51
|
Zhang Y, Zhang J, Zhao Y, Zhang Y, Liu L, Xu X, Wang X, Fu J. ChemR23 activation attenuates cognitive impairment in chronic cerebral hypoperfusion by inhibiting NLRP3 inflammasome-induced neuronal pyroptosis. Cell Death Dis 2023; 14:721. [PMID: 37932279 PMCID: PMC10628255 DOI: 10.1038/s41419-023-06237-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023]
Abstract
Neuroinflammation plays critical roles in vascular dementia (VaD), the second leading cause of dementia, which can be induced by chronic cerebral hypoperfusion (CCH). NLRP3 inflammasome-induced pyroptosis, the inflammatory programmed cell death, has been reported to contribute to the development of VaD. ChemR23 is a G protein-coupled receptor that has emerging roles in regulating inflammation. However, the role of ChemR23 signalling in NLRP3 inflammasome-induced pyroptosis in CCH remains elusive. In this study, a CCH rat model was established by permanent bilateral common carotid artery occlusion (BCCAO) surgery. Eight weeks after the surgery, the rats were intraperitoneally injected with the ChemR23 agonist Resolvin E1 (RvE1) or chemerin-9 (C-9). Additionally, primary rat hippocampal neurons and SH-SY5Y cells were adopted to mimic CCH injury in vitro. Our results showed that the levels of ChemR23 expression were decreased from the 8th week after BCCAO, accompanied by significant cognitive impairment. Further analysis revealed that CCH induced neuronal damage, synaptic injury and NLRP3-related pyroptosis activation in hippocampal neurons. However, pharmacologic activation of ChemR23 with RvE1 or C-9 counteracted these changes. In vitro experiments also showed that ChemR23 activation prevented primary neuron pyroptosis induced by chronic hypoxia. In addition, manipulating ChemR23 expression markedly regulated NLRP3 inflammasome-induced neuronal pyroptosis through PI3K/AKT/Nrf2 signalling in SH-SY5Y cells under hypoglycaemic and hypoxic conditions. Collectively, our data demonstrated that ChemR23 activation inhibits NLRP3 inflammasome-induced neuronal pyroptosis and improves cognitive function via the PI3K/AKT/Nrf2 signalling pathway in CCH models. ChemR23 may serve as a potential novel therapeutic target to treat CCH-induced cognitive impairment.
Collapse
Affiliation(s)
- Yaxuan Zhang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Jiawei Zhang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yao Zhao
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yueqi Zhang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Lan Liu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Xiaofeng Xu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Xiuzhe Wang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Jianliang Fu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
52
|
Xu Y, Yang Y, Chen X, Jiang D, Zhang F, Guo Y, Hu B, Xu G, Peng S, Wu L, Hu J. NLRP3 inflammasome in cognitive impairment and pharmacological properties of its inhibitors. Transl Neurodegener 2023; 12:49. [PMID: 37915104 PMCID: PMC10621314 DOI: 10.1186/s40035-023-00381-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
Cognitive impairment is a multifactorial and multi-step pathological process that places a heavy burden on patients and the society. Neuroinflammation is one of the main factors leading to cognitive impairment. The inflammasomes are multi-protein complexes that respond to various microorganisms and endogenous danger signals, helping to initiate innate protective responses in inflammatory diseases. NLRP3 inflammasomes produce proinflammatory cytokines (interleukin IL-1β and IL-18) by activating caspase-1. In this review, we comprehensively describe the structure and functions of the NLRP3 inflammasome. We also explore the intrinsic relationship between the NLRP3 inflammasome and cognitive impairment, which involves immune cell activation, cell apoptosis, oxidative stress, mitochondrial autophagy, and neuroinflammation. Finally, we describe NLRP3 inflammasome antagonists as targeted therapies to improve cognitive impairment.
Collapse
Affiliation(s)
- Yi Xu
- The Second Affiliated Hospital of Nanchang University, Department of the Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Yanling Yang
- The Second Affiliated Hospital of Nanchang University, Department of the Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Xi Chen
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Danling Jiang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fei Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yao Guo
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Bin Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Lidong Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Department of Thyroid and Hernia Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
53
|
Liu J, Liu S, Yu M, Li J, Xie Z, Gao B, Liu Y. Anti-inflammatory effect and mechanism of catalpol in various inflammatory diseases. Drug Dev Res 2023; 84:1376-1394. [PMID: 37534768 DOI: 10.1002/ddr.22096] [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: 04/10/2023] [Revised: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
Catalpol is a kind of iridoid glucoside, widely found in a variety of plants, mostly extracted from the rhizome of the traditional medicinal herb rehmanniae. It has various biological activities such as anti-inflammatory, antioxidant, and antitumor. The anti-inflammatory effects of catalpol have been demonstrated in a variety of diseases, such as neurological diseases, atherosclerosis, renal diseases, respiratory diseases, digestive diseases, bone and joint diseases, eye diseases, and periodontitis. The purpose of this review is to summarize the existing literature on the anti-inflammatory effects of catalpol in a variety of inflammatory diseases over the last decade and to focus on the anti-inflammatory mechanisms of catalpol.
Collapse
Affiliation(s)
- Jinyao Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shuang Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Mingyue Yu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jianing Li
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zunxuan Xie
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Boyang Gao
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuyan Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
54
|
Chen Y, Miao C, Zhao Y, Yang L, Wang R, Shen D, Ren N, Zhang Q. Inflammasomes in human reproductive diseases. Mol Hum Reprod 2023; 29:gaad035. [PMID: 37788097 DOI: 10.1093/molehr/gaad035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Inflammasomes are multi-protein complexes localized within immune and non-immune cells that induce caspase activation, proinflammatory cytokine secretion, and ultimately pyroptosis-a type of cell death. Inflammasomes are involved in a variety of human diseases, especially acute or chronic inflammatory diseases. In this review, we focused on the strong correlation between the NLRP3 inflammasome and various reproductive diseases, including ovarian aging or premature ovarian insufficiency, PCOS, endometriosis, recurrent spontaneous abortion, preterm labor, pre-eclampsia, and male subfertility, as well as the multifaceted role of NLRP3 in the pathogenesis and treatment of these diseases. In addition, we provide an overview of the structure and amplification of inflammasomes. This comprehensive review demonstrates the vital role of NLRP3 inflammasome activation in human reproductive diseases together with the underlying mechanisms, offers new insights for mechanistic studies of reproduction, and provides promising possibilities for the development of drugs targeting the NLRP3 inflammasome for the treatment of reproductive disorders in the future.
Collapse
Affiliation(s)
- Yun Chen
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenyun Miao
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhao
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Liuqing Yang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruye Wang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Dan Shen
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ning Ren
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Zhang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
55
|
Jin Y, Wei C, Huang X, Zhang D, Zhang L, Li X. Bioinformatics Analysis and Experimental Verification of Exercise for Aging Mice in Different Brain Regions Based on Transcriptome Sequencing. Life (Basel) 2023; 13:1988. [PMID: 37895370 PMCID: PMC10608440 DOI: 10.3390/life13101988] [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: 08/17/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
PURPOSE Physical exercise mitigates the effects of aging and cognitive decline. However, the precise neurobiological mechanisms underlying this phenomenon remain unclear. The primary aim of this study was to investigate the protective effect of exercise on age-related memory deficits in the prefrontal cortex (PFC) and hippocampus using bioinformatic analysis and biochemical verification. METHODS Young and aging mice were subjected to natural feeding or treadmill exercise (12 m/min, 8 weeks). Cognitive function was accessed using the Barnes maze and novel object recognition. Bioinformatic analysis was performed to identify co-expressed genes in different groups and brain regions. The selected genes and pathways were validated using RT-qPCR. RESULTS Regular exercise significantly ameliorated age-related cognitive deficits. Four up-regulated targets (Ifi27l2a, Irf7, Oas1b, Ifit1) and one down-regulation (Septin2) were reversed by exercise, demonstrating the underlying mechanisms of cognitive functions induced by aging with exercise in the hippocampus and PFC. The Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses indicated that the NOD-like receptor signaling pathway was inhibited in the neuroinflammation effects of exercise in aging mice in both brain regions. CONCLUSION Exercise enhances age-related learning and memory deficits. This beneficial effect may be attributed to the changes in five up/down-regulated genes and the NOD-like receptor signaling pathway in both the hippocampus and PFC. These findings establish the modulation of neuroinflammation as a pivotal molecular mechanism supporting exercise intervention in the brain aging process.
Collapse
Affiliation(s)
- Yu Jin
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (Y.J.); (C.W.); (X.H.); (D.Z.)
| | - Changling Wei
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (Y.J.); (C.W.); (X.H.); (D.Z.)
| | - Xiaohan Huang
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (Y.J.); (C.W.); (X.H.); (D.Z.)
| | - Deman Zhang
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (Y.J.); (C.W.); (X.H.); (D.Z.)
| | - Li Zhang
- Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China;
| | - Xue Li
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (Y.J.); (C.W.); (X.H.); (D.Z.)
| |
Collapse
|
56
|
Wang X, Shi YJ, Niu TY, Chen TT, Li HB, Wu SH, Li GL. Neuroprotective effect of 20 (S) - Protopanaxadiol (PPD) attenuates NLRP3 inflammasome-mediated microglial pyroptosis in vascular dementia rats. Neurosci Lett 2023; 814:137439. [PMID: 37579868 DOI: 10.1016/j.neulet.2023.137439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
20(S)-protopanaxadiol (PPD), one of the ginsenosides from Panax ginseng, has been reported to improve performance with dementia. This study aimed to investigate the neuroprotective effect of PPD attenuating NLRP3 inflammasome-mediated microglial pyroptosis in vascular dementia (VD) rats induced by bilateral common carotid artery ligation (2-VO). Male Sprague-Dawley rats (SPF, 150-180 g, n = 10/group) were randomly divided into PPD (20, 10, 5 mg/kg, subcutaneous injection once per day for 3 weeks), model, and vehicle-sham group. It was found that PPD significantly reversed 2-VO-induced cognitive impairment by decreasing escape latency and spontaneous alternation and increasing the number of crossing platforms, showing memory-improving effects. PPD improved the pathological morphology of brain tissue in VD rats. PPD significantly reduced the cerebral infarction area and the activation of microglia in the cortex and hippocampal DG, CA1, and CA3 area. Moreover, PPD could attenuate NLRP3 inflammasome-mediated microglial pyroptosis, inhibit the positive expression of NLRP3, decrease IL-1β, and IL-18 levels, and increase IL-10 levels in the brain cortex. PPD also significantly alleviated the neurotoxicity by decreasing the Aβ and p-Tau in hippocampal DG, CA1, and CA3 areas. In addition, the levels of NLRP3, ASC, and IL-1β in the cortex, APP, BACE1, and p-Tau in the hippocampus were significantly reduced by PPD. These results suggested that PPD hinders microglial activation to alleviate neuroinflammation of NLRP3 inflammasome and inhibits neurotoxicity of Aβ deposition and Tau phosphorylation in 2-VO-induced VD rats.
Collapse
Affiliation(s)
- Xue Wang
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ya-Jin Shi
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ting-Yuan Niu
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China
| | - Ting-Ting Chen
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Han-Bing Li
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China
| | - Su-Hui Wu
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China.
| | - Gen-Lin Li
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China.
| |
Collapse
|
57
|
Duan R, Hou J, Wang X, Huang Z, Cao H, Hu J, Peng Q, Duan H, Wang Q, Chen X. Prevotella histicola Transplantation Ameliorates Cognitive Impairment and Decreases Oxidative Stress in Vascular Dementia Rats. Brain Sci 2023; 13:1136. [PMID: 37626492 PMCID: PMC10452631 DOI: 10.3390/brainsci13081136] [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: 07/03/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Vascular dementia is a type of dementia from brain damage caused by cerebrovascular lesions and vascular risk factors. Prevotella histicola is a species of Prevotella, belonging to the category of obligate anaerobe. The purpose of our work was to study the protection of Prevotella histicola on cognitive function in rats subjected to vascular dementia (VaD) and investigate underlying molecular mechanisms. The rats were randomly divided into three groups: control group, 2VO group and 2VO + Prevotella histicola group. The VaD rats (the 2VO group and 2VO + Prevotella histicola group) were generated by bilateral common carotid artery occlusion (2VO). Rats in the 2VO+ Prevotella histicola group were administered with Prevotella histicola twice daily. In comparison with the rats in the 2VO group, rats in the 2VO + Prevotella histicola group presented an enhanced cognitive ability, increased synapse-associated protein expression, a downregulation of proinflammatory factors and an upregulation of neurotrophic factors. The relevant mechanism of the protective effect of Prevotella histicola may be associated with the inhibition of glial cell-associated inflammation by regulating phosphorylation of CaMKII. In conclusion, Prevotella histicola attenuates neurological impairments via regulating synapse-associated protein expression and the liberation of inflammatory elements in vascular dementia rats. The findings above might benefit the development of Prevotella histicola transplantation as a promising treatment of VaD.
Collapse
Affiliation(s)
- Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| | - Jiankang Hou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| | - Xixi Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| | - Zhihang Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| | - Haiming Cao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| | - Junya Hu
- Department of Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210006, China; (J.H.); (H.D.)
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| | - Huijie Duan
- Department of Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210006, China; (J.H.); (H.D.)
| | - Qingguang Wang
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Wuxi 214400, China
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (R.D.); (J.H.); (X.W.); (Z.H.); (H.C.); (Q.P.)
| |
Collapse
|
58
|
Abi-Ghanem C, Salinero AE, Kordit D, Mansour FM, Kelly RD, Venkataganesh H, Kyaw NR, Gannon OJ, Riccio D, Fredman G, Poitelon Y, Belin S, Kopec AM, Robison LS, Zuloaga KL. Sex differences in the effects of high fat diet on underlying neuropathology in a mouse model of VCID. Biol Sex Differ 2023; 14:31. [PMID: 37208759 PMCID: PMC10199629 DOI: 10.1186/s13293-023-00513-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Damage to the cerebral vasculature can lead to vascular contributions to cognitive impairment and dementia (VCID). A reduction in blood flow to the brain leads to neuropathology, including neuroinflammation and white matter lesions that are a hallmark of VCID. Mid-life metabolic disease (obesity, prediabetes, or diabetes) is a risk factor for VCID which may be sex-dependent (female bias). METHODS We compared the effects of mid-life metabolic disease between males and females in a chronic cerebral hypoperfusion mouse model of VCID. C57BL/6J mice were fed a control or high fat (HF) diet starting at ~ 8.5 months of age. Three months after diet initiation, sham or unilateral carotid artery occlusion surgery (VCID model) was performed. Three months later, mice underwent behavior testing and brains were collected to assess pathology. RESULTS We have previously shown that in this VCID model, HF diet causes greater metabolic impairment and a wider array of cognitive deficits in females compared to males. Here, we report on sex differences in the underlying neuropathology, specifically white matter changes and neuroinflammation in several areas of the brain. White matter was negatively impacted by VCID in males and HF diet in females, with greater metabolic impairment correlating with less myelin markers in females only. High fat diet led to an increase in microglia activation in males but not in females. Further, HF diet led to a decrease in proinflammatory cytokines and pro-resolving mediator mRNA expression in females but not males. CONCLUSIONS The current study adds to our understanding of sex differences in underlying neuropathology of VCID in the presence of a common risk factor (obesity/prediabetes). This information is crucial for the development of effective, sex-specific therapeutic interventions for VCID.
Collapse
Affiliation(s)
- Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Kordit
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Febronia M Mansour
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Gabrielle Fredman
- Department Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Yannick Poitelon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Sophie Belin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Ashley M Kopec
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Lisa S Robison
- Department of Psychology & Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL, 33314, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
59
|
Zheng J, Peng S, Cui L, Liu X, Li T, Zhao Z, Li Y, Hu Y, Zhang M, Xu L, Zhang J. Enriched environment attenuates hippocampal theta and gamma rhythms dysfunction in chronic cerebral hypoperfusion via improving imbalanced neural afferent levels. Front Cell Neurosci 2023; 17:985246. [PMID: 37265581 PMCID: PMC10231328 DOI: 10.3389/fncel.2023.985246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/27/2023] [Indexed: 06/03/2023] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is increasingly recognized as a common cognitive impairment-causing mechanism. However, no clinically effective drugs to treat cognitive impairment due to CCH have been identified. An abnormal distribution of neural oscillations was found in the hippocampus of CCH rats. By releasing various neurotransmitters, distinct afferent fibers in the hippocampus influence neuronal oscillations in the hippocampus. Enriched environments (EE) are known to improve cognitive levels by modulating neurotransmitter homeostasis. Using EE as an intervention, we examined the levels of three classical neurotransmitters and the dynamics of neural oscillations in the hippocampus of the CCH rat model. The results showed that EE significantly improved the balance of three classical neurotransmitters (acetylcholine, glutamate, and GABA) in the hippocampus, enhanced the strength of theta and slow-gamma (SG) rhythms, and dramatically improved neural coupling across frequency bands in CCH rats. Furthermore, the expression of the three neurotransmitter vesicular transporters-vesicular acetylcholine transporters (VAChT) and vesicular GABA transporters (VGAT)-was significantly reduced in CCH rats, whereas the expression of vesicular glutamate transporter 1 (VGLUT1) was abnormally elevated. EE partially restored the expression of the three protein levels to maintain the balance of hippocampal afferent neurotransmitters. More importantly, causal mediation analysis showed EE increased the power of theta rhythm by increasing the level of VAChT and VGAT, which then enhanced the phase amplitude coupling of theta-SG and finally led to an improvement in the cognitive level of CCH. These findings shed light on the role of CCH in the disruption of hippocampal afferent neurotransmitter balance and neural oscillations. This study has implications for our knowledge of disease pathways.
Collapse
Affiliation(s)
- Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sisi Peng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingling Cui
- Department of Anesthesiology, Tongren Hospital of Wuhan University, Wuhan, China
| | - Xi Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Li
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Zhenyu Zhao
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Yaqing Li
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miao Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linling Xu
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - JunJian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
60
|
Chai YL, Rajeev V, Poh L, Selvaraji S, Hilal S, Chen CP, Jo DG, Koo EH, Arumugam TV, Lai MKP. Chronic cerebral hypoperfusion alters the CypA-EMMPRIN-gelatinase pathway: Implications for vascular dementia. J Cereb Blood Flow Metab 2023; 43:722-735. [PMID: 36537035 PMCID: PMC10108186 DOI: 10.1177/0271678x221146401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 03/21/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) is postulated to underlie multiple pathophysiological processes in vascular dementia (VaD), including extracellular matrix dysfunction. While several extracellular matrix proteins, namely cyclophilin A (CypA), extracellular matrix metalloproteinase inducer (EMMPRIN) and gelatinases (matrix metalloproteinases, MMP-2 and -9) have been investigated in acute stroke, their involvement in CCH and VaD remains unclear. In this study, CypA-EMMPRIN-gelatinase proteins were analysed in a clinical cohort of 36 aged, cognitively unimpaired subjects and 48 VaD patients, as well as in a bilateral carotid artery stenosis mouse model of CCH. Lower CypA and higher EMMPRIN levels were found in both VaD serum and CCH mouse brain. Furthermore, gelatinases were differentially altered in CCH mice and VaD patients, with significant MMP-2 increase in CCH brain and serum, whilst serum MMP-9 was elevated in VaD but reduced in CCH, suggesting complex CypA-EMMPRIN-gelatinase regulatory mechanisms. Interestingly, subjects with cortical infarcts had higher serum MMP-2, while white matter hyperintensities, cortical infarcts and lacunes were associated with higher serum MMP-9. Taken together, our data indicate that perturbations of CypA-EMMPRIN signalling may be associated with gelatinase-mediated vascular sequelae, highlighting the potential utility of the CypA-EMMPRIN-gelatinase pathway as clinical biomarkers and therapeutic targets in VaD.
Collapse
Affiliation(s)
- Yuek Ling Chai
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| | - Vismitha Rajeev
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Luting Poh
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Saw Swee Hock School of Public
Health, National University of Singapore, Kent Ridge, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan
University, Suwon, Republic of Korea
| | - Edward H Koo
- Department of Medicine, National
University of Singapore, Kent Ridge, Singapore
- Graduate School for Integrative
Sciences and Engineering, National University of Singapore, Kent Ridge,
Singapore
- Department of Neurosciences,
University of California San Diego, San Diego, CA, USA
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan
University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology
and Disease Research, Department of Microbiology, Anatomy, Physiology and
Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe
University, Bundoora, VIC, Australia
| | - Mitchell KP Lai
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| |
Collapse
|
61
|
Zhang WJ, Li KY, Lan Y, Zeng HY, Chen SQ, Wang H. NLRP3 Inflammasome: A key contributor to the inflammation formation. Food Chem Toxicol 2023; 174:113683. [PMID: 36809826 DOI: 10.1016/j.fct.2023.113683] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/04/2023] [Accepted: 02/17/2023] [Indexed: 02/21/2023]
Abstract
Inflammation is an important part of the development of various organ diseases. The inflammasome, as an innate immune receptor, plays an important role in the formation of inflammation. Among various inflammasomes, the NLRP3 inflammasome is the most well studied. The NLRP3 inflammasome is composed of skeletal protein NLRP3, apoptosis-associated speck-like protein (ASC) and pro-caspase-1. There are three types of activation pathways: (1) "classical" activation pathway; (2) "non-canonical" activation pathway; (3) "alternative" activation pathway. The activation of NLRP3 inflammasome is involved in many inflammatory diseases. A variety of factors (such as genetic factors, environmental factors, chemical factors, viral infection, etc.) have been proved to activate NLRP3 inflammasome and promote the inflammatory response of the lung, heart, liver, kidney and other organs in the body. Especially, the mechanism of NLRP3 inflammation and its related molecules in its associated diseases remains not to be summarized, namely they may promote or delay inflammatory diseases in different cells and tissues. This article reviews the structure and function of the NLRP3 inflammasome and its role in various inflammations, including inflammations caused by chemically toxic substances.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Ke-Yun Li
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Yi Lan
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Han-Yi Zeng
- Department of Genetics, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Shui-Qin Chen
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, PR China.
| |
Collapse
|
62
|
Analysis of Potential Mechanism of Herbal Formula Taohong Siwu Decoction against Vascular Dementia Based on Network Pharmacology and Molecular Docking. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1235552. [PMID: 36726841 PMCID: PMC9886489 DOI: 10.1155/2023/1235552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/22/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023]
Abstract
Vascular dementia (VaD) is the second most prevalent dementia, which is attributable to neurovascular dysfunction. Currently, no approved pharmaceuticals are available. Taohong Siwu decoction (TSD) is a traditional Chinese medicine prescription with powerful antiapoptosis and anti-inflammatory properties. In this study, a network pharmacology approach together with molecular docking validation was used to explore the probable mechanism of action of TSD against VaD. A total of 44 active components, 202 potential targets of components, and 3,613 VaD-related targets including 161 intersecting were obtained. The potential chemical components including kaempferol, baicalein, beta-carotene, luteolin, quercetin, and beta-sitosterol involved in the inflammatory response, oxidative stress, and apoptosis might have potential therapeutic effects on the treatment of VaD. The potential core targets including AKT1, CASP3, IL1β, JUN, and TP53 associated with cell apoptosis and inflammatory might account for the essential therapeutic effects of TSD in VaD. The results indicated that TSD protected against VaD through multicomponent and multitarget modes. Though the detailed mechanism of action of various active ingredients needs to be further illustrated, TSD still showed a promising therapeutic agent for VaD due to its biological activity.
Collapse
|
63
|
Chong JR, Hilal S, Ashton NJ, Karikari TK, Reilhac A, Vrooman H, Schöll M, Zetterberg H, Blennow K, Chen CP, Lai MKP. Brain atrophy and white matter hyperintensities are independently associated with plasma neurofilament light chain in an Asian cohort of cognitively impaired patients with concomitant cerebral small vessel disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12396. [PMID: 36994314 PMCID: PMC10040495 DOI: 10.1002/dad2.12396] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 03/28/2023]
Abstract
Introduction Plasma neurofilament light chain (NfL) is a potential biomarker for neurodegeneration in Alzheimer's disease (AD), ischemic stroke, and non-dementia cohorts with cerebral small vessel disease (CSVD). However, studies of AD in populations with high prevalence of concomitant CSVD to evaluate associations of brain atrophy, CSVD, and amyloid beta (Aβ) burden on plasma NfL are lacking. Methods Associations were tested between plasma NfL and brain Aβ, medial temporal lobe atrophy (MTA) as well as neuroimaging features of CSVD, including white matter hyperintensities (WMH), lacunes, and cerebral microbleeds. Results We found that participants with either MTA (defined as MTA score ≥2; neurodegeneration [N]+WMH-) or WMH (cut-off for log-transformed WMH volume at 50th percentile; N-WMH+) manifested increased plasma NfL levels. Participants with both pathologies (N+WMH+) showed the highest NfL compared to N+WMH-, N-WMH+, and N-WMH- individuals. Discussion Plasma NfL has potential utility in stratifying individual and combined contributions of AD pathology and CSVD to cognitive impairment.
Collapse
Affiliation(s)
- Joyce R. Chong
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Saima Hilal
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemKent RidgeSingapore
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- King's College LondonInstitute of PsychiatryPsychology and NeuroscienceMaurice Wohl Institute Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
| | - Thomas K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Anthonin Reilhac
- Clinical Imaging Research CentreYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
| | - Henri Vrooman
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | - Michael Schöll
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
- Hong Kong Center for Neurodegenerative Diseasesthe Hong Kong University of Science and TechnologyHong Kong Science ParkShatinNew TerritoriesHong Kong SARChina
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
| | - Christopher P. Chen
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Department of Psychological MedicineYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
| | - Mitchell K. P. Lai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| |
Collapse
|
64
|
Li B, Du B, Gu Z, Wu C, Tan Y, Song C, Xu Y, Yin G, Gao X, Wang W, Sun X, Bi X. Correlations among peripheral blood markers, white matter hyperintensity, and cognitive function in patients with non-disabling ischemic cerebrovascular events. Front Aging Neurosci 2022; 14:1023195. [PMID: 36533171 PMCID: PMC9755852 DOI: 10.3389/fnagi.2022.1023195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/18/2022] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Both inflammation and cerebral white matter injury are closely associated with vascular cognitive impairment (VCI). The aim of this study was to analyze the correlation between peripheral serological markers, white matter injury, and cognitive function in patients with non-disabling ischemic cerebrovascular events (NICE); to identify potential biological markers for the diagnosis and prediction of VCI; and to provide a basis for the early diagnosis and intervention of VCI. METHODS We collected clinical data, along with demographic and medical history data, from 151 NICE patients. Fasting venous blood samples were collected. Based on the Montreal Cognitive Assessment (MoCA) after admission, we divided the patients into normal cognitive function (NCF) and VCI groups, and then classified them into mild white matter hyperintensity (mWMH) and severe white matter hyperintensity (sWMH) based on Fazekas scores. The differences in serological marker levels were compared between the cognitive function groups and the white matter hyperintensity groups. Binary logistic regression models and receiver operating characteristic curves were used to analyze the diagnostic predictive value of serological markers for VCI in patients with NICE and in the white matter hyperintensity subgroups. RESULTS Among 151 patients with NICE, 95 were male and 56 were female. Lymphocyte count (OR = 0.405, p = 0.010, 95% CI [0.201, 0.806]), red blood cell count (OR = 0.433, p = 0.010, 95% CI [0.228, 0.821]), and hemoglobin level (OR = 0.979, p = 0.046, 95% CI [0.958, 0.999]) were protective factors for cognitive function in patients with NICE. The sWMH group had a higher age, granulocyte/lymphoid ratio (NLR), and neutrophil percentage but a lower MoCA score, hemoglobin level, and lymphocyte count than the mWMH group. In the mWMH group, lymphocyte count (AUC = 0.713, p = 0.003, 95% CI [0.593, 0.833]) had an acceptable predictive value for the diagnosis of VCI, whereas white blood cell count (AUC = 0.672, p = 0.011, 95% CI [0.545, 0.799]), red blood cell count (AUC = 0.665, p = 0.014, 95% CI [0.545, 0.784]), and hemoglobin level (AUC = 0.634, p = 0.047, 95% CI [0.502, 0.765]) had marginal predictive value for the diagnosis of VCI. In the sWMH group, no significant differences were found in serological markers between the NCF and VCI groups. CONCLUSION Lymphocyte count, red blood cell count, and hemoglobin level were independent protective factors for cognitive function in patients with NICE; they can be used as potential biological markers to distinguish VCI in patients with NICE and are applicable to subgroups of patients with mWMH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xu Sun
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
65
|
Tregub PP, Averchuk AS, Baranich TI, Ryazanova MV, Salmina AB. Physiological and Pathological Remodeling of Cerebral Microvessels. Int J Mol Sci 2022; 23:12683. [PMID: 36293539 PMCID: PMC9603917 DOI: 10.3390/ijms232012683] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
There is growing evidence that the remodeling of cerebral microvessels plays an important role in plastic changes in the brain associated with development, experience, learning, and memory consolidation. At the same time, abnormal neoangiogenesis, and deregulated regulation of microvascular regression, or pruning, could contribute to the pathogenesis of neurodevelopmental diseases, stroke, and neurodegeneration. Aberrant remodeling of microvesselsis associated with blood-brain barrier breakdown, development of neuroinflammation, inadequate microcirculation in active brain regions, and leads to the dysfunction of the neurovascular unit and progressive neurological deficits. In this review, we summarize current data on the mechanisms of blood vessel regression and pruning in brain plasticity and in Alzheimer's-type neurodegeneration. We discuss some novel approaches to modulating cerebral remodeling and preventing degeneration-coupled aberrant microvascular activity in chronic neurodegeneration.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Federal State Budgetary Scientific Institution Research Center of Neurology, 125367 Moscow, Russia
| | | | | | | | | |
Collapse
|
66
|
Zhou Q, Le M, Yang Y, Wang W, Huang Y, Wang Q, Tian Y, Jiang M, Rao Y, Luo HB, Wu Y. Discovery of novel phosphodiesterase-1 inhibitors for curing vascular dementia: suppression of neuroinflammation by blocking NF-κB transcription regulation and activating cAMP/CREB axis. Acta Pharm Sin B 2022; 13:1180-1191. [PMID: 36970192 PMCID: PMC10031254 DOI: 10.1016/j.apsb.2022.09.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/01/2022] Open
Abstract
Vascular dementia (VaD) is the second commonest type of dementia which lacks of efficient treatments currently. Neuroinflammation as a prominent pathological feature of VaD, is highly involved in the development of VaD. In order to verify the therapeutic potential of PDE1 inhibitors against VaD, the anti-neuroinflammation, memory and cognitive improvement were evaluated in vitro and in vivo by a potent and selective PDE1 inhibitor 4a. Also, the mechanism of 4a in ameliorating neuroinflammation and VaD was systematically explored. Furthermore, to optimize the drug-like properties of 4a, especially for metabolic stability, 15 derivatives were designed and synthesized. As a result, candidate 5f, with a potent IC50 value of 4.5 nmol/L against PDE1C, high selectivity over PDEs, and remarkable metabolic stability, efficiently ameliorated neuron degeneration, cognition and memory impairment in VaD mice model by suppressing NF-κB transcription regulation and activating cAMP/CREB axis. These results further identified PDE1 inhibition could serve as a new therapeutic strategy for treatment of VaD.
Collapse
|
67
|
Jenkins TA. Metabolic Syndrome and Vascular-Associated Cognitive Impairment: a Focus on Preclinical Investigations. Curr Diab Rep 2022; 22:333-340. [PMID: 35737273 PMCID: PMC9314301 DOI: 10.1007/s11892-022-01475-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Metabolic syndrome is associated with an increased risk of vascular cognitive impairment or, in the more extreme, vascular dementia. Animal models are used to investigate the relationship between pathology and behaviour. This review summarizes the latest understanding of the role of the hippocampus and prefrontal cortex in vascular cognitive impairment, the influence of inflammation in this association while also commenting on some of the latest interventions proposed. RECENT FINDINGS Models of vascular cognitive impairment and vascular dementia, whether they develop from an infarct or non-infarct base, demonstrate increased neuroinflammation, reduced neuronal function and deficits in prefrontal and hippocampal-associated cognitive domains. Promising new research shows agents and environmental interventions that inhibit central oxidative stress and inflammation can reverse both pathology and cognitive dysfunction. While preclinical studies suggest that reversal of deficits in vascular cognitive impairment models is possible, replication in patients still needs to be demonstrated.
Collapse
Affiliation(s)
- Trisha A Jenkins
- Human Biosciences, School of Health and Biomedical Sciences, STEM College, RMIT University, Plenty Road, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
68
|
P2X7R/NLRP3 signaling pathway-mediated pyroptosis and neuroinflammation contributed to cognitive impairment in a mouse model of migraine. J Headache Pain 2022; 23:75. [PMID: 35780081 PMCID: PMC9250730 DOI: 10.1186/s10194-022-01442-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/10/2022] [Indexed: 01/15/2023] Open
Abstract
Migraine is the second most common form of headache disorder and the second leading cause of disability worldwide. Cognitive symptoms ranked second resulting in migraine-related disability, after pain. P2X7 receptor (P2X7R) was recently shown to be involved in hyperalgesia in migraine. However, the role of P2X7R in migraine-related cognitive impairment is still ill-defined. The aim of this study was to explore the molecular mechanisms underlying migraine-related cognitive impairment and the role of P2X7R in it. Here we used a well-established mouse model of migraine that triggered migraine attacks by application of inflammatory soup (IS) to the dura. Our results showed that repeated dural IS stimulation triggered upregulation of P2X7R, activation of NLRP3 inflammasome, release of proinflammatory cytokines (IL-1β and IL-18) and activation of pyroptotic cell death pathway. Gliosis (microgliosis and astrogliosis), neuronal loss and cognitive impairment also occurred in the IS-induced migraine model. No significant apoptosis or whiter matter damage was observed following IS-induced migraine attacks. These pathological changes occurred mainly in the cerebral cortex and to a less extent in the hippocampus, all of which can be prevented by pretreatment with a specific P2X7R antagonist Brilliant Blue G (BBG). Moreover, BBG can alleviate cognitive impairment following dural IS stimulation. These results identified P2X7R as a key contributor to migraine-related cognitive impairment and may represent a potential therapeutic target for mitigating cognitive impairment in migraine.
Collapse
|
69
|
Effect of Yisui Multipurpose Soup's Amelioration on D-Galactose-Induced Neuronal Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3372350. [PMID: 35754679 PMCID: PMC9217601 DOI: 10.1155/2022/3372350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022]
Abstract
This study clarified the regulatory effect of Yisui multipurpose Soup towards D-galactose-induced cognitive impairment cell model on the molecular level. We first constructed and cultured the cell model of cognitive impairment induced by D-galactose in neurons in vitro and then cultured the cells in the medium supplemented with different doses of drug-containing serum of Yisui multipurpose soup. Expressions of inflammatory cytokine tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), nitric oxide (NO), and interleukin-1β (IL-1β) were assessed by the ELISA and western blot, and cell apoptosis was determined by flow cytometry and TUNEL. The expression changes of apoptosis-related proteins Bcl-2 and Bax were estimated by immunofluorescence, qPCR, and western blot. Finally, we analyzed and made the network interaction diagram of Yisui multipurpose soup-components-targets through the network pharmacology method, from which we could learn that there were 1104 gene targets related to vascular cognitive impairment (VCI) and 1071 component targets of Yisui multipurpose soup. And there were 251 overlapping genes, mainly gathering in protein binding, protein modification, MAPK signaling pathway, and calcium signaling pathway. The expressions of TNF-α, iNOS, NO, and IL-1β were significantly decreased after the culture medium was replaced by medium containing drug serum. We also found that the effect of high-dose drug-containing serum on the expression of inflammatory factors was better than that of low dose. The Yisui multipurpose soup drug serum in the medium not only significantly increased Bcl-2 expression and effectively reduced Bax expression, but also inhibited the apoptosis of neurons induced by D-galactose. In conclusion, Yisui multipurpose soup could effectively protect D-galactose-induced neuronal cell cognitive impairment by orchestrating expressions of the inflammatory factors TNF-α, iNOS, NO, and IL-1β and the apoptosis-related proteins Bcl-2 and Bax.
Collapse
|
70
|
Discovering the Effects of Fisetin on NF-κB/NLRP-3/NRF-2 Molecular Pathways in a Mouse Model of Vascular Dementia Induced by Repeated Bilateral Carotid Occlusion. Biomedicines 2022; 10:biomedicines10061448. [PMID: 35740470 PMCID: PMC9221103 DOI: 10.3390/biomedicines10061448] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 12/26/2022] Open
Abstract
Vascular dementia (VaD) is the second leading cause of dementia. The majority of VaD patients have cognitive abnormalities, which are caused by cerebral hypoperfusion-induced ischemia, endothelial dysfunction, oxidative stress, and neuroinflammation. Natural products are receiving increasing attention for the treatment of neuroinflammatory diseases. The aim of this study was to investigate the molecular pathways underlying the protective effects of fisetin, a flavonoid present in many fruits and vegetables, in a mouse model of VaD induced by repeated ischemia-reperfusion (IR) of the total bilateral carotid artery. Here, we found that VaD caused brain injury, lipid peroxidation, and neuronal death in the hippocampus, as well as astrocyte and microglial activation, and reduced BDNF neurotrophic factor expression together with behavioral alterations. In addition, VaD induced the activation of inflammasome components (NLRP-3, ASC, and caspase 1), and their downstream products (IL-1β and IL-18) release and promote activation of apoptotic cell death. Fisetin attenuated histological injury, malondialdehyde levels, inflammasome pathway activation, apoptosis, as well as increased BDNF expression, reduced astrocyte, microglial activation, and cognitive deficits. In conclusion, the protective effects of fisetin could be due to the inhibition of the ROS-induced activation of NF-κB/NLRP3 inflammasome together with the activation of antioxidant Nrf2/HO-1, suggesting a possible crosstalk between these molecular pathways.
Collapse
|
71
|
Ballerini C, Njamnshi AK, Juliano SL, Kalaria RN, Furlan R, Akinyemi RO. Non-Communicable Neurological Disorders and Neuroinflammation. Front Immunol 2022; 13:834424. [PMID: 35769472 PMCID: PMC9235309 DOI: 10.3389/fimmu.2022.834424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/10/2022] [Indexed: 12/04/2022] Open
Abstract
Traumatic brain injury, stroke, and neurodegenerative diseases represent a major cause of morbidity and mortality in Africa, as in the rest of the world. Traumatic brain and spinal cord injuries specifically represent a leading cause of disability in the younger population. Stroke and neurodegenerative disorders predominantly target the elderly and are a major concern in Africa, since their rate of increase among the ageing is the fastest in the world. Neuroimmunology is usually not associated with non-communicable neurological disorders, as the role of neuroinflammation is not often considered when evaluating their cause and pathogenesis. However, substantial evidence indicates that neuroinflammation is extremely relevant in determining the consequences of non-communicable neurological disorders, both for its protective abilities as well as for its destructive capacity. We review here current knowledge on the contribution of neuroinflammation and neuroimmunology to the pathogenesis of traumatic injuries, stroke and neurodegenerative diseases, with a particular focus on problems that are already a major issue in Africa, like traumatic brain injury, and on emerging disorders such as dementias.
Collapse
Affiliation(s)
- Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alfred K. Njamnshi
- Brain Research Africa Initiative (BRAIN); Neurology Department, Central Hospital Yaounde/Faculty of Medicine and Biomedical Sciences (FMBS), The University of Yaounde 1, Yaounde, Cameroon
| | - Sharon L. Juliano
- Neuroscience, Uniformed Services University Hebert School (USUHS), Bethesda, MD, United States
| | - Rajesh N. Kalaria
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
- *Correspondence: Roberto Furlan, ; Rufus O. Akinyemi,
| | - Rufus O. Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Roberto Furlan, ; Rufus O. Akinyemi,
| |
Collapse
|
72
|
Berberine protects against chronic cerebral hypoperfusion-induced cognitive impairment and hippocampal damage via regulation of the ERK/Nrf2 pathway. J Chem Neuroanat 2022; 123:102119. [PMID: 35697268 DOI: 10.1016/j.jchemneu.2022.102119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/23/2022]
Abstract
Vascular cognitive impairment caused by chronic cerebral hypoperfusion (CCH) seriously affects the quality of life of elderly patients and places a great burden on society and family. With the development of traditional Chinese medicine (TCM), TCM approaches to the prevention and treatment of senile ischemic cerebrovascular disease has received increasing attention. In this study, rats with bilateral common carotid artery occlusion (BCCAO) were treated with berberine (BBR). Their learning and memory function, neuronal injury and repair, the extracellular regulatory protein kinase (ERK)/nuclear factor-E2-related factor 2 (Nrf2) signaling pathway, and impairment and improvement of the blood-brain barrier (BBB) were evaluated. This study found that BBR can alleviate the pathological injury to the brain, reduce neuronal loss and promote neuronal cell survival after CCH by interfering with the ERK/Nrf2 signaling pathway. BBR can reduce BBB injury in CCH rats by inhibiting the expression of VEGF-A and MMP-9 in plasma, which reveals a protective effect of BBR on vascular cognitive impairment. This study provides a new research direction for BBR in the treatment of ischemic cerebrovascular disease.
Collapse
|