1
|
Xing X, Zhang X, Fan J, Zhang C, Zhang L, Duan R, Hao H. Neuroprotective Effects of Melittin Against Cerebral Ischemia and Inflammatory Injury via Upregulation of MCPIP1 to Suppress NF-κB Activation In Vivo and In Vitro. Neurochem Res 2024; 49:348-362. [PMID: 37812268 PMCID: PMC10787673 DOI: 10.1007/s11064-023-04030-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/20/2023] [Accepted: 09/10/2023] [Indexed: 10/10/2023]
Abstract
Melittin, a principal constituent of honeybee venom, exhibits diverse biological effects, encompassing anti-inflammatory capabilities and neuroprotective actions against an array of neurological diseases. In this study, we probed the prospective protective influence of melittin on cerebral ischemia, focusing on its anti-inflammatory activity. Mechanistically, we explored whether monocyte chemotactic protein-induced protein 1 (MCPIP1, also known as ZC3H12A), a recently identified zinc-finger protein, played a role in melittin-mediated anti-inflammation and neuroprotection. Male C57/BL6 mice were subjected to distal middle cerebral artery occlusion to create a focal cerebral cortical ischemia model, with melittin administered intraperitoneally. We evaluated motor functions, brain infarct volume, cerebral blood flow, and inflammatory marker levels within brain tissue, employing quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assays, and western blotting. In vitro, an immortalized BV-2 microglia culture was stimulated with lipopolysaccharide (LPS) to establish an inflammatory cell model. Post-melittin exposure, cell viability, and cytokine expression were examined. MCPIP1 was silenced using siRNA in LPS-induced BV-2 cells, with the ensuing nuclear translocation of nuclear factor-κB assessed through cellular immunofluorescence. In vivo, melittin enhanced motor functions, diminished infarction, fostered blood flow restoration in ischemic brain regions, and markedly inhibited the expression of inflammatory cytokines (interleukin-1β, interleukin-6, tumor necrosis factor-α, and nuclear factor-κB). In vitro, melittin augmented MCPIP1 expression in LPS-induced BV-2 cells and ameliorated inflammation-induced cell death. The neuroprotective effect conferred by melittin was attenuated upon MCPIP1 knockdown. Our findings establish that melittin-induced tolerance to ischemic injury is intrinsically linked with its anti-inflammatory capacity. Moreover, MCPIP1 is, at the very least, partially implicated in this process.
Collapse
Affiliation(s)
- Xing Xing
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China.
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China.
| | - Jingyi Fan
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Ruisheng Duan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Hongyu Hao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
2
|
Sharma A, Jaiswal V, Park M, Lee HJ. Biogenic silver NPs alleviate LPS-induced neuroinflammation in a human fetal brain-derived cell line: Molecular switch to the M2 phenotype, modulation of TLR4/MyD88 and Nrf2/HO-1 signaling pathways, and molecular docking analysis. BIOMATERIALS ADVANCES 2023; 148:213363. [PMID: 36881963 DOI: 10.1016/j.bioadv.2023.213363] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Silver nanoparticles (AgNPs) have inconsistent findings against inflammation. Although a wealth of literature on the beneficial effects of green-synthesized AgNPs has been published, a detailed mechanistic study of green AgNPs on the protective effects against lipopolysaccharide (LPS)-induced neuroinflammation using human microglial cells (HMC3) has not yet been reported. For the first time, we studied the inhibitory effect of biogenic AgNPs on inflammation and oxidative stress induced by LPS in HMC3 cells. X-ray photoelectron spectroscopy, Fourier-transform infrared spectroscopy, and transmission electron microscopy were used to characterize AgNPs produced from honeyberry. Co-treatment with AgNPs significantly reduced mRNA expressions of inflammatory molecules such as interleukin (IL)-6 and tumor necrosis factor-α, while increasing the expressions of anti-inflammatory markers such as IL-10 and transforming growth factor (TGF)-β. HMC3 cells were also switched from M1 to M2, as shown by lower expression of M1 markers such as cluster of differentiation (CD)80, CD86, and CD68 and higher expression of M2 markers such as CD206, CD163, and triggering receptors expressed on myeloid cells (TREM2). Furthermore, AgNPs inhibited LPS-induced toll-like receptor (TLR)4 signaling, as evidenced by decreased expression of myeloid differentiation factor 88 (MyD88) and TLR4. In addition, AgNPs reduced the production of reactive oxygen species (ROS) and enhanced the expression of nuclear factor-E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), while decreasing the expression of inducible nitric oxide synthase. The docking score of the honeyberry phytoconstituents ranged from -14.93 to - 4.28 KJ/mol. In conclusion, biogenic AgNPs protect against neuroinflammation and oxidative stress by targeting TLR4/MyD88 and Nrf2/HO-1 signaling pathways in a LPS-induced in vitro model. Biogenic AgNPs could be utilized as potential nanomedicine against LPS-induced inflammatory disorders.
Collapse
Affiliation(s)
- Anshul Sharma
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Varun Jaiswal
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Miey Park
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea; Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Hae-Jeung Lee
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea; Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea.
| |
Collapse
|
3
|
The Yin and Yang of toll-like receptors in endothelial dysfunction. Int Immunopharmacol 2022; 108:108768. [DOI: 10.1016/j.intimp.2022.108768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
|
4
|
Lu J, Zhu B, Zhou F, Ding X, Qian C, Ding Z, Ye X. Polysaccharides From the Aerial Parts of Tetrastigma Hemsleyanum Diels et Gilg Induce Bidirectional Immunity and Ameliorate LPS-Induced Acute Respiratory Distress Syndrome in Mice. Front Pharmacol 2022; 13:838873. [PMID: 35370633 PMCID: PMC8965720 DOI: 10.3389/fphar.2022.838873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Tetrastigma hemsleyanum Diels et Gilg (Sanyeqing, SYQ) has traditionally been used to treat inflammation, high fever and improve immune function of patients. Polysaccharides have been proved to be one of the important components of SYQ. Previous studies have confirmed the antipyretic and antitumor effects of polysaccharides from SYQ (SYQP), and clarified that SYQP could enhance immunity through TLR4 signalling pathway. However, there were more possibilities for the mechanism by which SYQP exerted immunomodulatory effects and the role of SYQP in acute respiratory distress syndrome (ARDS) is elusive. The purpose of this study was further to explain the bidirectional modulation of immunity mechanism of SYQP in vitro and its effect in LPS-induced ARDS in vivo. Experimental results showed that SYQP significantly stimulated gene expressions of TLR1, TLR2 and TLR6 and secretion of cytokines in RAW264.7 cells. Individual or combined application of TLR2 antagonist C29 and TLR4 antagonist TAK-242 could reduce SYQP-mediated stimulation of cytokine secretion in RAW264.7 cells and mouse peritoneal macrophages (MPMs) to varying degrees. On the other hand, SYQP markedly inhibited the expression levels of inflammatory cytokines, NO, iNOS and COX-2 in LPS-treatment RAW264.7 cells. Moreover, in vivo results indicated that SYQP significantly reduced LPS-induced damage in ARDS mice through alleviating LPS-induced pulmonary morphological damage, inhibiting myeloperoxidase (MPO) expression levels, ameliorating the inflammatory cells in bronchoalveolar lavage fluid (BALF) and improving hematological status. Meanwhile, SYQP evidently reduced IL-6, TNF-α and IFN-γ secretion, the overexpression levels of TLR2 and TLR4, as well as the phosphorylation of NF-κB p65. In addition, SYQP reduced the phosphorylation of JAK2 and STAT1 and the overexpression of NLRP3, caspase-1, caspase-3 and caspase-8 in lung tissues of ARDS mice. In summary, our study confirmed that SYQP induced bidirectional immunity and ameliorated LPS-induced acute respiratory distress syndrome in mice through TLR2/TLR4-NF-κB, NLRP3/caspase and JAK/STAT signaling pathways, which provided a theoretical basis for further use of SYQP.
Collapse
Affiliation(s)
- Jingjing Lu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bingqi Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinghong Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaodong Qian
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Zhishan Ding, ; Xiaoqing Ye,
| | - Xiaoqing Ye
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Zhishan Ding, ; Xiaoqing Ye,
| |
Collapse
|
5
|
Markoutsa E, Mayilsamy K, Gulick D, Mohapatra SS, Mohapatra S. Extracellular vesicles derived from inflammatory-educated stem cells reverse brain inflammation-implication of miRNAs. Mol Ther 2022; 30:816-830. [PMID: 34371179 PMCID: PMC8821927 DOI: 10.1016/j.ymthe.2021.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/04/2021] [Accepted: 07/29/2021] [Indexed: 02/04/2023] Open
Abstract
Inflammation plays a key role in the development of age-related diseases. In Alzheimer's disease, neuronal cell death is attributed to amyloidbeta oligomers that trigger microglial activation. Stem cells have shown promise as therapies for inflammatory diseases- because of their paracrine activity combined with their ability to respond to the inflammatory environment. However, the mechanisms underlying stem cell-promoted neurological recovery are poorly understood. To elucidate these mechanisms, we first primed stem cells with the secretome of lipopolysaccharide- or amyloidbeta-activated microglia. Then, we compared the immunomodulatory effects of extracellular vesicles (EVs) secreted from primed and non-primed stem cells. Our results demonstrate that EVs from primed cells are more effective in inhibiting microglia and astrocyte activation, amyloid deposition, demyelination, memory loss and motor and anxiety-like behavioral dysfunction, compared to EVs from non-primed cells. MicroRNA (miRNA) profiling revealed the upregulation of at least 19 miRNAs on primed-stem cell EVs. The miRNA targets were identified, and KEGG pathway analysis showed that the overexpressed miRNAs target key genes on the toll-like receptor-4 (TLR4) signaling pathway. Overall, our results demonstrate that priming mesenchymal stem cells (MSCs) with the secretome of activated microglia results in the release of miRNAs from EVs with enhanced immune regulatory potential able to fight neuroinflammation.
Collapse
Affiliation(s)
- Eleni Markoutsa
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA,College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL, USA,Corresponding author: Eleni Markoutsa, Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Karthick Mayilsamy
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA
| | - Dannielle Gulick
- Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA
| | - Shyam S. Mohapatra
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA,College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL, USA
| | - Subhra Mohapatra
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA,Corresponding author: Subhra Mohapatra, Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
6
|
Song Y, Wu Z, Zhao P. The protective effects of activating Sirt1/NF-κB pathway for neurological disorders. Rev Neurosci 2021; 33:427-438. [PMID: 34757706 DOI: 10.1515/revneuro-2021-0118] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/08/2021] [Indexed: 12/30/2022]
Abstract
Sirt1, a member of the sirtuins family, is a nicotinamide adenosine dinucleotide (NAD+)-dependent deacetylase. It can be involved in the regulation of several processes including inflammatory response, apoptosis, oxidative stress, energy metabolism, and autophagy by exerting deacetylation. Nuclear factor-κB (NF-κB), a crucial nuclear transcription factor with specific DNA binding sequences, exists in almost all cells and plays a vital role in several biological processes involving inflammatory response, immune response, and apoptosis. As the hub of multiple intracellular signaling pathways, the activity of NF-κB is regulated by multiple factors. Sirt1 can both directly deacetylate NF-κB and indirectly through other molecules to inhibit its activity. We would like to emphasize that Sirt1/NF-κB is a signaling pathway that is closely related to neuroinflammation. Many recent studies have demonstrated the neuroprotective effects of Sirt1/NF-κB signaling pathway activation applied to the treatment of neurological related diseases. In this review, we focus on new advances in the neuroprotective effects of the Sirt1/NF-κB pathway. First, we briefly review Sirt1 and NF-κB, two key molecules of cellular metabolism. Next, we discuss the connection between NF-κB and neuroinflammation. In addition, we explore how Sirt1 regulates NF-κB in nerve cells and relevant evidence. Finally, we analyze the therapeutic effects of the Sirt1/NF-κB pathway in several common neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
7
|
Huang R, Hu Z, Chen X, Cao Y, Li H, Zhang H, Li Y, Liang L, Feng Y, Wang Y, Su W, Kong Z, Melgiri ND, Jiang L, Li X, Du J, Chen Y. The Transcription Factor SUB1 Is a Master Regulator of the Macrophage TLR Response in Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004162. [PMID: 34378353 PMCID: PMC8498911 DOI: 10.1002/advs.202004162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/24/2021] [Indexed: 12/26/2022]
Abstract
Toll-like receptor 2 and 4 (TLR2, TLR4) signaling is implicated in atherosclerotic plaque formation. The two-stage master regulator Virtual Inference of Protein-activity by Enriched Regulon (VIPER) analysis of macrophage TLR2 and TLR4 signature genes integrated with coexpression network genes derived from 371 patient-derived carotid specimens identifies activated RNA polymerase II transcriptional coactivator p15 (SUB1/Sub1, PC4) as a master regulon in the atherogenic TLR response. It is found that TLR2 and TLR4 signaling is proinflammatory and proatherosclerotic in chow-fed apolipoprotein E-deficient (ApoE-/- ) mice. Through transgenic myeloid-specific Sub1 knockout in ApoE-/- mice, it is discovered that these proatherosclerotic effects of TLR2 and TLR4 signaling are mediated by Sub1. Sub1 knockout in macrophages enhances anti-inflammatory M2 macrophage polarization and cholesterol efflux. Irradiated low density lipoprotein receptor-deficient (Ldlr-/- ) mice transplanted with Sub1-/- murine bone marrow display reduced atherosclerosis. Promoter analysis reveals Sub1-dependent activation of interferon regulatory factor 1 (Irf1) transcription in a casein kinase 2 (Ck2)-dependent manner, and Sub1-knockout macrophages display decreased Irf1 expression. Artificial Irf1 overexpression in Sub1-knockout macrophages enhances proinflammatory M1 skewing and lowers cholesterol clearance. In conclusion, the TLR master regulon Sub1, and its downstream effect on the transcription factor Irf1, promotes a proinflammatory M1 macrophage phenotype and enhances atherosclerotic burden in vivo.
Collapse
Affiliation(s)
- Rongzhong Huang
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zicheng Hu
- Institute of Ultrasound ImagingThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaorui Chen
- Department of Pulmonary and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Yu Cao
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Hongrong Li
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Hong Zhang
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Yongyong Li
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Liwen Liang
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Yuxing Feng
- Department of Rehabilitation and Pain MedicineThe Ninth People's Hospital of ChongqingChongqing400700China
| | - Ying Wang
- Department of Rehabilitation MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Wenhua Su
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Zerui Kong
- Department of Cardiothoracic SurgeryThe Affiliated Yan An Hospital of Kunming Medical UniversityKunming650000China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunming650500China
| | - ND Melgiri
- Impactys Foundation for Biomedical ResearchSan DiegoCA92121USA
| | - Lihong Jiang
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Xingsheng Li
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Jianlin Du
- Department of CardiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Yunqing Chen
- Department of CardiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| |
Collapse
|
8
|
Mohamed RA, Abdallah DM, El-brairy AI, Ahmed KA, El-Abhar HS. Palonosetron/Methyllycaconitine Deactivate Hippocampal Microglia 1, Inflammasome Assembly and Pyroptosis to Enhance Cognition in a Novel Model of Neuroinflammation. Molecules 2021; 26:5068. [PMID: 34443654 PMCID: PMC8401912 DOI: 10.3390/molecules26165068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022] Open
Abstract
Since westernized diet-induced insulin resistance is a risk factor in Alzheimer's disease (AD) development, and lipopolysaccharide (LPS) coexists with amyloid β (Aβ)1-42 in these patients, our AD novel model was developed to resemble sporadic AD by injecting LPS into high fat/fructose diet (HFFD)-fed rats. The neuroprotective potential of palonosetron and/or methyllycaconitine, 5-HT3 receptor and α7 nAChR blockers, respectively, was evaluated after 8 days of daily administration in HFFD/LPS rats. All regimens improved histopathological findings and enhanced spatial memory (Morris Water Maze); however, palonosetron alone or with methyllycaconitine promoted animal performance during novel object recognition tests. In the hippocampus, all regimens reduced the expression of glial fibrillary acidic protein and skewed microglia M1 to M2 phenotype, indicated by the decreased M1 markers and the enhanced M2 related parameters. Additionally, palonosetron and its combination regimen downregulated the expression of ASC/TMS1, as well as levels of inflammasome downstream molecules and abated cleaved caspase-1, interleukin (IL)-1β, IL-18 and caspase-11. Furthermore, ACh and 5-HT were augmented after being hampered by the insult. Our study speculates that blocking 5-HT3 receptor using palonosetron overrides methyllycaconitine to combat AD-induced neuroinflammation and inflammasome cascade, as well as to restore microglial function in a HFFD/LPS novel model for sporadic AD.
Collapse
Affiliation(s)
- Reem A. Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Dalaal M. Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| | - Amany I. El-brairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Kawkab A. Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Hanan S. El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| |
Collapse
|
9
|
Garcia-Contreras M, Thakor AS. Human adipose tissue-derived mesenchymal stem cells and their extracellular vesicles modulate lipopolysaccharide activated human microglia. Cell Death Discov 2021; 7:98. [PMID: 33972507 PMCID: PMC8110535 DOI: 10.1038/s41420-021-00471-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/05/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's disease (AD), are driven by neuroinflammation triggered by activated microglial cells; hence, the phenotypic regulation of these cells is an appealing target for intervention. Human adipose tissue-derived mesenchymal stem cells (hAD-MSCs) may be a potential therapeutic candidate to treat NDs given their immunomodulatory properties. Evidence suggests that the mechanism of action of hAD-MSCs is through their secretome, which includes secreted factors such as cytokines, chemokines, or growth factors as well as extracellular vesicles (EVs). Recently, EVs have emerged as important mediators in cell communication given, they can transfer proteins, lipids, and RNA species (i.e., miRNA, mRNA, and tRNAs) to modulate recipient cells. However, the therapeutic potential of hAD-MSCs and their secreted EVs has not been fully elucidated with respect to human microglia. In this study, we determined the therapeutic potential of different hAD-MSCs doses (200,000, 100,000, and 50,000 cells) or their secreted EVs (50, 20, or 10 µg/ml), on human microglial cells (HMC3) that were activated by lipopolysaccharides (LPS). Upregulation of inducible nitric oxide synthase (iNOS), an activation marker of HMC3 cells, was prevented when they were cocultured with hAD-MSCs and EVs. Moreover, hAD-MSCs inhibited the secretion of proinflammatory factors, such as IL-6, IL-8, and MCP-1, while their secreted EVs promoted the expression of anti-inflammatory mediators such as IL-10 or TIMP-1 in activated microglia. The present data therefore support a role for hAD-MSCs and their secreted EVs, as potential therapeutic candidates for the treatment of NDs.
Collapse
Affiliation(s)
- Marta Garcia-Contreras
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA.
| |
Collapse
|
10
|
CB2 receptors modulate seizure-induced expression of pro-inflammatory cytokines in the hippocampus but not neocortex. Mol Neurobiol 2021; 58:4028-4037. [PMID: 33907944 DOI: 10.1007/s12035-021-02395-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
We compared neuroinflammatory responses induced by nonconvulsive and convulsive seizures and analyzed the role that may be played by cannabinoid CB2 receptors in the neuroinflammatory response induced by generalized tonic-clonic seizures (GTCS). Using quantitative PCR, we analyzed expression of interleukin-1b, CCL2, interleukin-6, tumor necrosis factor (TNF), transforming growth factor beta 1 (TGFb1), fractalkine, and cannabinoid receptor type 2 in the neocortex, dorsal and ventral hippocampus, cortical leptomeninges, dura mater, and spleen in 3 and 6 h after induction of GTCS by a high dose of pentylenetetrazole (PTZ, 70 mg/kg) and absence-like activity by a low dose of PTZ (30 mg/kg). The low dose of PTZ had no effect on the gene expression 3 and 6 h after PTZ injection. In 3 and 6 h after high PTZ dose, the expression of CCL2 and TNF increased in the neocortex. Both ventral and dorsal parts of the hippocampus responded to seizures by elevation of CCL2 expression 3 h after PTZ. Cortical leptomeninges but not dura mater also had elevated CCL2 level and decreased TGFb1 expression 3 h after GTCS. Activation of CB2 receptors by HU308 suppressed an inflammatory response only in the dorsal hippocampus but not neocortex. Suppression of CB2 receptors by AM630 potentiated expression of inflammatory cytokines also in the hippocampus but not in the neocortex. Thus, we showed that GTCS, but not the absence-like activity, provoke inflammatory response in the neocortex, dorsal and ventral hippocampus, and cortical leptomeninges. Modulation of CB2 receptors changes seizure-induced neuroinflammation only in the hippocampus but not neocortex.
Collapse
|
11
|
Nair M, Jagadeeshan S, Katselis G, Luan X, Momeni Z, Henao-Romero N, Chumala P, Tam JS, Yamamoto Y, Ianowski JP, Campanucci VA. Lipopolysaccharides induce a RAGE-mediated sensitization of sensory neurons and fluid hypersecretion in the upper airways. Sci Rep 2021; 11:8336. [PMID: 33863932 PMCID: PMC8052339 DOI: 10.1038/s41598-021-86069-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Thoracic dorsal root ganglia (tDRG) contribute to fluid secretion in the upper airways. Inflammation potentiates DRG responses, but the mechanisms remain under investigation. The receptor for advanced glycation end-products (RAGE) underlies potentiation of DRG responses in pain pathologies; however, its role in other sensory modalities is less understood. We hypothesize that RAGE contributes to electrophysiological and biochemical changes in tDRGs during inflammation. We used tDRGs and tracheas from wild types (WT), RAGE knock-out (RAGE-KO), and with the RAGE antagonist FPS-ZM1, and exposed them to lipopolysaccharides (LPS). We studied: capsaicin (CAP)-evoked currents and action potentials (AP), tracheal submucosal gland secretion, RAGE expression and downstream pathways. In WT neurons, LPS increased CAP-evoked currents and AP generation, and it caused submucosal gland hypersecretion in tracheas from WT mice exposed to LPS. In contrast, LPS had no effect on tDRG excitability or gland secretion in RAGE-KO mice or mice treated with FPS-ZM1. LPS upregulated full-length RAGE (encoded by Tv1-RAGE) and downregulated a soluble (sRAGE) splice variant (encoded by MmusRAGEv4) in tDRG neurons. These data suggest that sensitization of tDRG neurons contributes to hypersecretion in the upper airways during inflammation. And at least two RAGE variants may be involved in these effects of LPS.
Collapse
Affiliation(s)
- Manoj Nair
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Santosh Jagadeeshan
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - George Katselis
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Xiaojie Luan
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Zeinab Momeni
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Nicolas Henao-Romero
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Paulos Chumala
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Julian S Tam
- Department of Medicine, Division of Respirology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, 920-8640, Japan
| | - Juan P Ianowski
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Verónica A Campanucci
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
12
|
Yao Y, Fu KY. Serum-deprivation leads to activation-like changes in primary microglia and BV-2 cells but not astrocytes. Biomed Rep 2020; 13:51. [PMID: 32974017 DOI: 10.3892/br.2020.1358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to determine the effect of serum deprivation on primary microglia, BV-2 cells and primary astrocytes. Cell morphology combined with the expression of phospho-(p-)38 and p-extracellular signal-regulated kinase (ERK) were assessed. Serum deprivation resulted in various alterations in the three cell cultures. Primary microglia and BV-2 cells exhibited alterations indicative of activation under serum treatment, as well as lipopolysaccharide (LPS) treatment. However, astrocytes did not react as fast. Regarding morphology, the processes present on the primary microglia and BV-2 cells became shorter and the cell bodies became larger, and more transparent vesicles were observed within the cell bodies, which indicated their increased phagocytic ability. At the protein level, p-p38 expression increased quickly within 1 h in the primary microglia culture in response to LPS treatment. Furthermore, the expression levels of p-p38 and p-ERK were elevated in both primary microglia and BV-2 cells under serum deprivation, as well as under LPS treatment, which was not observed in the primary astrocytes. These results suggest that serum deprivation may result in similar changes to cell morphology and the expression levels of p-p38 and p-ERK as LPS treatment in primary microglia and BV-2 cells. These observations suggest that primary microglia and BV-2 cells may become activated under serum deprivation, at least to a certain degree.
Collapse
Affiliation(s)
- Yao Yao
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Beijing 100050, P.R. China
| | - Kai-Yuan Fu
- Center for TMD and Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| |
Collapse
|
13
|
Adhikari Y, Jin X. Intraperitoneal injection of lipopolysaccharide prevents seizure-induced respiratory arrest in a DBA/1 mouse model of SUDEP. Epilepsia Open 2020; 5:386-396. [PMID: 32913947 PMCID: PMC7469803 DOI: 10.1002/epi4.12410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 03/30/2020] [Accepted: 05/03/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Sudden unexpected death in epilepsy (SUDEP) is the cause of premature death of 50% patients with chronic refractory epilepsy. Respiratory failure during seizures is regarded as an important mechanism of SUDEP. Previous studies have shown that abnormal serotonergic neurotransmission is involved in the pathogenesis of seizure-induced respiratory failure, while enhancing serotonergic neurotransmission in the brainstem suppresses it. Because peripheral inflammation is known to enhance serotonergic neuron activation and 5-HT synthesis and release, we investigated the effect of intraperitoneal lipopolysaccharide (LPS)-induced inflammation on the S-IRA susceptibility during audiogenic seizures in DBA/1 mice. METHODS After DBA/1 mice were primed by exposing to sound stimulation for three consecutive days, they were tested for seizure severity and seizure-induced respiratory arrest (S-IRA) induced by sound stimulation under different conditions. We determined the dose and time course of the effects of intraperitoneal administration of LPS on audiogenic seizures and S-IRA. The effects of blocking TLR4 or RAGE receptors and blocking 5-HT receptors on the LPS-induced effect on S-IRA were investigated. Statistical significance was evaluated using the Kruskal-Wallis test. RESULTS Intraperitoneal injection of LPS significantly had dose-dependent effects in reducing the incidence of S-IRA as well as seizure severity in DBA/1 mice. The protective effect of LPS on S-IRA peaked at 8-12 hours after LPS injection and was related to both reducing seizure severity and enhancing autoresuscitation. Blocking TLR4 or RAGE receptor with TAK-242 or FPS-ZM1, respectively, prior to LPS injection attenuated its effects on S-IRA and seizure severity. Injection of a nonselective 5-HT receptor antagonist, cyproheptadine, or a 5-HT3 receptor antagonist, ondansetron, was effective in blocking LPS-induced effect on S-IRA. Immunostaining results showed a significant increase in c-Fos-positive serotonergic neurons in the dorsal raphe. SIGNIFICANCE This is the first study that demonstrates the effect of intraperitoneal LPS injection-induced inflammation on reducing S-IRA susceptibility and provides additional evidence supporting the serotonin hypothesis on SUDEP. Our study suggests that inflammation may enhance brainstem 5-HT neurotransmission to promote autoresuscitation during seizure and prevent SUDEP.
Collapse
Affiliation(s)
- Yadav Adhikari
- Spinal Cord and Brain Injury Research GroupStark Neurosciences Research Institute. Indiana University School of MedicineIndianapolisIndianaUSA
| | - Xiaoming Jin
- Department of Anatomy, Cell Biology and PhysiologyStark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Neurological SurgeryStark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Spinal Cord and Brain Injury Research GroupStark Neurosciences Research Institute. Indiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
14
|
Zhao Y, Yang J, Li C, Zhou G, Wan H, Ding Z, Wan H, Zhou H. Role of the neurovascular unit in the process of cerebral ischemic injury. Pharmacol Res 2020; 160:105103. [PMID: 32739425 DOI: 10.1016/j.phrs.2020.105103] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Cerebral ischemic injury exhibits both high morbidity and mortality worldwide. Traditional research of the pathogenesis of cerebral ischemic injury has focused on separate analyses of the involved cell types. In recent years, the neurovascular unit (NVU) mechanism of cerebral ischemic injury has been proposed in modern medicine. Hence, more effective strategies for the treatment of cerebral ischemic injury may be provided through comprehensive analysis of brain cells and the extracellular matrix. However, recent studies that have investigated the function of the NVU in cerebral ischemic injury have been insufficient. In addition, the metabolism and energy conversion of the NVU depend on interactions among multiple cell types, which make it difficult to identify the unique contribution of each cell type. Therefore, in the present review, we comprehensively summarize the regulatory effects and recovery mechanisms of four major cell types (i.e., astrocytes, microglia, brain-microvascular endothelial cells, and neurons) in the NVU under cerebral ischemic injury, as well as discuss the interactions among these cell types in the NVU. Furthermore, we discuss the common signaling pathways and signaling factors that mediate cerebral ischemic injury in the NVU, which may help to provide a theoretical basis for the comprehensive elucidation of cerebral ischemic injury.
Collapse
Affiliation(s)
- Yu Zhao
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Guoying Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haofang Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
15
|
Tetramethylpyrazine attenuates endotoxin-induced retinal inflammation by inhibiting microglial activation via the TLR4/NF-κB signalling pathway. Biomed Pharmacother 2020; 128:110273. [PMID: 32460188 DOI: 10.1016/j.biopha.2020.110273] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/21/2022] Open
Abstract
Ocular inflammation is a common pathological condition of a series of retinal degenerative diseases. Tetramethylpyrazine (TMP), a Chinese herbal extraction, is widely used in the treatment of several ocular diseases in Eastern countries. However, the exact mechanisms correlating the vision protective effects of TMP have not been elucidated. Thus, this study aimed to investigate TMP's molecular targets in anti-inflammatory activity in endotoxin lipopolysaccharide (LPS)-induced retinal inflammation both in vitro and in vivo. The primary cultured retinal microglial cells were pretreated with TMP and then activated by LPS. We found pretreatment with TMP significantly inhibited LPS-induced upregulation of CD68, a marker of mononuclear microglia activation. The morphological changes induced by LPS were also inhibited by the TMP pretreatment. Moreover, Toll like receptor 4 (TLR4), phosphorylation of inhibitor of NF-κB alpha (p-IκB-α) and the translocation of nuclear factor kappa B p65 (NF-κB p65) were significantly downregulated in retinal microglial cells with TMP pretreatment, which indicated that TMP might suppress LPS-induced retinal microglial activation through TLR4/NF-κB signalling pathway. And these results were confirmed in vivo. Pretreatment with TMP inhibited microglial activation, migration and regeneration, especially in ganglion cell layer (GCL). In addition to the inhibition of TLR4, TMP significantly inhibited the translocation of NF-κB p-65 to the nucleus in vivo. The downstream genes of NF-κB, such as the pro-inflammatory cytokines interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β), were significantly downregulated by TMP pretreatment in the retina. Accordingly, the increased expression of cleaved caspase-3 and the decreased ratio of B-cell lymphoma-2 (Bcl-2) to Bcl-2 associated X Protein (Bax) were significantly attenuated by TMP. TUNEL assay also demonstrated that TMP exerted neuroprotective effects in the retina. Therefore, this study elucidated a novel mechanism that TMP inhibits retinal inflammation by inhibiting microglial activation via a TLR4/NF-κB signalling pathway.
Collapse
|
16
|
Niu X, Zang L, Li W, Xiao X, Yu J, Yao Q, Zhao J, Ye Z, Hu Z, Li W. Anti-inflammatory effect of Yam Glycoprotein on lipopolysaccharide-induced acute lung injury via the NLRP3 and NF-κB/TLR4 signaling pathway. Int Immunopharmacol 2019; 81:106024. [PMID: 31784404 DOI: 10.1016/j.intimp.2019.106024] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/23/2019] [Accepted: 11/02/2019] [Indexed: 12/13/2022]
Abstract
Acute lung injury (ALI) is a common lung disease accompanied by acute and persistent pulmonary inflammatory response syndrome, which leads to alveolar epithelial cells and capillary endothelial cell damage. Yam glycoprotein, separated from traditional Chinese yam, has been shown to have anti-inflammatory and immunomodulatory effects. In this experiment, we mainly studied the therapeutic effect and mechanism of a glycoprotein on the lipopolysaccharide (LPS)-induced ALI mice. An oral glycoprotein method was used to treat the mouse ALI model induced by LPS injection in the peritoneal cavity. Afterward, we measured the wet/dry (W/D) ratio, the activity of myeloperoxidase (MPO), the oxidative index superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-PX) and the production of inflammatory cytokines interleukin-1β (IL-1β), tumour necrosis factor-α (TNF-α), and interleukin-6 (IL-6) to evaluate the effect of yam glycoprotein on lung tissue changes. We examined the protein expression of TLR4, ASC, NF-κBp65, p-NF-κBp65, Caspase-1, IκB, NLRP3, p-IκB, and β-actin by western blot analysis. Immunohistochemical analyses of NLRP3 and p-p65 in lung tissue were carried out to assess the mechanism of glycoprotein action. This result suggests that glycoprotein markedly depressed LPS-induced lung W/D ratio, MPO activity, MDA content SOD and GSH-Px depletion, and the contents of inflammatory cytokines IL-1β, IL-6, and TNF-α. Moreover, glycoprotein blocked TLR4/NF-κBp65 signaling activation and NLRP3inflammasome expression in LPS-induced ALI mice. As this particular study shows, glycoprotein has a safeguarding effects on LPS-induced ALI mice, possibly via activating NLRP3inflammasome and TLR4/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - Wenqi Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - JinJin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - Qing Yao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - Jinmeng Zhao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China
| | - Zhaobo Ye
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhen Hu
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, PR China.
| |
Collapse
|