virus, c-Fos and NPSR immunostaining were employed to reveal the involved neurocircuits and targets of NPS action. The mice exposed to AP displayed an enhancement in defensive and risk assessment behaviors. NPS (0.1–1 nmol) intracerebroventricular (i.c.v.) injection significantly attenuated the AP-evoked defensive and risk assessment behaviors. NPSR antagonist [D-Val5]NPS at the dose of 40 nmol completely blocked the effect of 0.5 nmol of NPS which showed the best effective among dose range. The H129-H8-labeled neurons were observed in the bilateral posterodorsal medial amygdala (MePD) and posteroventral medial amygdala (MePV) 72 h after the virus injection into the unilateral olfactory bulb (OB), suggesting that the MePD and MePV receive olfactory information inputs from the OB. The percentage of H129-H8-labeled neurons that also express NPSR were 90.27 ± 3.56% and 91.67 ± 2.46% in the MePD and MePV, respectively. NPS (0.5 nmol, i.c.v.) remarkably increased the number of Fos immunoreactive (-ir) neurons in the MePD and MePV, and the majority of NPS-induced Fos-ir neurons also expressed NPSR. The behavior characteristic of NPS or with [D-Val5]NPS can be better replicated in MePD/MePV local injection within lower dose. The present findings demonstrated that NPS, via selective activation of the neurons bearing NPSR in the posterior medial amygdala, attenuates the AP-evoked defensive and risk assessment behaviors in mice.
Collapse
Affiliation(s)
- Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Lab of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| | - Can Wang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Ping Rao
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Hua-Dong Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Yan-Li Ren
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing Li
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chao-Yu Dong
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jun-Fan Xie
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xing-Wen Yang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fu-Qiang Xu
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Lab of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
27
|
Fang C, Zhang J, Wan Y, Li Z, Qi F, Dang Y, Li J, Wang Y. Neuropeptide S (NPS) and its receptor (NPSR1) in chickens: cloning, tissue expression, and functional analysis. Poult Sci 2021; 100:101445. [PMID: 34634709 PMCID: PMC8507198 DOI: 10.1016/j.psj.2021.101445] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/12/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022] Open
Abstract
Neuropeptide S (NPS) and its receptor neuropeptide S receptor 1 (NPSR1) have been suggested to regulate many physiological processes in the central nervous system (CNS), such as arousal, anxiety, and food intake in mammals and birds, however, the functionality and tissue expression of this NPS-NPSR1 system remain unknown in birds. Here, we cloned NPS and NPSR1 cDNAs from the chicken brain and reported their functionality and tissue expression. The cloned chicken NPS is predicted to encode a mature NPS peptide of 20 amino acids, which shows a remarkable sequence identity (∼94%) among tetrapod species examined, while NPSR1 encodes a receptor of 373 amino acids conserved across vertebrates. Using cell-based luciferase reporter systems, we demonstrated that chicken NPS could potently activate NPSR1 expressed in vitro and thus stimulates multiple signaling pathways, including calcium mobilization, cyclic adenosine monophosphate/protein kinase A (cAMP/PKA), and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling pathways, indicating that NPS actions could be mediated by NPSR1 in birds. Quantitative real-time PCR revealed that NPS and NPSR1 are widely expressed in chicken tissues, including the hypothalamus, and NPSR1 expression is likely controlled by a promoter upstream exon 1, which shows strong promoter activities in cultured DF-1 cells. Taken together, our data provide the first proof that the avian NPS-NPSR1 system is functional and helps to explore the conserved role of NPS and NPSR1 signaling in tetrapods.
Collapse
Affiliation(s)
- Chao Fang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China; The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiannan Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Yiping Wan
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Zejiao Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Feiyang Qi
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Yuanhao Dang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Juan Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Yajun Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
28
|
Identification of a Novel Neuropeptide S Receptor Antagonist Scaffold Based on the SHA-68 Core. Pharmaceuticals (Basel) 2021; 14:ph14101024. [PMID: 34681248 PMCID: PMC8538004 DOI: 10.3390/ph14101024] [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: 08/03/2021] [Revised: 09/22/2021] [Accepted: 09/25/2021] [Indexed: 11/16/2022] Open
Abstract
Activation of the neuropeptide S receptor (NPSR) system has been shown to produce anxiolytic-like actions, arousal, and enhance memory consolidation, whereas blockade of the NPSR has been shown to reduce relapse to substances of abuse and duration of anesthetics. We report here the discovery of a novel core scaffold (+) N-benzyl-3-(2-methylpropyl)-1-oxo-3-phenyl-1H,3H,4H,5H,6H,7H-furo[3,4-c]pyridine-5-carboxamide with potent NPSR antagonist activity in vitro. Pharmacokinetic parameters demonstrate that 14b reaches pharmacologically relevant levels in plasma and the brain following intraperitoneal (i.p.) administration, but is cleared rapidly from plasma. Compound 14b was able to block NPS (0.3 nmol)-stimulated locomotor activity in C57/Bl6 mice at 3 mg/kg (i.p.), indicating potent in vivo activity for the structural class. This suggests that 14b can serve as a useful tool for continued mapping of the pharmacological functions of the NPS receptor system.
Collapse
|
29
|
Tobinski AM, Rappeneau V. Role of the Neuropeptide S System in Emotionality, Stress Responsiveness and Addiction-Like Behaviours in Rodents: Relevance to Stress-Related Disorders. Pharmaceuticals (Basel) 2021; 14:ph14080780. [PMID: 34451877 PMCID: PMC8400992 DOI: 10.3390/ph14080780] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/22/2022] Open
Abstract
The neuropeptide S (NPS) and its receptor (NPSR1) have been extensively studied over the last two decades for their roles in locomotion, arousal/wakefulness and anxiety-related and fear-related behaviours in rodents. However, the possible implications of the NPS/NPSR1 system, especially those of the single nucleotide polymorphism (SNP) rs324981, in stress-related disorders and substance abuse in humans remain unclear. This is possibly due to the fact that preclinical and clinical research studies have remained separated, and a comprehensive description of the role of the NPS/NPSR1 system in stress-relevant and reward-relevant endpoints in humans and rodents is lacking. In this review, we describe the role of the NPS/NPSR1 system in emotionality, stress responsiveness and addiction-like behaviour in rodents. We also summarize the alterations in the NPS/NPSR1 system in individuals with stress-related disorders, as well as the impact of the SNP rs324981 on emotion, stress responses and neural activation in healthy individuals. Moreover, we discuss the therapeutic potential and possible caveats of targeting the NPS/NPSR1 system for the treatment of stress-related disorders. The primary goal of this review is to highlight the importance of studying some rodent behavioural readouts modulated by the NPS/NPSR1 system and relevant to stress-related disorders.
Collapse
|
30
|
Holanda VAD, Didonet JJ, Costa MBB, do Nascimento Rangel AH, da Silva ED, Gavioli EC. Neuropeptide S Receptor as an Innovative Therapeutic Target for Parkinson Disease. Pharmaceuticals (Basel) 2021; 14:ph14080775. [PMID: 34451872 PMCID: PMC8401573 DOI: 10.3390/ph14080775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 11/17/2022] Open
Abstract
Parkinson disease (PD) is a neurodegenerative disease mainly characterized by the loss of nigral dopaminergic neurons in the substantia nigra pars compacta. Patients suffering from PD develop severe motor dysfunctions and a myriad of non-motor symptoms. The treatment mainly consists of increasing central dopaminergic neurotransmission and alleviating motor symptoms, thus promoting severe side effects without modifying the disease’s progress. A growing body of evidence suggests a close relationship between neuropeptide S (NPS) and its receptor (NPSR) system in PD: (i) double immunofluorescence labeling studies showed that NPSR is expressed in the nigral tyrosine hydroxylase (TH)-positive neurons; (ii) central administration of NPS increases spontaneous locomotion in naïve rodents; (iii) central administration of NPS ameliorates motor and nonmotor dysfunctions in animal models of PD; (iv) microdialysis studies showed that NPS stimulates dopamine release in naïve and parkinsonian rodents; (v) central injection of NPS decreases oxidative damage to proteins and lipids in the rodent brain; and, (vi) 7 days of central administration of NPS protects from the progressive loss of nigral TH-positive cells in parkinsonian rats. Taken together, the NPS/NPSR system seems to be an emerging therapeutic strategy for alleviating motor and non-motor dysfunctions of PD and, possibly, for slowing disease progress.
Collapse
Affiliation(s)
- Victor A. D. Holanda
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN 59078-900, Brazil; (V.A.D.H.); (J.J.D.); (M.B.B.C.); (E.D.d.S.J.)
| | - Julia J. Didonet
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN 59078-900, Brazil; (V.A.D.H.); (J.J.D.); (M.B.B.C.); (E.D.d.S.J.)
| | - Manara B. B. Costa
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN 59078-900, Brazil; (V.A.D.H.); (J.J.D.); (M.B.B.C.); (E.D.d.S.J.)
| | | | - Edilson D. da Silva
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN 59078-900, Brazil; (V.A.D.H.); (J.J.D.); (M.B.B.C.); (E.D.d.S.J.)
| | - Elaine C. Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN 59078-900, Brazil; (V.A.D.H.); (J.J.D.); (M.B.B.C.); (E.D.d.S.J.)
- Correspondence:
| |
Collapse
|
31
|
Sinen O, Bülbül M, Derin N, Ozkan A, Akcay G, Aslan MA, Agar A. The effect of chronic neuropeptide-S treatment on non-motor parameters in experimental model of Parkinson's disease. Int J Neurosci 2021; 131:765-774. [PMID: 32441169 DOI: 10.1080/00207454.2020.1754213] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/28/2020] [Accepted: 03/28/2020] [Indexed: 10/24/2022]
Abstract
AIM Besides motor impairment, non-motor symptoms including cognitive decline, anxiety, and depression are observed in Parkinson's Disease (PD). The aim of this study was to investigate whether chronic administration of central neuropeptide-S (NPS) improves non-motor symptoms in 6-hydroxydopamine (6-OHDA)-induced parkinsonian rats. MATERIAL AND METHODS Experimental PD was utilized by unilateral stereotaxic injection of the 6-OHDA into the medial forebrain bundle (MFB), while the sham-operated animals underwent the same surgical procedures. NPS (1 nmol) or vehicle was daily administered through an intracerebroventricular (icv) cannula for 7 days. Radial arm maze (RAM) test was used to evaluate the working memory; whereas, elevated plus maze (EPM) test and sucrose preference test were used to monitor the anxiety and depression status, respectively. The levels of dopamine, glutamic acid, and glutamine was determined in harvested striatal and hippocampal tissue samples. The immunoreactivities for tyrosine hydroxylase (TH) was determined using immunohistochemistry. RESULTS In the RAM test, the 6-OHDA-induced increases in the reference and working memory errors were reduced by the central NPS administration. The decreased sucrose preference in the parkinsonian rats was increased by centrally administered NPS. The levels of dopamine levels in striatum and hippocampus were decreased in the parkinsonian rats, however, they were not altered by the centrally administered NPS. Additionally, NPS treatment significantly attenuated the 6-OHDA-induced loss of TH neuronal number. CONCLUSION Consequently, NPS appears to be a therapeutic candidate for the treatment of non-motor complications of PD.
Collapse
Affiliation(s)
- Osman Sinen
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Mehmet Bülbül
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Narin Derin
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Ayse Ozkan
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Guven Akcay
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Mutay Aydın Aslan
- Faculty of Medicine, Department of Medical Biochemistry, Akdeniz University, Antalya, Turkey
| | - Aysel Agar
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| |
Collapse
|
32
|
Bengoetxea X, Goedecke L, Remmes J, Blaesse P, Grosch T, Lesting J, Pape HC, Jüngling K. Human-Specific Neuropeptide S Receptor Variants Regulate Fear Extinction in the Basal Amygdala of Male and Female Mice Depending on Threat Salience. Biol Psychiatry 2021; 90:145-155. [PMID: 33902914 DOI: 10.1016/j.biopsych.2021.02.967] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/31/2021] [Accepted: 02/19/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND A nonsynonymous single nucleotide polymorphism in the neuropeptide S receptor 1 (NPSR1) gene (rs324981) results in isoleucine-to-asparagine substitution at amino acid 107. In humans, the ancestral variant (NPSR1 I107) is associated with increased anxiety sensitivity and risk of panic disorder, while the human-specific variant (NPSR1 N107) is considered protective against excessive anxiety. In rodents, neurobiological constituents of the NPS system have been analyzed in detail and their anxiolytic-like effects have been endorsed. However, their implication for anxiety and related disorders in humans remains unclear, as rodents carry only the ancestral NPSR1 I107 variant. METHODS We hypothesized that phenotypic correlates of NPSR1 variants manifest in fear-related circuits in the amygdala. We used CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/Cas9)-mediated gene editing to generate a "humanized" mouse strain, in which individuals express either NPSR1 I107 or NPSR1 N107. RESULTS Stimulation of NPSR1 evoked excitatory responses in principal neurons of the anterior basal amygdala with significant differences in magnitude between genotypes, resulting in synaptic disinhibition of putative extinction neurons in the posterior basal amygdala in mice expressing the human-specific hypofunctional N107 but not the ancestral I107 variant. N107 mice displayed improved extinction of conditioned fear, which was phenocopied after pharmacological antagonism of NPSR1 in the anterior basal amygdala of I107 mice. Differences in fear extinction between male and female mice were related to an interaction of Npsr1 genotype and salience of fear training. CONCLUSIONS The NPS system regulates extinction circuits in the amygdala depending on the Npsr1 genotype, contributing to sex-specific differences in fear extinction and high anxiety sensitivity of individuals bearing the ancestral NPSR1 I107 variant.
Collapse
Affiliation(s)
- Xabier Bengoetxea
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Lena Goedecke
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Jasmin Remmes
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Peter Blaesse
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Grosch
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Jörg Lesting
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Kay Jüngling
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany.
| |
Collapse
|
33
|
Dissociative Effects of Neuropeptide S Receptor Deficiency and Nasal Neuropeptide S Administration on T-Maze Discrimination and Reversal Learning. Pharmaceuticals (Basel) 2021; 14:ph14070643. [PMID: 34358069 PMCID: PMC8308873 DOI: 10.3390/ph14070643] [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: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
Cognitive flexibility refers to the ability to modify learned behavior in response to changes in the environment. In laboratory rodents, cognitive flexibility can be assessed in reversal learning, i.e., the change of contingencies, for example in T-maze discrimination learning. The present study investigated the role of the neuropeptide S (NPS) system in cognitive flexibility. In the first experiment, mice deficient of NPS receptors (NPSR) were tested in T-maze discrimination and reversal learning. In the second experiment, C57BL/6J mice were tested in the T-maze after nasal administration of NPS. Finally, the effect of nasal NPS on locomotor activity was evaluated. NPSR deficiency positively affected the acquisition of T-maze discrimination but had no effects on reversal learning. Nasal NPS administration facilitated reversal learning and supported an allocentric learning strategy without affecting acquisition of the task or locomotor activity. Taken together, the present data show that the NPS system is able to modulate both acquisition of T-maze discrimination and its reversal learning. However, NPSR deficiency only improved discrimination learning, while nasal NPS administration only improved reversal learning, i.e., cognitive flexibility. These effects, which at first glance appear to be contradictory, could be due to the different roles of the NPS system in the brain regions that are important for learning and cognitive flexibility.
Collapse
|
34
|
Park S, Flüthmann P, Wolany C, Goedecke L, Spenner HM, Budde T, Pape HC, Jüngling K. Neuropeptide S Receptor Stimulation Excites Principal Neurons in Murine Basolateral Amygdala through a Calcium-Dependent Decrease in Membrane Potassium Conductance. Pharmaceuticals (Basel) 2021; 14:ph14060519. [PMID: 34072275 PMCID: PMC8230190 DOI: 10.3390/ph14060519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 02/08/2023] Open
Abstract
Background: The neuropeptide S system, consisting of the 20 amino acid neuropeptide NPS and its G-protein-coupled receptor (GPCR) neuropeptide S receptor 1 (NPSR1), has been studied intensively in rodents. Although there is a lot of data retrieved from behavioral studies using pharmacology or genetic interventions, little is known about intracellular signaling cascades in neurons endogenously expressing the NPSR1. Methods: To elucidate possible G-protein-dependent signaling and effector systems, we performed whole-cell patch-clamp recordings on principal neurons of the anterior basolateral amygdala of mice. We used pharmacological interventions to characterize the NPSR1-mediated current induced by NPS application. Results: Application of NPS reliably evokes inward-directed currents in amygdalar neurons recorded in brain slice preparations of male and female mice. The NPSR1-mediated current had a reversal potential near the potassium reversal potential (EK) and was accompanied by an increase in membrane input resistance. GDP-β-S and BAPTA, but neither adenylyl cyclase inhibition nor 8-Br-cAMP, abolished the current. Intracellular tetraethylammonium or 4-aminopyridine reduced the NPS-evoked current. Conclusion: NPSR1 activation in amygdalar neurons inhibits voltage-gated potassium (K+) channels, most likely members of the delayed rectifier family. Intracellularly, Gαq signaling and calcium ions seem to be mandatory for the observed current and increased neuronal excitability.
Collapse
|
35
|
Chou Y, Hor CC, Lee MT, Lee H, Guerrini R, Calo G, Chiou L. Stress induces reinstatement of extinguished cocaine conditioned place preference by a sequential signaling via neuropeptide S, orexin, and endocannabinoid. Addict Biol 2021; 26:e12971. [PMID: 33078457 DOI: 10.1111/adb.12971] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Neurons containing neuropeptide S (NPS) and orexins are activated during stress. Previously, we reported that orexins released during stress, via orexin OX1 receptors (OX1 Rs), contribute to the reinstatement of cocaine seeking through endocannabinoid/CB1 receptor (CB1 R)-mediated dopaminergic disinhibition in the ventral tegmental area (VTA). Here, we further demonstrated that NPS released during stress is an up-stream activator of this orexin-endocannabinoid cascade in the VTA, leading to the reinstatement of cocaine seeking. Mice were trained to acquire cocaine conditioned place preference (CPP) by context-pairing cocaine injections followed by the extinction training with context-pairing saline injections. Interestingly, the extinguished cocaine CPP in mice was significantly reinstated by intracerebroventricular injection (i.c.v.) of NPS (1 nmol) in a manner prevented by intraperitoneal injection (i.p.) of SHA68 (50 mg/kg), an NPS receptor antagonist. This NPS-induced cocaine reinstatement was prevented by either i.p. or intra-VTA microinjection (i.vta.) of SB-334867 (15 mg/kg, i.p. or 15 nmol, i.vta.) and AM 251 (1.1 mg/kg, i.p. or 30 nmol, i.vta.), antagonists of OX1 Rs and CB1 Rs, respectively. Besides, NPS (1 nmol, i.c.v.) increased the number of c-Fos-containing orexin neurons in the lateral hypothalamus (LH) and increased orexin-A level in the VTA. The latter effect was blocked by SHA68. Furthermore, a 30-min restraint stress in mice reinstated extinguished cocaine CPP and was prevented by SHA68. These results suggest that NPS is released upon stress and subsequently activates LH orexin neurons to release orexins in the VTA. The released orexins then reinstate extinguished cocaine CPP via an OX1 R- and endocannabinoid-CB1 R-mediated signaling in the VTA.
Collapse
Affiliation(s)
- Yu‐Hsien Chou
- Graduate Institute of Pharmacology, College of Medicine National Taiwan University Taipei Taiwan
| | - Chia Chun Hor
- Graduate Institute of Pharmacology, College of Medicine National Taiwan University Taipei Taiwan
| | - Ming Tatt Lee
- Graduate Institute of Pharmacology, College of Medicine National Taiwan University Taipei Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine National Taiwan University Taipei Taiwan
- Faculty of Pharmaceutical Sciences UCSI University Kuala Lumpur Malaysia
| | - Hsin‐Jung Lee
- Graduate Institute of Pharmacology, College of Medicine National Taiwan University Taipei Taiwan
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA) University of Ferrara Ferrara Italy
| | - Girolamo Calo
- Section of Pharmacology, Department of Medical Sciences University of Ferrara Ferrara Italy
| | - Lih‐Chu Chiou
- Graduate Institute of Pharmacology, College of Medicine National Taiwan University Taipei Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine National Taiwan University Taipei Taiwan
- Graduate Institute of Acupuncture Sciences China Medical University Taichung Taiwan
| |
Collapse
|
36
|
Reinscheid RK, Ruzza C. Pharmacology, Physiology and Genetics of the Neuropeptide S System. Pharmaceuticals (Basel) 2021; 14:ph14050401. [PMID: 33922620 PMCID: PMC8146834 DOI: 10.3390/ph14050401] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 12/28/2022] Open
Abstract
The Neuropeptide S (NPS) system is a rather ‘young’ transmitter system that was discovered and functionally described less than 20 years ago. This review highlights the progress that has been made in elucidating its pharmacology, anatomical distribution, and functional involvement in a variety of physiological effects, including behavior and immune functions. Early on, genetic variations of the human NPS receptor (NPSR1) have attracted attention and we summarize current hypotheses of genetic linkage with disease and human behaviors. Finally, we review the therapeutic potential of future drugs modulating NPS signaling. This review serves as an introduction to the broad collection of original research papers and reviews from experts in the field that are presented in this Special Issue.
Collapse
Affiliation(s)
- Rainer K. Reinscheid
- Institute of Pharmacology & Toxicology, University Hospital Jena, Friedrich-Schiller University, 07747 Jena, Germany
- Institute of Physiology I, University Hospital Münster, Westfälische-Wilhelms University, 48149 Münster, Germany
- Correspondence: (R.K.R.); (C.R.)
| | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (R.K.R.); (C.R.)
| |
Collapse
|
37
|
Albanese V, Ruzza C, Marzola E, Bernardi T, Fabbri M, Fantinati A, Trapella C, Reinscheid RK, Ferrari F, Sturaro C, Calò G, Amendola G, Cosconati S, Pacifico S, Guerrini R, Preti D. Structure-Activity Relationship Studies on Oxazolo[3,4- a]pyrazine Derivatives Leading to the Discovery of a Novel Neuropeptide S Receptor Antagonist with Potent In Vivo Activity. J Med Chem 2021; 64:4089-4108. [PMID: 33733768 PMCID: PMC8041306 DOI: 10.1021/acs.jmedchem.0c02223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neuropeptide S modulates important neurobiological functions including locomotion, anxiety, and drug abuse through interaction with its G protein-coupled receptor known as neuropeptide S receptor (NPSR). NPSR antagonists are potentially useful for the treatment of substance abuse disorders against which there is an urgent need for new effective therapeutic approaches. Potent NPSR antagonists in vitro have been discovered which, however, require further optimization of their in vivo pharmacological profile. This work describes a new series of NPSR antagonists of the oxazolo[3,4-a]pyrazine class. The guanidine derivative 16 exhibited nanomolar activity in vitro and 5-fold improved potency in vivo compared to SHA-68, a reference pharmacological tool in this field. Compound 16 can be considered a new tool for research studies on the translational potential of the NPSergic system. An in-depth molecular modeling investigation was also performed to gain new insights into the observed structure-activity relationships and provide an updated model of ligand/NPSR interactions.
Collapse
Affiliation(s)
- Valentina Albanese
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Via Fossato di Mortara 17/19, 44121 Ferrara, Italy
- LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, 44121 Ferrara, Italy
| | - Erika Marzola
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Tatiana Bernardi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Martina Fabbri
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Anna Fantinati
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Claudio Trapella
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
- LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, 44121 Ferrara, Italy
| | - Rainer K Reinscheid
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
- Institute of Physiology I, University Hospital Münster, University of Münster, 48149 Münster, Germany
| | - Federica Ferrari
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Girolamo Calò
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti, 2, 35131 Padova, Italy
| | - Giorgio Amendola
- "DiSTABiF", Università della Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Sandro Cosconati
- "DiSTABiF", Università della Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Salvatore Pacifico
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
- LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, 44121 Ferrara, Italy
| | - Delia Preti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
38
|
The Neural Network of Neuropeptide S (NPS): Implications in Food Intake and Gastrointestinal Functions. Pharmaceuticals (Basel) 2021; 14:ph14040293. [PMID: 33810221 PMCID: PMC8065993 DOI: 10.3390/ph14040293] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
The Neuropeptide S (NPS), a 20 amino acids peptide, is recognized as the endogenous ligand of a previously orphan G protein-coupled receptor, now termed NPS receptor (NPSR). The limited distribution of the NPS-expressing neurons in few regions of the brainstem is in contrast with the extensive expression of NPSR in the rodent central nervous system, suggesting the involvement of this receptor in several brain functions. In particular, NPS promotes locomotor activity, behavioral arousal, wakefulness, and unexpectedly, at the same time, it exerts anxiolytic-like properties. Intriguingly, the NPS system is implicated in the rewarding properties of drugs of abuse and in the regulation of food intake. Here, we focus on the anorexigenic effect of NPS, centrally injected in different brain areas, in both sated and fasted animals, fed with standard or palatable food, and, in addition, on its influence in the gastrointestinal tract. Further investigations, regarding the role of the NPS/NPSR system and its potential interaction with other neurotransmitters could be useful to understand the mechanisms underlying its action and to develop novel pharmacological tools for the treatment of aberrant feeding patterns and obesity.
Collapse
|
39
|
Gasparini S, Resch JM, Gore AM, Peltekian L, Geerling JC. Pre-locus coeruleus neurons in rat and mouse. Am J Physiol Regul Integr Comp Physiol 2021; 320:R342-R361. [PMID: 33296280 PMCID: PMC7988775 DOI: 10.1152/ajpregu.00261.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022]
Abstract
Previously, we identified a population of neurons in the hindbrain tegmentum, bordering the locus coeruleus (LC). We named this population the pre-locus coeruleus (pre-LC) because in rats its neurons lie immediately rostral to the LC. In mice, however, pre-LC and LC neurons intermingle, making them difficult to distinguish. Here, we use molecular markers and anterograde tracing to clarify the location and distribution of pre-LC neurons in mice, relative to rats. First, we colocalized the transcription factor FoxP2 with the activity marker Fos to identify pre-LC neurons in sodium-deprived rats and show their distribution relative to surrounding catecholaminergic and cholinergic neurons. Next, we used sodium depletion and chemogenetic activation of the aldosterone-sensitive HSD2 neurons in the nucleus of the solitary tract (NTS) to identify the homologous population of pre-LC neurons in mice, along with a related population in the central lateral parabrachial nucleus. Using Cre-reporter mice for Pdyn, we confirmed that most of these sodium-depletion-activated neurons are dynorphinergic. Finally, after confirming that these neurons receive excitatory input from the NTS and paraventricular hypothalamic nucleus, plus convergent input from the inhibitory AgRP neurons in the arcuate hypothalamic nucleus, we identify a major, direct input projection from the medial prefrontal cortex. This new information on the location, distribution, and input to pre-LC neurons provides a neuroanatomical foundation for cell-type-specific investigation of their properties and functions in mice. Pre-LC neurons likely integrate homeostatic information from the brainstem and hypothalamus with limbic, contextual information from the cerebral cortex to influence ingestive behavior.
Collapse
Affiliation(s)
- Silvia Gasparini
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Jon M Resch
- Department of Medicine, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Anuradha M Gore
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Lila Peltekian
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Joel C Geerling
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
40
|
Kirouac GJ. The Paraventricular Nucleus of the Thalamus as an Integrating and Relay Node in the Brain Anxiety Network. Front Behav Neurosci 2021; 15:627633. [PMID: 33732118 PMCID: PMC7959748 DOI: 10.3389/fnbeh.2021.627633] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/28/2021] [Indexed: 12/25/2022] Open
Abstract
The brain anxiety network is composed of a number of interconnected cortical regions that detect threats and execute appropriate defensive responses via projections to the shell of the nucleus accumbens (NAcSh), dorsolateral region of the bed nucleus of the stria terminalis (BSTDL) and lateral region of the central nucleus of the amygdala (CeL). The paraventricular nucleus of the thalamus (PVT) is anatomically positioned to integrate threat- and arousal-related signals from cortex and hypothalamus and then relay these signals to neural circuits in the NAcSh, BSTDL, and CeL that mediate defensive responses. This review describes the anatomical connections of the PVT that support the view that the PVT may be a critical node in the brain anxiety network. Experimental findings are reviewed showing that the arousal peptides orexins (hypocretins) act at the PVT to promote avoidance of potential threats especially following exposure of rats to a single episode of footshocks. Recent anatomical and experimental findings are discussed which show that neurons in the PVT provide divergent projections to subcortical regions that mediate defensive behaviors and that the projection to the NAcSh is critical for the enhanced social avoidance displayed in rats exposed to footshocks. A theoretical model is proposed for how the PVT integrates cortical and hypothalamic signals to modulate the behavioral responses associated with anxiety and other challenging situations.
Collapse
Affiliation(s)
- Gilbert J. Kirouac
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
41
|
Xu W, Wang L, Yuan XS, Wang TX, Li WX, Qu WM, Hong ZY, Huang ZL. Sevoflurane depresses neurons in the medial parabrachial nucleus by potentiating postsynaptic GABA A receptors and background potassium channels. Neuropharmacology 2020; 181:108249. [PMID: 32931816 DOI: 10.1016/j.neuropharm.2020.108249] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/23/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022]
Abstract
Despite persistent clinical use for over 170 years, the neuronal mechanisms by which general anesthetics produce hypnosis remain unclear. Previous studies suggest that anesthetics exert hypnotic effects by acting on endogenous arousal circuits. Recently, it has been shown that the medial parabrachial nucleus (MPB) is a novel wake-promoting component in the dorsolateral pons. However, it is not known whether and how the MPB contributes to anesthetic-induced hypnosis. Here, we investigated the action of sevoflurane, a widely used volatile anesthetic agent that best represents the drug class of halogenated ethers, on MPB neurons in mice. Using in vivo fiber photometry, we found that the population activities of MPB neurons were inhibited during sevoflurane-induced loss of consciousness. Using in vitro whole-cell patch-clamp recordings, we revealed that sevoflurane suppressed the firing rate of MPB neurons in concentration-dependent and reversible manners. At a concentration equal to MAC of hypnosis, sevoflurane potentiated synaptic GABAA receptors (GABAA-Rs), and the inhibitory effect of sevoflurane on the firing rate of MPB neurons was completely abolished by picrotoxin, which is a selective GABAA-R antagonist. At a concentration equivalent to MAC of immobility, sevoflurane directly hyperpolarized MPB neurons and induced a significant decrease in membrane input resistance by increasing a basal potassium conductance. Moreover, pharmacological blockade of GABAA-Rs in the MPB prolongs induction and shortens emergence under sevoflurane inhalation at MAC of hypnosis. These results indicate that sevoflurane inhibits MPB neurons through postsynaptic GABAA-Rs and background potassium channels, which contributes to sevoflurane-induced hypnosis.
Collapse
Affiliation(s)
- Wei Xu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lu Wang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiang-Shan Yuan
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Tian-Xiao Wang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wen-Xian Li
- Department of Anesthesiology, The Eye, Ear, Nose and Throat Hospital of Fudan University, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zong-Yuan Hong
- Laboratory of Quantitative Pharmacology, Department of Pharmacology, Wannan Medical College, Wuhu, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Siminski N, Böhme S, Zeller JBM, Becker MPI, Bruchmann M, Hofmann D, Breuer F, Mühlberger A, Schiele MA, Weber H, Schartner C, Deckert J, Pauli P, Reif A, Domschke K, Straube T, Herrmann MJ. BNST and amygdala activation to threat: Effects of temporal predictability and threat mode. Behav Brain Res 2020; 396:112883. [PMID: 32860830 DOI: 10.1016/j.bbr.2020.112883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 12/28/2022]
Abstract
Recent animal and human studies highlight the uncertainty about the onset of an aversive event as a crucial factor for the involvement of the centromedial amygdala (CM) and bed nucleus of the stria terminalis (BNST) activity. However, studies investigating temporally predictable or unpredictable threat anticipation and confrontation processes are rare. Furthermore, the few existing fMRI studies analyzing temporally predictable and unpredictable threat processes used small sample sizes or limited fMRI paradigms. Therefore, we measured functional brain activity in 109 predominantly female healthy participants during a temporally predictable-unpredictable threat paradigm, which aimed to solve limited aspects of recent studies. Results showed higher BNST activity compared to the CM during the cue indicating that the upcoming confrontation is aversive relative to the cue indicating an upcoming neutral confrontation. Both the CM and BNST showed higher activity during the confrontation with unpredictable and aversive stimuli, but the reaction to aversive confrontation relative to neutral confrontation was stronger in the CM compared to the BNST. Additional modulation analyses by NPSR1 rs324981 genotype revealed higher BNST activity relative to the CM in unpredictable anticipation relative to predictable anticipation in T-carriers compared to AA carriers. Our results indicate that during the confrontation with aversive or neutral stimuli, temporal unpredictability modulates CM and BNST activity. Further, there is a differential activity concerning threat processing, as BNST is more involved when focussing on fear-related anticipation processes and CM is more involved when focussing on threat confrontation.
Collapse
Affiliation(s)
- N Siminski
- Center of Mental Health, Dept. of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - S Böhme
- Department of Clinical Psychology and Psychotherapy, Institute of Psychology, University of Regensburg, Regensburg, Germany; Department of Clinical Psychology and Psychotherapy, University of Erlangen, Erlangen, Germany
| | - J B M Zeller
- Center of Mental Health, Dept. of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - M P I Becker
- Institute of Medical Psychology and Systems Neuroscience, University of Muenster, Muenster, Germany
| | - M Bruchmann
- Institute of Medical Psychology and Systems Neuroscience, University of Muenster, Muenster, Germany
| | - D Hofmann
- Institute of Medical Psychology and Systems Neuroscience, University of Muenster, Muenster, Germany
| | - F Breuer
- Fraunhofer Institute for Integrated Circuits (IIS), Development Center for X-ray Technology (EZRT), Wuerzburg, Germany
| | - A Mühlberger
- Department of Clinical Psychology and Psychotherapy, Institute of Psychology, University of Regensburg, Regensburg, Germany
| | - M A Schiele
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - H Weber
- Center of Mental Health, Dept. of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - C Schartner
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - J Deckert
- Center of Mental Health, Dept. of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - P Pauli
- Department of Psychology (Biological Psychology, Clinical Psychology, and Psychotherapy), University of Wuerzburg, Wuerzburg, Germany
| | - A Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - K Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Basics in Neuro Modulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - T Straube
- Institute of Medical Psychology and Systems Neuroscience, University of Muenster, Muenster, Germany
| | - M J Herrmann
- Center of Mental Health, Dept. of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
43
|
Xing L, Shi G, Mostovoy Y, Gentry NW, Fan Z, McMahon TB, Kwok PY, Jones CR, Ptáček LJ, Fu YH. Mutant neuropeptide S receptor reduces sleep duration with preserved memory consolidation. Sci Transl Med 2020; 11:11/514/eaax2014. [PMID: 31619542 DOI: 10.1126/scitranslmed.aax2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022]
Abstract
Sleep is a crucial physiological process for our survival and cognitive performance, yet the factors controlling human sleep regulation remain poorly understood. Here, we identified a missense mutation in a G protein-coupled neuropeptide S receptor 1 (NPSR1) that is associated with a natural short sleep phenotype in humans. Mice carrying the homologous mutation exhibited less sleep time despite increased sleep pressure. These animals were also resistant to contextual memory deficits associated with sleep deprivation. In vivo, the mutant receptors showed increased sensitivity to neuropeptide S exogenous activation. These results suggest that the NPS/NPSR1 pathway might play a critical role in regulating human sleep duration and in the link between sleep homeostasis and memory consolidation.
Collapse
Affiliation(s)
- Lijuan Xing
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Guangsen Shi
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yulia Mostovoy
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nicholas W Gentry
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Zenghua Fan
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Thomas B McMahon
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Pui-Yan Kwok
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA.,Department of Dermatology, University of California San Francisco, San Francisco, CA 94143, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | | | - Louis J Ptáček
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA. .,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA.,Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA.,Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ying-Hui Fu
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA. .,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA.,Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA.,Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
44
|
Varga AG, Maletz SN, Bateman JT, Reid BT, Levitt ES. Neurochemistry of the Kölliker-Fuse nucleus from a respiratory perspective. J Neurochem 2020; 156:16-37. [PMID: 32396650 DOI: 10.1111/jnc.15041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
The Kölliker-Fuse nucleus (KF) is a functionally distinct component of the parabrachial complex, located in the dorsolateral pons of mammals. The KF has a major role in respiration and upper airway control. A comprehensive understanding of the KF and its contributions to respiratory function and dysfunction requires an appreciation for its neurochemical characteristics. The goal of this review is to summarize the diverse neurochemical composition of the KF, focusing on the neurotransmitters, neuromodulators, and neuropeptides present. We also include a description of the receptors expressed on KF neurons and transporters involved in each system, as well as their putative roles in respiratory physiology. Finally, we provide a short section reviewing the literature regarding neurochemical changes in the KF in the context of respiratory dysfunction observed in SIDS and Rett syndrome. By over-viewing the current literature on the neurochemical composition of the KF, this review will serve to aid a wide range of topics in the future research into the neural control of respiration in health and disease.
Collapse
Affiliation(s)
- Adrienn G Varga
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Sebastian N Maletz
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jordan T Bateman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Brandon T Reid
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Erica S Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| |
Collapse
|
45
|
Wang C, Xin L, Cai CC, Cong CY, Xie JF, Kong XP, Dong CY, Li J, Cui GF, Chen HL, Ren YL, Shao YF, Hou YP. Neuropeptide S Displays as a Key Neuromodulator in Olfactory Spatial Memory. Chem Senses 2020; 45:195-202. [PMID: 32010937 DOI: 10.1093/chemse/bjaa003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Neuropeptide S (NPS) is an endogenous peptide recently recognized to be presented in the brainstem and believed to play an important role in maintaining memory. The deletion of NPS or NPS receptor (NPSR) in mice shows a deficit in memory formation. Our recent studies have demonstrated that central administration of NPS facilitates olfactory function and ameliorates olfactory spatial memory impairment induced by muscarinic cholinergic receptor antagonist and N-methyl-D-aspartate receptor antagonist. However, it remains to be determined if endogenous NPS is an indispensable neuromodulator in the control of the olfactory spatial memory. In this study, we examined the effects of NPSR peptidergic antagonist [D-Val5]NPS (10 and 20 nmol, intracerebroventricular) and nonpeptidergic antagonist SHA 68 (10 and 50 mg/kg, intraperitoneal) on the olfactory spatial memory using computer-assisted 4-hole-board olfactory spatial memory test in mice. Furthermore, immunofluorescence was employed to identify the distributions of c-Fos and NPSR immunoreactive (-ir) neurons in olfactory system and hippocampal formation known to closely relate to the olfactory spatial memory. [D-Val5]NPS dosing at 20 nmol and SHA 68 dosing at 50 mg/kg significantly decreased the number of visits to the 2 odorants interchanged spatially, switched odorants, in recall trial, and simultaneously reduced the percentage of Fos-ir in NPSR-ir neurons, which were densely distributed in the anterior olfactory nucleus, piriform cortex, subiculum, presubiculum, and parasubiculum. These findings suggest that endogenous NPS is a key neuromodulator in olfactory spatial memory.
Collapse
Affiliation(s)
- Can Wang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Le Xin
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China.,Department of Anesthesiology, Lishan Hospital of the Anshan Central Hospital, Anshan, PR China
| | - Chen-Chen Cai
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Chao-Yu Cong
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Jun-Fan Xie
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Xiang-Pan Kong
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China.,Department of Human Anatomy, School of Medicine, Hunan Normal University, Changsha, PR China
| | - Chao-Yu Dong
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China.,Department of Gynaecology, The Third People's Hospital of Yunnan Province, Kunming, PR China
| | - Jing Li
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China.,Departments of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Gansu University of Chinese Medicine, Lanzhou, PR China
| | - Guang-Fu Cui
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Hai-Lin Chen
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Yan-Li Ren
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| |
Collapse
|
46
|
Neugebauer V, Mazzitelli M, Cragg B, Ji G, Navratilova E, Porreca F. Amygdala, neuropeptides, and chronic pain-related affective behaviors. Neuropharmacology 2020; 170:108052. [PMID: 32188569 DOI: 10.1016/j.neuropharm.2020.108052] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Neuropeptides play important modulatory roles throughout the nervous system, functioning as direct effectors or as interacting partners with other neuropeptide and neurotransmitter systems. Limbic brain areas involved in learning, memory and emotions are particularly rich in neuropeptides. This review will focus on the amygdala, a limbic region that plays a key role in emotional-affective behaviors and pain modulation. The amygdala is comprised of different nuclei; the basolateral (BLA) and central (CeA) nuclei and in between, the intercalated cells (ITC), have been linked to pain-related functions. A wide range of neuropeptides are found in the amygdala, particularly in the CeA, but this review will discuss those neuropeptides that have been explored for their role in pain modulation. Calcitonin gene-related peptide (CGRP) is a key peptide in the afferent nociceptive pathway from the parabrachial area and mediates excitatory drive of CeA neurons. CeA neurons containing corticotropin releasing factor (CRF) and/or somatostatin (SOM) are a source of long-range projections and serve major output functions, but CRF also acts locally to excite neurons in the CeA and BLA. Neuropeptide S (NPS) is associated with inhibitory ITC neurons that gate amygdala output. Oxytocin and vasopressin exert opposite (inhibitory and excitatory, respectively) effects on amygdala output. The opioid system of mu, delta and kappa receptors (MOR, DOR, KOR) and their peptide ligands (β-endorphin, enkephalin, dynorphin) have complex and partially opposing effects on amygdala function. Neuropeptides therefore serve as valuable targets to regulate amygdala function in pain conditions. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Bryce Cragg
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
47
|
Kolodziejczyk MH, Fendt M. Corticosterone Treatment and Incubation Time After Contextual Fear Conditioning Synergistically Induce Fear Memory Generalization in Neuropeptide S Receptor-Deficient Mice. Front Neurosci 2020; 14:128. [PMID: 32231512 PMCID: PMC7081924 DOI: 10.3389/fnins.2020.00128] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/31/2020] [Indexed: 12/18/2022] Open
Abstract
Fear memory generalization is a learning mechanism that promotes flexible fear responses to novel situations. While fear generalization has adaptive value, overgeneralization of fear memory is a characteristic feature of the pathology of anxiety disorders. The neuropeptide S (NPS) receptor (NPSR) has been shown to be associated with anxiety disorders and has recently been identified as a promising target for treating anxiety disorders. Moreover, stress hormones play a role in regulating both physiological and pathological fear memories and might therefore also be involved in anxiety disorders. However, little is known about the interplay between stress hormone and the NPS system in the development of overgeneralized fear. Here, we hypothesize that NPSR-deficient mice with high corticosterone (CORT) levels during the fear memories consolidation are more prone to develop generalized fear. To address this hypothesis, NPSR-deficient mice were submitted to a contextual fear conditioning procedure. Immediately after conditioning, mice received CORT injections (2.5 or 5 mg/kg). One day and 1 month later, the mice were tested for the specificity and strength of their fear memory, their anxiety level, and their startle response. Moreover, CORT blood levels were monitored throughout the experiment. Using this protocol, a specific contextual fear memory was observed in all experimental groups, despite the 5-mg/kg CORT-treated NPSR-deficient mice. This group of mice showed a generalization of contextual fear memory and a decreased startle response, and the females of this group had significantly less body weight gain. These findings indicate that interplay between CORT and the NPS system during the consolidation of fear memories is critical for the generalization of contextual fear.
Collapse
Affiliation(s)
- Malgorzata H Kolodziejczyk
- Neuropharmaclogy of Emotional Systems, Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Markus Fendt
- Neuropharmaclogy of Emotional Systems, Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
48
|
Lee MT, Chiu YT, Chiu YC, Hor CC, Lee HJ, Guerrini R, Calo G, Chiou LC. Neuropeptide S-initiated sequential cascade mediated by OX 1, NK 1, mGlu 5 and CB 1 receptors: a pivotal role in stress-induced analgesia. J Biomed Sci 2020; 27:7. [PMID: 31915019 PMCID: PMC6950992 DOI: 10.1186/s12929-019-0590-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/18/2019] [Indexed: 02/03/2023] Open
Abstract
Background Stress-induced analgesia (SIA) is an evolutionarily conserved phenomenon during stress. Neuropeptide S (NPS), orexins, substance P, glutamate and endocannabinoids are known to be involved in stress and/or SIA, however their causal links remain unclear. Here, we reveal an unprecedented sequential cascade involving these mediators in the lateral hypothalamus (LH) and ventrolateral periaqueductal gray (vlPAG) using a restraint stress-induced SIA model. Methods Male C57BL/6 mice of 8–12 week-old were subjected to intra-cerebroventricular (i.c.v.) and/or intra-vlPAG (i.pag.) microinjection of NPS, orexin-A or substance P alone or in combination with selective antagonists of NPS receptors (NPSRs), OX1 receptors (OX1Rs), NK1 receptors (NK1Rs), mGlu5 receptors (mGlu5Rs) and CB1 receptors (CB1Rs), respectively. Antinociceptive effects of these mediators were evaluated via the hot-plate test. SIA in mice was induced by a 30-min restraint stress. NPS levels in the LH and substance P levels in vlPAG homogenates were compared in restrained and unrestrained mice. Results NPS (i.c.v., but not i.pag.) induced antinociception. This effect was prevented by i.c.v. blockade of NPSRs. Substance P (i.pag.) and orexin-A (i.pag.) also induced antinociception. Substance P (i.pag.)-induced antinociception was prevented by i.pag. Blockade of NK1Rs, mGlu5Rs or CB1Rs. Orexin-A (i.pag.)-induced antinociception has been shown previously to be prevented by i.pag. blockade of OX1Rs or CB1Rs, and here was prevented by NK1R or mGlu5R antagonist (i.pag.). NPS (i.c.v.)-induced antinociception was prevented by i.pag. blockade of OX1Rs, NK1Rs, mGlu5Rs or CB1Rs. SIA has been previously shown to be prevented by i.pag. blockade of OX1Rs or CB1Rs. Here, we found that SIA was also prevented by i.c.v. blockade of NPSRs or i.pag. blockade of NK1Rs or mGlu5Rs. Restrained mice had higher levels of NPS in the LH and substance P in the vlPAG than unrestrained mice. Conclusions These results suggest that, during stress, NPS is released and activates LH orexin neurons via NPSRs, releasing orexins in the vlPAG. Orexins then activate OX1Rs on substance P-containing neurons in the vlPAG to release substance P that subsequently. Activates NK1Rs on glutamatergic neurons to release glutamate. Glutamate then activates perisynaptic mGlu5Rs to initiate the endocannabinoid retrograde inhibition of GABAergic transmission in the vlPAG, leading to analgesia.
Collapse
Affiliation(s)
- Ming Tatt Lee
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.,Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.,Faculty of Pharmaceutical Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Yu-Ting Chiu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Yu-Chun Chiu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chia Chun Hor
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Hsin-Jung Lee
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA), Ferrara, Italy
| | - Girolamo Calo
- Department of Medical Sciences and National Institute of Neurosciences, Section of Pharmacology, University of Ferrara, 44121, Ferrara, Italy
| | - Lih-Chu Chiou
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan. .,Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan. .,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan. .,Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
49
|
Kreutzmann JC, Khalil R, Köhler JC, Mayer D, Florido A, Nadal R, Andero R, Fendt M. Neuropeptide‐S‐receptor deficiency affects sex‐specific modulation of safety learning by pre‐exposure to electric stimuli. GENES BRAIN AND BEHAVIOR 2020; 19:e12621. [DOI: 10.1111/gbb.12621] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/28/2019] [Accepted: 10/22/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Judith C. Kreutzmann
- Institute for Pharmacology & ToxicologyOtto‐von‐Guericke University Magdeburg Magdeburg Germany
- Department of Systems Physiology of LearningLeibniz Institute for Neurobiology Magdeburg Germany
| | - Radwa Khalil
- Institute for Pharmacology & ToxicologyOtto‐von‐Guericke University Magdeburg Magdeburg Germany
| | - Jana C. Köhler
- Institute of PhysiologyOtto‐von‐Guericke University Magdeburg Magdeburg Germany
- Center of Behavioral Brain SciencesOtto‐von‐Guericke University Magdeburg Magdeburg Germany
| | - Dana Mayer
- Institute for Pharmacology & ToxicologyOtto‐von‐Guericke University Magdeburg Magdeburg Germany
| | - Antonio Florido
- Institut de NeurocièncesUniversitat Autònoma de Barcelona Bellaterra Spain
| | - Roser Nadal
- Institut de NeurocièncesUniversitat Autònoma de Barcelona Bellaterra Spain
- CIBERSAMInstituto de Salud Carlos III, Universitat Autònoma de Barcelona Bellaterra Spain
- Department of Psychobiology and Methodology in Health SciencesUniversitat Autònoma de Barcelona Bellaterra Spain
| | - Raül Andero
- Institut de NeurocièncesUniversitat Autònoma de Barcelona Bellaterra Spain
- CIBERSAMInstituto de Salud Carlos III, Universitat Autònoma de Barcelona Bellaterra Spain
- Department of Psychobiology and Methodology in Health SciencesUniversitat Autònoma de Barcelona Bellaterra Spain
| | - Markus Fendt
- Institute for Pharmacology & ToxicologyOtto‐von‐Guericke University Magdeburg Magdeburg Germany
- Center of Behavioral Brain SciencesOtto‐von‐Guericke University Magdeburg Magdeburg Germany
| |
Collapse
|
50
|
Holanda VAD, Oliveira MC, Souza LS, Lobão-Soares B, André E, Da Silva Junior ED, Guerrini R, Calo G, Ruzza C, Gavioli EC. Dopamine D 1 and D 2 receptors mediate neuropeptide S-induced antinociception in the mouse formalin test. Eur J Pharmacol 2019; 859:172557. [PMID: 31326375 DOI: 10.1016/j.ejphar.2019.172557] [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: 05/09/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/31/2022]
Abstract
Neuropeptide S (NPS) is the endogenous ligand of a G-protein coupled receptor named NPS receptor. The NPS system controls several biological functions, including anxiety, wakefulness, locomotor activity, food intake, and pain transmission. A growing body of evidence supports facilitatory effects for NPS over dopaminergic neurotransmission. The present study was aimed to investigate the role of dopamine receptors signaling in the antinociceptive effects of NPS in the mouse formalin test. The following dopamine receptor antagonists were employed: SCH 23390 (selective dopamine D1 antagonist, 0.05 mg/kg, ip), haloperidol (non-selective dopamine D2-like receptor antagonist; 0.03 mg/kg, ip), and sulpiride (selective dopamine D2-like receptor antagonist; 25 mg/kg, ip). Mice were pretreated with dopamine antagonists before the supraspinal administration of NPS (0.1 nmol, icv). Morphine (5 mg/kg, sc) and indomethacin (10 mg/kg, ip) were used as positive controls to set up the experimental conditions. Morphine-induced antinociceptive effects were observed during phases 1 and 2 of the test, while indomethacin was only active at phase 2. Central NPS significantly reduced formalin-induced nociception during both phases. The systemic administration of SCH 23390 slightly blocked the effects of NPS only during phase 2. Haloperidol prevented NPS-induced antinociceptive effects. Similar to haloperidol, sulpiride also counteracted the antinociceptive effects of NPS in both phases of the formalin test. In conclusion, the present findings suggest that the analgesic effects of NPS are linked with dopaminergic neurotransmission mainly through dopamine D2-like receptor signaling.
Collapse
Affiliation(s)
- Victor A D Holanda
- Department of Biophysics and Pharmacology, Federal University of Rio Grande Do Norte, Natal, RN, Brazil
| | - Matheus C Oliveira
- Department of Biophysics and Pharmacology, Federal University of Rio Grande Do Norte, Natal, RN, Brazil
| | - Lisiane S Souza
- Department of Biophysics and Pharmacology, Federal University of Rio Grande Do Norte, Natal, RN, Brazil
| | - Bruno Lobão-Soares
- Department of Biophysics and Pharmacology, Federal University of Rio Grande Do Norte, Natal, RN, Brazil
| | - Eunice André
- Department of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Edilson D Da Silva Junior
- Department of Biophysics and Pharmacology, Federal University of Rio Grande Do Norte, Natal, RN, Brazil
| | - Remo Guerrini
- Department of Chemistry and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Girolamo Calo
- Department of Medical Sciences, Section of Pharmacology, and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Chiara Ruzza
- Department of Medical Sciences, Section of Pharmacology, and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Elaine C Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande Do Norte, Natal, RN, Brazil.
| |
Collapse
|