1
|
Wang D, Wang G, Wang X, Ren Z, Jia C. Native Mass Spectrometry-Centric Approaches Revealed That Neuropeptides Frequently Interact with Amyloid-β. ACS Chem Neurosci 2024; 15:2719-2728. [PMID: 39066700 DOI: 10.1021/acschemneuro.4c00075] [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] [Indexed: 07/30/2024] Open
Abstract
Amyloid-β (Aβ) aggregates are recognized as initiators of Alzheimer's disease, and their interaction with the nervous system contributes to the progression of neurodegeneration. Herein, we investigated the frequency at which neuropeptides interact with Aβ and affect the aggregation kinetics and cytotoxicity of Aβ. To this end, we established a native mass spectrometry (MS)-centric workflow for screening Aβ-interacting neuropeptides, and six out of 12 neuropeptides formed noncovalent complexes with Aβ species in the MS gas phase. Thioflavin-T fluorescence assays and gel separation indicated that leptin and cerebellin decreased Aβ aggregation, whereas kisspeptin increased this process. In addition, leptin and cerebellin attenuated Aβ-induced cytotoxicity, which was independent of the influence of metal ions. Leptin can chelate copper from copper-bound Aβ species, reducing the cytotoxicity caused by the aggregation of Aβ and metal ion complexes. Overall, our study demonstrated that neuropeptides frequently interact with Aβ and revealed that leptin and cerebellin are potential inhibitors of Aβ aggregation, providing great insight into understanding the molecular mechanism of Aβ interacting with the nervous system and facilitating drug development.
Collapse
Affiliation(s)
- Danyang Wang
- Department of Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
- Beijing Proteome Research Center, State Key Laboratory of Medical Proteomics, Beijing Institute of Lifeomics, National Center for Protein Sciences-Beijing, Beijing 102206, China
| | - Guibin Wang
- Beijing Proteome Research Center, State Key Laboratory of Medical Proteomics, Beijing Institute of Lifeomics, National Center for Protein Sciences-Beijing, Beijing 102206, China
| | - Xiankun Wang
- Beijing Proteome Research Center, State Key Laboratory of Medical Proteomics, Beijing Institute of Lifeomics, National Center for Protein Sciences-Beijing, Beijing 102206, China
| | - Zhenhua Ren
- Department of Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chenxi Jia
- Department of Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
- Beijing Proteome Research Center, State Key Laboratory of Medical Proteomics, Beijing Institute of Lifeomics, National Center for Protein Sciences-Beijing, Beijing 102206, China
| |
Collapse
|
2
|
Xiao-Hang Q, Si-Yue C, Hui-Dong T. Multi-strain probiotics ameliorate Alzheimer's-like cognitive impairment and pathological changes through the AKT/GSK-3β pathway in senescence-accelerated mouse prone 8 mice. Brain Behav Immun 2024; 119:14-27. [PMID: 38548184 DOI: 10.1016/j.bbi.2024.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most prevalent type of dementia, still lacks disease-modifying treatment strategies. Recent evidence indicates that maintaining gut microbiota homeostasis plays a crucial role in AD. Targeted regulation of gut microbiota, including probiotics, is anticipated to emerge as a potential approach for AD treatment. However, the efficacy and mechanism of multi-strain probiotics treatment in AD remain unclear. METHODS In this study, 6-month-old senescence-accelerated-mouse-prone 8 (SAMP8) and senescence-accelerated-mouse-resistant 1 (SAMR1) were utilized. The SAMP8 mice were treated with probiotic-2 (P2, a probiotic mixture of Bifidobacterium lactis and Lactobacillus rhamnosus) and probiotic-3 (P3, a probiotic mixture of Bifidobacterium lactis, Lactobacillus acidophilus, and Lactobacillus rhamnosus) (1 × 109 colony-forming units) once daily for 8 weeks. Morris water maze (MWM) and novel object recognition (NOR) tests were employed to assess the memory ability. 16S sequencing was applied to determine the composition of gut microbiota, along with detecting serum short-chain fatty acids (SCFAs) concentrations. Neural injury, Aβ and Tau pathology, and neuroinflammation level were assessed through western blot and immunofluorescence. Finally, potential molecular mechanisms was explored through transcriptomic analysis and western blotting. RESULTS The MWM and NOR test results indicated a significant improvement in the cognitive level of SAMP8 mice treated with P2 and P3 probiotics compared to the SAMP8 control group. Fecal 16S sequencing revealed an evident difference in the α diversity index between SAMP8 and SAMR1 mice, while the α diversity of SAMP8 mice remained unchanged after P2 and P3 treatment. At the genus level, the relative abundance of ten bacteria differed significantly among the four groups. Multi-strain probiotics treatment could modulate serum SCFAs (valeric acid, isovaleric acid, and hexanoic acid) concentration. Neuropathological results demonstrated a substantial decrease in neural injury, Aβ and Tau pathology and neuroinflammation in the brain of SAMP8 mice treated with P3 and P2. Transcriptomic analysis identified the chemokine signaling pathway as the most significantly enriched signaling pathway between SAMP8 and SAMR1 mice. Western blot test indicated a significant change in the phosphorylation level of downstream AKT/GSK-3β between the SAMP8 and SAMR1 groups, which could be reversed through P2 and P3 treatment. CONCLUSIONS Multi-strain probiotics treatment can ameliorate cognitive impairment and pathological change in SAMP8 mice, including neural damage, Aβ and Tau pathology, and neuroinflammation. This effect is associated with the regulation of the phosphorylation of the AKT/GSK-3β pathway.
Collapse
Affiliation(s)
- Qian Xiao-Hang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Si-Yue
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tang Hui-Dong
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
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
|
4
|
Wallace CH, Oliveros G, Xie L, Serrano P, Rockwell P, Figueiredo-Pereira M. Potential Alzheimer's early biomarkers in a transgenic rat model and benefits of diazoxide/dibenzoylmethane co-treatment on spatial memory and AD-pathology. Sci Rep 2024; 14:3730. [PMID: 38355687 PMCID: PMC10867006 DOI: 10.1038/s41598-024-54156-z] [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: 10/16/2023] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Alzheimer's disease (AD) is the major form of dementia prevalent in older adults and with a high incidence in females. Identification of early biomarkers is essential for preventive intervention to delay its progression. Furthermore, due to its multifactorial nature, a multi-target approach could be therapeutically beneficial. Our studies included 4- (pre-pathology) and 11-month (mild-pathology) TgF344-AD rats, a transgenic Alzheimer's model that exhibits age-dependent AD progression. We identified two potential early biomarker genes for AD, early growth response 2 (EGR2) and histone 1H2AA (HIST1H2AA), in the hippocampus of 4-month females. Out of 17,168 genes analyzed by RNA sequencing, expression of these two genes was significantly altered in 4-month TgF344-AD rats compared to wild-type littermates. We also evaluated co-treatment with diazoxide (DZ), a potassium channel activator, and dibenzoylmethane (DIB), which inhibits eIF2α-P activity, on TgF344-AD and wild-type rats. DZ/DIB-treatment mitigated spatial memory deficits and buildup of hippocampal Aβ plaques and tau PHF in 11-month TgF344-AD rats but had no effect on wild-type littermates. To our knowledge, this preclinical study is the first to report EGR2 and HIST1H2AA as potential AD biomarkers in females, and the benefits of DZ/DIB-treatment in AD. Evaluations across multiple AD-related models is warranted to corroborate our findings.
Collapse
Affiliation(s)
- Charles H Wallace
- Department of Biological Sciences, Hunter College CUNY and Graduate Center, 695 Park Ave., New York, NY, USA
| | - Giovanni Oliveros
- Department of Biological Sciences, Hunter College CUNY and Graduate Center, 695 Park Ave., New York, NY, USA
| | - Lei Xie
- Department of Computer Sciences, Hunter College CUNY, New York, NY, USA
| | - Peter Serrano
- Department of Psychology, Hunter College CUNY, New York, NY, USA
| | - Patricia Rockwell
- Department of Biological Sciences, Hunter College CUNY and Graduate Center, 695 Park Ave., New York, NY, USA
| | - Maria Figueiredo-Pereira
- Department of Biological Sciences, Hunter College CUNY and Graduate Center, 695 Park Ave., New York, NY, USA.
| |
Collapse
|
5
|
Qian X, Hai W, Chen S, Zhang M, Jiang X, Tang H. Multi-omics data reveals aberrant gut microbiota-host glycerophospholipid metabolism in association with neuroinflammation in APP/PS1 mice. Gut Microbes 2023; 15:2282790. [PMID: 37992400 PMCID: PMC10730179 DOI: 10.1080/19490976.2023.2282790] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
Numerous studies have described the notable impact of gut microbiota on the brain in Alzheimer's disease (AD) via the gut - brain axis. However, the molecular mechanisms underlying the involvement of gut microbiota in the development of AD are limited. This study aimed to explore the potential mechanisms of gut microbiota in AD by integrating multi-omics data. In this study, APP/PS1 and WT mice at nine months of age were used as study mouse model. Cognitive function was assessed using the Morris water maze test. The levels of Aβ plaque and neuroinflammation in the brain were detected using immunofluorescence and PET/CT. In addition, we not only used 16S rRNA gene sequencing and metabolomics to explore the variation characteristics of gut microbiota and serum metabolism abundance, but also combined spatial metabolomics and transcriptomics to explore the change in the brain and identify their potential correlation. APP/PS1 mice showed significant cognitive impairment and amyloid-β deposits in the brain. The abundance of gut microbiota was significantly changed in APP/PS1 mice, including decreased Desulfoviobrio, Enterococcus, Turicibacter, and Ruminococcus and increased Pseudomonas. The integration of serum untargeted metabolomics and brain spatial metabolomics showed that glycerophospholipid metabolism was a common alteration pathway in APP/PS1 mice. Significant proliferation and activation of astrocyte and microglia were observed in APP/PS1 mice, accompanied by alterations in immune pathways. Integration analysis and fecal microbiota transplantation (FMT) intervention revealed potential association of gut microbiota, host glycerophospholipid metabolism, and neuroinflammation levels in APP/PS1 mice.
Collapse
Affiliation(s)
- Xiaohang Qian
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyue Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xufeng Jiang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huidong Tang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Nie Y, Liang J, Sun J, Li J, Zhai X, Zhao P. Orexin A alleviates LPS-induced acute lung injury by inhibiting macrophage activation through JNK-mediated autophagy. Int Immunopharmacol 2023; 124:111018. [PMID: 37801969 DOI: 10.1016/j.intimp.2023.111018] [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: 07/07/2023] [Revised: 09/16/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023]
Abstract
Crosstalk between the central nervous system and immune system by the neuroendocrine and autonomic nervous systems is critical during the inflammatory response. Exposure to endotoxin alters the activity of hypothalamic homeostatic systems, resulting in changed transmitter release within the brain. This study investigated the effects and cellular molecular mechanisms of neurogenic and exogenous orexin-A (OXA) in LPS-induced acute lung injury (ALI). We found the production of OXA in the hypothalamus and lungs was both decreased following LPS infection. LPS-induced lung injury including the destruction of the structure, inflammatory cell infiltration, and pro-inflammatory cytokines generation was aggravated in mice in which orexin neurons were lesioned with the neurotoxin orexin-saporin (orexin-SAP). Administration of exogenous OXA greatly improved lung pathology and reduced inflammatory response. Orexin receptors were found in cultured mouse bone marrow-derived macrophages (BMDMs) and lung macrophages (LMs), adoptive transfer of OXA-treated macrophages showed alleviative lung injury compared to adoptive transfer of macrophages without OXA treatment. Mechanistically, it is the induction of autophagy via JNK activation that is responsible for OXA to suppress macrophage-derived pro-inflammatory cytokine production. These findings highlight the importance of neuro-immune crosstalk and indicate that OXA may be a potential therapeutic agent in the treatment of ALI.
Collapse
Affiliation(s)
- Yunjuan Nie
- Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi 214000, Jiangsu Province, PR China; Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Junjie Liang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Jie Sun
- Department of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi 214000, PR China
| | - Jiao Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Xiaorun Zhai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Peng Zhao
- Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi 214000, Jiangsu Province, PR China; Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR 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: 0.7] [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
|
Qian XH, Liu XL, Chen G, Chen SD, Tang HD. Injection of amyloid-β to lateral ventricle induces gut microbiota dysbiosis in association with inhibition of cholinergic anti-inflammatory pathways in Alzheimer's disease. J Neuroinflammation 2022; 19:236. [PMID: 36171620 PMCID: PMC9520842 DOI: 10.1186/s12974-022-02599-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease and its pathogenesis is still unclear. There is dysbiosis of gut microbiota in AD patients. More importantly, dysbiosis of the gut microbiota has been observed not only in AD patients, but also in patients with mild cognitive impairment (MCI). However, the mechanism of gut microbiota dysbiosis in AD is poorly understood. Cholinergic anti-inflammatory pathway is an important pathway for the central nervous system (CNS) regulation of peripheral immune homeostasis, especially in the gut. Therefore, we speculated that dysfunction of cholinergic anti-inflammatory pathway is a potential pathway for dysbiosis of the gut microbiota in AD. METHODS In this study, we constructed AD model mice by injecting Aβ1-42 into the lateral ventricle, and detected the cognitive level of mice by the Morris water maze test. In addition, 16S rDNA high-throughput analysis was used to detect the gut microbiota abundance of each group at baseline, 2 weeks and 4 weeks after surgery. Furthermore, immunofluorescence and western blot were used to detect alteration of intestinal structure of mice, cholinergic anti-inflammatory pathway, and APP process of brain and colon in each group. RESULTS Aβ1-42 i.c.v induced cognitive impairment and neuron damage in the brain of mice. At the same time, Aβ1-42 i.c.v induced alteration of gut microbiota at 4 weeks after surgery, while there was no difference at the baseline and 2 weeks after surgery. In addition, changes in colon structure and increased levels of pro-inflammatory factors were detected in Aβ1-42 treatment group, accompanied by inhibition of cholinergic anti-inflammatory pathways. Amyloidogenic pathways in both the brain and colon were accelerated in Aβ1-42 treatment group. CONCLUSIONS The present findings suggested that Aβ in the CNS can induce gut microbiota dysbiosis, alter intestinal structure and accelerate the amyloidogenic pathways, which were related to inhibiting cholinergic anti-inflammatory pathways.
Collapse
Affiliation(s)
- Xiao-Hang Qian
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406, China
| | - Guang Chen
- The Second Hospital of Anhui Medical University, Anhui, 230601, China
| | - Sheng-di Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Dong Tang
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Medical Center on Aging of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
10
|
Markiewicz-Gospodarek A, Markiewicz R, Dobrowolska B, Rahnama M, Łoza B. Relationship of Neuropeptide S (NPS) with Neurocognitive, Clinical, and Electrophysiological Parameters of Patients during Structured Rehabilitation Therapy for Schizophrenia. J Clin Med 2022; 11:jcm11185266. [PMID: 36142912 PMCID: PMC9506378 DOI: 10.3390/jcm11185266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/29/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction: Neuropeptide S is a biomarker related to various neuropsychiatric and neurocognitive functions. Since the need to improve cognitive functions in schizophrenia is unquestionable, it was valuable to investigate the possible relationships of plasma levels of NPS with neurocognitive, psychopathological and EEG parameters in patients with schizophrenia. Aim: Relationships between the serum NPS level and neurocognitive, clinical, and electrophysiological parameters were investigated in patients diagnosed with schizophrenia who underwent structured rehabilitation therapy. Methods: Thirty-three men diagnosed with schizophrenia were randomized into two groups. The REH group (N16) consisted of patients who underwent structured rehabilitation therapy, the CON group (N17) continued its previous treatment. Additionally, the reference NPS serum results were checked in a group of healthy people (N15). In the study several tests assessing various neurocognitive functions were used: d2 Sustained-Attention Test (d2), Color Trails Test (CTT), Beck Cognitive Insight Scale (BCIS), Acceptance of Illness Scale (AIS), and General Self-Efficacy Scale (GSES). The clinical parameters were measured with Positive and Negative Syndrome Scale (PANSS) and electrophysiological parameters were analyzed with auditory evoked potentials (AEPs) and quantitative electroencephalography (QEEG). The NPS, neurocognitive, clinical, and electrophysiological results of REH and CON groups were recorded at the beginning (T1) and after a period of 3 months (T2). Results: A decreased level of NPS was associated with the improvement in specific complex indices of d2 and BCIS neurocognitive tests, as well as the improvement in the clinical state (PANSS). No correlation was observed between the level of NPS and the results of AEPs and QEEG measurements. Conclusions: A decreased level of NPS is possibly related to the improvement in metacognition and social cognition domains, as well as to clinical improvement during the rehabilitation therapy of patients with schizophrenia.
Collapse
Affiliation(s)
| | - Renata Markiewicz
- Department of Neurology, Neurological and Psychiatric Nursing, Medical University of Lublin, 20-093 Lublin, Poland
| | - Beata Dobrowolska
- Department of Holistic Care and Management in Nursing, Medical University of Lublin, 20-081 Lublin, Poland
| | - Mansur Rahnama
- Department of Oral Surgery, Medical University of Lublin, 20-093 Lublin, Poland
| | - Bartosz Łoza
- Department of Psychiatry, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
11
|
Yan QY, Lv JL, Shen XY, Ou-Yang XN, Yang JZ, Nie RF, Lu J, Huang YJ, Wang JY, Shen X. Patchouli alcohol as a selective estrogen receptor β agonist ameliorates AD-like pathology of APP/PS1 model mice. Acta Pharmacol Sin 2022; 43:2226-2241. [PMID: 35091686 PMCID: PMC9433381 DOI: 10.1038/s41401-021-00857-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022]
Abstract
Clinical evidence shows that postmenpausal women are almost twice as likely to develop Alzheimer's disease (AD) as men of the same age, and estrogen is closely related to the occurrence of AD. Estrogen receptor (ER) α is mainly expressed in the mammary gland and other reproductive organs like uterus while ERβ is largely distributed in the hippocampus and cardiovascular system, suggesting that ERβ selective agonist is a valuable drug against neurodegenerative diseases with low tendency in inducing cancers of breast and other reproductive organs. In this study we identified a natural product patchouli alcohol (PTA) as a selective ERβ agonist which improved the cognitive defects in female APP/PS1 mice, and explore the underlying mechanisms. Six-month-old female APP/PS1 mice were administered PTA (20, 40 mg · kg-1 · d-1, i.g.) for 90 days. We first demonstrated that PTA bound to ERβ with a dissociation constant (KD) of 288.9 ± 35.14 nM in microscale thermophoresis. Then we showed that PTA administration dose-dependently ameliorated cognitive defects evaluated in Morris water maze and Y-maze testes. Furthermore, PTA administration reduced amyloid plaque deposition in the hippocampus by promoting microglial phagocytosis; PTA administration improved synaptic integrity through enhancing BDNF/TrkB/CREB signaling, ameliorated oxidative stress by Catalase level, and regulated Bcl-2 family proteins in the hippocampus. The therapeutic effects of PTA were also observed in vitro: PTA (5, 10, 20 μM) dose-dependently increased phagocytosis of o-FAM-Aβ42 in primary microglia and BV2 cells through enhancing ERβ/TLR4 signaling; PTA treatment ameliorated o-Aβ25-35-induced reduction of synapse-related proteins VAMP2 and PSD95 in primary neurons through enhancing ERβ/BDNF/TrkB/CREB pathways; PTA treatment alleviated o-Aβ25-35-induced oxidative stress in primary neurons through targeting ERβ and increasing Catalase expression. Together, this study has addressed the efficacy of selective ERβ agonist in the amelioration of AD and highlighted the potential of PTA as a drug lead compound against the disease.
Collapse
Affiliation(s)
- Qiu-Ying Yan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian-Lu Lv
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xing-Yi Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xing-Nan Ou-Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Juan-Zhen Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui-Fang Nie
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu-Jie Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Ying Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
12
|
Relationship of Neuropeptide S with Clinical and Metabolic Parameters of Patients during Rehabilitation Therapy for Schizophrenia. Brain Sci 2022; 12:768. [PMID: 35741653 PMCID: PMC9221542 DOI: 10.3390/brainsci12060768&set/a 869781119+878628306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Neuropeptide S (NPS) is a factor associated with the central regulation of body weight, stress, anxiety, learning, memory consolidation, wakefulness-sleep cycle, and anti-inflammatory and neuroplastic effects. Its stress-reducing, anti-anxiety, arousal without anxiety, and pro-cognitive effects represent an interesting option for the treatment of neuropsychiatric disorders. The purpose of the study was to examine the potential associations of NPS levels in the blood with clinical and metabolic parameters during the rehabilitation therapy of patients with schizophrenia. Thirty-three male subjects diagnosed with schizophrenia were randomly divided into two groups. The rehabilitation group (REH, N16) consisted of patients who were subjected to structured, 3-month intensive rehabilitation therapy, and the control group (CON, N17) consisted of patients who were subjected to a standard support mechanism. Both groups continued their pharmacological treatment as usual. The NPS concentration, as well as clinical and metabolic parameters, were compared in both groups. Additionally, a group of healthy (H) males (N15) was tested for NPS reference scores. To look for the specificity and selectivity of the NPS relationship with clinical results, various factor models of the positive and negative syndrome scale (PANSS) were analyzed, including the original PANSS 2/3 model, its modified four-factor version, the male-specific four-factor model, and two five-factorial models validated in large groups in clinical and multi-ethnic studies. Results and conclusions: (1) Structured rehabilitation therapy, compared to unstructured supportive therapy, significantly reduced the level of schizophrenia disorders defined by various factor models derived from PANSS. (2) The clinical improvement within the 3-month rehabilitation therapy course was correlated with a significant decrease in neuropeptide S (NPS) serum level. (3) The excitement/Hostility (E/H) factor, which included schizophrenic symptoms of the psychotic disorganization, was specific and selective for the reduction in serum NPS, which was stable across all analyzed factor models. (4) The long-term relationship between serum NPS and clinical factors was not accompanied by basic metabolic parameters.
Collapse
|
13
|
Relationship of Neuropeptide S with Clinical and Metabolic Parameters of Patients during Rehabilitation Therapy for Schizophrenia. Brain Sci 2022. [DOI: 10.3390/brainsci12060768
expr 958893762 + 814326274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Neuropeptide S (NPS) is a factor associated with the central regulation of body weight, stress, anxiety, learning, memory consolidation, wakefulness–sleep cycle, and anti-inflammatory and neuroplastic effects. Its stress-reducing, anti-anxiety, arousal without anxiety, and pro-cognitive effects represent an interesting option for the treatment of neuropsychiatric disorders. The purpose of the study was to examine the potential associations of NPS levels in the blood with clinical and metabolic parameters during the rehabilitation therapy of patients with schizophrenia. Thirty-three male subjects diagnosed with schizophrenia were randomly divided into two groups. The rehabilitation group (REH, N16) consisted of patients who were subjected to structured, 3-month intensive rehabilitation therapy, and the control group (CON, N17) consisted of patients who were subjected to a standard support mechanism. Both groups continued their pharmacological treatment as usual. The NPS concentration, as well as clinical and metabolic parameters, were compared in both groups. Additionally, a group of healthy (H) males (N15) was tested for NPS reference scores. To look for the specificity and selectivity of the NPS relationship with clinical results, various factor models of the positive and negative syndrome scale (PANSS) were analyzed, including the original PANSS 2/3 model, its modified four-factor version, the male-specific four-factor model, and two five-factorial models validated in large groups in clinical and multi-ethnic studies. Results and conclusions: (1) Structured rehabilitation therapy, compared to unstructured supportive therapy, significantly reduced the level of schizophrenia disorders defined by various factor models derived from PANSS. (2) The clinical improvement within the 3-month rehabilitation therapy course was correlated with a significant decrease in neuropeptide S (NPS) serum level. (3) The excitement/Hostility (E/H) factor, which included schizophrenic symptoms of the psychotic disorganization, was specific and selective for the reduction in serum NPS, which was stable across all analyzed factor models. (4) The long-term relationship between serum NPS and clinical factors was not accompanied by basic metabolic parameters.
Collapse
|
14
|
Relationship of Neuropeptide S with Clinical and Metabolic Parameters of Patients during Rehabilitation Therapy for Schizophrenia. Brain Sci 2022; 12:brainsci12060768. [PMID: 35741653 PMCID: PMC9221542 DOI: 10.3390/brainsci12060768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 01/09/2023] Open
Abstract
Neuropeptide S (NPS) is a factor associated with the central regulation of body weight, stress, anxiety, learning, memory consolidation, wakefulness–sleep cycle, and anti-inflammatory and neuroplastic effects. Its stress-reducing, anti-anxiety, arousal without anxiety, and pro-cognitive effects represent an interesting option for the treatment of neuropsychiatric disorders. The purpose of the study was to examine the potential associations of NPS levels in the blood with clinical and metabolic parameters during the rehabilitation therapy of patients with schizophrenia. Thirty-three male subjects diagnosed with schizophrenia were randomly divided into two groups. The rehabilitation group (REH, N16) consisted of patients who were subjected to structured, 3-month intensive rehabilitation therapy, and the control group (CON, N17) consisted of patients who were subjected to a standard support mechanism. Both groups continued their pharmacological treatment as usual. The NPS concentration, as well as clinical and metabolic parameters, were compared in both groups. Additionally, a group of healthy (H) males (N15) was tested for NPS reference scores. To look for the specificity and selectivity of the NPS relationship with clinical results, various factor models of the positive and negative syndrome scale (PANSS) were analyzed, including the original PANSS 2/3 model, its modified four-factor version, the male-specific four-factor model, and two five-factorial models validated in large groups in clinical and multi-ethnic studies. Results and conclusions: (1) Structured rehabilitation therapy, compared to unstructured supportive therapy, significantly reduced the level of schizophrenia disorders defined by various factor models derived from PANSS. (2) The clinical improvement within the 3-month rehabilitation therapy course was correlated with a significant decrease in neuropeptide S (NPS) serum level. (3) The excitement/Hostility (E/H) factor, which included schizophrenic symptoms of the psychotic disorganization, was specific and selective for the reduction in serum NPS, which was stable across all analyzed factor models. (4) The long-term relationship between serum NPS and clinical factors was not accompanied by basic metabolic parameters.
Collapse
|
15
|
Shao YF, Wang C, Rao XP, Wang HD, Ren YL, Li J, Dong CY, Xie JF, Yang XW, Xu FQ, Hou YP. Neuropeptide S Attenuates the Alarm Pheromone-Evoked Defensive and Risk Assessment Behaviors Through Activation of Cognate Receptor-Expressing Neurons in the Posterior Medial Amygdala. Front Mol Neurosci 2022; 14:752516. [PMID: 35002616 PMCID: PMC8739225 DOI: 10.3389/fnmol.2021.752516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/09/2021] [Indexed: 11/24/2022] Open
Abstract
Neuropeptide S (NPS) acts by activating its cognate receptor (NPSR). High level expression of NPSR in the posterior medial amygdala suggests that NPS-NPSR system should be involved in regulation of social behaviors induced by social pheromones. The present study was undertaken to investigate the effects of central administration of NPS or with NPSR antagonist on the alarm pheromone (AP)-evoked defensive and risk assessment behaviors in mice. Furthermore, H129-H8, a novel high-brightness anterograde multiple trans-synaptic virus, c-Fos and NPSR immunostaining were employed to reveal the involved neurocircuits and targets of NPS action. The mice exposed to AP displayed an enhancement in defensive and risk assessment behaviors. NPS (0.1–1 nmol) intracerebroventricular (i.c.v.) injection significantly attenuated the AP-evoked defensive and risk assessment behaviors. NPSR antagonist [D-Val5]NPS at the dose of 40 nmol completely blocked the effect of 0.5 nmol of NPS which showed the best effective among dose range. The H129-H8-labeled neurons were observed in the bilateral posterodorsal medial amygdala (MePD) and posteroventral medial amygdala (MePV) 72 h after the virus injection into the unilateral olfactory bulb (OB), suggesting that the MePD and MePV receive olfactory information inputs from the OB. The percentage of H129-H8-labeled neurons that also express NPSR were 90.27 ± 3.56% and 91.67 ± 2.46% in the MePD and MePV, respectively. NPS (0.5 nmol, i.c.v.) remarkably increased the number of Fos immunoreactive (-ir) neurons in the MePD and MePV, and the majority of NPS-induced Fos-ir neurons also expressed NPSR. The behavior characteristic of NPS or with [D-Val5]NPS can be better replicated in MePD/MePV local injection within lower dose. The present findings demonstrated that NPS, via selective activation of the neurons bearing NPSR in the posterior medial amygdala, attenuates the AP-evoked defensive and risk assessment behaviors in mice.
Collapse
Affiliation(s)
- Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Lab of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| | - Can Wang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Ping Rao
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Hua-Dong Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Yan-Li Ren
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing Li
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chao-Yu Dong
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jun-Fan Xie
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xing-Wen Yang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fu-Qiang Xu
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Lab of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
16
|
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
|
17
|
Bhat US, Shahi N, Surendran S, Babu K. Neuropeptides and Behaviors: How Small Peptides Regulate Nervous System Function and Behavioral Outputs. Front Mol Neurosci 2021; 14:786471. [PMID: 34924955 PMCID: PMC8674661 DOI: 10.3389/fnmol.2021.786471] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
One of the reasons that most multicellular animals survive and thrive is because of the adaptable and plastic nature of their nervous systems. For an organism to survive, it is essential for the animal to respond and adapt to environmental changes. This is achieved by sensing external cues and translating them into behaviors through changes in synaptic activity. The nervous system plays a crucial role in constantly evaluating environmental cues and allowing for behavioral plasticity in the organism. Multiple neurotransmitters and neuropeptides have been implicated as key players for integrating sensory information to produce the desired output. Because of its simple nervous system and well-established neuronal connectome, C. elegans acts as an excellent model to understand the mechanisms underlying behavioral plasticity. Here, we critically review how neuropeptides modulate a wide range of behaviors by allowing for changes in neuronal and synaptic signaling. This review will have a specific focus on feeding, mating, sleep, addiction, learning and locomotory behaviors in C. elegans. With a view to understand evolutionary relationships, we explore the functions and associated pathophysiology of C. elegans neuropeptides that are conserved across different phyla. Further, we discuss the mechanisms of neuropeptidergic signaling and how these signals are regulated in different behaviors. Finally, we attempt to provide insight into developing potential therapeutics for neuropeptide-related disorders.
Collapse
Affiliation(s)
- Umer Saleem Bhat
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Navneet Shahi
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Siju Surendran
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
18
|
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
|
19
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
20
|
Kushikata T, Hirota K, Saito J, Takekawa D. Roles of Neuropeptide S in Anesthesia, Analgesia, and Sleep. Pharmaceuticals (Basel) 2021; 14:ph14050483. [PMID: 34069327 PMCID: PMC8158725 DOI: 10.3390/ph14050483] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Neuropeptide S (NPS) is an endogenous peptide that regulates various physiological functions, such as immune functions, anxiety-like behaviors, learning and memory, the sleep–wake rhythm, ingestion, energy balance, and drug addiction. These processes include the NPS receptor (NPSR1). The NPS–NPSR1 system is also significantly associated with the onset of disease, as well as these physiologic functions. For example, NPS is involved in bronchial asthma, anxiety and awakening disorders, and rheumatoid arthritis. In this review, among the various functions, we focus on the role of NPS in anesthesia-induced loss of consciousness; analgesia, mainly by anesthesia; and sleep–wakefulness. Progress in the field regarding the functions of endogenous peptides in the brain, including NPS, suggests that these three domains share common mechanisms. Further NPS research will help to elucidate in detail how these three domains interact with each other in their functions, and may contribute to improving the quality of medical care.
Collapse
Affiliation(s)
- Tetsuya Kushikata
- Department of Anesthesiology, Graduate School of Medicine, Hirosaki University, Zaifu 5, Hirosaki 0368562, Japan; (K.H.); (J.S.)
- Correspondence:
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Graduate School of Medicine, Hirosaki University, Zaifu 5, Hirosaki 0368562, Japan; (K.H.); (J.S.)
| | - Junichi Saito
- Department of Anesthesiology, Graduate School of Medicine, Hirosaki University, Zaifu 5, Hirosaki 0368562, Japan; (K.H.); (J.S.)
| | - Daiki Takekawa
- Department of Anesthesia, Hirosaki University Hospital, Honcho 53, Hirosaki 0368563, Japan;
| |
Collapse
|
21
|
Batran RZ, Gugnani KS, Maher TJ, Khedr MA. New quinolone derivatives as neuropeptide S receptor antagonists: Design, synthesis, homology modeling, dynamic simulations and modulation of Gq/Gs signaling pathways. Bioorg Chem 2021; 111:104817. [PMID: 33848721 DOI: 10.1016/j.bioorg.2021.104817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/16/2021] [Accepted: 03/06/2021] [Indexed: 01/04/2023]
Abstract
In a search for new neuropeptide S receptor antagonists, we have described a new series of quinolone-pyranopyrimidine hybrid derivatives aiming to modify the inhibitory characters towards NPSR to develop new therapeutic strategies against anxiety, addiction and food disorders. We identified six potent antagonists 3, 4b, 6, 8, 9 and 10 which counteracted the stimulatory effect of NPS at both Gq and Gs pathways, at low micromolar concentrations, through modulation of Ca2+ and cAMP signaling, respectively. Molecular docking predicted the orientation mode of the top active compounds; 10 and 4b with ΔG value of -23.94 and -23.87 kcal/mol, respectively that is considered good when compared to that of the reference compound ML154 (ΔG = -25.75 kcal/mol) . Molecular dynamic simulations confirmed the stability of binding of compound 10 to the homology model of NPSR as it reached the equilibrium after 4 ns at RMSD of 1.00 Å while ML154 was faster to achieve the equilibrium after 2 ns at RMSD of 1.00 Å.
Collapse
Affiliation(s)
- Rasha Z Batran
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El Bohouth St., Dokki, Giza, P.O. Box 12622, Egypt.
| | - Kuljeet S Gugnani
- Department of Pharmaceutical Sciences, MCPHS University, Boston, MA, USA
| | - Timothy J Maher
- Department of Pharmaceutical Sciences, MCPHS University, Boston, MA, USA
| | - Mohammed A Khedr
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, P.O. Box 11795, Egypt
| |
Collapse
|
22
|
Zhou Y, Zhu F, Liu Y, Zheng M, Wang Y, Zhang D, Anraku Y, Zou Y, Li J, Wu H, Pang X, Tao W, Shimoni O, Bush AI, Xue X, Shi B. Blood-brain barrier-penetrating siRNA nanomedicine for Alzheimer's disease therapy. SCIENCE ADVANCES 2020; 6:6/41/eabc7031. [PMID: 33036977 PMCID: PMC7546706 DOI: 10.1126/sciadv.abc7031] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/26/2020] [Indexed: 05/21/2023]
Abstract
Toxic aggregated amyloid-β accumulation is a key pathogenic event in Alzheimer's disease (AD), which derives from amyloid precursor protein (APP) through sequential cleavage by BACE1 (β-site APP cleavage enzyme 1) and γ-secretase. Small interfering RNAs (siRNAs) show great promise for AD therapy by specific silencing of BACE1. However, lack of effective siRNA brain delivery approaches limits this strategy. Here, we developed a glycosylated "triple-interaction" stabilized polymeric siRNA nanomedicine (Gal-NP@siRNA) to target BACE1 in APP/PS1 transgenic AD mouse model. Gal-NP@siRNA exhibits superior blood stability and can efficiently penetrate the blood-brain barrier (BBB) via glycemia-controlled glucose transporter-1 (Glut1)-mediated transport, thereby ensuring that siRNAs decrease BACE1 expression and modify relative pathways. Noticeably, Gal-NP@siBACE1 administration restored the deterioration of cognitive capacity in AD mice without notable side effects. This "Trojan horse" strategy supports the utility of RNA interference therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yutong Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Feiyan Zhu
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yang Liu
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| | - Yibin Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yan Zou
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Jia Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, China
| | - Haigang Wu
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Xiaobin Pang
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Olga Shimoni
- Institute for Biomedical Materials & Devices (IBMD), School of Mathematical and Physical Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China.
| | - Bingyang Shi
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
23
|
Abdul Manap AS, Madhavan P, Vijayabalan S, Chia A, Fukui K. Explicating anti-amyloidogenic role of curcumin and piperine via amyloid beta (A β) explicit pathway: recovery and reversal paradigm effects. PeerJ 2020; 8:e10003. [PMID: 33062432 PMCID: PMC7532763 DOI: 10.7717/peerj.10003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/30/2020] [Indexed: 12/28/2022] Open
Abstract
Previously, we reported the synergistic effects of curcumin and piperine in cell cultures as potential anti-cholinesterase and anti-amyloidogenic agents. Due to limited findings on the enrolment of these compounds on epigenetic events in AD, we aimed at elucidating the expression profiles of Aβ42-induced SH-SY5Y cells using microarray profiling. In this study, an optimized concentration of 35 µM of curcumin and piperine in combination was used to treat Aβ42 fibril and high-throughput microarray profiling was performed on the extracted RNA. This was then compared to curcumin and piperine used singularly at 49.11 µM and 25 µM, respectively. Our results demonstrated that in the curcumin treated group, from the top 10 upregulated and top 10 downregulated significantly differentially expressed genes (p < 0.05; fold change ≥ 2 or ≤ -2), there were five upregulated and three downregulated genes involved in the amyloidogenic pathway. While from top 10 upregulated and top 10 downregulated significantly differentially expressed genes (p < 0.05; fold change ≥ 2 or ≤ - 2) in the piperine treated group, there were four upregulated and three downregulated genes involved in the same pathway, whereas there were five upregulated and two downregulated genes involved (p < 0.05; fold change ≥ 2 or ≤ - 2) in the curcumin-piperine combined group. Four genes namely GABARAPL1, CTSB, RAB5 and AK5 were expressed significantly in all groups. Other genes such as ITPR1, GSK3B, PPP3CC, ERN1, APH1A, CYCS and CALM2 were novel putative genes that are involved in the pathogenesis of AD. We revealed that curcumin and piperine have displayed their actions against Aβ via the modulation of various mechanistic pathways. Alterations in expression profiles of genes in the neuronal cell model may explain Aβ pathology post-treatment and provide new insights for remedial approaches of a combined treatment using curcumin and piperine.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Shantini Vijayabalan
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Adeline Chia
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Koji Fukui
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| |
Collapse
|
24
|
Mechanisms of Electroacupuncture on Alzheimer’s Disease: A Review of Animal Studies. Chin J Integr Med 2020; 26:473-480. [DOI: 10.1007/s11655-020-3092-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2019] [Indexed: 12/22/2022]
|
25
|
Neugebauer V, Mazzitelli M, Cragg B, Ji G, Navratilova E, Porreca F. Amygdala, neuropeptides, and chronic pain-related affective behaviors. Neuropharmacology 2020; 170:108052. [PMID: 32188569 DOI: 10.1016/j.neuropharm.2020.108052] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Neuropeptides play important modulatory roles throughout the nervous system, functioning as direct effectors or as interacting partners with other neuropeptide and neurotransmitter systems. Limbic brain areas involved in learning, memory and emotions are particularly rich in neuropeptides. This review will focus on the amygdala, a limbic region that plays a key role in emotional-affective behaviors and pain modulation. The amygdala is comprised of different nuclei; the basolateral (BLA) and central (CeA) nuclei and in between, the intercalated cells (ITC), have been linked to pain-related functions. A wide range of neuropeptides are found in the amygdala, particularly in the CeA, but this review will discuss those neuropeptides that have been explored for their role in pain modulation. Calcitonin gene-related peptide (CGRP) is a key peptide in the afferent nociceptive pathway from the parabrachial area and mediates excitatory drive of CeA neurons. CeA neurons containing corticotropin releasing factor (CRF) and/or somatostatin (SOM) are a source of long-range projections and serve major output functions, but CRF also acts locally to excite neurons in the CeA and BLA. Neuropeptide S (NPS) is associated with inhibitory ITC neurons that gate amygdala output. Oxytocin and vasopressin exert opposite (inhibitory and excitatory, respectively) effects on amygdala output. The opioid system of mu, delta and kappa receptors (MOR, DOR, KOR) and their peptide ligands (β-endorphin, enkephalin, dynorphin) have complex and partially opposing effects on amygdala function. Neuropeptides therefore serve as valuable targets to regulate amygdala function in pain conditions. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Bryce Cragg
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
26
|
Zhou F, Yan XD, Wang C, He YX, Li YY, Zhang J, Wang ZJ, Cai HY, Qi JS, Wu MN. Suvorexant ameliorates cognitive impairments and pathology in APP/PS1 transgenic mice. Neurobiol Aging 2020; 91:66-75. [PMID: 32224066 DOI: 10.1016/j.neurobiolaging.2020.02.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/28/2020] [Accepted: 02/22/2020] [Indexed: 02/06/2023]
Abstract
Cognitive impairments and circadian rhythm disorders are the main clinical manifestations of Alzheimer's disease (AD). Orexin has been reported as abnormally elevated in the cerebrospinal fluid of AD patients, accompanied with cognitive impairments. Our recent research revealed that suvorexant, a dual orexin receptor antagonist, could improve behavioral circadian rhythm disorders in 9-month-old APP/PS1 mice. Here we further observed whether suvorexant could ameliorate the cognitive decline in APP/PS1 mice by using behavioral tests, and investigated the possible mechanisms by in vivo electrophysiological recording, western blot, and immunochemistry. The results showed that suvorexant treatment effectively ameliorated the cognitive impairments, alleviated in vivo hippocampal long-term potentiation suppression, restored the circadian phosphorylated CREB expression in the hippocampus, and reduced amyloid-β protein deposition in the hippocampus and cortex in APP/PS1 mice. These results indicate that the neuroprotective effects of suvorexant against AD are involved in the reduction of amyloid-β plaques, improvement of synaptic plasticity, and circadian expression of phosphorylated CREB, suggesting that suvorexant could be beneficial to the prevention and treatment of AD.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xu-Dong Yan
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Chun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ye-Xin He
- Department of Radiology, Shanxi Provincial People's Hospital, Taiyuan, People's Republic of China
| | - Yi-Ying Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jun Zhang
- Functional Laboratory Center, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|