1
|
Zhou L, Shi H, Xiao M, Liu W, Wang L, Zhou S, Chen S, Wang Y, Liu C. Remimazolam attenuates lipopolysaccharide-induced neuroinflammation and cognitive dysfunction. Behav Brain Res 2025; 476:115268. [PMID: 39322063 DOI: 10.1016/j.bbr.2024.115268] [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: 05/16/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE Remimazolam, a novel benzodiazepine, is widely used as an anesthetic in endoscopic procedures; however, its effects on cognitive function remain unclear, limiting its broader application in general anaesthesia. Neuroinflammation is a well-established key factor in the etiology and progression of cognitive dysfunction, including conditions such as Alzheimer's disease, Parkinson's disease, postoperative delirium, and postoperative cognitive dysfunction. Preclinical studies have demonstrated that remimazolam exerts anti-inflammatory and neuroprotective effects, and clinical reports indicate a reduced incidence of postoperative delirium in patients treated with remimazolam. Nevertheless, whether remimazolam improves cognitive function through its anti-inflammatory properties remains uncertain. This study aimed to investigate the neuroprotective effects of remimazolam and its underlying mechanism in a lipopolysaccharide (LPS)-induced model of neuroinflammation, neuronal injury, and cognitive dysfunction METHODS: C57BL/6 J male mice were administered LPS intraperitoneally to establish a model of neuroinflammation-induced cognitive impairment. A subset of mice received remimazolam via intraperitoneal injection 30 minutes prior to LPS administration. Cognitive performance was evaluated using behavioural tests, including the Morris Water Maze (MWM), Novel Object Recognition (NOR) test, and Open Field Test (OFT). Hippocampal tissues were analyzed by haematoxylin-eosin (HE) staining to assess structural changes. Inflammatory markers, including Interleukin (IL)-6, IL-1β, and tumor necrosis factor-α, were quantified using enzyme-linked immunosorbent assay (ELISA) and real-time quantitative PCR. Immunofluorescence was used to detect translocator protein (TSPO) and markers of microglia activation (IBA-1, CD16/32, and CD206). RESULTS (1) Remimazolam reversed LPS-induced cognitive deficits, as evidenced by shorter spatial exploration latency and increased platform crossings in the MWM, and an elevated recognition index in the NOR test. (2) Remimazolam improved hippocampal morphology, reducing LPS-induced neuronal damage. (3) Remimazolam significantly decreased levels of hippocampal inflammatory cytokines, inhibited microglial activation, promoted M2-type microglia polarization, and increased TSPO expression. CONCLUSION Remimazolam demonstrated neuroprotective and anti-neuroinflammatory effects in a mouse model of LPS-induced cognitive impairment. These effects are likely mediated through the regulation of TSPO, which inhibits microglial activation and promotes the polarization of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype.
Collapse
Affiliation(s)
- Leguang Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China; University of South China Hengyang Medical School Clinical Anatomy & Reproductive Medicine Application Institute, China
| | - Hongzhao Shi
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Mengzhe Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Wenjie Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Lijuan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Shangtao Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Shenghua Chen
- University of South China Hengyang Medical School Clinical Anatomy & Reproductive Medicine Application Institute, China
| | - Yan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China.
| | - Chengxi Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China.
| |
Collapse
|
2
|
Lu Y, Xu C, Xie K, Zhao B, Wang M, Qian C, Chen X, Gu L, Wu W, Lu R. The relationship between thiamin, folic acid and cognitive function in a rat model of uremia. Ren Fail 2024; 46:2329257. [PMID: 38482596 PMCID: PMC10946272 DOI: 10.1080/0886022x.2024.2329257] [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: 06/13/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
End-stage renal disease is a worldwide health burden, but the pathogenesis of uremia-associated cognitive impairment (CI) is poorly recognized. We hypothesized that uremia brings about deficiency of thiamin and folic acid and causes CI by inducing oxidative stress. Therefore, 24 Sprague-Dawley rats were randomly divided into two groups: a 5/6 nephrectomy group (n = 12) and a sham-operated group (n = 12). The Morris water maze was used to assess the cognitive function eight weeks post-surgery, and serum levels of thiamin, folic acid and homocysteine were detected subsequently. Brain and kidney tissues were collected for pathological examination and 8-Hydroxy-2'-deoxyguanosine (8-OHdG) immunochemistry staining. Results showed that the escape latency on training days 1-2 was longer, and the time in quadrant IV on experimental day 6 was significantly shorter in 5/6 nephrectomy group. Meanwhile, the uremic rats showed decreased thiamin, folic acid and increased homocysteine. We also found the time in quadrant IV was positively correlated with thiamin and folic acid level, while negatively correlated with the blood urea nitrogen and 8-OHdG positive cell proportion. Furthermore, in 5/6 nephrectomy group, the hippocampal neuron count was significantly reduced, and a greater proportion of 8-OHdG positive cells were detected. Pretreating LPS-stimulated rat microglial cells with thiamin or folic acid in vitro alleviated the inflammatory impairment in terms of cell viability and oxidative stress. In summary, we applied a uremic rat model and proved that uremia causes serum thiamin and folic acid deficiency, homocysteine elevation, along with neuron reduction and severe oxidative stress in hippocampus, finally leading to CI.
Collapse
Affiliation(s)
- Yifei Lu
- Department of Pharmacy, Shanghai University of Traditional Chinese Medicine, China
| | - Chenqi Xu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kewei Xie
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingru Zhao
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Minzhou Wang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Qian
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuemei Chen
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wangshu Wu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Renhua Lu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Sezer T, Okudan N, Belviranli M. Comparing the effect of high-intensity interval exercise and voluntary exercise training on cognitive functions in rats. Neurosci Lett 2024; 842:137993. [PMID: 39306028 DOI: 10.1016/j.neulet.2024.137993] [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: 07/29/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
It is known that exercise increases brain-derived neurotrophic factor (BDNF) levels in the hippocampus, the brain region responsible for learning and memory, resulting in improved cognitive functions and learning processes. However, it is claimed that different types of exercise cause different responses in the brain. It is thought that lactate and osteocalcin secreted in response to exercise are associated with an increase in BDNF levels. However, there are not enough studies on this subject. This study aimed to compare the effects of high-intensity interval training (HIIT) and voluntary exercise training on cognitive performance and molecular connections. Male rats were randomly divided into control, voluntary exercise training and HIIT groups. The voluntary exercise group had free access to the voluntary wheel for 8 weeks. The HIIT group performed HIIT on the treadmill 3 days a week for 8 weeks. The rats underwent open field (OF), elevated plus maze (EPM) and Morris water maze (MWM) tests 24 h after the last exercise training. Then, after blood was drawn under anesthesia, the rats were sacrificed and their hippocampus tissues were separated. Glucocorticoid and BDNF levels in the blood were evaluated by enzyme-linked immunosorbent assay (ELISA), and osteocalcin and BDNF expressions in the hippocampus were evaluated by real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). Neither voluntary exercise training nor HIIT had any significant effect on behavioral parameters assessed by OF, EPM and MWM tests. However, BDNF expression in hippocampus tissue was higher in the HIIT group than in the control group. In addition, osteocalcin expression in hippocampus tissue was higher in the HIIT and voluntary exercise groups than in the control group. In conclusion, according to the findings we obtained from this study, although it does not have a significant effect on cognitive functions, the effect of HIIT on brain functions seems to be more effective than voluntary exercise.
Collapse
Affiliation(s)
- Tuğba Sezer
- Selcuk University Faculty of Medicine, Department of Physiology, Konya, Turkey.
| | - Nilsel Okudan
- Selcuk University Faculty of Medicine, Department of Physiology, Konya, Turkey.
| | - Muaz Belviranli
- Selcuk University Faculty of Medicine, Department of Physiology, Konya, Turkey.
| |
Collapse
|
4
|
Fan LL, Fang H, Zheng JY, Qiu YH, Wu GL, Cai YF, Chen YB, Zhang SJ. Taohong Siwu decoction alleviates cognitive impairment by suppressing endoplasmic reticulum stress and apoptosis signaling pathway in vascular dementia rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118407. [PMID: 38824979 DOI: 10.1016/j.jep.2024.118407] [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: 03/14/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taohong Siwu Decoction (TSD), a classic traditional Chinese medicine formula, is used for the treatment of vascular diseases, including vascular dementia (VD). However, the mechanisms remain unclear. AIM OF STUDY This study aimed to investigate whether TSD has a positive effect on cognitive impairment in VD rats and to confirm that the mechanism of action is related to the Endoplasmic Reticulum stress (ERs) and cell apoptosis signaling pathway. MATERIALS AND METHODS A total of 40 male adult Sprague-Dawley rats were divided into four groups: sham-operated group (Sham), the two-vessel occlusion group (2VO), the 2VO treated with 4.5 g/kg/d TSD group (2VO + TSD-L), the 2VO treated with 13.5 g/kg/d TSD group (2VO + TSD-H). The rats underwent either 2VO surgery or sham surgery. Postoperative TSD treatment was given for 4 consecutive weeks. Behavioral tests were initiated at the end of gastrulation. Open-field test (OFT) was used to detect the activity level. The New Object Recognition test (NOR) was used to test long-term memory. The Morris water maze (MWM) test was used to examine the foundation of spatial learning and memory. As a final step, the hippocampus was taken for molecular testing. The protein levels of GRP78 (Bip), p-PERK, PERK, IRE1α, p-IRE1α, ATF6, eIF2α, p-eIF2α, ATF4, XBP1, Bcl-2 and Bax were determined by Western blot. Immunofluorescence visualizes molecular expression. RESULTS In the OFT, residence time in the central area was significantly longer in both TSD treatment groups compared to the 2VO group. In the NOR, the recognition index was obviously elevated in both TSD treatment groups. The 2VO group had a significantly longer escape latency and fewer times in crossing the location of the platform compared with the Sham group in MWM. TSD treatment reversed this notion. Pathologically, staining observations confirmed that TSD inhibited hippocampal neuronal loss and alleviated the abnormal reduction of the Nissl body. In parallel, TUNEL staining illustrated that TSD decelerated neuronal apoptosis. Western Blot demonstrated that TSD reduces the expression of ERs and apoptotic proteins. CONCLUSION In this study, the significant ameliorative effect on cognitive impairment of TSD has been determined by comparing the behavioral data of the 4 groups of rats. Furthermore, it was confirmed that this effect of TSD was achieved by suppressing the ERs-mediated apoptosis signaling pathway.
Collapse
Affiliation(s)
- Ling-Ling Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Hao Fang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jia-Yi Zheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yu-Hui Qiu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Guang-Liang Wu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China.
| | - Yun-Bo Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China.
| |
Collapse
|
5
|
Hong XY, Li S, Li T, Chen W, Li Y, Wang Z, Luo Y. Differential involvement of central and peripheral catecholamines between Alzheimer's disease and vascular dementia. Heliyon 2024; 10:e38843. [PMID: 39398044 PMCID: PMC11471233 DOI: 10.1016/j.heliyon.2024.e38843] [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: 05/08/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
Background and aim The important role of catecholamines has been gradually emphasized in the pathogenesis of neurodegenerative process. As the most prevalent form of cognitive dysfunction, Alzheimer's disease (AD) and vascular dementia (VaD) have the distinct pathological features and pathogenic mechanisms, however, the differential involvement of central and peripheral catecholamines between AD and VaD was still unclear. Methods Triple-transgenic AD (3 × Tg-AD) mice and chronic cerebral hypoperfusion (CCH) in rats induced by two-vessel occlusion (2VO) were used as the AD and VaD model in this study, respectively. The concentrations of catecholamines (dopamine, epinephrine and norepinephrine) and their metabolites (3-methoxytyramine, metanephrine and normetanephrine) in serum and five brain regions (hippocampus, cortex, corpus striatum, thalamus and pons) from 3 × Tg-AD mice and 2VO rats were quantitatively determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay. Results High expression and distribution of hippocampal dopamine, and epinephrine and norepinephrine in the cortex and thalamus were found in the early 3 × Tg-AD model, whereas chronic cerebral hypoperfusion induced by 2VO mainly affected the central noradrenergic and noradrenergic system, but not dopaminergic system. The increased serum levels of catecholamines were investigated in the 2VO rats, but not in the 3 × Tg-AD mice. Conclusion The differential expression and distribution of central catecholamines and their metabolites suggests the distinct catecholamines-related pathogenesis between AD and VaD. Peripheral catecholamine surge may be involved in the development of VaD, and the treatment strategy to prevent or reverse the effects of peripheral catecholamines may be protective for VaD.
Collapse
Affiliation(s)
- Xiao-Yue Hong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Siwei Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Tian Li
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Wei Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Zhuo Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| |
Collapse
|
6
|
Zhang S, Xie X, Xu Y, Mi J, Li Z, Guo Z, Xu G. Effects of transcranial magneto-acoustic stimulation on cognitive function and neural signal transmission in the hippocampal CA1 region of mice. Neuroscience 2024; 556:86-95. [PMID: 39047971 DOI: 10.1016/j.neuroscience.2024.01.028] [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: 09/12/2023] [Revised: 12/16/2023] [Accepted: 01/29/2024] [Indexed: 07/27/2024]
Abstract
As a new means of brain neuroregulation and research, transcranial magneto-acoustic stimulation (TMAS) uses the coupling effect of ultrasound and a static magnetic field to regulate neural activity in the corresponding brain areas. Calcium ions can promote the secretion of neurotransmitters and play a key role in the transmission of neural signals in brain cognition. In this study, to explore the effects of TMAS on cognitive function and neural signaling in the CA1 region of the hippocampus, TMAS was applied to male 2-month-old C57 mice with a magnetic field strength of 0.3 T and ultrasound intensity of 2.6 W/cm2. First, the efficiency of neural signaling in the CA1 region of the mouse hippocampus was detected by fiber photometry. Second, the effects of TMAS on cognitive function in mice were investigated through multiple behavioral experiments, including spatial learning and memory ability, anxiety and desire for novelty. The experimental results showed that TMAS could improve cognitive function in mice, and the efficiency of neural signaling in the CA1 area of the hippocampus was significantly increased during stimulation and maintained for one week after stimulation. In addition, the neural signaling efficiency in the CA1 area of the hippocampus increased in the open field (OF) experiment and recovered after one week, the neural signaling efficiency in the new object exploration (NOE) experiment was significantly enhanced, and the intensity slowed after one week. In conclusion, TMAS enhances cognitive performance and promotes neural signaling in the CA1 region of the mouse hippocampus.
Collapse
Affiliation(s)
- Shuai Zhang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China.
| | - Xiaofeng Xie
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Yihao Xu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Jinrui Mi
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Zichun Li
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Zhongsheng Guo
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Guizhi Xu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| |
Collapse
|
7
|
Cao Y, Yao W, Yang T, Yang M, Liu Z, Luo H, Cao Z, Chang R, Cui Z, Zuo H, Liu B. Elucidating the mechanisms of Buyang Huanwu Decoction in treating chronic cerebral ischemia: A combined approach using network pharmacology, molecular docking, and in vivo validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155820. [PMID: 39004032 DOI: 10.1016/j.phymed.2024.155820] [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: 01/23/2024] [Revised: 04/07/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE This study aimed to explore the potential mechanisms of Buyang Huanwu Decoction (BHD) in regulating the AKT/TP53 pathway and reducing inflammatory responses for the treatment of chronic cerebral ischemia (CCI) using UHPLC-QE-MS combined with network pharmacology, molecular docking techniques, and animal experiment validation. METHODS Targets of seven herbal components in BHD, such as Astragalus membranaceus, Paeoniae Rubra Radix, and Ligusticum chuanxiong, were identified through TCMSP and HERB databases. CCI-related targets were obtained from DisGeNET and Genecards, with an intersection analysis conducted to determine shared targets between the disease and the herbal components. Functional enrichment analysis of these intersecting targets was performed. Networks of gene ontology and pathway associations with these targets were constructed and visualized. A pharmacological network involving intersecting genes and active components was delineated. A protein-protein interaction network was established for these intersecting targets and visualized using Cytoscape 3.9.1. The top five genes from the PPI network and their corresponding active components underwent molecular docking. Finally, the 2-vessel occlusion (2-VO) induced CCI rat model was treated with BHD, and the network pharmacology findings were validated using Western blot, RT-PCR, behavioral tests, laser speckle imaging, ELISA, HE staining, Nissl staining, LFB staining, and immunohistochemistry and immunofluorescence. RESULTS After filtration and deduplication, 150 intersecting genes were obtained, with the top five active components by Degree value identified as Quercetin, Beta-Sitosterol, Oleic Acid, Kaempferol, and Succinic Acid. KEGG pathway enrichment analysis linked key target genes significantly with Lipid and atherosclerosis, AGE-RAGE signaling pathway, IL-17 signaling pathway, and TNF signaling pathway. The PPI network highlighted ALB, IL-6, AKT1, TP53, and IL-1β as key protein targets. Molecular docking results showed the strongest binding affinity between ALB and Beta-Sitosterol. Behavioral tests using the Morris water maze indicated that both medium and high doses of BHD could enhance spatial memory in 2-VO model rats, with high-dose BHD being more effective. Laser speckle results showed that BHD at medium and high doses could facilitate CBF recovery in CCI rats, demonstrating a dose-response relationship. HE staining indicated that all doses of BHD could reduce neuronal damage in the cortex and hippocampal CA1 region to varying extents, with the highest dose being the most efficacious. Nissl staining showed that nimodipine and medium and high doses of BHD could alleviate Nissl body damage. LFB staining indicated that nimodipine and medium and high doses of BHD could reduce the pathological damage to fiber bundles and myelin sheaths in the internal capsule and corpus callosum of CCI rats. ELISA results showed that nimodipine and BHD at medium and high doses could decrease the levels of TNF-α, IL-6, IL-17, and IL-1β in the serum of CCI rats (p < 0.05). Immunohistochemistry and immunofluorescence demonstrated that BHD could activate the AKT signaling pathway and inhibit TP53 in treating CCI. Western blot and RT-PCR results indicated that nimodipine and all doses of BHD could upregulate Akt1 expression and downregulate Alb, Tp53, Il-1β, and Il-6 expression in the hippocampus of CCI rats to varying degrees (p < 0.05). CONCLUSION BHD exerts therapeutic effects in the treatment of CCI by regulating targets, such as AKT1, ALB, TP53, IL-1β, and IL-6, and reducing inflammatory responses.
Collapse
Affiliation(s)
- Yue Cao
- College of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wanmei Yao
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, 030619, China
| | - Tao Yang
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Man Yang
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Zhuoxiu Liu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, 030619, China
| | - Huijuan Luo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, 030619, China
| | - Zhuoqing Cao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, 030619, China
| | - Ruifeng Chang
- Third Clinical College,Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Zhiyi Cui
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510000, China
| | - Haojie Zuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510000, China
| | - Biwang Liu
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; School of Fushan, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| |
Collapse
|
8
|
Melgar-Locatelli S, Mañas-Padilla MC, Castro-Zavala A, Rivera P, Del Carmen Razola-Díaz M, Monje FJ, Rodríguez-Pérez C, Castilla-Ortega E. Diet enriched with high-phenolic cocoa potentiates hippocampal brain-derived neurotrophic factor expression and neurogenesis in healthy adult micewith subtle effects on memory. Food Funct 2024; 15:8310-8329. [PMID: 39069830 DOI: 10.1039/d4fo01201a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Cocoa is widely known for its health benefits, but its neurocognitive impact remains underexplored. This preclinical study aimed to investigate the effects of cocoa and cocoa polyphenols on hippocampal neuroplasticity, cognitive function and emotional behavior. Seventy young-adult C57BL/6JRj male and female mice were fed either a standard diet (CTR) or a diet enriched with 10% high-phenolic content cocoa (HPC) or low-phenolic content cocoa (LPC) for at least four weeks. In a first experiment, behavioral tests assessing exploratory behavior, emotional responses and hippocampal-dependent memory were conducted four weeks into the diet, followed by animal sacrifice a week later. Adult hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) expression in the hippocampus and prefrontal cortex were evaluated using immunohistochemistry and western blot. In a different experiment, hippocampal synaptic response, long-term potentiation and presynaptic-dependent short-term plasticity were studied by electrophysiology. Cocoa-enriched diets had minimal effects on exploratory activity and anxiety-like behavior, except for reduced locomotion in the LPC group. Only the HPC diet enhanced object recognition memory, while place recognition memory and spatial navigation remained unaffected. The HPC diet also increased adult hippocampal neurogenesis, boosting the proliferation, survival and number of young adult-born neurons. However, both cocoa-enriched diets increased immobility in the forced swimming test and hippocampal BDNF expression. Hippocampal electrophysiology revealed no alterations in neuroplasticity among diets. The results were mostly unaffected by sex. Overall, the HPC diet demonstrated greater potential regarding cognitive and neuroplastic benefits, suggesting a key role of cocoa flavanols in dietary interventions aimed at enhancing brain health.
Collapse
Affiliation(s)
- Sonia Melgar-Locatelli
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
- Departamento de Nutrición y Bromatología, Universidad de Granada, Campus Universitario de Cartuja, Spain
| | - M Carmen Mañas-Padilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
- Universidad Internacional de la Rioja (UNIR), Spain
| | - Adriana Castro-Zavala
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Spain
| | - María Del Carmen Razola-Díaz
- Departamento de Nutrición y Bromatología, Universidad de Granada, Campus Universitario de Cartuja, Spain
- Instituto de Nutrición y Tecnología de los Alimentos 'José Mataix' (INYTA), Universidad de Granada, Granada, Spain
| | - Francisco J Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharma-cology, Medical University of Vienna, 1090 Vienna, Austria
| | - Celia Rodríguez-Pérez
- Departamento de Nutrición y Bromatología, Universidad de Granada, Campus Universitario de Cartuja, Spain
- Instituto de Nutrición y Tecnología de los Alimentos 'José Mataix' (INYTA), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain
| | - Estela Castilla-Ortega
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Spain
| |
Collapse
|
9
|
Wang L, Yang Z, Satoshi F, Prasanna X, Yan Z, Vihinen H, Chen Y, Zhao Y, He X, Bu Q, Li H, Zhao Y, Jiang L, Qin F, Dai Y, Zhang N, Qin M, Kuang W, Zhao Y, Jokitalo E, Vattulainen I, Kajander T, Zhao H, Cen X. Membrane remodeling by FAM92A1 during brain development regulates neuronal morphology, synaptic function, and cognition. Nat Commun 2024; 15:6209. [PMID: 39043703 PMCID: PMC11266426 DOI: 10.1038/s41467-024-50565-w] [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: 09/18/2023] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
The Bin/Amphiphysin/Rvs (BAR) domain protein FAM92A1 is a multifunctional protein engaged in regulating mitochondrial ultrastructure and ciliogenesis, but its physiological role in the brain remains unclear. Here, we show that FAM92A1 is expressed in neurons starting from embryonic development. FAM92A1 knockout in mice results in altered brain morphology and age-associated cognitive deficits, potentially due to neuronal degeneration and disrupted synaptic plasticity. Specifically, FAM92A1 deficiency impairs diverse neuronal membrane morphology, including the mitochondrial inner membrane, myelin sheath, and synapses, indicating its roles in membrane remodeling and maintenance. By determining the crystal structure of the FAM92A1 BAR domain, combined with atomistic molecular dynamics simulations, we uncover that FAM92A1 interacts with phosphoinositide- and cardiolipin-containing membranes to induce lipid-clustering and membrane curvature. Altogether, these findings reveal the physiological role of FAM92A1 in the brain, highlighting its impact on synaptic plasticity and neural function through the regulation of membrane remodeling and endocytic processes.
Collapse
Affiliation(s)
- Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Ziyun Yang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Fudo Satoshi
- Helsinki Institute of Life Science - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Xavier Prasanna
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Ziyi Yan
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Helena Vihinen
- Helsinki Institute of Life Science (HiLIFE) - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiumei He
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, 541004, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ying Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Feng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ni Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Weihong Kuang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Eija Jokitalo
- Helsinki Institute of Life Science (HiLIFE) - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Tommi Kajander
- Helsinki Institute of Life Science - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Hongxia Zhao
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland.
- School of Life Sciences, Guangxi Normal University, Guilin, China.
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, 541004, China.
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Gayger-Dias V, Menezes L, Da Silva VF, Stiborski A, Silva ACR, Sobottka TM, Quines-Silva VC, Pakulski-Souto B, Bobermin LD, Quincozes-Santos A, Leite MC, Gonçalves CA. Changes in Astroglial Water Flow in the Pre-amyloid Phase of the STZ Model of AD Dementia. Neurochem Res 2024; 49:1851-1862. [PMID: 38733521 DOI: 10.1007/s11064-024-04144-6] [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: 01/11/2024] [Revised: 03/04/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024]
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disease that is typically sporadic and has a high social and economic cost. We utilized the intracerebroventricular administration of streptozotocin (STZ), an established preclinical model for sporadic AD, to investigate hippocampal astroglial changes during the first 4 weeks post-STZ, a period during which amyloid deposition has yet to occur. Astroglial proteins aquaporin 4 (AQP-4) and connexin-43 (Cx-43) were evaluated, as well as claudins, which are tight junction (TJ) proteins in brain barriers, to try to identify changes in the glymphatic system and brain barrier during the pre-amyloid phase. Glial commitment, glucose hypometabolism and cognitive impairment were characterized during this phase. Astroglial involvement was confirmed by an increase in glial fibrillary acidic protein (GFAP); concurrent proteolysis was also observed, possibly mediated by calpain. Levels of AQP-4 and Cx-43 were elevated in the fourth week post-STZ, possibly accelerating the clearance of extracellular proteins, since these proteins actively participate in the glymphatic system. Moreover, although we did not see a functional disruption of the blood-brain barrier (BBB) at this time, claudin 5 (present in the TJ of the BBB) and claudin 2 (present in the TJ of the blood-cerebrospinal fluid barrier) were reduced. Taken together, data support a role for astrocytes in STZ brain damage, and suggest that astroglial dysfunction accompanies or precedes neuronal damage in AD.
Collapse
Affiliation(s)
- Vitor Gayger-Dias
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Leonardo Menezes
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Vanessa-Fernanda Da Silva
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Amanda Stiborski
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Ana Carolina Ribeiro Silva
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Thomas Michel Sobottka
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Vitória Cristine Quines-Silva
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Betina Pakulski-Souto
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Larissa Daniele Bobermin
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
- Graduate Program in Neurosciences, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - André Quincozes-Santos
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
- Graduate Program in Neurosciences, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Marina Concli Leite
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil
| | - Carlos-Alberto Gonçalves
- Biochemistry Laboratory 33, Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil.
- Graduate Program in Neurosciences, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos Street, 2600, Porto Alegre, 90035-003, Brazil.
| |
Collapse
|
11
|
Zhao Y, Ji G, Zhou S, Cai S, Li K, Zhang W, Zhang C, Yan N, Zhang S, Li X, Song B, Qu L. IGF2BP2-Shox2 axis regulates hippocampal-neuronal senescence to alleviate microgravity-induced recognition disturbance. iScience 2024; 27:109917. [PMID: 38812544 PMCID: PMC11134919 DOI: 10.1016/j.isci.2024.109917] [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: 01/22/2024] [Revised: 03/06/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
During space travel, microgravity leads to disturbances in cognitive function, while the underlying mechanism is still unclear. Simulated microgravity mice showed neuronal age-like changes in the hippocampus of our study. In the context of microgravity, we discovered m6A modification reshapes in the hippocampal region. When paired with RNA-seq and MeRIP-seq, Shox2 was found to be a powerful regulator in hippocampal neuron that respondes to microgravity. Decreased expression of senescence-associated secretory phenotype factors and improved genes related to synapses led to the restoration of memory function in the hippocampus upon increased expression of Shox2. Moreover, we discovered that IGF2BP2 was required for the m6A modification of the Shox2, and overexpressed IGF2BP2 in the hippocampus protected against both neuronal senescence and learning and memory decline caused by loss of gravity. Accordingly, our research identified the hippocampal IGF2BP2-Shox2 axis as a possible therapeutic approach to maintaining cognitive function during space travel.
Collapse
Affiliation(s)
- Yujie Zhao
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Guohua Ji
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Sihai Zhou
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Shiou Cai
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Kai Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Wanyu Zhang
- Basic Medical Sciences, Capital Medical University School, Beijing, China
| | - Chuanjie Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Na Yan
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Shuhui Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaopeng Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Bo Song
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Lina Qu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
12
|
Guo H, Li H, Jia Z, Ma S, Zhang J. Edaravone dexborneol attenuates cognitive impairment in a rat model of vascular dementia by inhibiting hippocampal oxidative stress and inflammatory responses and modulating the NMDA receptor signaling pathway. Brain Res 2024; 1833:148917. [PMID: 38582415 DOI: 10.1016/j.brainres.2024.148917] [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: 02/05/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Exploring the intricate pathogenesis of Vascular Dementia (VD), there is a noted absence of potent treatments available in the current medical landscape. A new brain-protective medication developed in China, Edaravone dexboeol (EDB), has shown promise due to its antioxidant and anti-inflammatory properties, albeit with a need for additional research to elucidate its role and mechanisms in VD contexts. In a research setup, a VD model was established utilizing Sprague-Dawley (SD) rats, subjected to permanent bilateral typical carotid artery occlusion (2VO). Behavioral assessment of the rats was conducted using the Bederson test and pole climbing test, while cognitive abilities, particularly learning and memory, were evaluated via the novel object recognition test and the Morris water maze test. Ensuing, the levels of malondialdehyde (MDA), superoxide dismutase (SOD), IL-1β, IL-6, IL-4, and tumor necrosis factor-α (TNF-α) were determined through Enzyme-Linked Immunosorbent Assay (ELISA). Synaptic plasticity-related proteins, synaptophysin (SYP), post-synaptic density protein 95 (PSD-95), and N-methyl-D-aspartate (NMDA) receptor proteins (NR1, NR2A, NR2B) were investigated via Western blotting technique. The findings imply that EDB has the potential to ameliorate cognitive deficiencies, attributed to VD, by mitigating oxidative stress, dampening inflammatory responses, and modulating the NMDA receptor signaling pathway, furnishing new perspectives into EDB's mechanism and proposing potential avenues for therapeutic strategies in managing VD.
Collapse
Affiliation(s)
- Hui Guo
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Haodong Li
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhisheng Jia
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shuyu Ma
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China; First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Jin Zhang
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan,China.
| |
Collapse
|
13
|
Yao D, Li T, Yu L, Hu M, He Y, Zhang R, Wu J, Li S, Kuang W, Yang X, Liu G, Xie Y. Selective degradation of hyperphosphorylated tau by proteolysis-targeting chimeras ameliorates cognitive function in Alzheimer's disease model mice. Front Pharmacol 2024; 15:1351792. [PMID: 38919259 PMCID: PMC11196765 DOI: 10.3389/fphar.2024.1351792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common chronic neurodegenerative diseases. Hyperphosphorylated tau plays an indispensable role in neuronal dysfunction and synaptic damage in AD. Proteolysis-targeting chimeras (PROTACs) are a novel type of chimeric molecule that can degrade target proteins by inducing their polyubiquitination. This approach has shown promise for reducing tau protein levels, which is a potential therapeutic target for AD. Compared with traditional drug therapies, the use of PROTACs to reduce tau levels may offer a more specific and efficient strategy for treating AD, with fewer side effects. In the present study, we designed and synthesized a series of small-molecule PROTACs to knock down tau protein. Of these, compound C8 was able to lower both total and phosphorylated tau levels in HEK293 cells with stable expression of wild-type full-length human tau (termed HEK293-htau) and htau-overexpressed mice. Western blot findings indicated that C8 degraded tau protein through the ubiquitin-proteasome system in a time-dependent manner. In htau-overexpressed mice, the results of both the novel object recognition and Morris water maze tests revealed that C8 markedly improved cognitive function. Together, our findings suggest that the use of the small-molecule PROTAC C8 to degrade phosphorylated tau may be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Dongping Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ting Li
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Mingxing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ye He
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruiming Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Junjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shuoyuan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Weihong Kuang
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020–2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Gongping Liu
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
14
|
Liu S, Yang X, Yuan M, Wang S, Fan H, Zou Q, Pu Y, Cai Z. High salt diet induces cognitive impairment and is linked to the activation of IGF1R/mTOR/p70S6K signaling. Metab Brain Dis 2024; 39:803-819. [PMID: 38771412 DOI: 10.1007/s11011-024-01358-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
A high-salt diet (HSD) has been associated with various health issues, including hypertension and cardiovascular diseases. However, recent studies have revealed a potential link between high salt intake and cognitive impairment. This study aims to investigate the effects of high salt intake on autophagy, tau protein hyperphosphorylation, and synaptic function and their potential associations with cognitive impairment. To explore these mechanisms, 8-month-old male C57BL/6 mice were fed either a normal diet (0.4% NaCl) or an HSD (8% NaCl) for 3 months, and Neuro-2a cells were incubated with normal medium or NaCl medium (80 mM). Behavioral tests revealed learning and memory deficits in mice fed the HSD. We further discovered that the HSD decreased autophagy, as indicated by diminished levels of the autophagy-associated proteins Beclin-1 and LC3, along with an elevated p62 protein level. HSD feeding significantly decreased insulin-like growth factor-1 receptor (IGF1R) expression in the brain of C57BL/6 mice and activated mechanistic target of rapamycin (mTOR) signaling. In addition, the HSD reduced synaptophysin and postsynaptic density protein 95 (PSD95) expression in the hippocampus and caused synaptic loss in mice. We also found amyloid β accumulation and hyperphosphorylation of tau protein at different loci both in vivo and in vitro. Overall, this study highlights the clinical significance of understanding the impact of an HSD on cognitive function. By targeting the IGF1R/mTOR/p70S6K pathway or promoting autophagy, it may be possible to mitigate the negative effects of high salt intake on cognitive function.
Collapse
Affiliation(s)
- Shu Liu
- Chongqing Medical University, 400042, Chongqing, China
- Chongqing institute Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
| | - Xu Yang
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
- Department of Neurology, Affiliated Hospital of Southwest Medical University, 646000, Sichuan, China
| | - Minghao Yuan
- Chongqing Medical University, 400042, Chongqing, China
- Chongqing institute Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
| | - Shengyuan Wang
- Chongqing Medical University, 400042, Chongqing, China
- Chongqing institute Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
| | - Haixia Fan
- Chongqing Medical University, 400042, Chongqing, China
- Chongqing institute Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
| | - Yinshuang Pu
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China
| | - Zhiyou Cai
- Chongqing Medical University, 400042, Chongqing, China.
- Chongqing institute Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China.
- Chongqing School, University of Chinese Academy of Sciences, 400714, Chongqing, Chongqing, China.
- Department of Neurology, Chongqing General Hospital, 400013, Chongqing, Chongqing, China.
- Department of Neurology, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, 400016, Chongqing, Chongqing, China.
| |
Collapse
|
15
|
Gao Z, Santos RB, Rupert J, Van Drunen R, Yu Y, Eckel‐Mahan K, Kolonin MG. Endothelial-specific telomerase inactivation causes telomere-independent cell senescence and multi-organ dysfunction characteristic of aging. Aging Cell 2024; 23:e14138. [PMID: 38475941 PMCID: PMC11296101 DOI: 10.1111/acel.14138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
It has remained unclear how aging of endothelial cells (EC) contributes to pathophysiology of individual organs. Cell senescence results in part from inactivation of telomerase (TERT). Here, we analyzed mice with Tert knockout specifically in EC. Tert loss in EC induced transcriptional changes indicative of senescence and tissue hypoxia in EC and in other cells. We demonstrate that EC-Tert-KO mice have leaky blood vessels. The blood-brain barrier of EC-Tert-KO mice is compromised, and their cognitive function is impaired. EC-Tert-KO mice display reduced muscle endurance and decreased expression of enzymes responsible for oxidative metabolism. Our data indicate that Tert-KO EC have reduced mitochondrial content and function, which results in increased dependence on glycolysis. Consistent with this, EC-Tert-KO mice have metabolism changes indicative of increased glucose utilization. In EC-Tert-KO mice, expedited telomere attrition is observed for EC of adipose tissue (AT), while brain and skeletal muscle EC have normal telomere length but still display features of senescence. Our data indicate that the loss of Tert causes EC senescence in part through a telomere length-independent mechanism undermining mitochondrial function. We conclude that EC-Tert-KO mice is a model of expedited vascular senescence recapitulating the hallmarks aging, which can be useful for developing revitalization therapies.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rafael Bravo Santos
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Joseph Rupert
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rachel Van Drunen
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
16
|
Javaid U, Afroz S, Ashraf W, Saghir KA, Alqahtani F, Anjum SMM, Ahmad T, Imran I. Ameliorative effect of Nyctanthes arbor-tristis L. by suppression of pentylenetetrazole-induced kindling in mice: An insight from EEG, neurobehavioral and in-silico studies. Biomed Pharmacother 2024; 175:116791. [PMID: 38776672 DOI: 10.1016/j.biopha.2024.116791] [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: 04/03/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Epilepsy is an abiding condition associated with recurrent seizure attacks along with associated neurological and psychological emanation owing to disparity of excitatory and inhibitory neurotransmission. The current study encompasses the assessment of the Nyctanthes arbor-tristis L. methanolic extract (Na.Cr) in the management of convulsive state and concomitant conditions owing to epilepsy. The latency of seizure incidence was assessed using pentylenetetrazol (PTZ) kindling models along with EEG in Na.Cr pretreated mice, trailed by behavior assessment (anxiety and memory), biochemical assay, histopathological alterations, chemical profiling through GCMS, and molecular docking. The chronic assessment of PTZ-induced kindled mice depicted salvation in a dose-related pattern and outcomes were noticeable with extract at 400 mg/kg. The extract at 400 mg/kg defends the progress of kindling seizures and associated EEG. Co-morbid conditions in mice emanating owing to epileptic outbreaks were validated by behavioral testing and the outcome depicted a noticeable defense related to anxiety (P<0.001) and cognitive deficit (P<0.001) at 400 mg/kg. The isolated brains were evaluated for oxidative stress and the outcome demonstrated a noticeable effect in a dose-dependent pattern. Treatment with Na.Cr. also preserved the brain from PTZ induced neuronal damage as indicated by histopathological analysis. Furthermore, the GCMS outcome predicted 28 compounds abundantly found in the plant. The results congregated in the current experiments deliver valued evidence about the defensive response apportioned by Na.Cr which might be due to decline in oxidative stress, AChE level, and GABAergic modulation. These activities may contribute to fundamental pharmacology and elucidate some mechanisms behind the activities of Nyctanthes arbor-tristis.
Collapse
Affiliation(s)
- Usman Javaid
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Syeda Afroz
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Khaled Ahmed Saghir
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Syed Muhammad Muneeb Anjum
- The Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore 75270, Pakistan
| | - Tanveer Ahmad
- Institut pour l'Avancée des Biosciences, Centre de Recherche UGA / INSERM U1209 / CNRS 5309, Université Grenoble Alpes, France
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan.
| |
Collapse
|
17
|
Zhang Z, Ding C, Fu R, Wang J, Zhao J, Zhu H. Low-frequency rTMS modulated the excitability and high-frequency firing in hippocampal neurons of the Alzheimer's disease mouse model. Brain Res 2024; 1831:148822. [PMID: 38408558 DOI: 10.1016/j.brainres.2024.148822] [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: 11/09/2023] [Revised: 02/05/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
Repetitive transcranial magnetic stimulation (rTMS), a non-invasive brain stimulation technique, holds potential for applications in the treatment of Alzheimer's disease (AD). This study aims to compare the therapeutic effects of rTMS at different frequencies on Alzheimer's disease and explore the alterations in neuronal electrophysiological properties throughout this process. APP/PS1 AD mice were subjected to two rTMS treatments at 0.5 Hz and 20 Hz, followed by assessments of therapeutic outcomes through the Novel Object Recognition (NOR) and Morris Water Maze (MWM) tests. Following this, whole-cell patch-clamp techniques were used to record action potential, voltage-gated sodium channel currents, and voltage-gated potassium channel currents in dentate gyrus granule neurons. The results show that AD mice exhibit significant cognitive decline compared to normal mice, along with a pronounced reduction in neuronal excitability and ion channel activity. Both frequencies of rTMS treatment partially reversed these changes, demonstrating similar therapeutic efficacy. Furthermore, the investigation indicates that low-frequency magnetic stimulation inhibited the concentrated firing of early action potentials in AD.
Collapse
Affiliation(s)
- Ze Zhang
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Tianjin 300130, China.
| | - Chong Ding
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Tianjin 300130, China; State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin 300130, China.
| | - Rui Fu
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Tianjin 300130, China.
| | - Jiale Wang
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Tianjin 300130, China.
| | - Junqiao Zhao
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Tianjin 300130, China.
| | - Haijun Zhu
- Key Laboratory of Digital Medical Engineering of Hebei Province, College of Electronic & Information Engineering, Hebei University, Baoding, Hebei 071002, China.
| |
Collapse
|
18
|
Mercier O, Quilichini PP, Magalon K, Gil F, Ghestem A, Richard F, Boudier T, Cayre M, Durbec P. Transient demyelination causes long-term cognitive impairment, myelin alteration and network synchrony defects. Glia 2024; 72:960-981. [PMID: 38363046 DOI: 10.1002/glia.24513] [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: 05/17/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
In the adult brain, activity-dependent myelin plasticity is required for proper learning and memory consolidation. Myelin loss, alteration, or even subtle structural modifications can therefore compromise the network activity, leading to functional impairment. In multiple sclerosis, spontaneous myelin repair process is possible, but it is heterogeneous among patients, sometimes leading to functional recovery, often more visible at the motor level than at the cognitive level. In cuprizone-treated mouse model, massive brain demyelination is followed by spontaneous and robust remyelination. However, reformed myelin, although functional, may not exhibit the same morphological characteristics as developmental myelin, which can have an impact on the activity of neural networks. In this context, we used the cuprizone-treated mouse model to analyze the structural, functional, and cognitive long-term effects of transient demyelination. Our results show that an episode of demyelination induces despite remyelination long-term cognitive impairment, such as deficits in spatial working memory, social memory, cognitive flexibility, and hyperactivity. These deficits were associated with a reduction in myelin content in the medial prefrontal cortex (mPFC) and hippocampus (HPC), as well as structural myelin modifications, suggesting that the remyelination process may be imperfect in these structures. In vivo electrophysiological recordings showed that the demyelination episode altered the synchronization of HPC-mPFC activity, which is crucial for memory processes. Altogether, our data indicate that the myelin repair process following transient demyelination does not allow the complete recovery of the initial myelin properties in cortical structures. These subtle modifications alter network features, leading to prolonged cognitive deficits in mice.
Collapse
Affiliation(s)
- Océane Mercier
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale P Quilichini
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Karine Magalon
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Florian Gil
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Antoine Ghestem
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Fabrice Richard
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Thomas Boudier
- Aix Marseille Univ, Turing Centre for Living Systems, Marseille, France
| | - Myriam Cayre
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale Durbec
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| |
Collapse
|
19
|
Li Y, Zhao X, Wang J, Yu Q, Ren J, Jiang Z, Jiao L. Characterization and anti-aging activities of polysaccharide from Rana dybowskii Guenther. Front Pharmacol 2024; 15:1370631. [PMID: 38606177 PMCID: PMC11007062 DOI: 10.3389/fphar.2024.1370631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024] Open
Abstract
Introduction: Rana dybowskii Guenther (RDG), as a traditional Chinese medicine, has been shown to have antioxidant effects. However, studies on the anti-aging effect of RDG are still limited. Methods: In this study, we prepared polysaccharides from the skin of RDG (RDGP) by hot water extraction, alcohol precipitation, ion-exchange chromatography and gel chromatography. The proteins were removed using the Sevage method in combination with an enzymatic method. The structural features were analyzed using high-performance gel permeation chromatography, β-elimination reaction and Fourier transform infrared spectra. The anti-aging effect of RDGP was investigated by using D-Gal to establish an aging model in mice, and pathological changes in the hippocampus were observed under a microscope. Results: We obtained the crude polysaccharide DGP from the skin of RDG, with a yield of 61.8%. The free protein was then removed by the Sevage method to obtain DGPI and deproteinated by enzymatic hydrolysis combined with the Sevage method to further remove the bound protein to obtain the high-purity polysaccharide DGPII. Then, DGPIa (1.03 × 105 Da) and DGPIIa (8.42 × 104 Da) were obtained by gel chromatography, monosaccharide composition analysis showed that they were composed of Man, GlcA, GalNAc, Glc, Gal, Fuc with molar ratios of 1: 4.22 : 1.55: 0.18 : 8.05: 0.83 and 0.74 : 1.78: 1: 0.28: 5.37 : 0.36, respectively. The results of the β-elimination reaction indicated the presence of O-glycopeptide bonds in DGPIa. The Morris water maze test indicated that mice treated with DGPIIa exhibited a significantly shorter escape latency and increased time spent in the target quadrant as well as an increase in the number of times they traversed the platform. Pathologic damage to the hippocampus was alleviated in brain tissue stained with hematoxylin-eosin. In addition, DGPIIa enhanced the activities of SOD, CAT, and GSH-Px and inhibited the level of MDA in the serum and brain tissues of aging mice. Discussion: These results suggest that RDGP has potential as a natural antioxidant and provide useful scientific information for anti-aging research.
Collapse
Affiliation(s)
- Yiping Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xuyan Zhao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Wang
- The Affiliated Hospital Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Qi Yu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Ren
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Ziye Jiang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Lili Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
20
|
Hernández-Rodríguez M, Mera Jiménez E, Nicolás-Vázquez MI, Miranda-Ruvalcaba R. Dihydroergotamine Increases Histamine Brain Levels and Improves Memory in a Scopolamine-Induced Amnesia Model. Int J Mol Sci 2024; 25:3710. [PMID: 38612521 PMCID: PMC11012231 DOI: 10.3390/ijms25073710] [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: 02/15/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The beneficial effects of increasing histamine levels on memory have acquired special interest due to their applicability to psychiatric conditions that cause memory impairments. In addition, by employing drug repurposing approaches, it was demonstrated that dihydroergotamine (DHE), an FDA drug approved to treat migraines, inhibits Histamine N Methyl Transferase (HNMT), the enzyme responsible for the inactivation of histamine in the brain. For this reason, in the present work, the effect of DHE on histamine levels in the hippocampus and its effects on memory was evaluated, employing the scopolamine-induced amnesia model, the Novel Object Recognition (NOR) paradigm, and the Morris Water Maze (MWM). Furthermore, the role of histamine 1 receptor (H1R) and histamine 2 receptor (H2R) antagonists in the improvement in memory produced by DHE in the scopolamine-induced amnesia model was evaluated. Results showed that the rats that received DHE (10 mg/kg, i.p.) showed increased histamine levels in the hippocampus after 1 h of administration but not after 5 h. In behavioral assays, it was shown that DHE (1 mg/kg, i.p.) administered 20 min before the training reversed the memory impairment produced by the administration of scopolamine (2 mg/kg, i.p.) immediately after the training in the NOR paradigm and MWM. Additionally, the effects in memory produced by DHE were blocked by pre-treatment with pyrilamine (20 mg/kg, i.p.) administered 30 min before the training in the NOR paradigm and MWM. These findings allow us to demonstrate that DHE improves memory in a scopolamine-induced amnesia model through increasing histamine levels at the hippocampus due to its activity as an HNMT inhibitor.
Collapse
Affiliation(s)
- Maricarmen Hernández-Rodríguez
- Laboratorio de Cultivo Celular, Neurofarmacología y Conducta, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Ciudad de México 11340, Mexico
| | - Elvia Mera Jiménez
- Laboratorio de Cultivo Celular, Neurofarmacología y Conducta, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Ciudad de México 11340, Mexico
| | - María Inés Nicolás-Vázquez
- Departamento de Ciencias Químicas, Facultad de Estudios Superiores Cuautitlán Campo 1, Universidad Nacional Autónoma de México, Avenida 1o de Mayo s/n, Colonia Santa María las Torres, Cuautitlán Izcalli 54740, Mexico; (M.I.N.-V.); (R.M.-R.)
| | - Rene Miranda-Ruvalcaba
- Departamento de Ciencias Químicas, Facultad de Estudios Superiores Cuautitlán Campo 1, Universidad Nacional Autónoma de México, Avenida 1o de Mayo s/n, Colonia Santa María las Torres, Cuautitlán Izcalli 54740, Mexico; (M.I.N.-V.); (R.M.-R.)
| |
Collapse
|
21
|
Xie Y, Wu Z, Qian Q, Yang H, Ma J, Luan W, Shang S, Li X. Apple polyphenol extract ameliorates sugary-diet-induced depression-like behaviors in male C57BL/6 mice by inhibiting the inflammation of the gut-brain axis. Food Funct 2024; 15:2939-2959. [PMID: 38406886 DOI: 10.1039/d3fo04606k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
To explore whether apple polyphenol extract (APE) ameliorates sugary-diet-induced depression-like behaviors, thirty male C57BL/6 mice (3-4 weeks old) were assigned to three groups randomly to receive different treatments for 8 consecutive weeks: (1) control group (CON), (2) S-HSD group (60% high sucrose diet feeding with 0.1 mg mL-1 sucralose solution as drinking water), and (3) S-APE group (S-HSD feeding with 500 mg per (kg bw day) APE solution gavage). The S-HSD group showed significant depression-like behaviors compared with the CON group, which was manifested by an increased number of buried marbles in the marble burying test, prolonged immobility time in both the tail suspension test and forced swimming test, and cognitive impairment based on the Morris water maze test. However, APE intervention significantly improved the depression-like behaviors by reducing serum levels of corticosterone and adrenocorticotropic hormone, and increasing the serum level of IL-10. Moreover, APE intervention inhibited the activation of the NF-κB inflammatory pathway, elevated colonic MUC-2 protein expression, and elevated the colonic and hippocampal tight junction proteins of occludin and ZO-1. Furthermore, APE intervention increased the richness and diversity of gut microbiota by regulating the composition of microbiota, with increased relative abundance of Firmicutes and Bacteroidota, decreased relative abundance of Verrucomicrobiota at the phylum level, significantly lowered relative abundance of Akkermansia at the genus level, and rebalanced abnormal relative abundance of Muribaculaceae_unclassified, Coriobacteriaceae_UCG-002, and Lachnoclostridium induced by S-HSD feeding. Thus, our study supports the potential application of APE as a dietary intervention for ameliorating depression-like behavioral disorders.
Collapse
Affiliation(s)
- Yisha Xie
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Zhengli Wu
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Qingfan Qian
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Hao Yang
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Jieyu Ma
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Wenxue Luan
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Siyuan Shang
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| | - Xinli Li
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, PR China.
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, PR China
| |
Collapse
|
22
|
Li S, Chen Y, Wang Y, Zhong X, Yu X, Kang Z, Li Y. Liproxstatin-1 alleviates ferroptosis in sevoflurane anesthesia-induced cognitive deficits of aged mice: The role oxidative stress. Synapse 2024; 78:e22286. [PMID: 38287474 DOI: 10.1002/syn.22286] [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: 06/25/2023] [Revised: 11/09/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024]
Abstract
In this study, we aimed to validate the hypothesis that the interplay between sevoflurane, oxidative stress and ferroptosis is crucial for the pathogenesis of sevoflurane-induced cognitive impairment in aged individuals. The mice with sevoflurane-induced cognitive impairment were used to explore the effects of sevoflurane on oxidative stress, iron homeostasis, and cognitive function in aged mice. Iron content and oxidative stress markers were analyzed in hippocampal tissue homogenates using specific assays. Additionally, the levels of iron death-related markers (Fth1 and Gpx4) were assessed by real-time PCR and Western blotting. Morris Water Maze and novel object recognition (NOR) tests were conducted to evaluate cognitive function. Sevoflurane exposure in aged mice resulted in a significant increase in iron overloading in the hippocampus, followed by a subsequent stabilization. Oxidative stress levels were elevated in the hippocampal tissue of sevoflurane-exposed mice, and a significant correlation was observed between iron death and oxidative stress. Liproxstatin-1, a ferroptosis inhibitor, effectively ameliorated the decline in memory and learning abilities induced by sevoflurane anesthesia. Liproxstatin-1 treatment reduced iron overload and oxidative stress in the hippocampal tissue of aged mice. The expression of Fth1 and Gpx4, iron death-related markers, was downregulated following Liproxstatin-1 intervention. Our findings suggest that sevoflurane anesthesia disrupts iron homeostasis, leading to increased oxidative stress and cognitive impairment in aged mice. These results highlight the potential of targeting iron-mediated processes to mitigate sevoflurane-induced cognitive impairment in the aging population.
Collapse
Affiliation(s)
- Shunyuan Li
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Yingle Chen
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Yingmei Wang
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Xianmei Zhong
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Xiaoquan Yu
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Zhenming Kang
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Yangyi Li
- Department of Anesthesiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
23
|
Fu S, Zhao X, Li Y, Fan X, Huang Z. Dexmedetomidine alleviates hippocampal neuronal loss and cognitive decline in rats undergoing open surgery under sevoflurane anaesthesia by suppressing CCAAT/enhancer-binding protein beta. Eur J Neurosci 2024; 59:36-53. [PMID: 37985440 DOI: 10.1111/ejn.16193] [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: 03/28/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Dexmedetomidine (Dex) may exert neuroprotective effects by attenuating inflammatory responses. However, whether Dex specifically improves postoperative cognitive dysfunction (POCD) by inhibiting microglial inflammation through what pathway remains unclear. In this study, the POCD model was constructed by performing open surgery after 3 h of continuous inhalation of 3% sevoflurane to rats, which were intraperitoneally injected with 25 μg/kg Dex .5 h before anaesthesia. The results displayed that Dex intervention decreased rat escape latency, maintained swimming speed and increased the number of times rats crossed the platform and the time spent in the target quadrant. Furthermore, the rat neuronal injury was restored, alleviated POCD modelling-induced rat hippocampal microglial activation and inhibited microglial M1 type polarization. Besides, we administered Dex injection and/or CCAAT/enhancer-binding protein beta (CEBPB) knockdown on the basis of sevoflurane exposure and open surgery and found that CEBPB was knocked down, resulting in the inability of Dex to function, which confirmed CEBPB as a target for Dex treatment. To sum up, Dex improved POCD by considering CEBPB as a drug target to activate the c-Jun N-terminal kinase (JNK)/p-38 signaling pathway, inhibiting microglial M1 polarization-mediated inflammation in the central nervous system.
Collapse
Affiliation(s)
- Shanshan Fu
- Department of Anesthesiology, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Xianghai Zhao
- Department of Anesthesiology, Stomatological Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Yingna Li
- Department of Anesthesiology, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Xinwen Fan
- Department of Anesthesiology, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Zeqing Huang
- Department of Anesthesiology, Cancer Hospital of Dalian University of Technology/Liaoning Cancer Hospital, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Xin Y, Chu T, Zhou S, Xu A. α5GABA A receptor: A potential therapeutic target for perioperative neurocognitive disorders, a review of preclinical studies. Brain Res Bull 2023; 205:110821. [PMID: 37984621 DOI: 10.1016/j.brainresbull.2023.110821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Perioperative neurocognitive disorders (PND) are a common complication in elderly patients following surgery, which not only prolongs the recovery period but also affects their future quality of life and imposes a significant burden on their family and society. Multiple factors, including aging, vulnerability, anesthetic drugs, cerebral oxygen desaturation, and severe pain, have been associated with PND. Unfortunately, no effective drug is currently available to prevent PND. α5 γ-aminobutyric acid subtype A (α5GABAA) receptors have been implicated in cognitive function modulation. Positive or negative allosteric modulators of α5GABAA receptors have been found to improve cognitive impairment under different conditions. Therefore, targeting α5GABAA receptors may represent a promising treatment strategy for PND. This review focuses on preclinical studies of α5GABAA receptors and the risk factors associated with PND, primarily including aging, anesthetics, and neuroinflammation. Specifically, positive allosteric modulators of α5GABAA receptors have improved cognitive function in aged experimental animals. In contrast, negative allosteric modulators of α5GABAA receptors have been found to facilitate cognitive recovery in aged or adult experimental animals undergoing anesthesia and surgery but not in aged experimental animals under anesthesia alone. The reasons for the discordant findings have yet to be elucidated. In preclinical studies, different strategies of drug administration, as well as various behavioral tests, may influence the stability of the results. These issues need to be carefully considered in future studies.
Collapse
Affiliation(s)
- Yueyang Xin
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Tiantian Chu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Siqi Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Aijun Xu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
25
|
Deng F, Zhao F, Wang W, Liu S, Wang Y. Serum lipidomics reveal the mechanism of memory disorder improvement by Qifu decoction. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4663-4673. [PMID: 37668283 DOI: 10.1039/d3ay00899a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Memory disorder (MD) is a neurodegenerative disease that seriously affects the quality of life of the elderly in China. It is characterized by cognitive deficits and psychiatric symptoms. In addition to oxidative damage, neurotransmitter disorders, and other factors, Ca2+ homeostasis and lipid metabolism are among the major pathways of MD etiology. Studies have shown that Ca2+ influx, causing Ca2+ overload, leads to neuronal apoptosis and alterations in lipid metabolites at all MD stages. Qifu decoction (QFD) is one of the classic compounds for the traditional treatment of dementia, which has been shown to significantly improve MD caused by dementia and Alzheimer's disease (AD). So far, it is not clear whether QFD can regulate Ca2+ homeostasis and lipids to improve MD. In this study, we developed a scopolamine hydrobromide MD mouse model and performed neurobehavioral experiments and examinations of brain tissue pathology, Ca2+ homeostasis-related factor levels, and non-targeted lipidomics to explore the mechanism of QFD action in improving MD. The results showed that four weeks of intragastric administration of QFD resulted in significant increases in the cognitive ability and spatial memory ability of the mice with MD. Furthermore, the damage to nerve cells was reduced, the levels of Ca2+ and CaM in the serum were decreased, whereas the content of CaMKII was increased, and the Ca2+ homeostasis was regulated. Non-targeted lipidomics detected four lipid subclasses and 17 potential differential metabolites. Metabolic pathway analysis revealed that QFD significantly regulated sphingolipid metabolism and improved MD. In summary, QFD improves scopolamine hydrobromide memory impairment in mice by regulating the Ca2+ signaling pathway and sphingolipid metabolism. This study provides new insights into the beneficial mechanism of QFD on MD from the perspective of lipidomics.
Collapse
Affiliation(s)
- Fanying Deng
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, 121 Daxue Road, Yuci District, Jinzhong, 030619, China.
| | - Fuxia Zhao
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, 121 Daxue Road, Yuci District, Jinzhong, 030619, China.
| | - Wenhui Wang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, 121 Daxue Road, Yuci District, Jinzhong, 030619, China.
| | - Shiqi Liu
- Schools of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yan Wang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, 121 Daxue Road, Yuci District, Jinzhong, 030619, China.
| |
Collapse
|
26
|
Greene SM, Klein PR, Alcala GA, Bustamante I, Bordas B, Johnson A, Vu V, Uhm SY, Gould GG. Aging to 24 months increased C57BL/6J mouse social sniffing and hippocampal Neto1 levels, and impaired female spatial learning. Behav Processes 2023; 211:104929. [PMID: 37586617 PMCID: PMC11441572 DOI: 10.1016/j.beproc.2023.104929] [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: 02/20/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023]
Abstract
Understanding how natural aging impacts rodent performance in translational behavior tests is critical to teasing apart impairments due to age-related decline from neurodegenerative disorder modeling. Reduced neuropilin and tolloid-like 1 (NETO1), an accessory protein of ionotropic glutamate receptors involved in synaptic plasticity, was associated with Alzheimer's disease, yet aging effects on Neto1 remain unclear. For these reasons, our goal was to characterize how Neto1 expression corresponded with social, repetitive, and spatial learning behaviors and stress response across the C57BL/6J mouse lifespan. We measured social preferences in three-chamber tests, and motor stereotypies by marble burying. Cognitive flexibility is typically assessed in the Morris water maze (MWM), wherein C57BL/6J mice exhibit deficits with age. However, fatigue or locomotor impairment may confound interpretation of MWM performance. Therefore, we used a less arduous water T-maze (WTM) to compare spatial learning flexibility in 2, 9-15, and 24-month-old male and female mice to test the hypothesis that deficits would emerge with age. In both sexes, 9-15-month-olds made more chamber entries during social preference tests, while 2-month-olds did less social sniffing than aged mice. No age or sex differences emerged in marble burying or serum corticosterone measurements. In 24-month-olds hippocampal Neto1was increased relative to 2-month-olds, and male cognitive flexibility was strong, while spatial learning and reversal learning of 24-month-old females was impaired in WTM irrespective of Neto1 expression. The WTM is a useful alternative assessment for cognitive flexibility deficits in aged mice, and the role of hippocampal Neto1 in promoting social sniffing is of interest.
Collapse
Affiliation(s)
- Susan M Greene
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; University of the Incarnate Word, 4301 Broadway, San Antonio, TX 78209, USA
| | - Preston R Klein
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gloria-Andrea Alcala
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; University of the Incarnate Word, 4301 Broadway, San Antonio, TX 78209, USA
| | - Isabela Bustamante
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Trinity University, One Trinity Place, San Antonio, TX 78212, USA
| | - Blanka Bordas
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA 24016, USA
| | - Alexia Johnson
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Howard University, Washington D.C. 20059, USA
| | - Vy Vu
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - So Yeon Uhm
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Georgianna G Gould
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
27
|
Wang F, Li Y, Tang D, Yang B, Tian T, Tian M, Meng N, Xie W, Zhang C, He Z, Zhu X, Ming D, Liu Y. Exploration of the SIRT1-mediated BDNF-TrkB signaling pathway in the mechanism of brain damage and learning and memory effects of fluorosis. Front Public Health 2023; 11:1247294. [PMID: 37711250 PMCID: PMC10499441 DOI: 10.3389/fpubh.2023.1247294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/16/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Fluoride is considered an environmental pollutant that seriously affects organisms and ecosystems, and its harmfulness is a perpetual public health concern. The toxic effects of fluoride include organelle damage, oxidative stress, cell cycle destruction, inflammatory factor secretion, apoptosis induction, and synaptic nerve transmission destruction. To reveal the mechanism of fluorosis-induced brain damage, we analyzed the molecular mechanism and learning and memory function of the SIRT1-mediated BDNF-TrkB signaling pathway cascade reaction in fluorosis-induced brain damage through in vivo experiments. Methods This study constructed rat models of drinking water fluorosis using 50 mg/L, 100 mg/L, and 150 mg/L fluoride, and observed the occurrence of dental fluorosis in the rats. Subsequently, we measured the fluoride content in rat blood, urine, and bones, and measured the rat learning and memory abilities. Furthermore, oxidative stress products, inflammatory factor levels, and acetylcholinesterase (AchE) and choline acetyltransferase (ChAT) activity were detected. The pathological structural changes to the rat bones and brain tissue were observed. The SIRT1, BDNF, TrkB, and apoptotic protein levels were determined using western blotting. Results All rats in the fluoride exposure groups exhibited dental fluorosis; decreased learning and memory abilities; and higher urinary fluoride, bone fluoride, blood fluoride, oxidative stress product, and inflammatory factor levels compared to the control group. The fluoride-exposed rat brain tissue had abnormal AchE and ChAT activity, sparsely arranged hippocampal neurons, blurred cell boundaries, significantly fewer astrocytes, and swollen cells. Furthermore, the nucleoli were absent from the fluoride-exposed rat brain tissue, which also contained folded neuron membranes, deformed mitochondria, absent cristae, vacuole formation, and pyknotic and hyperchromatic chromatin. The fluoride exposure groups had lower SIRT1, BDNF, and TrkB protein levels and higher apoptotic protein levels than the control group, which were closely related to the fluoride dose. The findings demonstrated that excessive fluoride caused brain damage and affected learning and memory abilities. Discussion Currently, there is no effective treatment method for the tissue damage caused by fluorosis. Therefore, the effective method for preventing and treating fluorosis damage is to control fluoride intake.
Collapse
Affiliation(s)
- Feiqing Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Yanju Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Dongxin Tang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Bo Yang
- Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Tingting Tian
- Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Mengxian Tian
- Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Na Meng
- Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Wei Xie
- Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Chike Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Zhixu He
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guiyang, Guizhou Province, China
| | - Xiaodong Zhu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Yang Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guiyang, Guizhou Province, China
| |
Collapse
|
28
|
Ponce-Lopez T, González Álvarez Tostado JA, Dias F, Montiel Maltez KH. Metformin Prevents NDEA-Induced Memory Impairments Associated with Attenuating Beta-Amyloid, Tumor Necrosis Factor-Alpha, and Interleukin-6 Levels in the Hippocampus of Rats. Biomolecules 2023; 13:1289. [PMID: 37759689 PMCID: PMC10526195 DOI: 10.3390/biom13091289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
N-nitrosodiethylamine (NDEA) is a potential carcinogen known to cause liver tumors and chronic inflammation, diabetes, cognitive problems, and signs like Alzheimer's disease (AD) in animals. This compound is classified as probably carcinogenic to humans. Usual sources of exposure include food, beer, tobacco, personal care products, water, and medications. AD is characterized by cognitive decline, amyloid-β (Aβ) deposit, tau hyperphosphorylation, and cell loss. This is accompanied by neuroinflammation, which involves release of microglial cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin 1β (IL-1β), by nuclear factor kappa B (NF-κB) upregulation; each are linked to AD progression. Weak PI3K/Akt insulin-signaling inhibits IRS-1 phosphorylation, activates GSK3β and promotes tau hyperphosphorylation. Metformin, an antihyperglycemic agent, has potent anti-inflammatory efficacy. It reduces proinflammatory cytokines such as IL-6, IL-1β, and TNF-α via NF-κB inhibition. Metformin also reduces reactive oxidative species (ROS) and modulates cognitive disorders reported due to brain insulin resistance links. Our study examined how NDEA affects spatial memory in Wistar rats. We found that all NDEA doses tested impaired memory. The 80 µg/kg dose of NDEA increased levels of Aβ1-42, TNF-α, and IL-6 in the hippocampus, which correlated with memory loss. Nonetheless, treatment with 100 mg/kg of metformin attenuated the levels of pro-inflammatory cytokines and Aβ1-42, and enhanced memory. It suggests that metformin may protect against NDEA-triggered memory issues and brain inflammation.
Collapse
Affiliation(s)
- Teresa Ponce-Lopez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Avenida Universidad Anáhuac 46, Lomas Anáhuac, Huixquilucan C.P. 52786, Estado de México, Mexico
| | | | | | | |
Collapse
|
29
|
Hu Z, Yuan Y, Tong Z, Liao M, Yuan S, Wu W, Tang Y, Wang Y, Tang C, Liu W. Aerobic Exercise Facilitates the Nuclear Translocation of SREBP2 by Activating AKT/SEC24D to Contribute Cholesterol Homeostasis for Improving Cognition in APP/PS1 Mice. Int J Mol Sci 2023; 24:12847. [PMID: 37629027 PMCID: PMC10454400 DOI: 10.3390/ijms241612847] [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: 07/18/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Impaired cholesterol synthesizing ability is considered a risk factor for the development of Alzheimer's disease (AD), as evidenced by reduced levels of key proteases in the brain that mediate cholesterol synthesis; however, cholesterol deposition has been found in neurons in tangles in the brains of AD patients. Although it has been shown that statins, which inhibit cholesterol synthesis, reduce the incidence of AD, this seems paradoxical for AD patients whose cholesterol synthesizing capacity is already impaired. In this study, we aimed to investigate the effects of aerobic exercise on cholesterol metabolism in the brains of APP/PS1 mice and to reveal the mechanisms by which aerobic exercise improves cognitive function in APP/PS1 mice. Our study demonstrates that the reduction of SEC24D protein, a component of coat protein complex II (COPII), is a key factor in the reduction of cholesterol synthesis in the brain of APP/PS1 mice. 12 weeks of aerobic exercise was able to promote the recovery of SEC24D protein levels in the brain through activation of protein kinase B (AKT), which in turn promoted the expression of mem-brane-bound sterol regulatory element-binding protein 2 (SREBP2) nuclear translocation and the expression of key proteases mediating cholesterol synthesis. Simultaneous aerobic exercise restored cholesterol transport capacity in the brain of APP/PS1 mice with the ability to efflux excess cholesterol from neurons and reduced neuronal lipid rafts, thereby reducing cleavage of the APP amyloid pathway. Our study emphasizes the potential of restoring intracerebral cholesterol homeostasis as a therapeutic strategy to alleviate cognitive impairment in AD patients.
Collapse
Affiliation(s)
- Zelin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yangqi Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Meiqing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yingzhe Tang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yirong Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Changfa Tang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
30
|
Liu M, Liu R, Yang M, Ba Y, Deng Q, Zhang Y, Han L, Gao L, Huang H. Combined exposure to lead and high-fat diet induced neuronal deficits in rats: Anti-neuroinflammatory role of SIRT1. Food Chem Toxicol 2023; 177:113857. [PMID: 37244597 DOI: 10.1016/j.fct.2023.113857] [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: 02/28/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Lead (Pb) exposure and high-fat diet (HFD) trigger neurotoxicity, which may involve neuroinflammation. However, the mechanism by which combined Pb and HFD exposure induces nucleotide oligomerization domain-like receptor family pyrin domain 3 (NLRP3) inflammasome activation has not been fully elucidated. MATERIAL AND METHODS The Sprague-Dawley (SD) rat model of exposure to Pb and HFD was established to reveal the influence of co-exposure on cognition and identify signaling clues that mediate neuroinflammation and synaptic dysregulation. PC12 cells was treated with Pb and PA in vitro. Silent information regulator 1 (SIRT1) agonist (SRT 1720) was employed as intervention agent. RESULTS Our results showed that Pb and HFD exposure induced cognitive impairment and lead to neurological damage in rats. Meanwhile, Pb and HFD could stimulate the NLRP3 inflammasome assembly and activate caspase 1, releasing proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18), further promoting neuronal cell activation and amplifying neuroinflammatory responses. Additionally, our findings suggest that SIRT1 plays a role in Pb and HFD induced neuroinflammation. However, the use of SRT 1720 agonists showed some potential in alleviating these impairments. CONCLUSION Pb exposure and HFD intake could induce neuronal damage through activation of the NLRP3 inflammasome pathway and synaptic dysregulation, while the NLRP3 inflammasome pathway may be rescued via activating SIRT1.
Collapse
Affiliation(s)
- Mengchen Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Rundong Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Mingzhi Yang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Yue Ba
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Qihong Deng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Yu Zhang
- State Key Laboratory of Microbial Technology, Qingdao, Shandong, 266000, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong, 250100, China; Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China.
| | - Lin Han
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong, 250100, China; Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, China.
| | - Lihua Gao
- Zhengzhou Center for Disease Control and Prevention, Zhengzhou, Henan, 450052, China.
| | - Hui Huang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| |
Collapse
|
31
|
Ferreira de Sá N, Camarini R, Suchecki D. One day away from mum has lifelong consequences on brain and behaviour. Neuroscience 2023:S0306-4522(23)00276-2. [PMID: 37352967 DOI: 10.1016/j.neuroscience.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
This chapter presents a brief overview of attachment theory and discusses the importance of the neonatal period in shaping an individual's physiological and behavioural responses to stress later in life, with a focus on the role of the parent-infant relationship, particularly in rodents. In rodents, the role of maternal behaviours goes far beyond nutrition, thermoregulation and excretion, acting as hidden regulators of the pup's physiology and development. In this review, we will discuss the inhibitory role of specific maternal behaviours on the ACTH and corticosterone (CORT) stress response. The interest of our group to explore the long-term consequences of maternal deprivation for 24 h (DEP) at different ages (3 days and 11 days) in rats was sparked by its opposite effects on ACTH and CORT levels. In early adulthood, DEP3 animals (males and females alike) show greater negative impact on affective behaviours and stress related parameters than DEP11, indicating that the latter is more resilient in tests of anxiety-like behaviour. These findings create an opportunity to explore the neurobiological underpinnings of vulnerability and resilience to stress-related disorders. The chapter also provides a brief historical overview and highlights the relevance of attachment theory, and how DEP helps to understand the effects of childhood parental loss as a risk factor for depression, schizophrenia, and PTSD in both childhood and adulthood. Furthermore, we present the concept of environmental enrichment (EE), its effects on stress responses and related behavioural changes and its benefits for rats previously subjected to DEP, along with the clinical implications of DEP and EE.
Collapse
Affiliation(s)
- Natália Ferreira de Sá
- Department of Psychobiology - Escola Paulista de Medicina, Universidade Federal de São Paulo
| | - Rosana Camarini
- Department of Pharmacology - Instituto de Ciências Biomédicas, Universidade de São Paulo
| | - Deborah Suchecki
- Department of Psychobiology - Escola Paulista de Medicina, Universidade Federal de São Paulo.
| |
Collapse
|
32
|
Mabry S, Wilson EN, Bradshaw JL, Gardner JJ, Fadeyibi O, Vera E, Osikoya O, Cushen SC, Karamichos D, Goulopoulou S, Cunningham RL. Sex and age differences in social and cognitive function in offspring exposed to late gestational hypoxia. RESEARCH SQUARE 2023:rs.3.rs-2507737. [PMID: 37333114 PMCID: PMC10275064 DOI: 10.21203/rs.3.rs-2507737/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Gestational sleep apnea affects 8-26% of pregnancies and can increase the risk for autism spectrum disorder (ASD) in offspring. ASD is a neurodevelopmental disorder associated with social dysfunction, repetitive behaviors, anxiety, and cognitive impairment. To examine the relationship between gestational sleep apnea and ASD-associated behaviors, we used a chronic intermittent hypoxia (CIH) protocol between gestational days (GD) 15-19 in pregnant rats to model late gestational sleep apnea. We hypothesized that late gestational CIH would produce sex- and age-specific social, mood, and cognitive impairments in offspring. Methods Timed pregnant Long-Evans rats were exposed to CIH or room air normoxia from GD 15-19. Behavioral testing of offspring occurred during either puberty or young adulthood. To examine ASD-associated phenotypes, we quantified ASD-associated behaviors (social function, repetitive behaviors, anxiety-like behaviors, and spatial memory and learning), hippocampal activity (glutamatergic NMDA receptors, dopamine transporter, monoamine oxidase-A, EGR-1, and doublecortin), and circulating hormones in offspring. Results Late gestational CIH induced sex- and age-specific differences in social, repetitive and memory functions in offspring. These effects were mostly transient and present during puberty. In female pubertal offspring, CIH impaired social function, increased repetitive behaviors, and increased circulating corticosterone levels, but did not impact memory. In contrast, CIH transiently induced spatial memory dysfunction in pubertal male offspring but did not impact social or repetitive functions. Long-term effects of gestational CIH were only observed in female offspring, wherein CIH induced social disengagement and suppression of circulating corticosterone levels in young adulthood. No effects of gestational CIH were observed on anxiety-like behaviors, hippocampal activity, circulating testosterone levels, or circulating estradiol levels, regardless of sex or age of offspring. Conclusions Our results indicate that hypoxia-associated pregnancy complications during late gestation can increase the risk for ASD-associated behavioral and physiological outcomes, such as pubertal social dysfunction, corticosterone dysregulation, and memory impairments.
Collapse
Affiliation(s)
- Steve Mabry
- UNTHSC: University of North Texas Health Science Center
| | | | | | | | | | - Edward Vera
- UNTHSC: University of North Texas Health Science Center
| | | | | | | | | | | |
Collapse
|
33
|
Aleksandrova Y, Munkuev A, Mozhaitsev E, Suslov E, Tsypyshev D, Chaprov K, Begunov R, Volcho K, Salakhutdinov N, Neganova M. Elaboration of the Effective Multi-Target Therapeutic Platform for the Treatment of Alzheimer's Disease Based on Novel Monoterpene-Derived Hydroxamic Acids. Int J Mol Sci 2023; 24:ijms24119743. [PMID: 37298694 DOI: 10.3390/ijms24119743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Novel monoterpene-based hydroxamic acids of two structural types were synthesized for the first time. The first type consisted of compounds with a hydroxamate group directly bound to acyclic, monocyclic and bicyclic monoterpene scaffolds. The second type included hydroxamic acids connected with the monoterpene moiety through aliphatic (hexa/heptamethylene) or aromatic linkers. An in vitro analysis of biological activity demonstrated that some of these molecules had powerful HDAC6 inhibitory activity, with the presence of a linker area in the structure of compounds playing a key role. In particular, it was found that hydroxamic acids containing a hexa- and heptamethylene linker and (-)-perill fragment in the Cap group exhibit excellent inhibitory activity against HDAC6 with IC50 in the submicromolar range from 0.56 ± 0.01 µM to 0.74 ± 0.02 µM. The results of the study of antiradical activity demonstrated the presence of moderate ability for some hydroxamic acids to scavenge 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2ROO• radicals. The correlation coefficient between the DPPH radical scavenging activity and oxygen radical absorbance capacity (ORAC) value was R2 = 0.8400. In addition, compounds with an aromatic linker based on para-substituted cinnamic acids, having a monocyclic para-menthene skeleton as a Cap group, 35a, 38a, 35b and 38b, demonstrated a significant ability to suppress the aggregation of the pathological β-amyloid peptide 1-42. The 35a lead compound with a promising profile of biological activity, discovered in the in vitro experiments, demonstrated neuroprotective effects on in vivo models of Alzheimer's disease using 5xFAD transgenic mice. Together, the results obtained demonstrate a potential strategy for the use of monoterpene-derived hydroxamic acids for treatment of various aspects of Alzheimer's disease.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, Chernogolovka 142432, Russia
| | - Aldar Munkuev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Evgenii Mozhaitsev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Evgenii Suslov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Dmitry Tsypyshev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Kirill Chaprov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, Chernogolovka 142432, Russia
| | - Roman Begunov
- Biology and Ecology Faculty of P. G. Demidov Yaroslavl State University, Matrosova Ave., 9, Yaroslavl 150003, Russia
| | - Konstantin Volcho
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Nariman Salakhutdinov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, Chernogolovka 142432, Russia
| |
Collapse
|
34
|
Kudara M, Kato-Ishikura E, Ikegaya Y, Matsumoto N. Ramelteon administration enhances novel object recognition and spatial working memory in mice. J Pharmacol Sci 2023; 152:128-135. [PMID: 37169477 DOI: 10.1016/j.jphs.2023.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Ramelteon is used to ameliorate sleep disorders that negatively affect memory performance; however, it remains unknown whether ramelteon strengthens neutral memories, which do not involve reward or punishment. To address this, we monitored behavior of mice treated with vehicle/ramelteon while they performed a novel object recognition task and a spontaneous alternation task. Object memory performance in the novel object recognition task was improved only if ramelteon was injected before training, suggesting that ramelteon specifically enhances the acquisition of object recognition memory. Ramelteon also enhanced spatial working memory in the spontaneous alternation task. Altogether, acute ramelteon treatment enhances memory in quasi-natural contexts.
Collapse
Affiliation(s)
- Mikuru Kudara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Eriko Kato-Ishikura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
35
|
Ma T, Feng L, Wei S, Wang Y, Li G, Lu Y, Zhang Y, Chu Y, Wang W, Zhang H. Antisense oligonucleotides targeting basal forebrain ATXN2 enhances spatial memory and ameliorates sleep deprivation-induced fear memory impairment in mice. Brain Behav 2023; 13:e3013. [PMID: 37072935 PMCID: PMC10275523 DOI: 10.1002/brb3.3013] [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: 07/19/2022] [Revised: 11/04/2022] [Accepted: 12/24/2022] [Indexed: 04/20/2023] Open
Abstract
INTRODUCTION Regulation of brain-derived neurotrophic factor (BDNF) in the basal forebrain ameliorates sleep deprivation-induced fear memory impairments in rodents. Antisense oligonucleotides (ASOs) targeting ATXN2 was a potential therapy for spinocerebellar ataxia, whose pathogenic mechanism associates with reduced BDNF expression. We tested the hypothesis that ASO7 targeting ATXN2 could affect BDNF levels in mouse basal forebrain and ameliorate sleep deprivation-induced fear memory impairments. METHODS Adult male C57BL/6 mice were used to evaluate the effects of ASO7 targeting ATXN2 microinjected into the bilateral basal forebrain (1 μg, 0.5 μL, each side) on spatial memory, fear memory and sleep deprivation-induced fear memory impairments. Spatial memory and fear memory were detected by the Morris water maze and step-down inhibitory avoidance test, respectively. Immunohistochemistry, RT-PCR, and Western blot were used to evaluate the changes of levels of BDNF, ATXN2, and postsynaptic density 95 (PSD95) protein as well as ATXN2 mRNA. The morphological changes in neurons in the hippocampal CA1 region were detected by HE staining and Nissl staining. RESULTS ASO7 targeting ATXN2 microinjected into the basal forebrain could suppress ATXN2 mRNA and protein expression for more than 1 month and enhance spatial memory but not fear memory in mice. BDNF mRNA and protein expression in basal forebrain and hippocampus was increased by ASO7. Moreover, PSD95 expression and synapse formation were increased in the hippocampus. Furthermore, ASO7 microinjected into the basal forebrain increased BDNF and PSD95 protein expression in the basal forebrain of sleep-deprived mice and counteracted sleep deprivation-induced fear memory impairments. CONCLUSION ASOs targeting ATXN2 may provide effective interventions for sleep deprivation-induced cognitive impairments.
Collapse
Affiliation(s)
- Tao Ma
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Long Feng
- Department of AnesthesiologyPLA General Hospital of Hainan HospitalHainanChina
| | - Shi‐Nan Wei
- PLA Rocket Force Characteristic Medical Center, Postgraduate Training Base of Jinzhou Medical UniversityBeijingChina
| | - Ying‐Ying Wang
- Department of AnesthesiologyBeijing Ditan Hospital, Capital Medical UniversityBeijingChina
| | - Guan‐Hua Li
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yan Lu
- Department of NeurologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Ying‐Xin Zhang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yang Chu
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Wei Wang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Hao Zhang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| |
Collapse
|
36
|
Li X, Gao Y, Han X, Tang S, Li N, Liu X, Ni X. Maresin1 ameliorates postoperative cognitive dysfunction in aged rats by potentially regulating the NF-κB pathway to inhibit astrocyte activation. Exp Gerontol 2023; 176:112168. [PMID: 37055002 DOI: 10.1016/j.exger.2023.112168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is one of the most serious postoperative complications in the elderly population. Perioperative central neuroinflammation is considered to be an important pathological mechanism of POCD, with the activation of astrocytes playing a key role in central neuroinflammation. Maresin1 (MaR1) is a specific pro-resolving mediator synthesized by macrophages in the resolution stage of inflammation, and provides unique anti-inflammatory and pro-resolution effects by limiting excessive neuroinflammation and promoting postoperative recovery. However, the question remains whether MaR1 can have a positive effect on POCD. The objective of this study was to investigate the protective effect of MaR1 on POCD cognitive function in aged rats after splenectomy. Morris water maze test and IntelliCage test showed that splenectomy could cause transient cognitive dysfunction in aged rats; however, the cognitive impairment of rats was significantly mitigated when MaR1 pretreatment was administered. MaR1 significantly alleviated the fluorescence intensity and protein expression of glial fibrillary acidic protein and central nervous system specific protein in the cornu ammonis 1 region of the hippocampus. Simultaneously, the morphology of astrocytes was also severely altered. Further experiments showed that MaR1 inhibited the mRNA and protein expression of several key proinflammatory cytokines-interleukin-1β, interleukin-6, and tumor necrosis factor-α in the hippocampus of aged rats following splenectomy. The molecular mechanism underlying this process was explored by evaluating expression of components of the nuclear factor κB (NF-κB) signaling pathway. MaR1 substantially inhibited the mRNA and protein expression of NF-κB p65 and κB inhibitor kinase β. Collectively, these results suggest that MaR1 ameliorated splenectomy-induced transient cognitive impairment in elderly rats, and this neuroprotective mechanism may occur through regulating the NF-κB pathway to inhibit astrocyte activation.
Collapse
Affiliation(s)
- Xiuhua Li
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yubo Gao
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xu Han
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Shaling Tang
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Na Li
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xing Liu
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xinli Ni
- Department of Anaesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
37
|
Lv JM, Zhang LM, Wang JX, Shao JJ, Guo XG, Wang LY, Kang LQ, Zheng WC, Xin Y, Song RX, Guo W, Zhang DX. Abdominal surgery plus sevoflurane exposure induces abnormal emotional changes and cognitive dysfunction in aged rats. Behav Brain Res 2023; 442:114328. [PMID: 36740076 DOI: 10.1016/j.bbr.2023.114328] [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: 10/11/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cognitive impairment, which includes perioperative psychological distress and cognitive dysfunction, can be determined by preoperative and post-operative neuropsychological tests. Several mechanisms have been proposed regarding the two-way communication between the immune system and the brain after surgery. We aimed to understand the mechanisms underlying perioperative neurocognitive disorders (PND) in elderly rats using an experimental abdominal surgery model. METHODS 24-month-old SD rats were exposed to the abdominal surgery model (AEL) under 3% anesthesia. On day 15 and day 30 post-surgery, fractional anisotropy (FA) using diffusion kurtosis imaging (DKI) was measured. From day 25 to day 30 post-surgery, behavioral tests, including open field test (OFT), Morris water maze (MWM), novel object recognition (NOR), force swimming test (FST), and elevated plus maze (EPM), were performed. Then, the rats were euthanized to perform pathological analysis and western blot measurement. RESULTS The rats exposed to AEL surgical treatment demonstrated significantly decreased time crossing the platform in the MWM, decreased recognition index in the NOR, reduced time in the open arm in the EPM, increased immobility time in the FST, and increased number of crossings in the OFT. Aged rats, after AEL exposure, further demonstrated decreased FA in the mPFC, nucleus accumbens (NAc), and hippocampus, together with reduced MAP2 intensity, attenuation of GAD65, VGlut2, CHAT, and phosphorylated P38MAPK expression, and increased reactive astrocytes and microglia. CONCLUSIONS In this study, the aged rats exposed to abdominal surgery demonstrated both emotional changes and cognitive dysfunction, which may be associated with neuronal degeneration and reduced phosphorylated P38MAPK.
Collapse
Affiliation(s)
- Jin-Meng Lv
- Department of Anesthesia and Trauma Research, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
| | - Jie-Xia Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China.
| | - Jing-Jing Shao
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Xin-Gui Guo
- Department of Medical Iconography, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China.
| | - Lu-Ying Wang
- Department of Anesthesia and Trauma Research, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
| | - Li-Qing Kang
- Department of Medical Iconography, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China.
| | - Wei-Chao Zheng
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
| | - Yue Xin
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China.
| | - Rong-Xin Song
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China.
| | - Wei Guo
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China.
| |
Collapse
|
38
|
Portero-Tresserra M, Galofré-López N, Pallares E, Gimenez-Montes C, Barcia C, Granero R, Rojic-Becker D, Vale-Martínez A, Martí-Nicolovius M, Guillazo-Blanch G. Effects of Caloric Restriction on Spatial Object Recognition Memory, Hippocampal Neuron Loss and Neuroinflammation in Aged Rats. Nutrients 2023; 15:nu15071572. [PMID: 37049417 PMCID: PMC10096994 DOI: 10.3390/nu15071572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Age-related neurobiological changes significantly affect hippocampal structure and function, such that the main cognitive impairments associated with aging are related to the integrity of this brain structure, including the deterioration in spatial object recognition (SOR) memory. Previous studies have shown that intrinsic factors such as neuroinflammation, as well as lifestyle factors such as diet, can affect aging-associated brain functions and cognitive performance. In this regard, caloric restriction (CR) produces beneficial effects on health and life expectancy, although its ability to slow down age-dependent effects on cognitive decline and hippocampus (HPC) functioning remains unclear. Therefore, we set out to evaluate the effects of CR on SOR memory in aged male Wistar rats, as well as those on hippocampal neuron loss, neurogenesis and inflammation. The data show that CR in aged rats attenuates the decline in SOR memory, age-associated hippocampal neuron loss, and age-dependent microglial activation. Furthermore, we found a significant reduction in neurogenesis in the dentate gyrus of the old animals relative to adult rats. These findings support the positive effect of CR on SOR memory, suggesting that it dampens hippocampal neuronal loss and reduces proinflammatory activity.
Collapse
Affiliation(s)
- Marta Portero-Tresserra
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Neus Galofré-López
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Elisabet Pallares
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Claudia Gimenez-Montes
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Carlos Barcia
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Roser Granero
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Divka Rojic-Becker
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Anna Vale-Martínez
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Margarita Martí-Nicolovius
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Gemma Guillazo-Blanch
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
39
|
Wu H, Wang Y, Fu H, Ji L, Li N, Zhang D, Su L, Hu Z. Maresin1 Ameliorates Sepsis-Induced Microglial Neuritis Induced through Blocking TLR4-NF-κ B-NLRP3 Signaling Pathway. J Pers Med 2023; 13:jpm13030534. [PMID: 36983716 PMCID: PMC10054512 DOI: 10.3390/jpm13030534] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/09/2023] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Objective: Neuroinflammation is a major etiology of cognitive dysfunction due to sepsis. Maresin1 (MaR1), identified as a docosahexaenoic acid (DHA)-derived metabolite from macrophages, has been demonstrated to exhibit potent neuroprotective and anti-inflammatory effects. Nevertheless, detailed functions and molecular mechanism of MaR1 in sepsis-induced cognitive dysfunction has not been fully elucidated. Here, we aimed to investigate potential neuroprotective effects of MaR1 on microglia-induced neuroinflammation in sepsis-induced cognitive impairment and to explore its anti-inflammatory mechanism. Methods: Different doses of MaR1 were administered to septic rats by via tail vein injection. The optimal dose was determined based on the 7-day survival rate of rats from each group. derived from macrophages with both anti-inflammatory to observe the ameliorative effects of MaR1 at optimal doses on cognitive dysfunction in septic rats. The effects of MaR1 on neuroinflammation-mediated microglial activation, neuronal apoptosis, and pro-inflammatory cytokine productions were in vivo and in vitro assayed, using Western blot, ELISA, TUNEL staining, Nissl staining, and the immunofluorescence method. To further elucidate anti-inflammatory machinery of MaR1, protein expressions of NLRP3 inflammatory vesicles and TLR4-NF-κB pathway-related proteins were subjected to Western blot assay. Results: After tail vein injection of MaR1 with different doses (2 ng/g, 4 ng/g, 8 ng/g), the results showed that 4 ng/g MaR1 treatment significantly increased the rats’ 7-day survival rate compared to the CLP controls. Therefore, subsequent experiments set 4 ng/g MaR1 as the optimal dose. Morris water maze experiments confirmed that MaR1 significantly reduced space memory dysfunction in rats. In addition, in CLP rats and LPS-stimulated BV2 microglia, MaR1 significantly reduced activated microglia and pro-inflammatory cytokines levels and neuronal apoptosis. Mechanically, MaR1 inhibits microglia-induced neuroinflammation through suppressing activations of NLRP3 inflammatory vesicles and TLR4-NF-κB signal pathway. Conclusion: Collectively, our findings suggested that MaR1 might be a prospective neuroprotective compound for prevention and treatment in the sepsis process.
Collapse
Affiliation(s)
- Huiping Wu
- School of Medicine, Soochow University, Suzhou 215006, China
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Ying Wang
- Operating Room, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121012, China
| | - Haiyan Fu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Lili Ji
- Emergency Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121012, China
| | - Na Li
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Dan Zhang
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Longxiang Su
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Beijing 100730, China
- Correspondence: (L.S.); (Z.H.)
| | - Zhansheng Hu
- School of Medicine, Soochow University, Suzhou 215006, China
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
- Correspondence: (L.S.); (Z.H.)
| |
Collapse
|
40
|
Chen L, Xiong L, Yao L, Pan J, Arzola E, Zhu X, Mei L, Xiong WC. Attenuation of Alzheimer's brain pathology in 5XFAD mice by PTH 1-34, a peptide of parathyroid hormone. Alzheimers Res Ther 2023; 15:53. [PMID: 36918976 PMCID: PMC10012528 DOI: 10.1186/s13195-023-01202-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and osteoporosis are two distinct diseases but often occur in the same patient. Their relationship remains poorly understood. Studies using Tg2576 AD animal model demonstrate bone deficits, which precede the brain phenotypes by several months, arguing for the independence of bone deficits on brain degeneration and raising a question if the bone deficits contribute to the AD development. To address this question, we investigated the effects of PTH1-34, a peptide of parathyroid hormone analog and a well-recognized effective anabolic therapy drug for patients with osteoporosis, on 5XFAD animal model. METHODS 5XFAD mice, an early onset β-amyloid (Aβ)-based AD mouse model, were treated with PTH1-34 intermittently [once daily injection of hPTH1-34 (50 μg/Kg), 5 days/week, starting at 2-month old (MO) for 2-3 month]. Wild type mice (C57BL/6) were used as control. The bone phenotypes were examined by microCT and evaluated by measuring serum bone formation and resorption markers. The AD relevant brain pathology (e.g., Aβ and glial activation) and behaviors were assessed by a combination of immunohistochemical staining analysis, western blots, and behavior tests. Additionally, systemic and brain inflammation were evaluated by serum cytokine array, real-time PCR (qPCR), and RNAscope. RESULTS A reduced trabecular, but not cortical, bone mass, accompanied with a decrease in bone formation and an increase in bone resorption, was detected in 5XFAD mice at age of 5/6-month old (MO). Upon PTH1-34 treatments, not only these bone deficits but also Aβ-associated brain pathologies, including Aβ and Aβ deposition levels, dystrophic neurites, glial cell activation, and brain inflammatory cytokines, were all diminished; and the cognitive function was improved. Further studies suggest that PTH1-34 acts on not only osteoblasts in the bone but also astrocytes in the brain, suppressing astrocyte senescence and expression of inflammatory cytokines in 5XFAD mice. CONCLUSIONS These results suggest that PTH1-34 may act as a senolytic-like drug, reducing systemic and brain inflammation and improving cognitive function, and implicate PTH1-34's therapeutic potential for patients with not only osteoporosis but also AD.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA.,Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Lingling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA
| | - Jinxiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Emily Arzola
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA
| | - Xiaojuan Zhu
- Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.
| |
Collapse
|
41
|
Blokland A, Sesia T. Delay-dependent forgetting in object recognition and object location test is dependent on strain and test. Behav Brain Res 2023; 437:114161. [PMID: 36257558 DOI: 10.1016/j.bbr.2022.114161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/01/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
The object recognition and object location task (ORT and OLT, respectively) have been applied in preclinical research to evaluate the effects of treatments on memory. Although both tasks look quite similar, they differ with respect to the brain structures involved in the memory performance. The characterization of the memory performance in both tasks is important to understand treatment effects. Since there are no previous studies that compared strain differences in delay-dependent forgetting in both tasks, Wistar and Long Evans rats were tested in both the ORT and the OLT at different intervals. The data showed that in the ORT the delay-dependent forgetting was similar for Wistar and Long Evans rats. However, the forgetting curve was different for both strains in the OLT: the Long Evans rats the forgetting took a longer interval. This study indicates that delay-dependent forgetting in the ORT and OLT is strain and test dependent. It is suggested that before testing treatments the forgetting curve of a specific strain should be tested in this type of tasks.
Collapse
Affiliation(s)
- Arjan Blokland
- Faculty of Psychology and Neuroscience, Department of Neuropsychology & Psychopharmacology, EURON, Maastricht University, the Netherlands; European Graduate School of Neuroscience (EURON), the Netherlands.
| | - Thibaut Sesia
- Department of Stereotactic and Functional Neurosurgery, University Hospital Cologne, Cologne, Germany; European Graduate School of Neuroscience (EURON), the Netherlands
| |
Collapse
|
42
|
Wang Z, Li T, Du M, Zhang L, Xu L, Song H, Zhang J. β-hydroxybutyrate improves cognitive impairment caused by chronic cerebral hypoperfusion via amelioration of neuroinflammation and blood-brain barrier damage. Brain Res Bull 2023; 193:117-130. [PMID: 36577190 DOI: 10.1016/j.brainresbull.2022.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Vascular cognitive impairment (VCI) is the second most common type of dementia after Alzheimer's disease (AD) in elderly people. Chronic cerebral hypoperfusion (CCH) is the early pathophysiological basis of VCI. β-Hydroxybutyrate (BHB) is one of the important components of ketone bodies, an intermediate product of endogenous energy metabolism, which can mitigate neuroinflammation in stroke and neurodegenerative diseases. The present study aimed to investigate whether BHB can improve cognitive impairment caused by CCH and the underlying mechanism. METHODS The CCH model was established by permanent bilateral common carotid artery occlusion (2VO). CCH rats were intraperitoneally injected with BHB (1.5 mmol/kg/d) every day for 8 consecutive weeks from 2 weeks before surgery. The hippocampal blood flow of rats was measured by using a laser Doppler velocimetry. Used the Morris water maze test (MWM) to assess spatial learning and memory of rats, and harvested brain tissues for molecular, biochemical, and pathological tests. RESULTS We found that BHB intervention for 8 weeks could effectively restore hippocampal blood flow and improve spatial learning and memory in CCH rats. BHB can protect the blood-brain barrier (BBB), as manifested by reducing the ultrastructural damage and leakage of the BBB, restoring the expression of tight junction-related proteins and reducing the expression of Matrix Metalloproteinases-9 (MMP-9). Additionally, after BHB intervention, microglia activation was reduced, oligodendrocyte motility was active, and the expression levels of pro-inflammatory factors such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), nuclear factor-κB (NF-κB) and advanced glycation end-products (RAGE) were lower, which also indicated that BHB had a beneficial effect in mitigating neuroinflammation. CONCLUSION BHB can improve the cognitive impairment caused by CCH. The potential mechanisms of BHB may be through reducing neuroinflammation and protecting BBB.
Collapse
Affiliation(s)
- Zhitian Wang
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| | - Tian Li
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| | - Miaoyu Du
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| | - Lei Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| | - Linling Xu
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| | - Hao Song
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan, Hubei 430071, China.
| |
Collapse
|
43
|
Maternal Hyperhomocysteinemia Produces Memory Deficits Associated with Impairment of Long-Term Synaptic Plasticity in Young Rats. Cells 2022; 12:cells12010058. [PMID: 36611852 PMCID: PMC9818716 DOI: 10.3390/cells12010058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Maternal hyperhomocysteinemia (HCY) is a common pregnancy complication caused by high levels of the homocysteine in maternal and fetal blood, which leads to the alterations of the cognitive functions, including learning and memory. In the present study, we investigated the mechanisms of these alterations in a rat model of maternal HCY. The behavioral tests confirmed the memory impairments in young and adult rats following the prenatal HCY exposure. Field potential recordings in hippocampal slices demonstrated that the long-term potentiation (LTP) was significantly reduced in HCY rats. The whole-cell patch-clamp recordings in hippocampal slices demonstrated that the magnitude of NMDA receptor-mediated currents did not change while their desensitization decreased in HCY rats. No significant alterations of glutamate receptor subunit expression except GluN1 were detected in the hippocampus of HCY rats using the quantitative real-time PCR and Western blot methods. The immunofluorescence microscopy revealed that the number of synaptopodin-positive spines is reduced, while the analysis of the ultrastructure of hippocampus using the electron microscopy revealed the indications of delayed hippocampal maturation in young HCY rats. Thus, the obtained results suggest that maternal HCY disturbs the maturation of hippocampus during the first month of life, which disrupts LTP formation and causes memory impairments.
Collapse
|
44
|
Fei‐Sun Y, Huang M, Qin H, Campos de SouzaHan S, Xue H, Wang Y, Wang Y. Protective effect of isoflurane preconditioning on neurological function in rats with HIE. IBRAIN 2022; 8:500-515. [PMID: 37786586 PMCID: PMC10528772 DOI: 10.1002/ibra.12081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 10/04/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is an important cause of neonatal death and disability, which can lead to long-term neurological and motor dysfunction. Currently, inhalation anesthetics are widely used in surgery, and some studies have found that isoflurane (ISO) may have a positive effect on neuroprotection. In this paper, we investigated whether ISO pretreatment has a neuroprotective effect on the neurological function of HIE rats. Here, 7-day-old neonatal rats were randomly divided into a sham group, a hypoxic-ischemic (HI) group, and an ISO pretreatment (pretreatment) group. The pretreatment group was pretreated with 2% ISO for 1 h, followed by the HI group to establish an HI animal model. The HI‑induced neurological injury was evaluated by Zea‑Longa scores and triphenyltetrazolium (TTC) staining. Neuronal number and histomorphological changes were observed with Nissl staining and Hematoxylin-eosin (HE) staining. In addition, motor learning memory function was evaluated by the Morris water maze (MWM), the Y-maze, and the rotarod tests. HI induced severe neurological dysfunction, brain infarction, and cell apoptosis as well as obvious neuron loss in neonatal rats. In the MWM, the rats in the pretreatment group showed a decrease in escape latency (p = 0.042), indicating that pretreatment with ISO could improve the learning ability of HI rats. The results of Nissl staining showed that in the HI group, there was an irregular arrangement of neurons and nuclear fixation; however, the cell damage was significantly reduced and the total number of neurons was increased after ISO pretreatment (p < 0.001). In conclusion, ISO pretreatment improved cognitive function and attenuated HI-induced reduction of Nissl-positive cells and spatial memory impairment, suggesting that pretreatment with ISO before HI modeling could reduce neuronal cell death in the hippocampus after HI.
Collapse
Affiliation(s)
- Yi Fei‐Sun
- Institute of Neurological Disease, National‐Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China HospitalSichuan UniversityChengduSichuanChina
- Center for Epigenetics and Induced Pluripotent Stem Cells, Kennedy Krieger InstituteJohns Hopkins UniversityBaltimoreUSA
| | - Miao Huang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Hao‐Yue Qin
- Department of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Senio Campos de SouzaHan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Han Xue
- School of Basic Medical SciencesJinzhou Medical UniversityJinzhouLiaoningChina
| | - Yu‐Ying Wang
- School of Basic Medical SciencesJinzhou Medical UniversityJinzhouLiaoningChina
| | - Yi‐Bo Wang
- School of Basic Medical SciencesJinzhou Medical UniversityJinzhouLiaoningChina
| |
Collapse
|
45
|
Zhang J, Liu Y, Li H, Hu Y, Yu S, Liu Q, Chen Y. Stellate Ganglion Block Improves Postoperative Cognitive Dysfunction in aged rats by SIRT1-mediated White Matter Lesion Repair. Neurochem Res 2022; 47:3838-3853. [DOI: 10.1007/s11064-022-03800-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/13/2022] [Accepted: 10/16/2022] [Indexed: 12/04/2022]
|
46
|
Xu X, Feng Y, Wang J, Salvi R, Yin X, Gao J, Chen Y. Auditory-limbic-cerebellum interactions and cognitive impairments in noise-induced hearing loss. CNS Neurosci Ther 2022; 29:932-940. [PMID: 36377461 PMCID: PMC9928548 DOI: 10.1111/cns.14028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
AIMS This study aimed to explore the neural substrate of hearing loss-related central nervous system in rats and its correlation with cognition. METHODS We identified the neural mechanism for these debilitating abnormalities by inducing a bilateral hearing loss animal model using intense broadband noise (122 dB of broadband noise for 2 h) and used the Morris water maze test to characterize the behavioral changes at 6 months post-noise exposure. Functional magnetic resonance imaging (fMRI) was conducted to clarify disrupted functional network using bilateral auditory cortex (ACx) as a seed. Structural diffusion tensor imaging (DTI) was applied to illustrate characteristics of fibers in ACx and hippocampus. Pearson correlation was computed behavioral tests and other features. RESULTS A deficit in spatial learning/memory, body weight, and negative correlation between them was observed. Functional connectivity revealed weakened coupling within the ACx and inferior colliculus, lateral lemniscus, the primary motor cortex, the olfactory tubercle, hippocampus, and the paraflocculus lobe of the cerebellum. The fiber number and mean length of ACx and different hippocampal subregions were also damaged in hearing loss rats. CONCLUSION A new model of auditory-limbic-cerebellum interactions accounting for noise-induced hearing loss and cognitive impairments is proposed.
Collapse
Affiliation(s)
- Xiao‐Min Xu
- Department of Radiology, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Yuan Feng
- Department of Radiology, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Jian Wang
- School of Human Communication DisordersDalhousie UniversityHalifaxNova ScotiaCanada
| | - Richard Salvi
- Center for Hearing and DeafnessUniversity at BuffaloBuffaloNew YorkUSA
| | - Xindao Yin
- Department of Radiology, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Jun Gao
- The Department of Neurobiology, Key Laboratory of Human Functional Genomics of JiangsuNanjing Medical UniversityNanjingChina
| | - Yu‐Chen Chen
- Department of Radiology, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| |
Collapse
|
47
|
Bittencourt LO, Damasceno-Silva RD, Aragão WAB, Eiró-Quirino L, Oliveira ACA, Fernandes RM, Freire MAM, Cartágenes SC, Dionizio A, Buzalaf MAR, Cassoli JS, Cirovic A, Cirovic A, Maia CDSF, Lima RR. Global Proteomic Profile of Aluminum-Induced Hippocampal Impairments in Rats: Are Low Doses of Aluminum Really Safe? Int J Mol Sci 2022; 23:ijms232012523. [PMID: 36293377 PMCID: PMC9603961 DOI: 10.3390/ijms232012523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/12/2022] [Accepted: 09/29/2022] [Indexed: 11/21/2022] Open
Abstract
Hippocampus is the brain area where aluminum (Al) accumulates in abundance and is widely associated with learning and memory. In the present study, we evaluate behavioral, tissue, and proteomic changes in the hippocampus of Wistar rats caused by exposure to doses that mimic human consumption of aluminum chloride (AlCl3) in urban areas. For this, male Wistar rats were divided into two groups: Control (distilled water) and AlCl3 (8.3 mg/kg/day), both groups were exposed orally for 60 days. After the Al exposure protocol, cognitive functions were assessed by the Water maze test, followed by a collection for analysis of the global proteomic profile of the hippocampus by mass spectrometry. Aside from proteomic analysis, we performed a histological analysis of the hippocampus, to the determination of cell body density by cresyl violet staining in Cornu Ammonis fields (CA) 1 and 3, and hilus regions. Our results indicated that exposure to low doses of aluminum chloride triggered a decreased cognitive performance in learning and memory, being associated with the deregulation of proteins expression, mainly those related to the regulation of the cytoskeleton, cellular metabolism, mitochondrial activity, redox regulation, nervous system regulation, and synaptic signaling, reduced cell body density in CA1, CA3, and hilus.
Collapse
Affiliation(s)
- Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rakhel Dayanne Damasceno-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Luciana Eiró-Quirino
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Ana Carolina Alves Oliveira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Marco Aurelio M. Freire
- Graduate Program in Health and Society, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoro 59610210, Brazil
| | - Sabrina Carvalho Cartágenes
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru 05508060, Brazil
| | | | - Juliana Silva Cassoli
- Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Ana Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, 11000 Belgrade, Serbia
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
- Correspondence:
| |
Collapse
|
48
|
Wanga L, Zhang H, Xie C. Down-regulation of miR-204-5p ameliorates sevoflurane-induced brain injury in neonatal rats through targeting VCAM1. Toxicol Mech Methods 2022; 33:307-315. [PMID: 36177783 DOI: 10.1080/15376516.2022.2128705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
To confirm the regulation of miR-204-5p on VCAM1 and its effect on sevoflurane-induced brain injury in neonatal rats. We adopted the sevoflurane-induced brain injury model, and the double luciferase reporter gene assay was applied to explore the targeting relationship between vascular adhesion factor 1 (VCAM1) and miR-204-5p. RT-qPCR was applied to assess the levels of miR-204-5. VCAM1, LC3, P62 and cleaved-caspase 3 levels in the hippocampus were estimated by western blot. The number of autophagosomes in the cerebral cortex was assessed via Transmission electron microscopy (TEM), and histopathological changes within the hippocampus by HE staining. miR-204-5p levels were remarkably increased, but VCAM1 expression was decreased after neonatal rat brain injury. Furthermore, miR-204-5p directly targeted VCAM1. The escape latency, swimming distance, autophagosome number, neuronal apoptosis ratio, LC3 II and Cleaved-caspase 3 expression were reduced after miR-204-5p inhibition interference, whereas crossing times, and P62 expression increased in the sevoflurane-induced brain injury model. Furthermore down-regulation of VCAM1 reversed the trend of these indices. These results suggest that down-regulation of miR-204-5p ameliorates sevoflurane-induced cognitive impairment and hippocampal pathology and inhibits neuronal autophagy and apoptosis by targeting VCAM1.
Collapse
Affiliation(s)
- Lingang Wanga
- Department of Anesthesiology, The First People's Hospital of Xiaoshan District, Hangzhou City, Zhejiang 311200, China
| | - Hui Zhang
- Department of Anesthesiology, The First People's Hospital of Xiaoshan District, Hangzhou City, Zhejiang 311200, China
| | - Chenyao Xie
- Department of Anesthesiology, The First People's Hospital of Xiaoshan District, Hangzhou City, Zhejiang 311200, China
| |
Collapse
|
49
|
Xu J, Li Y, Tian B, Liu H, Wu S, Wang W. The effects and mechanism of environmental enrichment on MK-801 induced cognitive impairment in rodents with schizophrenia. Front Cell Neurosci 2022; 16:1024649. [PMID: 36246525 PMCID: PMC9556631 DOI: 10.3389/fncel.2022.1024649] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
Schizophrenia is a severe mental disorder characterized by positive, negative, and cognitive symptoms. Cognitive symptoms are a kind of symptoms with high incidence and great impact on patients. There is no effective treatment in clinical practice. N-methyl-d-aspartic acid (NMDA) receptor hypofunction may be an important cause of cognitive symptoms. MK-801 (also named Dizocilpine), a noncompetitive antagonist of NMDA receptor, is often used to construct a model of NMDA receptor dysfunction. In terms of treatment, environmental enrichment (EE) as an environmental intervention can effectively improve the symptoms of cognitive impairment in rodents. In this paper, we first briefly introduce the background of cognitive symptoms and EE in schizophrenia, and then investigate the manifestations of MK-801 induced cognitive impairment, the improvement of EE on these cognitive impairments based on the MK-801 induced schizophrenia rodent models, and the possible mechanism of EE in improving cognitive symptoms. This article reviews the literature in recent years, which provides an important reference for MK-801 to construct a cognitive symptom model of schizophrenia and the mechanism of EE in improving cognitive symptoms of schizophrenia.
Collapse
|
50
|
de Oliveira Marques C, Sesterheim P, Gayger Dias V, da Silva VF, Rodrigues L, Gonçalves CA. Hypothesizing that the intranasal administration of streptozotocin would be a valid model of Alzheimer’s disease-like dementia. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|