1
|
Wu Y, Yang L, Jiang W, Zhang X, Yao Z. Glycolytic dysregulation in Alzheimer's disease: unveiling new avenues for understanding pathogenesis and improving therapy. Neural Regen Res 2025; 20:2264-2278. [PMID: 39101629 PMCID: PMC11759019 DOI: 10.4103/nrr.nrr-d-24-00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/11/2024] [Accepted: 06/20/2024] [Indexed: 08/06/2024] Open
Abstract
Alzheimer's disease poses a significant global health challenge owing to the progressive cognitive decline of patients and absence of curative treatments. The current therapeutic strategies, primarily based on cholinesterase inhibitors and N-methyl-D-aspartate receptor antagonists, offer limited symptomatic relief without halting disease progression, highlighting an urgent need for novel research directions that address the key mechanisms underlying Alzheimer's disease. Recent studies have provided insights into the critical role of glycolysis, a fundamental energy metabolism pathway in the brain, in the pathogenesis of Alzheimer's disease. Alterations in glycolytic processes within neurons and glial cells, including microglia, astrocytes, and oligodendrocytes, have been identified as significant contributors to the pathological landscape of Alzheimer's disease. Glycolytic changes impact neuronal health and function, thus offering promising targets for therapeutic intervention. The purpose of this review is to consolidate current knowledge on the modifications in glycolysis associated with Alzheimer's disease and explore the mechanisms by which these abnormalities contribute to disease onset and progression. Comprehensive focus on the pathways through which glycolytic dysfunction influences Alzheimer's disease pathology should provide insights into potential therapeutic targets and strategies that pave the way for groundbreaking treatments, emphasizing the importance of understanding metabolic processes in the quest for clarification and management of Alzheimer's disease.
Collapse
Affiliation(s)
- You Wu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijie Yang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Wanrong Jiang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xinyuan Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhaohui Yao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Liu MW, Zhang Y, Xiong GF, Zhang BR, Zhang QJ, Gao SJ, Zhu YL, Zhang LM. Dexmedetomidine for the treatment of sepsis-associated encephalopathy: Mechanism and prospects. Biomed Pharmacother 2025; 188:118209. [PMID: 40424824 DOI: 10.1016/j.biopha.2025.118209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a severe central nervous system complication that is secondary to sepsis and is characterized by a poor prognosis, high mortality, and multiple systemic manifestations. Although the specific etiology remains incompletely understood, SAE typically presents with varying degrees of neurological dysfunction. The complex mechanisms underlying SAE significantly influence patient outcomes. Recent studies have emphasized the roles of bloodbrain barrier (BBB) disruption, microglial activation, mitochondrial dysfunction, apoptosis, inflammatory responses, and oxidative stress in the development and progression of SAE. Dexmedetomidine, a highly selective α2-adrenergic receptor agonist initially employed as an anesthetic adjunct, has attracted increasing attention for its therapeutic potential in SAE. Its notable pharmacological properties include anti-inflammatory activity, modulation of microglial responses, regulation of immune function, stabilization of mitochondrial activity, inhibition of apoptosis, and maintenance of hemodynamic stability in patients with sepsis. In addition, dexmedetomidine supports gastrointestinal homeostasis and offers multiorgan protection. This review consolidates current findings regarding the protective mechanisms of dexmedetomidine in sepsis-induced brain injury and provides insight into its potential clinical applications in the management of SAE.
Collapse
Affiliation(s)
- Ming-Wei Liu
- Department of Emergency, Dali Bai Autonomous Prefecture People's Hospital, Dali 671000, China.
| | - Ye Zhang
- Department of Traditional Chinese Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Gui-Fei Xiong
- Department of Pain Management, People's Hospital of Kaiyuan City, KaiYuan, Hani-Yi Autonomous Prefecture of Honghe, 661600, China.
| | - Bin-Ran Zhang
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming 650032, China.
| | - Qiu-Juan Zhang
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming 650032, China.
| | - Shu-Ji Gao
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming 650032, China.
| | - Yan-Lin Zhu
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming 650032, China.
| | - Lin-Ming Zhang
- Department of Neurology, The First Hospital Affiliated To Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
3
|
Duan C, Lin W, Zhang M, Xue B, Sun W, Jin Y, Zhang X, Guo H, Yuan Q, Yu M, Liu Q, Wang N, Wang H, Wu H, Wang S. Nardostachys jatamansi Extract and Nardosinone Exert Neuroprotective Effects by Suppressing Glucose Metabolic Reprogramming and Modulating T Cell Infiltration. Cells 2025; 14:644. [PMID: 40358168 PMCID: PMC12071694 DOI: 10.3390/cells14090644] [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: 04/05/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Nardostachys jatamansi DC. (Gansong), a widely utilized herb in traditional Chinese medicine, has been historically employed in the management of various neuropsychiatric disorders. Nardosinone (Nar), a sesquiterpenoid compound, has been identified as one of the principal bioactive constituents of N. jatamansi. This study investigated the effects of ethyl acetate extract (NJ-1A) from N. jatamansi and its active constituent nardosinone on neuroinflammatory mediator release, glucose metabolic reprogramming, and T cell migration using both in vitro and in vivo experimental models. METHODS Lipopolysaccharide(LPS)-induced BV-2 microglial cells and a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/p)-induced male C57BL/6N mouse chronic model of Parkinson's disease were applied. RESULTS Both NJ-1A and Nar could significantly suppress LPS-induced production of M1 pro-inflammatory factors or markers in microglia and could inhibit the glycolytic process and promote oxidative phosphorylation via the AKT/mTOR signaling pathway. Furthermore, they exhibited the capacity to attenuate chemokine release from activated microglia, consequently reducing T cell migration. In vivo experiments revealed that NJ-1A and Nar effectively inhibited microglial activation, diminished T cell infiltration, and mitigated the loss of tyrosine hydroxylase (TH)-positive dopaminergic neurons in the substantia nigra of MPTP-induced mice. CONCLUSIONS NJ-1A and nardosinone exert neuroprotective effects through the modulation of microglial polarization states, regulation of metabolic reprogramming, and suppression of T cell infiltration.
Collapse
Affiliation(s)
- Congyan Duan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Weifang Lin
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Mingjie Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Bianxia Xue
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (B.X.); (H.G.); (Q.Y.)
| | - Wangjie Sun
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Yang Jin
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Xiaoxu Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Hong Guo
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (B.X.); (H.G.); (Q.Y.)
| | - Qing Yuan
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (B.X.); (H.G.); (Q.Y.)
| | - Mingyu Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Qi Liu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Naixuan Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Hong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| | - Honghua Wu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (B.X.); (H.G.); (Q.Y.)
| | - Shaoxia Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai Dist., Tianjin 301617, China; (C.D.); (W.L.); (M.Z.); (W.S.); (Y.J.); (X.Z.); (M.Y.); (Q.L.); (N.W.); (H.W.)
| |
Collapse
|
4
|
Qian N, Zhang CX, Fang GD, Qiu S, Song Y, Yuan M, Wang DL, Cheng XR. Interventional Effects of Edible Bird's Nest and Free Sialic Acids on LPS-Induced Brain Inflammation in Mice. Nutrients 2025; 17:531. [PMID: 39940389 PMCID: PMC11819919 DOI: 10.3390/nu17030531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Objectives: Our study investigated the effects and mechanisms of edible bird's nest (EBN) and free sialic acids (SA) on LPS-induced brain inflammation in mice. Methods: The experiment divided the mice into four groups: control group (CON), lipopolysaccharide group (LPS), EBN intervention group (EBN, 200 mg/kg/d in dry EBN), and sialic acid intervention group (SA, dosage was calibrated based on the concentration of sialic acid in EBN). Results: The results showed that LPS caused a decrease followed by upregulation in body weight in female mice, and EBN exhibited renal protective effects. In the Morris water maze, the learning and memory abilities of mice in the LPS group first declined and then recovered. At the same time, the escape latency improved in the EBN and SA groups. In the Open field test, both the EBN and SA groups exhibited anti-anxiety and anti-depressive effects. Immunohistochemistry in the hippocampus showed significant cell damage in the LPS group, while the damage was alleviated in the EBN and SA groups. LPS promoted the expression of TICAM1 and MYD88 in the NF-κB pathway, while both the EBN and SA groups could inhibit the expression of TICAM1. Conclusions: The study has found that both EBN and SA exhibited noteworthy anti-inflammatory effects, indicating that the main active component in EBN that provides neuroprotective effects is SA. The bound SA in EBN confers additional effects, supporting the development of prevention and treatment strategies for brain inflammation.
Collapse
Affiliation(s)
- Nan Qian
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (N.Q.); (C.-X.Z.); (G.-D.F.); (Y.S.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Chen-Xi Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (N.Q.); (C.-X.Z.); (G.-D.F.); (Y.S.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Guan-Dong Fang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (N.Q.); (C.-X.Z.); (G.-D.F.); (Y.S.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Shuang Qiu
- Hebei Edible Bird’s Nest Fresh Stew Technology Innovation Center, Langfang 065700, China; (S.Q.); (M.Y.)
| | - Yu Song
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (N.Q.); (C.-X.Z.); (G.-D.F.); (Y.S.)
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou 646000, China
| | - Man Yuan
- Hebei Edible Bird’s Nest Fresh Stew Technology Innovation Center, Langfang 065700, China; (S.Q.); (M.Y.)
| | - Dong-Liang Wang
- Hebei Edible Bird’s Nest Fresh Stew Technology Innovation Center, Langfang 065700, China; (S.Q.); (M.Y.)
| | - Xiang-Rong Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (N.Q.); (C.-X.Z.); (G.-D.F.); (Y.S.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
5
|
Škandík M, Friess L, Vázquez-Cabrera G, Keane L, Grabert K, Cruz De Los Santos M, Posada-Pérez M, Baleviciute A, Cheray M, Joseph B. Age-associated microglial transcriptome leads to diminished immunogenicity and dysregulation of MCT4 and P2RY12/P2RY13 related functions. Cell Death Discov 2025; 11:16. [PMID: 39828750 PMCID: PMC11743796 DOI: 10.1038/s41420-025-02295-1] [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: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The aging process is marked by a time-dependent deterioration in cellular functions, particularly the immune and neural systems. Understanding the phenotype acquisition of microglia, the sentinel immune cells of the brain, is crucial for understanding the nature of age-related neurological diseases. However, the specific phenotype adopted by microglia during aging remains a subject of debate and is contingent on the chosen experimental model. To address these unresolved questions, we employed a novel and highly controlled approach utilizing long-term cultivated BV-2 microglia, exempted from additional external stimuli. Our findings revealed that aged microglial cells, in comparison to their younger counterparts, acquire a distinct gene expression profile, primarily characterized by alterations in microglial immune response. Indeed, pro-inflammatory stimulated aged and young BV-2 microglia exhibited similar transcriptomic profiles, yet the response intensity to the stimulus was markedly muted in the aged microglia. Functional neurotoxic assays confirmed diminished neuronal death in coculture with aged, activated microglia, underscoring a compromised immune response. Furthermore, a subsequent comparative analysis of aged BV-2 microglia with established transcriptomic microglial datasets from aged mice and humans identified 13 overlapping genes, laying the foundation for identifying core microglial aging signature. Particularly noteworthy were SLC16A3 and P2RY13, which consistently exhibited upregulation and downregulation, respectively, across all datasets. Additionally, four other genes-CAPG, LGALS3BP, NRIP1, and P2RY12-were found to share regulatory patterns in response to both aging and extrinsic activation. An in-depth investigation focused on SLC16A3, encoding the high-affinity lactate transporter MCT4, revealed disruptions in extracellular acidification rate and lactate concentration with age. Microglial purine sensing and motility capacities, regulated by P2RY12/P2RY13, displayed age-related alterations. Remarkably, protein analysis in human brain tissue validated the observed upregulation of MCT4 and downregulation of P2RY12 in aged microglia. In conclusion, our study unveils a distinct phenotype in aged microglia characterized by compromised immune responsiveness. Through the integration of in vitro cultured BV-2 microglia with primary microglia datasets, we identify critical molecular determinants of microglial cellular aging confirmed in human-aged brain tissue. This comprehensive approach offers potential insights for understanding and potentially reprogramming aged microglia, with implications for combating age-related neurological disorders.
Collapse
Affiliation(s)
- Martin Škandík
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Friess
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Lily Keane
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kathleen Grabert
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Cruz De Los Santos
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mercedes Posada-Pérez
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China
| | - Austeja Baleviciute
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China.
| |
Collapse
|
6
|
Li L, Chen Q, Qin Y, Yu G, Qi T, Sui H, Qi X, Huang L. Regulation of TREM2 on BV2 inflammation through PI3K/AKT/mTOR pathway. Biotechnol Genet Eng Rev 2024; 40:4040-4061. [PMID: 37125903 DOI: 10.1080/02648725.2023.2204719] [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: 02/26/2023] [Accepted: 04/13/2023] [Indexed: 05/02/2023]
Abstract
This work sought to determine how lipopolysaccharide (LPS)-induced pro-inflammatory factor production in BV2 microglia was influenced by myeloid cell 2 (TREM2) expressions. LPS (0.1, 1, and 10 µg/mL) induced inflammation in BV2 cells, MTT and QPCR were used to detect the occurrence of inflammation; TREM2 activation and inhibition vectors were used to activate and inhibit TREM2; Cell Proliferation was detected using CCK-8 and cell cloning experiments. LY294002 was used to inhibit the activity of PI3K/AKT signal pathway; Western blot and ELISA were used to detect cell polarization and signal pathway changes. CCK-8 and cell clone experiments found that the activation of TERM2 can promote the proliferation of BV2 cells; and the activation of TERM2 can promote the expression of IL6, IL1β, TNFα and the expression of M2 cell phenotype molecules Arg-1 and CD206. The effect of adding LY294002 signaling pathway by TERM2 activation was inhibited, indicating that TERM2 can affect the occurrence of inflammation by regulating the activity of PI3K/AKT signaling pathway. Finally, Western blotting and ELISA showed that activation of TERM2 can promote the expression of Arg-1 and CD206 in BV2 cells, and promote the transformation of BV2 cells to M2 polarization. TERM2 can affect the inflammatory response in microglia through the PI3K/AKT signaling pathway, suggesting that TERM2 may be a target for the treatment of inflammatory response in glial cells. This study provides a treatment plan for alleviating the impact of inflammation on central nervous system.
Collapse
Affiliation(s)
- Li Li
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Qingyou Chen
- Department of Electrical Biology, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Yinghui Qin
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Guangna Yu
- Medical examination center, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Tingting Qi
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Hesong Sui
- Department of Orthopedic surgery, Qiqihar Jianhua Hospital, Qiqihar, China
| | - Xin Qi
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Lijuan Huang
- Department of Neurology, the Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| |
Collapse
|
7
|
Zhu J, Zhang Y. Dexmedetomidine inhibits the migration, invasion, and glycolysis of glioblastoma cells by lactylation of c-myc. Neurol Res 2024; 46:1105-1112. [PMID: 39193894 DOI: 10.1080/01616412.2024.2395069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is a brain tumor with poor prognosis. Dexmedetomidine (Dex) regulates the biological behaviors of tumor cells to accelerate or decelerate cancer progression. OBJECTIVE We investigated the effects of Dex on the migration, invasion, and glycolysis in GBM. METHODS The concentration of Dex was determined using the cell counting kit-8 assay. The impacts of Dex on biological behaviors of GBM cells were assessed using Transwell assay, XF96 extracellular flux analysis, and western blot. The expression of c-Myc was examined using reverse transcription-quantitative polymerase chain reaction. The lactylation or stability of c-Myc was measured by western blot after immunoprecipitation or cycloheximide treatment. RESULTS We found that Dex (200 nM) inhibited GBM cell viability, migration, invasion, and glycolysis. C-Myc was highly expressed in GBM cells and was decreased by Dex treatment. Moreover, Dex suppressed lactylated c-Myc levels via suppressing glycolysis, thereby reducing the protein stability of c-Myc. Sodium lactate treatment abrogated the effects of Dex on the biological behaviors of GBM cells. CONCLUSION Dex suppressed the migration, invasion, and glycolysis of GBM cells via inhibiting lactylation of c-Myc and suppressing the c-Myc stability, suggesting that Dex may be a novel therapeutic drug for GBM treatment.
Collapse
Affiliation(s)
- Jianglian Zhu
- Neurological Disease Center, The Third Affiliated Hospital of Chongqing Medical University, Yubei District, Chongqing, China
| | - Yundong Zhang
- Neurological Disease Center, The Third Affiliated Hospital of Chongqing Medical University, Yubei District, Chongqing, China
| |
Collapse
|
8
|
Zhang G, Zhao A, Zhang X, Zeng M, Wei H, Yan X, Wang J, Jiang X, Dai Y. Glycolytic reprogramming in microglia: A potential therapeutic target for ischemic stroke. Cell Signal 2024; 124:111466. [PMID: 39419195 DOI: 10.1016/j.cellsig.2024.111466] [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: 06/24/2024] [Revised: 09/17/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Ischemic stroke is currently the second leading cause of mortality worldwide, with limited treatment options available. As resident immune cells, microglia promptly respond to cerebral ischemic injury, influencing neuroinflammatory damage and neurorepair. Studies suggest that microglia undergo metabolic reprogramming from mitochondrial oxidative phosphorylation to glycolysis in response to ischemia, significantly impacting their function during ischemic stroke. Therefore, this study aims to investigate the roles and regulatory mechanisms involved in this process, aiming to identify a new therapeutic target or potential drug candidate.
Collapse
Affiliation(s)
- Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jie Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yongna Dai
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
9
|
Sadeghdoust M, Das A, Kaushik DK. Fueling neurodegeneration: metabolic insights into microglia functions. J Neuroinflammation 2024; 21:300. [PMID: 39551788 PMCID: PMC11571669 DOI: 10.1186/s12974-024-03296-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system, emerge in the brain during early embryonic development and persist throughout life. They play essential roles in brain homeostasis, and their dysfunction contributes to neuroinflammation and the progression of neurodegenerative diseases. Recent studies have uncovered an intricate relationship between microglia functions and metabolic processes, offering fresh perspectives on disease mechanisms and possible treatments. Despite these advancements, there are still significant gaps in our understanding of how metabolic dysregulation affects microglial phenotypes in these disorders. This review aims to address these gaps, laying the groundwork for future research on the topic. We specifically examine how metabolic shifts in microglia, such as the transition from oxidative phosphorylation and mitochondrial metabolism to heightened glycolysis during proinflammatory states, impact the disease progression in Alzheimer's disease, multiple sclerosis, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Additionally, we explore the role of iron, fatty and amino acid metabolism in microglial homeostasis and repair. Identifying both distinct and shared metabolic adaptations in microglia across neurodegenerative diseases could reveal common therapeutic targets and provide a deeper understanding of disease-specific mechanisms underlying multiple CNS disorders.
Collapse
Affiliation(s)
- Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University of Newfoundland, 300 Prince Phillip Dr. St. John's, St. John's, NL, A1B 3V6, Canada
| | - Aysika Das
- Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University of Newfoundland, 300 Prince Phillip Dr. St. John's, St. John's, NL, A1B 3V6, Canada
| | - Deepak Kumar Kaushik
- Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University of Newfoundland, 300 Prince Phillip Dr. St. John's, St. John's, NL, A1B 3V6, Canada.
| |
Collapse
|
10
|
Wang S, Wu L, Xie Y, Ge S, Wu Y, Chen L, Yi L, Yang J, Duan F, Huang L. Erjingpill bionic cerebrospinal fluid alleviates LPS-induced inflammatory response in BV2 cells by inhibiting glycolysis via mTOR. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118412. [PMID: 38824976 DOI: 10.1016/j.jep.2024.118412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Erjingpill, a well-known prescription documented in the classic Chinese medical text "Shengji Zonglu," has been proven to have effective alleviating effects on neuroinflammation in Alzheimer's disease (AD). Although the alterations in microglial cell glycolysis are known to play a crucial role in the development of neuroinflammation, it remains unclear whether the anti-neuroinflammatory effects of Erjingpill are associated with its impact on microglial cell glycolysis. AIM OF THE STUDY This study aims to determine whether Erjingpill exerts anti-neuroinflammatory effects by influencing microglial cell glycolysis. MATERIALS AND METHODS Firstly, Erjingpill decoction was prepared into an Erjingpill bionic cerebrospinal fluid (EBCF) through a process of in vitro intestinal absorption, hepatocyte incubation, and blood-brain barrier (BBB) transcytosis. Subsequently, UPLC/Q-TOF-MS/MS technology was used to analyze the compounds in Erjingpill and EBCF. Next, an in vitro neuroinflammation model was established by LPS-induced BV2 cells. The impact of EBCF on BV2 cell proliferation activity was evaluated using the CCK-8 assay, while the NO release was assessed using the Griess assay. Additionally, mRNA levels of pro-inflammatory factors (IL-1β, IL-6, TNF-α, and COX-2), anti-inflammatory factors (IL-10, IL-4, Arg-1, and TGF-β), M1 microglial markers (iNOS, CD86), M2 microglial markers (CD36, CD206), and glycolytic enzymes (HK2, GLUT1, PKM, and LDHA) were measured using qPCR. Furthermore, protein expression of microglial activation marker Iba-1, M1 marker iNOS, and M2 marker CD206 were identified through immunofluorescence, while concentrations of pro-inflammatory cytokines IL-1β and TNF-α were measured using ELISA. Enzymatic activity of glycolytic enzymes (HK, PK, and LDH) was assessed using assay kits, and the protein levels of pro-inflammatory factors (IL-1β, iNOS, and COX-2), anti-inflammatory factors (IL-10 and Arg-1), and key glycolytic proteins GLUT1 and PI3K/AKT/mTOR were detected by Western blot. RESULTS Through the analysis of Erjingpill and EBCF, 144 compounds were identified in Erjingpill and 40 compounds were identified in EBCF. The results demonstrated that EBCF effectively inhibited the elevation of inflammatory factors and glycolysis levels in LPS-induced BV2 cells, promoted polarization of M1 microglial cells towards the M2 phenotype, and suppressed the PI3K/AKT/mTOR inflammatory pathway. Moreover, EBCF alleviated LPS-induced BV2 cell inflammatory response by modulating mTOR to inhibit glycolysis. CONCLUSIONS EBCF exhibits significant anti-neuroinflammatory effects, likely attributed to its modulation of mTOR to inhibit microglial cell glycolysis. This study furnishes experimental evidence supporting the clinical utilization of Erjingpill for preventing and treating AD.
Collapse
Affiliation(s)
- Shuaikang Wang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Li Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Yongyan Xie
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Shuchao Ge
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Yi Wu
- Jiangxi Provincial Institute of Food and Drug Inspection and Testing, Nanchang, Jiangxi, 330004, China.
| | - Liping Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Longgen Yi
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Jie Yang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Feipeng Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Liping Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China; Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| |
Collapse
|
11
|
Li R, Zhang Y, Zhu Q, Wu Y, Song W. The role of anesthesia in peri‑operative neurocognitive disorders: Molecular mechanisms and preventive strategies. FUNDAMENTAL RESEARCH 2024; 4:797-805. [PMID: 39161414 PMCID: PMC11331737 DOI: 10.1016/j.fmre.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/21/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
Peri-operative neurocognitive disorders (PNDs) include postoperative delirium (POD) and postoperative cognitive dysfunction (POCD). Children and the elderly are the two populations most vulnerable to the development of POD and POCD, which results in both high morbidity and mortality. There are many factors, including neuroinflammation and oxidative stress, that are associated with POD and POCD. General anesthesia is a major risk factor of PNDs. However, the molecular mechanisms of PNDs are poorly understood. Dexmedetomidine (DEX) is a useful sedative agent with analgesic properties, which significantly improves POCD in elderly patients. In this review, the current understanding of anesthesia in PNDs and the protective effects of DEX are summarized, and the underlying mechanisms are further discussed.
Collapse
Affiliation(s)
- Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qinxin Zhu
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Yili Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| | - Weihong Song
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
| |
Collapse
|
12
|
Huang Q, Wang Y, Chen S, Liang F. Glycometabolic Reprogramming of Microglia in Neurodegenerative Diseases: Insights from Neuroinflammation. Aging Dis 2024; 15:1155-1175. [PMID: 37611905 PMCID: PMC11081147 DOI: 10.14336/ad.2023.0807] [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: 06/06/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Neurodegenerative diseases (ND) are conditions defined by progressive deterioration of the structure and function of the nervous system. Some major examples include Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS). These diseases lead to various dysfunctions, like impaired cognition, memory, and movement. Chronic neuroinflammation may underlie numerous neurodegenerative disorders. Microglia, an important immunocell in the brain, plays a vital role in defending against neuroinflammation. When exposed to different stimuli, microglia are activated and assume different phenotypes, participating in immune regulation of the nervous system and maintaining tissue homeostasis. The immunological activity of activated microglia is affected by glucose metabolic alterations. However, in the context of chronic neuroinflammation, specific alterations of microglial glucose metabolism and their mechanisms of action remain unclear. Thus, in this paper, we review the glycometabolic reprogramming of microglia in ND. The key molecular targets and main metabolic pathways are the focus of this research. Additionally, this study explores the mechanisms underlying microglial glucose metabolism reprogramming in ND and offers an analysis of the most recent therapeutic advancements. The ultimate aim is to provide insights into the development of potential treatments for ND.
Collapse
Affiliation(s)
- Qi Huang
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Yanfu Wang
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fengxia Liang
- Department of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
13
|
Wang G, Shen J, Guan Q, Lin Y, Zhai L, Shen H. LncRNA-AC020978 Promotes Metabolic Reprogramming in M1 Microglial Cells in Postoperative Cognitive Disorder via PKM2. Mol Neurobiol 2024; 61:2459-2467. [PMID: 37897635 DOI: 10.1007/s12035-023-03729-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
The present work aimed to explore the role of long non-coding RNA (lncRNA)-AC020978 in postoperative cognitive disorder (POCD) and the underlying mechanism. The POCD mouse model was constructed through isoflurane anesthesia + abbreviated laparotomy. The AC020978 expression in brain tissue was silenced after lentivirus injection, then Morris water maze test was conducted to detect the cognitive disorder level, flow cytometry was performed to analyze M1 macrophage level, ELISA was carried out to measure inflammatory factor levels, H&E, Nissl and immunohistochemical staining was performed to detect the pathological changes in brain tissue, and Western blotting assay was adopted to detect protein expression. In addition, microglial cells were cultured in vitro, after lentivirus infection, the effect of AC020978 on the M1 polarization of microglial cells and glycolysis was observed. AC020978 overexpression promoted POCD progression and aggravated cognitive disorder in mice; in addition, the proportion of peripheral and central M1 cells increased, the inflammatory factor levels were upregulated, and microglial cells were activated. By contrast, AC020978 silencing led to cognitive disorder in mice and suppressed microglial cell activation and M1 polarization. In vitro experimental results indicated that AC020978 promoted the expression and phosphorylation of PKM2, which promoted inflammatory response through enhancing microglial cell glycolysis and M1 polarization. AC020978 interacts with PKM2 to promote the glycolysis and M1 polarization of microglial cells, thus regulating cognitive disorder and central inflammation in POCD.
Collapse
Affiliation(s)
- Genghuan Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jian Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qiaobing Guan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yingcong Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Liping Zhai
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.
| | - Heping Shen
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.
| |
Collapse
|
14
|
Wang X, Zhang R, Chen B, Zhang T, Jin X, Gao P. Preliminary evaluation of the efficacy and safety of brimonidine for deep sedation. Fundam Clin Pharmacol 2024; 38:139-151. [PMID: 37612481 DOI: 10.1111/fcp.12944] [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: 02/10/2023] [Revised: 05/16/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Although brimonidine is currently used in the clinical treatment of glaucoma and rosacea, research of the deep sedative effect on animals after systemic administration is reported firstly and has shown promising results. METHODS The median effective dose (ED50 ), the median lethal dose (LD50 ), and the therapeutic index of brimonidine for deep sedation and formalin stimulation assay were determined by various animal experiments. The effect of synergistic anesthesia in rabbits with brimonidine and chloral hydrate was preliminarily evaluated. RESULTS The ED50 of brimonidine for highly effective sedation by intraperitoneal injection in rats was calculated to be 2.05 mg kg-1 with a 95% confidence interval (CI) of 1.87 to 2.25 mg kg-1 . The ED50 of brimonidine for deep sedation by intravenous and intrarectal injection in rabbits was calculated to be 0.087 mg kg-1 with a 95% CI of 0.084 to 0.091 mg kg-1 and 1.65 mg kg-1 with a 95% CI of 1.43 to 1.91 mg kg-1 , respectively. The LD50 of intraperitoneal brimonidine injection in rats was calculated to be 468 mg kg-1 with a 95% CI of 441 to 497 mg kg-1 and a therapeutic index of 228. Brimonidine has a certain analgesic and heart rate lowering effects. CONCLUSION The results confirmed that brimonidine has deep sedation and analgesic effects after systemic administration and has high safety. It can be used in combination with other types of sedative drugs to achieve better effects.
Collapse
Affiliation(s)
- Xiaohui Wang
- Laboratory of Traditional Chinese Medicine Preparations, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Rui Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Bin Chen
- Laboratory of Medical Biomaterials, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Ting Zhang
- Pharmacokinetics Laboratory, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Xinghua Jin
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Ping Gao
- Laboratory of Medical Biomaterials, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| |
Collapse
|
15
|
Wang Y, Zhao M, Cui J, Lian H, Hao Z, Lou L, Jia X, Zhao W, Shen H, Xing L, Zhang X. Ochratoxin A-enhanced glycolysis induces inflammatory responses in human gastric epithelium cells through mTOR/HIF-1α signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115868. [PMID: 38142590 DOI: 10.1016/j.ecoenv.2023.115868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Ochratoxin A (OTA) is a mycotoxin commonly found in several food commodities worldwide with potential nephrotoxic, hepatotoxic and carcinogenic effects. We previously showed for the first time that OTA treatment enhanced glycolysis in human gastric epithelium (GES-1) cells in vitro. Here, we found that OTA exposure activated inflammatory responses, evidenced by increasing of NF-κB signaling pathway-related protein (p-p65 and p-IκBα) expressions and elevating of inflammatory cytokine (IL-1β and IL-6) mRNA expressions in GES-1 cells. To elucidate the role of glycolysis in inflammatory effects triggered by OTA, we pretreated GES-1 cells with glycolysis inhibitor (2-deoxy-D-glucose, 2-DG) before OTA exposure. The result showed that 2-DG reduced the protein expressions of p-p65 and p-IκBα and alleviated the mRNA expressions of inflammatory cytokines in OTA-treated GES-1 cells. Furthermore, OTA activated the mTOR/HIF-1α pathway by increasing the protein expressions of p-mTOR, p-eIF4E and HIF-1α, and inhibition of mTOR with rapamycin or silencing HIF-1α with siRNA significantly attenuated OTA-enhanced glycolysis by reducing glycolysis related genes and thereby decreasing inflammatory effects of GES-1 cells. These results demonstrate that OTA activates inflammatory responses in GES-1 cells and this is controlled by mTOR/HIF-1α pathway-mediated glycolysis enhancement. Our findings provide a novel mechanistic view into OTA-induced gastric cytotoxicity.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Man Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Jinfeng Cui
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Hongguang Lian
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Zengfang Hao
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Lei Lou
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Xin Jia
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Wei Zhao
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Haitao Shen
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Lingxiao Xing
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China; Department of Pathology, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
16
|
Xie J, Tuo P, Zhang W, Wang S. Inhibition of the TLR4/NF-κB pathway promotes the polarization of LPS-induced BV2 microglia toward the M2 phenotype. Neuroreport 2023; 34:834-844. [PMID: 37938926 DOI: 10.1097/wnr.0000000000001961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
This study aimed to investigate whether the inhibition of the TLR4/NF-κB pathway can promote lipopolysaccharide (LPS)-induced microglial polarization from the M1 to M2 phenotype, and thus exert neuroprotection. LPS-induced microglia were used as a model for inflammation in vitro. TLR4-specific inhibitor resatorvid (TAK-242) and NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) were used to verify the effect of the TLR4/NF-κB pathway on microglia activation and polarization. Cell proliferation was measured by cell counting, and nitric oxide (NO) and reactive oxygen species (ROS) release was measured using the Griess reagent and ROS kit, respectively. Immunofluorescence and RT-qPCR analyses were used to detect the expression of microglial activation markers, phenotypic markers, related pathway molecules, and inflammatory factors. TLR4 specific inhibitor TAK-242 and NF-κB inhibitor PDTC alleviated LPS-induced microglia over-activation by inhibiting the TLR4/NF-κB pathway, and reduced LPS-stimulated cell proliferation and the release of NO, ROS, TNF-a, and IL-6 and IL-1β. Meanwhile, TAK-242 and PDTC promoted LPS-induced polarization of microglia from M1 to M2 phenotype, decreased the expression of microglial activation marker Iba1 and M1 phenotypic markers (TNF-a and CD86), and increased the expression of M2 phenotypic markers (Arg-1 and CD206). The mechanism may be related to inhibiting the TLR4/NF-κB pathway. The inhibition of the TLR4/NF-κB pathway can promote LPS-induced polarization of BV2 microglia from M1 phenotype to M2 phenotype.
Collapse
Affiliation(s)
- Jiehong Xie
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | | | | |
Collapse
|
17
|
Sun XR, Yao ZM, Chen L, Huang J, Dong SY. Metabolic reprogramming regulates microglial polarization and its role in cerebral ischemia reperfusion. Fundam Clin Pharmacol 2023; 37:1065-1078. [PMID: 37339781 DOI: 10.1111/fcp.12928] [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: 08/14/2022] [Revised: 05/12/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023]
Abstract
The brain is quite sensitive to changes in energy supply because of its high energetic demand. Even small changes in energy metabolism may be the basis of impaired brain function, leading to the occurrence and development of cerebral ischemia/reperfusion (I/R) injury. Abundant evidence supports that metabolic defects of brain energy during the post-reperfusion period, especially low glucose oxidative metabolism and elevated glycolysis levels, which play a crucial role in cerebral I/R pathophysiology. Whereas research on brain energy metabolism dysfunction under the background of cerebral I/R mainly focuses on neurons, the research on the complexity of microglia energy metabolism in cerebral I/R is just emerging. As resident immune cells of the central nervous system, microglia activate rapidly and then transform into an M1 or M2 phenotype to correspond to changes in brain homeostasis during cerebral I/R injury. M1 microglia release proinflammatory factors to promote neuroinflammation, while M2 microglia play a neuroprotective role by secreting anti-inflammatory factors. The abnormal brain microenvironment promotes the metabolic reprogramming of microglia, which further affects the polarization state of microglia and disrupts the dynamic equilibrium of M1/M2, resulting in the aggravation of cerebral I/R injury. Increasing evidence suggests that metabolic reprogramming is a key driver of microglial inflammation. For example, M1 microglia preferentially produce energy through glycolysis, while M2 microglia provide energy primarily through oxidative phosphorylation. In this review, we highlight the emerging significance of regulating microglial energy metabolism in cerebral I/R injury.
Collapse
Affiliation(s)
- Xiao-Rong Sun
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Zi-Meng Yao
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Lei Chen
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Shu-Ying Dong
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, China
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu, China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| |
Collapse
|
18
|
Chen H, Guo Z, Sun Y, Dai X. The immunometabolic reprogramming of microglia in Alzheimer's disease. Neurochem Int 2023; 171:105614. [PMID: 37748710 DOI: 10.1016/j.neuint.2023.105614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder (NDD). In the central nervous system (CNS), immune cells like microglia could reprogram intracellular metabolism to alter or exert cellular immune functions in response to environmental stimuli. In AD, microglia could be activated and differentiated into pro-inflammatory or anti-inflammatory phenotypes, and these differences in cellular phenotypes resulted in variance in cellular energy metabolism. Considering the enormous energy requirement of microglia for immune functions, the changes in mitochondria-centered energy metabolism and substrates of microglia are crucial for the cellular regulation of immune responses. Here we reviewed the mechanisms of microglial metabolic reprogramming by analyzing their flexible metabolic patterns and changes that occurred in their metabolism during the development of AD. Further, we summarized the role of drugs in modulating immunometabolic reprogramming to prevent neuroinflammation, which may shed light on a new research direction for AD treatment.
Collapse
Affiliation(s)
- Hongli Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Zichen Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| |
Collapse
|
19
|
Litke R, Vicari J, Huang BT, Shapiro L, Roh KH, Silver A, Talreja P, Palacios N, Yoon Y, Kellner C, Kaniskan H, Vangeti S, Jin J, Ramos-Lopez I, Mobbs C. Novel small molecules inhibit proteotoxicity and inflammation: Mechanistic and therapeutic implications for Alzheimer's Disease, healthspan and lifespan- Aging as a consequence of glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544352. [PMID: 37398396 PMCID: PMC10312632 DOI: 10.1101/2023.06.12.544352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Inflammation drives many age-related, especially neurological, diseases, and likely mediates age-related proteotoxicity. For example, dementia due to Alzheimer's Disease (AD), cerebral vascular disease, many other neurodegenerative conditions is increasingly among the most devastating burdens on the American (and world) health system and threatens to bankrupt the American health system as the population ages unless effective treatments are developed. Dementia due to either AD or cerebral vascular disease, and plausibly many other neurodegenerative and even psychiatric conditions, is driven by increased age-related inflammation, which in turn appears to mediate Abeta and related proteotoxic processes. The functional significance of inflammation during aging is also supported by the fact that Humira, which is simply an antibody to the pro-inflammatory cytokine TNF-a, is the best-selling drug in the world by revenue. These observations led us to develop parallel high-throughput screens to discover small molecules which inhibit age-related Abeta proteotoxicity in a C. elegans model of AD AND LPS-induced microglial TNF-a. In the initial screen of 2560 compounds (Microsource Spectrum library) to delay Abeta proteotoxicity, the most protective compounds were, in order, phenylbutyrate, methicillin, and quetiapine, which belong to drug classes (HDAC inhibitors, beta lactam antibiotics, and tricyclic antipsychotics, respectably) already robustly implicated as promising to protect in neurodegenerative diseases, especially AD. RNAi and chemical screens indicated that the protective effects of HDAC inhibitors to reduce Abeta proteotoxicity are mediated by inhibition of HDAC2, also implicated in human AD, dependent on the HAT Creb binding protein (Cbp), which is also required for the protective effects of both dietary restriction and the daf-2 mutation (inactivation of IGF-1 signaling) during aging. In addition to methicillin, several other beta lactam antibiotics also delayed Abeta proteotoxicity and reduced microglial TNF-a. In addition to quetiapine, several other tricyclic antipsychotic drugs also delayed age-related Abeta proteotoxicity and increased microglial TNF-a, leading to the synthesis of a novel congener, GM310, which delays Abeta as well as Huntingtin proteotoxicity, inhibits LPS-induced mouse and human microglial and monocyte TNF-a, is highly concentrated in brain after oral delivery with no apparent toxicity, increases lifespan, and produces molecular responses highly similar to those produced by dietary restriction, including induction of Cbp inhibition of inhibitors of Cbp, and genes promoting a shift away from glycolysis and toward metabolism of alternate (e.g., lipid) substrates. GM310, as well as FDA-approved tricyclic congeners, prevented functional impairments and associated increase in TNF-a in a mouse model of stroke. Robust reduction of glycolysis by GM310 was functionally corroborated by flux analysis, and the glycolytic inhibitor 2-DG inhibited microglial TNF-a and other markers of inflammation, delayed Abeta proteotoxicity, and increased lifespan. These results support the value of phenotypic screens to discover drugs to treat age-related, especially neurological and even psychiatric diseases, including AD and stroke, and to clarify novel mechanisms driving neurodegeneration (e.g., increased microglial glycolysis drives neuroinflammation and subsequent neurotoxicity) suggesting novel treatments (selective inhibitors of microglial glycolysis).
Collapse
|
20
|
Gao J, Su G, Chen W, Wu Q, Liu J, Liu J, Chai M, Dong Y, Wang H, Chen L, Zhang Z, Wang M. Mechanism of ligusticum cycloprolactam against neuroinflammation based on network pharmacology and experimental verification. Clin Exp Pharmacol Physiol 2023. [PMID: 37308175 DOI: 10.1111/1440-1681.13784] [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: 10/06/2022] [Revised: 04/05/2023] [Accepted: 04/26/2023] [Indexed: 06/14/2023]
Abstract
Ligustilide, a natural phthalide mainly derived from chuanxiong rhizomes and Angelica Sinensis roots, possesses anti-inflammatory activity, particularly in the context of the nervous system. However, its application is limited because of its unstable chemical properties. To overcome this limitation, ligusticum cycloprolactam (LIGc) was synthesized through structural modification of ligustilide. In this study, we combined network pharmacological methods with experimental verification to investigate the anti-neuroinflammatory effects and mechanisms of ligustilide and LIGc. Based on our network pharmacology analysis, we identified four key targets of ligustilide involved in exerting an anti-inflammatory effect, with the nuclear factor (NF)-κB signal pathway suggested as the main signalling pathway. To verify these results, we examined the expression of inflammatory cytokines and inflammation-related proteins, analysed the phosphorylation level of NF-κB, inhibitor of κBα (IκBα) and inhibitor of κB kinase α and β (IKKα+β), and evaluated the effect of BV2 cell-conditioned medium on HT22 cells in vitro. Our results, demonstrate for the first time that LIGc can downregulate the activation of the NF-κB signal pathway in BV2 cells induced by lipopolysaccharide, suppress the production of inflammatory cytokines and reduce nerve injury in HT22 cells mediated by BV2 cells. These findings suggest that LIGc inhibits the neuroinflammatory response mediated by BV2 cells, providing strong scientific support for the development of anti-inflammatory drugs based on natural ligustilide or its derivatives. However, there are some limitations to our current study. In the future, further experiments using in vivo models may provide additional evidence to support our findings.
Collapse
Affiliation(s)
- Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qionghui Wu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Junxi Liu
- Chinese Academy of Sciences Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Jifei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Miao Chai
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ying Dong
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - He Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lixia Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
21
|
Sidheeque Hassan V, Hanifa M, Navik U, Bali A. Exogenous fetuin-A protects against sepsis-induced myocardial injury by inhibiting oxidative stress and inflammation in mice. Fundam Clin Pharmacol 2023; 37:607-617. [PMID: 36647295 DOI: 10.1111/fcp.12870] [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: 09/07/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
Sepsis-induced myocardial injury is a consequence of septicemia and is one of the major causes of death in intensive care units. A serum glycoprotein called fetuin-A is secreted largely by the liver, tongue, placenta, and adipose tissue. Fetuin-A has a variety of biological and pharmacological properties. The anti-inflammatory and antioxidant glycoprotein fetuin-A has shown its efficacy in a number of inflammatory disorders including sepsis. However, its protective role against sepsis-induced myocardial injury remains elusive. The purpose of this work is to explore the role of fetuin-A in mouse models of myocardial injury brought on by cecal ligation and puncture (CLP). CLP significantly induced the myocardial injury assessed in terms of elevated myocardial markers (serum CK-MB, cTnI levels), inflammatory markers (IL-6, TNF-α) in the serum, and oxidative stress markers (increased MDA levels and decreased reduced glutathione) in heart tissue homogenate following 24 h of ligation and puncture. Further, hematoxylin and eosin (H&E) staining showed considerable histological alterations in the myocardial tissue of sepsis-developed mice. Interestingly, fetuin-A pretreatment (50 and 100 mg/kg) for 4 days before the CLP procedure significantly improved the myocardial injury and was evaluated in perspective of a reduction in the CK-MB, cTnI levels, IL-6, and TNF-α in sepsis-developed animals. Fetuin-A pretreatment significantly attenuated the oxidative stress and improved the myocardial morphology in a dose-dependent manner. The present study provides preliminary evidence that fetuin-A exerts protection against sepsis-induced cardiac dysfunction in vivo via suppression of inflammation and oxidative damage.
Collapse
Affiliation(s)
- V Sidheeque Hassan
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Mohd Hanifa
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
22
|
Liu X, Jiang N, Zhou W. Various Energetic Metabolism of Microglia in Response to Different Stimulations. Molecules 2023; 28:molecules28114501. [PMID: 37298976 DOI: 10.3390/molecules28114501] [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: 03/31/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The activation of the microglia plays an important role in the neuroinflammation induced by different stimulations associated with Alzheimer's disease (AD). Different stimulations, such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs) and cytokines, trigger a consequence of activation in the microglia with diverse changes of the microglial cell type response in AD. The activation of the microglia is often accompanied by metabolic changes in response to PAMPs, DAMPs and cytokines in AD. Actually, we do not know the distinct differences on the energetic metabolism of microglia when subject to these stimuli. This research assessed the changes of the cell type response and energetic metabolism in mouse-derived immortalized cells (BV-2 cells) induced by a PAMP (LPS), DAMPs (Aβ and ATP) and a cytokine (IL-4) in mouse-derived immortalized cells (BV-2 cells) and whether the microglial cell type response was improved by targeting the metabolism. We uncovered that LPS, a proinflammatory stimulation of PAMPs, modified the morphology from irregular to fusiform, with stronger cell viability, fusion rates and phagocytosis in the microglia accompanied by a metabolic shift to the promotion of glycolysis and the inhibition of oxidative phosphorylation (OXPHOS). Aβ and ATP, which are two known kinds of DAMPs that trigger microglial sterile activation, induced the morphology from irregular to amoebic, and significantly decreased others in the microglia, accompanied by boosting or reducing both glycolysis and OXPHOS. Monotonous pathological changes and energetic metabolism of microglia were observed under IL-4 exposure. Further, the inhibition of glycolysis transformed the LPS-induced proinflammatory morphology and decreased the enhancement of LPS-induced cell viability, the fusion rate and phagocytosis. However, the promotion of glycolysis exerted a minimal effect on the changes of morphology, the fusion rate, cell viability and phagocytosis induced by ATP. Our study reveals that microglia induced diverse pathological changes accompanied by various changes in the energetic metabolism in response to PAMPs, DAMPs and cytokines, and it may be a potential application of targeting the cellular metabolism to interfere with the microglia-mediated pathological changes in AD.
Collapse
Affiliation(s)
- Xiaohui Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasure, Beijing 100850, China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasure, Beijing 100850, China
| | - Wenxia Zhou
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasure, Beijing 100850, China
| |
Collapse
|
23
|
Afridi R, Suk K. Microglial Responses to Stress-Induced Depression: Causes and Consequences. Cells 2023; 12:1521. [PMID: 37296642 PMCID: PMC10252665 DOI: 10.3390/cells12111521] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Chronic stress is a major risk factor for various psychiatric diseases, including depression; it triggers various cellular and structural changes, resulting in the alteration of neurocircuitry and subsequent development of depression. Accumulating evidence suggests that microglial cells orchestrate stress-induced depression. Preclinical studies of stress-induced depression revealed microglial inflammatory activation in regions of the brain that regulate mood. Although studies have identified several molecules that trigger inflammatory responses in microglia, the pathways that regulate stress-induced microglial activation remain unclear. Understanding the exact triggers that induce microglial inflammatory activation can help find therapeutic targets in order to treat depression. In the current review, we summarize the recent literature on possible sources of microglial inflammatory activation in animal models of chronic stress-induced depression. In addition, we describe how microglial inflammatory signaling affects neuronal health and causes depressive-like behavior in animal models. Finally, we propose ways to target the microglial inflammatory cascade to treat depressive disorders.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
24
|
Knobel P, Litke R, Mobbs CV. Biological age and environmental risk factors for dementia and stroke: Molecular mechanisms. Front Aging Neurosci 2022; 14:1042488. [PMID: 36620763 PMCID: PMC9813958 DOI: 10.3389/fnagi.2022.1042488] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Since the development of antibiotics and vaccination, as well as major improvements in public hygiene, the main risk factors for morbidity and mortality are age and chronic exposure to environmental factors, both of which can interact with genetic predispositions. As the average age of the population increases, the prevalence and costs of chronic diseases, especially neurological conditions, are rapidly increasing. The deleterious effects of age and environmental risk factors, develop chronically over relatively long periods of time, in contrast to the relatively rapid deleterious effects of infectious diseases or accidents. Of particular interest is the hypothesis that the deleterious effects of environmental factors may be mediated by acceleration of biological age. This hypothesis is supported by evidence that dietary restriction, which universally delays age-related diseases, also ameliorates deleterious effects of environmental factors. Conversely, both age and environmental risk factors are associated with the accumulation of somatic mutations in mitotic cells and epigenetic modifications that are a measure of "biological age", a better predictor of age-related morbidity and mortality than chronological age. Here we review evidence that environmental risk factors such as smoking and air pollution may also drive neurological conditions, including Alzheimer's Disease, by the acceleration of biological age, mediated by cumulative and persistent epigenetic effects as well as somatic mutations. Elucidation of such mechanisms could plausibly allow the development of interventions which delay deleterious effects of both aging and environmental risk factors.
Collapse
Affiliation(s)
- Pablo Knobel
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rachel Litke
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charles V. Mobbs
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States,*Correspondence: Charles V. Mobbs,
| |
Collapse
|
25
|
Zhang Z, Mu X, Zhou X. Dexmedetomidine alleviates inflammatory response and oxidative stress injury of vascular smooth muscle cell via α2AR/GSK-3β/MKP-1/NRF2 axis in intracranial aneurysm. BMC Pharmacol Toxicol 2022; 23:81. [PMID: 36273189 PMCID: PMC9588221 DOI: 10.1186/s40360-022-00607-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 08/08/2022] [Indexed: 02/01/2023] Open
Abstract
Vascular smooth muscle cell (VSMC) phenotypic modulation regulates the initiation and progression of intracranial aneurysm (IA). Dexmedetomidine (DEX) is suggested to play neuroprotective roles in patients with craniocerebral injury. Therefore, we investigated the biological functions of DEX and its mechanisms against IA formation and progression in the current study. The rat primary VSMCs were isolated from Sprague-Dawley rats. IA and superficial temporal artery (STA) tissue samples were obtained from patients with IA. Flow cytometry was conducted to identify the characteristics of isolated VSMCs. Hydrogen peroxide (H2O2) was used to mimic IA-like conditions in vitro. Cell viability was detected using CCK-8 assays. Wound healing and Transwell assays were performed to detect cell motility. ROS production was determined by immunofluorescence using DCFH-DA probes. Western blotting and RT-qPCR were carried out to measure gene expression levels. Inflammation responses were determined by measuring inflammatory cytokines. Immunohistochemistry staining was conducted to measure α2-adrenergic receptor levels in tissue samples. DEX alleviated the H2O2-induced cytotoxicity, attenuated the promoting effects of H2O2 on cell malignancy, and protected VSMCs against H2O2-induced oxidative damage and inflammation response. DEX regulated the GSK-3β/MKP-1/NRF2 pathway via the α2AR. DEX alleviates the inflammatory responses and oxidative damage of VSMCs by regulating the GSK-3β/MKP-1/NRF2 pathway via the α2AR in IA.
Collapse
Affiliation(s)
- Ze Zhang
- grid.452458.aDepartment of Anesthesiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000 Hebei China
| | - Xiue Mu
- grid.452458.aDepartment of Anesthesiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000 Hebei China
| | - Xiaohui Zhou
- grid.452458.aDepartment of Anesthesiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000 Hebei China
| |
Collapse
|
26
|
Kim JE, Yang SJ. miR-30a-5p Augments the Anti-inflammatory Effects of Dexmedetomidine in LPS-induced BV2 Cells. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2022. [DOI: 10.15324/kjcls.2022.54.3.201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Ji-Eun Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Korea
| |
Collapse
|
27
|
Dexmedetomidine Mitigates Microglial Activation Associated with Postoperative Cognitive Dysfunction by Modulating the MicroRNA-103a-3p/VAMP1 Axis. Neural Plast 2022; 2022:1353778. [PMID: 35494481 PMCID: PMC9042642 DOI: 10.1155/2022/1353778] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Surgery-induced microglial activation is critical in mediating postoperative cognitive dysfunction (POCD) in elderly patients, where the important protective effect of dexmedetomidine has been indicated. However, the mechanisms of action of dexmedetomidine during the neuroinflammatory response that underlies POCD remain largely unknown. We found that lipopolysaccharide (LPS) induced substantial inflammatory responses in primary and BV2 microglial cells. The screening of differentially expressed miRNAs revealed that miR-103a-3p was downregulated in these cell culture models. Overexpression of miR-103a-3p mimics and inhibitors suppressed and enhanced the release of inflammatory factors, respectively. VAMP1 expression was upregulated in LPS-treated primary and BV-2 microglial cells, and it was validated as a downstream target of miR-103-3p. VAMP1-knockdown significantly inhibited the LPS-induced inflammatory response. Dexmedetomidine treatment markedly inhibited LPS-induced inflammation and the expression of VAMP1, and miR-103a-3p expression reversed this inhibition. Moreover, dexmedetomidine mitigated microglial activation and the associated inflammatory response in a rat model of surgical trauma that mimicked POCD. In this model, dexmedetomidine reversed miR-103a-3p and VAMP1 expression; this effect was abolished by miR-103a-3p overexpression. Taken together, the data show that miR-103a-3p/VAMP1 is critical for surgery-induced microglial activation of POCD.
Collapse
|
28
|
Wen W, Gong X, Cheung H, Yang Y, Cai M, Zheng J, Tong X, Zhang M. Dexmedetomidine Alleviates Microglia-Induced Spinal Inflammation and Hyperalgesia in Neonatal Rats by Systemic Lipopolysaccharide Exposure. Front Cell Neurosci 2021; 15:725267. [PMID: 34955749 PMCID: PMC8692868 DOI: 10.3389/fncel.2021.725267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
Noxious stimulus and painful experience in early life can induce cognitive deficits and abnormal pain sensitivity. As a major component of the outer membrane of gram-negative bacteria, lipopolysaccharide (LPS) injection mimics clinical symptoms of bacterial infections. Spinal microglial activation and the production of pro-inflammatory cytokines have been implicated in the pathogenesis of LPS-induced hyperalgesia in neonatal rats. Dexmedetomidine (DEX) possesses potent anti-neuroinflammatory and neuroprotective properties through the inhibition of microglial activation and microglial polarization toward pro-inflammatory (M1) phenotype and has been widely used in pediatric clinical practice. However, little is known about the effects of DEX on LPS-induced spinal inflammation and hyperalgesia in neonates. Here, we investigated whether systemic LPS exposure has persistent effects on spinal inflammation and hyperalgesia in neonatal rats and explored the protective role of DEX in adverse effects caused by LPS injection. Systemic LPS injections induced acute mechanical hyperalgesia, increased levels of pro-inflammatory cytokines in serum, and short-term increased expressions of pro-inflammatory cytokines and M1 microglial markers in the spinal cord of neonatal rats. Pretreatment with DEX significantly decreased inflammation and alleviated mechanical hyperalgesia induced by LPS. The inhibition of M1 microglial polarization and microglial pro-inflammatory cytokines expression in the spinal cord may implicate its neuroprotective effect, which highlights a new therapeutic target in the treatment of infection-induced hyperalgesia in neonates and preterm infants.
Collapse
Affiliation(s)
- Wen Wen
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingrui Gong
- Department of Anesthesiology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, China
| | - Hoiyin Cheung
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyan Yang
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meihua Cai
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Tong
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mazhong Zhang
- Department of Anesthesiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Brain Science of Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Potential therapeutic effects of boswellic acids/Boswellia serrata extract in the prevention and therapy of type 2 diabetes and Alzheimer's disease. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:2167-2185. [PMID: 34542667 DOI: 10.1007/s00210-021-02154-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022]
Abstract
The link between diabetes and cognitive dysfunction has been reported in many recent articles. There is currently no disease-modifying treatment available for cognitive impairment. Boswellia serrata (B. serrata) is used traditionally to treat chronic inflammatory diseases such as type 2 diabetes (T2D), insulin resistance (IR), and Alzheimer's disease (AD). This review aims to highlight current research on the potential use of boswellic acids (BAs)/B. serrata extract in T2D and AD. We reviewed the published information through June 2021. Studies have been collected through a search on online electronic databases (Academic libraries as PubMed, Scopus, Web of Science, and Egyptian Knowledge Bank). Accumulating evidence in preclinical and small human clinical studies has indicated that BAs/B. serrata extract has potential therapeutic effect in T2D and AD. According to most of the authors, the potential therapeutic effects of BAs/B. serrata extract in T2D and AD can be attributed to immunomodulatory, anti-inflammatory, antioxidant activity, and elimination of the senescent cells. BAs/B. serrata extract may act by inhibiting the IκB kinase/nuclear transcription factor-κB (IKK/NF-κB) signaling pathway and increasing the formation of selective anti-inflammatory LOX-isoform modulators. In conclusion, BAs/B. serrata extract may have positive therapeutic effects in prevention and therapy of T2D and AD. However, more randomized controlled trials with effective, large populations are needed to show a definitive conclusion about therapeutic efficacy of BAs/B. serrata extract in T2D and AD.
Collapse
|
30
|
Propofol Ameliorates Microglia Activation by Targeting MicroRNA-221/222-IRF2 Axis. J Immunol Res 2021; 2021:3101146. [PMID: 34423051 PMCID: PMC8373515 DOI: 10.1155/2021/3101146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022] Open
Abstract
Background Propofol is a widely used intravenous anesthetic drug with potential neuroprotective effect in diverse diseases of neuronal injuries such as traumatic brain injury and ischemic stroke. However, the underlying molecular mechanism remains largely unknown. Methods Real-time qPCR, enzyme-linked immunosorbent assay, and Western blotting were used to identify the expression pattern of miR-221/222, inflammatory genes, cytokines, and IRF2. The biological roles and mechanisms of propofol in microglia activation were determined in BV2 cells and primary microglia. Bioinformatic analysis and luciferase reporter assay were used to confirm the regulatory role of miR-221/222 in Irf2 expression. Results We found that miR-221 and miR-222 were downstream targets of propofol and were consistently upregulated in lipopolysaccharide- (LPS-) primed BV2 cells. Gain- and loss-of-function studies revealed that miR-221 and miR-222 were profoundly implicated in microglia activation. Then, interferon regulatory factor 2 (Irf2) was identified as a direct target gene of miR-221/222. IRF2 protein levels were reduced by miR-221/222 and increased by propofol treatment. Ectopic expression of IRF2 attenuated the proinflammatory roles induced by LPS in BV2 cells. More importantly, the suppressive effects of propofol on LPS-primed activation of BV2 cells or primary mouse microglia involved the inhibition of miR-221/222-IRF2 axis. Conclusions Our study highlights the critical function of miR-221/222, which inhibited Irf2 translation, in the anti-inflammatory effects of propofol, and provides a new perspective for the molecular mechanism of propofol-mediated neuroprotective effect.
Collapse
|
31
|
Cui Y, Qu Y, Yin K, Zhang X, Lin H. Selenomethionine ameliorates LPS-induced intestinal immune dysfunction in chicken jejunum. Metallomics 2021; 13:6127319. [PMID: 33693770 DOI: 10.1093/mtomcs/mfab003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 11/14/2022]
Abstract
Selenomethionine (SeMet) is a widely used food supplement. However, the research on the effect of SeMet on intestinal immune function is not enough. Therefore, in this experiment, SeMet was added to the diet of chickens, and lipopolysaccharide (LPS) was used as harmful stimulation to study the effect of SeMet on intestinal immune function in chickens. We chose chicken jejunum as the research object. The results showed that LPS treatment decreased the expressions of selenoproteins and induced inflammatory reaction, cytokine disorder, decreases of immunoglobulin levels, heat shock protein expression disorder, and decreases of defensin expression levels in jejunum. However, dietary SeMet can effectively alleviate the above injury caused by LPS. Our results showed that SeMet could improve the intestinal immunity in chickens, and feeding SeMet could alleviate the intestinal immune dysfunction caused by LPS. The application range of SeMet in feed can be roughly given through our experiment; i.e. 0.35-0.5 mg/kg SeMet was effective. We speculated that dietary SeMet could effectively alleviate the intestinal immune dysfunction caused by harmful stimulation and help to resist the further damage caused by harmful stimulation.
Collapse
Affiliation(s)
- Yuan Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yingying Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Kai Yin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xintong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.,Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P. R. China
| |
Collapse
|
32
|
Corrigendum. Fundam Clin Pharmacol 2020; 34:749. [DOI: 10.1111/fcp.12595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|