1
|
Chen ZH, Wu HT, Yao Z, Liu Q, Zhang HM, Li XC, Yao LQ, Yang X. Using ML techniques to predict extubation outcomes for patients with central nervous system injuries in the Yun-Gui Plateau. Sci Rep 2025; 15:17773. [PMID: 40404881 PMCID: PMC12098664 DOI: 10.1038/s41598-025-98861-9] [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: 11/29/2024] [Accepted: 04/15/2025] [Indexed: 05/24/2025] Open
Abstract
No predictive models have been reported for tracheostomy extubation success in plateau region rehabilitation departments. Hence, the primary objective of this retrospective study was to evaluate the predictive capabilities of different models for extubation outcomes in CNS injury patients in plateau rehabilitation departments, as well as investigate the influence of clinical features on these outcomes. Data were collected from 501 adult tracheostomy patients in the Department of Rehabilitation Medicine, including 196 successful extubations. Logistic regression was employed to identify the significant features linked to extubation outcomes from a pool of 31 variables. A total of eight independent models and a weighted posterior voting ensemble model were developed. Hyperparameter optimization and tenfold cross-validation were used to assist in choosing model parameters. Random forest (ACC = 84.15, AUC = 0.85), extra trees (83.17%, 0.87), K-NN (82.18%, 0.85), and gradient boosting (81.19%, 0.85) performed well. An ensemble model (85.15%, 0.87) combining random forest, Gaussian naive Bayes, and K-NN via the WPV method was developed. Dysphagia and low GCS scores have been linked to increased difficulty in extubation, as indicated by SHAP values and previous studies. Moreover, there could be a relationship between chronic inflammation and albumin levels in patients, which may collectively impact extubation success. This study evaluated the effectiveness of conventional models for predicting extubation outcomes and analyzed the factors influencing extubation results at high altitudes, laying the groundwork for clinical use and future research. Nevertheless, further research will see advantages in using multicentric approaches and broadening clinical indicators.
Collapse
Affiliation(s)
- Zi-Han Chen
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China
| | - Hao-Tian Wu
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China
| | - Zhou Yao
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China
| | - Qian Liu
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China
| | - Hong-Mei Zhang
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China
| | - Xiao-Chen Li
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China
| | - Li-Qing Yao
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China.
- Kunming Medical University Rehabilitation College, Chenggong District, Kunming, 650500, Yunnan, China.
| | - Xue Yang
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650000, Yunnan, China.
| |
Collapse
|
2
|
Chen A, Duan Y, Zhou S, Du F, Peng H, Zeng D, Wang J, Wu Y, Shi S, Li S, Yu S, Zhang J. Mesenchymal Stem Cells Restore Endothelial Integrity and Alleviate Emotional Impairments in a Diabetic Mouse Model via Inhibition of MMP-9 Activity. Int J Mol Sci 2025; 26:3355. [PMID: 40244194 PMCID: PMC11989596 DOI: 10.3390/ijms26073355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Diabetes mellitus (DM) has reached pandemic prevalence, significantly impacting global health. Accumulating evidence has highlighted a bidirectional relationship between diabetes and depression, with blood-brain barrier (BBB) disruption playing a pivotal role in the pathogenesis of and therapeutic approaches to both disorders. Mesenchymal stem cells (MSCs) have emerged as a promising cell-based therapeutic strategy for DM; however, their potential to mitigate DM-associated emotional deficits remains unclear. This study investigates whether MSCs can restore BBB integrity and improve emotional deficits in a diabetic mouse model via matrix metalloprotein-9 (MMP-9) inhibition. We used biochemical, molecular, and behavioral analyses to assess BBB function, inflammation, and emotional behavior. Our results demonstrated that diabetic conditions induce BBB dysfunction, characterized by the MMP-9-mediated degradation of tight junction (TJ) proteins claudin-5 (Cldn5) and occludin (Ocln), alongside neuroinflammation and emotional impairments. Notably, MSC administration restored BBB integrity and attenuated neuroinflammation by suppressing MMP-9 activity and upregulating TJ proteins. Importantly, MSC treatment not only alleviated anxiety- and depressive-like behaviors but also enhanced glycemic control in DMmodels. These findings elucidate the mechanistic basis of MSC therapy for DM-related neuropsychiatric complications and, crucially, reveal its dual therapeutic efficacy in concurrently ameliorating both neuropsychiatric symptoms and metabolic dysfunction in DM models. This synergistic therapeutic effect provides a translational rationale for advancing MSC-based therapies into clinical applications.
Collapse
Affiliation(s)
- Aoying Chen
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Yuhan Duan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Shaocong Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Fangzhou Du
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Huiyu Peng
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
| | - Dongao Zeng
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Jingwen Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Shuaiguang Shi
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Shikai Li
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
| | - Shuang Yu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Jingzhong Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (A.C.); (Y.D.); (S.Z.); (H.P.); (D.Z.); (S.S.); (S.L.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (F.D.); (J.W.); (Y.W.)
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
3
|
Li Z, Fu J, Jiang K, Gao J, Guo Y, Li C, Zhao L, Nam J, Gao H. Hyperbaric Oxygen Improves Cognitive Impairment Induced by Hypoxia via Upregulating the Expression of Oleic Acid and MBOAT2 of Mice. Antioxidants (Basel) 2024; 13:1320. [PMID: 39594462 PMCID: PMC11591255 DOI: 10.3390/antiox13111320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Cognitive impairment (CI) causes severe impairment of brain function and quality of life of patients, which brings a great burden to society. Cerebral hypoxia is an important factor in the pathogenesis of CI. Hyperbaric oxygen (HBO) therapy may mitigate hypoxia-induced CI, but its efficacy and mechanisms are not fully understood. In this study, a mice model of CI induced by hypoxia environment was established, then behavioral tests, pathological examination, metabolomic and lipidomic analyses, and molecular biology were used to assess the impact of HBO on hypoxia-induced CI. HBO was found to alleviate CI and pathological damage of hypoxia mice. Metabolomic, lipidomic, and molecular biology analyses showed that HBO increased the levels of oleic acid (OA) and membrane-bound O-acyltransferase 2 (MBOAT2), thereby altering the composition of membrane phospholipids (PLs) and reducing hypoxia-induced neuronal ferroptosis (FPT) to interfere with cognitive function in mice. In vitro experiments confirmed that OA and MBOAT2 led to membrane PL remodeling in a mutually dependent manner, affecting cell resistance to hypoxia-FPT. The results emphasized the combined effect value of OA and MBOAT2 in HBO for hypoxia-induced CI, and provided a novel perspective for the treatment of CI by HBO.
Collapse
Affiliation(s)
- Zhen Li
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jun Fu
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
- Innocation Academy of Testing Technology, Research and Experiment Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Kaiyuan Jiang
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Jie Gao
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Yuejun Guo
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Chen Li
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Liangcai Zhao
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hongchang Gao
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
- Innocation Academy of Testing Technology, Research and Experiment Center, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
4
|
Zhan L, Qiu M, Zheng J, Lai M, Lin K, Dai J, Sun W, Xu E. Fractalkine/CX3CR1 axis is critical for neuroprotection induced by hypoxic postconditioning against cerebral ischemic injury. Cell Commun Signal 2024; 22:457. [PMID: 39327578 PMCID: PMC11426015 DOI: 10.1186/s12964-024-01830-4] [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: 04/29/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Microglial activation-mediated neuroinflammation is a major contributor to neuronal damage after cerebral ischemia. The Fractalkine (FKN)/CX3C chemokine receptor 1 (CX3CR1) axis plays a critical role in regulating microglial activation and neuroinflammation. The aim of this study is to ascertain the role and mechanism of FKN/CX3CR1 axis in hypoxic postconditioning (HPC)-induced anti-inflammatory and neuroprotective effects on transient global cerebral ischemia (tGCI). We found that HPC suppressed microglial activation and alleviated neuroinflammation in hippocampal CA1 after tGCI. Meanwhile, HPC upregulated the expression of FKN and CX3CR1 in neurons, but it downregulated the expression of CX3CR1 in glial cells after tGCI. In addition, the overexpression of FKN induced by the administration of FKN-carried lentivirus reduced microglial activation and inhibited neuroinflammation in CA1 after tGCI. Furthermore, silencing CX3CR1 with CX3CRi-carried lentivirus in CA1 after tGCI suppressed microglial activation and neuroinflammation and exerted neuroprotective effects. Finally, the overexpression of FKN caused a marked increase of neuronal CX3CR1 receptors, upregulated the phosphorylation of Akt, and reduced neuronal loss of rats in CA1 after tGCI. These findings demonstrated that HPC protected against neuronal damage in CA1 of tGCI rats through inhibiting microglial activation and activating Akt signaling pathway via FKN/CX3CR1 axis.
Collapse
Affiliation(s)
- Lixuan Zhan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Meiqian Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Jianhua Zheng
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Meijing Lai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Kunqin Lin
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Jiahua Dai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Weiwen Sun
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - En Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China.
| |
Collapse
|
5
|
Cui C, Jiang X, Wang Y, Li C, Lin Z, Wei Y, Ni Q. Cerebral Hypoxia-Induced Molecular Alterations and Their Impact on the Physiology of Neurons and Dendritic Spines: A Comprehensive Review. Cell Mol Neurobiol 2024; 44:58. [PMID: 39105862 PMCID: PMC11303443 DOI: 10.1007/s10571-024-01491-4] [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: 05/28/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
This article comprehensively reviews how cerebral hypoxia impacts the physiological state of neurons and dendritic spines through a series of molecular changes, and explores the causal relationship between these changes and neuronal functional impairment. As a severe pathological condition, cerebral hypoxia can significantly alter the morphology and function of neurons and dendritic spines. Specifically, dendritic spines, being the critical structures for neurons to receive information, undergo changes such as a reduction in number and morphological abnormalities under hypoxic conditions. These alterations further affect synaptic function, leading to neurotransmission disorders. This article delves into the roles of molecular pathways like MAPK, AMPA receptors, NMDA receptors, and BDNF in the hypoxia-induced changes in neurons and dendritic spines, and outlines current treatment strategies. Neurons are particularly sensitive to cerebral hypoxia, with their apical dendrites being vulnerable to damage, thereby affecting cognitive function. Additionally, astrocytes and microglia play an indispensable role in protecting neuronal and synaptic structures, regulating their normal functions, and contributing to the repair process following injury. These studies not only contribute to understanding the pathogenesis of related neurological diseases but also provide important insights for developing novel therapeutic strategies. Future research should further focus on the dynamic changes in neurons and dendritic spines under hypoxic conditions and their intrinsic connections with cognitive function.
Collapse
Affiliation(s)
- Chao Cui
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Xue Jiang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Yumei Wang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Chao Li
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Youzhen Wei
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China.
- Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200000, China.
| | - Qingbin Ni
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China.
| |
Collapse
|
6
|
Ni W, Niu Y, Cao S, Fan C, Fan J, Zhu L, Wang X. Intermittent hypoxia exacerbates anxiety in high-fat diet-induced diabetic mice by inhibiting TREM2-regulated IFNAR1 signaling. J Neuroinflammation 2024; 21:166. [PMID: 38956653 PMCID: PMC11218348 DOI: 10.1186/s12974-024-03160-1] [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: 03/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.
Collapse
MESH Headings
- Animals
- Mice
- Diet, High-Fat/adverse effects
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Anxiety/etiology
- Anxiety/metabolism
- Signal Transduction/physiology
- Signal Transduction/drug effects
- Hypoxia/metabolism
- Hypoxia/complications
- Male
- Mice, Inbred C57BL
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/psychology
- Receptor, Interferon alpha-beta/metabolism
- Receptor, Interferon alpha-beta/genetics
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Microglia/metabolism
- STAT1 Transcription Factor/metabolism
- Sleep Apnea, Obstructive/complications
- Sleep Apnea, Obstructive/metabolism
- Sleep Apnea, Obstructive/psychology
Collapse
Affiliation(s)
- Wenyu Ni
- Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Qidong Liver Cancer Institute, No.9, Seyuan Road, Chongchuan District, Nantong, Jiangsu, 226000, China
| | - Yun Niu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Sitong Cao
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chunsun Fan
- Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Qidong Liver Cancer Institute, No.9, Seyuan Road, Chongchuan District, Nantong, Jiangsu, 226000, China
| | - Jian Fan
- Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Qidong Liver Cancer Institute, No.9, Seyuan Road, Chongchuan District, Nantong, Jiangsu, 226000, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Xueting Wang
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Medical Research Center Affiliated Hospital 2 of Nantong University, Nantong, China.
| |
Collapse
|
7
|
Chen J, Wei X, Wu X, Zhang Q, Xia G, Xia H, Shang H, Lin S. Disorder of neuroplasticity aggravates cognitive impairment via neuroinflammation associated with intestinal flora dysbiosis in chronic heart failure. Aging (Albany NY) 2024; 16:10882-10904. [PMID: 38968172 PMCID: PMC11272129 DOI: 10.18632/aging.205960] [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: 12/12/2023] [Accepted: 03/28/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Chronic heart failure (CHF) impairs cognitive function, yet its effects on brain structure and underlying mechanisms remain elusive. This study aims to explore the mechanisms behind cognitive impairment. METHODS CHF models in rats were induced by ligation of the left anterior descending coronary artery. Cardiac function was analyzed by cardiac ultrasound and hemodynamics. ELISA, immunofluorescence, Western blot, Golgi staining and transmission electron microscopy were performed on hippocampal tissues. The alterations of intestinal flora under the morbid state were investigated via 16S rRNA sequencing. The connection between neuroinflammation and synapses is confirmed by a co-culture system of BV2 microglia and HT22 cells in vitro. Results: CHF rats exhibited deteriorated cognitive behaviors. CHF induced neuronal structural disruption, loss of Nissl bodies, and synaptic damage, exhibiting alterations in multiple parameters. CHF rats showed increased hippocampal levels of inflammatory cytokines and activated microglia and astrocytes. Furthermore, the study highlights dysregulated PDE4-dependent cAMP signaling and intestinal flora dysbiosis, closely associated with neuroinflammation, and altered synaptic proteins. In vitro, microglial neuroinflammation impaired synaptic plasticity via PDE4-dependent cAMP signaling. CONCLUSIONS Neuroinflammation worsens CHF-related cognitive impairment through neuroplasticity disorder, tied to intestinal flora dysbiosis. PDE4 emerges as a potential therapeutic target. These findings provide insightful perspectives on the heart-gut-brain axis.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaohong Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xuefen Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qian Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Guiyang Xia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Huan Xia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Sheng Lin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
8
|
Huang JM, Zhao N, Hao XN, Li SY, Wei D, Pu N, Peng GH, Tao Y. CX3CL1/CX3CR1 Signaling Mediated Neuroglia Activation Is Implicated in the Retinal Degeneration: A Potential Therapeutic Target to Prevent Photoreceptor Death. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 38231527 PMCID: PMC10795588 DOI: 10.1167/iovs.65.1.29] [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: 09/08/2023] [Accepted: 12/17/2023] [Indexed: 01/18/2024] Open
Abstract
Purpose Retinal degeneration (RD) is a large cluster of retinopathies that is characterized by the progressive photoreceptor death and visual impairments. CX3CL1/CX3CR1 signaling has been documented to mediate the microglia activation and gliosis reaction during neurodegeneration. We intend to verify whether the CX3CL1/CX3CR1 signaling is involved in the RD pathology. Methods A pharmacologically induced RD mice model was established. AZD8797, a CX3CR1 antagonist, was injected into the vitreous cavity of an RD model to modulate the neuroglia activation. Then, the experimental animals were subjected to functional, morphological, and behavioral analysis. Results The CX3CL1/CX3CR1 signaling mediated neuroglia activation was implicated in the photoreceptor demise of an RD model. Intravitreal injection of AZD8797 preserved the retinal structure and enhanced the photoreceptor survival through inhibiting the CX3CL1/CX3CR1 expressions. Fundus photography showed that the distribution of retinal vessel was clear, and the severity of lesions was alleviated by AZD8797. In particular, these morphological benefits could be translated into remarkable functional improvements, as evidenced by the behavioral test and electroretinogram (mf-ERG) examination. A mechanism study showed that AZD8797 mitigated the microglia activation and migration in the degenerative retinas. The Müller cell hyper-reaction and secondary gliosis response were also suppressed by AZD8797. Conclusions The neuroinflammation is implicated in the photoreceptor loss of RD pathology. Targeting the CX3CL1/CX3CR1 signaling may serve as an effective therapeutic strategy. Future refinements of these findings may cast light into the discovery of new medications for RD.
Collapse
Affiliation(s)
- Jie-Min Huang
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Zhao
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiao-Na Hao
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Si-Yu Li
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Dong Wei
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ning Pu
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Guang-Hua Peng
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ye Tao
- Department of Physiology and Neurobiology, Laboratory of Visual Cell Differentiation and Regulation, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Wang X, Sun H, Wang X, Lan J, Guo Y, Liu W, Cui L, Ji X. More severe initial manifestations and worse short-term functional outcome of intracerebral hemorrhage in the plateau than in the plain. J Cereb Blood Flow Metab 2024; 44:94-104. [PMID: 37708253 PMCID: PMC10905638 DOI: 10.1177/0271678x231201088] [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: 04/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 09/16/2023]
Abstract
Intracerebral hemorrhage (ICH) is one of the most devastating forms of stroke. However, studies on ICH at high altitude are insufficient. We aimed to compare the initial manifestations, imaging features and short-term functional outcomes of ICH at different altitudes, and further explore the effect of altitude on the severity and prognosis of ICH. We retrospectively recruited ICH patients from January 2018 to July 2021 from two centers at different altitudes in China. Information regarding to clinical manifestations, neuroimages, and functional outcomes at discharge were collected and analyzed. Association between altitude and initial severity, neuroimages, and short-term prognosis of ICH were also investigated. A total of 724 patients with 400 lowlanders and 324 highlanders were enrolled. Compared with patients from the plain, those at high altitude were characterized by more severe preliminary manifestations (P < 0.0001), larger hematoma volume (P < 0.001) and poorer short-term functional outcome (P < 0.0001). High altitude was independently associated with dependency at discharge (adjusted P = 0.024), in-hospital mortality (adjusted P = 0.049) and gastrointestinal hemorrhage incidence (adjusted P = 0.017). ICH patients from high altitude suffered from more serious initial manifestations and worse short-term functional outcome than lowlanders. Control of blood pressure, oxygen supplementation and inhibition of inflammation may be critical for ICH at high altitude.
Collapse
Affiliation(s)
- Xiaoyin Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Haochen Sun
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xian Wang
- Department of Health Management, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Lan
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yong Guo
- Department of Neurology, Yushu People’s Hospital, Yushu, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Lili Cui
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xunming Ji
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|