1
|
Zhang Z, Wang H, Tao B, Shi X, Chen G, Ma H, Peng R, Zhang J. Attenuation of Blood-Brain Barrier Disruption in Traumatic Brain Injury via Inhibition of NKCC1 Cotransporter: Insights into the NF-κB/NLRP3 Signaling Pathway. J Neurotrauma 2025; 42:814-831. [PMID: 39879999 DOI: 10.1089/neu.2023.0580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
Following traumatic brain injury (TBI), inhibition of the Na+-K+-Cl- cotransporter1 (NKCC1) has been observed to alleviate damage to the blood-brain barrier (BBB). However, the underlying mechanism for this effect remains unclear. This study aimed to investigate the mechanisms by which inhibiting the NKCC1 attenuates disruption of BBB integrity in TBI. The TBI model was induced in C57BL/6 mice through a controlled cortical impact device, and an in vitro BBB model was established using Transwell chambers. Western blot (WB) analysis was used to evaluate NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome and nuclear factor-kappaB (NF-κB) pathway proteins. Flow cytometry and transendothelial electrical resistance (TEER) were employed to assess endothelial cell apoptosis levels and BBB integrity. ELISA was utilized to measure cytokines interleukin-1β (IL-1β) and matrix metalloproteinase-9 (MMP-9). Immunofluorescence techniques were used to evaluate protein levels and the nuclear translocation of the rela (p65) subunit. The Evans blue dye leakage assay and the brain wet-dry weight method were utilized to assess BBB integrity and brain swelling. Inhibition of NKCC1 reduced the level of NLRP3 inflammasome and the secretion of IL-1β and MMP-9 in microglia. Additionally, NKCC1 inhibition suppressed the activation of the NF-κB signaling pathway, which in turn decreased the level of NLRP3 inflammasome. The presence of NLRP3 inflammasome in BV2 cells led to compromised BBB integrity within an inflammatory milieu. Following TBI, an upregulation of NLRP3 inflammasome was observed in microglia, astrocytes, vascular endothelial cells, and neurons. Furthermore, inhibiting NKCC1 resulted in a decrease in the positive rate of NLRP3 inflammasome in microglia and the levels of inflammatory cytokines IL-1β and MMP-9 after TBI, which correlated with BBB damage and the development of cerebral edema. These findings demonstrate that the suppression of the NKCC1 cotransporter protein alleviates BBB disruption through the NF-κB/NLRP3 signaling pathway following TBI.
Collapse
Affiliation(s)
- Zehan Zhang
- Department of Neurosurgery, PLA Air Force Hospital of Southern Theatre Command, Guangzhou, China
| | - Hui Wang
- Department of Experimental Pathology, Institute of Radiation Medicine, Beijing, China
| | - Bingyan Tao
- Department of Neurosurgery, 961th Hospital of Joint Logistics Support Force, Qiqihaer, China
| | - Xudong Shi
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Guilin Chen
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hengchao Ma
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ruiyun Peng
- Department of Experimental Pathology, Institute of Radiation Medicine, Beijing, China
| | - Jun Zhang
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Anton PE, Twardy S, Nagpal P, Moreno JA, Burchill MA, Chatterjee A, Busquet N, Mesches M, Kovacs EJ, McCullough RL. Suppression of NF-κB/NLRP3 by nanoligomer therapy mitigates ethanol and advanced age-related neuroinflammation. J Leukoc Biol 2025; 117:qiaf024. [PMID: 40036603 PMCID: PMC12022636 DOI: 10.1093/jleuko/qiaf024] [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: 12/15/2024] [Accepted: 02/02/2025] [Indexed: 03/06/2025] Open
Abstract
Binge alcohol use is increasing among aged adults (>65 yr). Alcohol-related toxicity in aged adults is associated with neurodegeneration; yet, the molecular underpinnings of this age-related sensitivity to alcohol are not well described. Studies utilizing rodent models of neurodegenerative disease reveal heightened activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Nod-like receptor 3 (NLRP3) mediate microglia activation and associated neuronal injury. Our group, and others, have implicated hippocampal-resident microglia as key producers of inflammatory mediators; yet, the link between inflammation and neurodegeneration has not been established in models of binge ethanol exposure and advanced age. Here, we report binge ethanol increased the proportion of NLRP3+ microglia in the hippocampus of aged (18 to 20 mo) female C57BL/6N mice compared with young (3 to 4 mo). In primary microglia, ethanol-induced expression of reactivity markers and NLRP3 inflammasome activation were more pronounced in microglia from aged mice compared with young. Using a NLRP3-specific inhibitor (OLT1177) and a novel brain-penetrant Nanoligomer that inhibits NF-κB and NLRP3 translation (SB_NI_112), we find ethanol-induced microglial reactivity can be attenuated by OLT1177 and SB_NI_112 in microglia from aged mice. In a model of intermittent binge ethanol exposure, SB_NI_112 prevented ethanol-mediated microglia reactivity, IL-1β production, and tau hyperphosphorylation in the hippocampus of aged mice. These data suggest early indicators of neurodegeneration occurring with advanced age and binge ethanol exposure are driven by NF-κB and NLRP3. Further investigation is warranted to explore the use of targeted immunosuppression via Nanoligomers to attenuate neuroinflammation after alcohol consumption in the aging populations.
Collapse
Affiliation(s)
- Paige E Anton
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Shannon Twardy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Matthew A Burchill
- GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | | - Nicolas Busquet
- Animal Behavior & In Vivo Neurophysiology Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Neurology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Michael Mesches
- Animal Behavior & In Vivo Neurophysiology Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Neurology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Elizabeth J Kovacs
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Division of GI Trauma and Endocrine Surgery, Department of Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Veterans' Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, United States
| | - Rebecca L McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
3
|
Zhu G, Zhang H, Xie R, Younis MR, Fu S, Wang X, Liu B, Li K, Lui S, Wu M. Monitoring Acidification Preceding Aβ Deposition in Alzheimer's Disease. Adv Healthc Mater 2025; 14:e2404907. [PMID: 40103521 DOI: 10.1002/adhm.202404907] [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: 12/06/2024] [Revised: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Amyloid beta (Aβ) is the primary early biomarker of Alzheimer's disease (AD), and since an acidic environment promotes Aβ aggregation, acidification plays a crucial role in AD progression. In this study, a novel acid-responsive near-infrared (NIR) fluorescent probe alongside multiple molecular biology techniques to investigate the temporal relationship between acidification and Aβ deposition, as well as the underlying mechanisms of acidification is employed. By monitoring 2- to 11-month-old APP/PS1 mice and wild-type (WT) mice, it is detected significant fluorescence signal in APP/PS1 mice beginning at 3 months preceding Aβ deposition at 5 months, and peaking at 5 months, followed by cognitive deficits at 8 months. Additionally, elevated monocarboxylate transporter 4 (MCT4) protein expression in 3-month-old APP/PS1 mice indicated disruption of astrocyte-neuron lactate shuttle (ANLS) homeostasis. Overall, this findings first demonstrate that acidification precedes Aβ deposition, peaks at the onset of Aβ deposition, and diminishes thereafter, with early acidification likely driven by the disruption of ANLS.
Collapse
Affiliation(s)
- Guannan Zhu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Hong Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Ruoxi Xie
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California, 90095, USA
| | - Shengxiang Fu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Xiaoze Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Beibei Liu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, No. 29, Wangjiang Road, Chengdu, 610064, China
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| |
Collapse
|
4
|
Liu C, Zhao Y, Zhang W, Dao JJ, Li Q, Huang J, Li ZF, Ma YK, Qiao CM, Cui C, Chen SX, Yu L, Shen YQ, Zhao WJ. Targeted activation of ErbB4 receptor ameliorates neuronal deficits and neuroinflammation in a food-borne polystyrene microplastic exposed mouse model. J Neuroinflammation 2025; 22:86. [PMID: 40089796 PMCID: PMC11910855 DOI: 10.1186/s12974-025-03406-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
The impact of polystyrene microplastics (PS-MPs) on the nervous system has been documented in the literature. Numerous studies have demonstrated that the activation of the epidermal growth factor receptor 4 (ErbB4) is crucial in neuronal injury and regeneration processes. This study investigated the role of targeted activation of ErbB4 receptor through a small molecule agonist, 4-bromo-1-hydroxy-2-naphthoic acid (C11H7BrO3, E4A), in mitigating PS-MPs-induced neuronal injury. The findings revealed that targeted activation of ErbB4 receptor significantly ameliorated cognitive behavioral deficits in mice exposed to PS-MPs. Furthermore, E4A treatment upregulated the expression of dedicator of cytokinesis 3 (DOCK3) and Sirtuin 3 (SIRT3) and mitigated mitochondrial and synaptic dysfunction within the hippocampus of PS-MPs-exposed mice. E4A also diminished the activation of the TLR4-NF-κB-NLRP3 signaling pathway, consequently reducing neuroinflammation. In vitro experiments demonstrated that E4A partially alleviated PS-MPs-induced hippocampal neuronal injury and its effects on microglial inflammation. In conclusion, the findings of this study indicate that targeted activation of ErbB4 receptor may mitigate neuronal damage and subsequent neuroinflammation, thereby alleviating hippocampal neuronal injury induced by PS-MPs exposure and ameliorating cognitive dysfunction. These results offer valuable insights for the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Chong Liu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yan Zhao
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang, Shandong, China
| | - Wei Zhang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, Guizhou, China
| | - Ji-Ji Dao
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Qian Li
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Jia Huang
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang, Shandong, China
| | - Zhen-Feng Li
- Experimental Center for Medical Research, Shandong Second Medical University, Weifang, Shandong, China
| | - Yu-Ke Ma
- Rehabilitation Therapy, Medical School, Weifang University of Science and Technology, Weifang, Shandong, China
| | - Chen-Meng Qiao
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chun Cui
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Shuang-Xi Chen
- The First Affiliated Hospital, Department of Neurology, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Yu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang, Shandong, China
| | - Yan-Qin Shen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei-Jiang Zhao
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
- Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Dadao, Binhu District, Wuxi, Jiangsu, 214122, P.R. China.
| |
Collapse
|
5
|
Zhang X, Zhang A, Li R, Jiao M, Wen R, Zheng R. Electroacupuncture regulates ferroptosis to improve postoperative cognitive dysfunction in mice through mediating GRX1/GSK-3β/Nrf2 axis. Brain Res Bull 2025; 222:111234. [PMID: 39889835 DOI: 10.1016/j.brainresbull.2025.111234] [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: 12/11/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The incidence of postoperative cognitive dysfunction (POCD) is higher happening in the elderly. It has been reported electroacupuncture (EA) was beneficial to the treatment of POCD, but its specific regulatory mechanism is still unclear. METHODS Through partial hepatectomy in mice, POCD model of mice was established. Baihui acupoint (GV20) was selected for targeted point of EA therapy. Morris water maze (MWM) was applied to evaluate cognitive impairment of mice. HE staining was used to examine cell arrangement and cell morphology in hippocampus of mice. RT-qPCR, western blot and IHC were employed to detect abundance of genes and proteins. MDA, GSH and iron levels was measured by some commercial kits. RESULTS Our findings revealed that partial hepatectomy surgery impaired learning and memory ability of mice, promoted ferroptosis. inhibited GRX1 and inactivated GSK-3β/Nrf2 pathway. However, EA therapy abolished these effects. In addition, GRX1 silencing and erastin abolished EA-mediated alterations of improving POCD in mice. CONCLUSION EA suppressed ferroptosis by regulating GRX1/GSK-3β/Nrf2 pathway to improve postoperative cognitive dysfunction of POCD mice.
Collapse
Affiliation(s)
- Xiaqing Zhang
- First Department of Anesthesiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, No.2 Weiyang West Road, Xianyang City, Shaanxi Province 712000, PR China
| | - Afen Zhang
- First Department of Anesthesiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, No.2 Weiyang West Road, Xianyang City, Shaanxi Province 712000, PR China
| | - Rui Li
- First Department of Anesthesiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, No.2 Weiyang West Road, Xianyang City, Shaanxi Province 712000, PR China
| | - Mingna Jiao
- First Department of Anesthesiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, No.2 Weiyang West Road, Xianyang City, Shaanxi Province 712000, PR China
| | - Rou Wen
- First Department of Anesthesiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, No.2 Weiyang West Road, Xianyang City, Shaanxi Province 712000, PR China
| | - Rongzhi Zheng
- First Department of Anesthesiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, No.2 Weiyang West Road, Xianyang City, Shaanxi Province 712000, PR China.
| |
Collapse
|
6
|
Putnam GL, Maitta RW. Alpha synuclein and inflammaging. Heliyon 2025; 11:e41981. [PMID: 39897785 PMCID: PMC11786851 DOI: 10.1016/j.heliyon.2025.e41981] [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: 02/16/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
The α-synuclein protein is an established molecule in Lewy body pathology, especially Parkinson's disease (PD). While the pathological role of α-synuclein (α-syn) in PD has been well described, novel evidence may suggest that α-syn interacts with inflammasomes in response to aging. As age is an inevitable physiological state and is also considered the greatest risk factor for PD, this calls for investigation into how α-syn, aging, and PD could be linked. There is a growing amount of data regarding α-syn normal function in the body that includes involvement in cellular transport such as protein complexes assembly, vesicular trafficking, neurotransmitter release, as well as immune cell maturation. Regarding abnormal α-syn, a number of autosomal dominant mutations have been identified as causes of familial PD, however, symptomatology may not become apparent until later in life due to compensatory mechanisms in the dopaminergic response. This potentially links age-related physiological changes not only as a risk factor for PD, but for the concept of "inflammaging ". This is defined as chronic inflammation that accompanies aging observed in many neurodegenerative pathologies, that include α-syn's ability to form oligomers and toxic fibrils seen in PD. This oligomeric α-syn stimulates pro-inflammatory signals, which may worsen PD symptoms and propagate chronic inflammation. Thus, this review will explore a potential link between α-syn's role in the immune system, inflammaging, and PD.
Collapse
Affiliation(s)
| | - Robert W. Maitta
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
7
|
Li B, Du L, Wu S, Yin Y. Protective effects of taurine on heat Stress-Induced cognitive impairment through Npas4 and Lcn2. Int Immunopharmacol 2024; 143:113376. [PMID: 39405930 DOI: 10.1016/j.intimp.2024.113376] [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: 06/15/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
Heat stress (HS) induces various pathophysiological responses in the brain, encompassing neuroinflammation and cognitive impairments. Although taurine has been reported to possess anti-inflammatory and cognitive-enhancing properties, its role and mechanisms in HS-induced cognitive impairment remain unclear. This study supplemented mice exposed to HS with taurine to assess its effect on cognitive function in a HS-induced mouse model. The results revealed that taurine ameliorated cognitive deficits following HS in mice and mitigated HS-induced astrocyte and microglia activation as well as blood-brain barrier (BBB) damage in the hippocampus. Mechanistically, Mechanistically, transcriptome sequencing was employed to identify that taurine regulates neuronal PAS domain protein (Npas4) and lipocalin 2 (Lcn2) during HS. Taurine was found to modulate hippocampal inflammation and influence cognitive function by upregulating Npas4 and downregulating Lcn2 after HS. Subsequently, molecular docking and AnimalTFDB database calculations were conducted, revealing that taurine might regulate the expression of Npas4 and Lcn2 by modulating the regulatory transcription factors (TFs) RE1 silencing transcription factor (REST) and nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1). Our findings demonstrate that taurine enhances the recovery of cognitive function through Npas4 and Lcn2 following HS, providing a theoretical basis for the clinical application of taurine in preventing or treating HS-induced cognitive impairment.
Collapse
Affiliation(s)
- Bin Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
8
|
Ji Y, Ma Y, Ma Y, Wang Y, Zhao X, Xu L, Ge S. An Amino Acids and Dipeptide Injection Inhibits the TNF-α/HMGB1 Inflammatory Signaling Pathway to Reduce Pyroptosis and M1 Microglial Polarization in POCD Mice: the Gut to the Brain. Mol Neurobiol 2024; 61:10097-10114. [PMID: 38700653 DOI: 10.1007/s12035-024-04209-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 04/27/2024] [Indexed: 11/24/2024]
Abstract
Peripheral surgery-induced neural inflammation is a key pathogenic mechanism of postoperative cognitive dysfunction (POCD). However, the mechanism underlying neuroinflammation and associated neural injury remains elusive. Surgery itself can lead to gut damage, and the occurrence of POCD is accompanied by high levels of TNF-α in the serum and blood‒brain barrier (BBB) damage. Reductions in stress, inflammation and protein loss have been emphasized as strategies for enhanced recovery after surgery (ERAS). We designed an amino acids and dipeptide (AAD) formula for injection that could provide intestinal protection during surgery. Through the intraoperative infusion of AAD based on the ERAS concept, we aimed to explore the effect of AAD injection on POCD and its underlying mechanism from the gut to the brain. Here, we observed that AAD injection ameliorated neural injury in POCD, in addition to restoring the function of the intestinal barrier and BBB. We also found that TNF-α levels decreased in the ileum, blood and hippocampus. Intestinal barrier protectors and TNF-α inhibitors also alleviated neural damage. AAD injection treatment decreased HMGB1 production, pyroptosis, and M1 microglial polarization and increased M2 polarization. In vitro, AAD injection protected the impaired gut barrier and decreased TNF-α production, alleviating damage to the BBB by stimulating cytokine transport in the body. HMGB1 and Caspase-1 inhibitors decreased pyroptosis and M1 microglial polarization and increased M2 polarization to protect TNF-α-stimulated microglia in vitro. Collectively, these findings suggest that the gut barrier-TNF-α-BBB-HMGB1-Caspase-1 inflammasome-pyroptosis-M1 microglia pathway is a novel mechanism of POCD related to the gut-brain axis and that intraoperative AAD infusion is a potential treatment for POCD.
Collapse
Affiliation(s)
- Yelong Ji
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yuanyuan Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yimei Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Ying Wang
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Xining Zhao
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Li Xu
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Shengjin Ge
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
9
|
Bu X, Guo H, Gao W, Zhang L, Hou J, Li B, Xia Z, Wang W. Neuroprotection of celastrol against postoperative cognitive dysfunction through dampening cGAS-STING signaling. Exp Neurol 2024; 382:114987. [PMID: 39369806 DOI: 10.1016/j.expneurol.2024.114987] [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: 07/14/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Neuroinflammation is a central player in postoperative cognitive dysfunction (POCD), an intractable and highly confounding neurological complication with finite therapeutic options. Celastrol, a quinone methide triterpenoid, is a bioactive ingredient extracted from Tripterygium wilfordii with talented anti-inflammatory capacity. However, it is unclear whether celastrol can prevent anesthesia/surgery-evoked cognitive deficits in an inflammation-specific manner. The STING agonist 5,6-dimethylxanthenone-4-acetic acid (DMXAA) was used to determine whether celastrol possesses neuroprotection dependent on the STING pathway in vivo and in vitro. Isoflurane and laparotomy triggered cGAS-STING activation, caspase-3/GSDME-dependent pyroptosis, and enhanced Iba-1 immunoreactivity. Celastrol improved cognitive performance and decreased the levels of cGAS, 2'3'-cGAMP, STING, NF-κB phosphorylation, Iba-1, TNF-α, IL-6, and IFN-β. Downregulation of cleaved caspase-3 and N-GSDME was observed in the hippocampus of POCD mice and HT22 cells after celastrol administration, accompanied by limited secretion of pyroptosis-pertinent pro-inflammatory cytokines IL-1β and IL-18. DMXAA neutralized the favorable influences of celastrol on cognitive function, as confirmed by the activation of the STING/caspase-3/GSDME axis. These findings implicate celastrol as a therapeutic agent for POCD through anti-inflammation and anti-pyroptosis.
Collapse
Affiliation(s)
- Xueshan Bu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Hui Guo
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, Hubei Province 430070, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Lei Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Bixi Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, Hubei Province 430070, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China.
| | - Wei Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China.
| |
Collapse
|
10
|
Bonfante S, Netto MB, de Oliveira Junior AN, Mathias K, Machado RS, Joaquim L, Cidreira T, da Silva MG, Daros GC, Danielski LG, Gava F, da Silva Lemos I, Matiola RT, Córneo E, Prophiro JS, de Bitencourt RM, Catalão CHR, da Silva Generoso J, Streck EL, Dal-Pizzol F, Barichello T, Petronilho F. Oxidative stress and mitochondrial dysfunction contributes to postoperative cognitive dysfunction in elderly rats dependent on NLRP3 activation. Metab Brain Dis 2024; 40:1. [PMID: 39535569 DOI: 10.1007/s11011-024-01425-5] [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: 10/25/2023] [Accepted: 08/25/2024] [Indexed: 11/16/2024]
Abstract
Postoperative cognitive dysfunction (POCD), a complication following procedures such as orthopedic surgery, is associated with a worsened prognosis, especially in the elderly population. Several mechanisms have been proposed for communication between the immune system and the brain after surgery. In an experimental tibial fracture (TF) model, we aimed to understand the role of the NLR family pyrin domain containing 3 (NLRP3) on oxidative stress and mitochondrial dysfunction as mechanisms underlying POCD in aged and adult rats. Adult or aged male Wistar rats were subjected to the TF model and received intracerebroventricular saline or MCC950 (140 ng/kg), a specific small-molecule inhibitor that selectively blocks activation of the NLRP3 inflammasome. We followed the control (sham) and TF groups treated with MCC950 or saline for seven days to determine cognitive function and survival. The prefrontal cortex and hippocampus were isolated for NLRP3 evaluation, cytokine analysis, oxidative stress measurements, myeloperoxidase activity, nitric oxide formation, mitochondrial respiratory chain enzymes, and succinate dehydrogenase (SDH) activity. Seven days after TF induction, NLRP3 levels increased in the hippocampus and prefrontal cortex in both ages, showed an enhancement in aged rats compared to adults, and experienced a reversal with MCC950 administration. The administration of MCC950 restored memory, IL-1β and IL-10 levels, nitrite/nitrate, lipid peroxidation in the hippocampus and prefrontal cortex, and preserved catalase activity in the prefrontal cortex in aged rats. At the same age, the complex I activity alteration in both regions and complex II, IV, and SDH in the prefrontal cortex were reversed. In conclusion, NLRP3 activation contributes to POCD development because it is intrinsically involved in mitochondrial dysfunction and oxidative stress after orthopedic surgery in aged rats.
Collapse
Affiliation(s)
- Sandra Bonfante
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | | | | | | | | | | | | | | | | | - Lucinéia Gainski Danielski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Fernanda Gava
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Isabela da Silva Lemos
- Laboratory of Neurometabolic Diseases, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Rafaela Tezza Matiola
- Laboratory of Neurometabolic Diseases, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Emily Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | | | | | - Carlos Henrique Rocha Catalão
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
- Faillace Department of Psychiatry and Behavioural Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Jaqueline da Silva Generoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Emílio Luiz Streck
- Laboratory of Neurometabolic Diseases, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Faillace Department of Psychiatry and Behavioural Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil.
- Laboratório de Neurobiologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciuma, SC, Brazil.
| |
Collapse
|
11
|
Lu J, Zhang Y, Hao Q, Zhou H, Zong Y. IDO-Kynurenine pathway mediates NLRP3 inflammasome activation-induced postoperative cognitive impairment in aged mice. Int J Neurosci 2024; 134:1309-1319. [PMID: 37746906 DOI: 10.1080/00207454.2023.2262741] [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: 06/26/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
AIM Postoperative cognitive dysfunction (POCD) is a common postoperative complication, especially in elderly patients. It extends hospital stay, increases the mortality rate and are heavy burdens to the family and society. Accumulating research has indicated that overactivation of pyrin domain-containing protein 3 (NLRP3) inflammasomes is related to POCD andplays a critical role in activating pro-inflammatory cytokines. According to existing studies, indoleamine 2,3-dioxygenase (IDO) is potently up-regulated by inflammatory factors, tryptophan in brain is mainly catalyzed by IDO to kynurenine (KYN), KYN metabolism may contribute to the development of depressive disorder and memory deficits. Hence, this study elucidated whether IDO-Kynurenine pathway mediates NLRP3 inflammasome activation-induced postoperative cognitive impairment in aged mice. MATERIAL AND METHODS POCD model was established in aged C57BL/6J mice by exploratory laparotomy under isoflurane anesthesia. Learning and memory were determined using Morris water maze. RESULTS The data showed that IDO and kynurenine aminotransferase-II (KAT-II) mRNA in hippocampus was up-regulated, and NLRP3, caspase recruitment domain (ASC), interleukin-1b (IL-1b) and IDO overexpressed, KYN levels increased after anesthesia and surgery. NLRP3 inflammasome inhibitor (MCC950) reversed NLRP3, ASC, IL-1b and IDO overexpression, and the elevation of KYN levels. To clarify the role of IDO-Kynurenine pathway in postoperative cognitive impairment, IDO inhibitor (1-methyl-Ltryptophan 1-MT) reduced the elevation of KYN and kynurenic acid (KYNA) levels, reduction of tryptophan (TRP), as well as improved learning and memory abilities. Finally, KAT-II inhibitor (PF-04859989) reduced brain KYNA levels and restored the cognitive impairment. CONCLUSION These results reveal that IDO-Kynurenine pathway mediates NLRP3 inflammasome activation-induced postoperative cognitive impairment.
Collapse
Affiliation(s)
- Jian Lu
- Department of Anesthesiology, the Second Hospital of Jiaxing, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Ye Zhang
- Department of Anesthesiology, the Second Hospital of Jiaxing, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Qian Hao
- Department of Anesthesiology, the Second Hospital of Jiaxing, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Hongmei Zhou
- Department of Anesthesiology, the Second Hospital of Jiaxing, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Youming Zong
- Department of Anesthesiology, the Second Hospital of Jiaxing, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| |
Collapse
|
12
|
Ying J, Deng X, Du R, Ding Q, Tian H, Lin Y, Zhou B, Gao W. Mitochondrial modulation treating postoperative cognitive dysfunction neuroprotection via DRP1 inhibition by Mdivi1. Sci Rep 2024; 14:26155. [PMID: 39478015 PMCID: PMC11525678 DOI: 10.1038/s41598-024-75548-1] [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: 05/11/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
This study investigated the role of mitochondrial dynamics in postoperative cognitive dysfunction (POCD) and assessed the therapeutic potential of mitochondrial modulation, particularly through the inhibition of dynamin-related protein 1 (DRP1) with Mdivi-1. Our findings indicated that DRP1 inhibition substantially mitigated neuroinflammation mediated by microglial cells, contributing to improved cognitive function in POCD models. The administration of Mdivi-1 led to a notable decrease in mitochondrial fission, reduced reactive oxygen species (ROS) production, and stabilization of mitochondrial membrane potential, all of which correlate with diminished neuroinflammation, as evidenced by lower NOD-like receptor family pyrin domain containing 3 (NLRP3)/ interleukin-1β (IL-1β) expression in microglial cells. Importantly, Mdivi-1 treatment was also found to enhance synaptic plasticity, increasing synaptic spine density in the hippocampal region of POCD mice. This improvement in mitochondrial health and synaptic integrity was paralleled by enhanced cognitive performance, as demonstrated in Y-maze tests. These results underscored the critical role of mitochondrial dynamics in the pathophysiology of POCD and suggested that targeting mitochondrial dysfunction, specifically through DRP1 inhibition, could be an effective approach for POCD treatment.
Collapse
Affiliation(s)
- Jun Ying
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaobing Deng
- Department of Anesthesiology, Lushan Rehabilitation and Recuperation Center, PLA Joint Logistics Support Force, Jiujiang, China
| | - Ruini Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiyang Ding
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Tian
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
13
|
Ma Z, Niu H, Qi H, Li Y. Functional role of lncRNA MEG3 on pyroptosis through interacting with EZH2 and YTHDC1 in postoperative cognitive dysfunction. Brain Res Bull 2024; 217:111060. [PMID: 39236791 DOI: 10.1016/j.brainresbull.2024.111060] [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: 02/29/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND The molecular biology mechanisms underlying postoperative cognitive dysfunction (POCD) remain unclear, resulting in a lack of specific therapeutic targets and limited clinical treatment options. The NLRP3 pyroptotic pathway, induced by neuroinflammation, is known to promote the development of POCD. Research has shown that lncRNA MEG3 exacerbates cell pyroptosis in various neurological injuries, though the precise mechanism remains to be investigated. METHODS In vitro and in vivo models of POCD were established through treatment with sevoflurane. Gene and protein expression were investigated using qRT-PCR, Western blot analysis, ELISA, and histological staining. Additionally, cell viability and injury were assessed through CCK-8 and LDH assays. Hippocampal-dependent memory and cognitive abilities were evaluated using the Morris Water Maze (MWM) test. Furthermore, the interactions between MEG3 and EZH2/YTHDC1 were validated through RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP). RESULTS Our findings reveal that sevoflurane significantly reduced MEG3 and pyroptosis-related proteins in mice. The overexpression of MEG3 protected mice against sevoflurane-induced cognitive dysfunction and reversed sevoflurane-induced pyroptosis in hippocampal neurons. MEG3 induced the downregulation of NLRP3 expression and reduced mRNA stability through its interaction with EZH2/YTHDC1. CONCLUSION In conclusion, our study elucidates that MEG3 inhibits the NLRP3 inflammasome and hippocampal neuron pyroptosis through the recruitment of EZH2/YTHDC1. These findings shed light on the underlying mechanism of MEG3 in the regulation of POCD and suggest that MEG3 could serve as a potential therapeutic target for the treatment of POCD.
Collapse
Affiliation(s)
- Zijian Ma
- Department of Anesthesiology, South area of the Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067055, China
| | - Haifei Niu
- Department of Ultrasound, the Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, China
| | - Haiqi Qi
- Department of Anesthesiology, South area of the Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067055, China
| | - Yan Li
- Department of Anesthesiology, South area of the Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067055, China.
| |
Collapse
|
14
|
Que M, Li S, Xia Q, Li X, Luo X, Zhan G, Luo A. Microbiota-gut-brain axis in perioperative neurocognitive and depressive disorders: Pathogenesis to treatment. Neurobiol Dis 2024; 200:106627. [PMID: 39111702 DOI: 10.1016/j.nbd.2024.106627] [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: 06/04/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
An increasing number of people undergo anesthesia and surgery. Perioperative neurocognitive and depressive disorders are common central nervous system complications with similar pathogeneses. These conditions pose a deleterious threat to human health and a significant societal burden. In recent years, numerous studies have focused on the role of the gut microbiota and its metabolites in the central nervous system via the gut-brain axis. Its involvement in perioperative neurocognitive and depressive disorders has attracted considerable attention. This review aimed to elucidate the role of the gut microbiota and its metabolites in the pathogenesis of perioperative neurocognitive and depressive disorders, as well as the value of targeted interventions and treatments.
Collapse
Affiliation(s)
- Mengxin Que
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaofeng Zhan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Xi X, Zhang R, Chi Y, Zhu Z, Sun R, Gong W. TXNIP Regulates NLRP3 Inflammasome-Induced Pyroptosis Related to Aging via cAMP/PKA and PI3K/Akt Signaling Pathways. Mol Neurobiol 2024; 61:8051-8068. [PMID: 38460079 DOI: 10.1007/s12035-024-04089-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/02/2024] [Indexed: 03/11/2024]
Abstract
Aging is an inevitable natural process with time-dependent dysfunction and the occurrence of various diseases, which impose heavy burdens on individuals, families, and society. It has been reported that NLRP3 inflammasome-induced pyroptosis contributes significantly to age-related diseases and aging, while TXNIP is suggested to be involved in regulating pyroptosis mediated by NLRP3. However, the mechanism between TXNIP and NLRP3 inflammasome is still unclear. In this study, we used HT-22 cells to explore the effect of TXNIP on pyroptosis and its potential association with the aging. Also, we delved into the underlying mechanisms. Our findings revealed that TXNIP significantly augmented pyroptosis in HT-22 cells, primarily by enhancing the activation of the NLRP3 inflammasome and promoting the release of proinflammatory cytokines. Remarkably, as TXNIP levels increased, we observed a corresponding rise in the number of p16-positive cells, which is indicative of aging. Furthermore, we conducted experiments to modulate the improvement of TXNIP on NLRP3 inflammasome-induced pyroptosis, that is, the PI3K activator 740 Y-P and the PKA activator DC2797 inhibited the effect, while the PI3K inhibitor LY294002 and the PKA inhibitor H89 enhanced the effect. In conclusion, our study demonstrated that TXNIP regulates NLRP3 inflammasome-induced pyroptosis in HT-22 cells related to aging via the PI3K/Akt and cAMP/PKA pathways.
Collapse
Affiliation(s)
- Xiaoshuang Xi
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Rong Zhang
- The Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yijia Chi
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Ziman Zhu
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Ruifeng Sun
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
16
|
Li M, Zhang R, Wu S, Cheng L, Fu H, Qu L. Isoflurane anesthesia decreases excitability of inhibitory neurons in the basolateral amygdala leading to anxiety‑like behavior in aged mice. Exp Ther Med 2024; 28:399. [PMID: 39171147 PMCID: PMC11336806 DOI: 10.3892/etm.2024.12688] [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: 03/13/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Anxiety after surgery can be a major factor leading to postoperative cognitive dysfunction, particularly in elderly patients. The role of inhibitory neurons in the basolateral amygdala (BLA) in anxiety-like behaviors in aged mice following isoflurane anesthesia remains unclear. Therefore, the present study aimed to investigate the role of inhibitory neurons in isoflurane-treated mice. A total of 30 C57BL/6 mice (age, 13 months) were allocated into the control and isoflurane anesthesia groups (15 mice/group) and were then subjected to several neurological assessments. Behavioral testing using an elevated plus maze test showed that aged mice in the isoflurane anesthesia group displayed significant anxiety-like behavior, since they spent more time in the closed arm, exhibited more wall climbing behavior and covered more distance. In addition, whole-cell patch-clamp recording revealed that the excitability of the BLA excitatory neurons was notably increased following mice anesthesia with isoflurane, while that of inhibitory neurons was markedly reduced. Following mice treatment with diazepam, the excitability of the BLA inhibitory neurons was notably increased compared with that of the excitatory neurons, which was significantly attenuated. Overall, the results of the current study indicated that anxiety-like behavior could occur in aged mice after isoflurane anesthesia, which could be caused by a reduced excitability of the inhibitory neurons in the BLA area. This process could enhance excitatory neuronal activity in aged mice, thus ultimately promoting the onset of anxiety-like behaviors.
Collapse
Affiliation(s)
- Mengyuan Li
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Ruijiao Zhang
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Shiyin Wu
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Liqin Cheng
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Huan Fu
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Liangchao Qu
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
- Department of Anesthesia and Surgery, People's Hospital of Ganjiang New District, Nanchang, Jiangxi 341099, P.R. China
| |
Collapse
|
17
|
Wang T, Sun G, Tao B. Updated insights into the NLRP3 inflammasome in postoperative cognitive dysfunction: emerging mechanisms and treatments. Front Aging Neurosci 2024; 16:1480502. [PMID: 39411285 PMCID: PMC11474915 DOI: 10.3389/fnagi.2024.1480502] [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: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) poses a significant threat to patients undergoing anesthesia and surgery, particularly elderly patients. It is characterized by diminished cognitive functions post surgery, such as impaired memory and decreased concentration. The potential risk factors for POCD include age, surgical trauma, anesthetic type, and overall health condition; however, the precise mechanisms underlying POCD remain elusive. Recent studies suggest that neuroinflammation might be a primary pathogenic factor. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes are implicated in exacerbating POCD by promoting the release of inflammatory factors and proteins that initiate pyroptosis, further influencing the disease process. The regulation of NLRP3 inflammasome activity, including its activation and degradation, is tightly controlled through multiple pathways and mechanisms. In addition, autophagy, a protective mechanism, regulates the NLRP3 inflammasome to control the progression of POCD. This review reviews recent findings on the role of the NLRP3 inflammasome in POCD pathogenesis and discusses therapeutic strategies aimed at reducing NLRP3 sources, inhibiting cellular pyroptosis, and enhancing autophagy.
Collapse
Affiliation(s)
| | | | - Bingdong Tao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Xue D, Guo X, Liu J, Li Y, Liu L, Liao G, Zhang M, Cao J, Liu Y, Lou J, Li H, Mi W, Wang L, Fu Q. Tryptophan-rich diet and its effects on Htr7 + Tregs in alleviating neuroinflammation and cognitive impairment induced by lipopolysaccharide. J Neuroinflammation 2024; 21:241. [PMID: 39334486 PMCID: PMC11437714 DOI: 10.1186/s12974-024-03239-9] [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: 01/23/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Neuroinflammation is a vital pathogenic mechanism for neurodegenerative diseases such as Alzheimer's, schizophrenia, and age-related cognitive decline. Regulatory T cells (Tregs) exhibit potent anti-inflammatory properties and can modulate neurodegenerative diseases arising from central nervous system inflammatory responses. However, the role of Tregs in neuroinflammation-related cognitive dysfunction remains unclear. It is highly plausible that Htr7+ Tregs expressing unique genes associated with the nervous system, including the Htr7 gene encoding the serotonin receptor 5-HT7, play a pivotal role. METHODS Mice were given a tryptophan-rich diet (with a tryptophan content of 0.6%) or a normal diet (with a tryptophan content of 0.16%). The neuroinflammation-mediated cognitive dysfunction model was established by intracerebroventricular injection of lipopolysaccharide (LPS) in 8-week-old C57BL/6J mice. The activation and infiltration of Tregs were measured using flow cytometry. Primary Tregs were cocultured separately with primary CD8+ T cells and primary microglia for in vitro validation of the impact of 5-HT and 5-HT7 receptor on Tregs. Prior to their transfer into recombination activating gene 1 (Rag1-/-) mice, Tregs were ex vivo transfected with lentivirus to knock down the expression of Htr7. RESULTS In this study, the tryptophan-rich diet was found to reverse LPS-induced cognitive impairment and reduce the levels of 5-HT in peripheral blood. The tryptophan-rich diet led to increased levels of 5-HT in peripheral blood, which in turn promoted the proliferation and activation of Htr7+ Tregs. Additionally, the tryptophan-rich diet was also shown to attenuate LPS-mediated neuroinflammation by activating Htr7+ Tregs. Furthermore, 5-HT and 5-HT7 receptor were found to enhance the immunosuppressive effect of Tregs on CD8+ T cells and microglia. In Rag1-/- mice, Htr7+ Tregs were shown to alleviate LPS-induced neuroinflammation and cognitive impairment. CONCLUSIONS Our research revealed the ability of Htr7+ Tregs to mitigate neuroinflammation and prevent neuronal damage by suppressing the infiltration of CD8+ T cells into the brain and excessive activation of microglia, thereby ameliorating LPS-induced cognitive impairment. These insights may offer novel therapeutic targets involving Tregs for neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Dinghao Xue
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xu Guo
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jingjing Liu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Anesthesiology, Chinese People's Armed Police Force Hospital of Beijing, Beijing, 100027, China
| | - Yanxiang Li
- Department of Anesthesiology, The 71st Group Army Hospital of CPLA Army, Xuzhou, 221004, China
| | - Luyu Liu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guosong Liao
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Mingru Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jiangbei Cao
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanhong Liu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jingsheng Lou
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hao Li
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weidong Mi
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Long Wang
- Department of Pain Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qiang Fu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
19
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
20
|
Bu X, Gong P, Zhang L, Song W, Hou J, Li Q, Wang W, Xia Z. Pharmacological inhibition of cGAS ameliorates postoperative cognitive dysfunction by suppressing caspase-3/GSDME-dependent pyroptosis. Neurochem Int 2024; 178:105788. [PMID: 38843953 DOI: 10.1016/j.neuint.2024.105788] [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: 05/15/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Neuroinflammation is a major driver of postoperative cognitive dysfunction (POCD). The cyclic GMP-AMP synthase-stimulator of interferon gene (cGAS-STING) signaling is a prominent alarming device for aberrant double-stranded DNA (dsDNA) that has emerged as a key mediator of neuroinflammation in cognitive-related diseases. However, the role of the cGAS-STING pathway in the pathogenesis of POCD remains unclear. A POCD model was developed in male C57BL/6J mice by laparotomy under isoflurane (Iso) anesthesia. The cGAS inhibitor RU.521 and caspase-3 agonist Raptinal were delivered by intraperitoneal administration. BV2 cells were exposed to Iso and lipopolysaccharide (LPS) in the absence or presence of RU.521, and then cocultured with HT22 cells in the absence or presence of Raptinal. Cognitive function was assessed using the Morris water maze test and novel object recognition test. Immunofluorescence assays were used to observe the colocalization of dsDNA and cGAS. The downstream proteins and pro-inflammatory cytokines were detected using the Western blot and enzyme-linked immunosorbent assay (ELISA). Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was used to assess the degree of cell death in the hippocampus following anesthesia/surgery treatment. Isoflurane/laparotomy and Iso + LPS significantly augmented the levels of cGAS in the hippocampus and BV2 cells, accompanied by mislocalized dsDNA accumulation in the cytoplasm. RU.521 alleviated cognitive impairment, diminished the levels of 2'3'-cGAMP, cGAS, STING, phosphorylated NF-κB p65 and NF-κB-pertinent pro-inflammatory cytokines (TNFα and IL-6), and repressed pyroptosis-associated elements containing cleaved caspase-3, N-GSDME, IL-1β and IL-18. These phenotypes could be rescued by Raptinal in vivo and in vitro. These findings suggest that pharmacological inhibition of cGAS mitigates neuroinflammatory burden of POCD by dampening caspase-3/GSDME-dependent pyroptosis, providing a potential therapeutic strategy for POCD.
Collapse
Affiliation(s)
- Xueshan Bu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ping Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Anesthesiology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Lei Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wenqin Song
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Qingwen Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
21
|
李 晋, 许 丽, 李 敏, 宋 怡, 张 静, 贾 龙. [Correlations between serum BDNF, IL-18 and hs-CRP levels in patients with acute cerebral infarction and vascular cognitive impairment]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2024; 56:708-714. [PMID: 39041569 PMCID: PMC11284472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Indexed: 07/24/2024]
Abstract
OBJECTIVE To explore the correlations between serum levels of brain-derived neurotrophic factor (BDNF), interleukin-18 (IL-18) and hypersensitivity C-reactive protein (hs-CRP) in patients with acute cerebral infarction and vascular cognitive impairment (VCI), and to provide some clinical bases for early prevention of VCI. METHODS A total of 160 patients with acute cerebral infarction admitted in Department of Neurology of Jincheng People' s Hospital from May 2019 to April 2020 were enrolled in this study and were devided into three groups according to whether or not combined with cognitive impairment, including no cognitive impairment group (NCI, 57 cases), vascular cognitive impairment no dementia group (VCIND, 56 cases) and vascular dementia group (VaD, 47 cases). The cognitive function of all the patients were evaluated by Montreal cognitive assessment (MoCA). The National Institute of Health stroke scale (NIHSS) was used to assess the degree of neurological deficit (mild-, moderate-, severe-neurologic deficit group). The infarct size was calculated by Pullicino' s method (small-, middle-, large-infarct group). The levels of serum BDNF and IL-18 were measured by enzyme-linked immunosorbent assay (ELISA), and serum levels of hs-CRP were measured by immunoturbidimetry during the acute phase (0-7 d), recovery period (15-30 d) and 6 months after cerebral infarction. The effects of varying degrees of neurological deficits and different size of infarction on BDNF, IL-18 and hs-CRP were observed. The levels of serum BDNF, IL-18 and hs-CRP in the patients of the three groups with acute, convalescent and six-month cerebral infarction were compared, and their correlations with VCI were analyzed. RESULTS Serum BDNF level and MoCA scores in mild-neurologic deficit group and small-infarct group were significantly higher than those in moderate- and severe-deficit group, middle- and large-infarct group, respectively (P < 0.05). Their levels of IL-18 and hs-CRP were significantly lower than those in moderate- and severe-deficit group, middle- and large-infarct group, respectively (P < 0.05). The levels of serum BDNF in NCI group, VCIND group and VaD group during the acute phase, convalescence and 6 months after cerebral infarction were in a significant decline, and the differences during the acute phase and recovery period were statistically significant (P < 0.05). The levels of IL-18 and hs-CRP during the acute phase, recovery period and 6 months after cerebral infarction showed a significant increasing trend with significance (P < 0.05). Correlation analysis revealed that the levels of BDNF was positively correlated with MoCA scores but negatively correlated with the severity of cognitive impairment while the expression levels of IL-18 and hs-CRP were negatively correlated with MoCA scores but positively correlated with the severity of cognitive impairment. CONCLUSION Serum BDNF, IL-18 and hs-CRP are involved in the pathological process of occurrence and development of VCI in the patients with acute cerebral infarction. BDNF has a protective effect on VCI while IL-18 and hs-CRP cause severe cognitive impairment. The levels of serum BDNF、IL-18 and hs-CRP in the patients with acute ischemic cerebral infarction are closely related to the severity of cognitive impairment and can be used as biomarkers of early diagnosis of VCI.
Collapse
Affiliation(s)
- 晋娜 李
- 晋城市人民医院神经内科,山西晋城 048000Department of Neurology, Jincheng People' s Hospital, Jincheng 048000, Shanxi, China
| | - 丽娜 许
- 晋城市人民医院神经内科,山西晋城 048000Department of Neurology, Jincheng People' s Hospital, Jincheng 048000, Shanxi, China
| | - 敏 李
- 晋城市人民医院神经内科,山西晋城 048000Department of Neurology, Jincheng People' s Hospital, Jincheng 048000, Shanxi, China
| | - 怡 宋
- 长治医学院研究生学院,山西长治 046000Graduate Institute of Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - 静 张
- 长治医学院研究生学院,山西长治 046000Graduate Institute of Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - 龙斌 贾
- 晋城市人民医院神经内科,山西晋城 048000Department of Neurology, Jincheng People' s Hospital, Jincheng 048000, Shanxi, China
| |
Collapse
|
22
|
Lin K, Peng F, He K, Qian Z, Mei X, Su Z, Wujimaiti Y, Xia X, Zhang T. Research progress on intestinal microbiota regulating cognitive function through the gut-brain axis. Neurol Sci 2024; 45:3711-3721. [PMID: 38632176 DOI: 10.1007/s10072-024-07525-5] [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: 11/28/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
The intestinal microbiota community is a fundamental component of the human body and plays a significant regulatory role in maintaining overall health and in the management disease states.The intestinal microbiota-gut-brain axis represents a vital connection in the cognitive regulation of the central nervous system by the intestinal microbiota.The impact of intestinal microbiota on cognitive function is hypothesized to manifest through both the nervous system and circulatory system. Imbalances in intestinal microbiota during the perioperative period could potentially contribute to perioperative neurocognitive dysfunction. This article concentrates on a review of existing literature to explore the potential influence of intestinal microbiota on brain and cognitive functions via the nervous and circulatory systems.Additionally, it summarizes recent findings on the impact of perioperative intestinal dysbacteriosis on perioperative neurocognitive dysfunction and suggests novel approaches for prevention and treatment of this condition.
Collapse
Affiliation(s)
- Kaijie Lin
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Feng Peng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China
| | - Kunyang He
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Zhengyu Qian
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xuan Mei
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Zhikun Su
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | | | - Xun Xia
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China.
- The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China.
| | - Tianyao Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China.
- The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Zhang X, Yang Y, Ma X, Cao H, Sun Y. Probiotics relieve perioperative postoperative cognitive dysfunction induced by cardiopulmonary bypass through the kynurenine metabolic pathway. Sci Rep 2024; 14:12822. [PMID: 38834581 DOI: 10.1038/s41598-024-59275-1] [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: 12/04/2023] [Accepted: 04/09/2024] [Indexed: 06/06/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) has become the popular critical post-operative consequences, especially cardiopulmonary bypass surgery, leading to an increased risk of mortality. However, no therapeutic effect about POCD. Probiotics are beneficial bacteria living in the gut and help to reduce the risk of POCD. However, the detailed mechanism is still not entirely known. Therefore, our research aims to uncover the effect and mechanism of probiotics in relieving POCD and to figure out the possible relationship between kynurenine metabolic pathway. 36 rats were grouped into three groups: sham operated group (S group, n = 12), Cardiopulmonary bypass group (CPB group, n = 12), and probiotics+CPB (P group, n = 12). After CPB model preparation, water maze test and Garcia score scale was performed to identify the neurological function. Immunofluorescence and Hematoxylin and eosin staining has been used for hippocampal neurons detection. Brain injury related proteins, oxidative stress factors, and inflammatory factors were detected using enzyme-linked immunosorbent assays (ELISA). Neuronal apoptosis was detected by TdT-mediated dUTP nick end-labeling (TUNEL) staining and western blot. High-performance liquid chromatography/mass spectrometry (HPLC/MS) was performed to detect the key factors of the kynurenine metabolic pathway. Our results demonstrated that probiotics improved neurological function of post-CPB rats. The administration of probiotics ameliorated memory and learning in spatial terms CPB rats (P < 0.05). Hematoxylin and eosin (H&E) staining data, S-100β and neuron-specific enolase (NSE) data convinced that probiotics agonists reduced brain damage in CPB rats (P < 0.05). Moreover, probiotics regulated inflammatory factors, meanwhile attenuated hippocampal neuronal apoptosis. Probiotics alleviated POCD in rats with CPB through regulation of kynurenine metabolic signaling pathway.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, Hubei, China
- Postgraduate Training Base, The General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, 121013, Liaoning, China
| | - Yanzhang Yang
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, 024000, Inner Mongolia, China
| | - Xinyi Ma
- Postgraduate Training Base, The General Hospital of Northern Theater Command, Dalian Medical University, Dalian, 116051, Liaoning, China
| | - Huijuan Cao
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China
| | - Yingjie Sun
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
24
|
Lin QC, Wang J, Wang XL, Pan C, Jin SW, Char S, Tao YX, Cao H, Li J. Hippocampal HDAC6 promotes POCD by regulating NLRP3-induced microglia pyroptosis via HSP90/HSP70 in aged mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167137. [PMID: 38527593 DOI: 10.1016/j.bbadis.2024.167137] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Postoperative Cognitive Dysfunction (POCD) has attracted increased attention, but its precise mechanism remains to be explored. This study aimed to figure out whether HDAC6 could regulate NLRP3-induced pyroptosis by modulating the functions of HSP70 and HSP90 in microglia to participate in postoperative cognitive dysfunction in aged mice. METHODS Animal models of postoperative cognitive dysfunction in aged mice were established by splenectomy under sevoflurane anesthesia. Morris water maze was used to examine the cognitive function and motor ability. Sixteen-months-old C57BL/6 male mice were randomly divided into six groups: control group (C group), sham surgery group (SA group), splenectomy group (S group), splenectomy + HDAC6 inhibitor ACY-1215 group (ACY group), splenectomy + HDAC6 inhibitor ACY-1215 + HSP70 inhibitor Apoptozole group (AP group), splenectomy + solvent control group (SC group). The serum and hippocampus of mice were taken after mice were executed. The protein levels of HDAC6, HSP90, HSP70, NLRP3, GSDMD-N, cleaved-Caspase-1 (P20), IL-1β were detected by western blotting. Serum IL-1β, IL-6 and S100β were measured using ELISA assay, and cell localization of HDAC6 was detected by immunofluorescence. In vitro experiments, BV2 cells were used to validate whether this mechanism worked in microglia. The protein levels of HDAC6, HSP90, HSP70, NLRP3, GSDMD-N, P20, IL-1β were detected by western blotting and the content of IL-1β in the supernatant was measured using ELISA assay. The degree of acetylation of HSP90, the interaction of HSP70, HSP90 and NLRP3 were analyzed by coimmunoprecipitation assay. RESULTS Splenectomy under sevoflurane anesthesia in aged mice could prolong the escape latency, reduce the number of crossing platforms, increase the expression of HDAC6 and activate the NLRP3 inflammasome to induce pyroptosis in hippocampus microglia. Using ACY-1215 could reduce the activation of NLRP3 inflammasome, the pyroptosis of microglia and the degree of spatial memory impairment. Apoptozole could inhibit the binding of HSP70 to NLRP3, reduce the degradation of NLRP3 and reverse the protective effect of HDAC6 inhibitors. The results acquired in vitro experiments closely resembled those in vivo, LPS stimulation led to the pyroptosis of BV2 microglia cells and the release of IL-1β due to the activation of the NLRP3 inflammasome, ACY-1215 showed the anti-inflammatory effect and Apoptozole exerted the opposite effect. CONCLUSIONS Our findings suggest that hippocampal HDAC6 promotes POCD by regulating NLRP3-induced microglia pyroptosis via HSP90/HSP70 in aged mice.
Collapse
Affiliation(s)
- Qi-Cheng Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiao Wang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xin-Lin Wang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chi Pan
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shao-Wu Jin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Steven Char
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Hong Cao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jun Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
25
|
Guan S, Li Y, Xin Y, Wang D, Lu P, Han F, Xu H. Deciphering the dual role of N-methyl-D-Aspartate receptor in postoperative cognitive dysfunction: A comprehensive review. Eur J Pharmacol 2024; 971:176520. [PMID: 38527701 DOI: 10.1016/j.ejphar.2024.176520] [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: 12/04/2023] [Revised: 03/03/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication following surgery, adversely impacting patients' recovery, increasing the risk of negative outcomes, prolonged hospitalization, and higher mortality rates. The N-methyl-D-aspartate (NMDA) receptor, crucial for learning, memory, and synaptic plasticity, plays a significant role in the development of POCD. Various perioperative factors, including age and anesthetic use, can reduce NMDA receptor function, while surgical stress, inflammation, and pain may lead to its excessive activation. This review consolidates preclinical and clinical research to explore the intricate relationship between perioperative factors affecting NMDA receptor functionality and the onset of POCD. It discusses the influence of aging, anesthetic administration, perioperative injury, pain, and inflammation on the NMDA receptor-related pathophysiology of POCD. The comprehensive analysis presented aims to identify effective treatment targets for POCD, contributing to the improvement of patient outcomes post-surgery.
Collapse
Affiliation(s)
- Shaodi Guan
- Department of Anesthesiology and Pain Medicine, 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, Wuhan, 430030, China
| | - Yali Li
- Department of Anesthesiology and Pain Medicine, 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, Wuhan, 430030, China
| | - Yueyang Xin
- Department of Anesthesiology and Pain Medicine, 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, Wuhan, 430030, China
| | - Danning Wang
- Department of Anesthesiology and Pain Medicine, 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, Wuhan, 430030, China
| | - Pei Lu
- Department of Anesthesiology and Pain Medicine, 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, Wuhan, 430030, China
| | - Fanglong Han
- Department of Anesthesiology, Xiangyang Maternal and Child Health Hospital, Xiangyang, 441003, China
| | - Hui Xu
- Department of Anesthesiology and Pain Medicine, 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, Wuhan, 430030, China.
| |
Collapse
|
26
|
Devlin BA, Nguyen DM, Grullon G, Clark MJ, Ceasrine AM, Deja M, Shah A, Ati S, Finn A, Ribeiro D, Schaefer A, Bilbo SD. Neuron Derived Cytokine Interleukin-34 Controls Developmental Microglia Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.589920. [PMID: 38766127 PMCID: PMC11100801 DOI: 10.1101/2024.05.10.589920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Neuron-microglia interactions dictate the development of neuronal circuits in the brain. However, the factors that support and broadly regulate these processes across developmental stages are largely unknown. Here, we find that IL34, a neuron-derived cytokine, is upregulated in development and plays a critical role in supporting and maintaining neuroprotective, mature microglia in the anterior cingulate cortex (ACC) of mice. We show that IL34 mRNA and protein is upregulated in neurons in the second week of postnatal life and that this increase coincides with increases in microglia number and expression of mature, homeostatic markers, e.g., TMEM119. We also found that IL34 mRNA is higher in more active neurons, and higher in excitatory (compared to inhibitory) neurons. Genetic KO of IL34 prevents the functional maturation of microglia and results in an anxiolytic phenotype in these mice by adulthood. Acute, low dose blocking of IL34 at postnatal day (P)15 in mice decreased microglial TMEM119 expression and increased aberrant microglial phagocytosis of thalamocortical synapses within the ACC. In contrast, viral overexpression of IL34 early in life (P1-P8) caused early maturation of microglia and prevented microglial phagocytosis of thalamocortical synapses during the appropriate neurodevelopmental refinement window. Taken together, these findings establish IL34 as a key regulator of neuron-microglia crosstalk in postnatal brain development, controlling both microglial maturation and synapse engulfment.
Collapse
|
27
|
Yang Y, Hang W, Li J, Liu T, Hu Y, Fang F, Yan D, McQuillan PM, Wang M, Hu Z. Effect of General Anesthetic Agents on Microglia. Aging Dis 2024; 15:1308-1328. [PMID: 37962460 PMCID: PMC11081156 DOI: 10.14336/ad.2023.1108] [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: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
The effects of general anesthetic agents (GAAs) on microglia and their potential neurotoxicity have attracted the attention of neuroscientists. Microglia play important roles in the inflammatory process and in neuromodulation of the central nervous system. Microglia-mediated neuroinflammation is a key mechanism of neurocognitive dysfunction during the perioperative period. Microglial activation by GAAs induces anti-inflammatory and pro-inflammatory effects in microglia, suggesting that GAAs play a dual role in the mechanism of postoperative cognitive dysfunction. Understanding of the mechanisms by which GAAs regulate microglia may help to reduce the incidence of postoperative adverse effects. Here, we review the actions of GAAs on microglia and the consequent changes in microglial function. We summarize clinical and animal studies associating microglia with general anesthesia and describe how GAAs interact with neurons via microglia to further explore the mechanisms of action of GAAs in the nervous system.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China.
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA.
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Patrick M. McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA.
| | - Mi Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
28
|
Li N, Ma Y, Li C, Sun M, Qi F. Dexmedetomidine alleviates sevoflurane-induced neuroinflammation and neurocognitive disorders by suppressing the P2X4R/NLRP3 pathway in aged mice. Int J Neurosci 2024; 134:511-521. [PMID: 36066545 DOI: 10.1080/00207454.2022.2121921] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Microglia-mediated inflammation is associated with perioperative neurocognitive disorders (PNDs) caused by sevoflurane. Dexmedetomidine has been reported to protect against sevoflurane-induced cognitive impairment. In this study, we investigated the effects and underlying mechanisms of dexmedetomidine on sevoflurane-induced microglial neuroinflammation and PNDs. METHODS Wild-type and purinergic ionotropic 4 receptor (P2X4R) overexpressing C57/BL6 mice were intraperitoneally injected with 20 μg/kg dexmedetomidine or an equal volume of normal saline 2 h prior to sevoflurane exposure. The Morris water maze (MWM) test was performed to assess cognitive function. Immunofluorescence staining was employed to detect microglial activation. The expression levels of proinflammatory cytokines were measured by real-time quantitative PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). The protein levels of P2X4R and NOD-like receptor protein 3 (NLRP3) were detected by Western Blotting. RESULTS Sevoflurane increased the number of microglia, upregulated the levels of proinflammatory cytokines, elevated the protein levels of P2X4R and NLRP3 in the hippocampus and induced cognitive decline, while pretreatment with dexmedetomidine downregulated the protein levels of P2X4R and NLRP3, alleviated sevoflurane-induced microglial neuroinflammation and improved cognitive dysfunction. Moreover, overexpression of P2X4R weakened the neuroprotective effect of dexmedetomidine. CONCLUSIONS Dexmedetomidine protected against sevoflurane-induced neuroinflammation and neurocognitive disorders by suppressing the P2X4R/NLRP3 pathway.
Collapse
Affiliation(s)
- Ning Li
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Yufeng Ma
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Chuangang Li
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Manyi Sun
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Feng Qi
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
29
|
Pei MQ, Xu LM, Yang YS, Chen WC, Chen XL, Fang YM, Lin S, He HF. Latest advances and clinical application prospects of resveratrol therapy for neurocognitive disorders. Brain Res 2024; 1830:148821. [PMID: 38401770 DOI: 10.1016/j.brainres.2024.148821] [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: 09/11/2023] [Revised: 01/13/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Neurocognitive disorders, such as Alzheimer's disease, vascular dementia, and postoperative cognitive dysfunction, are non-psychiatric brain syndromes in which a significant decline in cognitive function causes great trauma to the mental status of the patient. The lack of effective treatments for neurocognitive disorders imposes a considerable burden on society, including a substantial economic impact. Over the past few decades, the identification of resveratrol, a natural plant compound, has provided researchers with an opportunity to formulate novel strategies for the treatment of neurocognitive disorders. This is because resveratrol effectively protects the brain of those with neurocognitive disorders by targeting some mechanisms such as inflammation and oxidative stress. This article reviews the status of recent research investigating the use of resveratrol for the treatment of different neurocognitive disorders. By examining the possible mechanisms of action of resveratrol and the shared mechanisms of different neurocognitive disorders, treatments for neurocognitive disorders may be further clarified.
Collapse
Affiliation(s)
- Meng-Qin Pei
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Li-Ming Xu
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Yu-Shen Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Wei-Can Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Xin-Li Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Yu-Ming Fang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China
| | - Shu Lin
- Center of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China; Neuroendocrinology Group, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - He-Fan He
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, Fujian Province, China.
| |
Collapse
|
30
|
Zhang XJ, Wang Z, Chen JW, Yuan SY, Zhao L, Zhong JY, Chen JJ, Lin WJ, Wu WS. The neuroprotective effect of near infrared light therapy in aged mice with postoperative neurocognitive disorder by upregulating IRF7. J Affect Disord 2024; 349:297-309. [PMID: 38211750 DOI: 10.1016/j.jad.2024.01.074] [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: 10/04/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
BACKGROUND Postoperative neurocognitive disorder (PND) is a common central nervous system complication after undergoing surgery and anesthesia especially in elderly patients, while the therapeutic options are very limited. This study was carried out to investigate the beneficial effects of transcranial near infrared light (NIRL) which was employed to the treatment of PND and propose the involved mechanisms. METHODS The PND mice were established through left carotid artery exposure under isoflurane anesthesia and received transcranial NIRL treatment. Behavioral testing was performed to evaluate the cognitive function of PND mice after transcranial NIRL therapy. Changes in the transcriptomic profiles of prefrontal cortex (PFC) and hippocampus (HP) were identified by next generation sequencing (NGS), and the molecular mechanisms involved were examined by both in vivo mouse model and in vitro cell culture studies. RESULTS We found that transcranial NIRL therapy effectively ameliorated learning and memory deficit induced by anesthesia and surgery in aged mice. Specifically, we identified down-regulation of interferon regulatory factor 7 (IRF7) in the brains of PND mice that was mechanistically associated with increased pro-inflammatory M1 phenotype of microglia and elevated neuroinflammatory. NIRL treatment produced protective effects through the upregulation of IRF7 expression and reversing microglial phenotypes from pro-inflammatory to neuroprotective, resulting in reduced brain damage and improved cognitive function in PND mice. CONCLUSION Our results indicate that transcranial NIRL is an effective and safe therapy for PND via alleviating neuroinflammation, and IRF7 plays a key transcription factor in regulating the M1-to-M2 switch of microglia.
Collapse
Affiliation(s)
- Xiao-Jun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Jia-Wei Chen
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Shang-Yan Yuan
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Le Zhao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Jun-Ying Zhong
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Jun-Jun Chen
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Medical Research Center of Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wen-Si Wu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
31
|
Anton PE, Nagpal P, Moreno J, Burchill MA, Chatterjee A, Busquet N, Mesches M, Kovacs EJ, McCullough RL. NF-κB/NLRP3 Translational Inhibition by Nanoligomer Therapy Mitigates Ethanol and Advanced Age-Related Neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582114. [PMID: 38464118 PMCID: PMC10925165 DOI: 10.1101/2024.02.26.582114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Binge alcohol use is increasing among aged adults (>65 years). Alcohol-related toxicity in aged adults is associated with neurodegeneration, yet the molecular underpinnings of age-related sensitivity to alcohol are not well described. Studies utilizing rodent models of neurodegenerative disease reveal heightened activation of Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Nod like receptor 3 (NLRP3) mediate microglia activation and associated neuronal injury. Our group, and others, have implicated hippocampal-resident microglia as key producers of inflammatory mediators, yet the link between inflammation and neurodegeneration has not been established in models of binge ethanol exposure and advanced age. Here, we report binge ethanol increased the proportion of NLRP3+ microglia in the hippocampus of aged (18-20 months) female C57BL/6N mice compared to young (3-4 months). In primary microglia, ethanol-induced expression of reactivity markers and NLRP3 inflammasome activation were more pronounced in microglia from aged mice compared to young. Making use of an NLRP3-specific inhibitor (OLT1177) and a novel brain-penetrant Nanoligomer that inhibits NF-κB and NLRP3 translation (SB_NI_112), we find ethanol-induced microglial reactivity can be attenuated by OLT1177 and SB_NI_112 in microglia from aged mice. In a model of intermittent binge ethanol exposure, SB_NI_112 prevented ethanol-mediated microglia reactivity, IL-1β production, and tau hyperphosphorylation in the hippocampus of aged mice. These data suggest early indicators of neurodegeneration occurring with advanced age and binge ethanol exposure are NF-κB- and NLRP3-dependent. Further investigation is warranted to explore the use of targeted immunosuppression via Nanoligomers to attenuate neuroinflammation after alcohol consumption in the aged.
Collapse
Affiliation(s)
- Paige E. Anton
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Julie Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Matthew A. Burchill
- GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Nicolas Busquet
- Animal Behavior & In Vivo Neurophysiology Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Neurology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora Colorado
| | - Michael Mesches
- Animal Behavior & In Vivo Neurophysiology Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Neurology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora Colorado
| | - Elizabeth J. Kovacs
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of GI Trauma and Endocrine Surgery, Department of Surgery, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Veterans’ Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO
| | - Rebecca L. McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
- Alcohol Research Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
32
|
Lu J, Zong Y, Tao X, Dai H, Song J, Zhou H. Anesthesia/surgery-induced learning and memory dysfunction by inhibiting mitophagy-mediated NLRP3 inflammasome inactivation in aged mice. Exp Brain Res 2024; 242:417-427. [PMID: 38145993 PMCID: PMC10805997 DOI: 10.1007/s00221-023-06724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/15/2023] [Indexed: 12/27/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common postoperative complication, not only affects the quality of life of the elderly and increases the mortality rate, but also brings a greater burden to the family and society. Previous studies demonstrated that Nod-like receptor protein 3 (NLRP3) inflammasome participates in various inflammatory and neurodegenerative diseases. However, possible mitophagy mechanism in anesthesia/surgery-elicited NLRP3 inflammasome activation remains to be elucidated. Hence, this study clarified whether mitophagy dysfunction is related to anesthesia/surgery-elicited NLRP3 inflammasome activation. POCD model was established in aged C57BL/6 J mice by tibial fracture fixation under isoflurane anesthesia. Morris Water Maze (MWM) was used to evaluate learning and memory abilities. We found that in vitro experiments, lipopolysaccharide (LPS) significantly facilitated NLRP3 inflammasome activation and mitophagy inhibition in BV2 cells. Rapamycin restored mitophagy and improved mitochondrial function, and inhibited NLRP3 inflammasome activation induced by LPS. In vivo experiments, anesthesia and surgery caused upregulation of hippocampal NLRP3, caspase recruitment domain (ASC) and interleukin-1β (IL-1 β), and downregulation of microtubule-associated protein light chain 3II (LC3II) and Beclin1 in aged mice. Olaparib inhibited anesthesia/surgery-induced NLRP3, ASC, and IL-1β over-expression in the hippocampus, while upregulated the expression of LC3II and Beclin1. Furthermore, Olaparib improved cognitive impairment in older mice. These results revealed that mitophagy was involved in NLRP3 inflammasome-mediated anesthesia/surgery-induced cognitive deficits in aged mice. Overall, our results suggested that mitophagy was related in NLRP3 inflammasome-induced cognitive deficits after anesthesia and surgery in aged mice. Activating mitophagy may have clinical benefits in the prevention of cognitive impairment induced by anesthesia and surgery in elderly patients.
Collapse
Affiliation(s)
- Jian Lu
- Department of Anesthesiology, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Youming Zong
- Department of Anesthesiology, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Xiaoyan Tao
- Department of Nursing, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Hongyu Dai
- Department of Anesthesiology, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Jiale Song
- Department of Anesthesiology, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Hongmei Zhou
- Department of Anesthesiology, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China.
| |
Collapse
|
33
|
Ge Y, Yang J, Chen J, Dai M, Dou X, Yao S, Yao C, Lin Y. Absence in CX3CR1 receptor signaling promotes post-ischemic stroke cognitive function recovery through suppressed microglial pyroptosis in mice. CNS Neurosci Ther 2024; 30:e14551. [PMID: 38421089 PMCID: PMC10850801 DOI: 10.1111/cns.14551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Post-stroke cognitive impairment (PSCI) is a major source of morbidity and mortality after stroke, but the pathological mechanisms remain unclear. Previous studies have demonstrated that the CX3CR1 receptor plays a crucial role in maintaining an early protective microenvironment after stroke, but whether it persistently influences cognitive dysfunction in the chronic phase requires further investigation. METHODS Mouse was used to establish a middle cerebral artery occlusion (MCAO)/reperfusion model to study PSCI. Cognitive function was assessed by the Morris water maze (MWM) and the novel object recognition test. Neurogenesis was assessed by immunofluorescence staining with Nestin+ /Ki67+ and DCX+ /BrdU+ double-positive cells. The cerebral damage was monitored by [18 F]-DPA-714 positron emission tomography, Nissel, and TTC staining. The pyroptosis was histologically, biochemically, and electron microscopically examined. RESULTS Upon MCAO, at 28 to 35 days, CX3CR1 knockout (CX3CR1-/- ) mice had better cognitive behavioral performance both in MWM and novel object recognition test than their CX3CR1+/- counterparts. Upon MCAO, at 7 days, CX3CR1-/- mice increased the numbers of Nestin+ /Ki67+ and DCX+ /BrdU+ cells, and meanwhile it decreased the protein expression of GSDMD, NLRP3 inflammasome subunit, caspase-1, mature IL-1β/IL-18, and p-P65 in the hippocampus as compared with CX3CR1+/- mice. In addition, CX3CR1-/- mice could reverse infarct volume in the hippocampus region post-stroke. CONCLUSION Our study demonstrated that CX3CR1 gene deletion was beneficial to PSCI recovery. The mechanism might lie in inhibited pyroptosis and enhanced neurogenesis. CX3CR1 receptor may serve as a therapeutic target for improving the PSCI.
Collapse
Affiliation(s)
- Yangyang Ge
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jiayi Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoke Dou
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chenye Yao
- Department of Neurology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
34
|
Lee H, Kim SY, Lim Y. Solanum melongena extract supplementation protected skeletal muscle and brain damage by regulation of BDNF/PGC1α/irisin pathway via brain function-related myokines in high-fat diet induced obese mice. J Nutr Biochem 2024; 124:109537. [PMID: 38030047 DOI: 10.1016/j.jnutbio.2023.109537] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
In this study, we investigated the protective effects of SM on skeletal muscle and brain damage by regulation of BDNF/PGC1α/irisin pathway via brain function related myokines in high-fat diet-induced OB mice. OB was induced by high-fat diet for 6 weeks. SM extract (SME) was administered with 200 mg/kg BW (LSM) and 500 mg/kg BW (HSM) by oral gavage every day for 12 weeks. Behavior tests such as grip strength, Y-maze, and passive avoidance test were conducted to analyze muscle and cognitive function. Histopathological changes in skeletal muscle and brain were examined by hematoxylin and eosin staining and the protein levels of biomarkers related to oxidative stress, inflammation, protein degradation, neuro-plasticity, and cell cycling were measured by western blot. SME regulated morphological changes (muscle cross-sectional area: 1.23%, 1.40%; density of neurons in hippocampus:1.74%, 1.73%) in T2DM mice. Importantly, SME supplementation significantly increased several muscle-derived myokines which might influence the expression of neuronal markers in OB mice (FGF21: 1.27%, 1.34%; PGC1α: 1.0%, 1.32%; IRISIN: 1.9%, 1.08%; BDNF: 1.35%, 1.23%). Accordingly, SME activated hippocampal neurotrophic factors including BDNF (1.0%, 1.2%) and its associated PGC1α/irisin pathway (PGC1α :1.1%, 1.1%; IRISIN:1.1%, 0.9%) significantly. This study demonstrated the possibliy that protective myokines increased by SME supplementation may contribute to neuro-protection in OB mice. Taken together, the current study suggests that SME can be used to prevent skeletal muscle and brain damage in OB by protecting against oxidative stress and inflammatin via modulation of the BDNF/PGC1α/irisin pathway in the therapeutic approach of obese patients.
Collapse
Affiliation(s)
- Heaji Lee
- Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea
| | - Sun Yeou Kim
- Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-gu, Incheon, Republic of Korea
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Muscat SM, Butler MJ, Bettes MN, DeMarsh JW, Scaria EA, Deems NP, Barrientos RM. Post-operative cognitive dysfunction is exacerbated by high-fat diet via TLR4 and prevented by dietary DHA supplementation. Brain Behav Immun 2024; 116:385-401. [PMID: 38145855 PMCID: PMC10872288 DOI: 10.1016/j.bbi.2023.12.028] [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: 10/22/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023] Open
Abstract
Post-operative cognitive dysfunction (POCD) is an abrupt decline in neurocognitive function arising shortly after surgery and persisting for weeks to months, increasing the risk of dementia diagnosis. Advanced age, obesity, and comorbidities linked to high-fat diet (HFD) consumption such as diabetes and hypertension have been identified as risk factors for POCD, although underlying mechanisms remain unclear. We have previously shown that surgery alone, or 3-days of HFD can each evoke sufficient neuroinflammation to cause memory deficits in aged, but not young rats. The aim of the present study was to determine if HFD consumption before surgery would potentiate and prolong the subsequent neuroinflammatory response and memory deficits, and if so, to determine the extent to which these effects depend on activation of the innate immune receptor TLR4, which both insults are known to stimulate. Young-adult (3mo) & aged (24mo) male F344xBN F1 rats were fed standard chow or HFD for 3-days immediately before sham surgery or laparotomy. In aged rats, the combination of HFD and surgery caused persistent deficits in contextual memory and cued-fear memory, though it was determined that HFD alone was sufficient to cause the long-lasting cued-fear memory deficits. In young adult rats, HFD + surgery caused only cued-fear memory deficits. Elevated proinflammatory gene expression in the hippocampus of both young and aged rats that received HFD + surgery persisted for at least 3-weeks after surgery. In a separate experiment, rats were administered the TLR4-specific antagonist, LPS-RS, immediately before HFD onset, which ameliorated the HFD + surgery-associated neuroinflammation and memory deficits. Similarly, dietary DHA supplementation for 4 weeks prior to HFD onset blunted the neuroinflammatory response to surgery and prevented development of persistent memory deficits. These results suggest that HFD 1) increases risk of persistent POCD-associated memory impairments following surgery in male rats in 2) a TLR4-dependent manner, which 3) can be targeted by DHA supplementation to mitigate development of persistent POCD.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Emmanuel A Scaria
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Department of Psychiatry & Behavioral Health, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
36
|
Wang B, Chen SM, Yang SQ, Jiang JM, Zhang P, Zou W, Tang XQ. GDF11 mediates H 2S to prevent chronic stress-induced cognitive impairment by reducing hippocampal NLRP3/caspase-1-dependent pyroptosis. J Affect Disord 2024; 344:600-611. [PMID: 37827256 DOI: 10.1016/j.jad.2023.10.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/17/2023] [Accepted: 10/08/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND We previously revealed that hydrogen sulfide (H2S) attenuates chronic stress-induced cognitive impairment, but the underlying mechanism needs to be further clarified. Growth differentiation factor 11 (GDF11) plays an important regulatory role in cognitive function and that hippocampal NLRP3/caspase-1-mediated pyroptosis contributes to the pathogenesis of cognitive impairment. Hence, this research aimed to explore whether promoting GDF11 levels and suppressing hippocampal NLRP3/caspase-1-mediated pyroptosis mediate H2S to alleviate chronic stress-induced cognitive impairment. METHODS Sprague-Dawley rats were subjected to unpredictable chronic mild stress lasting four weeks to establish an animal model of chronic stress-induced cognitive impairment. Behavioral performance was assessed by the Y-maze test and the novel object recognition test. The expression levels of proteins were analyzed by Western blot analysis. The levels of IL-1β and IL-18 in the hippocampus were measured by ELISA. RESULTS NaHS upregulated the expression of GDF11 in the hippocampus of chronic unpredictable mild stress (CUMS)-exposed rats. Silencing GDF11 blocked NaHS-improved cognitive impairment in CUMS-exposed rats, according to the Y-maze test and the novel object recognition test. Furthermore, NaHS mitigated NLRP3/caspase-1-mediated pyroptosis in the hippocampus of CUMS-exposed rats and this effect was reversed by silencing GDF11. Moreover, overexpression of GDF11 alleviated CUMS-induced cognitive impairment and NLRP3/caspase-1-mediated hippocampal pyroptosis. CONCLUSIONS GDF11 mediates H2S to attenuate chronic stress-induced cognitive impairment via inhibiting hippocampal NLRP3/caspase-1-mediated pyroptosis.
Collapse
Affiliation(s)
- Bo Wang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China; The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Si-Min Chen
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - San-Qiao Yang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Jia-Mei Jiang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Ping Zhang
- The Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Wei Zou
- The Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China.
| | - Xiao-Qing Tang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China; Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China; The Second Affiliated Hospital, Institute of Cerebral Disease, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China.
| |
Collapse
|
37
|
Zhang S, Liu C, Sun J, Li Y, Lu J, Xiong X, Hu L, Zhao H, Zhou H. Bridging the Gap: Investigating the Link between Inflammasomes and Postoperative Cognitive Dysfunction. Aging Dis 2023; 14:1981-2002. [PMID: 37450925 PMCID: PMC10676784 DOI: 10.14336/ad.2023.0501] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 05/01/2023] [Indexed: 07/18/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a cluster of cognitive problems that may arise after surgery. POCD symptoms include memory loss, focus inattention, and communication difficulties. Inflammasomes, intracellular multiprotein complexes that control inflammation, may have a significant role in the development of POCD. It has been postulated that the NLRP3 inflammasome promotes cognitive impairment by triggering the inflammatory response in the brain. Nevertheless, there are many gaps in the current literature to understand the underlying pathophysiological mechanisms and develop future therapy. This review article underlines the limits of our current knowledge about the NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome and POCD. We first discuss inflammasomes and their types, structures, and functions, then summarize recent evidence of the NLRP3 inflammasome's involvement in POCD. Next, we propose a hypothesis that suggests the involvement of inflammasomes in multiple organs, including local surgical sites, blood circulation, and other peripheral organs, leading to systemic inflammation and subsequent neuronal dysfunction in the brain, resulting in POCD. Research directions are then discussed, including analyses of inflammasomes in more clinical POCD animal models and clinical trials, studies of inflammasome types that are involved in POCD, and investigations into whether inflammasomes occur at the surgical site, in circulating blood, and in peripheral organs. Finally, we discuss the potential benefits of using new technologies and approaches to study inflammasomes in POCD. A thorough investigation of inflammasomes in POCD might substantially affect clinical practice.
Collapse
Affiliation(s)
- Siyu Zhang
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, China.
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Joint Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Jintao Sun
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Yang Li
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Jian Lu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| | - Heng Zhao
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Joint Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Hongmei Zhou
- Anesthesiology Department, Zhejiang Chinese Medical University, Hangzhou, China.
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China.
| |
Collapse
|
38
|
Huang Y, Yang Y, Ye C, Liu Z, Wei F. The m 6A Reader YTHDF1 Improves Sevoflurane-Induced Neuronal Pyroptosis and Cognitive Dysfunction Through Augmenting CREB-BDNF Signaling. Neurochem Res 2023; 48:3625-3638. [PMID: 37572160 DOI: 10.1007/s11064-023-04007-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/12/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
Sevoflurane is one of the most widely used anesthetics in surgery which is the main cause of postoperative cognitive dysfunction (POCD). Previous reports confirmed that YTHDF1 is differently expressed in sevoflurane-induced POCD, while the roles and mechanistic details remain unclear. The molecular expressions were assessed using qRT-PCR, western blot, immunofluorescence and immunohistochemistry. Pathological change in the hippocampus tissues was analyzed using HE staining. Cognitive ability in rats was measured using MWM test. Hippocampal neuronal viability and apoptosis were measured by MTT assay and flow cytometry, respectively. The levels of pro-inflammatory cytokines were assessed using ELISA. The interaction between YTHDF1 and CREB was analyzed by RNA immunoprecipitation assay. YTHDF1 was significantly decreased in hippocampus tissues by sevoflurane exposure, and its overexpression could improve sevoflurane-induced neuron damage and cognitive dysfunction. Meanwhile, YTHDF1 upregulation repressed sevoflurane-induced activation of NLRP3 inflammation and pyroptosis in hippocampus tissues. Subsequently, YTHDF1 directly interacted to CREB mRNA to augment its stability and translation via a m6A-dependent manner, thus activating CREB/BDNF pathway. In addition, the inactivation of CREB/BDNF pathway could reverse the protective effects of YTHDF1 overexpression on sevoflurane-mediated neuronal damage and pyroptosis. These findings revealed that YTHDF1 improved sevoflurane-induced neuronal pyroptosis and cognitive dysfunction through activating CREB-BDNF signaling.
Collapse
Affiliation(s)
- Yuanlu Huang
- Department of Anesthesiology and Operation, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, No.1519 Dongyue Avenue, Nanchang, Jiangxi Province, 330052, P.R. China
| | - Yuxuan Yang
- Department of Anesthesiology and Operation, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, No.1519 Dongyue Avenue, Nanchang, Jiangxi Province, 330052, P.R. China
| | - Changsheng Ye
- Department of Anesthesiology and Operation, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, No.1519 Dongyue Avenue, Nanchang, Jiangxi Province, 330052, P.R. China
| | - Ziye Liu
- Department of Anesthesiology and Operation, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, No.1519 Dongyue Avenue, Nanchang, Jiangxi Province, 330052, P.R. China
| | - Fusheng Wei
- Department of Anesthesiology and Operation, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, No.1519 Dongyue Avenue, Nanchang, Jiangxi Province, 330052, P.R. China.
| |
Collapse
|
39
|
Xu Y, Yang Y, Chen X, Jiang D, Zhang F, Guo Y, Hu B, Xu G, Peng S, Wu L, Hu J. NLRP3 inflammasome in cognitive impairment and pharmacological properties of its inhibitors. Transl Neurodegener 2023; 12:49. [PMID: 37915104 PMCID: PMC10621314 DOI: 10.1186/s40035-023-00381-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
Cognitive impairment is a multifactorial and multi-step pathological process that places a heavy burden on patients and the society. Neuroinflammation is one of the main factors leading to cognitive impairment. The inflammasomes are multi-protein complexes that respond to various microorganisms and endogenous danger signals, helping to initiate innate protective responses in inflammatory diseases. NLRP3 inflammasomes produce proinflammatory cytokines (interleukin IL-1β and IL-18) by activating caspase-1. In this review, we comprehensively describe the structure and functions of the NLRP3 inflammasome. We also explore the intrinsic relationship between the NLRP3 inflammasome and cognitive impairment, which involves immune cell activation, cell apoptosis, oxidative stress, mitochondrial autophagy, and neuroinflammation. Finally, we describe NLRP3 inflammasome antagonists as targeted therapies to improve cognitive impairment.
Collapse
Affiliation(s)
- Yi Xu
- The Second Affiliated Hospital of Nanchang University, Department of the Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Yanling Yang
- The Second Affiliated Hospital of Nanchang University, Department of the Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Xi Chen
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Danling Jiang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fei Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yao Guo
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Bin Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Lidong Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Department of Thyroid and Hernia Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
40
|
Liu B, Zhu X, Zhou Q, Su Y, Qian Y, Ma Z, Gu X, Xia T. Activating ryanodine receptor improves isoflurane-induced cognitive dysfunction. Brain Res Bull 2023; 204:110790. [PMID: 37852420 DOI: 10.1016/j.brainresbull.2023.110790] [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: 07/30/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is characterized by impaired learning and memory. 6 h duration isoflurane anesthesia is an important factor to induce POCD, and the dysfunction of ryanodine receptor (RyR) in the hippocampus may be involved in this process. We investigated the expression of RyR3 in the hippocampus of mice after 6-h duration isoflurane anesthesia, as well as the improvement of RyR receptor agonist caffeine on POCD mice, while attempting to identify the underlying molecular mechanism. MATERIALS We constructed a POCD model using 8-week-old male C57BL/6J mice that were exposed to 6-h duration isoflurane. Prior to the three-day cognitive behavioral experiment, RyR agonist caffeine were injected. Fear conditioning and location memory tests were used in behavioral studies. We also exposed the mouse neuroblastoma cell line Neuro-2a (N2A) to 6-h duration isoflurane exposure to simulate the conditions of in vivo cognitive dysfunction. We administered ryanodine receptor agonist (caffeine) and inhibitor (ryanodine) to N2a cells. Following that, we performed a series of bioinformatics analysis to discover proteins that are involved in the development of cognitive dysfunction. Rt-PCR and Western blot were used to assess mRNA level and protein expression. RESULTS 6-h duration isoflurane anesthesia induced cognitive dysfunction and increased RyR3 mRNA levels in hippocampus. The mRNA levels of RyR3 in cultured N2a cells after anesthesia were comparable to those in vivo, and the RyR agonist caffeine corrected the expression of some cognitive-related phenotypic proteins that were disturbed after anesthesia. Intraperitoneal injection of RyR agonist caffeine can improve cognitive function after isoflurane anesthesia in mice, and bioinformatics analyses suggest that CaMKⅣ may be involved in the molecular mechanism. CONCLUSION Ryanodine receptor agonist caffeine may improve cognitive dysfunction in mice after isoflurane anesthesia.
Collapse
Affiliation(s)
- Binwen Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Xurui Zhu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Qingyun Zhou
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Yan Su
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Yue Qian
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
41
|
Liu Y, Yang W, Xue J, Chen J, Liu S, Zhang S, Zhang X, Gu X, Dong Y, Qiu P. Neuroinflammation: The central enabler of postoperative cognitive dysfunction. Biomed Pharmacother 2023; 167:115582. [PMID: 37748409 DOI: 10.1016/j.biopha.2023.115582] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Abstract
The proportion of advanced age patients undergoing surgical procedures is on the rise owing to advancements in surgical and anesthesia technologies as well as an overall aging population. As a complication of anesthesia and surgery, older patients frequently suffer from postoperative cognitive dysfunction (POCD), which may persist for weeks, months or even longer. POCD is a complex pathological process involving multiple pathogenic factors, and its mechanism is yet unclear. Potential theories include inflammation, deposition of pathogenic proteins, imbalance of neurotransmitters, and chronic stress. The identification, prevention, and treatment of POCD are still in the exploratory stages owing to the absence of standardized diagnostic criteria. Undoubtedly, comprehending the development of POCD remains crucial in overcoming the illness. Neuroinflammation is the leading hypothesis and a crucial component of the pathological network of POCD and may have complex interactions with other mechanisms. In this review, we discuss the possible ways in which surgery and anesthesia cause neuroinflammation and investigate the connection between neuroinflammation and the development of POCD. Understanding these mechanisms may likely ensure that future treatment options of POCD are more effective.
Collapse
Affiliation(s)
- Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning province, China
| | - Wei Yang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning province, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning province, China
| | - Juntong Chen
- Zhejiang University School of Medicine, Hangzhou 311121, Zhejiang province, China
| | - Shiqing Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Shijie Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Xiaohui Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning province, China.
| | - Youjing Dong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China.
| | - Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China.
| |
Collapse
|
42
|
Luo Y, Liu J, Hong Y, Peng S, Meng S. Sevoflurane-induced hypotension causes cognitive dysfunction and hippocampal inflammation in mice. Behav Brain Res 2023; 455:114672. [PMID: 37716552 DOI: 10.1016/j.bbr.2023.114672] [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: 05/24/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Abstract
Sevoflurane commonly adopted for anesthetic in clinical practice, however, its influences on cerebral blood flow and cognitive function remain controversial. Herein, the sevoflurane-induced hypotension on arterial blood pressure, cerebral blood flow, cognitive function, and hippocampal inflammation was investigated in mice. A significant decrease in arterial blood pressure and cerebral blood flow was indicated by the sevoflurane anesthesia treatment. Moreover, sevoflurane-induced hypotension was associated with the impaired cognitive function and the increased levels of NLRP3 inflammasome activation and oxidative stress in hippocampus. These findings suggest that sevoflurane-induced hypotension may lead to the cognitive dysfunction and hippocampal inflammation.
Collapse
Affiliation(s)
- Yuelian Luo
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiayi Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yu Hong
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shuling Peng
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Shiyu Meng
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
43
|
Cheon SY, Kim MY, Kim J, Kim EJ, Kam EH, Cho I, Koo BN, Kim SY. Hyperammonemia induces microglial NLRP3 inflammasome activation via mitochondrial oxidative stress in hepatic encephalopathy. Biomed J 2023; 46:100593. [PMID: 37059364 PMCID: PMC10498413 DOI: 10.1016/j.bj.2023.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/10/2023] [Accepted: 04/04/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND The role of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in the pathogenesis of hepatic encephalopathy (HE) is unclear. Mitochondrial reactive oxygen species (mtROS) is a signal for NLRP3 inflammasome activation. Therefore, we aimed to determine whether mtROS-dependent NLRP3 inflammasome activation is involved in HE, using in vivo and in vitro models. METHODS Bile duct ligation (BDL) in C57/BL6 mice was used as an in vivo HE model. NLRP3 activation was assessed in the hippocampus. Immunofluorescence staining was performed to determine the cellular source of NLRP3 in the hippocampal tissue. For the in vitro experiment, BV-2 microglial cells were primed with lipopolysaccharide (LPS), followed by ammonia treatment. NLRP3 activation and mitochondrial dysfunction were measured. Mito-TEMPO was used to suppress mtROS production. RESULTS BDL mice showed cognitive impairment with hyperammonemia. Both the priming and activation steps of NLRP3 inflammasome activation were processed in the hippocampus of BDL mice. Moreover, intracellular ROS levels increased in the hippocampus, and NLRP3 was mainly expressed in the microglia of the hippocampus. In LPS-primed BV-2 cells, ammonia treatment induced NLRP3 inflammasome activation and pyroptosis, with elevation of mtROS and altered mitochondrial membrane potential. Pretreatment with Mito-TEMPO suppressed mtROS production and the subsequent NLRP3 inflammasome activation and pyroptosis under LPS and ammonia treatment in BV-2 cells. CONCLUSIONS Hyperammonemia in HE may be involved in mtROS overproduction and subsequent NLRP3 inflammasome activation. Further studies using NLRP3-specific inhibitor or NLRP3 knockout mice are needed to elucidate the important role of NLRP3 inflammasome in HE development.
Collapse
Affiliation(s)
- So Yeong Cheon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - Min-Yu Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeongmin Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Kam
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Inja Cho
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - So Yeon Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Li X, Wang G, Li W, Wang X, Wu J, He Y, Li X, Sun X, Zhang M, Guo Y. Histone deacetylase 9 plays a role in sevoflurane-induced neuronal differentiation inhibition by inactivating cAMP-response element binding protein transcription and inhibiting the expression of neurotrophin-3. FASEB J 2023; 37:e23164. [PMID: 37688590 DOI: 10.1096/fj.202300168r] [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: 01/31/2023] [Revised: 07/02/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023]
Abstract
Postoperative cognitive decline (POCD) is a common and serious complication following anesthesia and surgery; however, the precise mechanisms of POCD remain unclear. Our previous research showed that sevoflurane impairs adult hippocampal neurogenesis (AHN) and thus cognitive function in the aged brain by affecting neurotrophin-3 (NT-3) expression; however, the signaling mechanism involved remains unexplored. In this study, we found a dramatic decrease in the proportion of differentiated neurons with increasing concentrations of sevoflurane, and the inhibition of neural stem cell differentiation was partially reversed after the administration of exogenous NT-3. Understanding the molecular underpinnings by which sevoflurane affects NT-3 is key to counteracting cognitive dysfunction. Here, we report that sevoflurane administration for 2 days resulted in upregulation of histone deacetylase 9 (HDAC9) expression, which led to transcriptional inactivation of cAMP-response element binding protein (CREB). Due to the colocalization of HDAC9 and CREB within cells, this may be related to the interaction between HDAC9 and CREB. Anyway, this ultimately led to reduced NT-3 expression and inhibition of neural stem cell differentiation. Furthermore, knockdown of HDAC9 rescued the transcriptional activation of CREB after sevoflurane exposure, while reversing the downregulation of NT-3 expression and inhibition of neural stem cell differentiation. In summary, this study identifies a unique mechanism by which sevoflurane can inhibit CREB transcription through HDAC9, and this process reduces NT-3 levels and ultimately inhibits neuronal differentiation. This finding may reveal a new strategy to prevent sevoflurane-induced neuronal dysfunction.
Collapse
Affiliation(s)
- Xinlei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Wei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xu Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Jiangnan Wu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yingxue He
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaowei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaobin Sun
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yanjing Guo
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
45
|
Wu H, Li D, Zhang T, Zhao G. Novel Mechanisms of Perioperative Neurocognitive Disorders: Ferroptosis and Pyroptosis. Neurochem Res 2023; 48:2969-2982. [PMID: 37289349 DOI: 10.1007/s11064-023-03963-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Perioperative neurocognitive disorders (PNDs) are some of the most common postoperative complications among the elderly and susceptible individuals, which significantly worsens the clinical outcome of patients. However, the prevention and treatment strategies of PNDs are difficult to determine and implement since the pathogenesis of PNDs is not well understood. The development of living organisms is associated with active and organized cell death, which is essential for maintaining the homeostasis of life. Ferroptosis is a programmed cell death (different from apoptosis and necrosis) mainly caused by an imbalance in the generation and degradation of intracellular lipid peroxides due to iron overload. Pyroptosis is an inflammatory cell death characterized by the creation of membrane holes mediated by the gasdermin (GSDM) family, followed by cell lysis and the release of pro-inflammatory cytokines. Ferroptosis and pyroptosis are involved in the pathogenesis of various central nervous system (CNS) diseases. Furthermore, ferroptosis and pyroptosis are closely associated with the occurrence and development of PNDs. This review summarizes the main regulatory mechanisms of ferroptosis and pyroptosis and the latest related to PNDs. Based on the available evidence, potential intervention strategies that can alleviate PNDs by inhibiting ferroptosis and pyroptosis have also been provided.
Collapse
Affiliation(s)
- Hang Wu
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China
| | - Dongmei Li
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China
| | - Te Zhang
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China
| | - Guoqing Zhao
- Department of Anaesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin, China.
- Jilin University, 2699 Forward Avenue, Changchun, Jilin, China.
| |
Collapse
|
46
|
Zeng X, Li J, Shan W, Lai Z, Zuo Z. Gut microbiota of old mice worsens neurological outcome after brain ischemia via increased valeric acid and IL-17 in the blood. MICROBIOME 2023; 11:204. [PMID: 37697393 PMCID: PMC10496352 DOI: 10.1186/s40168-023-01648-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/09/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND Aging is a significant risk factor for ischemic stroke and worsens its outcome. However, the mechanisms for this worsened neurological outcome with aging are not clearly defined. RESULTS Old C57BL/6J male mice (18 to 20 months old) had a poorer neurological outcome and more severe inflammation after transient focal brain ischemia than 8-week-old C57BL/6J male mice (young mice). Young mice with transplantation of old mouse gut microbiota had a worse neurological outcome, poorer survival curve, and more severe inflammation than young mice receiving young mouse gut microbiota transplantation. Old mice and young mice transplanted with old mouse gut microbiota had an increased level of blood valeric acid. Valeric acid worsened neurological outcome and heightened inflammatory response including blood interleukin-17 levels after brain ischemia. The increase of interleukin-17 caused by valeric acid was inhibited by a free fatty acid receptor 2 antagonist. Neutralizing interleukin-17 in the blood by its antibody improved neurological outcome and attenuated inflammatory response in mice with brain ischemia and receiving valeric acid. Old mice transplanted with young mouse feces had less body weight loss and better survival curve after brain ischemia than old mice transplanted with old mouse feces or old mice without fecal transplantation. CONCLUSIONS These results suggest that the gut microbiota-valeric acid-interleukin-17 pathway contributes to the aging-related changes in the outcome after focal brain ischemia and response to stimulus. Valeric acid may activate free fatty acid receptor 2 to increase interleukin-17.
Collapse
Affiliation(s)
- Xianzhang Zeng
- Department of Anesthesiology, University of Virginia, 1 Hospital Drive, PO Box 800710, Charlottesville, VA 22901 USA
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, Harbin, 150001 Heilongjiang People’s Republic of China
| | - Jun Li
- Department of Anesthesiology, University of Virginia, 1 Hospital Drive, PO Box 800710, Charlottesville, VA 22901 USA
| | - Weiran Shan
- Department of Anesthesiology, University of Virginia, 1 Hospital Drive, PO Box 800710, Charlottesville, VA 22901 USA
| | - Zhongmeng Lai
- Department of Anesthesiology, University of Virginia, 1 Hospital Drive, PO Box 800710, Charlottesville, VA 22901 USA
- Department of Anesthesiology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001 People’s Republic of China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, 1 Hospital Drive, PO Box 800710, Charlottesville, VA 22901 USA
- Departments of Neuroscience and Neurosurgery, University of Virginia, Charlottesville, VA 22901 USA
| |
Collapse
|
47
|
Zhang F, Xu J, Hu Y, Fang J, Yang M, Huang K, Xu W, He X. Diallyl trisulfide ameliorates bone loss and alters specific gut microbiota and serum metabolites in natural aging mice. Food Funct 2023; 14:7642-7653. [PMID: 37540026 DOI: 10.1039/d3fo01840g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Aging is a major cause of bone loss and osteoporosis. Diallyl trisulfide (DATS), one of the main organic sulfides in garlic oil, has been shown to alleviate arthritis in mice. However, further research is still needed to determine how DATS affects bone formation and bone loss in aging mice. Here, we established a mouse model of natural aging for dietary DATS intervention. DATS treatment improved the bone microstructure, including the disorganized arrangement of bone trabeculae and promoted collagen synthesis, as confirmed by micro-CT and histological analyses. The abundance of beneficial bacteria for bone formation, such as Clostridiaceae and Erysipelotrichaceae, and the microbial diversity and community richness were all altered by DATS, according to 16S rRNA sequencing data. 14 potential biomarkers and 9 important metabolic pathways were examined using serum metabolomics analysis. Additionally, there has been a significant reduction in sphingosine, which is directly associated with bone metabolism. The level of sphingosine and relative abundance of Clostridium were found to be negatively correlated by correlation analysis, indicating that bacteria may regulate bone reconstruction via influencing metabolites. Furthermore, Runx2 and β-catenin gene expression levels increased in bones, which may be related to the ameliorative mechanism of DATS. Our results suggested that DATS may prevent age-related bone loss by upregulating osteogenic gene expression through altering gut microbes and serum metabolism.
Collapse
Affiliation(s)
- Feng Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Jia Xu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Yanzhou Hu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Jingjing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
| | - Minglan Yang
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing, 100083, China
| | - Wentao Xu
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing, 100083, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100191, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering; China Agricultural University, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing, 100083, China
| |
Collapse
|
48
|
Zhuang Y, Xu J, Zheng K, Zhang H. Research progress of postoperative cognitive dysfunction in cardiac surgery under cardiopulmonary bypass. IBRAIN 2023; 10:290-304. [PMID: 39346790 PMCID: PMC11427806 DOI: 10.1002/ibra.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 10/01/2024]
Abstract
Cardiopulmonary bypass (CPB) is often used in cardiothoracic surgery because its nonphysiological state causes pathophysiological changes in the body, causing multiorgan and multitissue damage to varying degrees. Postoperative cognitive dysfunction (POCD) is a common central nervous system complication after cardiac surgery. The etiology and mechanism of POCD are not clear. Neuroinflammation, brain mitochondrial dysfunction, cerebral embolism, ischemia, hypoxia, and other factors are related to the pathogenesis of POCD. There is a close relationship between CPB and POCD, as CPB can cause inflammation, hypoxia and reperfusion injury, and microemboli formation, all of which can trigger POCD. POCD increases medical costs, seriously affects patients' quality of life, and increases mortality. Currently, there is a lack of effective treatment methods for POCD. Commonly used methods include preoperative health management, reducing inflammation response during surgery, preventing microemboli formation, and implementing individualized rehabilitation programs after surgery. Strengthening preventive measures can minimize the occurrence of POCD and its adverse effects.
Collapse
Affiliation(s)
- Yi‐Ming Zhuang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Ji‐Yang Xu
- Department of AnesthesiologyJudicial Police Hospital of Guizhou ProvinceGuiyangChina
| | - Kun Zheng
- Department of AnesthesiologyGuizhou Provincial People's HospitalGuiyangChina
| | - Hong Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
49
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
50
|
Tang L, Wang Y, Gong X, Xiang J, Zhang Y, Xiang Q, Li J. Integrated transcriptome and metabolome analysis to investigate the mechanism of intranasal insulin treatment in a rat model of vascular dementia. Front Pharmacol 2023; 14:1182803. [PMID: 37256231 PMCID: PMC10225696 DOI: 10.3389/fphar.2023.1182803] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Introduction: Insulin has an effect on neurodegenerative diseases. However, the role and mechanism of insulin in vascular dementia (VD) and its underlying mechanism are unknown. In this study, we aimed to investigate the effects and mechanism of insulin on VD. Methods: Experimental rats were randomly assigned to control (CK), Sham, VD, and insulin (INS) + VD groups. Insulin was administered by intranasal spray. Cognitive function was evaluated using the Morris's water maze. Nissl's staining and immunohistochemical staining were used to assess morphological alterations. Apoptosis was evaluated using TUNEL-staining. Transcriptome and metabolome analyses were performed to identify differentially expressed genes (DEGs) and differentially expressed metabolites (DEMs), respectively. Results: Insulin significantly improved cognitive and memory functions in VD model rats (p < 0.05). Compared with the VD group, the insulin + VD group exhibited significantly reduced the number of Nissl's bodies numbers, apoptosis level, GFAP-positive cell numbers, apoptosis rates, and p-tau and tau levels in the hippocampal CA1 region (p < 0.05). Transcriptomic analysis found 1,257 and 938 DEGs in the VD vs. CK and insulin + VD vs. VD comparisons, respectively. The DEGs were mainly enriched in calcium signaling, cAMP signaling, axon guidance, and glutamatergic synapse signaling pathways. In addition, metabolomic analysis identified 1 and 14 DEMs between groups in negative and positive modes, respectively. KEGG pathway analysis indicated that DEGs and DEMs were mostly enriched in metabolic pathway. Conclusion: Insulin could effectively improve cognitive function in VD model rats by downregulating tau and p-tau expression, inhibiting astrocyte inflammation and neuron apoptosis, and regulating genes involved in calcium signaling, cAMP signaling, axon guidance, and glutamatergic synapse pathways, as well as metabolites involved in metabolic pathway.
Collapse
Affiliation(s)
- Liang Tang
- Department of Basic Biology, Changsha Medical College, Changsha, China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, China
| | - Yan Wang
- Department of Basic Biology, Changsha Medical College, Changsha, China
| | - Xujing Gong
- Department of Basic Biology, Changsha Medical College, Changsha, China
| | - Ju Xiang
- Department of Basic Biology, Changsha Medical College, Changsha, China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, China
- School of Computer and Communication Engineering, Changsha University of Science and Technology, Changsha, China
| | - Yan Zhang
- Department of Basic Biology, Changsha Medical College, Changsha, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, China
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Qin Xiang
- Department of Basic Biology, Changsha Medical College, Changsha, China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, China
| | - Jianming Li
- Department of Basic Biology, Changsha Medical College, Changsha, China
- Center for Neuroscience and Behavior, Changsha Medical College, Changsha, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical College, Changsha, China
| |
Collapse
|