1
|
Han M, Dong Y, Wang S, Huang X, Bai C, Gai Z. Regulation of gut microbiota and serum neurotransmitters in mice by Streptococcus thermophilus GA8- and Lacticaseibacillus rhamnosus HAO9-fermented milk containing high levels of gamma-aminobutyric acid. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8050-8058. [PMID: 38828862 DOI: 10.1002/jsfa.13634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/26/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) is an important neurotransmitter in the human body, with several negative emotions reported as being associated with GABA dysregulation. This study investigates the safety and modulatory effects of GABA-enriched milk, fermented by Streptococcus thermophilus GA8 and Lacticasebacillus rhamnosus HAO9, on the gut microbiota and neurotransmitter profiles in mice. RESULTS Through rigorous culturing and fermentation processes, we achieved consistent GABA production in milk, with concentrations reaching 4.6 and 8.5 g L-1 for GA8-fermented and co-fermented milk, respectively, after 48 h. Using SPF male C57BL/6J mice, we administered either mono-culture or combined-culture milk treatments and monitored physiological impacts. The treatments did not affect mouse body weight but induced significant changes in gut microbiota composition. Beta diversity analysis revealed distinct microbial profiles between treatment groups, highlighting fermentation-specific microbial shifts, such as an increase in Verrucomicrobia for the GA8 group and a modulation in Saccharibacteria_genera_incertae_sedis for the GA8 + HAO9 group. Serum neurotransmitter levels were elevated in both treatment groups, with significant increases in l-glutamine, l-tryptophan and, notably, serotonin hydrochloride in the GA8 + HAO9 group. Correlation analysis identified a positive association between specific bacterial genera and neurotransmitter levels, suggesting a probiotic effect on neuroactive substances. CONCLUSION These findings suggest that fermented milk has potential as a probiotic supplement for mood improvement and stress relief, highlighting its role in modulating the gut-brain axis. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Mei Han
- Department of Food Science, Shanghai Business School, Shanghai, China
| | - Yao Dong
- Department of Research and Development, Wecare Probiotics Co., Ltd, Suzhou, China
| | - Shuo Wang
- APC Microbiome Ireland, Cork, Ireland
| | | | - Chen Bai
- Department of Food Science, Shanghai Business School, Shanghai, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd, Suzhou, China
| |
Collapse
|
2
|
Peng F, Lu J, Su K, Liu X, Luo H, He B, Wang C, Zhang X, An F, Lv D, Luo Y, Su Q, Jiang T, Deng Z, He B, Xu L, Guo T, Xiang J, Gu C, Wang L, Xu G, Xu Y, Li M, Kelley KW, Cui B, Liu Q. Oncogenic fatty acid oxidation senses circadian disruption in sleep-deficiency-enhanced tumorigenesis. Cell Metab 2024; 36:1598-1618.e11. [PMID: 38772364 DOI: 10.1016/j.cmet.2024.04.018] [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: 11/02/2022] [Revised: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Circadian disruption predicts poor cancer prognosis, yet how circadian disruption is sensed in sleep-deficiency (SD)-enhanced tumorigenesis remains obscure. Here, we show fatty acid oxidation (FAO) as a circadian sensor relaying from clock disruption to oncogenic metabolic signal in SD-enhanced lung tumorigenesis. Both unbiased transcriptomic and metabolomic analyses reveal that FAO senses SD-induced circadian disruption, as illustrated by continuously increased palmitoyl-coenzyme A (PA-CoA) catalyzed by long-chain fatty acyl-CoA synthetase 1 (ACSL1). Mechanistically, SD-dysregulated CLOCK hypertransactivates ACSL1 to produce PA-CoA, which facilitates CLOCK-Cys194 S-palmitoylation in a ZDHHC5-dependent manner. This positive transcription-palmitoylation feedback loop prevents ubiquitin-proteasomal degradation of CLOCK, causing FAO-sensed circadian disruption to maintain SD-enhanced cancer stemness. Intriguingly, timed β-endorphin resets rhythmic Clock and Acsl1 expression to alleviate SD-enhanced tumorigenesis. Sleep quality and serum β-endorphin are negatively associated with both cancer development and CLOCK/ACSL1 expression in patients with cancer, suggesting dawn-supplemented β-endorphin as a potential chronotherapeutic strategy for SD-related cancer.
Collapse
Affiliation(s)
- Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinyu Liu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Fan An
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yuanyuan Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Qitong Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Tonghui Jiang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Ziqian Deng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lingzhi Xu
- Department of Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jin Xiang
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ling Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guowang Xu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Ying Xu
- Cambridge-Soochow University Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Mindian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine and Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Leem KH, Kim S, Kim HW, Park HJ. Downregulation of microRNA-330-5p induces manic-like behaviors in REM sleep-deprived rats by enhancing tyrosine hydroxylase expression. CNS Neurosci Ther 2023; 29:1525-1536. [PMID: 36794530 PMCID: PMC10173715 DOI: 10.1111/cns.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/13/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
AIM In our pilot study, we found an increase in tyrosine hydroxylase (Th) mRNA expression in the prefrontal cortex of 72-h REM sleep-deprived (SD) rats, a mania model. Additionally, the expression levels of miR-325-3p, miR-326-3p, and miR-330-5p, the predicted target miRNAs on TH, were significantly decreased. Based on these results, in this study, we investigated whether miRNA-325-3p, miR-326-3p, and miR-330-5p modulate TH and manic-like behaviors in SD rats. METHODS Manic-like behaviors were assessed using the open field test (OFT) and elevated plus-maze (EPM) test. The direct binding activity of miRNAs to the 3'-untranslated region (3'-UTR) of the Th gene was measured in HEK-293 cells using a luciferase reporter system. We also examined mRNA and protein expression of TH after intracerebroventricular (ICV) injection of miR-330-5p agomir to SD rats, along with manic-like behaviors. RESULTS We observed an upregulation in mRNA and protein expression of TH and downregulation in miRNA-325-3p, miR-326-3p, and miR-330-5p expressions in the prefrontal cortex of SD rats, together with increased manic-like behaviors. The luciferase reporter assay showed that miR-330-5p could repress TH expression through direct binding to its target site in the 3'-UTR of Th, whereas miR-326-3p and miR-330-5p could not. In addition, ICV injection of miR-330-5p agomir alleviated the increase in TH expression in the prefrontal cortex of SD rats and manic-like behaviors. CONCLUSIONS TH expression regulation through miR-330-5p may be implicated in the pathophysiology of mania in SD rats.
Collapse
Affiliation(s)
- Kang Hyun Leem
- Department of Herbology, College of Korean MedicineSemyung UniversityJecheonKorea
| | - Sanga Kim
- Department of Pharmacology, School of MedicineKyung Hee UniversitySeoulKorea
| | - Hee Won Kim
- Department of Medical Engineering, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Hae Jeong Park
- Department of Pharmacology, School of MedicineKyung Hee UniversitySeoulKorea
| |
Collapse
|
4
|
Zhang L, Zhang Y, Jiang X, Mao L, Xia Y, Fan Y, Li N, Jiang Z, Qin X, Jiang Y, Liu G, Qiu F, Zhang J, Zou Z, Chen C. Disruption of the lung-gut-brain axis is responsible for cortex damage induced by pulmonary exposure to zinc oxide nanoparticles. Toxicology 2023; 485:153390. [PMID: 36535435 DOI: 10.1016/j.tox.2022.153390] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
Increasing evidence shows that gut microbiota is important for host health in response to metal nanomaterials exposure. However, the effect of gut microbiota on the cortex damage caused by pulmonary exposure to zinc oxide nanoparticles (ZnONPs) remains mainly unknown. In this study, a total of 48 adult C57BL/6J mice were intratracheally instilled with 0.6 mg/kg ZnONPs in the presence or absence of antibiotics (ABX) treatment. Besides, 24 mice were treated with or without fecal microbiota transplantation (FMT) after the intraperitoneal administration of ABX. Our results demonstrated for the first time that dysbiosis induced by ABX treatment significantly aggravated cortex damage induced by pulmonary exposure to ZnONPs. Such damage might highly occur through the induction of oxidative stress, manifested by the enhancement of antioxidative enzymes and products of lipid peroxidation. However, ferroptosis was not involved in this process. Interestingly, our data revealed that ABX treatment exacerbated the alterations of gut-brain peptides (including Sst, Sstr2, and Htr4) induced by ZnONPs in both gut and cortex tissues. Moreover, fecal microbiota transplantation (FMT) was able to alleviate cerebral cortex damage, oxidative stress, and alterations of gut-brain peptides induced by pulmonary exposure to ZnONPs. The results together indicate that pulmonary exposure to ZnONPs causes cerebral cortex damage possibly via the disruption of the lung-gut-brain axis. These findings not only propose valuable insights into the mechanism of ZnONPs neurotoxicity but also provide a potential therapeutic method against brain disorders induced by pulmonary exposure to ZnONPs. AVAILABILITY OF DATA AND MATERIALS: The datasets used and/or analyzed during the current study are available from the The corresponding author on reasonable request.
Collapse
Affiliation(s)
- Lingbing Zhang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yandan Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuejun Jiang
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China; Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Yinzhen Fan
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Na Li
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Ziqi Jiang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yu Jiang
- Department of Respiratory Medicine, The University‑Town Affiliated Hospital of Chongqing Medical University, Chongqing 401331, PR China
| | - Gang Liu
- Department of Emergency, The University‑Town Affiliated Hospital of Chongqing Medical University, Chongqing 401331, PR China
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Chengzhi Chen
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China; Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
5
|
Shabani L, Abbasi M, Azarnew Z, Amani AM, Vaez A. Neuro-nanotechnology: diagnostic and therapeutic nano-based strategies in applied neuroscience. Biomed Eng Online 2023; 22:1. [PMID: 36593487 PMCID: PMC9809121 DOI: 10.1186/s12938-022-01062-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Artificial, de-novo manufactured materials (with controlled nano-sized characteristics) have been progressively used by neuroscientists during the last several decades. The introduction of novel implantable bioelectronics interfaces that are better suited to their biological targets is one example of an innovation that has emerged as a result of advanced nanostructures and implantable bioelectronics interfaces, which has increased the potential of prostheses and neural interfaces. The unique physical-chemical properties of nanoparticles have also facilitated the development of novel imaging instruments for advanced laboratory systems, as well as intelligently manufactured scaffolds and microelectrodes and other technologies designed to increase our understanding of neural tissue processes. The incorporation of nanotechnology into physiology and cell biology enables the tailoring of molecular interactions. This involves unique interactions with neurons and glial cells in neuroscience. Technology solutions intended to effectively interact with neuronal cells, improved molecular-based diagnostic techniques, biomaterials and hybridized compounds utilized for neural regeneration, neuroprotection, and targeted delivery of medicines as well as small chemicals across the blood-brain barrier are all purposes of the present article.
Collapse
Affiliation(s)
- Leili Shabani
- grid.412571.40000 0000 8819 4698Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- grid.412571.40000 0000 8819 4698Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeynab Azarnew
- grid.412571.40000 0000 8819 4698Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- grid.412571.40000 0000 8819 4698Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- grid.412571.40000 0000 8819 4698Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Turan I, Sayan Ozacmak H, Ozacmak VH, Ergenc M, Bayraktaroğlu T. The effects of glucagon-like peptide 1 receptor agonist (exenatide) on memory impairment, and anxiety- and depression-like behavior induced by REM sleep deprivation. Brain Res Bull 2021; 174:194-202. [PMID: 34146656 DOI: 10.1016/j.brainresbull.2021.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
Previous investigations have shown that REM sleep deprivation impairs the hippocampus-dependent memory, long-term potentiation and causing mood changes. The aim of the present study was to explore the effects of exenatide on memory performance, anxiety- and depression like behavior, oxidative stress markers, and synaptic protein levels in REM sleep deprived rats. A total of 40 male Wistar rats were randomly divided to control, exenatide-treated control, sleep deprivation (SD), wide platform (WP) and exenatide-treated SD groups. During experiments, exenatide treatment (0.5 μg/kg, subcutaneously) was applied daily in a single dose for 9 days. Modified multiple platform method was employed to generate REM sleep deprivation for 72 h. The Morris water maze test was used to assess memory performance. Anxiety- and depression-like behaviors were evaluated by open field test (OFT), elevated plus maze (EPM) forced swimming test (FST), respectively 72 h after REMSD. The levels of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and postsynaptic density proteins 95 (PSD95) were measured in tissues of hippocampus and prefrontal cortex. The content of malondialdehyde (MDA) and reduced glutathione (GSH) were also measured. In the present study, an impairment in memory was observed in SD rats at the 24th hour of SD in compare to those of other groups. REMSD increased depression-like behavior in FST as well as the number of rearing and crossing square in OFT. Anxiety is the most common comorbid condition with depressive disorders. Contents of CaMKII and PSD95 decreased in hippocampus of SD rats. Exenatide treatment improved the impaired memory of SD rats and increased CaMKII content in hippocampus There was no difference in MDA and GSH levels among groups. Exenatide treatment also diminished locomotor activity in OFT. In conclusion, treatment with exenatide, at least in part, prevented from these cognitive and behavioral changes possibly through normalizing CaMKII levels in the hippocampus.
Collapse
Affiliation(s)
- Inci Turan
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Physiology, Zonguldak, Turkey.
| | - Hale Sayan Ozacmak
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Physiology, Zonguldak, Turkey
| | - V Haktan Ozacmak
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Physiology, Zonguldak, Turkey
| | - Meryem Ergenc
- Zonguldak Bulent Ecevit University Faculty of Medicine, Institute of Health Sciences Department of Physiology, Zonguldak, Turkey
| | - Taner Bayraktaroğlu
- Zonguldak Bulent Ecevit Unıversity Faculty of Medicine, Department of Endocrinology, Zonguldak, Turkey
| |
Collapse
|
7
|
Sun Z, Bo Q, Mao Z, Li F, He F, Pao C, Li W, He Y, Ma X, Wang C. Reduced Plasma Dopamine-β-Hydroxylase Activity Is Associated With the Severity of Bipolar Disorder: A Pilot Study. Front Psychiatry 2021; 12:566091. [PMID: 33995135 PMCID: PMC8115127 DOI: 10.3389/fpsyt.2021.566091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Dopamine-β-hydroxylase (DβH) is an enzyme converting dopamine to norepinephrine, a key neurotransmitter in mood disorders, such as major depressive disorder (MDD) and bipolar disorder (BD). Due to overlapping symptomology of unipolar and bipolar depression, the present study attempted to explorer if the plasma DβH activity could discriminate the depressive episodes of BD from MDD. The aim of this study was to compare the plasma DβH activity among MDD patients (n = 104), BD patients (n = 101), and healthy controls (n = 160). Clinical characteristics and cognitive function were assessed using the Young Mania Rating Scale (YMRS), Hamilton Depression Scale (HAM-D), Hamilton Anxiety Scale (HAM-A), Patient Health Questionnaire-9 (PHQ-9), and Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Our data showed a lower plasma DβH activity in patients with BD, not MDD, than that in controls. For the BD patients, the plasma DβH activities were negatively correlated with HAM-D scores and HAM-A scores. However, there was no significant correlation between plasma DβH activity and severity of depressive symptoms in MDD patients. No significant correlation between DβH activities and cognitive assessments neither in BD nor in MDD patients. The present study provides evidence that BD is associated with decreased circulating DβH activity.
Collapse
Affiliation(s)
- Zuoli Sun
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qijing Bo
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhen Mao
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Feng Li
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fan He
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Christine Pao
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wenbiao Li
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yi He
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xin Ma
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders and Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Sharma A, Muresanu DF, Sahib S, Tian ZR, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Bryukhovetskiy I, Manzhulo I, Patnaik R, Wiklund L, Sharma HS. Concussive head injury exacerbates neuropathology of sleep deprivation: Superior neuroprotection by co-administration of TiO 2-nanowired cerebrolysin, alpha-melanocyte-stimulating hormone, and mesenchymal stem cells. PROGRESS IN BRAIN RESEARCH 2020; 258:1-77. [PMID: 33223033 DOI: 10.1016/bs.pbr.2020.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sleep deprivation (SD) is common in military personnel engaged in combat operations leading to brain dysfunction. Military personnel during acute or chronic SD often prone to traumatic brain injury (TBI) indicating the possibility of further exacerbating brain pathology. Several lines of evidence suggest that in both TBI and SD alpha-melanocyte-stimulating hormone (α-MSH) and brain-derived neurotrophic factor (BDNF) levels decreases in plasma and brain. Thus, a possibility exists that exogenous supplement of α-MSH and/or BDNF induces neuroprotection in SD compounded with TBI. In addition, mesenchymal stem cells (MSCs) are very portent in inducing neuroprotection in TBI. We examined the effects of concussive head injury (CHI) in SD on brain pathology. Furthermore, possible neuroprotective effects of α-MSH, MSCs and neurotrophic factors treatment were explored in a rat model of SD and CHI. Rats subjected to 48h SD with CHI exhibited higher leakage of BBB to Evans blue and radioiodine compared to identical SD or CHI alone. Brain pathology was also exacerbated in SD with CHI group as compared to SD or CHI alone together with a significant reduction in α-MSH and BDNF levels in plasma and brain and enhanced level of tumor necrosis factor-alpha (TNF-α). Exogenous administration of α-MSH (250μg/kg) together with MSCs (1×106) and cerebrolysin (a balanced composition of several neurotrophic factors and active peptide fragments) (5mL/kg) significantly induced neuroprotection in SD with CHI. Interestingly, TiO2 nanowired delivery of α-MSH (100μg), MSCs, and cerebrolysin (2.5mL/kg) induced enhanced neuroprotection with higher levels of α-MSH and BDNF and decreased the TNF-α in SD with CHI. These observations are the first to show that TiO2 nanowired administration of α-MSH, MSCs and cerebrolysin induces superior neuroprotection following SD in CHI, not reported earlier. The clinical significance of our findings in light of the current literature is discussed.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
9
|
Antimania-Like Effect of Panax ginseng Regulating the Glutamatergic Neurotransmission in REM-Sleep Deprivation Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3636874. [PMID: 33123570 PMCID: PMC7586145 DOI: 10.1155/2020/3636874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/29/2020] [Accepted: 09/28/2020] [Indexed: 11/17/2022]
Abstract
Previous studies have shown the therapeutic properties of ginseng and ginsenosides on hyperactive and impulsive behaviors in several psychiatric diseases. Herein, we investigated the effect of Panax ginseng Meyer (PG) on hyperactive/impulsive behaviors in a manic-like animal model, sleep deprivation (SD) rats. Male rats were sleep-deprived for 48 h, and PG (200 mg/kg) was administered for 4 days, from 2 days prior to the start of SD to the end date of SD. The elevated plus maze (EPM) test showed that PG alleviated the increased frequency of entries into and spent time within open arms by SD. In order to investigate the molecular mechanism on this effect of PG, we assessed differentially expressed genes (DEGs) in the prefrontal cortex of PG-treated SD rats using RNA sequencing (RNA-seq) and performed gene-enrichment analysis for DEGs. The gene-enrichment analysis showed that PG most prominently affected the glutamatergic synapse pathway. Among the glutamatergic synapse pathway genes, particularly, PG enhanced the expressions of glutamate transporter Slc1a3 and Slc1a2 reduced in SD rats. Moreover, we found that PG could inhibit the SD-induced phosphorylation of the NR2A subunit of the NMDA receptor. These results suggested that PG might have a therapeutic effect against the manic-like behaviors, regulating the glutamatergic neurotransmission.
Collapse
|
10
|
Tai F, Wang C, Deng X, Li R, Guo Z, Quan H, Li S. Treadmill exercise ameliorates chronic REM sleep deprivation-induced anxiety-like behavior and cognitive impairment in C57BL/6J mice. Brain Res Bull 2020; 164:198-207. [PMID: 32877716 DOI: 10.1016/j.brainresbull.2020.08.025] [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: 02/19/2020] [Revised: 07/12/2020] [Accepted: 08/22/2020] [Indexed: 10/23/2022]
Abstract
Various sleep disorders have deleterious effects on mental and cognitive performance. Exercise, as an alternative therapeutic strategy, exerts beneficial impacts on human health. In the present study, we aimed to evaluate the effects of 4 weeks treadmill exercise (4W-TE) on anxiety-like behavior and cognitive performance in mice exposed to 2 months REM sleep deprivation (2M-SD) (20 h per day). Behavioral performance of mice in elevated plus maze test (EPM), open field test (OFT), Y maze test (YM) and Morris water maze test (MWM) was recorded and analyzed 28 h after the last day of sleep deprivation. After behavioral tests, various neurotransmitters including norepinephrine (NE), dopamine (DA), serotonin (5-HT) and γ-aminobutyric acid (GABA) in mouse hippocampus were quantified using high performance liquid chromatography. The hippocampal levels of insulin-like growth factor-1 (IGF-1) and brain derived neurotrophic factor (BDNF) were further detected using ELISA. Behavioral data indicated that 2M-SD exposure induced anxiety-like behaviors and cognitive impairment, as evidenced by the decreased open-arm entries in EPM, reduced central area travels in OFT, declined spontaneous alteration in YM and prolonged escaping latency in MWM. In addition, 2M-SD exposure increased NE and DA, decreased 5-HT and GABA, and reduced IGF-1 and BDNF levels in mouse hippocampus. Interestingly, all these behavioral, neurochemical and neurobiological changes can be ameliorated by 4W-TE training. In summary, these findings confirm the beneficial impacts of exercise on health and provide further experimental evidence for future application of exercise as an alternative therapy against the mental and cognitive problems in patients with sleep disorders.
Collapse
Affiliation(s)
- Feng Tai
- School of Physical Education, Liaoning Normal University, Dalian, 116029, China
| | - Che Wang
- Department of Medicinal Chemistry, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Xin Deng
- Department of Physical Education, Harbin Engineering University, Haerbin, 150001, China
| | - Ruojin Li
- Department of Medicinal Chemistry, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Zimeng Guo
- School of Physical Education, Liaoning Normal University, Dalian, 116029, China
| | - Haiying Quan
- School of Physical Education, Liaoning Normal University, Dalian, 116029, China.
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China; Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
11
|
Kordestani-Moghadam P, Nasehi M, Vaseghi S, Khodagholi F, Zarrindast MR. The role of sleep disturbances in depressive-like behavior with emphasis on α-ketoglutarate dehydrogenase activity in rats. Physiol Behav 2020; 224:113023. [PMID: 32574661 DOI: 10.1016/j.physbeh.2020.113023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/01/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Sleep disorders may induce anxiety- and depressive-like behaviors. Furthermore, sleep disorders can alter the function of α-KGDH (α-ketoglutarate dehydrogenase), which is involved in the citric acid cycle. In this study, we evaluated the effect of two models of sleep deprivation (SD) including total SD (TSD) and partial SD (PSD), and two models of napping combined with each models of SD on rats' performance in Forced Swim Test (FST) and α-KGDH activity in both hemispheres of the amygdala. 64 male Wistar rats were used in this study. A modified water box was also used to induce SD. The results showed that, immobility was increased in 48-hour PSD group, indicating a possible depressive-like behavior. Swimming time was also increased following 48-hour TSD. However, climbing time was decreased in 48-hour PSD/TSD groups. Additionally, α-KGDH activity was increased in the left amygdala in 48-hour TSD and PSD groups. In conclusion, PSD may increase depressive-like behavior. TSD and PSD can decrease swimming time but increase climbing time, and these effects may be related to serotonergic and noradrenergic transmissions, respectively. Increase in α-KGDH activity in the left amygdala may be related to the brain's need for more energy during prolonged wakefulness. α-KGDH activity in the right amygdala was unaffected probably due to a decrease in alertness following SD.
Collapse
Affiliation(s)
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Department of Pharmacology School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroendocrinology Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Chauhan N, Soni S, Agrawal P, Balhara YPS, Jain U. Recent advancement in nanosensors for neurotransmitters detection: Present and future perspective. Process Biochem 2020. [DOI: 10.1016/j.procbio.2019.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
13
|
Chen Q, Peng Y, Lin Y, Li S, Huang X, Chen LW. Atypical Sleep and Postoperative Delirium in the Cardiothoracic Surgical Intensive Care Unit: A Pilot Prospective Study. Nat Sci Sleep 2020; 12:1137-1144. [PMID: 33324127 PMCID: PMC7733439 DOI: 10.2147/nss.s275698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Postoperative delirium (POD) is a very common and serious neurological complication in patients admitted to the cardiothoracic surgical intensive care unit (CSICU). We aimed to identify a novel potential sleep-based marker for POD and investigate the relevance between atypical sleep and POD. PATIENTS AND METHODS This was a prospective, observational study of patients admitted to the CSICU between December 2019 and February 2020 at our center. Sleep characteristics from 21:00 on postoperative day 1 to 07:00 on postoperative day 2 were assessed using polysomnography (PSG). POD from the end of PSG monitoring until postoperative day 5 was evaluated using the Confusion Assessment Method for the Intensive Care Unit. RESULTS This analysis included 20 patients admitted to the CSICU. The incidence of atypical sleep was 45.0%. Compared to patients without delirium, those with delirium had less delta power, less percentage REM sleep, and a higher proportion of atypical sleep and REM sleep loss (P < 0.05). CONCLUSION The presence of atypical sleep and the absence of REM sleep were associated with POD in patients admitted to the CSICU.
Collapse
Affiliation(s)
- Qiong Chen
- Department of Nursing, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Yanchun Peng
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Yanjuan Lin
- Department of Nursing, Union Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Sailan Li
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Xizhen Huang
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Liang-Wan Chen
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
14
|
Yu R, Yang W, Qi D, Gong L, Li C, Li Y, Jiang H. Targeted neurotransmitter metabolomics profiling of oleanolic acid in the treatment of spontaneously hypertensive rats. RSC Adv 2019; 9:23276-23288. [PMID: 35514525 PMCID: PMC9067294 DOI: 10.1039/c9ra02377a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
Essential hypertension (EH) is a prevalent chronic medical condition and a major risk factor for cardiovascular morbidity and mortality. Neurotransmitters are involved in the physiological process of blood pressure regulation in the body. Studies have shown that oleanolic acid (OA) can effectively regulate neurotransmitter-related metabolic disorders caused by EH, but the mechanism is still unclear. Here, we studied the neurotransmitter metabolic profiles in five brain regions by targeted metabolomics approaches in spontaneously hypertensive rats (SHRs) treated with OA and vehicle. Samples from five brain regions (hippocampus, striatum, hypothalamus, temporal lobe, and frontal lobe) were collected from the control group, the spontaneously hypertensive rat (SHR) group, and the OA group. Targeted metabolomics based on UPLC-Q-Exactive-MS was employed to characterize the dramatically changed neurotransmitters in the brain regions of SHRs treated with OA and vehicle. The expressions of the key enzymes involved in the neurotransmitter metabolism were detected by the reverse transcription-polymerase chain reaction (RT-PCR). The metabolomic profiles of SHRs pre-protected by OA were significantly different from those of unprotected SHRs. A total of 18 neurotransmitters could be confirmed as significantly altered metabolites, which were involved in tyrosine and glutamate metabolism as well as other pathways. The results showing seven key enzymes in neurotransmitter metabolism further validated the changes in the metabolic pathways. OA could effectively restore tyrosine metabolism in the striatum and hypothalamus, glutamate metabolism in the hippocampus, striatum and temporal lobe, cholinergic metabolism in the striatum, and histidine metabolism in the hypothalamus due to its inhibition of inflammatory reactions, structural damage of the neuronal cells, and increase in sedative activity. This study indicated that brain region-targeted metabolomics can provide a powerful tool to further investigate the possible mechanism of OA in EH.
Collapse
Affiliation(s)
- Ruixue Yu
- School of Pharmaceutical Sciences, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine Jinan 250355 China
| | - Wenqing Yang
- Experience Center of Shandong University of Traditional Chinese Medicine Jinan 250355 Shandong China
| | - Dongmei Qi
- Experience Center of Shandong University of Traditional Chinese Medicine Jinan 250355 Shandong China
| | - Lili Gong
- Experience Center of Shandong University of Traditional Chinese Medicine Jinan 250355 Shandong China
| | - Chao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine Jinan China
| | - Yunlun Li
- School of Pharmaceutical Sciences, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine Jinan 250355 China
| | - Haiqiang Jiang
- Experience Center of Shandong University of Traditional Chinese Medicine Jinan 250355 Shandong China
| |
Collapse
|
15
|
Yang SQ, Jiang L, Lan F, Wei HJ, Xie M, Zou W, Zhang P, Wang CY, Xie YR, Tang XQ. Inhibited Endogenous H 2S Generation and Excessive Autophagy in Hippocampus Contribute to Sleep Deprivation-Induced Cognitive Impairment. Front Psychol 2019; 10:53. [PMID: 30733697 PMCID: PMC6353847 DOI: 10.3389/fpsyg.2019.00053] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 01/09/2019] [Indexed: 12/21/2022] Open
Abstract
Background and Aim: Sleep deprivation (SD) causes deficit of cognition, but the mechanisms remain to be fully established. Hydrogen sulfide (H2S) plays an important role in the formation of cognition, while excessive and prolonged autophagy in hippocampus triggers cognitive disorder. In this work, we proposed that disturbances in hippocampal endogenous H2S generation and autophagy might be involved in SD-induced cognitive impairment. Methods: After treatment of adult male wistar rats with 72-h SD, the Y-maze test, object location test (OLT), novel object recognition test (NORT) and the Morris water maze (MWM) test were performed to determine the cognitive function. The autophagosome formation was observed with electron microscope. Generation of endogenous H2S in the hippocampus of rats was detected using unisense H2S microsensor method. The expressions of cystathionine-β-synthase (CBS), 3-mercaptopyruvate sulfurtransferase (3-MST), beclin-1, light chain LC3 II/LC3 I, and p62 in the hippocampus were assessed by western blotting. Results: The Y-maze, OLT, NORT, and MWM test demonstrated that SD-exposed rats exhibited cognitive dysfunction. SD triggered the elevation of hippocampal autophagy as evidenced by enhancement of autophagosome, up-regulations of beclin-1 and LC3 II/LC3 I, and down-regulation of p62. Meanwhile, the generation of endogenous H2S and the expressions of CBS and 3-MST (H2S producing enzyme) in the hippocampus of SD-treated rats were reduced. Conclusion: These results suggested that inhibition of endogenous H2S generation and excessiveness of autophagy in hippocampus are involved in SD-induced cognitive impairment.
Collapse
Affiliation(s)
- San-Qiao Yang
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Li Jiang
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China.,Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Fang Lan
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China.,Department of Neurology, First Affiliated Hospital of University of South China, Hengyang, China
| | - Hai-Jun Wei
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Ming Xie
- Department of Neurology, First Affiliated Hospital of University of South China, Hengyang, China
| | - Wei Zou
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Ping Zhang
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Chun-Yan Wang
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Yu-Rong Xie
- College of Chemistry and Chemical Engineering, University of South China, Hengyang, China
| | - Xiao-Qing Tang
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China.,Department of Neurology, First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
16
|
Sharma A, Muresanu DF, Ozkizilcik A, Tian ZR, Lafuente JV, Manzhulo I, Mössler H, Sharma HS. Sleep deprivation exacerbates concussive head injury induced brain pathology: Neuroprotective effects of nanowired delivery of cerebrolysin with α-melanocyte-stimulating hormone. PROGRESS IN BRAIN RESEARCH 2019; 245:1-55. [PMID: 30961865 DOI: 10.1016/bs.pbr.2019.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|