1
|
Yang D, Su J, Chen Y, Chen G. The NF-κB pathway: Key players in neurocognitive functions and related disorders. Eur J Pharmacol 2024; 984:177038. [PMID: 39369877 DOI: 10.1016/j.ejphar.2024.177038] [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: 06/21/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Perioperative neurocognitive disorder (PND) is a common complication of surgical anesthesia, yet its precise etiology remains unclear. Neuroinflammation is a key feature of PND, influenced by both patient -related and surgical variables. The nuclear factor-κB (NF-κB) transcription factor family plays a critical role in regulating the body's immunological proinflammatory response, which is pivotal in the development of PND. Surgery and anesthesia trigger the activation of the NF-κB signaling pathway, leading to the initiation of inflammatory cascades, disruption of the blood-brain barrier, and neuronal injury. Immune cells and glial cells are central to these pathological processes in PND. Furthermore, this study explores the interactions between NF-κB and various signaling molecules, including Tlr4, P2X, α7-nAChR, ROS, HIF-1α, PI3K/Ak, MicroRNA, Circular RNA, and histone deacetylases, within the context of PND. Targeting NF-κB as a therapeutic approach for PND shows promise as a potential treatment strategy.
Collapse
Affiliation(s)
- Danfeng Yang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Junwei Su
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
2
|
Liu T, Zhou L, Dong R, Qu Y, Liu Y, Song W, Lv J, Wu S, Peng W, Shi L. Isomalto-Oligosaccharide Potentiates Alleviating Effects of Intermittent Fasting on Obesity-Related Cognitive Impairment during Weight Loss and the Rebound Weight Gain. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23875-23892. [PMID: 39431286 DOI: 10.1021/acs.jafc.4c07351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Obesity-related cognitive dysfunction poses a significant threat to public health. The present study demonstrated mitigating effects of intermittent fasting (IF) and its combination with isomalto-oligosaccharides and IF (IF + IMO) on cognitive impairments induced by a high-fat-high-fructose (HFHF) diet in mice, with IF + IMO exhibiting superior effects. Transcriptomic analysis of the hippocampus revealed that the protective effects on cognition might be attributed to the suppression of toll-like receptor 4 (TLR4)/NFκB signaling, oxidative phosphorylation, and neuroinflammation. Moreover, both IF and IF + IMO modulated the gut microbiome and promoted the production of short-chain fatty acids, with IF + IMO displaying more pronounced effects. IF + IMO-modulated gut microbiota, metabolites, and molecular targets associated with cognitive impairments were further corroborated using human data from public databases Gmrepo and gutMgene. Furthermore, the fecal microbiome transplantation confirmed the direct impacts of IF + IMO-derived microbiota on improving cognition functions by suppressing TLR4/NFκB signaling and increasing BDNF levels. Notably, prior exposure to IF + IMO prevented weight-regain-induced cognitive decline, suppressed TLR4/NFκB signaling and inflammatory cytokines in the hippocampus, and mitigated weight regain-caused gut dysbacteriosis without altering body weight. Our study underscores that IMO-augmented alleviating effects of IF on obesity-related cognitive impairment particularly during weight-loss and weight-regain periods, presenting a novel nutritional strategy to tackle obesity-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Lanqi Zhou
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Rui Dong
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Yizhe Qu
- School of Physical Education, Shaanxi Normal University, Xi'an 710062, China
| | - Yuan Liu
- School of Physical Education, Shaanxi Normal University, Xi'an 710062, China
| | - Wei Song
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Jiayao Lv
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Shan Wu
- College of Food Science and Technology, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Wen Peng
- Nutrition and Health Promotion Center, Department of Public Health, Medical College, Qinghai University, Xining 810016, Qinghai, China
- Qinghai Provincial Key Laboratory of Traditional Chinese Medicine Research for Glucolipid Metabolic Diseases, Xining 810016, Qinghai, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| |
Collapse
|
3
|
Du J, Yin Y, Wu D, Diao C, Zhao T, Peng F, Li N, Wang D, Shi J, Wang L, Kong L, Zhou W, Hao A. SIRT6 modulates lesion microenvironment in LPC induced demyelination by targeting astrocytic CHI3L1. J Neuroinflammation 2024; 21:243. [PMID: 39342313 PMCID: PMC11438192 DOI: 10.1186/s12974-024-03241-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Demyelination occurs widely in the central nervous system (CNS) neurodegenerative diseases, especially the multiple sclerosis (MS), which with a complex and inflammatory lesion microenvironment inhibiting remyelination. Sirtuin6 (SIRT6), a histone/protein deacetylase is of interest for its promising effect in transcriptional regulation, cell cycling, inflammation, metabolism and longevity. Here we show that SIRT6 participates in the remyelination process in mice subjected to LPC-induced demyelination. Using pharmacological SIRT6 inhibitor or activator, we found that SIRT6 modulated LPC-induced damage in motor or cognitive function. Inhibition of SIRT6 impaired myelin regeneration, exacerbated neurological deficits, and decreased oligodendrocyte precursor cells (OPCs) proliferation and differentiation, whereas activation of SIRT6 reversed behavioral performance in mice, demonstrating a beneficial effect of SIRT6. Importantly, based on RNA sequencing analysis of the corpus callosum tissues, it was further revealed that SIRT6 took charge in regulation of glial activation during remyelination, and significant alterations in CHI3L1 were obtained, a glycoprotein specifically secreted by astrocytes. Impaired proliferation and differentiation of OPCs could be induced in vitro using supernatants from reactive astrocyte, especially when SIRT6 was inhibited. Mechanistically, SIRT6 regulates the secretion of CHI3L1 from reactive astrocytes by histone-H3-lysine-9 acetylation (H3K9Ac). Adeno-associated virus-overexpression of SIRT6 (AAV-SIRT6-OE) in astrocytes improved remyelination and functional recovery after LPC-induced demyelination, whereas together with AAV-CHI3L1-OE inhibits this therapeutic effect. Collectively, our data elucidate the role of SIRT6 in remyelination and further reveal astrocytic SIRT6/CHI3L1 as the key regulator for improving the remyelination environment, which may be a potential target for MS therapy.
Collapse
Affiliation(s)
- Jingyi Du
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yue Yin
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dong Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Can Diao
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Tiantian Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Fan Peng
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Naigang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dongshuang Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Jiaming Shi
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Liyan Wang
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China.
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
4
|
Zhang Z, Chen H, Han L, Liu K, Du S, Gao R. Inhibition of the NLRP3/caspase-1 cascade related pyroptosis relieved propofol-induced neuroinflammation and cognitive impairment in developing rats. Free Radic Biol Med 2024; 225:87-97. [PMID: 39341300 DOI: 10.1016/j.freeradbiomed.2024.09.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 08/31/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Numerous preclinical studies have demonstrated that prolonged exposure to propofol (A general anaesthetics) can lead to hippocampus injury in immature brains and impact long-term learning and memory functions. Neuroinflammation plays a pivotal role in the impairment of brain function associated with early exposure to anesthetic drugs. Nevertheless, the involvement of hippocampal pyroptosis and neuroinflammation mediated by the NLRP3/caspase-1 signaling cascade in propofol-induced developmental neurotoxicity remains unclear. METHODS Postnatal day (PND) 7 SD rats, PC12 cells, and HAPI cells were used to establish propofol neurotoxicity models in vivo and in vitro, respectively. We examined the potential hippocampal injury and cognitive dysfunction caused by propofol in neonatal rats through the NLRP3/caspase-1 signaling pathway using MCC950 and VX765 to inhibit the pathway. This investigation involved assessing histological changes in the hippocampus, behavioral performance in adulthood, NLRP3-related pyroptosis indicators, and neuroinflammatory cytokines. RESULTS Both in vivo and in vitro studies have demonstrated that exposure to propofol activates the NLRP3/caspase-1 signaling cascade in the hippocampus of PND7 rats, leading to pyroptosis, neuroinflammation, and subsequent hippocampal injury and behavioral changes in adulthood. However, MCC950 and VX765 inhibit the NLRP3/caspase-1 signaling cascade, reversing the developmental neurotoxicity of propofol. CONCLUSION Our study findings suggest that negative regulation of NLRP3/caspase-1 activation may serve as a potential therapeutic strategy for developmental neuroinflammation induced by propofol.
Collapse
Affiliation(s)
- Zhiheng Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Hui Chen
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Lin Han
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Kai Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Shan Du
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China.
| | - Ruifeng Gao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
5
|
Yang ZZ, Liu WQ, Yu HD, Yu SX, Li YR, Wang YF, Yao TF, Li WZ, Sun D, Niu L, Liu XZ, Zuo ZF. Inhibition of GZMB activity ameliorates cognitive dysfunction by reducing demyelination in diabetic mice. Free Radic Biol Med 2024; 225:53-62. [PMID: 39326683 DOI: 10.1016/j.freeradbiomed.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Diabetic cognitive dysfunction (DCD) has attracted increased attention, but its precise mechanism remains to be explored. Oligodendrocytes form myelin sheaths that wrap around axons. Granzyme B (GZMB) can cause axonal degeneration of the central nervous system. However, the role of GZMB in diabetic cognitive dysfunction (DCD) has not been reported. This study aimed to investigate whether GZMB promotes demyelination and participates in DCD by regulating the endoplasmic reticulum stress function of oligodendrocytes. METHODS Streptozotocin was injected intraperitoneally to establish a diabetic model in C57BL/6 mice. The mice were randomly divided into four groups: control group, diabetic group, diabetic + SerpinA3N group, and diabetic + saline treatment group. We performed the Morris water maze test to assess the learning and memory abilities of the mice. An immunofluorescence assay was performed to detect the expression sites of GZMB and OLIG2 in the hippocampal CA1 region. Luxol Fast Blue staining and electron microscopy were performed to detect the myelin number and myelin plate densities. Immunohistochemistry was used to detect the expression levels of MBP and CNPase. Protein blotting was used to assess the expression levels of GZMB, PERK, p-PERK, eIF2α, p-eIF2α, NLRP3, Caspase-1, GSDMD-N, IL-1β, and IL-18 as well as MBP and CNPase. RESULTS The GZMB inhibitor SerpinA3N reduces escape latency and increases the traversing platforms and residence time in the target area, improving DCD in mice. It also reduces endoplasmic reticulum stress in hippocampal oligodendrocytes and focal prolapse, further promoting MBP and CNPase expression and reducing demyelination. CONCLUSIONS Our findings suggest that inhibition of GZMB activity modulates oligodendrocyte endoplasmic reticulum stress and pyroptosis, reduces demyelination, and ameliorates diabetes-related cognitive impairment.
Collapse
Affiliation(s)
- Zheng-Zhong Yang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Wen-Qiang Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Hong-Dan Yu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Sheng-Xue Yu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Ya-Ru Li
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Yu-Fei Wang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Tie-Feng Yao
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Wan-Ze Li
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Die Sun
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Lin Niu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Xue-Zheng Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China.
| | - Zhong-Fu Zuo
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
6
|
Qin X, He J, Chen H, Cai X. Exploring the Potential Role of Dexmedetomidine in Reducing Postoperative Cognitive Dysfunction in Elderly Hip Fracture Patients. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:484-494. [PMID: 39129701 PMCID: PMC11319741 DOI: 10.62641/aep.v52i4.1596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
BACKGROUND Hip fractures are prevalent in the elderly; however, Postoperative Cognitive Dysfunction (POCD) is a possible complication of hip fracture surgery in elderly patients. This study examines the influence and the underlying mechanism of dexmedetomidine on POCD in elderly patients following hip fracture surgery. METHODS The retrospective study involved elderly patients with hip fracture who were treated at the Fifth Affiliated Hospital of Xinjiang Medical University from October 2021 to August 2022. During the surgery procedures, dexmedetomidine was administrated and the peripheral blood samples were collected from the patients. Inflammatory factors were measured using Enzyme-linked immunosorbent assay (ELISA), while pyroptosis-related proteins were detected through quantitative reverse transcription PCR (RT-qPCR) and western blot. Additionally, the levels of CD4+T and CD8+T cells were assessed using flow cytometry. An aged rats hip fracture model was established to further investigate the impact of dexmedetomidine on postoperative mobility, cognition function, pyroptosis and immune cells in rats. RESULTS Postoperative cognitive function in patients did not show significant alteration when compared with pre-operation levels (p > 0.05). There were notable reduction in the levels of interleukin-18 (IL-18), Caspase-3, Gasdermin-D (GSDMD) and NLR Family Pyrin Domain Containing 3 (NLRP3) (p < 0.001), accompanied by an increase in the proportion of CD4+T cells and an decrease in CD8+T cells after operation (p < 0.01). In aged rats, postoperative exploratory activities increased compared to their preoperative state. Compared with preoperative levels, the levels of interleukin-1β (IL-1β), IL-18, Caspase-3, GSDMD, and NLRP3 were significantly decreased (p < 0.001), the proportion of CD4+T cells was increased, and the proportion of CD8+T cells was decreased postoperatively (p < 0.01). CONCLUSIONS Although there was no significant alteration in postoperative cognitive function in patients, dexmedetomidine may still play a role in mitigating POCD potentially due to its effects on reducing immune inflammation and pyroptosis markers. Further research is needed to fully understand the underlying mechanisms and its clinical implications.
Collapse
Affiliation(s)
- Xingang Qin
- Department of Anesthesiology, The Fifth Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Jianbo He
- Department of Anesthesiology, The Fifth Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Hong Chen
- Department of Anesthesiology, The Fifth Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Xiaoli Cai
- Department of Anesthesiology, The Fifth Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| |
Collapse
|
7
|
Zhao D, Hu M, Liu S. Glial cells in the mammalian olfactory bulb. Front Cell Neurosci 2024; 18:1426094. [PMID: 39081666 PMCID: PMC11286597 DOI: 10.3389/fncel.2024.1426094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
The mammalian olfactory bulb (OB), an essential part of the olfactory system, plays a critical role in odor detection and neural processing. Historically, research has predominantly focused on the neuronal components of the OB, often overlooking the vital contributions of glial cells. Recent advancements, however, underscore the significant roles that glial cells play within this intricate neural structure. This review discus the diverse functions and dynamics of glial cells in the mammalian OB, mainly focused on astrocytes, microglia, oligodendrocytes, olfactory ensheathing cells, and radial glia cells. Each type of glial contributes uniquely to the OB's functionality, influencing everything from synaptic modulation and neuronal survival to immune defense and axonal guidance. The review features their roles in maintaining neural health, their involvement in neurodegenerative diseases, and their potential in therapeutic applications for neuroregeneration. By providing a comprehensive overview of glial cell types, their mechanisms, and interactions within the OB, this article aims to enhance our understanding of the olfactory system's complexity and the pivotal roles glial cells play in both health and disease.
Collapse
Affiliation(s)
| | | | - Shaolin Liu
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, Department of Biomedical Sciences, University of Georgia College of Veterinary Medicine, Athens, GA, United States
| |
Collapse
|
8
|
Hanrieder J. Lipid imaging of Alzheimer's disease pathology. J Neurochem 2024; 168:1175-1178. [PMID: 38372595 DOI: 10.1111/jnc.16079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024]
Abstract
Alzheimer's disease (AD) affects one in eight individuals over 65 and poses an immense societal challenge. AD pathology is characterized by the formation of beta-amyloid plaques and Tau tangles in the brain. While some disease-modifying treatments targeting beta-amyloid are emerging, the exact chain of events underlying the pathogenesis of this disease remains unclear. Brain lipids have long been implicated in AD pathology, though their role in AD pathogenesis remains not fully resolved. Significant advancements in mass spectrometry imaging (MSI) allow to detail spatial lipid regulations in biological tissues at the low um scale. In this issue, Huang et al. resolve spatial lipid patterns in human AD brain and genetic mouse models using desorption electrospray ionization (DESI)-based MSI integrated with other spatial techniques such as imaging mass cytometry of correlative protein signatures. Those spatial multiomics experiments identify plaque-associated lipid regulations that are dependent on progressing plaque pathology in both mouse models and the human brain. Of those lipid species, particularly pro-inflammatory lysophospholipids have been implicated in AD pathology through their interaction with both aggregating Aβ and microglial activation through lipid sensing surface receptors. Together, this study provides further insight into how brain lipid homeostasis is linked to progressing AD pathology, and thereby highlights the potential of MSI-based spatial lipidomics as an emerging spatial biology technology for biomedical research.
Collapse
Affiliation(s)
- Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Science for Life Laboratory (SciLife), University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
9
|
Wei J, Zheng W, Teng C, An X, Li L, Zhong P, Peng C, Zhuge S, Akoto Ampadu J, Yu C, Cai X. Exogenous NADPH could mitigate pyroptosis-induced brain injury in fetal mice exposed to gestational intermittent hypoxia. Int Immunopharmacol 2024; 135:112311. [PMID: 38781607 DOI: 10.1016/j.intimp.2024.112311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Obstructive Sleep Apnea (OSA) during pregnancy is characterized by intermittent hypoxia (IH) during sleep and will lead to the rise of oxidative stress in the fetal body. Pyroptosis, a type of inflammatory and programmable cell death mediated by Gasdermin D (GSDMD), plays a substantial role in oxygen deprivation's contribution to neural system damage. Existing research shows that Nicotinamide Adenine Dinucleotide Phosphate (NADPH) plays a protective role in alleviating brain tissue pyroptosis. We speculate that exogenous NADPH may play a protective role in OSA during pregnancy. METHODS A model of GIH group was established to simulate the pathophysiological mechanisms of OSA during pregnant and AIR group was established by giving the same frequency. Sham group was established by injecting NS and the NADPH group was established and given exogenous NADPH. We utilized the Morris Water Maze to assess cognitive function impairment, Luxol Fast Blue (LBF) staining to confirm myelin sheath formation, TUNEL staining to examine cell death in fetal mice brain tissue, and Western blotting to detect pertinent protein expressions. RESULTS The GIH group offspring exhibited decreases in spatial learning and memory abilities, reduced numbers of oligodendrocytes and formed myelin, as well as increased expression of pyroptosis-related proteins. The NADPH group offspring showed restoration in spatial learning and memory abilities increased counts of oligodendrocytes and formed myelin sheaths, in addition to decreased expression of pyroptosis-related. CONCLUSIONS This study demonstrates that early injection of exogenous NADPH can alleviate the damage to fetal brain development caused by gestational intermittent hypoxia (GIH).
Collapse
Affiliation(s)
- Jiayun Wei
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Weikun Zheng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Chenjiong Teng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xueqian An
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Lingling Li
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Peipei Zhong
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Chenlei Peng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Shurui Zhuge
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Janet Akoto Ampadu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Chenyi Yu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang 325027, PR China; The second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
10
|
Li H, Qian J, Wang Y, Wang J, Mi X, Qu L, Song N, Xie J. Potential convergence of olfactory dysfunction in Parkinson's disease and COVID-19: The role of neuroinflammation. Ageing Res Rev 2024; 97:102288. [PMID: 38580172 DOI: 10.1016/j.arr.2024.102288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder that affects 7-10 million individuals worldwide. A common early symptom of PD is olfactory dysfunction (OD), and more than 90% of PD patients suffer from OD. Recent studies have highlighted a high incidence of OD in patients with SARS-CoV-2 infection. This review investigates the potential convergence of OD in PD and COVID-19, particularly focusing on the mechanisms by which neuroinflammation contributes to OD and neurological events. Starting from our fundamental understanding of the olfactory bulb, we summarize the clinical features of OD and pathological features of the olfactory bulb from clinical cases and autopsy reports in PD patients. We then examine SARS-CoV-2-induced olfactory bulb neuropathology and OD and emphasize the SARS-CoV-2-induced neuroinflammatory cascades potentially leading to PD manifestations. By activating microglia and astrocytes, as well as facilitating the aggregation of α-synuclein, SARS-CoV-2 could contribute to the onset or exacerbation of PD. We also discuss the possible contributions of NF-κB, the NLRP3 inflammasome, and the JAK/STAT, p38 MAPK, TLR4, IL-6/JAK2/STAT3 and cGAS-STING signaling pathways. Although olfactory dysfunction in patients with COVID-19 may be reversible, it is challenging to restore OD in patients with PD. With the emergence of new SARS-CoV-2 variants and the recurrence of infections, we call for continued attention to the intersection between PD and SARS-CoV-2 infection, especially from the perspective of OD.
Collapse
Affiliation(s)
- Hui Li
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Junliang Qian
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Youcui Wang
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Juan Wang
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Xiaoqing Mi
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Le Qu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Ning Song
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China.
| | - Junxia Xie
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China.
| |
Collapse
|
11
|
Hou P, Yang Y, Li Z, Ye D, Chen L, Feng T, Zeng J, Wei L, Wang S. TAK-3 Inhibits Lipopolysaccharide-Induced Neuroinflammation in Traumatic Brain Injury Rats Through the TLR-4/NF-κB Pathway. J Inflamm Res 2024; 17:2147-2158. [PMID: 38617382 PMCID: PMC11015848 DOI: 10.2147/jir.s454099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Purpose The activation of the inflammatory response is regarded as a pivotal factor in the pathogenesis of TBI. Central nervous system infection often leads to the exacerbation of neuroinflammation following TBI, primarily caused by Gram-negative bacteria. This study aims to elucidate the effects of the novel anti-inflammatory drug TAK-3 on LPS-induced neuroinflammation in TBI rats. Methods In conjunction with the rat controlled cortical impact model, we administered local injections of Lipopolysaccharide to the impact site. Subsequently, interventions were implemented through intraperitoneal injections of TAK-3 and NF-κB activitor2 to modulate the TLR4/NF-κB axis The impact of LPS on neurological function was assessed using mNSS, open field test, and brain water content measurement. Inflammatory markers, including TNF-α, IL-1β, IL-6 and IL-10 were assessed to evaluate the condition of neuritis by Elisa. The activation of the TLR-4/NF-κB signaling pathway was detected by immunofluorescence staining and Western blot to assess the anti-inflammatory effects of TAK-3. Results The administration of LPS exacerbated neurological damage in rats with TBI, as evidenced by a reduction in motor activity and an increase in anxiety-like behavior. Furthermore, LPS induced disruption of the blood-brain barrier integrity and facilitated the development of brain edema. The activation of microglia and astrocytes by LPS at the cellular and molecular levels has been demonstrated to induce a significant upregulation of neuroinflammatory factors. The injection of TAK-3 attenuated the neuroinflammatory response induced by LPS. Conclusion The present study highlights the exacerbating effects of LPS on neuroinflammation in TBI through activation of the TLR-4/NF-κB signaling pathway. TAK-3 can modulate the activity of this signaling axis, thereby attenuating neuroinflammation and ultimately reducing brain tissue damage.
Collapse
Affiliation(s)
- Pengwei Hou
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University (The 900TH Hospital), Fuzhou, Fujian Province, People’s Republic of China
| | - Yang Yang
- Fuzhou General Teaching Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, People’s Republic of China
| | - Ziqi Li
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University (The 900TH Hospital), Fuzhou, Fujian Province, People’s Republic of China
| | - Dan Ye
- Fuzhou General Teaching Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, People’s Republic of China
| | - Li Chen
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University (The 900TH Hospital), Fuzhou, Fujian Province, People’s Republic of China
| | - Tianshun Feng
- Department of Neurosurgery, Dongfang Affiliated Hospital of Xiamen University School of Medicine, Xiamen University, Xiamen, Fujian Province, People’s Republic of China
| | - Jiateng Zeng
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Liangfeng Wei
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University (The 900TH Hospital), Fuzhou, Fujian Province, People’s Republic of China
| | - Shousen Wang
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University (The 900TH Hospital), Fuzhou, Fujian Province, People’s Republic of China
- Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, Fujian Province, People’s Republic of China
| |
Collapse
|
12
|
Xu L, Min H, Saha A, Gunaratne A, Schwartzman J, Parrott R, Kurtzberg J, Filiano AJ. Mesenchymal stromal cells suppress microglial activation and tumor necrosis factor production. Cytotherapy 2024; 26:185-193. [PMID: 38054911 DOI: 10.1016/j.jcyt.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/03/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND AIMS White matter diseases are commonly associated with microglial activation and neuroinflammation. Mesenchymal stromal cells (MSCs) have immunomodulatory properties and thus have the potential to be developed as cell therapy for white matter disease. MSCs interact with resident macrophages to alter the trajectory of inflammation; however, the impact MSCs have on central nervous system macrophages and the effect this has on the progression of white matter disease are unclear. METHODS In this study, we utilized numerous assays of varying complexity to model different aspects of white matter disease. These assays ranged from an in vivo spinal cord acute demyelination model to a simple microglial cell line activation assay. Our goal was to investigate the influence of human umbilical cord tissue MSCs on the activation of microglia. RESULTS MSCs reduced the production of tumor necrosis factor (TNF) by microglia and decreased demyelinated lesions in the spinal cord after acute focal injury. To determine if MSCs could directly suppress the activation of microglia and to develop an efficient potency assay, we utilized isolated primary microglia from mouse brains and the Immortalized MicroGlial Cell Line (IMG). MSCs suppressed the activation of microglia and the release of TNF after stimulation with lipopolysaccharide, a toll-like receptor agonist. CONCLUSIONS In this study, we demonstrated that MSCs altered the immune response after acute injury in the spinal cord. In numerous assays, MSCs suppressed activation of microglia and release of the pro-inflammatory cytokine TNF. Of these assays, IMG could be standardized and used as an effective potency assay to determine the efficacy of MSCs for treating white matter disease or other neuroinflammatory conditions associated with microglial activation.
Collapse
Affiliation(s)
- Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Hyunjung Min
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Arjun Saha
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Aruni Gunaratne
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | | | - Roberta Parrott
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Anthony J Filiano
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Neurosurgery, Duke University, Durham, North Carolina, USA; Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA; Department of Pathology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
13
|
Perpiñá-Clérigues C, Mellado S, Galiana-Roselló C, Fernández-Regueras M, Marcos M, García-García F, Pascual M. Novel insight into the lipid network of plasma extracellular vesicles reveal sex-based differences in the lipidomic profile of alcohol use disorder patients. Biol Sex Differ 2024; 15:10. [PMID: 38273378 PMCID: PMC10809459 DOI: 10.1186/s13293-024-00584-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Alcohol use disorder (AUD) is one of the most common psychiatric disorders, with the consumption of alcohol considered a leading cause of preventable deaths worldwide. Lipids play a crucial functional role in cell membranes; however, we know little about the role of lipids in extracellular vesicles (EVs) as regulatory molecules and disease biomarkers. METHODS We employed a sensitive lipidomic strategy to characterize lipid species from the plasma EVs of AUD patients to evaluate functional roles and enzymatic activity networks to improve the knowledge of lipid metabolism after alcohol consumption. We analyzed plasma EV lipids from AUD females and males and healthy individuals to highlight lipids with differential abundance and biologically interpreted lipidomics data using LINEX2, which evaluates enzymatic dysregulation using an enrichment algorithm. RESULTS Our results show, for the first time, that AUD females exhibited more significant substrate-product changes in lysophosphatidylcholine/phosphatidylcholine lipids and phospholipase/acyltransferase activity, which are potentially linked to cancer progression and neuroinflammation. Conversely, AUD males suffer from dysregulated ceramide and sphingomyelin lipids involving sphingomyelinase, sphingomyelin phosphodiesterase, and sphingomyelin synthase activity, which relates to hepatotoxicity. Notably, the analysis of plasma EVs from AUD females and males demonstrates enrichment of lipid ontology terms associated with "negative intrinsic curvature" and "positive intrinsic curvature", respectively. CONCLUSIONS Our methodological developments support an improved understanding of lipid metabolism and regulatory mechanisms, which contribute to the identification of novel lipid targets and the discovery of sex-specific clinical biomarkers in AUD.
Collapse
Affiliation(s)
- Carla Perpiñá-Clérigues
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Cristina Galiana-Roselló
- Department of Inorganic Chemistry, Institute of Molecular Science, University of Valencia, 46980, Paterna, Spain
| | - María Fernández-Regueras
- Hospital Universitario de Burgos, 09006, Burgos, Spain
- Hospital Universitario de Salamanca, 37007, Salamanca, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Francisco García-García
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain.
| |
Collapse
|
14
|
Ji Y, Ma Y, Ma Y, Wang Y, Zhao X, Jin D, Xu L, Ge S. Rutin prevents pyroptosis and M1 microglia via Nrf2/Mac-1/caspase-1-mediated inflammasome axis to improve POCD. Int Immunopharmacol 2024; 127:111290. [PMID: 38064815 DOI: 10.1016/j.intimp.2023.111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/05/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Neuroinflammation following peripheral surgery plays a key role in postoperative cognitive dysfunction (POCD) development and there is no effective therapy to inflammation-mediated cognitive impairment. Recent studies showed that rutin, a natural flavonoid compound, conferred neuroprotection. However, the effects and mechanisms of rutin on cognition of surgical and aged mice and LPS-induced BV2 need deeper exploration. METHODS The effect of rutin in vivo and vitro were evaluated by Morris water maze test, HE stainin, Golgi-Cox staining, IF, IHC, RT-PCR, Flow Cytometer and Western blotting. In vivo, aged mice were treated with rutin and surgery. In vitro, rutin, Nrf2 knockdown, MAC-1 overexpression and VX765, a caspase-1 inhibitor, were administration on BV2 microglial cells. RESULTS Surgery led to compensatory increase in nuclear Nrf2 and rutin could further increase it. Neural damage was accompanied with high level in MAC-1, caspase-1-mediated pyroptosis and M1 microglia, while rutin recovered the process. Nrf2 inhibition abolished the effect of rutin with the increase of MAC-1, caspase-1-mediated pyroptosis and M1 microglia. Activation of MAC-1 abrogated protection of rutin by increase in pyroptosis and M1 microglia. Finally, we found that treatment with VX765 improved injury and increased M2 microglia against overexpression of MAC-1. CONCLUSIONS Our study indicated that rutin may be a potential therapy in POCD and exerted neural protection via Nrf2/ Mac-1/ caspase-1-mediated inflammasome axis to regulate pyroptosis and microglial polarization.
Collapse
Affiliation(s)
- Yelong Ji
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Yuanyuan Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Yimei Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Ying Wang
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Xining Zhao
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Danfeng Jin
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Li Xu
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Shengjin Ge
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China.
| |
Collapse
|
15
|
Zhu TT, Wang H, Liu PM, Gu HW, Pan WT, Zhao MM, Hashimoto K, Yang JJ. Clemastine-induced enhancement of hippocampal myelination alleviates memory impairment in mice with chronic pain. Neurobiol Dis 2024; 190:106375. [PMID: 38092269 DOI: 10.1016/j.nbd.2023.106375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Patients with chronic pain often experience memory impairment, but the underlying mechanisms remain elusive. The myelin sheath is crucial for rapid and accurate action potential conduction, playing a pivotal role in the development of cognitive abilities in the central nervous system. The study reveals that myelin degradation occurs in the hippocampus of chronic constriction injury (CCI) mice, which display both chronic pain and memory impairment. Using fiber photometry, we observed diminished task-related neuronal activity in the hippocampus of CCI mice. Interestingly, the repeated administration with clemastine, which promotes myelination, counteracts the CCI-induced myelin loss and reduced neuronal activity. Notably, clemastine specifically ameliorates the impaired memory without affecting chronic pain in CCI mice. Overall, our findings highlight the significant role of myelin abnormalities in CCI-induced memory impairment, suggesting a potential therapeutic approach for treating memory impairments associated with neuropathic pain.
Collapse
Affiliation(s)
- Ting-Ting Zhu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - He Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Pan-Miao Liu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Han-Wen Gu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei-Tong Pan
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Ming-Ming Zhao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Kenji Hashimoto
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
16
|
Arneth B. Regulatory T Cells in Multiple Sclerosis Diagnostics-What Do We Know So Far? J Pers Med 2023; 14:29. [PMID: 38248730 PMCID: PMC10821144 DOI: 10.3390/jpm14010029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disorder that affects the central nervous system (CNS) through inflammation. MS symptoms become acute if the disease progresses to the relapsing phase. AIM This review aimed to evaluate the role played by regulatory T cells (Tregs) in the pathogenesis of MS. METHODS This review used scholarly journal articles obtained from PubMed, PsycINFO, and CINAHL with different search parameters such as 'regulatory T cells', 'multiple sclerosis', and 'current knowledge'. The process of searching for articles was limited to those that had publication dates falling between 2010 and 2020. RESULTS Tregs play a role in the pathogenesis of MS. This conclusion is supported by animal disease models and environmental factors that can underlie Treg alterations in MS. Despite the knowledge of the role played by Tregs in MS pathogenesis, the specific subsets of Tregs involved in MS development remain incompletely understood. DISCUSSION This review provides an essential link between Tregs and MS activity. Targeting Tregs could be an efficient way to establish new treatment methods for MS management. CONCLUSION MS is a complex condition affecting many people worldwide. Research has shown that Tregs can influence MS development and progression. More investigations are needed to understand how Tregs affect the pathogenesis of MS.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Philipps University Marburg, 35043 Marburg, Germany;
- Institute of Laboratory Medicine and Pathobiochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
- Hospital of the Universities of Giessen and Marburg, 35392 Giessen, Germany
| |
Collapse
|
17
|
Kong G, Xiong W, Li C, Xiao C, Wang S, Li W, Chen X, Wang J, Chen S, Zhang Y, Gu J, Fan J, Jin Z. Treg cells-derived exosomes promote blood-spinal cord barrier repair and motor function recovery after spinal cord injury by delivering miR-2861. J Nanobiotechnology 2023; 21:364. [PMID: 37794487 PMCID: PMC10552208 DOI: 10.1186/s12951-023-02089-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
The blood-spinal cord barrier (BSCB) is a physical barrier between the blood and the spinal cord parenchyma. Current evidence suggests that the disruption of BSCB integrity after spinal cord injury can lead to secondary injuries such as spinal cord edema and excessive inflammatory response. Regulatory T (Treg) cells are effective anti-inflammatory cells that can inhibit neuroinflammation after spinal cord injury, and their infiltration after spinal cord injury exhibits the same temporal and spatial characteristics as the automatic repair of BSCB. However, few studies have assessed the relationship between Treg cells and spinal cord injury, emphasizing BSCB integrity. This study explored whether Treg affects the recovery of BSCB after SCI and the underlying mechanism. We confirmed that spinal cord angiogenesis and Treg cell infiltration occurred simultaneously after SCI. Furthermore, we observed significant effects on BSCB repair and motor function in mice by Treg cell knockout and overexpression. Subsequently, we demonstrated the presence and function of exosomes in vitro. In addition, we found that Treg cell-derived exosomes encapsulated miR-2861, and miR-2861 regulated the expression of vascular tight junction (TJs) proteins. The luciferase reporter assay confirmed the negative regulation of IRAK1 by miR-2861, and a series of rescue experiments validated the biological function of IRAKI in regulating BSCB. In summary, we demonstrated that Treg cell-derived exosomes could package and deliver miR-2861 and regulate the expression of IRAK1 to affect BSCB integrity and motor function after SCI in mice, which provides novel insights for functional repair and limiting inflammation after SCI.
Collapse
Affiliation(s)
- Guang Kong
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wu Xiong
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cong Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenyu Xiao
- Nanjing Medical University, Nanjing, Jiangsu, China
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Siming Wang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenbo Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiangjun Chen
- Nanjing Medical University, Nanjing, Jiangsu, China
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juan Wang
- Nanjing Medical University, Nanjing, Jiangsu, China
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sheng Chen
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yongjie Zhang
- Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jun Gu
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.
| | - Jin Fan
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Zhengshuai Jin
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|