1
|
Brockway DF, Crowley NA. Emerging pharmacological targets for alcohol use disorder. Alcohol 2024; 121:103-114. [PMID: 39069210 DOI: 10.1016/j.alcohol.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/27/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Alcohol Use Disorder (AUD) remains a challenging condition with limited effective treatment options; however new technology in drug delivery and advancements in pharmacology have paved the way for discovery of novel therapeutic targets. This review explores emerging pharmacological targets that offer new options for the management of AUD, focusing on the potential of somatostatin (SST), vasoactive intestinal peptide (VIP), glucagon-like peptide-1 (GLP-1), nociceptin (NOP), and neuropeptide S (NPS). These targets have been selected based on recent advancements in preclinical and clinical research, which suggest their significant roles in modulating alcohol consumption and related behaviors. SST dampens cortical circuits, and targeting both the SST neurons and the SST peptide itself presents promise for treating AUD and various related comorbidities. VIP neurons are modulated by alcohol and targeting the VIP system presents an unexplored avenue for addressing alcohol exposure at various stages of development. GLP-1 interacts with the dopaminergic reward system and reduces alcohol intake. Nociceptin modulates mesolimbic circuitry and agonism and antagonism of nociceptin receptor offers a complex but promising approach to reducing alcohol consumption. NPS stands out for its anxiolytic-like effects, particularly relevant for the anxiety associated with AUD. This review aims to synthesize the current understanding of these targets, highlighting their potential in developing more effective and personalized AUD therapies, and underscores the importance of continued research in identifying and validating novel targets for treatment of AUD and comorbid conditions.
Collapse
Affiliation(s)
- Dakota F Brockway
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA; Penn State Neuroscience Institute, Penn State University, University Park, PA, 16802, USA.
| | - Nicole A Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA; Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA; Penn State Neuroscience Institute, Penn State University, University Park, PA, 16802, 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
|
Huang Y, Wojciechowski A, Feldman K, Ettaro R, Veros K, Ritter M, Carvalho Costa P, DiStasio J, Peirick JJ, Reissner KJ, Runyon SP, Clark SD. RTI-263, a biased neuropeptide S receptor agonist that retains an anxiolytic effect, attenuates cocaine-seeking behavior in rats. Neuropharmacology 2023; 241:109743. [PMID: 37820934 DOI: 10.1016/j.neuropharm.2023.109743] [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: 07/31/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
Neuropeptide S (NPS) is a neuromodulatory peptide that acts via a G protein-coupled receptor. Centrally administered NPS suppresses anxiety-like behaviors in rodents while producing a paradoxical increase in arousal. In addition, NPS increases drug-seeking behavior when administered during cue-induced reinstatement. Conversely, an NPS receptor (NPSR) antagonist, RTI-118, decreases cocaine-seeking behavior. A biased NPSR ligand, RTI-263, produces anxiolytic-like effects and has memory-enhancing effects similar to those of NPS but without the increase in arousal. In the present study, we show that RTI-263 decreased cocaine seeking by both male and female rats during cue-induced reinstatement. However, RTI-263 did not modulate the animals' behaviors during natural reward paradigms, such as palatable food intake, feeding during a fasting state, and cue-induced reinstatement of sucrose seeking. Therefore, NPSR biased agonists are a potential pharmacotherapy for substance use disorder because of the combined benefits of decreased drug seeking and the suppression of anxiety.
Collapse
Affiliation(s)
- Yuanli Huang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Alaina Wojciechowski
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Kyle Feldman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Robert Ettaro
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Kaliana Veros
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Morgan Ritter
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Paula Carvalho Costa
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Jacob DiStasio
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA
| | - Jennifer J Peirick
- Laboratory Animal Facilities, University at Buffalo, Buffalo, NY 14214, USA
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott P Runyon
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC 27709, USA
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
6
|
Yang S, Zhu G. Phytotherapy of abnormality of fear memory: A narrative review of mechanisms. Fitoterapia 2023; 169:105618. [PMID: 37482307 DOI: 10.1016/j.fitote.2023.105618] [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: 05/23/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
It is generally believed that in post-traumatic stress disorder (PTSD), the high expression of fear memory is mainly determined by amygdala hyperactivity and hippocampus hypoactivity. In this review, we firstly updated the mechanisms of fear memory, and then searched the experimental evidence of phytotherapy for fear memory in the past five years. Based on the summary of those experimental studies, we further discussed the future research strategies of plant medicines, including the study of the mechanism of specific brain regions, the optimal time for the prevention and treatment of fear memory-related diseases such as PTSD, and the development of new drugs with active components of plant medicines. Accordingly, plant medicines play a clear role in improving fear memory abnormalities and have the drug development potential in the treatment of fear-related disorders.
Collapse
Affiliation(s)
- Shaojie Yang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230061, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
ÖZKAN A, BÜLBÜL M, DERİN N, SİNEN O, AKÇAY G, PARLAK H, AYDIN ASLAN M, AĞAR A. Neuropeptide-S affects cognitive impairment and depression-like behavior on MPTP induced experimental mouse model of Parkinson’s disease. Turk J Med Sci 2021; 51:3126-3135. [PMID: 34289654 PMCID: PMC10734830 DOI: 10.3906/sag-2105-74] [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: 05/07/2021] [Revised: 12/13/2021] [Accepted: 07/21/2021] [Indexed: 11/03/2022] Open
Abstract
Background/aim The present study proposes to investigate the effect of neuropeptide–S (NPS) on cognitive functions and depression-like behavior of MPTP-induced experimental model of Parkinson’s disease (PD). Materials and methods Three-month-old C57BL/6 mice were randomly divided into three groups as; Control, Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and MPTP + NPS 0.1 nmol (received intraperitoneal injection of MPTP and intracerebroventricular injection of NPS, 0.1 nmol for seven days). The radial arm maze and pole tests were carried out, and the levels of tyrosine hydroxylase (TH) were determined using western blotting. A mass spectrometer was used to measure the levels of dopamine, glutamic acid, and glutamine. Results The T-turn and time to descend enhanced in MPTP group, while these parameters were decreased by NPS treatment. In the MPTP group, the number of working memory errors (WME) and reference memory errors (RME) increased, whereas NPS administration decreased both parameters. Sucrose preference decreased in the MPTP group while increasing in the NPS group. MPTP injection significantly reduced dopamine, glutamic acid, and glutamine levels. NPS treatment restored the MPTP-induced reduction in glutamine and glutamic acid levels. Conclusion NPS may be involved in the future treatment of cognitive impairments and depression-like behaviors in PD.
Collapse
Affiliation(s)
- Ayşe ÖZKAN
- Department of Physiology, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Mehmet BÜLBÜL
- Department of Physiology, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Narin DERİN
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Osman SİNEN
- Department of Physiology, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Güven AKÇAY
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Hande PARLAK
- Department of Physiology, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Mutay AYDIN ASLAN
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| | - Aysel AĞAR
- Department of Physiology, Faculty of Medicine, Akdeniz University, Antalya,
Turkey
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Si W, Liu X, Pape HC, Reinscheid RK. Neuropeptide S-Mediated Modulation of Prepulse Inhibition Depends on Age, Gender, Stimulus-Timing, and Attention. Pharmaceuticals (Basel) 2021; 14:489. [PMID: 34065431 PMCID: PMC8160819 DOI: 10.3390/ph14050489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Conflicting reports about the role of neuropeptide S (NPS) in animal models of psychotic-like behavior and inconsistent results from human genetic studies seeking potential associations with schizophrenia prompted us to reevaluate the effects of NPS in the prepulse inhibition (PPI) paradigm in mice. Careful examination of NPS receptor (NPSR1) knockout mice at different ages revealed that PPI deficits are only expressed in young male knockout animals (<12 weeks of age), that can be replicated in NPS precursor knockout mice and appear strain-independent, but are absent in female mice. PPI deficits can be aggravated by MK-801 and alleviated by clozapine. Importantly, treatment of wildtype mice with a centrally-active NPSR1 antagonist was able to mimic PPI deficits. PPI impairment in young male NPSR1 and NPS knockout mice may be caused by attentional deficits that are enhanced by increasing interstimulus intervals. Our data reveal a substantial NPS-dependent developmental influence on PPI performance and confirm a significant role of attentional processes for sensory-motor gating. Through its influence on attention and arousal, NPS appears to positively modulate PPI in young animals, whereas compensatory mechanisms may alleviate NPS-dependent deficits in older mice.
Collapse
Affiliation(s)
- Wei Si
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA; (W.S.); (X.L.)
| | - Xiaobin Liu
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA; (W.S.); (X.L.)
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische-Wilhelms University, 48149 Münster, Germany;
| | - Rainer K. Reinscheid
- Institute of Physiology I, Westfälische-Wilhelms University, 48149 Münster, Germany;
- Institute of Pharmacology and Toxicology, Friedrich-Schiller University, 07747 Jena, Germany
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Reinscheid RK, Mafessoni F, Lüttjohann A, Jüngling K, Pape HC, Schulz S. Neandertal introgression and accumulation of hypomorphic mutations in the neuropeptide S (NPS) system promote attenuated functionality. Peptides 2021; 138:170506. [PMID: 33556445 DOI: 10.1016/j.peptides.2021.170506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/14/2021] [Accepted: 02/03/2021] [Indexed: 12/21/2022]
Abstract
The neuropeptide S (NPS) system plays an important role in fear and fear memory processing but has also been associated with allergic and inflammatory diseases. Genes for NPS and its receptor NPSR1 are found in all tetrapods. Compared to non-human primates, several non-synonymous single-nucleotide polymorphisms (SNPs) occur in both human genes that collectively result in functional attenuation, suggesting adaptive mechanisms in a human context. To investigate historic and geographic origins of these hypomorphic mutations and explore genetic signs of selection, we analyzed ancient genomes and worldwide genotype frequencies of four prototypic SNPs in the NPS system. Neandertal and Denisovan genomes contain exclusively ancestral alleles for NPSR1 while all derived alleles occur in ancient genomes of anatomically modern humans, indicating that they arose in modern Homo sapiens. Worldwide genotype frequencies for three hypomorphic NPSR1 SNPs show significant regional homogeneity but follow a gradient towards increasing derived allele frequencies that supports an out-of-Africa scenario. Increased density of high-frequency polymorphisms around the three NPSR1 loci suggests weak or possibly balancing selection. A hypomorphic mutation in the NPS precursor, however, was detected at high frequency in Eurasian Neandertal genomes and shows genetic signatures indicating that it was introgressed into the human gene pool, particularly in Southern Europe, by interbreeding with Neandertals. We discuss potential evolutionary scenarios including behavior and immune-based natural selection.
Collapse
Affiliation(s)
- Rainer K Reinscheid
- Institute of Pharmacology & Toxicology, Friedrich-Schiller-University, Jena, Germany; Institute of Physiology I, Westfälische-Wilhelms-University, Münster, Germany.
| | | | - Annika Lüttjohann
- Institute of Physiology I, Westfälische-Wilhelms-University, Münster, Germany
| | - Kay Jüngling
- Institute of Physiology I, Westfälische-Wilhelms-University, Münster, Germany
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische-Wilhelms-University, Münster, Germany
| | - Stefan Schulz
- Institute of Pharmacology & Toxicology, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
24
|
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
|
25
|
Zhang R, Lao K, Lu B, Guo H, Cheng J, Chen P, Gou X. (m)RVD-hemopressin (α) and (m)VD-hemopressin (α) improve the memory-impairing effect of scopolamine in novel object and object location recognition tasks in mice. Peptides 2021; 136:170442. [PMID: 33171279 DOI: 10.1016/j.peptides.2020.170442] [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: 09/08/2020] [Revised: 10/28/2020] [Accepted: 11/04/2020] [Indexed: 11/21/2022]
Abstract
Dysfunction of cholinergic system plays an important role in disease associated with cognitive blockage, such as Alzheimer's disease (AD). Central administration of scopolamine, an antagonist of acetylcholine receptor, could induce memory impairment in mice. Endocannabinoid system was also implicated in AD, as two peptides agonists of cannabinoid 1 receptor (CB1R), (m)RVD-hemopressin (α) (RVD) and (m)VD-hemopressin (α) (VD) have been reported to inhibit the AD-relating impairment in animal and cell models. More than one-third of the cholinergic cells expressed CB1R, so we speculated that RVD and VD might have ability to inhibit the memory-impairing effect of scopolamine. Our results showed RVD and VD ameliorated the memory toxicity of scopolamine, and the effects of the two peptides could be blocked by CB1R antagonists hemopressin (Hp) and AM251 in novel object and object location recognition tasks in mice. This study suggested that RVD and VD might be potential compounds for the treatment of the disease associated with impairment of cholinergic system.
Collapse
Affiliation(s)
- Ruisan Zhang
- Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, China
| | - Kejing Lao
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Baiyu Lu
- Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, China
| | - Huifang Guo
- Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, China
| | - Jianghong Cheng
- Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, China
| | - Peng Chen
- Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, China.
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, China; Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China.
| |
Collapse
|
26
|
Borgonetti V, Les F, López V, Galeotti N. Attenuation of Anxiety-Like Behavior by Helichrysum stoechas (L.) Moench Methanolic Extract through Up-Regulation of ERK Signaling Pathways in Noradrenergic Neurons. Pharmaceuticals (Basel) 2020; 13:ph13120472. [PMID: 33348565 PMCID: PMC7766703 DOI: 10.3390/ph13120472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023] Open
Abstract
The long-term use of anxiolytic and antidepressant drugs can cause a plethora of side effects and the use of complementary and alternative medicine, which is generally considered safer than conventional medicine, is consistently increasing. Helichrysum stoechas (L.) Moench methanolic extract (HSE) has shown MAO-A inhibitory properties in previous studies. With the aim of obtaining innovative and safer therapies for mood disorders, this study investigated the potential activity of HSE in the management of anxiety- and depression-related symptoms. HSE showed dose-dependent (30-100 mg/kg p.o.) anxiolytic-like activity in the light dark box and marble burying tests, without any antidepressant-like activity, as shown by the results of the tail suspension test. Additionally, HSE did not have any effect on the modulation of pain, which highlights its selectivity in the control of anxiety-related behavior. At active doses, HSE did not produce any sedative effect or result in impaired motor coordination and memory functions. Western blotting experiments showed the ability of HSE to counteract the reduction in the phosphorylation of ERK44/42, to restore brain-derived neurotrophic factor (BDNF) expression and to return cyclic AMP response element binding (CREB) levels to basal levels in noradrenergic hippocampal neurons of mice exposed to an anxiety-related environment, which indicates a protective role against anxiety behavior. These results suggest that oral administration of HSE might represent an interesting opportunity for the management of anxiety disorders.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy;
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Zaragoza, Spain;
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Víctor López
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Zaragoza, Spain;
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, 50013 Zaragoza, Spain
- Correspondence: (V.L.); (N.G.)
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy;
- Correspondence: (V.L.); (N.G.)
| |
Collapse
|
27
|
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
|
28
|
Chauveau F, Claverie D, Lardant E, Varin C, Hardy E, Walter A, Canini F, Rouach N, Rancillac A. Neuropeptide S promotes wakefulness through the inhibition of sleep-promoting ventrolateral preoptic nucleus neurons. Sleep 2020; 43:5547657. [PMID: 31403694 DOI: 10.1093/sleep/zsz189] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/26/2019] [Indexed: 12/22/2022] Open
Abstract
STUDY OBJECTIVES The regulation of sleep-wake cycles is crucial for the brain's health and cognitive skills. Among the various substances known to control behavioral states, intraventricular injection of neuropeptide S (NPS) has already been shown to promote wakefulness. However, the NPS signaling pathway remains elusive. In this study, we characterized the effects of NPS in the ventrolateral preoptic nucleus (VLPO) of the hypothalamus, one of the major brain structures regulating non-rapid eye movement (NREM) sleep. METHODS We combined polysomnographic recordings, vascular reactivity, and patch-clamp recordings in mice VLPO to determine the NPS mode of action. RESULTS We demonstrated that a local infusion of NPS bilaterally into the anterior hypothalamus (which includes the VLPO) significantly increases awakening and specifically decreases NREM sleep. Furthermore, we established that NPS application on acute brain slices induces strong and reversible tetrodotoxin (TTX)-sensitive constriction of blood vessels in the VLPO. This effect strongly suggests that the local neuronal network is downregulated in the presence of NPS. At the cellular level, we revealed by electrophysiological recordings and in situ hybridization that NPSR mRNAs are only expressed by non-Gal local GABAergic neurons, which are depolarized by the application of NPS. Simultaneously, we showed that NPS hyperpolarizes sleep-promoting neurons, which is associated with an increased frequency in their spontaneous IPSC inputs. CONCLUSION Altogether, our data reveal that NPS controls local neuronal activity in the VLPO. Following the depolarization of local GABAergic neurons, NPS indirectly provokes feed-forward inhibition onto sleep-promoting neurons, which translates into a decrease in NREM sleep to favor arousal.
Collapse
Affiliation(s)
- Frédéric Chauveau
- IRBA (Armed Biomedical Research Institute), Brétigny-sur-Orge, France
| | - Damien Claverie
- IRBA (Armed Biomedical Research Institute), Brétigny-sur-Orge, France
| | - Emma Lardant
- IRBA (Armed Biomedical Research Institute), Brétigny-sur-Orge, France
| | - Christophe Varin
- Brain Plasticity Unit, CNRS, UMR 8249, ESPCI-ParisTech, PSL Research University, Paris, France.,Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Eléonore Hardy
- Neuroglial Interactions in Cerebral Physiopathology, CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Labex Memolife, PSL Research University, Paris, France
| | - Augustin Walter
- Neuroglial Interactions in Cerebral Physiopathology, CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Labex Memolife, PSL Research University, Paris, France
| | - Frédéric Canini
- IRBA (Armed Biomedical Research Institute), Brétigny-sur-Orge, France.,Ecole du Val de Grâce, Laveran, Paris
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Labex Memolife, PSL Research University, Paris, France
| | - Armelle Rancillac
- Brain Plasticity Unit, CNRS, UMR 8249, ESPCI-ParisTech, PSL Research University, Paris, France.,Neuroglial Interactions in Cerebral Physiopathology, CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
29
|
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
|
30
|
Alshogran OY, Al-Eitan LN, Altawalbeh SM, Khalil AA, Alqudah MAY, Oweis AO, Aman HA, Alhawari HH. Investigating the Contribution of NPSR1, IL-6 and BDNF Polymorphisms to Depressive and Anxiety Symptoms in Hemodialysis Patients. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109657. [PMID: 31132388 DOI: 10.1016/j.pnpbp.2019.109657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/20/2022]
Abstract
AIMS Psychological symptoms are prevalent in hemodialysis (HD) patients. Previous investigations showed that brain-derived neurotrophic factor (BDNF) and interleukin-6 (IL-6) as well as the interaction with neuropeptide S receptor 1 (NPSR1) are linked to the development of psychological distress. This study examined the association of polymorphisms of genes encoding these proteins with depression and anxiety in a representative group of Jordanian HD patients. METHODS A total of 302 HD patients were involved in the study and categorized into three groups based on the Hospital Anxiety and Depression Scale, HADS-D or HADS-A scores as follows: normal (<7), mild (8-10) and moderate-severe (11-21). Single nucleotide polymorphism (SNP) of NPSR1 Asn107Ile (rs324981), IL-6 G174C (rs1800795), and BDNF Val66Met (rs6265) was genotyped using blood samples. RESULTS The frequency of Ile-allele of NPSR1 Asn107Ile was significantly higher in patients with moderate-severe HADS-A scores versus normal (53% vs. 40.8%, p = .035). Using ordinal regression analysis, Asn-allele of NPSR1 polymorphism was nominally significantly associated with a lower risk of anxiety (OR = 0.57, CI: 0.33-0.97, p = .038) after adjusting for other covariates. A marginally significant difference in genotype distribution of IL-6 G174C was observed among patients according to HADS-D scores (p = .05). Furthermore, carriers of IL-6174 CC genotype showed lower median IL-6 serum concentration versus carriers of GG genotype (5.2 vs. 1.35 pg/mL, p < .05). CONCLUSIONS The results support the genetic role of NPSR1 in the pathogenesis of anxiety and suggest that carriers of NPSR1 Ile-allele are at increased risk of anxiety in HD patients. Neither BDNF Val66Met nor IL-6 G174C were linked to psychological symptoms. Future studies among other ethnicities are necessary to verify the observations.
Collapse
Affiliation(s)
- Osama Y Alshogran
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan.
| | - Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan; Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Shoroq M Altawalbeh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Amani A Khalil
- Department of Clinical Nursing, School of Nursing, The University of Jordan, Amman 11942, Jordan
| | - Mohammad A Y Alqudah
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ashraf O Oweis
- Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Hatem A Aman
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Hussein H Alhawari
- Department of Internal Medicine, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
31
|
Zhao P, Qian X, Nie Y, Sun N, Wang Z, Wu J, Wei C, Ma R, Wang Z, Chai G, Li Y. Neuropeptide S Ameliorates Cognitive Impairment of APP/PS1 Transgenic Mice by Promoting Synaptic Plasticity and Reducing Aβ Deposition. Front Behav Neurosci 2019; 13:138. [PMID: 31293402 PMCID: PMC6603143 DOI: 10.3389/fnbeh.2019.00138] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/06/2019] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating disease in the elderly with no known effective treatment. It is characterized by progressive deterioration of memory and cognition. Many new potential targets are being investigated to develop effective therapeutic strategies for AD. Neuropeptide S (NPS) is an endogenous peptide in the central nervous system, which has been shown to play a beneficial role in learning and memory. However, whether NPS can ameliorate cognitive deficits in AD remains unclear. In this study, we examined the effects of NPS treatment on the cognitive behaviors and pathological hallmarks in 8-month-old APPswe/PS1dE9 (APP/PS1) AD mice. We found that the APP/PS1 mice exhibited lower levels of NPS receptors (NPSRs) in the hippocampal area, and NPS administration increased c-Fos expression in the hippocampus and cortex, which suggests the NPS/NPSR system may contribute to the pathogenesis of AD. After an intracerebroventricular injection of NPS (1 nmol) for 2 weeks, we found NPS treatment ameliorated spatial memory deficits and promoted dendrite ramification and spine generation in hippocampal CA1 neurons, which was accompanied by the upregulation of postsynaptic density protein 95 (PSD95) and synapsin1. We also demonstrated that the injection of NPS decreased Aβ plaque deposits by decreasing the γ-secretase activity and the phosphorylation of APP at Thr668. Furthermore, application of NPS reversed the deficits in hippocampal late-phase long-term potentiation (LTP). These findings suggest NPS attenuated cognitive deficits by reducing pathological features in APP/PS1 mice, and NPS might be a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Peng Zhao
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Xiaohang Qian
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Yunjuan Nie
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Na Sun
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | | | - Jiajun Wu
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Chen Wei
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Ruikun Ma
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Zhe Wang
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Gaoshang Chai
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Yuqing Li
- Wuxi Medical School, Jiangnan University, Wuxi, China.,Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
32
|
Sanna MD, Les F, Lopez V, Galeotti N. Lavender ( Lavandula angustifolia Mill.) Essential Oil Alleviates Neuropathic Pain in Mice With Spared Nerve Injury. Front Pharmacol 2019; 10:472. [PMID: 31143116 PMCID: PMC6521744 DOI: 10.3389/fphar.2019.00472] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
Low treatment efficacy represents an important unmet need in neuropathic pain patients and there is an urgent need to develop a more effective pharmacotherapy. An increasing number of patients choose complementary medicine to relieve pain. Lavender essential oil (LEO) is approved by the European Medicines Agency as herbal medicine to relieve anxiety and stress. However, the capability of LEO to relieve other nervous system disorders such as neuropathic pain has never been established. Our work aimed to evaluate the antineuropathic properties of lavender on a spared nerve injury (SNI) model of neuropathic pain in mice. An acute oral administration of LEO (100 mg/kg) alleviated SNI-induced mechanical allodynia, evaluated in the von Frey test, with an intensity comparable to the reference drug pregabalin. Investigations into the mechanism of action showed that LEO markedly decreased the phosphorylation of ERK1, ERK2, and JNK1, and decreased the levels of iNOS in the spinal cord; involvement of the endocannabinoid system was also detected using in vitro inhibition of the FAAH and MALG enzymes as well as in vivo experiments with the CB1 antagonist. Conversely, no effect on P38 phosphorylation and NF-kB activation was detected. These antihyperalgesic effects appeared at the same dose able to induce antidepressant-like, anxiolytic-like, and anorexic effects. In addition, gavage with LEO did not significantly alter animals' gross behavior, motor coordination, or locomotor activity, nor impaired memory functions. Oral administration of LEO could represent a therapeutic approach in the management of neuropathic pain states.
Collapse
Affiliation(s)
- Maria Domenica Sanna
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Victor Lopez
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Nicoletta Galeotti
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
33
|
Bülbül M, Sinen O, Özkan A, Aslan MA, Ağar A. Central neuropeptide-S treatment improves neurofunctions of 6-OHDA-induced Parkinsonian rats. Exp Neurol 2019; 317:78-86. [PMID: 30825442 DOI: 10.1016/j.expneurol.2019.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/12/2019] [Accepted: 02/26/2019] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is characterized by degeneration of the dopaminergic neurons in substantia nigra (SN). The motor symptoms of PD include tremor, rigidity, bradykinesia and postural impairment. In rodents, central administration of neuropeptide-S (NPS) has been shown to induce locomotor activity, dopamine release and neuronal survival by decreasing lipid peroxidation, additionally, the NPS receptor (NPSR) was detected in SN. Accumulating findings suggest that central NPS may ameliorate the parkinsonian symptoms, however, this has been explored incompletely due to the scarcity of experimental studies. Therefore, the present study was designed to test whether central NPS treatment exerts protective and/or alleviative effects on 6-OHDA-induced rat experimental PD model. Adult male Wistar rats received acute (alleviate; 10 nmol, icv) or chronic (protective; 1 nmol, icv for 7 days) NPS treatment following the central injection of 6-OHDA in medial forebrain bundle. Motor performance tests and in vivo nigral microdialysis were performed before and 7 days after the central 6-OHDA injection. The immunoreactivities for tyrosine hydroxylase (TH), NPSR, 4-hydroxynonenal (4-HNE) and c-Fos were detected by immunohistochemistry in frozen SN sections. Our double immunofluorescence labeling studies demonstrated that NPSR is present in the nigral TH-positive neurons. Central NPS injection caused a remarkable c-Fos expression in SN; whereas, no change was observed following vehicle injection. In both chronic and acute treatment groups, the 6-OHDA-induced motor dysfunction and impaired nigral dopamine release were improved significantly. However, only chronic, but not acute treatment restored the loss of nigral TH-positive cells, while decreasing the 4-HNE immunoreactivity in SN. Our findings demonstrate that central NPS treatment not only exerts a neuroprotective action on nigral dopaminergic neurons, it also improves the striatal dopaminergic signaling. Therefore, the present study candidates the NPSR agonism as a novel therapeutic approach for PD treatment.
Collapse
Affiliation(s)
- Mehmet Bülbül
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Osman Sinen
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Ayşe Özkan
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Mutay Aydın Aslan
- Faculty of Medicine, Department of Medical Biochemistry, Akdeniz University, Antalya, Turkey
| | - Aysel Ağar
- Faculty of Medicine, Department of Physiology, Akdeniz University, Antalya, Turkey.
| |
Collapse
|
34
|
Zhang ZR, Tao YX. Physiology, pharmacology, and pathophysiology of neuropeptide S receptor. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 161:125-148. [PMID: 30711025 DOI: 10.1016/bs.pmbts.2018.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neuropeptide S receptor 1 (NPSR1), originally named G protein-coupled receptor 154 (GPR154), was deorphanized in 2002 with neuropeptide S identified as the endogenous ligand. NPSR1 is primarily expressed in bronchus, brain as well as immune cells. It regulates multiple physiological processes, including immunoregulation, locomotor activity, anxiety, arousal, learning and memory, and food intake and energy balance. SNPs of NPSR1 are significantly associated with several diseases, including asthma, anxiolytic and arousal disorders, and rheumatoid arthritis. This chapter will summarize studies on NPSR1, including its molecular structure, tissue distribution, physiology, pharmacology, and pathophysiology.
Collapse
Affiliation(s)
- Zheng-Rui Zhang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States; Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States; Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States.
| |
Collapse
|
35
|
He Q, Shen Z, Ren L, Wang X, Qian M, Zhu J, Shen X. Association of NPSR1 rs324981 polymorphism and treatment response to antidepressants in Chinese Han population with generalized anxiety disorder. Biochem Biophys Res Commun 2018; 504:137-142. [PMID: 30190127 DOI: 10.1016/j.bbrc.2018.08.145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/24/2018] [Indexed: 11/16/2022]
Abstract
In previous studies, neuropeptide S (NPS) and its cognate receptor (NPSR) have been involved in the pathogenesis of anxiety disorders in previous studies. Here, we aimed to investigate the association of NPSR1 polymorphism with generalized anxiety disorder (GAD) and its treatment response in Chinese Han population. Three hundred and thirty seven patients and one hundred and seventy seven healthy controls were involved in our study for 8 weeks. Further, Hamilton Anxiety Scale (HAMA) was used to assess anxiety symptom at baseline and the 1st, 2nd, 4th, 8th week. And all participants were genotyped for NPSR1 (rs324981) variants by polymerase chain reaction. Using Repeated-measures analysis, it showed significant reduction on HAMA scores in patients treated with escitalopram (F = 1.03, P = 0.362) and venlafaxine (F = 0.27, P = 0.763) respectively through 8 weeks treatment. Additionally, patients with AA and TT homozygous genotypes treated with venlafaxine XR had a higher reduction of HAMA scores compared to AT heterozygotic carriers (F = 4.18, P = 0.004), while no significant differences were found in patients treated with escitalopram (F = 1.05, P = 0.383). Thus, our study provides preliminary evidence that NPSR1 AA and TT homozygous genotypes have better treatment responses to venlafaxine XR in Chinese GAD patients, but not to escitalopram. Further studies are needed to verify the observation.
Collapse
Affiliation(s)
- Qianqian He
- Department of Neurosis and Psychosomatic Diseases, Huzhou 3rd Hospital, Huzhou, Zhejiang, 313000, PR China.
| | - Zhongxia Shen
- Department of Neurosis and Psychosomatic Diseases, Huzhou 3rd Hospital, Huzhou, Zhejiang, 313000, PR China.
| | - Lie Ren
- Department of Neurosis and Psychosomatic Diseases, Huzhou 3rd Hospital, Huzhou, Zhejiang, 313000, PR China.
| | - Xing Wang
- Department of Neurosis and Psychosomatic Diseases, Huzhou 3rd Hospital, Huzhou, Zhejiang, 313000, PR China.
| | - Mincai Qian
- Department of Neurosis and Psychosomatic Diseases, Huzhou 3rd Hospital, Huzhou, Zhejiang, 313000, PR China.
| | - Jianying Zhu
- Department of Radiology, Huzhou 3rd Hospital, Huzhou, 313000, PR China.
| | - Xinhua Shen
- Department of Neurosis and Psychosomatic Diseases, Huzhou 3rd Hospital, Huzhou, Zhejiang, 313000, PR China.
| |
Collapse
|
36
|
Grund T, Neumann ID. Brain neuropeptide S: via GPCR activation to a powerful neuromodulator of socio-emotional behaviors. Cell Tissue Res 2018; 375:123-132. [PMID: 30112573 DOI: 10.1007/s00441-018-2902-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/21/2018] [Indexed: 12/19/2022]
Abstract
Neuropeptide S (NPS) has attracted the attention of the scientific community due to its potent anxiolytic-like and fear-attenuating effects studied in rodents. Therefore, NPS might represent a treatment option for neuropsychiatric disorders, such as anxiety disorders, even more so as single nucleotide polymorphisms in the human NPS receptor gene have been associated with increased anxiety traits that contribute to the pathogenesis of fear- and anxiety-related disorders. However, the signaling mechanisms underlying the behavioral effects of NPS and the interaction with other brain neuropeptides are still rather unknown. To illuminate how NPS modulates the expression of selected emotional and social behaviors, the present review focuses on neuroanatomical and electrophysiological studies, as well as intracellular signaling mechanisms following NPS receptor stimulation in rodents. We will also discuss interactions of the NPS system with two well-described neuropeptides, namely corticotropin-releasing factor and oxytocin, which may contribute to the fear- and anxiety-reducing effects.
Collapse
Affiliation(s)
- Thomas Grund
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040, Regensburg, Germany.
| |
Collapse
|
37
|
Jiang J, Peng Y, Liang X, Li S, Chang X, Li L, Chang M. Centrally Administered Cortistation-14 Induces Antidepressant-Like Effects in Mice via Mediating Ghrelin and GABA A Receptor Signaling Pathway. Front Pharmacol 2018; 9:767. [PMID: 30072893 PMCID: PMC6060333 DOI: 10.3389/fphar.2018.00767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/25/2018] [Indexed: 12/22/2022] Open
Abstract
Cortistatin-14 (CST-14), a recently discovered cyclic neuropeptide, can bind to all five cloned somatostatin receptors (SSTRs) and ghrelin receptor to exert its biological activities and co-exists with GABA within the cortex and hippocampus. However, the role of CST-14 in the control of depression processes is not still clarified. Here, we tested the behavioral effects of CST-14 in the in a variety of classical rodent models of depression [forced swimming test (FST), tail suspension test (TST) and novelty-suppressed feeding test]. In the models of depression, CST-14 produced antidepressant-like effects, and does not altered locomotor activity levels. And, we found that CST-14 mRNA and BDNF mRNA were significantly decreased in the hippocampus and cortex after mice exposed to stress. Further data show that i.c.v. administration of CST-14 produce rapid antidepressant effects, and does not altered locomotor activity levels. Then these antidepressant-like effects were significantly reversed by [D-Lys3]GHRP-6 (ghrelin receptor antagonist), but not c-SOM (SSTRs antagonist). Meanwhile, the effects of some neurotransmitter blockers indicates that only GABAA system, but not CRF1 receptor, α/β-adrenergic receptor, is involved in the antidepressant effect of CST-14. The effects of the mTOR inhibitor (rapamycin), the PI3K inhibitor (LY294002) and the p-ERK1/2 inhibitor (U0126) suggesting that the ERK/mTOR or PI3K/Akt/mTOR signaling pathway is not involved in the antidepressant effects of CST-14. Interestingly, intranasal administration of CST-14 led to reducing depressive-like behavior, and near-infrared fluorescent experiments showed the real-time in vivo bio-distribution in brain after intranasal infusion of Cy7.5-CST-14. Taken all together, the results of present study point to a role for CST-14 in the modulation of depression processes via the ghrelin and GABAA receptor, and suggest cortistation may represent a novel strategy for the treatment of depression disorders. Highlights:CST-14 and BDNF mRNA are decreased in hippocampus and cortex once mice exposed to stress. i.c.v. or intranasal administration of CST-14 produce rapid antidepressant effects. NIR fluorescence imaging detected the brain uptake and distribution after intranasal CST-14. Antidepressant effects of CST-14 were only related to ghrelin and GABAA system. Co-injection of CST-14 and NPS produce antidepressant effect, and do not impair memory.
Collapse
Affiliation(s)
- JinHong Jiang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - YaLi Peng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - XueYa Liang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - Shu Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - Xin Chang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - LongFei Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - Min Chang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Life Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| |
Collapse
|
38
|
Bülbül M, Travagli RA. Novel transmitters in brain stem vagal neurocircuitry: new players on the pitch. Am J Physiol Gastrointest Liver Physiol 2018; 315:G20-G26. [PMID: 29597355 PMCID: PMC6109706 DOI: 10.1152/ajpgi.00059.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The last few decades have seen a major increase in the number of neurotransmitters and neuropeptides recognized as playing a role in brain stem neurocircuits, including those involved in homeostatic functions such as stress responsiveness, gastrointestinal motility, feeding, and/or arousal/wakefulness. This minireview will focus on the known physiological role of three of these novel neuropeptides, i.e., apelin, nesfatin-1, and neuropeptide-S, with a special emphasis on their hypothetical roles in vagal signaling related to gastrointestinal motor functions.
Collapse
Affiliation(s)
- Mehmet Bülbül
- 1Faculty of Medicine, Department of Physiology, Akdeniz UniversityAntalya, Turkey
| | - R. Alberto Travagli
- 2Department of Neural and Behavioral Neurosciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
39
|
Jiang JH, Peng YL, Zhang PJ, Xue HX, He Z, Liang XY, Chang M. The ventromedial hypothalamic nucleus plays an important role in anxiolytic-like effect of neuropeptide S. Neuropeptides 2018; 67:36-44. [PMID: 29195839 DOI: 10.1016/j.npep.2017.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/08/2023]
Abstract
Neuropeptide S (NPS), the endogenous neuropeptide ligand of NPSR, has been reported to regulate anxiety-related behavior involved in multiple brain regions, including amygdale, locus coeruleus and Barrington's nucleus. However, little research has been conducted on the anxiolytic-like behaviors of NPS on the hypothalamus, which was an important area in defensive behavior. Here, we investigated a role of hypothalamus in anxiolytic-like behaviors of NPS. We found that NPSR protein of mouse distributed mainly in the ventromedial hypothalamus (VMH). And in the single prolonged stress model (SPS), the results showed that NPS mRNA of the mice exposed to SPS was significantly higher than control, while NPSR mRNA was remarkable lower than control in hypothalamus. Further studies found that NPS intra-VMH infusion dose-dependently (1, 10 and 100pmol) induced anxiolytic effects, using elevated plus maze and open field tests. These anxiolytic effects could be blocked by NPSR antagonist (SHA68), but not by picrotoxin (a GABAA receptor antagonist) and sacolfen (a GABAB receptor antagonist). Meanwhile, our data showed that the expression of c-Fos was significantly increased in VMH after NPS delivered into the lateral ventricles. These results cast a new light on the hypothalamic nucleus in the anxiolytic-like effect of NPS system.
Collapse
Affiliation(s)
- Jin Hong Jiang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Ya Li Peng
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Pei Jiang Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Hong Xiang Xue
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Zhen He
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Xue Ya Liang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - M Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
40
|
Zhu R, Liu X, Shi Y, Wang X, Xue L, Zhao H. Propranolol can induce PTSD-like memory impairments in rats. Brain Behav 2018; 8:e00905. [PMID: 29484264 PMCID: PMC5822589 DOI: 10.1002/brb3.905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction One hallmark symptom of post-traumatic stress disorder (PTSD) is an inability to restrict fear responses to the appropriate predictor. An infusion of glucocorticoids (GCs) after a high-intensity shock has been shown to induce PTSD-like memory impairments. In addition to GCs, noradrenergic signalling is also recognized as a key biomarker underlying PTSD symptomatology. Methods To explore the role of the noradrenergic system in PTSD-like memory impairments, in this study, various doses of the β-adrenoceptor antagonist propranolol were systemically or bilaterally injected into the dorsal hippocampus immediately after unpaired cue-shock contextual fear conditioning, and then the rats were tested 24 h later. Results Interestingly, we found that only low-dose propranolol could induce PTSD-like memory impairments, as rats showed reduced freezing to the correct predictor and generalized fear responses to the safe cues, accompanied by increased NE levels in the hippocampus and altered neural activity within the frontal-subcortical circuit. Conclusion These findings demonstrate that the noradrenergic system is involved in regulating the consolidation of contextual fear memory and that propranolol can dose-dependently induce PTSD-like memory impairments.
Collapse
Affiliation(s)
- Rong‐Ting Zhu
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Xiang‐Hui Liu
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Yan‐Wei Shi
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Xiao‐Guang Wang
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Li Xue
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Hu Zhao
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
41
|
Domschke K, Akhrif A, Romanos M, Bajer C, Mainusch M, Winkelmann J, Zimmer C, Neufang S. Neuropeptide S Receptor Gene Variation Differentially Modulates Fronto-Limbic Effective Connectivity in Childhood and Adolescence. Cereb Cortex 2018; 27:554-566. [PMID: 26503268 DOI: 10.1093/cercor/bhv259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The neuropeptide S (NPS) system contributes to the pathogenesis of anxiety. The more active T allele of the functional rs324981 variant in the neuropeptide S receptor gene (NPSR1) is associated with panic disorder (PD) and distorted cortico-limbic activity during emotion processing in healthy adults and PD patients. This study investigated the influence of NPSR1 genotype on fronto-limbic effective connectivity within the developing brain. Sixty healthy subjects (8-21 years) were examined using an emotional go-nogo task and fMRI. Fronto-limbic connectivity was determined using Dynamic Causal Modeling. In A allele carriers, connectivity between the right middle frontal gyrus (MFG) and the right amygdala was higher in older (≥14 years) than that in younger (<14 years) probands, whereas TT homozygotes ≥14 years showed a reduction of fronto-limbic connectivity between the MFG and both the amygdala and the insula. Fronto-limbic connectivity varied between NPSR1 genotypes in the developing brain suggesting a risk-increasing effect of the NPSR1T allele for anxiety-related traits via impaired top-down control of limbic structures emerging during adolescence. Provided robust replication in longitudinal studies, these findings may constitute valuable biomarkers for early targeted prevention of anxiety disorders.
Collapse
Affiliation(s)
| | - Atae Akhrif
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Marcel Romanos
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Christina Bajer
- Department of Diagnostical and Interventional Neuroradiology.,Department of Neurology and
| | - Margrit Mainusch
- Department of Diagnostical and Interventional Neuroradiology.,Department of Neurology and
| | - Juliane Winkelmann
- Institute of Human Genetics, Technical University Munich, Munich, Germany.,Institute of Human Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Claus Zimmer
- Department of Diagnostical and Interventional Neuroradiology
| | - Susanne Neufang
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
42
|
Abstract
SummaryAs many as 10% of the population experience post-traumatic stress disorder (PTSD) at some time in their lives. It often runs a severe, chronic and treatment-resistant course. This article reviews the evidence base for typically recommended treatments such as cognitive-behavioural therapy (CBT), eye movement desensitisation and reprocessing and selective serotonin reuptake inhibitors (SSRIs). It tabulates the major randomised controlled trials of SSRIs and trauma-focused CBT and reviews research on novel treatments such as ketamine, MDMA, quetiapine, propranolol and prazosin.
Collapse
|
43
|
Cohen H, Vainer E, Zeev K, Zohar J, Mathé AA. Neuropeptide S in the basolateral amygdala mediates an adaptive behavioral stress response in a rat model of posttraumatic stress disorder by increasing the expression of BDNF and the neuropeptide YY1 receptor. Eur Neuropsychopharmacol 2018; 28:159-170. [PMID: 29157796 DOI: 10.1016/j.euroneuro.2017.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/17/2017] [Accepted: 11/03/2017] [Indexed: 01/19/2023]
Abstract
Neuropeptide S (NPS) is a regulatory peptide that has anxiolytic and arousal-promoting effects in rodents. We used an animal model of posttraumatic stress disorder (PTSD) to assess long-term behavioral effects of a single dose of NPS, microinjected into the basolateral amygdala (BLA) 1h following exposure to predator-scent stress (PSS). To elucidate the molecular mechanism by which NPS attenuates behavioral stress responses, expression levels of neuropeptide Y (NPY), NPY-Y1 receptor (NPY-Y1R), and brain-derived neurotrophic factor (BDNF) were evaluated in the hippocampus. The behavioral and molecular effects of NPS receptor antagonist (NPS-RA), NPY-Y1R antagonist (NPY-Y1RA), or both administered centrally were evaluated in the same manner. Circulating corticosterone levels were measured at different time points following PSS-exposure. Immediate post-exposure treatment with NPS had a marked protective effect; BLA microinfusion of NPS completely abolished the extreme behavioral response to PSS, restored the decreased expression of BDNF and, unexpectedly, PY-Y1R, but didn't affect the decreased expression of NPY. BLA microinfusion of both NPY-Y1RA and NPS-RA together had an additive effect, which completely prevented the anxiolytic effects of NPS in rats exposed to PSS and disrupted the expression of NPY-Y1R in the hippocampus following NPS infusion. It may therefore be hypothesized that NPS acts, directly or indirectly, on both the NPY-Y1R and NPS receptors and that the cross-talk between NPS and NPY-Y1R may be necessary for the anxiolytic effects of NPS post-exposure. The NPS system might thus contribute to a potential endogenous mechanism underlying the shift towards adaptive behavioral response and thereby might be relevant as a pharmacological target for attenuating stress-related sequelae.
Collapse
Affiliation(s)
- Hagit Cohen
- Ministry of Health Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 4600, Beer-Sheva 84170, Israel.
| | - Ella Vainer
- Ministry of Health Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 4600, Beer-Sheva 84170, Israel
| | - Kaplan Zeev
- Ministry of Health Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 4600, Beer-Sheva 84170, Israel
| | - Joseph Zohar
- Division of Psychiatry, The State of Israel Ministry of Health, The Chaim Sheba Medical Center, Ramat-Gan, Israel, Sackler Medical School, Tel-Aviv University, Israel
| | - Aleksander A Mathé
- Karolinska Institutet, Department of Clinical Neuroscience, Karolinska Institutet, Sankt Görans Hospital, SE-11281 Stockholm, Sweden.
| |
Collapse
|
44
|
Thomasson J, Canini F, Poly-Thomasson B, Trousselard M, Granon S, Chauveau F. Neuropeptide S overcomes short term memory deficit induced by sleep restriction by increasing prefrontal cortex activity. Eur Neuropsychopharmacol 2017; 27:1308-1318. [PMID: 28941995 DOI: 10.1016/j.euroneuro.2017.08.431] [Citation(s) in RCA: 7] [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: 12/02/2016] [Revised: 07/27/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
Sleep restriction (SR) impairs short term memory (STM) that might be related to different processes. Neuropeptide S (NPS), an endogenous neuropeptide that improves short term memory, activates arousal and decreases anxiety is likely to counteract the SR-induced impairment of STM. The objective of the present study was to find common cerebral pathways in sleep restriction and NPS action in order to ultimately antagonize SR effect on memory. The STM was assessed using a spontaneous spatial alternation task in a T-maze. C57-Bl/6J male mice were distributed in 4 groups according to treatment (0.1nmol of NPS or vehicle intracerebroventricular injection) and to 20h-SR. Immediately after behavioural testing, regional c-fos immunohistochemistry was performed and used as a neural activation marker for spatial short term memory (prefrontal cortex, dorsal hippocampus) and emotional reactivity (basolateral amygdala and ventral hippocampus). Anxiety-like behaviour was assessed using elevated-plus maze task. Results showed that SR impaired short term memory performance and decreased neuronal activation in cingular cortex.NPS injection overcame SR-induced STM deficits and increased neuronal activation in infralimbic cortex. SR spared anxiety-like behavior in the elevated-plus maze. Neural activation in basolateral nucleus of amygdala and ventral hippocampus were not changed after SR.In conclusion, the present study shows that NPS overcomes SR-induced STM deficits by increasing prefrontal cortex activation independently of anxiety-like behaviour.
Collapse
Affiliation(s)
- Julien Thomasson
- Institut de Recherche Biomédicale des Armées Brétigny-sur-Orge, France
| | - Frédéric Canini
- Institut de Recherche Biomédicale des Armées Brétigny-sur-Orge, France; Ecole du Val de Grâce, 1 Place Laveran, Paris, France
| | | | - Marion Trousselard
- Institut de Recherche Biomédicale des Armées Brétigny-sur-Orge, France; Ecole du Val de Grâce, 1 Place Laveran, Paris, France
| | - Sylvie Granon
- Institut des Neurosciences Paris-Saclay (Neuro-PSI), CNRS UMR 9197, Université Paris-Saclay, Orsay, France
| | - Frédéric Chauveau
- Institut de Recherche Biomédicale des Armées Brétigny-sur-Orge, France.
| |
Collapse
|
45
|
Jinushi K, Kushikata T, Kudo T, Calo G, Guerrini R, Hirota K. Central noradrenergic activity affects analgesic effect of Neuropeptide S. J Anesth 2017; 32:48-53. [PMID: 29128909 DOI: 10.1007/s00540-017-2427-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/02/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Neuropeptide S (NPS) is an endogenous neuropeptide controlling anxiolysis, wakefulness, and analgesia. NPS containing neurons exist near to the locus coeruleus (LC) involved in the descending anti-nociceptive system. NPS interacts with central noradrenergic neurons; thus brain noradrenergic signaling may be involved in NPS-induced analgesia. We tested NPS analgesia in noradrenergic neuron-lesioned rats using a selective LC noradrenergic neurotoxin, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4). METHODS A total 66 male Sprague-Dawley rats weighing 350-450 g were used. Analgesic effects of NPS were evaluated using hot-plate and tail-flick test with or without DSP-4. The animal allocated into 3 groups; hot-plate with NPS alone intracerebroventricular (icv) (0.0, 1.0, 3.3, and 10.0 nmol), tail-flick NPS alone icv (0.0 and 10.0 nmol), and hot-plate with NPS and DSP-4 (0 or 50 mg/kg ip). In hot-plate with NPS and DSP-4 group, noradrenaline content in the cerebral cortex, pons, hypothalamus, were measured. RESULTS NPS 10 nmol icv prolonged hot plate (%MPE) but not tail flick latency at 30 and 40 min after administration. DSP-4 50 mg/kg decreased noradrenaline content in the all 3 regions. The NA depletion inhibited NPS analgesic effect in the hot plate test but not tail flick test. There was a significant correlation between hot plate latency (percentage of maximum possible effect: %MPE) with NPS 10 nmol and NA content in the cerebral cortex (p = 0.017, r 2 = 0.346) which noradrenergic innervation arisen mainly from the LC. No other regions had the correlation. CONCLUSIONS NPS analgesia interacts with LC noradrenergic neuronal activity.
Collapse
Affiliation(s)
- Kei Jinushi
- Department of Anesthesiology, Hirosaki University Hospital, Hirosaki, 036-8563, Japan
| | - Tetsuya Kushikata
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Zaifu 5, Hirosaki, 036-8562, Japan.
| | - Takashi Kudo
- Department of Anesthesiology, Hirosaki University Hospital, Hirosaki, 036-8563, Japan
| | - Girolamo Calo
- Section of Pharmacology, Department of Medical Science, National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, LTTA, University of Ferrara, 44121, Ferrara, Italy
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Zaifu 5, Hirosaki, 036-8562, Japan
| |
Collapse
|
46
|
Ensho T, Nakahara K, Suzuki Y, Murakami N. Neuropeptide S increases motor activity and thermogenesis in the rat through sympathetic activation. Neuropeptides 2017; 65:21-27. [PMID: 28433253 DOI: 10.1016/j.npep.2017.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 04/05/2017] [Accepted: 04/08/2017] [Indexed: 12/01/2022]
Abstract
The central role of neuropeptide S (NPS), identified as the endogenous ligand for GPR154, now named neuropeptide S receptor (NPSR), has not yet been fully clarified. We examined the central role of NPS for body temperature, energy expenditure, locomotor activity and adrenal hormone secretion in rats. Intracerebroventricular (icv) injection of NPS increased body temperature in a dose-dependent manner. Energy consumption and locomotor activity were also significantly increased by icv injection of NPS. In addition, icv injection of NPS increased the peripheral blood concentration of adrenalin and corticosterone. Pretreatment with the β1- and β2-adrenergic receptor blocker timolol inhibited the NPS-induced increase of body temperature. The expression of both NPS mRNA in the brainstem and NPSR mRNA in the hypothalamus showed a nocturnal rhythm with a peak occurring during the first half of the dark period. To examine whether the endogenous NPS is involved in regulation of body temperature, NPSR antagonist SHA68 was administered one hour after darkness. SHA68 attenuated the nocturnal rise of body temperature. These results suggest that NPS contributes to the regulation of the sympathetic nervous system.
Collapse
Affiliation(s)
- Takuya Ensho
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | - Keiko Nakahara
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan.
| | - Yoshihiro Suzuki
- Laboratory of Animal Health Science, School of Veterinary Medicine, Kitasato University, Aomori 034-8628, Japan
| | - Noboru Murakami
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| |
Collapse
|
47
|
Role of G Protein-Coupled Receptors in the Regulation of Structural Plasticity and Cognitive Function. Molecules 2017; 22:molecules22071239. [PMID: 28737723 PMCID: PMC6152405 DOI: 10.3390/molecules22071239] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Cognition and other higher brain functions are known to be intricately associated with the capacity of neural circuits to undergo structural reorganization. Structural remodelling of neural circuits, or structural plasticity, in the hippocampus plays a major role in learning and memory. Dynamic modifications of neuronal connectivity in the form of dendritic spine morphology alteration, as well as synapse formation and elimination, often result in the strengthening or weakening of specific neural circuits that determine synaptic plasticity. Changes in dendritic complexity and synapse number are mediated by cellular processes that are regulated by extracellular signals such as neurotransmitters and neurotrophic factors. As many neurotransmitters act on G protein-coupled receptors (GPCRs), it has become increasingly apparent that GPCRs can regulate structural plasticity through a myriad of G protein-dependent pathways and non-canonical signals. A thorough understanding of how GPCRs exert their regulatory influence on dendritic spine morphogenesis may provide new insights for treating cognitive impairment and decline in various age-related diseases. In this article, we review the evidence of GPCR-mediated regulation of structural plasticity, with a special emphasis on the involvement of common as well as distinct signalling pathways that are regulated by major neurotransmitters.
Collapse
|
48
|
Intrahippocampal injection of Cortistatin-14 impairs recognition memory consolidation in mice through activation of sst 2 , ghrelin and GABA A/B receptors. Brain Res 2017; 1666:38-47. [DOI: 10.1016/j.brainres.2017.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 02/03/2023]
|
49
|
Roncacè V, Polli FS, Zojicic M, Kohlmeier KA. Neuropeptide S (NPS) is a neuropeptide with cellular actions in arousal and anxiety-related nuclei: Functional implications for effects of NPS on wakefulness and mood. Neuropharmacology 2017; 126:292-317. [PMID: 28655610 DOI: 10.1016/j.neuropharm.2017.06.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/02/2017] [Accepted: 06/23/2017] [Indexed: 12/27/2022]
Abstract
Neuropeptide S (NPS) is a peptide recently recognized to be present in the CNS, and believed to play a role in vigilance and mood control, as behavioral studies have shown it promotes arousal and has an anxiolytic effect. Although NPS precursor is found in very few neurons, NPS positive fibers are present throughout the brain stem. Given the behavioral actions of this peptide and the wide innervation pattern, we examined the cellular effects of NPS within two brain stem nuclei known to play a critical role in anxiety and arousal: the dorsal raphe (DR) and laterodorsal tegmentum (LDT). In mouse brain slices, NPS increased cytoplasmic levels of calcium in DR and LDT cells. Calcium rises were independent of action potential generation, reduced by low extracellular levels of calcium, attenuated by IP3 - and ryanodine (RyR)-dependent intracellular calcium store depletion, and eliminated by the receptor (NPSR) selective antagonist, SHA 68. NPS also exerted an effect on the membrane of DR and LDT cells inducing inward and outward currents, which were driven by an increase in conductance, and eliminated by SHA 68. Membrane actions of NPS were found to be dependent on store-mediated calcium as depletion of IP3 and RyR stores eliminated NPS-induced currents. Finally, NPS also had actions on synaptic events, suggesting facilitation of glutamatergic and GABAergic presynaptic transmission. When taken together, actions of NPS influenced the excitability of DR and LDT neurons, which could play a role in the anxiolytic and arousal-promoting effects of this peptide.
Collapse
Affiliation(s)
- Vincenzo Roncacè
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen Ø, Denmark
| | - Filip Souza Polli
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen Ø, Denmark
| | - Minella Zojicic
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen Ø, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
50
|
Liu X, Si W, Garau C, Jüngling K, Pape HC, Schulz S, Reinscheid RK. Neuropeptide S precursor knockout mice display memory and arousal deficits. Eur J Neurosci 2017; 46:1689-1700. [PMID: 28548278 DOI: 10.1111/ejn.13613] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/02/2017] [Accepted: 05/18/2017] [Indexed: 12/11/2022]
Abstract
Activation of neuropeptide S (NPS) signaling has been found to produce arousal, wakefulness, anxiolytic-like behaviors, and enhanced memory formation. In order to further study physiological functions of the NPS system, we generated NPS precursor knockout mice by homologous recombination in embryonic stem cells. NPS-/- mice were viable, fertile, and anatomically normal, when compared to their wild-type and heterozygous littermates. The total number of NPS neurons-although no longer synthesizing the peptide - was not affected by the knockout, as analyzed in NPS-/- /NPSEGFP double transgenic mice. Analysis of behavioral phenotypes revealed significant deficits in exploratory activity in NPS-/- mice. NPS precursor knockout mice displayed attenuated arousal in the hole board test, visible as reduced total nose pokes and number of holes inspected, that was not confounded by increased repetitive or stereotypic behavior. Importantly, long-term memory was significantly impaired in NPS-/- mice in the inhibitory avoidance paradigm. NPS precursor knockout mice displayed mildly increased anxiety-like behaviors in three different tests measuring responses to stress and novelty. Interestingly, heterozygous littermates often presented behavioral deficits similar to NPS-/- mice or displayed intermediate phenotype. These observations may suggest limited ligand availability in critical neural circuits. Overall, phenotypical changes in NPS-/- mice are similar to those observed in NPS receptor knockout mice and support earlier findings that suggest major functions of the NPS system in arousal, regulation of anxiety and stress, and memory formation.
Collapse
Affiliation(s)
- Xiaobin Liu
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA.,Department of Pharmaceutical Science, University of North Texas Health Sciences Center, Fort Worth, TX, USA
| | - Wei Si
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Celia Garau
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Kay Jüngling
- Institute of Physiology I, University Hospital Münster, Westfälische-Wilhems-University, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Hans-Christian Pape
- Institute of Physiology I, University Hospital Münster, Westfälische-Wilhems-University, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Stefan Schulz
- Institute of Pharmacology & Toxicology, Friedrich-Schiller-University, Jena, Germany
| | - Rainer K Reinscheid
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA.,Institute of Physiology I, University Hospital Münster, Westfälische-Wilhems-University, Robert-Koch-Str. 27a, D-48149, Münster, Germany.,Institute of Pharmacology & Toxicology, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|