51
|
Jia H, Qi X, Fu L, Wu H, Shang J, Qu M, Yang C, Wang J. NLRP3
inflammasome inhibitor ameliorates ischemic stroke by reprogramming the phenotype of microglia/macrophage in a murine model of distal middle cerebral artery occlusion. Neuropathology 2022; 42:181-189. [PMID: 35434787 DOI: 10.1111/neup.12802] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/31/2022]
Affiliation(s)
- Hongning Jia
- Department of Neurology Cangzhou Central Hospital Cangzhou China
| | - Xiaoyuan Qi
- Department of Neurology Cangzhou Central Hospital Cangzhou China
| | - Lan Fu
- Department of Imaging Cangzhou Central Hospital Cangzhou China
| | - Huijun Wu
- Department of Neurology Cangzhou Central Hospital Cangzhou China
| | - Jinxing Shang
- Department of Neurosurgery Cangzhou Central Hospital Cangzhou China
| | - Mingwei Qu
- Department of Neurology Cangzhou Central Hospital Cangzhou China
| | - Chaoping Yang
- Department of Neurology Cangzhou Central Hospital Cangzhou China
| | - Jianping Wang
- Department of Neurology Cangzhou Central Hospital Cangzhou China
| |
Collapse
|
52
|
Zhu T, Wang L, Wang LP, Wan Q. Therapeutic targets of neuroprotection and neurorestoration in ischemic stroke: Applications for natural compounds from medicinal herbs. Biomed Pharmacother 2022; 148:112719. [DOI: 10.1016/j.biopha.2022.112719] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
|
53
|
Wang L, Fan X, Chen Y, Liang X, Shen W, Zhang Y. Efficacy and Safety of Xingnaojing Injection for Emergency Treatment of Acute Ischemic Stroke: A Systematic Review and Meta-Analysis. Front Pharmacol 2022; 13:839305. [PMID: 35401232 PMCID: PMC8987164 DOI: 10.3389/fphar.2022.839305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/21/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Xingnaojing injection (XNJ) is derived from a traditional Chinese prescription named Angong Niuhuang pill. As an adjuvant treatment widely used in acute ischemic stroke (AIS), XNJ has proven to be effective with certain clinical evidence. The aim of this study is to collect the latest evidence and evaluate efficacy and safety of XNJ for emergency treatment of AIS. Methods: We searched seven literature databases and two clinical trial registries from their inception to November 14, 2021 for randomized controlled trials (RCTs) examining the efficacy of XNJ for AIS. Two reviewers independently selected relevant trials, extracted data, and assessed the risk of bias. We pooled data into a meta-analysis using RevMan 5.4 software. Results: Thirty-eight RCTs were included in this review, with a total of 3,677 participants. XNJ plus conventional treatments (CTs) showed a significant advantage, compared with CTs alone, in improving functional independence at 14 days (RR = 1.70, 95% CI = 1.03 to 2.81, p = 0.04), neurological function (MDNIHSS < 6h = −3.81, 95% CI = −5.25 to −2.38, p < 0.00001; MDNIHSS < 24h = −3.75, 95% CI = −4.92 to −2.59, p < 0.00001; MDNIHSS < 72h = −3.74, 95% CI = −5.48 to −2.00, p < 0.0001; MDNIHSS < 14d = −1.97, 95% CI = −3.25 to −0.69, p = 0.003), and activities of daily living on the Barthel index (MDBI-14day = 9.97, 95% CI = 9.29 to 10.65, p < 0.00001; MDBI-30day = 10.04, 95% CI = 5.82, to 14.26, p < 0.00001). In addition, the results showed that XNJ plus CTs was superior to CTs alone in reducing IL-6, TNF-α, hs-CRP, and MMP-9. Regarding safety of XNJ, the incidence of adverse reactions in the XNJ group was lower than that in the control group (RR = 0.57, 95% CI = 0.38 to 0.87, p = 0.009). The certainty of evidence was evaluated as low or very low for all. Conclusion: XNJ appears to be effective and safe for emergency treatment of AIS. The first 72 h after the onset of stroke, in particular the first 6 hours, may be the optimum initiation time. However, further high-quality RCTs are warranted to determine an appropriate initiation time. Systematic Review Registration: [https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=233211], identifier [CRD42021233211].
Collapse
Affiliation(s)
| | | | | | | | - Wei Shen
- *Correspondence: Wei Shen, ; Yunling Zhang,
| | | |
Collapse
|
54
|
Gao T, Huang F, Wang W, Xie Y, Wang B. Interleukin-10 genetically modified clinical-grade mesenchymal stromal cells markedly reinforced functional recovery after spinal cord injury via directing alternative activation of macrophages. Cell Mol Biol Lett 2022; 27:27. [PMID: 35300585 PMCID: PMC8931978 DOI: 10.1186/s11658-022-00325-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Background After spinal cord injury (SCI), dysregulated or nonresolving inflammatory processes can severely disturb neuronal homeostasis and drive neurodegeneration. Although mesenchymal stromal cell (MSC)-based therapies have showed certain therapeutic efficacy, no MSC therapy has reached its full clinical goal. In this study, we examine interleukin-10 (IL10) genetically modified clinical-grade MSCs (IL10-MSCs) and evaluate their clinical safety, effectiveness, and therapeutic mechanism in a completely transected SCI mouse model. Methods We established stable IL10-overexpressing human umbilical-cord-derived MSCs through electric transduction and screened out clinical-grade IL10-MSCs according to the criteria of cell-based therapeutic products, which were applied to mice with completely transected SCI by repeated tail intravenous injections. Then we comprehensively investigated the motor function, histological structure, and nerve regeneration in SCI mice, and further explored the potential therapeutic mechanism after IL10-MSC treatment. Results IL10-MSC treatment markedly reinforced locomotor improvement, accompanied with decreased lesion volume, regeneration of axons, and preservation of neurons, compared with naïve unmodified MSCs. Further, IL10-MSC transplantation increased the ratio of microglia to infiltrated alternatively activated macrophages (M2), and reduced the ratio of classically activated macrophages (M1) at the injured spinal cord, meanwhile increasing the percentage of Treg and Th2 cells, and reducing the percentage of Th1 cells in the peripheral circulatory system. In addition, IL10-MSC administration could prevent apoptosis and promote neuron differentiation of neural stem cells (NSCs) under inflammatory conditions in vitro. Conclusions IL10-MSCs exhibited a reliable safety profile and demonstrated promising therapeutic efficacy in SCI compared with naïve MSCs, providing solid support for future clinical application of genetically engineered MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00325-9.
Collapse
Affiliation(s)
- Tianyun Gao
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Feifei Huang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Wenqing Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuanyuan Xie
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
55
|
A Single Administration of Riluzole Applied Acutely After Spinal Cord Injury Attenuates Pro-inflammatory Activity and Improves Long-Term Functional Recovery in Rats. J Mol Neurosci 2022; 72:730-740. [PMID: 34988900 DOI: 10.1007/s12031-021-01947-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/16/2021] [Indexed: 10/19/2022]
Abstract
After spinal cord injury (SCI), emergency treatment intervention can minimize tissue damage, which is closely related to the recovery of long-term function. Here, we examined whether the administration of a single dose of riluzole (6 mg/kg) immediately after SCI was a critical window for the drug to exert its regulatory effect and limit long-term neurological deficits. The animals were sacrificed 1 day after administration for investigation of neuronal survival and a potential neuroinflammatory response, and sacrificed in the 6th week for assessment of neurological function. Riluzole applied in a single dose immediately post-SCI decreased the mRNA level of interleukin-1β at 6 h, reduced the destruction of neurons, and reduced the activation of microglia/macrophage M1 expression at day 1 post-SCI. Additionally, riluzole-treated rats showed higher expressions of interleukin-33 and its receptor ST2 in microglia/macrophages of the spinal cord than vehicle-treated rats, suggesting that this signaling pathway might be involved in microglia/macrophage-mediated inflammation. At 6 weeks, riluzole-treated rats exhibited higher motor function scores than vehicle-treated controls. In addition, riluzole-treated rats exhibited higher expression of GAP43 protein and shorter N1 peak latency and larger N1-P1 amplitude in motor-evoked potentials, compared to vehicle-treated rats. Together, these data suggested that early application of riluzole after SCI could be crucial for long-term functional recovery, so it may represent a promising therapeutic candidate within the critical therapeutic window for acute SCI.
Collapse
|
56
|
Cianciulli A, Calvello R, Ruggiero M, Panaro MA. Inflammaging and Brain: Curcumin and Its Beneficial Potential as Regulator of Microglia Activation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020341. [PMID: 35056657 PMCID: PMC8780663 DOI: 10.3390/molecules27020341] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 01/09/2023]
Abstract
Inflammaging is a term used to describe the tight relationship between low-grade chronic inflammation and aging that occurs during physiological aging in the absence of evident infection. This condition has been linked to a broad spectrum of age-related disorders in various organs including the brain. Inflammaging represents a highly significant risk factor for the development and progression of age-related conditions, including neurodegenerative diseases which are characterized by the progressive dysfunction and degeneration of neurons in the brain and peripheral nervous system. Curcumin is a widely studied polyphenol isolated from Curcuma longa with a variety of pharmacologic properties. It is well-known for its healing properties and has been extensively used in Asian medicine to treat a variety of illness conditions. The number of studies that suggest beneficial effects of curcumin on brain pathologies and age-related diseases is increasing. Curcumin is able to inhibit the formation of reactive-oxygen species and other pro-inflammatory mediators that are believed to play a pivotal role in many age-related diseases. Curcumin has been recently proposed as a potential useful remedy against neurodegenerative disorders and brain ageing. In light of this, our current review aims to discuss the potential positive effects of Curcumin on the possibility to control inflammaging emphasizing the possible modulation of inflammaging processes in neurodegenerative diseases.
Collapse
|
57
|
Li R, Zhou Y, Zhang S, Li J, Zheng Y, Fan X. The natural (poly)phenols as modulators of microglia polarization via TLR4/NF-κB pathway exert anti-inflammatory activity in ischemic stroke. Eur J Pharmacol 2022; 914:174660. [PMID: 34863710 DOI: 10.1016/j.ejphar.2021.174660] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/04/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidences suggest that inflammation plays a key role in the pathogenesis of stroke, a devastating disease second only to cardiac ischemia as a cause of death worldwide. Microglia are the first non-neuronal cells on the scene during the innate immune response to acute ischemic stroke. Microglia respond to acute brain injury by activating and developing classic M1-like (pro-inflammatory) or alternative M2-like (anti-inflammatory) phenotypes. M1 microglia produce pro-inflammatory cytokines to exacerbate neural death, astrocyte apoptosis, and blood brain barrier (BBB) disruption, while M2 microglia play the opposite role. NF-κB, a central regulator of the inflammatory response, was responsible for microglia M1 and M2 polarization. NF-κB p65 and p50 form a heterodimer to initiate a pro-inflammatory cytokine response, which enhances M1 activation and impair M2 response of microglia. TLR4, expressed on the surface of microglia, plays an important role in activating NF-κB, ultimately causing the M1 response of microglia. Therefore, modulation of microglial phenotypes via TLR4/NF-κB signaling pathway may be a promising therapeutic approach for ischemic stroke. Dietary (poly)phenols are present in various foods, which have shown promising protective effects on ischemic stroke. In vivo studies strongly suggest that many (poly)phenols have a pronounced impact on ischemic stroke, as demonstrated by lower neuroinflammation. Thus, this review focuses on the anti-inflammatory properties of dietary (poly)phenols and discusses their effects on the polarization of microglia through modulating TLR4/NF-κB signaling pathway in the ischemic stroke.
Collapse
Affiliation(s)
- Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jieying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
58
|
Nguyen HD, Oh H, Kim MS. The association between curry-rice consumption and hypertension, type 2 diabetes, and depression: The findings from KNHANES 2012-2016. Diabetes Metab Syndr 2022; 16:102378. [PMID: 34974329 DOI: 10.1016/j.dsx.2021.102378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND AIMS We aim to evaluate the association between curry-rice consumption, cardiovascular diseases (CVDs), type 2 diabetes (T2DB), arthritis, and depression. METHODS 17,625 participants aged ≥18 years were recruited to obtain data on sociodemographic characteristics, lifestyle, medical history, current medications, family history, and food consumption. The association between curry-rice consumption, CVDs, T2DB, arthritis, and depression was examined using multivariable-adjusted analyses. RESULTS In the logistic model, risks of elevated triglyceride (OR 0.89; 95% CI, 0.82-0.97, p = 0.006), elevated HbA1c (OR 0.81; 95% CI, 0.73-0.91, p < 0.001), and elevated glucose (OR 0.86; 95% CI, 0.79-0.94, p < 0.001) were significantly lower in the high curry-rice consumption group than in the low curry-rice consumption group. Risk of hypertension (OR 0.88; 95% CI, 0.78-0.98, p = 0.044), T2DB (OR, 0.82; 95% CI, 0.68-0.98, p < 0.001), and depression (OR 0.82; 95% CI, 0.70-0.97, p = 0.026) was significantly lower among the high curry-rice consumption group than in the low curry-rice consumption group. These findings were consistent with the results of the analysis when curry-rice consumption was treated as a continuous variable. CONCLUSIONS The potential health benefits resulting from the intake of curry-rice via an ordinary diet could protect the public from the burden of non-communicable diseases (NCDs) and mental health. These results highlight an ongoing need to understand the role of curry-rice in NCDs and mental health.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, Jeonnam, Republic of Korea
| | - Hojin Oh
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, Jeonnam, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, Jeonnam, Republic of Korea.
| |
Collapse
|
59
|
Kingsbury C, Shear A, Heyck M, Sadanandan N, Zhang H, Gonzales-Portillo B, Cozene B, Sheyner M, Navarro-Torres L, García-Sánchez J, Lee JY, Borlongan CV. Inflammation-relevant microbiome signature of the stroke brain, gut, spleen, and thymus and the impact of exercise. J Cereb Blood Flow Metab 2021; 41:3200-3212. [PMID: 34427146 PMCID: PMC8669279 DOI: 10.1177/0271678x211039598] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Stroke remains a significant unmet need in the clinic with few therapeutic options. We, and others, have implicated the role of inflammatory microbiota in stroke secondary cell death. Elucidating this inflammation microbiome as a biomarker may improve stroke diagnosis and treatment. Here, adult Sprague-Dawley rats performed 30 minutes of exercise on a motorized treadmill for 3 consecutive days prior to transient middle cerebral artery occlusion (MCAO). Stroke animals that underwent exercise showed 1) robust behavioral improvements, 2) significantly smaller infarct sizes and increased peri-infarct cell survival and 3) decreasing trends of inflammatory microbiota BAC303, EREC482, and LAB158 coupled with significantly reduced levels of inflammatory markers ionized calcium binding adaptor molecule 1, tumor necrosis factor alpha, and mouse monoclonal MHC Class II RT1B in the brain, gut, spleen, and thymus compared to non-exercised stroke rats. These results suggest that a specific set of inflammatory microbiota exists in central and peripheral organs and can serve as a disease biomarker and a therapeutic target for stroke.
Collapse
Affiliation(s)
- Chase Kingsbury
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alex Shear
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matt Heyck
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Henry Zhang
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bella Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Blaise Cozene
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michael Sheyner
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Lisset Navarro-Torres
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Julián García-Sánchez
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
60
|
Ran Y, Qie S, Gao F, Ding Z, Yang S, Tian G, Liu Z, Xi J. Baicalein ameliorates ischemic brain damage through suppressing proinflammatory microglia polarization via inhibiting the TLR4/NF-κB and STAT1 pathway. Brain Res 2021; 1770:147626. [PMID: 34418356 DOI: 10.1016/j.brainres.2021.147626] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/07/2021] [Accepted: 08/14/2021] [Indexed: 01/12/2023]
Abstract
Microglial polarization mediated neuroinflammation plays an important role in the pathological process of stroke. The aim of this study is to determine whether baicalein indirectly ameliorates neuronal injury through modulating microglial polarization after stroke and if so, then by what mechanism. The effects of baicalein on microglial polarization were revealed through the middle cerebral artery occlusion mouse model (MCAO, n = 6), the lipopolysaccharide (LPS) + interferon-γ (IFN-γ) and oxygen-glucose deprivation (OGD) induced neuroinflammatory microglia model (BV2, n = 3), respectively. Mice were treated with baicalein (100 mg/kg, i.g.) after reperfusion, and followed by daily administrations for 3 days. Results showed that the infarct volumes at 3 d in vehicle and baicalein-treated MCAO mice were 91.18 ± 4.02% and 55.36 ± 4.10%. Baicalein improved sensorimotor functions (p < 0.01) after MCAO. Real-time PCR revealed that baicalein decreased proinflammatory markers expression (p < 0.05), while elevated the anti-inflammatory markers (p < 0.05) in vivo and in vitro. Both western blot and immunofluorescent staining further confirmed that baicalein reduced proinflammatory marker CD16 levels (p < 0.01) and enhanced anti-inflammatory marker CD206 or Arg-1 levels (p < 0.05). Notably, baicalein suppressed the release of proinflammatory cytokines (p < 0.05) and nitric oxide (NO, p < 0.001). Mechanistically, baicalein prevented increases in TLR4 protein levels (p < 0.001), the phosphorylation of IKBα and p65 (p < 0.01), and the nuclear translocation of NF-κB p65 (p < 0.05). The NF-κB inhibitor, BAY 11-7085, enhanced the inhibitory effect of baicalein on the proinflammatory microglial polarization. Baicalein also inhibited the phosphorylation of signal transducer and activator of transcription 1 (STAT1, p < 0.001). A microglia-neuron co-culture system revealed that baicalein driven neuroprotection against OGD induced neuronal damage through modulating microglial polarization (p < 0.05). Baicalein indirectly ameliorates neuronal injury after stroke by polarizing microglia toward the anti-inflammatory phenotype via inhibition of the TLR4/NF-κB pathway and down-regulation of phosphorylated STAT1, suggesting that baicalein might serve a potential therapy for stroke.
Collapse
Affiliation(s)
- Yuanyuan Ran
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China
| | - Fuhai Gao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China
| | - Zitong Ding
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China
| | - Shuiqing Yang
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China
| | - Guiqin Tian
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China.
| | - Jianing Xi
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing 100144, PR China.
| |
Collapse
|
61
|
Lu J, Wang J, Yu L, Cui R, Zhang Y, Ding H, Yan G. Treadmill Exercise Attenuates Cerebral Ischemia-Reperfusion Injury by Promoting Activation of M2 Microglia via Upregulation of Interleukin-4. Front Cardiovasc Med 2021; 8:735485. [PMID: 34692788 PMCID: PMC8532515 DOI: 10.3389/fcvm.2021.735485] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/13/2021] [Indexed: 01/07/2023] Open
Abstract
Background: Exercise has been proven to be an effective therapy for stroke by reducing the microglia-initiated proinflammatory response. Few studies, however, have focused on the phenotypic changes in microglia cells caused by exercise training. The present study was designed to evaluate the influence of treadmill exercise on microglia polarization and the molecular mechanisms involved. Methods: Male Sprague-Dawley rats were randomly assigned into 3 groups: sham, MCAO and exercise. The middle cerebral artery occlusion (MCAO) and exercise groups received MCAO surgery and the sham group a sham operation. The exercise group also underwent treadmill exercise after the surgery. These groups were studied after 4 and 7 days to evaluate behavioral performance using a modified neurological severity score (mNSS), and infarct conditions using 2,3,5-triphenyl tetrazolium chloride. Quantitative real-time polymerase chain reaction (qRT-PCR) and Luminex was employed to determine the expressions of markers of microglia phenotypes. Western blotting was employed to identify the phosphorylation levels of Janus kinase1 (JAK1) and signal transducer and activator of transcription 6 (STAT6). Immunofluorescence was conducted to identify microglia phenotypes. Results: Treadmill exercise was found to improve neurobehavioral outcomes, mainly motor and balance functions, reduce infarct volumes and significantly increase interleukin-4 (IL-4) expression. In addition, treadmill exercise inhibited M1 microglia and promoted M2 microglia activation as evidenced by decreased M1 and increased M2 markers. Furthermore, an obvious increase in p-JAK1 and p-STAT6 was observed in the exercise group. Conclusions: Treadmill exercise ameliorates cerebral ischemia-reperfusion injury by enhancing IL-4 expression to promote M2 microglia polarization, possibly via the JAK1-STAT6 pathway.
Collapse
Affiliation(s)
- Juanjuan Lu
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China.,School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jie Wang
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Long Yu
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Rong Cui
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China.,School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Ying Zhang
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Hanqing Ding
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guofeng Yan
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
62
|
Curcumin Ameliorates White Matter Injury after Ischemic Stroke by Inhibiting Microglia/Macrophage Pyroptosis through NF- κB Suppression and NLRP3 Inflammasome Inhibition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1552127. [PMID: 34630845 PMCID: PMC8497115 DOI: 10.1155/2021/1552127] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
NLRP3 inflammasome-mediated pyroptosis is a proinflammatory programmed cell death pathway, which plays a vital role in functional outcomes after stroke. We previously described the beneficial effects of curcumin against stroke-induced neuronal damage through modulating microglial polarization. However, the impact of curcumin on microglial pyroptosis remains unknown. Here, stroke was modeled in mice by middle cerebral artery occlusion (MCAO) for 60 minutes and treated with curcumin (150 mg/kg) intraperitoneally immediately after reperfusion, followed by daily administrations for 7 days. Curcumin ameliorated white matter (WM) lesions and brain tissue loss 21 days poststroke and improved sensorimotor function 3, 10, and 21 days after stroke. Furthermore, curcumin significantly reduced the number of gasdermin D+ (GSDMD+) Iba1+ and caspase-1+Iba1+ microglia/macrophage 21 days after stroke. In vitro, lipopolysaccharide (LPS) with ATP treatment was used to induce pyroptosis in primary microglia. Western blot revealed a decrease in pyroptosis-related proteins, e.g., GSDMD-N, cleaved caspase-1, NLRP3, IL-1β, and IL-18, following in vitro or in vivo curcumin treatment. Mechanistically, both in vivo and in vitro studies confirmed that curcumin inhibited the activation of the NF-κB pathway. NLRP3 knocked down by siRNA transfection markedly increased the inhibitory effects of curcumin on microglial pyroptosis and proinflammatory responses, both in vitro and in vivo. Furthermore, stereotaxic microinjection of AAV-based NLRP3 shRNA significantly improved sensorimotor function and reduced WM lesion following curcumin treatment in MCAO mice. Our study suggested that curcumin reduced stroke-induced WM damage, improved functional outcomes, and attenuated microglial pyroptosis, at least partially, through suppression of the NF-κB/NLRP3 signaling pathway, further supporting curcumin as a potential therapeutic drug for stroke.
Collapse
|
63
|
Moya Gómez A, Font LP, Brône B, Bronckaers A. Electromagnetic Field as a Treatment for Cerebral Ischemic Stroke. Front Mol Biosci 2021; 8:742596. [PMID: 34557522 PMCID: PMC8453690 DOI: 10.3389/fmolb.2021.742596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/04/2021] [Indexed: 11/24/2022] Open
Abstract
Cerebral stroke is a leading cause of death and adult-acquired disability worldwide. To this date, treatment options are limited; hence, the search for new therapeutic approaches continues. Electromagnetic fields (EMFs) affect a wide variety of biological processes and accumulating evidence shows their potential as a treatment for ischemic stroke. Based on their characteristics, they can be divided into stationary, pulsed, and sinusoidal EMF. The aim of this review is to provide an extensive literature overview ranging from in vitro to even clinical studies within the field of ischemic stroke of all EMF types. A thorough comparison between EMF types and their effects is provided, as well as an overview of the signal pathways activated in cell types relevant for ischemic stroke such as neurons, microglia, astrocytes, and endothelial cells. We also discuss which steps have to be taken to improve their therapeutic efficacy in the frame of the clinical translation of this promising therapy.
Collapse
Affiliation(s)
- Amanda Moya Gómez
- UHasselt Hasselt University, BIOMED, Diepenbeek, Belgium.,Department of Biomedical Engineering, Faculty of Telecommunications, Informatics and Biomedical Engineering, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Lena Pérez Font
- Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Bert Brône
- UHasselt Hasselt University, BIOMED, Diepenbeek, Belgium
| | | |
Collapse
|
64
|
Moretti RL, Dias EN, Kiel SG, Augusto MCM, Rodrigues PS, Sampaio ACS, Medeiros LS, Martins MFM, Suffredini IB, Cardoso CV, Bondan EF. Behavioral and morphological effects of resveratrol and curcumin in rats submitted to doxorubicin-induced cognitive impairment. Res Vet Sci 2021; 140:242-250. [PMID: 34536813 DOI: 10.1016/j.rvsc.2021.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 08/08/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022]
Abstract
Doxorubicin (DOX) is known to cause cognitive impairments in patients submitted to long-term chemotherapy (deficits also known as chemobrain). Therefore, there is an urgent need for therapeutic strategies capable of returning cancer survivors back to their previous quality of life. The present study investigated whether resveratrol (RSV) or curcumin (CUR) administration could affect mnemonic function and brain morphological changes following DOX administration in rats. Male Wistar rats were divided into 4 groups: DOX group (2.5 mg/kg/week for 4 weeks, i.p., plus distilled water for 28 days, oral gavage - OG), DOX + RSV group (DOX, 2.5 mg/kg/week for 4 weeks, i.p., plus RSV, 10 mg/kg/day for 28 days, OG), DOX + CUR group (DOX, 2.5 mg/kg/week for 4 weeks, i.p., plus CUR, 100 mg/kg/day for 28 days, OG) and control (CTR) group (0.9% saline solution weekly for 4 weeks, i.p., plus distilled water for 28 days, OG). Behavioral analyses (open field - OF - and the novel object recognition test - NORT) were performed. Brains were collected and analyzed by hematoxylin-eosin and luxol fast blue staining techniques and by immunohistochemistry for GFAP (glial fibrillary acidic protein) expression in astrocytes and Iba1 (ionized calcium-binding adaptor molecule 1) expression in microglia. DOX-injected rats presented short-term and long-term memory impairments as seen in the NORT at 3 and 24 h after habituation and increased GFAP and Iba1 expression, respectively, in astrocytes and microglia of the frontal cortex, hypothalamus and hippocampus. Such cognitive deficits were prevented by CUR at both periods and by RSV at 24 h. DOX-induced astrogliosis and microgliosis were avoided by RSV and CUR. No signs of demyelination or neuronal loss were found in any group. Thus, CUR and RSV prevented memory loss, astrogliosis and microgliosis induced by DOX monotherapy.
Collapse
Affiliation(s)
- R L Moretti
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - E N Dias
- Department of Veterinary Medicine, University Cruzeiro do Sul, Avenida Tenente, Laudelino Ferreira do Amaral, 700, São Paulo, SP, 08060-000, Brazil
| | - S G Kiel
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - M C M Augusto
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - P S Rodrigues
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - A C S Sampaio
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - L S Medeiros
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - M F M Martins
- Department of Veterinary Medicine, University Cruzeiro do Sul, Avenida Tenente, Laudelino Ferreira do Amaral, 700, São Paulo, SP, 08060-000, Brazil
| | - I B Suffredini
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - C V Cardoso
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil
| | - E F Bondan
- Graduate Program in Environmental and Experimental Pathology, University Paulista, Rua Doutor Bacelar, 1212, 4th Floor, São Paulo, SP, 04026-002, Brazil; Department of Veterinary Medicine, University Cruzeiro do Sul, Avenida Tenente, Laudelino Ferreira do Amaral, 700, São Paulo, SP, 08060-000, Brazil.
| |
Collapse
|
65
|
Zhang T, Chen X, Qu Y, Ding Y. Curcumin Alleviates Oxygen-Glucose-Deprivation/Reperfusion-Induced Oxidative Damage by Regulating miR-1287-5p/LONP2 Axis in SH-SY5Y Cells. Anal Cell Pathol (Amst) 2021; 2021:5548706. [PMID: 34589382 PMCID: PMC8476263 DOI: 10.1155/2021/5548706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/30/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress-induced neuronal damage is a main cause of ischemia/reperfusion injury. Curcumin (Cur), the principal constituent extracted from dried rhizomes of Curcuma longa L. (turmeric), exhibits excellent antioxidant effects. Previous studies have indicated that miR-1287-5p was downregulated in patients with ischemic stroke. Additionally, we predicted that Lon Peptidase 2, Peroxisomal (LONP2), which is involved in oxidative stress regulation, is targeted by miR-1287-5p. The aim of the current study is to investigate the effect of Cur on ischemia/reperfusion damage and its underlying mechanism. To mimic ischemia/reperfusion damage environment, SH-SY5Y cells were subjected to oxygen-glucose-deprivation/reperfusion (OGD/R). OGD/R treatment downregulated miR-1287-5p and upregulated LONP2 in SH-SY5Y cells, but Cur alleviated OGD/R-induced oxidative damage and reversed the effect of OGD/R on the expression of miR-1287-5p and LONP2. Furthermore, we confirmed the interactive relationship between miR-1287-5p and LONP2 (negative regulation). We revealed that miR-1287-5p overexpression alleviated OGD/R-induced oxidative damage alleviation, similar to the effect of Cur. MiR-1287-5p inhibition accentuated OGD/R-induced oxidative damage in SH-SY5Y cells, which was reversed by Cur. The expression of LONP2 in OGD/R-treated SH-SY5Y cells was decreased by miR-1287-5p overexpression and increased by miR-1287-5p inhibition, and Cur counteracted the increase in LONP2 expression induced by miR-1287-5p inhibition. In conclusion, we suggest that Cur alleviates OGD/R-induced oxidative damage in SH-SY5Y cells by regulating the miR-1287-5p/LONP2 axis. The findings provide a theoretical basis for the clinical application of curcumin.
Collapse
Affiliation(s)
- Teng Zhang
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Provincial Hospital Traditional Chinese Medicine, Wuhan 430074, China
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Province Traditional Chinese Medicine Research Institute, Wuhan 430074, China
| | - Xiaomin Chen
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Provincial Hospital Traditional Chinese Medicine, Wuhan 430074, China
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Province Traditional Chinese Medicine Research Institute, Wuhan 430074, China
| | - Yueqing Qu
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Provincial Hospital Traditional Chinese Medicine, Wuhan 430074, China
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Province Traditional Chinese Medicine Research Institute, Wuhan 430074, China
| | - Yanbing Ding
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Provincial Hospital Traditional Chinese Medicine, Wuhan 430074, China
- Department of Traditional Chinese Medicine Encephalopathy, Hubei Province Traditional Chinese Medicine Research Institute, Wuhan 430074, China
| |
Collapse
|
66
|
Zhao X, Zhao M, Pang B, Zhu Y, Liu J. Diagnostic value of combined serological markers in the detection of acute cerebral infarction. Medicine (Baltimore) 2021; 100:e27146. [PMID: 34516506 PMCID: PMC8428755 DOI: 10.1097/md.0000000000027146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/17/2021] [Indexed: 01/05/2023] Open
Abstract
To evaluate the value of the combination schemes of 10 serological markers in the clinical diagnosis of acute cerebral infarction.The level of total cholesterol, triglycerides, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol, high-sensitivity C-reactive protein, homocysteine (HCY), lipoprotein-related phospholipase A2, ischemia-modified albumin, complement C1q, and lipoprotein a were analyzed in 154 patients with acute ischemic cerebral infarction. The optimized diagnostic combination for acute cerebral infarction was explored by calculating the maximum area under the receiver operating characteristic curves (AUC).The levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, high-sensitivity C-reactive protein, HCY, lipoprotein-related phospholipase A2, ischemia-modified albumin, complement C1q, and lipoprotein a were significantly higher in the patient vs the control group. Moreover, the positive rate of HCY reached 89.9%. The analysis of the receiver operating characteristic curve of each index and their combinations showed that the minimum AUC of HDL-C alone was 0.543, while the maximum AUC of HCY was 0.853. A multiple logistic regression analysis indicated that HDL-C was a slightly significant variate in the diagnosis of acute cerebral infarction.The value of individual serological markers in the diagnosis of acute cerebral infarction was slightly significant, while the combination of the markers significantly improved the efficiency of its diagnosis.
Collapse
Affiliation(s)
- Xiaowen Zhao
- Department of Clinical Laboratory, Liaocheng People's Hospital, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong, China
| | - Min Zhao
- Department of Clinical Laboratory, Liaocheng People's Hospital, Shandong, China
| | - Baojun Pang
- Department of Clinical Laboratory, Liaocheng People's Hospital, Shandong, China
| | - Yingnan Zhu
- Department of Clinical Laboratory, Liaocheng People's Hospital, Shandong, China
| | - Jizhu Liu
- Department of Clinical Laboratory, Liaocheng People's Hospital, Shandong, China
| |
Collapse
|
67
|
Tong C, Wu H, Gu D, Li Y, Fan Y, Zeng J, Ding W. Effect of curcumin on the non-alcoholic steatohepatitis via inhibiting the M1 polarization of macrophages. Hum Exp Toxicol 2021; 40:S310-S317. [PMID: 34470535 DOI: 10.1177/09603271211038741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a global medical problem and macrophages' activation is closely related to the pathogenesis of NASH. Curcumin is a polyphenol from turmeric with significant anti-inflammatory activity. OBJECTIVE The objective of present study was to observe the effect of curcumin on macrophages' activation and secretion of pro-inflammatory cytokines in NASH. METHODS Hematoxylin and eosin and TUNEL staining were used to observe the hepatic function. RT-PCR was conducted to evaluate the hepatic mRNA expression of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). Flow cytometry was adopted to detect the M1 polarization of macrophages. The RAW264.7 macrophage was pretreated with different doses of curcumin, and then lipopolysaccharide (LPS) and interferon-γ (IFN-γ) were given to activate the M1 macrophage. The activation ratio of M1 macrophage was observed by flow cytometry, and IL-1β and TNF-α expression was detected by RT-PCR and ELISA. RESULTS After treatment with curcumin, the activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), the mRNA expression of TNF-α and IL-1β, and M1 polarization of macrophages were significantly decreased. Hematoxylin and eosin and TUNEL staining showed that inflammation and apoptosis in the liver were improved. What is more, curcumin can effectively inhibit M1 macrophage activation induced by lipopolysaccharide and IFN-γ and reduce the secretion of IL-1β and TNF-α. CONCLUSION Curcumin can effectively improve NASH and reduce hepatic cell necrosis by inhibiting the M1 polarization of macrophages and the secretion of inflammatory factors.
Collapse
Affiliation(s)
- Cong Tong
- Department of Gastroenterology, Northern Jiangsu People's Hospital, 38043Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Hongfei Wu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Da Gu
- Department of Gastroenterology, Northern Jiangsu People's Hospital, 38043Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yanian Li
- 36674Dalian Medical University, Dalian, Liaoning Province, China
| | - Yuhan Fan
- Department of Gastroenterology, Northern Jiangsu People's Hospital, 38043Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiahui Zeng
- Department of Gastroenterology, Northern Jiangsu People's Hospital, 38043Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Ding
- Department of Traditional Chinese Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
68
|
Yu F, Huang T, Ran Y, Li D, Ye L, Tian G, Xi J, Liu Z. New Insights Into the Roles of Microglial Regulation in Brain Plasticity-Dependent Stroke Recovery. Front Cell Neurosci 2021; 15:727899. [PMID: 34421544 PMCID: PMC8374071 DOI: 10.3389/fncel.2021.727899] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/13/2021] [Indexed: 01/07/2023] Open
Abstract
Stroke remains the leading cause of long-term disability worldwide with significant long-term sequelae. However, there is no highly effective treatment to enhance post-stroke recovery despite extensive efforts in exploring rehabilitative therapies. Neurorehabilitation is recognized as the cornerstone of functional restoration therapy in stroke, where treatments are focused on neuroplastic regulation to reverse neural structural disruption and improve neurofunctional networks. Post-stroke neuroplasticity changes begin within hours of symptom onset and reaches a plateau by 3 to 4 weeks within the global brain in animal studies. It plays a determining role in spontaneous stroke recovery. Microglia are immediately activated following cerebral ischemia, which has been found both proximal to the primary ischemic injury and at the remote brain regions which have functional connections to the primary injury area. Microglia exhibit different activation profiles based on the microenvironment and adaptively switch their phenotypes in a spatiotemporal manner in response to brain injuries. Microglial activation coincides with neuroplasticity after stroke, which provides the fundamental base for the microglia-mediated inflammatory responses involved in the entire neural network rewiring and brain repair. Microglial activation exerts important effects on spontaneous recovery after stroke, including structural and functional reestablishment of neurovascular networks, neurogenesis, axonal remodeling, and blood vessel regeneration. In this review, we focus on the crosstalk between microglial activation and endogenous neuroplasticity, with a special focus on the plastic alterations in the whole brain network and their implications for structural and functional restoration after stroke. We then summarize recent advances in the impacts of microglial phenotype polarization on brain plasticity, trying to discuss the potential efficacy of microglia-based extrinsic restorative interventions in promoting post-stroke recovery.
Collapse
Affiliation(s)
- Fang Yu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Anesthesiology, Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Tingting Huang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Ran
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Da Li
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Lin Ye
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Guiqin Tian
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Jianing Xi
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
69
|
Subedi L, Gaire BP. Neuroprotective Effects of Curcumin in Cerebral Ischemia: Cellular and Molecular Mechanisms. ACS Chem Neurosci 2021; 12:2562-2572. [PMID: 34251185 DOI: 10.1021/acschemneuro.1c00153] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite being a major global health concern, cerebral ischemia/stroke has limited therapeutic options. Tissue plasminogen activator (tPA) is the only available medication to manage acute ischemic stroke, but this medication is associated with adverse effects and has a narrow therapeutic time window. Curcumin, a polyphenol that is abundantly present in the rhizome of the turmeric plant (Curcuma longa), has shown promising neuroprotective effects in animal models of neurodegenerative diseases, including cerebral ischemia. In the central nervous system (CNS), neuroprotective effects of curcumin have been experimentally validated in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and cerebral ischemia. Curcumin can exert pleiotropic effects in the postischemic brain including antioxidant, anti-inflammatory, antiapoptotic, vasculoprotective, and direct neuroprotective efficacies. Importantly, neuroprotective effects of curcumin has been reported in both ischemic and hemorrhagic stroke models. A broad-spectrum neuroprotective efficacy of curcumin suggested that curcumin can be an appealing therapeutic strategy to treat cerebral ischemia. In this review, we aimed to address the pharmacotherapeutic potential of curcumin in cerebral ischemia including its cellular and molecular mechanisms of neuroprotection revealing curcumin as an appealing therapeutic candidate for cerebral ischemia.
Collapse
Affiliation(s)
- Lalita Subedi
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Bhakta Prasad Gaire
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| |
Collapse
|
70
|
Nguyen HD, Kim MS. Effects of heavy metal, vitamin, and curry consumption on metabolic syndrome during menopause: a Korean community-based cross-sectional study. Menopause 2021; 28:949-959. [PMID: 34284432 DOI: 10.1097/gme.0000000000001825] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the associations between metabolic syndrome (MetS) during menopause and serum heavy metal levels and vitamin and curry consumption. METHODS A data set of 7,131 pre- and postmenopausal women aged ≥ 20 years collected between 2009 and 2017 was used to obtain information on sociodemographic, lifestyles, family histories, food intakes, and serum heavy metal levels and MetS. Logistic regression was used to identify associations between the presence of MetS and risk factors and to predict risks of MetS based on marginal effects. RESULTS Our results show that postmenopausal women had a higher risk of MetS than premenopausal women. During postmenopause elevations in the levels of serum cadmium by one unit increased the risk of MetS by 33% (OR 1.33; 95% CI, 1.03-1.72, P = 0.028). Risks of MetS in pre- and postmenopausal women, when serum Hb levels increased by 1 unit increased 21% (OR 1.21; 95% CI, 1.09-1.33, P < 0.001) and 26% (OR 1.26; 95% CI, 1.16-1.38, P < 0.001), respectively. Furthermore, the risk of MetS risk in pre- and postmenopausal women was increased 2.49-fold and 2.79-fold by a 1% increase in HbA1c level (OR 2.49; 95% CI, 1.97-3.16, P < 0.001) and (OR 2.79; 95% CI, 2.30-3.38, P < 0.001), respectively. High curry consumption reduced the risk of MetS significantly more than low curry consumption (OR 0.60; 95% CI, 0.39-0.91, P = 0.017) in premenopausal women. Furthermore, an increase in daily vitamin B2 intake by 1 mg reduced the risk of MetS by 45% (OR 0.55; 95% CI, 0.32-0.94, P = 0.028) in postmenopausal women. CONCLUSION Vitamin B2 and curry supplementation may protect against MetS. Further work is needed to reduce risk factors associated with heavy metals and determine the effects of vitamins and curry consumption on MetS during menopause.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, Jeonnam, Republic of Korea
| | | |
Collapse
|
71
|
Farkhondeh T, Samarghandian S, Roshanravan B, Peivasteh-Roudsari L. Impact of Curcumin on Traumatic Brain Injury and Involved Molecular Signaling Pathways. Recent Pat Food Nutr Agric 2021; 11:137-144. [PMID: 31288732 DOI: 10.2174/2212798410666190617161523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/14/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
Abstract
Traumatic Brain Injury (TBI) is one of the main causes of mortality and morbidity worldwide with no suitable treatment. The present study was designed to review the present literature about the protective effects of curcumin and the underlying mechanism against TBI. All published English language papers from beginning to 2019 were selected in this study. The findings indicate that curcumin may be effective against TBI outcomes by modulating the molecular signaling pathways involved in oxidative stress, inflammation, apoptosis, and autophagy. However, more experimental studies should be done to identify all mechanisms involved in the pathogenesis of TBI. Patents for Curcumin and chronic inflammation and traumatic brain injury management (WO2017097805A1 and US9101580B2) were published. In conclusion, the present study confirmed the potential therapeutic impact of curcumin for treating TBI.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Babak Roshanravan
- Medical Student, Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Leila Peivasteh-Roudsari
- Devision of Food Safety and Hygiene, Department of Environmental Health, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
72
|
Ran Y, Ye L, Ding Z, Gao F, Yang S, Fang B, Liu Z, Xi J. Melatonin Protects Against Ischemic Brain Injury by Modulating PI3K/AKT Signaling Pathway via Suppression of PTEN Activity. ASN Neuro 2021; 13:17590914211022888. [PMID: 34120482 PMCID: PMC8207287 DOI: 10.1177/17590914211022888] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide with limited therapeutic options. Melatonin can attenuate ischemic brain damage with improved functional outcomes. However, the cellular mechanisms of melatonin-driven neuroprotection against post-stroke neuronal death remain unknown. Here, distal middle cerebral artery occlusion (dMCAO) was performed in C57BL/6j mice to develop an ischemic stroke in vivo model. Melatonin was injected intraperitoneally immediately after ischemia, and 24 and 48 hours later. Melatonin treatment, with 5 to 20 mg/kg, elicited a dose-dependent decrease in infarct volume and concomitant increase in sensorimotor function. At the molecular level, phosphorylation of PTEN and Akt were increased, whereas PTEN activity was decreased in melatonin treated animals 72 hours after dMCAO. At the cellular level, oxygenglucose deprivation (OGD) challenge of neuronal cell line Neuro-2a (N2a) and primary neurons supported melatonin’s direct protection against neuronal cell death. Melatonin treatment reduced LDH release and neuronal apoptosis at various time points, markedly increased Akt phosphorylation in neuronal membrane, but significantly suppressed it in the cytoplasm of post-OGD neurons. Mechanistically, melatonin-induced Akt phosphorylation and neuronal survival was blocked by Wortmannin, a potent PIP3 inhibitor, exposing increased PI3K/Akt activation as a central player in melatonin-driven neuroprotection. Finally, PTEN knock-down through siRNA significantly inhibited PI3K/Akt activation and cell survival following melatonin treatment, suggesting that melatonin protection against ischemic brain damage, is at least partially, dependent on modulation of the PTEN/PI3K/Akt signaling axis.
Collapse
Affiliation(s)
- Yuanyuan Ran
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Lin Ye
- School of Materials Science and Engineering, Beijing Institute of Technology, Haidian, China
| | - Zitong Ding
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Fuhai Gao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Shuiqing Yang
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Boyan Fang
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Jianing Xi
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| |
Collapse
|
73
|
Duc HN, Oh H, Kim MS. Effects of Antioxidant Vitamins, Curry Consumption, and Heavy Metal Levels on Metabolic Syndrome with Comorbidities: A Korean Community-Based Cross-Sectional Study. Antioxidants (Basel) 2021; 10:808. [PMID: 34069726 PMCID: PMC8161368 DOI: 10.3390/antiox10050808] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
The burden of metabolic syndrome (MetS) has increased worldwide, especially during the COVID-19 pandemic, and this phenomenon is related to environmental, dietary, and lifestyle risk factors. We aimed to determine the association between the levels of serum heavy metals, hs-CRP, vitamins, and curry intake and to predict risks of MetS based on marginal effects. A data set of 60,256 Koreans aged ≥ 15 years between 2009 and 2017 was used to obtain information on sociodemographic, lifestyle, family history characteristics, MetS, food intake survey, and serum heavy metals. Daily intake of vitamins was measured by a one-day 24 h recall, and curry consumption was calculated using a food frequency questionnaire. Serum heavy metal levels were quantified by graphite furnace atomic absorption spectrometry and using a mercury analyzer. We found that vitamin B1, B2, B3, C, and A intakes were significantly lower in subjects with than without MetS. In contrast, serum levels of Pb, Hg, Cd, vitamin A, E, and hs-CRP were significantly higher in subjects with MetS. The risk of MetS was significantly lower for high curry consumers than low curry consumers (adjusted odds ratio 0.85, 95%CI 0.74-0.98). The risks of MetS were reduced by 12% and 1%, when vitamin B1 and C intakes increased by one mg, respectively, but were increased by 14%, 3%, and 9%, when serum levels of Pb, Hg, and hs-CRP increased by one unit. These results show that the potential health benefits resulting from vitamin and curry intakes could protect the public against the dual burden of communicable and non-communicable diseases. Further studies are required to reduce risk factors associated with serum heavy metal levels and to determine whether interactions between vitamin and curry consumption influence the presence of MetS.
Collapse
Affiliation(s)
| | | | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 57922, Korea; (H.N.D.); (H.O.)
| |
Collapse
|
74
|
Amato S, Arnold A. Modeling Microglia Activation and Inflammation-Based Neuroprotectant Strategies During Ischemic Stroke. Bull Math Biol 2021; 83:72. [PMID: 33982158 DOI: 10.1007/s11538-021-00905-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/26/2021] [Indexed: 11/28/2022]
Abstract
Neural inflammation immediately follows the onset of ischemic stroke. During this process, microglial cells can be activated into two different phenotypes: the M1 phenotype, which can worsen brain injury by producing pro-inflammatory cytokines; or the M2 phenotype, which can aid in long term recovery by producing anti-inflammatory cytokines. In this study, we formulate a nonlinear system of differential equations to model the activation of microglia post-ischemic stroke, which includes bidirectional switching between the microglia phenotypes, as well as the interactions between these cells and the cytokines that they produce. Further, we explore neuroprotectant-based modeling strategies to suppress the activation of the detrimental M1 phenotype, while promoting activation of the beneficial M2 phenotype. Through use of global sensitivity techniques, we analyze the effects of the model parameters on the ratio of M1 to M2 microglia and the total number of activated microglial cells in the system over time. Results demonstrate the significance of bidirectional microglia phenotype switching on the ratio of M1 to M2 microglia, in both the absence and presence of neuroprotectant terms. Simulations further suggest that early inhibition of M1 activation and support of M2 activation leads to a decreased minimum ratio of M1 to M2 microglia and allows for a larger number of M2 than M1 cells for a longer time period.
Collapse
Affiliation(s)
- Sara Amato
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Andrea Arnold
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA, USA.
| |
Collapse
|
75
|
Liu Y, Cheng Y, Zhang W, Tian H. Neuroprotective Effect of a New Free Radical Scavenger HL-008 in an Ischemia-Reperfusion Injury Rat Model. Neuroscience 2021; 465:105-115. [PMID: 33915200 DOI: 10.1016/j.neuroscience.2021.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/28/2022]
Abstract
Oxidative stress plays a critical role in cerebral ischemia-reperfusion injury. We have previously developed a powerful antioxidant, HL-008. This study aimed to investigate the neuroprotective function of HL-008. HL-008 efficacy in vitro and in vivo was evaluated using a PC-12 cell oxidative stress model induced by hydrogen peroxide and a rat model of middle cerebral artery occlusion, respectively. The MTT assay was used to analyze cell viability. 2,3,5-Triphenyltetrazolium chloride and Hematoxylin and Eosin staining, immunofluorescence, western blot, and proteomics were used to evaluate the infarction volume, brain tissue morphology, apoptosis, inflammation, and related pathways. Indicators related to oxidative levels were also detected. HL-008 significantly reduced the cerebral infarction volume induced by ischemia-reperfusion, improved the neurological score, alleviated oxidative stress and inflammation in the brain tissue, reduced glial cell activation, inhibited brain tissue apoptosis by influencing multiple signaling pathways, and had a neuroprotective effect. If HL-008 is successfully developed, it could significantly improve stroke patients' quality of life.
Collapse
Affiliation(s)
- Yahong Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, China
| | - Ying Cheng
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia.
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, China.
| |
Collapse
|
76
|
Zhang D, Feng Y, Pan H, Xuan Z, Yan S, Mao Y, Xiao X, Huang X, Zhang H, Zhou F, Chen B, Chen X, Liu H, Yan X, Liang H, Cui W. 9-Methylfascaplysin exerts anti-ischemic stroke neuroprotective effects via the inhibition of neuroinflammation and oxidative stress in rats. Int Immunopharmacol 2021; 97:107656. [PMID: 33895476 DOI: 10.1016/j.intimp.2021.107656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/23/2021] [Accepted: 04/03/2021] [Indexed: 02/04/2023]
Abstract
OBJECTIVES This study was aimed to investigate the neuroprotective effects of 9-methylfascaplysin, a novel marine derivative derived from sponge, against middle cerebral artery occlusion/reperfusion (MCAO)-induced motor impairments, neuroinflammation and oxidative stress in rats. METHODS Neurological and behavioral tests were used to evaluate behavioral changes. The 2, 3, 5-triphenyltetrazolium chloride staining was used to determine infarct size and edema extent. Activated microglia/macrophage was analyzed by immunohistochemical staining of Iba-1. RT-PCR and ELISA were used to measure the expression of inducible nitric oxide synthase, tumor necrosis factor-α, interleukin-1β, CD16 and CD206. Western blotting analysis was performed to explore the activation of nuclear factor-κB (NF-κB) and NLRP3. The levels of oxidative stress were studied by evaluating the activities of superoxide dismutase, catalase and glutathione peroxidase. RESULTS Post-occlusion intracerebroventricular injection of 9-methylfascaplysin significantly attenuated motor impairments and infarct size in MCAO rats. Moreover, 9-methylfascaplysin reduced the activation of microglia/macrophage in ischemic penumbra as evidenced by the decreased Iba-1-positive area and the reduced expression of pro-inflammatory factors. Furthermore, 9-methylfascaplysin inhibited MCAO-induced oxidative stress and activation of NF-κB and NLRP3 inflammasome. CONCLUSION All the results suggested that 9-methylfascaplysin might produce neuroprotective effects against MCAO via the reduction of oxidative stress and neuroinflammation, simultaneously, possibly via the inhibition of NF-κB and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Difan Zhang
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Yi Feng
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hanbo Pan
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Zhenquan Xuan
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Sicheng Yan
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Yuechun Mao
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Xiao Xiao
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Xinghan Huang
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hui Zhang
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Fei Zhou
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Bojun Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Xiaowei Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hao Liu
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Xiaojun Yan
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hongze Liang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China; Ningbo Kangning Hospital, Ningbo 315020, China.
| |
Collapse
|
77
|
Xue Y, Nie D, Wang LJ, Qiu HC, Ma L, Dong MX, Tu WJ, Zhao J. Microglial Polarization: Novel Therapeutic Strategy against Ischemic Stroke. Aging Dis 2021; 12:466-479. [PMID: 33815877 PMCID: PMC7990355 DOI: 10.14336/ad.2020.0701] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke, which is the second highest cause of death and the leading cause of disability, represents ~71% of all strokes globally. Some studies have found that the key elements of the pathobiology of stroke is immunity and inflammation. Microglia are the first line of defense in the nervous system. After stroke, the activated microglia become a double-edged sword, with distinct phenotypic changes to the deleterious M1 types and neuroprotective M2 types. Therefore, ways to promote microglial polarization toward M2 phenotype after stroke have become the focus of attention in recent years. In this review, we discuss the process of microglial polarization, summarize the alternation of signaling pathways and epigenetic regulation that control microglial polarization in ischemic stroke, aiming to find the potential mechanisms by which microglia can be transformed into the M2 polarized phenotype.
Collapse
Affiliation(s)
- Yimeng Xue
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
| | - Ding Nie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Lin-Jian Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
| | - Han-Cheng Qiu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Ming-Xin Dong
- Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
| | - Wen-Jun Tu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
78
|
Liu C, Liu S, Xiong L, Zhang L, Li X, Cao X, Xue J, Li L, Huang C, Huang Z. Genistein-3'-sodium sulfonate Attenuates Neuroinflammation in Stroke Rats by Down-Regulating Microglial M1 Polarization through α7nAChR-NF-κB Signaling Pathway. Int J Biol Sci 2021; 17:1088-1100. [PMID: 33867831 PMCID: PMC8040300 DOI: 10.7150/ijbs.56800] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/22/2021] [Indexed: 12/22/2022] Open
Abstract
Microglial M1 depolarization mediated prolonged inflammation contributing to brain injury in ischemic stroke. Our previous study revealed that Genistein-3'-sodium sulfonate (GSS) exerted neuroprotective effects in ischemic stroke. This study aimed to explore whether GSS protected against brain injury in ischemic stroke by regulating microglial M1 depolarization and its underlying mechanisms. We established transient middle cerebral artery occlusion and reperfusion (tMCAO) model in rats and used lipopolysaccharide (LPS)-stimulated BV2 microglial cells as in vitro model. Our results showed that GSS treatment significantly reduced the brain infarcted volume and improved the neurological function in tMCAO rats. Meanwhile, GSS treatment also dramatically reduced microglia M1 depolarization and IL-1β level, reversed α7nAChR expression, and inhibited the activation of NF-κB signaling in the ischemic penumbra brain regions. These effects of GSS were further verified in LPS-induced M1 depolarization of BV2 cells. Furthermore, pretreatment of α7nAChR inhibitor (α-BTX) significantly restrained the neuroprotective effect of GSS treatment in tMCAO rats. α-BTX also blunted the regulating effects of GSS on neuroinflammation, M1 depolarization and NF-κB signaling activation. This study demonstrates that GSS protects against brain injury in ischemic stroke by reducing microglia M1 depolarization to suppress neuroinflammation in peri-infarcted brain regions through upregulating α7nAChR and thereby inhibition of NF-κB signaling. Our findings uncover a potential molecular mechanism for GSS treatment in ischemic stroke.
Collapse
Affiliation(s)
- Chaoming Liu
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Pathobiology, JiangXi College of Traditional Chinese Medicine, Fuzhou, 344000, China
| | - Song Liu
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Lijiao Xiong
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Limei Zhang
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Xiao Li
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Xingling Cao
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jinhua Xue
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Liangdong Li
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Cheng Huang
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Zhihua Huang
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
79
|
Subedi L, Gaire BP. Phytochemicals as regulators of microglia/macrophages activation in cerebral ischemia. Pharmacol Res 2021; 165:105419. [DOI: 10.1016/j.phrs.2021.105419] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/16/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022]
|
80
|
Hou K, Li G, Yu J, Xu K, Wu W. Receptors, Channel Proteins, and Enzymes Involved in Microglia-mediated Neuroinflammation and Treatments by Targeting Microglia in Ischemic Stroke. Neuroscience 2021; 460:167-180. [PMID: 33609636 DOI: 10.1016/j.neuroscience.2021.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022]
Abstract
Stroke is the largest contributor to global neurological disability-adjusted life-years, posing a huge economic and social burden to the world. Though pharmacological recanalization with recombinant tissue plasminogen activator and mechanical thrombectomy have greatly improved the prognosis of patients with ischemic stroke, clinically, there is still no effective treatment for the secondary injury caused by cerebral ischemia. In recent years, more and more evidences show that neuroinflammation plays a pivotal role in the pathogenesis and progression of ischemic cerebral injury. Microglia are brain resident innate immune cells and act the role peripheral macrophages. They play critical roles in mediating neuroinflammation after ischemic stroke. Microglia-mediated neuroinflammation is not an isolated process and has complex relationships with other pathophysiological processes as oxidative/nitrative stress, excitotoxicity, necrosis, apoptosis, pyroptosis, autophagy, and adaptive immune response. Upon activation, microglia differentially express various receptors, channel proteins, and enzymes involved in promoting or inhibiting the inflammatory processes, making them the targets of intervention for ischemic stroke. To inhibit microglia-related neuroinflammation and promote neurological recovery after ischemic stroke, numerous biochemical agents, cellular therapies, and physical methods have been demonstrated to have therapeutic potentials. Though accumulating experimental evidences have demonstrated that targeting microglia is a promising approach in the treatment of ischemic stroke, the clinical progress is slow. Till now, no clinical study could provide convincing evidence that any biochemical or physical therapies could exert neuroprotective effect by specifically targeting microglia following ischemic stroke.
Collapse
Affiliation(s)
- Kun Hou
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Guichen Li
- Department of Neurology, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Jinlu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Kan Xu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Wei Wu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| |
Collapse
|
81
|
Fu CY, Zhong CR, Yang YT, Zhang M, Li WA, Zhou Q, Zhang F. Sirt1 activator SRT2104 protects against oxygen-glucose deprivation/reoxygenation-induced injury via regulating microglia polarization by modulating Sirt1/NF-κB pathway. Brain Res 2021; 1753:147236. [PMID: 33412146 DOI: 10.1016/j.brainres.2020.147236] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 01/12/2023]
Abstract
Cerebral ischemic/reperfusion injury is the most common neurological disorder and the second leading cause of death worldwide. Modulating microglia polarization from pro-inflammatory M1 phenotype to anti-inflammatory M2 state has been suggested as a potential therapeutic approach in the treatment of this injury. SRT2104, a novel activator of histone deacetylase Sirtuin-1 (Sirt1), has recently been shown to have anti-inflammation properties. However, the effect of SRT2104 on cerebral ischemic/reperfusion injury has not been elucidated. Here, we found that SRT2104 inhibited neuron and microglia death directly and indirectly through microglia condition medium from an oxygen glucose deprivation/reoxygenation (OGD/R) -induced cell injury models. Moreover, SRT2104 treatment modulated the microglia polarization shift from the M1 phenotype and skewed toward the M2 phenotype. Additionally, we found that SRT2104 could significant inhibit the activation of NF-κB and enhanced Sirt1 expression in microglia. Mechanism studies using the BV2 microglial cell line confirmed that knockdown Sirt1 significantly reduced the effect of SRT2104 on the activation of NF-κB pathway and microglial phenotype shift. Altogether, our result shows SRT2104 protect OGD/R-induced injury through shifting microglia phenotype, which may have potential in further studies as a novel neuroprotective agent for cerebral ischemic/reperfusion injury therapy.
Collapse
Affiliation(s)
- Chuan-Yi Fu
- Neurointerventional Department, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Chun-Rong Zhong
- Health Care Center Area Four, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Yuan-Tao Yang
- Neurointerventional Department, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Mao Zhang
- Neurointerventional Department, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Wen-An Li
- Neurointerventional Department, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Qing Zhou
- Neurointerventional Department, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Fan Zhang
- Neurointerventional Department, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China.
| |
Collapse
|
82
|
Seibel R, Schneider RH, Gottlieb MGV. Effects of Spices (Saffron, Rosemary, Cinnamon, Turmeric and Ginger) in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:347-357. [PMID: 34279199 DOI: 10.2174/1567205018666210716122034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 01/14/2021] [Accepted: 03/17/2021] [Indexed: 12/06/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent dementia in the elderly, causing disability, physical, psychological, social, and economic damage to the individual, their families, and caregivers. Studies have shown some spices, such as saffron, rosemary, cinnamon, turmeric, and ginger, have antioxidant and anti-inflammatory properties that act in inhibiting the aggregation of acetylcholinesterase and amyloid in AD. For this reason, spices have been studied as beneficial sources against neurodegenerative diseases, including AD. In this sense, this study aims to present a review of some spices (Saffron, Rosemary, Cinnamon, Turmeric and Ginger) and their bioactive compounds, most consumed and investigated in the world regarding AD. In this article, scientific evidence is compiled in clinical trials in adults, the elderly, animals, and in vitro, on properties considered neuroprotective, having no or negative effects on neuroprotection of these spices and their bioactive compounds. The importance of this issue is based on the pharmacological treatment for AD that is still not very effective. In addition, the recommendations and prescriptions of these spices are still permeated by questioning and lack of robust evidence of their effects on neurodegeneration. The literature search suggests all spices included in this article have bioactive compounds with anti-inflammatory and antioxidant actions associated with neuroprotection. To date, the amounts of spice ingestion in humans are not uniform, and there is no consensus on its indication and chronic consumption guarantees safety and efficacy in neuroprotection. Therefore, clinical evidence on this topic is necessary to become a formal adjuvant treatment for AD.
Collapse
Affiliation(s)
- Raquel Seibel
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre/RS, Brazil
| | - Rodolfo H Schneider
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre/RS, Brazil
| | - Maria G V Gottlieb
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre/RS, Brazil
| |
Collapse
|
83
|
Zhang W, Tian T, Gong SX, Huang WQ, Zhou QY, Wang AP, Tian Y. Microglia-associated neuroinflammation is a potential therapeutic target for ischemic stroke. Neural Regen Res 2021; 16:6-11. [PMID: 32788440 PMCID: PMC7818879 DOI: 10.4103/1673-5374.286954] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia-associated neuroinflammation plays an important role in the pathophysiology of ischemic stroke. Microglial activation and polarization, and the inflammatory response mediated by these cells play important roles in the development, progression and outcome of brain injury after ischemic stroke. Currently, there is no effective strategy for treating ischemic stroke in clinical practice. Therefore, it is clinically important to study the role and regulation of microglia in stroke. In this review, we discuss the involvement of microglia in the neuroinflammatory process in ischemic stroke, with the aim of providing a better understanding of the relationship between ischemic stroke and microglia.
Collapse
Affiliation(s)
- Wan Zhang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Tian Tian
- Department of Clinical Laboratory, the First Hospital of Changsha, Changsha, Hunan Province, China
| | - Shao-Xin Gong
- Department of Pathology, the First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Qin-Yi Zhou
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| |
Collapse
|
84
|
Parrella E, Gussago C, Porrini V, Benarese M, Pizzi M. From Preclinical Stroke Models to Humans: Polyphenols in the Prevention and Treatment of Stroke. Nutrients 2020; 13:nu13010085. [PMID: 33383852 PMCID: PMC7823436 DOI: 10.3390/nu13010085] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Polyphenols are an important family of molecules of vegetal origin present in many medicinal and edible plants, which represent important alimentary sources in the human diet. Polyphenols are known for their beneficial health effects and have been investigated for their potential protective role against various pathologies, including cancer, brain dysfunctions, cardiovascular diseases and stroke. The prevention of stroke promoted by polyphenols relies mainly on their effect on cardio- and cerebrovascular systems. However, a growing body of evidence from preclinical models of stroke points out a neuroprotective role of these molecules. Notably, in many preclinical studies, the polyphenolic compounds were effective also when administered after the stroke onset, suggesting their possible use in promoting recovery of patients suffering from stroke. Here, we review the effects of the major polyphenols in cellular and in vivo models of both ischemic and hemorrhagic stroke in immature and adult brains. The results from human studies are also reported.
Collapse
|
85
|
ZHENG X, GAN H, LI L, HU X, FANG Y, CHU L. [Astragaloside Ⅳ inhibits inflammation after cerebral ischemia in rats through promoting microglia/macrophage M2 polarization]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020; 49:679-686. [PMID: 33448170 PMCID: PMC10412416 DOI: 10.3785/j.issn.1008-9292.2020.12.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/09/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To investigate the effects of astragaloside Ⅳ (AS-Ⅳ) on microglia/macrophage M1/M2 polarization and inflammatory response after cerebral ischemia in rats. METHODS Forty eight male SD rats were randomly divided into sham operation control group, model control group and AS-Ⅳ group with 16 rats in each. Focal cerebral ischemia model was induced by occlusion of the right middle cerebral artery (MCAO) using the intraluminal filament. After ischemia induced, the rats in AS-Ⅳ group were intraperitoneally injected with 40 mg/kg AS-Ⅳ once a day for 3 days. The neurological functions were evaluated by the modified neurological severity score (mNSS) and the corner test on d1 and d3 after modelling. The infarct volume was measured by 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining on d3 after ischemia. The expression of M1 microglia/macrophage markers CD86, inducible nitric oxide synthase (iNOS) and pro-inflammatory factors TNF-α, IL-1β, IL-6, M2 microglia/macrophages markers CD206, arginase-1 (Arg-1), chitinase-like protein (YM1/2) and anti-inflammatory factors interleukin-10 (IL-10) and transforming growth factor beta (TGF-β) was detected by real-time RT-PCR. The expression of CD16/32/Iba1 and CD206/Iba1 was determined by double labeling immunefluorescence method in the peripheral area of cerebral ischemia. RESULTS Compared with model control group, AS-Ⅳ treatment improved neurological function recovery and reduced infarct volume after ischemia (P<0.05 or P<0.01). The qRT-PCR results showed that AS-Ⅳ treatment down-regulated the expression of CD86, iNOS, TNF-α, IL-1β, IL-6 mRNA (all P<0.01), and up-regulated the expression of CD206, Arg-1, YM1/2, IL-10 and TGF-β mRNA (all P<0.01). Furthermore, the results of immunefluorescence labeling showed that AS-Ⅳ treatment reduced the number of CD16/32+/Iba1+ cells (P<0.05) and increased the number of CD206+/Iba1+ cells (P<0.01) after cerebral ischemia. CONCLUSIONS The findings suggest that AS-Ⅳ ameliorates brain injury after cerebral ischemia in rats, which may be related to inhibiting inflammation through promoting the polarization of the microglia/macrophage from M1 to M2 phenotype in the ischemic brain.
Collapse
|
86
|
Qie S, Ran Y, Lu X, Su W, Li W, Xi J, Gong W, Liu Z. Candesartan modulates microglia activation and polarization via NF-κB signaling pathway. Int J Immunopathol Pharmacol 2020; 34:2058738420974900. [PMID: 33237822 PMCID: PMC7691946 DOI: 10.1177/2058738420974900] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Microglia are diverse cells that acquire different functional phenotypes in
response to microenvironment in which they reside. Several transcriptional
regulators have been identified that regulate different microglia phenotypes.
They are mainly stimulated into two opposing phenotypes, classically (M1) and
alternatively (M2) phenotype. Regulating microglia polarization from M1 to M2
state has been suggested as a potential therapeutic approach in treatment of CNS
disorders. Candesartan, an angiotensin II type I receptors antagonist, exerts
beneficial effects for antioxidant, anti-inflammation, neurotrophic, and
anti-apoptotic function. However, the effect of candesartan on microglia
polarization and underlying mechanisms remain unknown. In this study, the
resting microglia were stimulated to M1 microglia with lipopolysaccharide (LPS)
and interferon-γ (IFN-γ), and then treated with vehicle or candesartan for 24 h.
RT-PCR was utilized to detect the mRNA expression of microglia phenotype markers
and inflammatory cytokines. Microglia phenotype markers and toll-like receptor 4
(TLR4)/nuclear factor kappa B (NF-κB) pathway were determined by western blot. A
neuron-microglia co-culture system was used to determine whether candesartan
could ameliorate the neurotoxic effect of M1 microglia to oxygen-glucose
deprivation (OGD) neuron. Candesartan treatment reduced the expression of M1
markers, and increased M2 markers. Meanwhile, candesartan reduced fluorescence
intensity and protein level of M1 marker and enhanced M2 marker. Candesartan
also regulated the neuroinflammatory response via reducing the release of
pro-inflammatory cytokines and increasing anti-inflammatory cytokines in LPS +
IFN-γ stimulated BV2 cells. Candesartan markedly inhibited the protein level of
TLR4, the phosphorylation of IKBα and p65, and suppressed nuclear translocation
of NF-κB p65. BAY 11-7085, a NF-κB inhibitor, remarkably enlarged the inhibitory
effect of candesartan on NF-κB pathway. In addition, M1 phenotype microglia
exacerbated post-OGD N2a cells death and LDH release, whereas candesartan
reversed such neurotoxic effect. Candesartan treatment may ameliorate
stroke-induced neuronal damage through shifting microglia to M2 phenotype in a
TLR4/NF-κB-dependent manner.
Collapse
Affiliation(s)
- Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Ran
- Department of Research, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiaosheng Lu
- Department of Plastic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Wei Su
- Department of Neurosurgery, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Wei Li
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Jianing Xi
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Zongjian Liu
- Department of Research, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
87
|
Chelluboina B, Vemuganti R. Therapeutic potential of nutraceuticals to protect brain after stroke. Neurochem Int 2020; 142:104908. [PMID: 33220386 DOI: 10.1016/j.neuint.2020.104908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Stroke leads to significant neuronal death and long-term neurological disability due to synergistic pathogenic mechanisms. Stroke induces a change in eating habits and in many cases, leads to undernutrition that aggravates the post-stroke pathology. Proper nutritional regimen remains a major strategy to control the modifiable risk factors for cardiovascular and cerebrovascular diseases including stroke. Studies indicate that nutraceuticals (isolated and concentrated form of high-potency natural bioactive substances present in dietary nutritional components) can act as prophylactic as well as adjuvant therapeutic agents to prevent stroke risk, to promote ischemic tolerance and to reduce post-stroke consequences. Nutraceuticals are also thought to regulate blood pressure, delay neurodegeneration and improve overall vascular health. Nutraceuticals potentially mediate these effects by their powerful antioxidant and anti-inflammatory properties. This review discusses the studies that have highlighted the translational potential of nutraceuticals as stroke therapies.
Collapse
Affiliation(s)
- Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
| |
Collapse
|
88
|
Jiang Q, Geng X, Warren J, Eugene Paul Cosky E, Kaura S, Stone C, Li F, Ding Y. Hypoxia Inducible Factor-1α (HIF-1α) Mediates NLRP3 Inflammasome-Dependent-Pyroptotic and Apoptotic Cell Death Following Ischemic Stroke. Neuroscience 2020; 448:126-139. [PMID: 32976985 DOI: 10.1016/j.neuroscience.2020.09.036] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Stroke is a major cause of death and long-term disability. Recent evidence suggests that hypoxia-inducible factor 1α (HIF-1α), a transcription factor that regulates oxygen levels, plays a key role in neurological outcomes after ischemic stroke. Accordingly, we investigated the mechanism of HIF-1α on pyroptotic and apoptotic cells during ischemia/reperfusion (I/R). Adult Sprague-Dawley rats underwent 2 h of middle cerebral artery occlusion (MCAO). The rats were then exposed to 6 or 24 h of reperfusion, with or without YC-1 (HIF-1α inhibitor, 5 mg/kg). Infarct volumes, along with mRNA and protein quantities of HIF-1α, NLRP3, IL-1β, IL-18, Caspase-1, and co-localization of HIF-1α, and NLRP3, were assessed. We measured apoptotic and pyroptotic cell death, gasdermin D (GSDMD) activation and lactate dehydrogenase (LDH) activity, and the infiltration of neutrophils and macrophages after ischemic stroke. HIF-1α mRNA and NLRP3 inflammasome components were increased after 24 h of reperfusion. YC-1 significantly reduced the mRNA and protein expression of NLRP3, IL-1β, IL-18, and caspase-1; significantly decreased infarction and pyroptotic cell death after 24 h of reperfusion; attenuated the neuroinflammatory response by reducing infiltration of CD68- and MPO-positive cells after 24 h of reperfusion; and reduced apoptotic cell death following ischemic stroke. We found that HIF-1α likely regulates inflammatory responses through the NLRP3 inflammasome complex, thus influencing both apoptotic and pyroptotic cell death after stroke. These findings suggest that future investigations are needed regarding HIF-1α and its role as a potential molecular target in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Qian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Jonathan Warren
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Shawn Kaura
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Christopher Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI, USA.
| |
Collapse
|
89
|
Marques M, Cordeiro M, Marinho M, Vian C, Vaz G, Alves B, Jardim R, Hort M, Dora C, Horn A. Curcumin-loaded nanoemulsion improves haemorrhagic stroke recovery in wistar rats. Brain Res 2020; 1746:147007. [DOI: 10.1016/j.brainres.2020.147007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/16/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
|
90
|
Qing J, Zhang Z, Novák P, Zhao G, Yin K. Mitochondrial metabolism in regulating macrophage polarization: an emerging regulator of metabolic inflammatory diseases. Acta Biochim Biophys Sin (Shanghai) 2020; 52:917-926. [PMID: 32785581 DOI: 10.1093/abbs/gmaa081] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Indexed: 12/24/2022] Open
Abstract
As a major type of immune cells with heterogeneity and plasticity, macrophages are classically divided into inflammatory (M1) and alternative/anti-inflammatory (M2) types and play a crucial role in the progress of the inflammatory diseases. Recent studies have shown that metabolism is an important determinant of macrophage phenotype. Mitochondria, one of the most important compartments involving cell metabolism, are closely associated with the regulation of cell functions. In most types of cell, mitochondrial oxidative phosphorylation (OXPHOS) is the primary mode of cellular energy production. However, mitochondrial OXPHOS is inhibited in activated M1 macrophages, rendering them unable to be converted into M2 phenotype. Thus, mitochondrial metabolism is a crucial regulator in macrophage functions. This review summarizes the roles of mitochondria in macrophage polarization and analyzes the molecular mechanisms underlying mitochondrial metabolism and function, which may provide new approaches for the treatment of metabolic inflammatory diseases.
Collapse
Affiliation(s)
- Jina Qing
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
- Research Lab of translational medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Zizhen Zhang
- School of Medicine, Hunan Polytechnic of Environment and Biology, Hengyang 421001, China
| | - Petr Novák
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan 511518, China
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
- Research Lab of translational medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
91
|
Hamada T, Aratake T, Higashi Y, Ueba Y, Shimizu T, Shimizu S, Yawata T, Ueba T, Nakamura R, Akizawa T, Fujieda M, Saito M. Zinc-aggravated M1 microglia regulate astrocytic engulfment via P2×7 receptors. J Trace Elem Med Biol 2020; 61:126518. [PMID: 32353820 DOI: 10.1016/j.jtemb.2020.126518] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/03/2020] [Accepted: 03/27/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Glial cells such as astrocytes and microglia play an important role in the central nervous system via communication between these glial cells. Activated microglia can exhibit either the inflammatory M1 phenotype or the anti-inflammatory M2 phenotype, which influences astrocytic neuroprotective functions, including engulfment of cell debris. Recently, extracellular zinc has been shown to promote the inflammatory M1 phenotype in microglia through intracellular zinc accumulation and reactive oxygen species (ROS) generation. PURPOSE Here, we investigated whether the zinc-enhanced inflammatory M1 phenotype of microglia affects the astrocytic engulfing activity. METHODS Engulfing activity was assessed in astrocytes treated with microglial-conditioned medium (MCM) from lipopolysaccharide (LPS)-activated or from ZnCl2-pretreated LPS-activated M1 microglia. The effect of zinc on microglia phenotype was also validated using the zinc chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) and the ROS scavenger Trolox. RESULTS Although treatment of astrocytes with LPS showed no significant effect on the engulfing activity, MCM from LPS-induced M1 microglia increased the beads uptake by astrocytes. This increased uptake activity was suppressed when MCM from LPS-induced M1 microglia pretreated with ZnCl2 was applied to astrocytes, which was further abolished by the intracellular zinc chelator TPEN and the ROS scavenger Trolox. In addition, expression of P2×7 receptors (P2×7R) was increased in astrocytes treated with MCM derived from M1 microglia but not in the M1 microglia pretreated with ZnCl2. CONCLUSION These findings suggest that zinc pre-treatment abolishes the ability of LPS-induced M1 microglia to increase the engulfing activity in astrocytes via alteration of astrocytic P2×7R.
Collapse
Affiliation(s)
- Tomoya Hamada
- Department of Pediatrics, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan; Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Takaaki Aratake
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan; Research Fellow of Japan Society for the Promotion of Science, Japan
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Yusuke Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Toshio Yawata
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Tetsuya Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Rina Nakamura
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Toshifumi Akizawa
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Mikiya Fujieda
- Department of Pediatrics, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| |
Collapse
|
92
|
Du X, Xu Y, Chen S, Fang M. Inhibited CSF1R Alleviates Ischemia Injury via Inhibition of Microglia M1 Polarization and NLRP3 Pathway. Neural Plast 2020; 2020:8825954. [PMID: 32908485 PMCID: PMC7474788 DOI: 10.1155/2020/8825954] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/31/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
Ischemia cerebral stroke is one of the common neurological diseases with severe inflammatory response and neuron death. The inhibition of colony-stimulating factor 1 receptor (CSF1R) which especially expressed in microglia/macrophage exerted neuroprotection in stroke. However, the underlying neuroinflammatory regulation effects of CSF1R in ischemia stroke are not clear. In this study, cerebral ischemia stroke mice model was established. The C57/B6J mice were administered with Ki20227, a CSF1R inhibitor, by gavage for 7 consecutive days (0.002 mg/kg/day) before modeling. The Rota-Rod test and neurobehavioral score test were investigated to assess neurobehavioral functions. The area of infarction was assessed by 2, 3, 5-triphenyltetrazolium chloride (TTC) staining. The mRNA expressions of M1/M2 microglia markers were evaluated by real-time PCR. Immunofluorescence and Western blot were utilized to detect the changes of Iba1 and NLRP3 pathway proteins. Results showed that neurobehavioral function improvement was demonstrated by an increased stay time on the Rota-Rod test and a decreased neurobehavioral score in the Ki20227 treatment group. The area of infarction reduced in Ki20227 group when compared to the stroke group. Moreover, the mRNA expression of M1 microglia markers (TNF-α and iNOS) decreased while M2 microglia markers (IL-10 and Arg-1) increased. Meanwhile, compared to the stroke and stroke+PBS group, Ki20227 administration downregulated the expression of NLRP3, active caspase 1, and NF-κB protein in the ischemia penumbra of Ki20227 treatment group mice. In short, the CSF1R inhibitor, Ki20227, played vital neuroprotective roles in ischemia cerebral stroke mice, and the mechanisms may be via inhibiting microglia M1 polarization and NLRP3 inflammasome pathway activation. Our study provides a potential new target for the treatment of ischemic stroke injury.
Collapse
Affiliation(s)
- Xiaoxue Du
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310006 Zhejiang, China
- Translational Medicine Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006 Zhejiang, China
| | - Yuzhen Xu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Shijia Chen
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310006 Zhejiang, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310006 Zhejiang, China
| |
Collapse
|
93
|
A preclinical randomized controlled study of ischemia treated with Ginkgo biloba extracts: Are complex components beneficial for treating acute stroke? Curr Res Transl Med 2020; 68:197-203. [PMID: 32814684 DOI: 10.1016/j.retram.2020.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 07/02/2020] [Accepted: 07/21/2020] [Indexed: 11/22/2022]
Abstract
The rigorous design of preclinical experimental studies of candidate neuroprotectants for the treatment of acute ischemic stroke is crucial for the success of subsequent randomized clinical trials. The efficacy of Ginkgo biloba extracts (GBEs) in complex mixtures for the treatment of acute ischemic stroke remains unclear. In this preclinical randomized controlled trail (pRCT), the effects of a novel (n)GBE containing pinitol versus traditional (t)GBE without pinitol were evaluated on the mouse models of acute transient and permanent stroke, separately. The sample size, an important aspect of study design, was calculated based on our experimental data. Mice with ischemia that were induced by transient middle cerebral artery occlusion (tMCAO) or permanent distal middle cerebral artery occlusion (pdMCAO), were treated with vehicle, nGBE, tGBE, or pinitol alone by tail-vein injection. Our results showed that nGBE significantly reduced infarct size in mice with tMCAO compared with vehicle-treated control mice. Both nGBE and tGBE significantly reduced infarct size in mice with pdMCAO compared with the vehicle-treated controls. None of the three treatments rescued weight loss or prevented the neurological deficits in either the tMCAO- or pdMCAO-model mice. These findings suggest that nGBE, which includes all of the components of tGBE and pinitol, is neuroprotective in two ischemic stroke models. Additional studies of complex GBE mixtures for stroke treatment compared to single component medications are undergoing evaluation.
Collapse
|
94
|
Sheikholeslami MA, Parvardeh S, Ghafghazi S, Moini Zanjani T, Sabetkasaei M. The Attenuating Effect of Curcumin on Morphine Dependence in Rats: The Involvement of Spinal Microglial Cells and Inflammatory Cytokines. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:198-207. [PMID: 32802100 PMCID: PMC7393048 DOI: 10.22037/ijpr.2019.111701.13309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
New evidence suggests an important role for spinal glial cells in the development of opioid dependence. Curcumin, a component of the Curcuma Longa, has shown to act as a suppressor of microglial cells. The main goal of this study was to explore the attenuating effects of curcumin on morphine dependence with a focus on spinal microglial cells and inflammatory cytokines. In order to induce morphine dependence in male Wistar rats, morphine was administered intraperitoneally (i.p.) once daily for 9 days in an increasing dose of 10, 20, and 40 mg/kg. Curcumin (2.5, 5, and 10 mg/kg, i.p.) was given from the days 10th to 18th. Naloxone-precipitated abstinence syndrome was used to assess the behavioral symptoms of morphine dependence. Immunofluorescence staining of Iba1 and ELISA test were used to measure spinal microglial activity and inflammatory cytokines levels, respectively. The results showed that curcumin (2.5, 5, and 10 mg/kg) significantly decreased jumping, leaning, and diarrhea in morphine-dependent rats. In addition, the spinal concentration of TNF-α and IL-6 was reduced by curcumin (2.5, 5, and 10 mg/kg) significantly. Moreover, curcumin showed a potent attenuating effect on the number of Iba1 positive cells in rats which were subjected to morphine dependence. The results of this study demonstrated that curcumin exerts a remarkable reducing effect on morphine dependence in rats. The findings showed that the therapeutic effect of curcumin on morphine dependence is mediated through the suppression of activated microglial cells and reduction of inflammatory cytokines levels in the spinal cord.
Collapse
Affiliation(s)
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shiva Ghafghazi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Taraneh Moini Zanjani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sabetkasaei
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
95
|
He R, Jiang Y, Shi Y, Liang J, Zhao L. Curcumin-laden exosomes target ischemic brain tissue and alleviate cerebral ischemia-reperfusion injury by inhibiting ROS-mediated mitochondrial apoptosis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111314. [PMID: 32919674 DOI: 10.1016/j.msec.2020.111314] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022]
Abstract
The pathogenesis of ischemic cerebrovascular disease has revealed that ischemia-reperfusion (I/R) injury often leads to aggravation of metabolic oxidative stress and blood-brain barrier (BBB) destruction, eventually causing secondary brain tissue damage. Accumulated reactive oxygen species (ROS) in focal ischemia activate mitochondria-mediated apoptosis and damage the BBB by degrading tight junction proteins (TJPs). Herein, we report macrophage-derived exosomes (Ex) loaded with curcumin (cur) as a multifunctional biomimetic delivery vehicle (Ex-cur) for targeting ischemic brain tissue and alleviating cerebral I/R injury by inhibiting ROS-mediated mitochondrial apoptosis in a transient cerebral ischemia rat model. The design principle relies on unique features of macrophage-derived exosomes and the natural ingredient cur. Specifically, cur can be entrapped within exosomes when incubated with murine macrophage RAW264.7 cells, and its stability is subsequently significantly improved. The resultant Ex-cur can target ischemic regions by leveraging the targeting migration capability of Ex driven by inflammation. Accumulated Ex-cur in ischemic regions is experimentally proven to be highly effective at reducing ROS accumulation by virtue of the antioxidant properties of cur. Using Ex-cur to down-regulate ROS accumulation in lesions, we alleviate BBB damage and suppress mitochondria-mediated neuronal apoptosis, which is confirmed by a series of relevant protein analysis. These findings demonstrate good therapeutic efficacy of Ex-cur for treating I/R injury, providing experimental evidence for the potential clinical benefits of Ex-cur for other modes of neuroprotection.
Collapse
Affiliation(s)
- Ruyi He
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yibing Jiang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jia Liang
- Life Science Institution, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| |
Collapse
|
96
|
Chen S, Chen H, Du Q, Shen J. Targeting Myeloperoxidase (MPO) Mediated Oxidative Stress and Inflammation for Reducing Brain Ischemia Injury: Potential Application of Natural Compounds. Front Physiol 2020; 11:433. [PMID: 32508671 PMCID: PMC7248223 DOI: 10.3389/fphys.2020.00433] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress and inflammation are two critical pathological processes of cerebral ischemia-reperfusion injury. Myeloperoxidase (MPO) is a critical inflammatory enzyme and therapeutic target triggering both oxidative stress and neuroinflammation in the pathological process of cerebral ischemia-reperfusion injury. MPO is presented in infiltrated neutrophils, activated microglial cells, neurons, and astrocytes in the ischemic brain. Activation of MPO can catalyze the reaction of chloride and H2O2 to produce HOCl. MPO also mediates oxidative stress by promoting the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), modulating the polarization and inflammation-related signaling pathways in microglia and neutrophils. MPO can be a therapeutic target for attenuating oxidative damage and neuroinflammation in ischemic stroke. Targeting MPO with inhibitors or gene deficiency significantly reduced brain infarction and improved neurological outcomes. This article discusses the important roles of MPO in mediating oxidative stress and neuroinflammation during cerebral ischemia-reperfusion injury and reviews the current understanding of the underlying mechanisms. Furthermore, we summarize the active compounds from medicinal herbs with potential as MPO inhibitors for anti-oxidative stress and anti-inflammation to attenuate cerebral ischemia-reperfusion injury, and as adjunct therapeutic agents for extending the window of thrombolytic treatment. We highlight that targeting MPO could be a promising strategy for alleviating ischemic brain injury, which merits further translational study.
Collapse
Affiliation(s)
- Shuang Chen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Qiaohui Du
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
97
|
Li F, Zhao H, Han Z, Wang R, Tao Z, Fan Z, Zhang S, Li G, Chen Z, Luo Y. Xuesaitong May Protect Against Ischemic Stroke by Modulating Microglial Phenotypes and Inhibiting Neuronal Cell Apoptosis via the STAT3 Signaling Pathway. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:115-123. [PMID: 30426907 DOI: 10.2174/1871527317666181114140340] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/20/2018] [Accepted: 11/11/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Xuesaitong mainly comprises Panax notoginseng saponins and has shown a promising feature in an acute ischemic stroke model; however, its effect on long-term recovery following stroke, and the related mechanisms, are unknown. METHODS The objective of this study was to investigate the long-term protective effects of xuesaitong against ischemic stroke and its effect on microglial polarization. Experimental cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) for 45 min, and C57BL/6 mice were immediately injected with xuesaitong or vehicle through the caudal vein at the onset of cerebral reperfusion consecutively for 14 days. The animals were randomly divided into three groups: a sham-operated group, vehicle-treated group and xuesaitong-treated group at a dose of 15μg/g. Subsequently, 2,3,5-triphenyltetrazolium chloride staining was used to assess infarct volume, and adhesive removal tests and balance beam tests were used to evaluate neurological deficits at days 1, 3, 7 and 14 following ischemia. Reverse-transcriptase polymerase chain reaction and immunofluorescence staining for M1 markers (CD16, iNOS) and M2 markers (CD206, arginase-1) were performed to characterize phenotypic changes in microglia. Elisa was used to determine the release of pro-inflammatory and anti-inflammatory cytokines. TUNEL staining was conducted to detect neuronal cell apoptosis, and western blots were used to determine the activation of signal transducer and activator of transcription 3 (STAT3). RESULTS Our results revealed that xuesaitong treatment, compared with vehicle treatment, significantly reduced cerebral infarct volume 1 and 3 days after MCAO and resulted in significant improvements in long-term neurological outcomes. Furthermore, xuesaitong treatment, compared with vehicle treatment, significantly reduced M1 markers and elevated M2 markers 7 and 14 days after MCAO at both the mRNA and protein level in ipsilateral brain tissue. This finding was also accompanied by a reduction in neuronal cell apoptosis and p-STAT3 transcription factor levels in the xuesaitong-treated group compared with the vehicle-treated group. CONCLUSION We demonstrated that xuesaitong has long-term neuroprotective effects against ischemic stroke, possibly by promoting the polarization of microglia to an M2 phenotype and by inhibiting neuronal cell death via down-regulation of the STAT3 signaling pathway, providing new evidence that xuesaitong might be a promising therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Fangfang Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Haiping Zhao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Ziping Han
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Zhen Tao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Zhibin Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Sijia Zhang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Guangwen Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Zhigang Chen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
98
|
Jiang CT, Wu WF, Deng YH, Ge JW. Modulators of microglia activation and polarization in ischemic stroke (Review). Mol Med Rep 2020; 21:2006-2018. [PMID: 32323760 PMCID: PMC7115206 DOI: 10.3892/mmr.2020.11003] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke is one of the leading causes of mortality and disability worldwide. However, there is a current lack of effective therapies available. As the resident macrophages of the brain, microglia can monitor the microenvironment and initiate immune responses. In response to various brain injuries, such as ischemic stroke, microglia are activated and polarized into the proinflammatory M1 phenotype or the anti‑inflammatory M2 phenotype. The immunomodulatory molecules, such as cytokines and chemokines, generated by these microglia are closely associated with secondary brain damage or repair, respectively, following ischemic stroke. It has been shown that M1 microglia promote secondary brain damage, whilst M2 microglia facilitate recovery following stroke. In addition, autophagy is also reportedly involved in the pathology of ischemic stroke through regulating the activation and function of microglia. Therefore, this review aimed to provide a comprehensive overview of microglia activation, their functions and changes, and the modulators of these processes, including transcription factors, membrane receptors, ion channel proteins and genes, in ischemic stroke. The effects of autophagy on microglia polarization in ischemic stroke were also reviewed. Finally, future research areas of ischemic stroke and the implications of the current knowledge for the development of novel therapeutics for ischemic stroke were identified.
Collapse
Affiliation(s)
- Cheng-Ting Jiang
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wan-Feng Wu
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yi-Hui Deng
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jin-Wen Ge
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
99
|
Yuan Y, Wu C, Ling EA. Heterogeneity of Microglia Phenotypes: Developmental, Functional and Some Therapeutic Considerations. Curr Pharm Des 2020; 25:2375-2393. [PMID: 31584369 DOI: 10.2174/1381612825666190722114248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Microglia play a pivotal role in maintaining homeostasis in complex brain environment. They first exist as amoeboid microglial cells (AMCs) in the developing brain, but with brain maturation, they transform into ramified microglial cells (RMCs). In pathological conditions, microglia are activated and have been classified into M1 and M2 phenotypes. The roles of AMCs, RMCs and M1/M2 microglia phenotypes especially in pathological conditions have been the focus of many recent studies. METHODS Here, we review the early development of the AMCs and RMCs and discuss their specific functions with reference to their anatomic locations, immunochemical coding etc. M1 and M2 microglia phenotypes in different neuropathological conditions are also reviewed. RESULTS Activated microglia are engaged in phagocytosis, production of proinflammatory mediators, trophic factors and synaptogenesis etc. Prolonged microglia activation, however, can cause damage to neurons and oligodendrocytes. The M1 and M2 phenotypes featured prominently in pathological conditions are discussed in depth. Experimental evidence suggests that microglia phenotype is being modulated by multiple factors including external and internal stimuli, local demands, epigenetic regulation, and herbal compounds. CONCLUSION Prevailing views converge that M2 polarization is neuroprotective. Thus, proper therapeutic designs including the use of anti-inflammatory drugs, herbal agents may be beneficial in suppression of microglial activation, especially M1 phenotype, for amelioration of neuroinflammation in different neuropathological conditions. Finally, recent development of radioligands targeting 18 kDa translocator protein (TSPO) in activated microglia may hold great promises clinically for early detection of brain lesion with the positron emission tomography.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, 4 Medical Drive, MD10, National University of Singapore, 117594, Singapore
| |
Collapse
|
100
|
Sharifi-Rad M, Lankatillake C, Dias DA, Docea AO, Mahomoodally MF, Lobine D, Chazot PL, Kurt B, Boyunegmez Tumer T, Catarina Moreira A, Sharopov F, Martorell M, Martins N, Cho WC, Calina D, Sharifi-Rad J. Impact of Natural Compounds on Neurodegenerative Disorders: From Preclinical to Pharmacotherapeutics. J Clin Med 2020; 9:1061. [PMID: 32276438 PMCID: PMC7231062 DOI: 10.3390/jcm9041061] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/29/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Among the major neurodegenerative disorders (NDDs), Alzheimer's disease (AD) and Parkinson's disease (PD), are a huge socioeconomic burden. Over many centuries, people have sought a cure for NDDs from the natural herbals. Many medicinal plants and their secondary metabolites are reported with the ability to alleviate the symptoms of NDDs. The major mechanisms identified, through which phytochemicals exert their neuroprotective effects and potential maintenance of neurological health in ageing, include antioxidant, anti-inflammatory, antithrombotic, antiapoptotic, acetylcholinesterase and monoamine oxidase inhibition and neurotrophic activities. This article review the mechanisms of action of some of the major herbal products with potential in the treatment of NDDs according to their molecular targets, as well as their regional sources (Asia, America and Africa). A number of studies demonstrated the beneficial properties of plant extracts or their bioactive compounds against NDDs. Herbal products may potentially offer new treatment options for patients with NDDs, which is a cheaper and culturally suitable alternative to conventional therapies for millions of people in the world with age-related NDDs.
Collapse
Affiliation(s)
- Mehdi Sharifi-Rad
- Department of Medical Parasitology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran;
| | - Chintha Lankatillake
- School of Health and Biomedical Sciences, RMIT University, Bundoora, PO Box 71, VIC 3083, Australia;
| | - Daniel A. Dias
- School of Health and Biomedical Sciences, RMIT University, Bundoora, PO Box 71, VIC 3083, Australia;
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Mohamad Fawzi Mahomoodally
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam; or
- Department of Health Sciences, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius;
| | - Devina Lobine
- Department of Health Sciences, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius;
| | - Paul L. Chazot
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
| | - Begum Kurt
- Graduate Program of Biomolecular Sciences, Institute of Natural and Applied Sciences, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey;
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey;
| | - Ana Catarina Moreira
- Pulmonology Department, Hospital Garcia de Orta, EPE Almada, 2801-951 Lisboa, Portugal;
| | - Farukh Sharopov
- Department of Pharmaceutical Technology, Avicenna Tajik State Medical University, Rudaki 139, Dushanbe 734003, Tajikistan;
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepcion, Concepcion 4070386, Chile;
- Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, Concepcion 4070386, Chile
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1991953381, Iran
| |
Collapse
|