1
|
Ding Y, Wang Y, Qi M, Zhang X, Wu D. Pioglitazone Modulates Microglia M1/M2 Polarization Through PPAR-γ Pathway and Exerts Neuroprotective Effects in Experimental Subarachnoid Hemorrhage. Mol Neurobiol 2025; 62:5930-5946. [PMID: 39668302 DOI: 10.1007/s12035-024-04664-w] [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: 11/29/2023] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Subarachnoid hemorrhage (SAH), a subtype of hemorrhagic stroke primarily resulting from the rupture of intracranial aneurysms, remains a significant contributor to disability and mortality, notwithstanding advancements in treatment. This study investigates the neuroprotective effects of pioglitazone in SAH, focusing on the PPAR-γ pathway and its potential role in mitigating early brain injury (EBI) following SAH. Neuroprotective efficacy was assessed through neurobehavioral assessment, brain water content analysis, TUNEL, immunofluorescence, western blotting, and inflammatory factor assay. Results indicate that pioglitazone treatment effectively mitigated brain edema, reduced neuronal death, and enhanced short-term neurobehavioral function in SAH-afflicted rats. Furthermore, pioglitazone demonstrated sustained improvements in long-term neurobehavioral function and decreased neuronal loss post-SAH. Mechanistically, SAH induced the polarization of microglia towards the M1 phenotype and the release of pro-inflammatory cytokines. Conversely, pioglitazone treatment predominantly shifted microglia polarization towards the M2 phenotype, eliciting a notable release of anti-inflammatory cytokines. Notably, the positive effects of pioglitazone were nullified by the PPAR-γ inhibitor T0070907. In conclusion, our findings suggest that pioglitazone may alleviate neuroinflammation by modulating microglia M1/M2 polarization through the PPAR-γ pathway, thereby conferring neuroprotection against SAH injury and positing itself as a potential therapeutic agent for SAH treatment.
Collapse
Affiliation(s)
- Yingying Ding
- Department of Neurosurgery, The Affiliated Yixing Hospital of Jiangsu University, Jiangsu Province, Yixing, 214200, China
| | - Yue Wang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA) Jiangsu Province, Wuxi, 214044, China
| | - Ming Qi
- Department of Neurosurgery, The Affiliated Yixing Hospital of Jiangsu University, Jiangsu Province, Yixing, 214200, China
| | - Xu Zhang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA) Jiangsu Province, Wuxi, 214044, China
| | - Da Wu
- Department of Neurosurgery, The Affiliated Yixing Hospital of Jiangsu University, Jiangsu Province, Yixing, 214200, China.
| |
Collapse
|
2
|
Zhao XP, Duan L, Zhao QR, Lv X, Tian NY, Yang SL, Dong K. NLRP3 inflammasome as a therapeutic target in doxorubicin-induced cardiotoxicity: role of phytochemicals. Front Pharmacol 2025; 16:1567312. [PMID: 40313623 PMCID: PMC12043718 DOI: 10.3389/fphar.2025.1567312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Doxorubicin (DOX) has received widespread attention as a broad-spectrum antitumor drug. However, it has been a recognized challenge that long-term DOX injections can lead to severe cardiotoxicity. There are numerous interventions to DOX-induced cardiotoxicity, and the most cost-effective is phytochemicals. It has been reported that phytochemicals have complex and diverse biological properties, facilitating the mitigation of DOX-induced cardiotoxicity. DOX-induced cardiotoxicity has numerous pathological mechanisms, and the nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome-mediated cardiomyocyte pyroptosis is one of them. This review initially presents an overview of the pathological mechanisms that underlie cardiotoxicity induced by DOX. Subsequently, we present a comprehensive elucidation of the structure and activation of the NLRP3 inflammasome. Finally, we provide a detailed summary of phytochemicals that can mitigate DOX-induced cardiotoxicity by influencing the expression of the NLRP3 inflammasome in cardiomyocytes.
Collapse
Affiliation(s)
- Xiao-Peng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Lian Duan
- China Volleyball College, Beijing Sport University, Beijing, China
- College of Physical Education, Yanshan University, Qinhuangdao, China
| | - Qian-Ru Zhao
- Shenyang Sports Research and Medical Center, Shenyang Sports Development Center, Shenyang, China
| | - Xing Lv
- Department of Rehabilitation, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Nai-Yuan Tian
- College of Physical Education, Yanshan University, Qinhuangdao, China
| | - Sheng-Lei Yang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Kun Dong
- College of Physical Education, Yanshan University, Qinhuangdao, China
| |
Collapse
|
3
|
Shu H, Liao Q, Chen Z, Liang M, Zhang S, Liu J, Wu Y, Hu P, Luo M, Zhu W, Zhu X, Yang L, Yan T. Flavonoids serve as a promising therapeutic agent for ischemic stroke. Brain Res 2025; 1853:149528. [PMID: 39999903 DOI: 10.1016/j.brainres.2025.149528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Ischemic stroke (IS) continues to be a major public health concern and is characterized by significantly high mortality and disabling rates. Inhibiting nerve cells death and enhancing the repair of ischemic tissue are important treatment concepts for IS. Currently, the mainstream treatment strategies mainly focus on short-term care, which underscores the urgent need for novel therapeutic strategies for long-term care. Emerging data reveal that flavonoids have surfaced as promising candidates for IS patients' long-term care. Flavonoids can alleviate neuroinflammation and anti-apoptosis due to their characteristic pharmacological mechanisms. Clinical evidence suggests that long-term flavonoids intake improves IS patients' long-term outcomes. Though the effect of flavonoids in IS treatment has been explored for decades, the neuroprotective pharmacodynamics have not been well established. Thereby, the aim of current review is to summarize the pathways involved in neuroprotective effect of flavonoids. This review will also advance the potential of flavonoids as a viable clinical candidate for the treatment of IS.
Collapse
Affiliation(s)
- Hongxin Shu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiuye Liao
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihao Chen
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Mingyu Liang
- School of life sciences, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si Zhang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junzhe Liu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yanze Wu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ping Hu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming Luo
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenping Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Li Yang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tengfeng Yan
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
4
|
Jasim MH, Saadoon Abbood R, Sanghvi G, Roopashree R, Uthirapathy S, Kashyap A, Sabarivani A, Ray S, Mustafa YF, Yasin HA. Flavonoids in the regulation of microglial-mediated neuroinflammation; focus on fisetin, rutin, and quercetin. Exp Cell Res 2025; 447:114537. [PMID: 40147710 DOI: 10.1016/j.yexcr.2025.114537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Neuroinflammation is a critical mechanism in central nervous system (CNS) inflammatory disorders, encompassing conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), traumatic brain injury (TBI), encephalitis, spinal cord injury (SCI), and cerebral stroke. Neuroinflammation is characterized by increased blood vessel permeability, leukocyte infiltration, glial cell activation, and elevated production of inflammatory mediators, such as chemokines and cytokines. Microglia act as the resident macrophages of the central nervous system, serving as the principal defense mechanism in brain tissue. After CNS injury, microglia modify their morphology and downregulate genes that promote homeostatic functions. Despite comprehensive transcriptome analyses revealing specific gene modifications in "pathological" microglia, microglia's precise protective or harmful functions in neurological disorders remain insufficiently comprehended. Accumulating data suggests that the polarization of microglia into the M1 proinflammatory phenotype or the M2 antiinflammatory phenotype may serve as a sensible therapeutic strategy for neuroinflammation. Flavonoids, including rutin, fisetin, and quercetin, function as crucial chemical reservoirs with unique structures and diverse actions and are extensively used to modulate microglial polarization in treating neuroinflammation. This paper highlights the detrimental effects of neuroinflammation seen in neurological disorders such as stroke. Furthermore, we investigate their therapeutic benefits in alleviating neuroinflammation via the modulation of macrophage polarization.
Collapse
Affiliation(s)
- Mohannad Hamid Jasim
- Biology Department, College of Education, University of Fallujah, Fallujah, Iraq.
| | - Rosull Saadoon Abbood
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India.
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq.
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq.
| |
Collapse
|
5
|
Hassan MAM, Fahmy MI, Azzam HN, Ebrahim YM, El-Shiekh RA, Aboulmagd YM. Multifaceted therapeutic potentials of catalpol, an iridoid glycoside: an updated comprehensive review. Inflammopharmacology 2025:10.1007/s10787-025-01694-1. [PMID: 40097877 DOI: 10.1007/s10787-025-01694-1] [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: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Catalpol, classified as an iridoid glucoside, is recognized for its significant role in medicine, particularly in the treatment of various conditions such as diabetes mellitus, neuronal disorders, and inflammatory diseases. This review aims to evaluate the biological implications of catalpol and the mechanisms underlying its diverse pharmacological effects. A thorough exploration of existing literature was conducted utilizing the keyword "Catalpol" across prominent public domains like Google Scholar, PubMed, and EKB. Catalpol has demonstrated a diverse array of pharmacological effects in experimental models, showcasing its anti-diabetic, cardiovascular-protective, neuroprotective, anticancer, hepatoprotective, anti-inflammatory, and antioxidant properties. In summary, catalpol manifests a spectrum of biological effects through a myriad of mechanisms, prominently featuring its anti-inflammatory and antioxidant capabilities. Its diverse pharmacological profile underscores its potential for therapeutic applications across a range of conditions. Further research is warranted to fully elucidate the clinical implications of catalpol and optimize its use in medical practice.
Collapse
Affiliation(s)
- Mennat-Allah M Hassan
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Hany N Azzam
- Pharmacy Practice Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Yara M Aboulmagd
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
6
|
Qian Q, Wu J, Wang C, Yang Z, Cheng Y, Zheng Y, Wang X, Wang H. 6-PPD triggered lipid metabolism disorder and inflammatory response in larval zebrafish (Danio rerio) by regulating PPARγ/NF-κB pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125785. [PMID: 39900129 DOI: 10.1016/j.envpol.2025.125785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/05/2025]
Abstract
As a synthetic rubber antioxidant, the environmental monitoring concentrations of N-(1,3-Dimethylbutyl)-N'-phenyl-p-phenylenediamine (6-PPD) have exceeded the risk threshold, attracting widespread attention. Although investigations into the harmful effects on zebrafish have commenced, a comprehensive exploration of its toxicological impacts and underlying molecular mechanisms remains to be conducted. By using zebrafish as a model, this study systematically evaluated 6-PPD-induced lipid metabolism disorders and inflammation response following environmental exposure. Bioinformatics analysis revealed that 6-PPD target genes enriched in the hepatitis B pathway, indicating potential hepatic toxicity via inflammatory pathways. Therefore, we hypothesize that 6-PPD could trigger hepatotoxicity through the crosstalk between lipid metabolism and inflammation. Further experiments substantiated this hypothesis by showing lipid accumulation in the liver following 6-PPD exposure, along with elevated triglyceride (TG) and total cholesterol (TC) levels, and imbalanced expression of lipid metabolism-related marker genes. Additionally, 6-PPD exposure induced the accumulation of reactive oxygen species (ROS) and inhibited the differentiation and maturation of immune cells, resulting in immune evasion. Most of these abnormalities were exacerbated in a dose-dependent manner with increasing concentrations of 6-PPD. The addition of the PPARγ pathway agonist puerarin (PUE) or NF-κB pathway inhibitor quinazoline (QNZ) to 6-PPD exposure group mitigated these toxic effects, validating our conjecture that lipid metabolism disorder and inflammatory responses may result from the regulation of the PPARγ/NF-κB pathway.
Collapse
Affiliation(s)
- Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ji Wu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Cuizhen Wang
- Sanquan College of Xinxiang Medical University, Xinxiang, 453513, China
| | - Zheng Yang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Ying Cheng
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
7
|
Zhang Y, Lv S, Huang P, Xiao L, Lin N, Huang E. Network pharmacology study on the mechanism of berberine in Alzheimer's disease model. NPJ Sci Food 2025; 9:16. [PMID: 39900946 PMCID: PMC11790853 DOI: 10.1038/s41538-025-00378-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: 06/24/2024] [Accepted: 01/06/2025] [Indexed: 02/05/2025] Open
Abstract
Research indicated that berberine (BBR) plays a protective role in modulating Alzheimer's disease (AD). This study aimed to explore the target genes of BBR associated with AD therapy using a network pharmacology study. Through network pharmacology analysis, two main potential target genes, β-amyloid precursor protein (APP) and peroxisome proliferator-activated receptor gamma (PPARG), of BBR for AD therapy were screened out. Further experiments demonstrated that BV2 and C8-D1A treated with BBR were decreased in the mRNA and protein expression of APP and presenilin 1 while PPARG was increased with a reduction in the NF-κB pathway. A similar result was shown in vivo. Through a network pharmacology study, this study supported that BBR played a protective role in the AD mice model via blocking APP processing and amyloid plaque formation. It also promotes PPARG expression to blockage of NF-κB pathway-mediated inflammatory response and neuroinflammation.
Collapse
Affiliation(s)
- Yaoyi Zhang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Shuai Lv
- Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Pinyuan Huang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lingmin Xiao
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Nan Lin
- Fujian Key Laboratory of Vascular Aging, Department of Geriatrics, Fujian Institute of Geriatrics, Fujian Clinical Research Center for Senile Vascular Aging and Brain Aging, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - En Huang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China.
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
8
|
Lin XX, Yang PQ, Li XJ, Xu ZZ, Wu HT, Hu SM, Yang XL, Ding Y, Yu WZ. Network pharmacology‑based analysis and in vitro experimental verification of the inhibitory role of luteoloside on gastric cancer cells via the p53/p21 pathway. Oncol Lett 2025; 29:76. [PMID: 39650229 PMCID: PMC11622105 DOI: 10.3892/ol.2024.14822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/16/2024] [Indexed: 12/11/2024] Open
Abstract
The present study aimed to investigate the inhibitory effect of luteoloside on the proliferation, migration and invasion of gastric cancer (GC) cells based on network pharmacology and in vitro experiments. GC-associated targets were obtained from the GeneCards and Online Mendelian Inheritance in Man databases. Gene Ontology functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were performed using the Database for Annotation, Visualization and Integrated Discovery. Protein-protein interaction (PPI) networks and herb-active ingredient-target gene-signaling pathway networks were constructed using the Search Tool for the Retrieval of Interacting Genes and proteins and Cytoscape software to analyze core target genes and pathways. In addition, the alkaline comet assay was performed to assess DNA damage, demonstrating that luteoloside induces DNA double-strand breaks in a concentration-dependent manner, as indicated by increased comet tail lengths. γ-H2AX detection through western blot analysis further corroborated these findings, showing significant upregulation of this DNA damage marker in luteoloside-treated GC cells. The human GC cell line NCI-N87 was utilized for in vitro experiments to investigate the impact of different doses of luteoloside on cell proliferation, invasion and migration using Cell Counting Kit-8, scratch-wound and Transwell assays, respectively. The underlying molecular mechanism of luteoloside was explored using western blot analysis. The successfully constructed PPI network revealed the p53, Akt1, Bcl-2 and Caspase-3 proteins as the core targets, all of which showed good binding activity with luteoloside. The in vitro experiments demonstrated that luteoloside treatment significantly inhibited GC-cell proliferation, migration and invasion. The western blot results showed notable concentration-dependent upregulation of p53 and p21 protein expression and downregulation of Bcl-2 protein expression following luteoloside treatment. Overall, luteoloside inhibited the proliferation, migration and invasion of GC cells by activating the p53/p21 signaling pathway.
Collapse
Affiliation(s)
- Xin-Xing Lin
- Department of General Surgery, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Pei-Qing Yang
- Department of Gastroenterology, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Xiao-Jun Li
- Department of General Surgery, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Zhong-Zhen Xu
- Department of Gastroenterology, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Hai-Tao Wu
- Department of General Surgery, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Shun-Ming Hu
- Department of Gastroenterology, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Xiao-Lei Yang
- Department of General Surgery, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Yong Ding
- Department of General Surgery, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| | - Wei-Zhou Yu
- Department of Gastroenterology, Dafeng People's Hospital, Yancheng, Jiangsu 224100, P.R. China
| |
Collapse
|
9
|
Tian HY, Lei YX, Zhou JT, Liu LJ, Yang T, Zhou Y, Ge JW, Xu C, Mei ZG. Insight into interplay between PANoptosis and autophagy: novel therapeutics in ischemic stroke. Front Mol Neurosci 2025; 17:1482015. [PMID: 39846000 PMCID: PMC11751022 DOI: 10.3389/fnmol.2024.1482015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
PANoptosis is a novelly defined mode of programmed cell death that involves the activation of multiple cellular death pathways, including pyroptosis, apoptosis, and necroptosis, triggering robust inflammatory reactions. Autophagy is a crucial cellular process that maintains cellular homeostasis and protects cells from various stresses. PANoptosis and autophagy, both vital players in the intricate pathological progression of ischemic stroke (IS), a brain ailment governed by intricate cell death cascades, have garnered attention in recent years for their potential interplay. While mounting evidence hints at a crosstalk between these two processes in IS, the underlying mechanisms remain elusive. Therefore, this review delves into and dissects the intricate mechanisms that underpin the intersection of PANoptosis and autophagy in this devastating condition. In conclusion, the crosstalk between PANoptosis and autophagy in IS presents a promising target for the development of novel stroke therapies. Understanding the interplay between these two pathways offers a much-needed insight into the underlying mechanisms of IS and opens the possibility for new therapeutic strategies.
Collapse
Affiliation(s)
- He-Yan Tian
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Shenzhen, China
| | - Yun-Xing Lei
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Shenzhen, China
| | - Jing-Tao Zhou
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Shenzhen, China
| | - Long-Jun Liu
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Shenzhen, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jin-Wen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Hunan Academy of Traditional Chinese Medicine, Changsha, China
| | - Chen Xu
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Shenzhen, China
| | - Zhi-Gang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
10
|
Muhammad AJ, Al-Baqami FF, Alanazi FE, Alattar A, Alshaman R, Rehman NU, Riadi Y, Shah FA. The Interplay of Carveol and All-Trans Retinoic Acid (ATRA) in Experimental Parkinson's Disease: Role of Inflammasome-Mediated Pyroptosis and Nrf2. Neurochem Res 2024; 49:3118-3130. [PMID: 39190122 DOI: 10.1007/s11064-024-04226-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/21/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
Parkinson's disease (PD) is a debilitating and the second most common neurodegenerative disorder with a high prevalence. PD has a multifaceted etiology characterized by an altered redox state and an excessive inflammatory response. Extensive research has consistently demonstrated the role of the nuclear factor E2-related factor (Nrf2) and inflammasomes, notably NLRP3 in neurodegenerative diseases. In this study, our focus was on exploring the potential neuroprotective properties of carveol in Parkinson's disease. Our findings suggest that carveol may exhibit these effects through Nrf2 and by suppressing pyroptosis. Male albino mice were treated with carveol, and the animal PD model was induced through a single intranigral dose of 2 µg/2µl lipopolysaccharide (LPS). To further demonstrate the essential role of the Nrf2 pathway, we utilized all-trans retinoic acid (ATRA) to inhibit the Nrf2. Our finding showed the induction of pyroptosis as evidenced by increased levels of NLRP3 and other inflammatory mediators, including IL-1β, iNOS, p-NFKB, and apoptotic cell death indicated by positive fluoro Jade B (FJB) staining. Moreover, increased levels of lipid peroxides and reactive oxygen species indicated a significant rise in oxidative stress due to LPS. The administration of carveol mitigates oxidative stress and suppresses inflammatory pathways through the augmentation of intrinsic antioxidant defenses, primarily via the activation of the Nrf2. Conversely, ATRA reversed carveol protective effects by increasing FJB-positive cells, inflammatory and oxidative biomarkers. Taken together, our findings suggest that carveol mitigated LPS-induced Parkinson-like symptoms, partially through the activation of the Nrf2 and downregulation of pyroptosis notably NLRP3.
Collapse
Affiliation(s)
- Asmaa Jan Muhammad
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Faisal F Al-Baqami
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia
| | - Fawaz E Alanazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Najeeb Ur Rehman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia
| | - Yassine Riadi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy Prince Sattam Bin Abdul Aziz University, Al-Kharj, Saudi Arabia
| | - Fawad Ali Shah
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia.
| |
Collapse
|
11
|
Guo X, Qin Y, Feng Z, Li H, Yang J, Su K, Mao R, Li J. Investigating the anti-inflammatory effects of icariin: A combined meta-analysis and machine learning study. Heliyon 2024; 10:e35307. [PMID: 39170422 PMCID: PMC11336647 DOI: 10.1016/j.heliyon.2024.e35307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Objective The objectives of this study were to define the superiority of icariin and its derivatives' anti-inflammatory activities and to create a reference framework for evaluating preclinical evidence. This method combines machine learning and meta-analysis to identify underlying biological pathways. Methods Data came from PubMed, Embase, Web of Science, and the Cochrane Library. SYRCLE was used to evaluate the risk of bias in a subset of research. Meta-analysis and detailed subgroup analyses, categorized by species, genders, disease type, dosage, and treatment duration, were performed using R and STATA 15.0 software to derive nuanced insights. Employing R software (version 4.2.3) and the tidymodels package, the analysis focused on constructing a model and selecting features, with TNF-α as the dependent variable. This approach aims to identify significant predictors of drug efficacy. An in-depth literature facilitated the synthesis of anti-inflammatory mechanisms attributed to icariin and its constituent compounds. Results Following a meticulous search and selection process, 19 studies, involving 370 and 260 animals were included in the meta-analysis and machine-learning assessment, respectively. The findings revealed that icariin and its derivatives markedly reduced inflammation markers, including TNF-α and IL-1β. Additionally, machine-learning outcomes, with TNF-α as the target variable, indicated enhanced anti-inflammatory effects of icariin across respiratory, urological, neurological, and digestive disease types. These effects were more pronounced at doses exceeding 27.52 mg/kg/day and treatment durations beyond 31.22 days. Conclusion Strong anti-inflammatory effects are exhibited by icariiin and its derivatives, which are especially beneficial in the management of digestive, neurological, pulmonary, and urinary conditions. Effective for periods longer than 31.22 days and at dosages more than 27.52 mg/kg/day. Subsequent research will involve more targeted animal experiments and safety assessments to obtain more comprehensive preclinical evidence.
Collapse
Affiliation(s)
- Xiaochuan Guo
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Yanqin Qin
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhenzhen Feng
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Haibo Li
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Jingfan Yang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Kailin Su
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Ruixiao Mao
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Jiansheng Li
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| |
Collapse
|
12
|
Huang HC, Shi YJ, Vo TLT, Hsu TH, Song TY. The Anti-Inflammatory Effects and Mechanism of the Submerged Culture of Ophiocordyceps sinensis and Its Possible Active Compounds. J Fungi (Basel) 2024; 10:523. [PMID: 39194849 DOI: 10.3390/jof10080523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
The pharmacological effects of the fruiting body of Ophiocordyceps sinensis (O. sinensis) such as antioxidant, anti-virus, and immunomodulatory activities have already been described, whereas the anti-inflammatory effects and active components of the submerged culture of O. sinesis (SCOS) still need to be further verified. This study aimed to investigate the active compounds in the fermented liquid (FLOS), hot water (WEOS), and 50-95% (EEOS-50, EEOS-95) ethanol extracts of SCOS and their anti-inflammatory effects and potential mechanisms in lipopolysaccharide (LPS)-stimulated microglial BV2 cells. The results demonstrated that all of the SCOS extracts could inhibit NO production in BV2 cells. EEOS-95 exhibited the strongest inhibitory effects (71% inhibitory ability at 500 µg/mL), and its ergosterol, γ-aminobutyric acid (GABA), total phenolic, and total flavonoid contents were significantly higher than those of the other extracts (18.60, 18.60, 2.28, and 2.14 mg/g, p < 0.05, respectively). EEOS-95 also has a strong inhibitory ability against IL-6, IL-1β, and TNF-α with an IC50 of 617, 277, and 507 µg/mL, respectively, which is higher than that of 1 mM melatonin. The anti-inflammatory mechanism of EEOS-95 seems to be associated with the up-regulation of PPAR-γ/Nrf-2/HO-1 antioxidant-related expression and the down-regulation of NF-κB/COX-2/iNOS pro-inflammatory expression signaling. In summary, we demonstrated that EEOS-95 exhibits neuroinflammation-mediated neurodegenerative disorder activities in LPS-induced inflammation in brain microglial cells.
Collapse
Affiliation(s)
- Hsien-Chi Huang
- PhD Program of Biotechnology and Bioindustry, College of Biotechnology and Bioresources, Da-Yeh University, Changhua 515, Taiwan
| | - Yu-Juan Shi
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Thuy-Lan-Thi Vo
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Tai-Hao Hsu
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Tuzz-Ying Song
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| |
Collapse
|
13
|
Ghosh A, Khanam N, Nath D. Solid lipid nanoparticle: A potent vehicle of the kaempferol for brain delivery through the blood-brain barrier in the focal cerebral ischemic rat. Chem Biol Interact 2024; 397:111084. [PMID: 38823537 DOI: 10.1016/j.cbi.2024.111084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Kaempferol is major flavonoid present in Convolvulus pluricaulis. This phytochemical protects the brain against oxidative stress, neuro-inflammation, neurotoxicity, neurodegeneration and cerebral ischemia induced neuronal destruction. Kaempferol is poorly water soluble. Our study proved that solid lipid nanoparticles (SLNs) were efficient carrier of kaempferol through blood-brain barrier (BBB). Kaempferol was incorporated into SLNs prepared from stearic acid with polysorbate 80 by the process of ultrasonication. Mean particle size and zeta potential of kaempferol loaded solid lipid nanoparticles (K-SLNs) were 451.2 nm and -15.0 mV. Atomic force microscopy showed that K-SLNs were spherical in shape. Fourier transformed infrared microscopy (FTIR) showed that both stearic acid and kaempferol were present in K-SLNs. X-ray diffraction (XRD) and differential scanning calorimetry (DSC) revealed that the matrices of K-SLNs were in untidy crystalline state. Entraptment efficiency of K-SLNs was 84.92%. In-vitro drug release percentage was 93.24%. Kaempferol loaded solid lipid nanoparticles (K-SLNs) showed controlled release profile. In-vitro uptake study showed significant efficiency of K-SLNs to cross blood-brain barrier (BBB). After oral administration into the focal cerebral ischemic rat, accumulation of fluorescent labeled K-SLNs was observed in the brain cortex which confirmed its penetrability into the brain. It significantly decreased the neurological deficit, infarct volume and level of reactive oxygen species (ROS) and decreased the level of pro-inflammatory mediators like NF-κB and p-STAT3. Damaged neurons and brain texture were improved. This study indicated increased bioavailability of kaempferol into the brain tissue through SLNs formulation.
Collapse
Affiliation(s)
- Ashutosh Ghosh
- Department of Zoology, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Nasima Khanam
- Department of Zoology, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Debjani Nath
- Department of Zoology, University of Kalyani, Nadia, West Bengal, 741235, India.
| |
Collapse
|
14
|
Cheng N, Cheng X, Tan F, Liang Y, Xu L, Wang J, Tan J. Electroacupuncture attenuates cerebral ischemia/reperfusion injury by regulating oxidative stress, neuronal death and neuroinflammation via stimulation of PPAR-γ. Acupunct Med 2024; 42:133-145. [PMID: 38351622 DOI: 10.1177/09645284231211600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
BACKGROUND Oxidative stress and inflammatory responses play essential roles in cerebral ischemia/reperfusion (I/R) injury. Electroacupuncture (EA) is widely used as a rehabilitation method for stroke in China; however, the underlying mechanism of action remains unclear. Peroxisome proliferator-activated receptor gamma (PPAR-γ) has been reported to impact anti-inflammatory and anti-oxidative effects. OBJECTIVE This study investigated the role of PPAR-γ in EA-mediated effects and aimed to illuminate its possible mechanisms in cerebral I/R. METHODS In this study, male Sprague-Dawley (SD) rats with middle cerebral artery occlusion/reperfusion (MCAO/R) injury were treated with EA at LI11 and ST36 for 30 min daily after MCAO/R for seven consecutive days. The neuroprotective effects of EA were measured by neurobehavioral evaluation, triphenyltetrazolium chloride staining, hematoxylin-eosin staining and transmission electron microscopy. Oxidative stress, inflammatory factors, neural apoptosis and microglial activation were examined by enzyme-linked immunosorbent assay, immunofluorescence and reverse transcriptase polymerase chain reaction. Western blotting was used to assess PPAR-γ-mediated signaling. RESULTS We found that EA significantly alleviated cerebral I/R-induced infarct volume, decreased neurological scores and inhibited I/R-induced oxidative stress, inflammatory responses and microglial activation. EA also increased PPAR-γ protein expression. Furthermore, the protective effects of EA were reversed by injection of the PPAR-γ antagonist T0070907. CONCLUSION EA attenuates cerebral I/R injury by regulating oxidative stress, neuronal death and neuroinflammation via stimulation of PPAR-γ.
Collapse
Affiliation(s)
- Nanfang Cheng
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Xinyuan Cheng
- The Fourth Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Feng Tan
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Yangui Liang
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Lihong Xu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Jian Wang
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Jiuqing Tan
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| |
Collapse
|
15
|
Lang W, Wen X, Zhang S, Liang X, Chen L, Zhang D, Zhou R, Ali I, Hu X, Zhang H, Cheng M. Cynaroside ameliorates methotrexate-induced enteritis in rats through inhibiting NLRP3 inflammasome activation. Front Immunol 2024; 15:1405084. [PMID: 38835771 PMCID: PMC11148340 DOI: 10.3389/fimmu.2024.1405084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction Cynaroside exhibits various biological properties, including anti-inflammatory, antiviral, antitumor, and cardioprotective effects. However, its involvement in methotrexate (MTX)-induced intestinal inflammation remains inadequately understood. Thus, we investigated the impact of cynaroside on MTX-induced intestinal inflammation and its potential mechanisms. Methods To assess the protective potential of cynaroside against intestinal inflammation, Sprague-Dawley rats were subjected to a regimen of 7 mg/kg MTX for 3 days, followed by treatment with cynaroside at varying doses (10, 20, or 40 mg/kg). Histopathological evaluations were conducted alongside measurements of inflammatory mediators to elucidate the involvement of the NLRP3 inflammasome in alleviating intestinal inflammation. Results Administration of 7 mg/kg MTX resulted in decreased daily food intake, increased weight loss, and elevated disease activity index in rats. Conversely, treatment with cynaroside at 20 or 40 mg/kg ameliorated the reductions in body weight and daily food intake and suppressed the MTX-induced elevation in the disease activity index. Notably, cynaroside administration at 20 or 40 mg/kg attenuated inflammatory cell infiltration, augmented goblet cell numbers and lowered serum levels of tumor necrosis factor-α, interleukin (IL)-1β, and IL-18, as well as the CD68-positive cell rate in the intestines of MTX-induced rats. Furthermore, cynaroside downregulated the expression levels of NLRP3, cleaved caspase 1, and cleaved IL-1β in MTX-induced rats. Discussion Collectively, our findings indicated that cymaroside alleviates intestinal inflammatory injury by inhibiting the activation of NLRP3 inflammasome in MTX-induced rats.
Collapse
Affiliation(s)
- Wuying Lang
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
- Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd., Shangluo, China
- Key Research Laboratory for Standardized Planting and Quality Improvement of Bulk Chinese Medicinal Materials in Shangluo, Shangluo University, Shangluo, China
| | - Xin Wen
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shuangqi Zhang
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
| | - Xuhua Liang
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
- Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd., Shangluo, China
- Key Research Laboratory for Standardized Planting and Quality Improvement of Bulk Chinese Medicinal Materials in Shangluo, Shangluo University, Shangluo, China
| | - Lin Chen
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
- Key Research Laboratory for Standardized Planting and Quality Improvement of Bulk Chinese Medicinal Materials in Shangluo, Shangluo University, Shangluo, China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Co. Ltd., Shangluo, China
| | - Ruina Zhou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ihsan Ali
- College of Veterinary Science Faculty of Animal Husbandry and Veterinary Science, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Xuansheng Hu
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
| | - Haihua Zhang
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Min Cheng
- College of Biology Pharmacy and Food Engineering, Shangluo University, Shangluo, China
- Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd., Shangluo, China
| |
Collapse
|
16
|
Zhou Y, Huang Y, Ye W, Chen Z, Yuan Z. Cynaroside improved depressive-like behavior in CUMS mice by suppressing microglial inflammation and ferroptosis. Biomed Pharmacother 2024; 173:116425. [PMID: 38490155 DOI: 10.1016/j.biopha.2024.116425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024] Open
Abstract
Depression is a common mental health disorder, and in recent years, the incidence of various forms of depression has been on the rise. Most medications for depression are highly dependency-inducing and can lead to relapse upon discontinuation. Therefore, novel treatment modalities and therapeutic targets are urgently required. Traditional Chinese medicine (TCM) offers advantages in the treatment of depression owing to its multi-target, multi-dimensional approach that addresses the root cause of depression by regulating organ functions and balancing Yin and Yang, with minimal side effects. Cynaroside (CNS), an extract from the traditional Chinese herb honeysuckle, is a flavonoid compound with antioxidant properties. In this study, network pharmacology identified 44 potential targets of CNS associated with depression and several highly correlated inflammatory signaling pathways. CNS alleviated LPS-induced M1 polarization and the release of inflammatory factors in BV-2 cells. Transcriptomic analysis and validation revealed that CNS reduced inflammatory polarization, lipid peroxidation, and ferroptosis via the IRF1/SLC7A11/GPX4 signaling pathway. In vivo experiments showed that CNS treatment had effects similar to those of fluoxetine (FLX). It effectively ameliorated anxiety-, despair-, and anhedonia-like states in chronic unpredictable mild stress (CUMS)-induced mice and reduced microglial activation in the hippocampus. Thus, we conclude that CNS exerts its therapeutic effect on depression by inhibiting microglial cells from polarizing into the M1 phenotype and reducing inflammation and ferroptosis levels. This study provides further evidence that CNS is a potential antidepressant, offering new avenues for the treatment of depression.
Collapse
Affiliation(s)
- Yiwei Zhou
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yuhan Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Wei Ye
- School Of Chinese Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Zijie Chen
- Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Zhengzhong Yuan
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
17
|
Zhang D, Zhang J, Ma Z, Wu Q, Liu M, Fan T, Ding L, Ren D, Wen A, Wang J. Luteoloside inhibits Aβ1-42 fibrillogenesis, disintegrates preformed fibrils, and alleviates amyloid-induced cytotoxicity. Biophys Chem 2024; 306:107171. [PMID: 38194817 DOI: 10.1016/j.bpc.2023.107171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/17/2023] [Accepted: 12/30/2023] [Indexed: 01/11/2024]
Abstract
Abnormal aggregation and fibrillogenesis of amyloid-β protein (Aβ) can cause Alzheimer's disease (AD). Thus, the discovery of effective drugs that inhibit Aβ fibrillogenesis in the brain is crucial for the treatment of AD. Luteoloside, as one of the polyphenolic compounds, is found to have a certain therapeutic effect on nervous system diseases. However, it remains unknown whether luteoloside is a potential drug for treating AD by modulating Aβ aggregation pathway. In this study, we performed diverse biophysical and biochemical methods to explore the inhibition of luteoloside on Aβ1-42 which is linked to AD. The results demonstrated that luteoloside efficiently prevented amyloid oligomerization and cross-β-sheet formation, reduced the rate of amyloid growth and the length of amyloid fibrils in a dose-dependent manner. Moreover, luteoloside was able to influence aggregation and conformation of Aβ1-42 during different fiber-forming phases, and it could disintegrate already preformed fibrils of Aβ1-42 and convert them into nontoxic aggregates. Furthermore, luteoloside protected cells from amyloid-induced cytotoxicity and hemolysis, and attenuated the level of reactive oxygen species (ROS). The molecular docking study showed that luteoloside interacted with Aβ1-42 mainly via Conventional Hydrogen Bond, Carbon Hydrogen Bond, Pi-Pi T-shaped, Pi-Alkyl and Pi-Anion, thereby possibly preventing it from forming the aggregates. These observations indicate that luteoloside, a natural anti-oxidant molecule, may be applicable as an effective inhibitor of Aβ, and promote further exploration of the therapeutic strategy against AD.
Collapse
Affiliation(s)
- Di Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Juanli Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zhongying Ma
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Qianwen Wu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Meiyou Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Tingting Fan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Likun Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Danjun Ren
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
18
|
Cheng Y, Zhang Y, Huang P, Cheng Q, Ding H. Luteolin ameliorates pentetrazole-induced seizures through the inhibition of the TLR4/NF-κB signaling pathway. Epilepsy Res 2024; 201:107321. [PMID: 38382229 DOI: 10.1016/j.eplepsyres.2024.107321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/21/2023] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
Epilepsy represents a prevalent neurological disorder in the population, and the existing antiepileptic drugs (AEDs) often fail to adequately control seizures. Inflammation is recognized as a pivotal factor in the pathophysiology of epilepsy. Luteolin, a natural flavonoid extract, possesses anti-inflammatory properties and exhibits promising neuroprotective activity. Nevertheless, the precise molecular mechanisms underlying the antiepileptic effects of luteolin remain elusive. In this study, we established a rat model of epilepsy using pentylenetetrazole (PTZ) to induce seizures. A series of behavioral experiments were conducted to assess behavioral abilities and cognitive function. Histological techniques, including HE staining, Nissl staining, and TUNEL staining, were employed to assess hippocampal neuronal damage. Additionally, Western blotting, RT-qPCR, and ELISA were utilized to analyze the expression levels of proteins involved in the TLR4/IκBα/NF-κB signaling pathway, transcription levels of apoptotic factors, and levels of inflammatory cytokines, respectively. Luteolin exhibited a dose-dependent reduction in seizure severity, prolonged the latency period of seizures, and shortened seizure duration. Furthermore, luteolin prevented hippocampal neuronal damage in PTZ-induced epileptic rats and partially restored behavioral function and learning and memory abilities. Lastly, PTZ kindling activated the TLR4/IκBα/NF-κB pathway, leading to elevated levels of the cytokines TNF-α, IL-6 and IL-1β, which were attenuated by luteolin. Luteolin exerted anticonvulsant and neuroprotective activities in the PTZ-induced epileptic model. Its mechanism was associated with the inhibition of the TLR4/IκBα/NF-κB pathway, alleviating the immune-inflammatory response in the post-epileptic hippocampus.
Collapse
Affiliation(s)
- Yahong Cheng
- College of Medicine and Health Science, Wuhan Polytechnic University, No.68, Xuefu South Road, Wuhan, Hubei 430023, PR China
| | - Yiyuan Zhang
- Wuhan University School of Pharmaceutical Sciences, Wuhan University, No.185 Donghu Road, Wuhan, Hubei 430072, PR China
| | - Puxin Huang
- Wuhan University School of Pharmaceutical Sciences, Wuhan University, No.185 Donghu Road, Wuhan, Hubei 430072, PR China
| | - Qingzhou Cheng
- College of Medicine and Health Science, Wuhan Polytechnic University, No.68, Xuefu South Road, Wuhan, Hubei 430023, PR China.
| | - Hong Ding
- Wuhan University School of Pharmaceutical Sciences, Wuhan University, No.185 Donghu Road, Wuhan, Hubei 430072, PR China.
| |
Collapse
|
19
|
Ji-Eun K, Paras Man P, Jang S, Yi HK. Anti-inflammatory effect of luteoloside against methylglyoxal induced human dental pulp cells. J Appl Biomed 2024; 22:33-39. [PMID: 38505968 DOI: 10.32725/jab.2024.002] [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: 07/14/2023] [Accepted: 02/12/2024] [Indexed: 03/21/2024] Open
Abstract
PURPOSE The aim of this study was to investigate whether luteoloside, a flavonoid, could protect human dental pulp cells (HDPCs) against inflammation and oxidative stress induced by methylglyoxal (MGO), one of the advanced glycated end products (AGE) substances. METHODS HDPCs were stimulated with MGO and treated with luteoloside. MTT assay was used to determine cell viability. Protein expression was measured via western blotting. Reactive oxygen species (ROS) were measured with a Muse Cell Analyzer. Alkaline phosphatase activity (ALP) and Alizarin red staining were used for mineralization assay. RESULTS Luteoloside down-regulated the expression of inflammatory molecules such as ICAM-1, VCAM-1, TNF-α, IL-1β, MMP-2, MMP-9, and COX-2 in MGO-induced HDPCs without showing any cytotoxicity. It attenuated ROS formation and enhanced osteogenic differentiation such as ALP activity and Alizarin red staining in MGO-induced HDPCs. Overall, luteoloside showed protective actions against inflammation and oxidative stress in HDPCs induced by MGO through its anti-inflammatory, anti-oxidative, and osteogenic activities by down-regulating p-JNK in the MAPK pathway. CONCLUSION These results suggest that luteoloside might be a potential adjunctive therapeutic agent for treating pulpal pathological conditions in patients with diabetes mellitus.
Collapse
Affiliation(s)
| | | | - Sungil Jang
- Jeonbuk National University, School of Dentistry, Institute of Oral Bioscience, Departments of Oral Biochemistry, Jeonju, Korea
| | | |
Collapse
|
20
|
Zhou J, Sun F, Zhang W, Feng Z, Yang Y, Mei Z. Novel insight into the therapeutical potential of flavonoids from traditional Chinese medicine against cerebral ischemia/reperfusion injury. Front Pharmacol 2024; 15:1352760. [PMID: 38487170 PMCID: PMC10937431 DOI: 10.3389/fphar.2024.1352760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Cerebral ischemia/reperfusion injury (CIRI) is a major contributor to poor prognosis of ischemic stroke. Flavonoids are a broad family of plant polyphenols which are abundant in traditional Chinese medicine (TCM) and have beneficial effects on several diseases including ischemic stroke. Accumulating studies have indicated that flavonoids derived from herbal TCM are effective in alleviating CIRI after ischemic stroke in vitro or in vivo, and exhibit favourable therapeutical potential. Herein, we systematically review the classification, metabolic absorption, neuroprotective efficacy, and mechanisms of TCM flavonoids against CIRI. The literature suggest that flavonoids exert potential medicinal functions including suppressing excitotoxicity, Ca2+ overloading, oxidative stress, inflammation, thrombin's cellular toxicity, different types of programmed cell deaths, and protecting the blood-brain barrier, as well as promoting neurogenesis in the recovery stage following ischemic stroke. Furthermore, we identified certain matters that should be taken into account in future research, as well as proposed difficulties and opportunities in transforming TCM-derived flavonoids into medications or functional foods for the treatment or prevention of CIRI. Overall, in this review we aim to provide novel ideas for the identification of new prospective medication candidates for the therapeutic strategy against ischemic stroke.
Collapse
Affiliation(s)
- Jing Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Feiyue Sun
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhitao Feng
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Yi Yang
- The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
21
|
Monsalvo-Maraver LA, Ovalle-Noguez EA, Nava-Osorio J, Maya-López M, Rangel-López E, Túnez I, Tinkov AA, Tizabi Y, Aschner M, Santamaría A. Interactions Between the Ubiquitin-Proteasome System, Nrf2, and the Cannabinoidome as Protective Strategies to Combat Neurodegeneration: Review on Experimental Evidence. Neurotox Res 2024; 42:18. [PMID: 38393521 PMCID: PMC10891226 DOI: 10.1007/s12640-024-00694-3] [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: 11/19/2023] [Revised: 01/13/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024]
Abstract
Neurodegenerative disorders are chronic brain diseases that affect humans worldwide. Although many different factors are thought to be involved in the pathogenesis of these disorders, alterations in several key elements such as the ubiquitin-proteasome system (UPS), the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, and the endocannabinoid system (ECS or endocannabinoidome) have been implicated in their etiology. Impairment of these elements has been linked to the origin and progression of neurodegenerative disorders, while their potentiation is thought to promote neuronal survival and overall neuroprotection, as proved with several experimental models. These key neuroprotective pathways can interact and indirectly activate each other. In this review, we summarize the neuroprotective potential of the UPS, ECS, and Nrf2 signaling, both separately and combined, pinpointing their role as a potential therapeutic approach against several hallmarks of neurodegeneration.
Collapse
Affiliation(s)
- Luis Angel Monsalvo-Maraver
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico.
| | - Enid A Ovalle-Noguez
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
| | - Jade Nava-Osorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
| | - Marisol Maya-López
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
- Doctorado en Ciencias Biológicas y de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Edgar Rangel-López
- Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Isaac Túnez
- Instituto de Investigaciones Biomédicas Maimonides de Córdoba (IMIBIC), Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Red Española de Excelencia en Estimulación Cerebral (REDESTIM), Córdoba, Spain
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Yaroslavl State University, Yaroslavl, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abel Santamaría
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
22
|
Chen J, Wang Y, Li M, Zhu X, Liu Z, Chen Q, Xiong K. Netrin-1 Alleviates Early Brain Injury by Regulating Ferroptosis via the PPARγ/Nrf2/GPX4 Signaling Pathway Following Subarachnoid Hemorrhage. Transl Stroke Res 2024; 15:219-237. [PMID: 36631632 DOI: 10.1007/s12975-022-01122-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023]
Abstract
Subarachnoid hemorrhage (SAH) is a type of stroke with high morbidity and mortality. Netrin-1 (NTN-1) can alleviate early brain injury (EBI) following SAH by enhancing peroxisome proliferator-activated receptor gamma (PPARγ), which is an important transcriptional factor modulating lipid metabolism. Ferroptosis is a newly discovered type of cell death related to lipid metabolism. However, the specific function of ferroptosis in NTN-1-mediated neuroprotection following SAH is still unclear. This study aimed to evaluate the neuroprotective effects and the possible molecular basis of NTN-1 in SAH-induced EBI by modulating neuronal ferroptosis using the filament perforations model of SAH in mice and the hemin-stimulated neuron injury model in HT22 cells. NTN-1 or a vehicle was administered 2 h following SAH. We examined neuronal death, brain water content, neurological score, and mortality. NTN-1 treatment led to elevated survival probability, greater survival of neurons, and increased neurological score, indicating that NTN-1-inhibited ferroptosis ameliorated neuron death in vivo/in vitro in response to SAH. Furthermore, NTN-1 treatment enhanced the expression of PPARγ, nuclear factor erythroid 2-related factor 2 (Nrf2), and glutathione peroxidase 4 (GPX4), which are essential regulators of ferroptosis in EBI after SAH. The findings show that NTN-1 improves neurological outcomes in mice and protects neurons from death caused by neuronal ferroptosis. Furthermore, the mechanism underlying NTN-1 neuroprotection is correlated with the inhibition of ferroptosis, attenuating cell death via the PPARγ/Nrf2/GPX4 pathway and coenzyme Q10-ferroptosis suppressor protein 1 (CoQ10-FSP1) pathway.
Collapse
Affiliation(s)
- Junhui Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China
- Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, China
| | - Yuhai Wang
- Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China
| | - Xun Zhu
- Department of Neurosurgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhuanghua Liu
- Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, 214044, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China.
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
23
|
Li W, Wang X, Chen Y, Ding Y, Ling X, Yuan B, Tao J. Luteolin-7-O-glucoside promotes macrophage release of IFN-β by maintaining mitochondrial function and corrects the disorder of glucose metabolism during RSV infection. Eur J Pharmacol 2024; 963:176271. [PMID: 38113965 DOI: 10.1016/j.ejphar.2023.176271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/26/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
Respiratory syncytial virus (RSV) pneumonia is the main cause of acute bronchiolitis in infants. Luteolin-7-O-glucoside (LUT-7G) is a natural flavonoid, which exists in a variety of plants and has the potential to treat viral pneumonia. We established RSV pneumonia mouse models and RSV-infected cell models. Clodronate liposomes were used to deplete macrophages. We used HE staining and immunohistochemistry to determine inflammatory damage and virus replication. We detected the expression levels of inflammatory factors and IFN-β through qPCR and ELISA. JC-1 kit was used for detecting the cell mitochondrial Membrane potential (MMP). ROS, SOD, and MDA kits were used for detecting intracellular oxidative stress damage. Metabolites of TCA in lung tissue and serum of mice were detected by GC-MS. Pharmacodynamic studies have shown that intervention with LUT-7G can alleviate lung tissue damage caused by RSV infection, inhibit RSV replication, and downregulate TNF-α, IL-1β, and IL-6 mRNA expression. LUT-7G upregulated the IFN-β content and the expression of IFN-β, ISG15, and OAS1 mRNA. In vitro, LUT-7G inhibited RSV-induced cell death, reversed the RSV-induced decrease of MMP and decreased intracellular oxidative stress. Target metabonomics showed that RSV infection upregulated the levels of glycolysis and TCA metabolites in lung tissue and serum, while LUT-7G could improve the disorder of glucose metabolism. The results indicate that LUT-7G can promote the release of IFN-β in the lung, alleviate inflammatory damage, and inhibit RSV replication during RSV infection. These effects may be achieved by protecting the mitochondrial function of alveolar macrophages and correcting the disorder of glucose metabolism.
Collapse
Affiliation(s)
- Weifeng Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China; Jiangsu Key Laboratory of Paediatric Respiratory Disease, Institute of Paediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xuan Wang
- Jiangsu Vocational College of Medicine, Yancheng, 224000, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yanzhen Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Oncology Department, Nanjing, 210023, China.
| | - Yali Ding
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China; Jiangsu Key Laboratory of Paediatric Respiratory Disease, Institute of Paediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaoyin Ling
- Affiliated Hospital of Nantong University, Nantong, 226000, China.
| | - Bin Yuan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China.
| | - Jialei Tao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China.
| |
Collapse
|
24
|
Titus C, Hoque MT, Bendayan R. PPAR agonists for the treatment of neuroinflammatory diseases. Trends Pharmacol Sci 2024; 45:9-23. [PMID: 38065777 DOI: 10.1016/j.tips.2023.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 01/07/2024]
Abstract
Peroxisome proliferator-activated receptors [PPARs; PPARα, PPARβ/δ (also known as PPARδ), and PPARγ] widely recognized for their important role in glucose/lipid homeostasis, have recently received significant attention due to their additional anti-inflammatory and neuroprotective effects. Several newly developed PPAR agonists have shown high selectivity for specific PPAR isoforms in vitro and in vivo, offering the potential to achieve desired therapeutic outcomes while reducing the risk of adverse effects. In this review, we discuss the latest preclinical and clinical studies of the activation of PPARs by synthetic, natural, and isoform-specific (full, partial, and dual) agonists for the treatment of neuroinflammatory diseases, including HIV-associated neurocognitive disorders (HAND), Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and cerebral ischemia.
Collapse
Affiliation(s)
- Celene Titus
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON M5S 3M2, Canada
| | - Md Tozammel Hoque
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON M5S 3M2, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON M5S 3M2, Canada.
| |
Collapse
|
25
|
Xu G, Liu G, Wang Z, Li Y, Fang W. Circular RNAs: Promising Treatment Targets and Biomarkers of Ischemic Stroke. Int J Mol Sci 2023; 25:178. [PMID: 38203348 PMCID: PMC10779226 DOI: 10.3390/ijms25010178] [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: 11/01/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Ischemic stroke is one of the most significant causes of morbidity and mortality worldwide. However, there is a dearth of effective drugs and treatment methods for ischemic stroke. Significant numbers of circular RNAs (circRNAs) exhibit abnormal expression following ischemic stroke and are considered potential therapeutic targets. CircRNAs have emerged as promising biomarkers due to their stable expression in peripheral blood and their potential significance in ischemic stroke diagnosis and prognosis. This review provides a summary of 31 circRNAs involved in the pathophysiological processes of apoptosis, autophagy, inflammation, oxidative stress, and angiogenesis following ischemic stroke. Furthermore, we discuss the mechanisms of action of said circRNAs and their potential clinical applications. Ultimately, circRNAs exhibit promise as both therapeutic targets and biomarkers for ischemic stroke.
Collapse
Affiliation(s)
| | | | | | - Yunman Li
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (G.X.); (G.L.); (Z.W.)
| | - Weirong Fang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (G.X.); (G.L.); (Z.W.)
| |
Collapse
|
26
|
Xiong L, Huang W, Liu Y, Zhao H, Wang Y, Jin Y, Zhang L, Zhang Y. Study on Antipyretic Properties of Phenolics in Lonicerae Japonicae Flos Based on Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectrometry Combined with Network Pharmacology. J Food Biochem 2023; 2023:1-17. [DOI: 10.1155/2023/8883860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Objective. To identify and quantify the active phenolic components in Lonicerae japonicae flos (LJF) for fever treatment and their mechanism of action using network pharmacology and molecular docking. Methods. Based on qualitative analysis of LJF, 194 phenolics were obtained, including 81 phenolic acids and 113 flavonoids. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were used to identify potential targets for these components to interact with fever. Molecular docking with microsomal PGE2 synthase-1, EP1, EP2, EP3, and EP4 targets was used to determine antipyretic components. The antipyretic efficacy of the main components was verified by in vivo experiments. Finally, high-performance liquid chromatography-tandem mass spectrometry was used to quantify the main antipyretic components of LJF. Results. Phenolics in LJF may prevent and treat fever by participating in calcium signaling, regulating TRP channels, and cAMP signaling. Luteolin-7-O-glucoside, apigenin-7-O-glucoside, 3,5-O-dicaffeoylquinic acid, luteolin, and other components have a good docking effect with PGE2 synthase-1 and its four subtypes. 3,5-O-dicaffeoylquinic acid, luteolin-7-O-glucoside, and apigenin-7-O-glucoside have good antipyretic effects in a yeast-induced pyrexia model. The content of these antipyretic components varies with the developmental period of LJF. Phenolic acids are the main components that distinguish the different developmental periods of LJF. Conclusion. The potential antipyretic components and molecular mechanisms of phenolics provide a basis for the traditional medicinal effects and future development and utilization of LJF.
Collapse
Affiliation(s)
- Lewen Xiong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wenjing Huang
- Weifang Traditional Chinese Medicine Hospital, Weifang, China
| | - Yan Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hongwei Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yang Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ying Jin
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Longfei Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yongqing Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
27
|
Liu H, Zhang J, Yan X, An D, Lei H. The Anti-atherosclerosis Mechanism of Ziziphora clinopodioides Lam. Based On Network Pharmacology. Cell Biochem Biophys 2023; 81:515-532. [PMID: 37523140 DOI: 10.1007/s12013-023-01151-2] [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] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
We investigated the mechanisms underlying the effects of Ziziphora clinopodioides Lam. (ZCL) on atherosclerosis (AS) using network pharmacology and in vitro validation.We collected the active components of ZCL and predicted their targets in AS. We constructed the protein-protein interaction, compound-target, and target-compound-pathway networks, and performed GO and KEGG analyses. Molecular docking of the active components and key targets was constructed with Autodock and Pymol software. Validation was performed with qRT-PCR, ELISA, and Western blot.We obtained 80 components of ZCL. The network analysis identified that 14 components and 37 genes were involved in AS. Then, 10 key nodes in the PPI network were identified as the key targets of ZCL because of their importance in network topology. The binding energy of 8 components (Cynaroside, α-Spinasterol, Linarin, Kaempferide, Acacetin, Genkwanin, Chrysin, and Apiin) to 4 targets (MMP9, TP53, AKT1, SRC) was strong and <-1 kJ/mol. In addition, 13 of the 14 components were flavonoids and thus total flavonoids of Ziziphora clinopodioides Lam. (ZCF) were used for in vitro validation. We found that ZCF reduced eNOS, P22phox, gp91phox, and PCSK9 at mRNA and protein levels, as well as the levels of IL-1β, TNF-α, and IL-6 proteins in vitro (P < 0.05).We successfully predicted the active components, targets, and mechanisms of ZCL in treating AS using network pharmacology. We confirmed that ZCF may play a role in AS by modulating oxidative stress, lipid metabolism, and inflammatory response via Cynaroside, Linarin, Kaempferide, Acacetin, Genkwanin, Chrysin, and Apiin.
Collapse
Affiliation(s)
- Hongbing Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, 102488, Beijing, China
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China
| | - Jianxin Zhang
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China
| | - Xuehua Yan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, 102488, Beijing, China
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China
| | - Dongqing An
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China.
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, 102488, Beijing, China.
| |
Collapse
|
28
|
Hong WM, Xie YW, Zhao MY, Yu TH, Wang LN, Xu WY, Gao S, Cai HB, Guo Y, Zhang F. Vasoprotective Effects of Hyperoside against Cerebral Ischemia/Reperfusion Injury in Rats: Activation of Large-Conductance Ca 2+-Activated K + Channels. Neural Plast 2023; 2023:5545205. [PMID: 37609123 PMCID: PMC10442186 DOI: 10.1155/2023/5545205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/29/2023] [Accepted: 07/19/2023] [Indexed: 08/24/2023] Open
Abstract
Hyperoside (Hyp), a kind of Chinese herbal medicine, exerts multiple therapeutic effects on many diseases. However, the role and mechanisms of Hyp in vascular pathophysiology in ischemic stroke need to be further established. The study aimed to investigate the role of (large-conductance Ca2+-activated K+) BK channels on the vasoprotection of Hyp against cerebral ischemia and reperfusion (I/R) injury in rats. The concentration gradient of Hyp was pretreated in both the middle cerebral artery occlusion and reperfusion model and oxygen-glucose deprivation/reoxygenation (OGD/R) model of primary vascular smooth muscle cells (VSMCs) in rats. A series of indicators were detected, including neurological deficit score, infarct volume, malondialdehyde (MDA), superoxide dismutase (SOD), cerebral blood flow (CBF), cell viability, membrane potential, and BK channels α- and β1-subunits expression. The results showed that Hyp significantly reduced infarct volume and ameliorated neurological dysfunction in I/R-injured rats. Besides, the effects of I/R-induced reduction of BK channels α- and β1-subunits expression were significantly reversed by Hyp in endothelial-denudated cerebral basilar arteries. Furthermore, the protective effect against I/R-induced increases of MDA and reduction of SOD as well as CBF induced by Hyp was significantly reversed by iberiotoxin (IbTX). In OGD/R-injured VSMCs, downregulated cellular viability and BK channels β1-subunits expression were remarkably reversed by Hyp. However, neither OGD/R nor Hyp affected BK channels α-subunits expression, and Hyp failed to induced hyperpolarization of VSMCs. Moreover, the protective effect against OGD/R-induced reduction of cell viability and SOD level and increases of MDA production induced by Hyp was significantly reversed by IbTX in VSMCs. The study indicates that Hyp has the therapeutic potential to improve vascular outcomes, and the mechanism is associated with suppressing oxidative stress and improving CBF through upregulating BK channels.
Collapse
Affiliation(s)
- Wen-Ming Hong
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- School of Nursing, Anhui Medical University, Hefei 230032, China
- Open Project of Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Yue-Wu Xie
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Meng-Yu Zhao
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Tian-Hang Yu
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Li-Na Wang
- School of Nursing, Anhui Medical University, Hefei 230032, China
| | - Wan-Yan Xu
- School of Nursing, Anhui Medical University, Hefei 230032, China
| | - Shen Gao
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Hua-Bao Cai
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yan Guo
- Department of Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Fang Zhang
- School of Nursing, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
29
|
Chen X, Shi W, Xie Y, Wang Y, Yao Q, Ke H, Xu X, Liu H, Liu P, Zhou X. Hepatic Krüppel-like factor 14 regulates lipid metabolism in nonalcoholic steatohepatitis mice. FASEB J 2023; 37:e23070. [PMID: 37389939 DOI: 10.1096/fj.202300448r] [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: 03/09/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
Excessive lipid accumulation is a critical characteristic in the development of nonalcoholic steatohepatitis (NASH). The underlying molecular mechanism, however, is unclear. In this study, we explored whether and how Krüppel-like factor 14 (KLF14) affects hepatic lipid metabolism in NASH. KLF14 expression was detected in NASH patients and mice fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD). Adeno-associated viruses and adenoviruses were used to alter hepatic KLF14 expression in vivo or in vitro to investigate how KLF14 functions in lipid regulation. The molecular mechanisms were explored using RNA-seq, luciferase reporter, and ChIP assays. The fatty liver phenotype was analyzed histopathologically, and serum and hepatocyte biochemical parameters were measured. The NASH mouse model developed quickly in C57BL/6J mice fed a CDAHFD for 8 weeks. We found that KLF14 expression was decreased in NASH patients and CDAHFD mice. Oleic acid and palmitic acid treatment also reduced KLF14 levels in hepatocytes. KLF14 knockdown downregulated the genes involved in fatty acid oxidation, promoting the progression of hepatic steatosis. In contrast, hepatic KLF14 overexpression alleviated lipid accumulation and oxidative stress in CDAHFD mice. These effects resulted from direct activation of the PPARα signaling pathway. PPARα inhibition diminished the KLF14 overexpression-reduced protective effects against steatosis in OA&PA-treated MPHs and AAV-KLF14-infected CDAHFD mice. These data reveal that hepatic KLF14 regulates lipid accumulation and oxidative stress through the KLF14-PPARα pathway as NASH progresses. KLF14 may be a novel therapeutic target for hepatic steatosis.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Wenjie Shi
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Yunwu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qian Yao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Huajing Ke
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Hui Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Pi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The People's Hospital of Longhua, Shenzhen, China
| | - Xiaojiang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Zhao X, Tian Z, Sun M, Dong D. Nrf2: a dark horse in doxorubicin-induced cardiotoxicity. Cell Death Discov 2023; 9:261. [PMID: 37495572 PMCID: PMC10372151 DOI: 10.1038/s41420-023-01565-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Being a broad-spectrum anticancer drug, doxorubicin is indispensable for clinical treatment. Unexpectedly, its cardiotoxic side effects have proven to be a formidable obstacle. Numerous studies are currently devoted to elucidating the pathological mechanisms underlying doxorubicin-induced cardiotoxicity. Nrf2 has always played a crucial role in oxidative stress, but numerous studies have demonstrated that it also plays a vital part in pathological mechanisms like cell death and inflammation. Numerous studies on the pathological mechanisms associated with doxorubicin-induced cardiotoxicity demonstrate this. Several clinical drugs, natural and synthetic compounds, as well as small molecule RNAs have been demonstrated to prevent doxorubicin-induced cardiotoxicity by activating Nrf2. Consequently, this study emphasizes the introduction of Nrf2, discusses the role of Nrf2 in doxorubicin-induced cardiotoxicity, and concludes with a summary of the therapeutic modalities targeting Nrf2 to ameliorate doxorubicin-induced cardiotoxicity, highlighting the potential value of Nrf2 in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaopeng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, 110102, China
| | - Zheng Tian
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, 110102, China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, 110102, China.
| | - Dan Dong
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
31
|
Pluta R, Miziak B, Czuczwar SJ. Apitherapy in Post-Ischemic Brain Neurodegeneration of Alzheimer's Disease Proteinopathy: Focus on Honey and Its Flavonoids and Phenolic Acids. Molecules 2023; 28:5624. [PMID: 37570596 PMCID: PMC10420307 DOI: 10.3390/molecules28155624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Neurodegeneration of the brain after ischemia is a major cause of severe, long-term disability, dementia, and mortality, which is a global problem. These phenomena are attributed to excitotoxicity, changes in the blood-brain barrier, neuroinflammation, oxidative stress, vasoconstriction, cerebral amyloid angiopathy, amyloid plaques, neurofibrillary tangles, and ultimately neuronal death. In addition, genetic factors such as post-ischemic changes in genetic programming in the expression of amyloid protein precursor, β-secretase, presenilin-1 and -2, and tau protein play an important role in the irreversible progression of post-ischemic neurodegeneration. Since current treatment is aimed at preventing symptoms such as dementia and disability, the search for causative therapy that would be helpful in preventing and treating post-ischemic neurodegeneration of Alzheimer's disease proteinopathy is ongoing. Numerous studies have shown that the high contents of flavonoids and phenolic acids in honey have antioxidant, anti-inflammatory, anti-apoptotic, anti-amyloid, anti-tau protein, anticholinesterase, serotonergic, and AMPAK activities, influencing signal transmission and neuroprotective effects. Notably, in many preclinical studies, flavonoids and phenolic acids, the main components of honey, were also effective when administered after ischemia, suggesting their possible use in promoting recovery in stroke patients. This review provides new insight into honey's potential to prevent brain ischemia as well as to ameliorate damage in advanced post-ischemic brain neurodegeneration.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (S.J.C.)
| | | | | |
Collapse
|
32
|
Baicalein ameliorates Alzheimer's disease via orchestration of CX3CR1/NF-κB pathway in a triple transgenic mouse model. Int Immunopharmacol 2023; 118:109994. [PMID: 37098656 DOI: 10.1016/j.intimp.2023.109994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/15/2023]
Abstract
Alzheimer's disease (AD) is a common chronic neurodegenerative disease. Some studies have suggested that dysregulation of microglia activation and the resulting neuroinflammation play an important role in the development of AD pathology. Activated microglia have both M1 and M2 phenotypes and inhibition of M1 phenotype while stimulating M2 phenotype has been considered as a potential treatment for neuroinflammation-related diseases. Baicalein is a class of flavonoids with anti-inflammatory, antioxidant and other biological activities, but its role in AD and the regulation of microglia are limited. The purpose of this study was to investigate the effect of baicalein on the activation of microglia in AD model mice and the related molecular mechanism. Our results showed that baicalein significantly improved the learning and memory ability and AD-related pathology of 3 × Tg-AD mice, inhibited the level of pro-inflammatory factors TNF-α, IL-1β and IL-6, promoted the production of anti-inflammatory factors IL-4 and IL-10, and regulated the microglia phenotype through CX3CR1/NF-κB signaling pathway. In conclusion, baicalein can regulate the phenotypic transformation of activated microglia and reduce neuroinflammation through CX3CR1/NF-κB pathway, thereby improving the learning and memory ability of 3 × Tg-AD mice.
Collapse
|
33
|
El-Sayed EK, Ibrahim RR, Ahmed AA, Khattab MA, Chen LY, Lai KH, Shaarawy FSE, Tawfik NF, Moharram FA. Quercus coccinea Münchh leaves polyphenols: Appraisal acute lung injury induced by lipopolysaccharide in mice. Biomed Pharmacother 2023:114765. [PMID: 37246132 DOI: 10.1016/j.biopha.2023.114765] [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: 04/04/2023] [Accepted: 04/21/2023] [Indexed: 05/30/2023] Open
Abstract
Genus Quercus is a well-known source for its polyphenolic content and important biological activity. Plants belonging to the Quercus genus were traditionally used in asthma, inflammatory diseases, wound healing, acute diarrhea, and hemorrhoid. Our work intended to study the polyphenolic profile of the Q. coccinea (QC) leaves and to assess the protective activity of its 80% aqueous methanol extract (AME) against lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Together, the potential molecular mechanism was investigated. Nineteen polyphenolic compounds (1-18), including tannins, flavone, and flavonol glycosides. Phenolic acids and aglycones were purified and identified from the AME of QC leaves. Treatment with AME of QC showed an anti-inflammatory effect evidenced by a remarkable decline in the count of white blood cells and neutrophils which was in harmony with decreasing the levels of high mobility group box-1, nuclear factor kappa B, tumor necrosis factor-α, and interleukin 1 beta. In addition, the antioxidant activity of QC was documented through the significant reduction in malondialdehyde level and elevation of reduced glutathione level and superoxide dismutase activity. Furthermore, the mechanism involved in the pulmonary protective effect of QC involved the downregulation of the TLR4/MyD88 pathway. The AME of QC showed a protective effect against LPS-induced ALI through the powerful anti-inflammatory and antioxidant activities which are linked to its abundancy with polyphenols.
Collapse
Affiliation(s)
- Elsayed K El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt
| | - Reham R Ibrahim
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Asmaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt
| | - Mohamed A Khattab
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lo-Yun Chen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Fatheya S El Shaarawy
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Nashwa F Tawfik
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Fatma A Moharram
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
34
|
Han C, Guan L, Xu L. Protective effect of luteoloside against Toxoplasma gondii-induced liver injury through inhibiting TLR4/NF-κB and P2X7R/NLRP3 and enhancing Nrf2/HO-1 signaling pathway. Parasitol Res 2023; 122:1333-1342. [PMID: 37046028 DOI: 10.1007/s00436-023-07833-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Toxoplasma gondii (T. gondii) infection can cause liver injury by inducing inflammation and oxidative stress. The Chinese herbal extract luteoloside (Lut) has considerable anti-inflammatory and antioxidant properties, but its effects on the liver injury during T. gondii infection have not been reported. This study investigated the hepatoprotective effects of Lut by treating T. gondii-infected mice with 0-200 mg/kg doses of Lut and further examined the expression of key proteins in the inflammation and oxidative stress-related pathways in the liver to investigate the potential mechanism of the hepatoprotective effects of Lut. Results showed that Lut remarkably reduced serum ALT and AST levels, considerably decreased inflammatory factors TNF-α, IL-6, and IL-1β, as well as oxidative products MDA, and greatly increased antioxidant enzymes SOD and GSH. The expression of key proteins TLR4, Myd88, TRAF6, p-NF-κB p65 in the TLR4/NF-κB pathway and P2X7R, NLRP3, caspase 1, IL-1β, IL-18 in the P2X7R/NLRP3 pathway were significantly decreased in the liver. And the expression of key proteins Nrf2, HO-1, NQO-1, and GCLC in the Nrf2/HO-1 antioxidant-related pathway was significantly upregulated. In conclusion, Lut attenuated T. gondii-induced liver injury by inhibiting the inflammatory response and enhancing antioxidant capacity. The hepatoprotective mechanisms of Lut are involved in inhibiting TLR4/NF-κB and P2X7R/NLRP3 inflammatory signaling pathways, as well as enhancing the Nrf2/HO-1 antioxidant pathway. These findings not only provide some reference for further exploring the specific hepatoprotective mechanism of Lut during T. gondii infection, but also provide some theoretical basis for the future clinical application of Lut as a hepatoprotective drug in T. gondii infection.
Collapse
Affiliation(s)
- Chengquan Han
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China
| | - Lizeng Guan
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China
| | - Lu Xu
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China.
| |
Collapse
|
35
|
Sun S, Yu A, Cheng R, Wang L, He T, Xu X, Song R, Shan D, Lv F, Zhong X, Deng Q, Li X, He Y, Zheng Y, Ren X, Xia Q, She G. Similarities and differences between Ziqin and Kuqin in anti-inflammatory, analgesic, and antioxidant activities and their core chemical composition based on the zebrafish model and spectrum-effect relationship. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116049. [PMID: 36529251 DOI: 10.1016/j.jep.2022.116049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/16/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria baicalensis (SB) is a traditional Chinese medicine (TCM). In the clinical application of TCM, SB has been divided into two specifications (Ziqin and Kuqin) for a long time. At present, the Chinese Pharmacopoeia Commission no longer distinguishes between the two. However, the two specifications of medicinal materials and pieces are still in circulation in the market. AIM OF THE STUDY This work aimed at investigating the similarities and differences between Ziqin and Kuqin in anti-inflammatory, analgesic, and antioxidant activities and their material basis. It will provide a new angle for relevant regulations to formulate the specifications and standards of SB. MATERIALS AND METHODS Here we investigated the similarities and differences between Ziqin and Kuqin in anti-inflammatory, analgesic, and antioxidant activities related to four zebrafish models and three chemical tests. The chemical fingerprints of SB (Ziqin and Kuqin) were profiled by HPLC. Meanwhile, UHPLC-Q-TOF/MS was used to identify the chemical constituents of Ziqin and Kuqin. The main effect-related compounds of SB, Ziqin, and Kuqin were screened out by spectrum-effect relationship. Finally, six monomeric compounds were validated experimentally using the zebrafish inflammation model induced by CuSO4. RESULTS Both Ziqin and Kuqin had significant anti-inflammatory, analgesic, and antioxidant activities. Kuqin had better anti-inflammatory and analgesic activities, while Ziqin had better antioxidant activity. HPLC fingerprint and UHPLC-Q-TOF/MS evaluation showed that the chemical composition types and main components of Ziqin and Kuqin were basically the same, while the contents and proportions of chemical components in Ziqin and Kuqin were different. By spectrum-effect relationship, compounds X1, X2 (luteoloside), X3, X4 (baicalin), X6 (wogonoside), X7 (baicalein), X8 (wogonin), and X9 (oroxylin A) were the same active chemical constituents of Ziqin and Kuqin. The core components of anti-inflammatory and analgesia activities in Kuqin were compounds X1, X2, X3, X5, X6, X7, X8, and X9. The antioxidant core active components of Ziqin were compounds X2, X3, X4, X6, X7, and X9. Among them, luteoloside, baicalin, wogonoside, baicalein, wogonin, and oroxylin A were validated successfully with good anti-inflammatory effects. CONCLUSIONS This study revealed that Ziqin and kuqin have high similarity in chemical composition, but their proportions and active core components are different. This may be one of the main reasons why they have the same activity but different activity trends. These findings will help to improve the understanding of the different clinical applications of Ziqin and Kuqin, and provide a reference for the formulation of quality standards and their further research.
Collapse
Affiliation(s)
- Siqi Sun
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China; Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, 250000, PR China.
| | - Axiang Yu
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Ruiyang Cheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 102488, PR China.
| | - Le Wang
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Ting He
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Xiao Xu
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Ruolan Song
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Dongjie Shan
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Fang Lv
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Xiangjian Zhong
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Qingyue Deng
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Xianxian Li
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Yingyu He
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Yuan Zheng
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Xueyang Ren
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, 250000, PR China.
| | - Gaimei She
- School of ChineseMateria Medica, Beijing University of Chinese Medicine, Beijing, 102488, PR China.
| |
Collapse
|
36
|
Anti-Inflammatory Effects of Flavonoids in Common Neurological Disorders Associated with Aging. Int J Mol Sci 2023; 24:ijms24054297. [PMID: 36901731 PMCID: PMC10001833 DOI: 10.3390/ijms24054297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Aging reduces homeostasis and contributes to increasing the risk of brain diseases and death. Some of the principal characteristics are chronic and low-grade inflammation, a general increase in the secretion of proinflammatory cytokines, and inflammatory markers. Aging-related diseases include focal ischemic stroke and neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). Flavonoids are the most common class of polyphenols and are abundantly found in plant-based foods and beverages. A small group of individual flavonoid molecules (e.g., quercetin, epigallocatechin-3-gallate, and myricetin) has been used to explore the anti-inflammatory effect in vitro studies and in animal models of focal ischemic stroke and AD and PD, and the results show that these molecules reduce the activated neuroglia and several proinflammatory cytokines, and also, inactivate inflammation and inflammasome-related transcription factors. However, the evidence from human studies has been limited. In this review article, we highlight the evidence that individual natural molecules can modulate neuroinflammation in diverse studies from in vitro to animal models to clinical studies of focal ischemic stroke and AD and PD, and we discuss future areas of research that can help researchers to develop new therapeutic agents.
Collapse
|
37
|
Luteoloside Induces G0/G1 Phase Arrest of Neuroblastoma Cells by Targeting p38 MAPK. Molecules 2023; 28:molecules28041748. [PMID: 36838737 PMCID: PMC9966487 DOI: 10.3390/molecules28041748] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Luteoloside has shown anti-inflammatory, antiviral, and antitumor properties. However, the effect and mechanism of luteoloside on neuroblastoma cells remain unknown. The proliferation of human neuroblastoma cells (SH-SY5Y and SK-N-AS) treated with different concentrations of luteoloside (0, 12.5, 25, and 50 μM) was detected by the MTT assay and colony formation assay. Cell apoptosis and cell cycle were examined by Hoechst staining and flow cytometry. A subcutaneous tumorigenesis model was established in nude mice to evaluate the effect of luteoloside on tumor growth in vivo. Bioinformatics, molecular docking techniques, and cellular thermal shift assays were utilized to predict the potential targets of luteoloside in neuroblastoma. The p38 MAPK inhibitor SB203580 was used to confirm the role of p38 MAPK. Luteoloside inhibited the proliferation of neuroblastoma cells in vitro and in vivo. Luteoloside slightly induced cellular G0/G1 phase arrest and reduced the expression levels of G0/G1 phase-related genes and the proteins cyclin D1, CDK4, and C-myc, which are downregulated by p38 MAPK pathways. Meanwhile, p38 was identified as the target of luteoloside, and inhibition of p38 MAPK reversed the inhibitory effect of luteoloside on neuroblastoma cells. Luteoloside is a potential anticancer drug for treating neuroblastoma by activating p38 MAPK.
Collapse
|
38
|
Zhang X, Li M, Yue Y, Zhang Y, Wu A. Luteoloside Prevents Sevoflurane-induced Cognitive Dysfunction in Aged Rats via Maintaining Mitochondrial Function and Dynamics in Hippocampal Neurons. Neuroscience 2023; 516:42-53. [PMID: 36764603 DOI: 10.1016/j.neuroscience.2023.01.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/11/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is characterized by impaired cognitive function, such as decreased learning and memory after anesthesia and surgery. This study aimed to explore the effect of luteoloside, a flavonoid extracted from natural herbs, on sevoflurane-induced cognitive dysfunction. Aged Sprague-Dawley male rats (20 months old) were treated with luteoloside for 7 days prior to sevoflurane exposure. After evaluation using an open field, novel object recognition, and Y-maze tests, it was determined that luteoloside effectively prevented sevoflurane-induced cognitive dysfunction. Sevoflurane exposure led to hippocampal neuron apoptosis in vivo (n = 6) and in vitro (n = 3), while this injury was prevented by luteoloside in a dose-dependent manner. Mechanistically, luteoloside maintained mitochondrial function and dynamics, as evidenced by the restored adenosine triphosphate (ATP) production and mitochondrial membrane potential as well as the upregulated levels of mitochondrial fission (optic atrophy protein 1 (Opa1) and mitofusin1 (Mfn1)) and downregulated mitochondrial fusion (mitochondrial fission 1 (Fisl) and dynamin-related protein 1 (Drp1)) factors. Notably, silencing Opa1 blocked the protective effect of luteoloside on hippocampal neurons and mitochondrial function. In summary, luteoloside prevented sevoflurane-induced cognitive dysfunction in aged rats, which may be achieved by regulating mitochondrial dynamics. Our study reveals the potential of luteoloside in preventing POCD in aged patients.
Collapse
Affiliation(s)
- Xuena Zhang
- Department of Anesthesiology Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Mingying Li
- Department of Anesthesiology Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yun Yue
- Department of Anesthesiology Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ying Zhang
- College of Control and Computer Engineering, North China Electric Power University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
39
|
He Z, Li X, Zhang H, Liu X, Han S, Abdurahman A, Shen L, Du X, Li N, Yang X, Liu Q. A novel vanadium complex VO(p-dmada) inhibits neuroinflammation induced by lipopolysaccharide. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
40
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
41
|
Li L, Pan G, Fan R, Li D, Guo L, Ma L, Liang H, Qiu J. Luteolin alleviates inflammation and autophagy of hippocampus induced by cerebral ischemia/reperfusion by activating PPAR gamma in rats. BMC Complement Med Ther 2022; 22:176. [PMID: 35778706 PMCID: PMC9248165 DOI: 10.1186/s12906-022-03652-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Luteolin, a flavonoid compound with anti-inflammatory activity, has been reported to alleviate cerebral ischemia/reperfusion (I/R) injury. However, its potential mechanism remains unclear. Methods The binding activity of luteolin to peroxisome proliferator-activated receptor gamma (PPARγ) was calculated via molecular docking analysis. Rats were subjected to middle cerebral artery occlusion and reperfusion (MCAO/R). After reperfusion, vehicle, 25 mg/kg/d luteolin, 50 mg/kg/d luteolin, 10 mg/kg/d pioglitazone, 50 mg/kg/d luteolin combined with 10 mg/kg/d T0070907 (PPARγ inhibitor) were immediately orally treatment for 7 days. ELISA, TTC staining, H&E staining, immunohistochemistry, immunofluorescence and transmission electron microscope methods were performed to evaluate the inflammation and autophagy in damaged hippocampal region. The PPARγ, light chain 3 (LC3) B-II/LC3B-I and p-nuclear factor-κB (NF-κB) p65 proteins expression levels in damaged hippocampal region were analyzed. Results Luteolin showed good PPARγ activity according to docking score (score = − 8.2). Luteolin treatment downregulated the infarct area and the pro-inflammatory cytokines levels caused by MCAO/R injury. Moreover, luteolin administration ameliorated neuroinflammation and autophagy in damaged hippocampal region. Pioglitazone plays protective roles similar to luteolin. T0070907 concealed the neuroprotective roles of 50 mg/kg/d luteolin. Conclusions Luteolin exerts neuroprotective roles against inflammation and autophagy of hippocampus induced by cerebral I/R by activating PPARγ in rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03652-8.
Collapse
|
42
|
Nrf2 Regulates Oxidative Stress and Its Role in Cerebral Ischemic Stroke. Antioxidants (Basel) 2022; 11:antiox11122377. [PMID: 36552584 PMCID: PMC9774301 DOI: 10.3390/antiox11122377] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
Cerebral ischemic stroke is characterized by acute ischemia in a certain part of the brain, which leads to brain cells necrosis, apoptosis, ferroptosis, pyroptosis, etc. At present, there are limited effective clinical treatments for cerebral ischemic stroke, and the recovery of cerebral blood circulation will lead to cerebral ischemia-reperfusion injury (CIRI). Cerebral ischemic stroke involves many pathological processes such as oxidative stress, inflammation, and mitochondrial dysfunction. Nuclear factor erythroid 2-related factor 2 (Nrf2), as one of the most critical antioxidant transcription factors in cells, can coordinate various cytoprotective factors to inhibit oxidative stress. Targeting Nrf2 is considered as a potential strategy to prevent and treat cerebral ischemia injury. During cerebral ischemia, Nrf2 participates in signaling pathways such as Keap1, PI3K/AKT, MAPK, NF-κB, and HO-1, and then alleviates cerebral ischemia injury or CIRI by inhibiting oxidative stress, anti-inflammation, maintaining mitochondrial homeostasis, protecting the blood-brain barrier, and inhibiting ferroptosis. In this review, we have discussed the structure of Nrf2, the mechanisms of Nrf2 in cerebral ischemic stroke, the related research on the treatment of cerebral ischemia through the Nrf2 signaling pathway in recent years, and expounded the important role and future potential of the Nrf2 pathway in cerebral ischemic stroke.
Collapse
|
43
|
Luteoloside pretreatment attenuates anoxia-induced damage in cardiomyocytes by regulating autophagy mediated by 14-3-3η and the AMPKα-mTOR/ULK1 pathway. Mol Cell Biochem 2022; 478:1475-1486. [DOI: 10.1007/s11010-022-04611-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 11/04/2022] [Indexed: 11/17/2022]
|
44
|
El Gazzar WB, Allam MM, Shaltout SA, Mohammed LA, Sadek AM, Nasr HE. Pioglitazone modulates immune activation and ameliorates inflammation induced by injured renal tubular epithelial cells via PPARγ/miRNA‑124/STAT3 signaling. Biomed Rep 2022; 18:2. [PMID: 36544854 PMCID: PMC9756109 DOI: 10.3892/br.2022.1584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Acute kidney injury (AKI) is commonly a result of renal ischemia reperfusion injury (IRI), which produces clinical complications characterized by the rapid deterioration of renal function, leading to chronic kidney disease and increases the risk of morbidity and mortality. Currently, only supportive treatment is available. AKI, which is accompanied by immune activation and inflammation, is caused by proximal tubular injury. The present study investigated the role of tubular epithelial cells as drivers of inflammation in renal IRI and their potential function as antigen-presenting cells, as well as the molecular mechanisms by which peroxisome proliferator-activated receptor-γ (PPARγ) agonists [such as pioglitazone (Pio)] exert reno-protective action in renal IRI. A total of 50 Wistar male albino rats were divided into five groups: Sham + DMSO, Sham + Pio, IRI + DMSO, IRI + prophylactic preoperative (pre) Pio and IRI + postoperative Pio. The histopathological changes in renal tissue samples and the renal epithelial cell expression of CD86, miRNA-124, STAT3, pro-inflammatory cytokines, inducible nitric oxide synthase (iNOS) and Arginase-II were analyzed by immunohistochemistry, reverse transcription-quantitative PCR, western blotting and ELISA respectively. IRI was a potent inducer for CD86 immunoexpression. An ameliorative action of Pio was demonstrated via decreased CD86 immunoexpression, upregulation of miRNA-124, decreased STAT3 expression and beneficial anti-inflammatory effects. The tubular epithelium served a notable role in the inflammatory response in renal IRI. Pio exerted its anti-inflammatory effects via PPARγ/miRNA-124/STAT3 signaling.
Collapse
Affiliation(s)
- Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt,Correspondence to: Dr Walaa Bayoumie El Gazzar, Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan
| | - Mona Maher Allam
- Department of Physiology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Sherif Ahmed Shaltout
- Department of Pharmacology, Public Health and Clinical Skills, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan,Department of Pharmacology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Lina Abdelhady Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Ashraf Mohamed Sadek
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan,Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Hend Elsayed Nasr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| |
Collapse
|
45
|
Choudhary N, Tewari D, Nabavi SF, Kashani HRK, Lorigooini Z, Filosa R, Khan FB, Masoudian N, Nabavi SM. Plant based food bioactives: A boon or bane for neurological disorders. Crit Rev Food Sci Nutr 2022; 64:3279-3325. [PMID: 36369694 DOI: 10.1080/10408398.2022.2131729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Neurological disorders are the foremost occurring diseases across the globe resulting in progressive dysfunction, loss of neuronal structure ultimately cell death. Therefore, attention has been drawn toward the natural resources for the search of neuroprotective agents. Plant-based food bioactives have emerged as potential neuroprotective agents for the treatment of neurodegenerative disorders. This comprehensive review primarily focuses on various plant food bioactive, mechanisms, therapeutic targets, in vitro and in vivo studies in the treatment of neurological disorders to explore whether they are boon or bane for neurological disorders. In addition, the clinical perspective of plant food bioactives in neurological disorders are also highlighted. Scientific evidences point toward the enormous therapeutic efficacy of plant food bioactives in the prevention or treatment of neurological disorders. Nevertheless, identification of food bioactive components accountable for the neuroprotective effects, mechanism, clinical trials, and consolidation of information flow are warranted. Plant food bioactives primarily act by mediating through various pathways including oxidative stress, neuroinflammation, apoptosis, excitotoxicity, specific proteins, mitochondrial dysfunction, and reversing neurodegeneration and can be used for the prevention and therapy of neurodegenerative disorders. In conclusion, the plant based food bioactives are boon for neurological disorders.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, Adesh Institute of Pharmacy and Biomedical Sciences, Adesh University, Bathinda, Punjab, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Seyed Fazel Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| | - Hamid Reza Khayat Kashani
- Department of Neurosurgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rosanna Filosa
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain, 15551 United Arab Emirates
| | - Nooshin Masoudian
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
| | - Seyed Mohammad Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| |
Collapse
|
46
|
Chen Y, Peng F, Xing Z, Chen J, Peng C, Li D. Beneficial effects of natural flavonoids on neuroinflammation. Front Immunol 2022; 13:1006434. [PMID: 36353622 PMCID: PMC9638012 DOI: 10.3389/fimmu.2022.1006434] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/03/2022] [Indexed: 12/05/2022] Open
Abstract
Neuroinflammation is the fundamental immune response against multiple factors in the central nervous system and is characterized by the production of inflammatory mediators, activated microglia and astrocytes, and the recruitment of innate and adaptive immune cells to inflammatory sites, that contributes to the pathological process of related brain diseases, such as Alzheimer’s disease, Parkinson’s disease, depression, and stroke. Flavonoids, as a species of important natural compounds, have been widely revealed to alleviate neuroinflammation by inhibiting the production of pro-inflammatory mediators, elevating the secretion of anti-inflammatory factors, and modulating the polarization of microglia and astrocyte, mainly via suppressing the activation of NLRP3 inflammasome, as well as NF-κB, MAPK, and JAK/STAT pathways, promoting Nrf2, AMPK, BDNF/CREB, Wnt/β-Catenin, PI3k/Akt signals and SIRT1-mediated HMGB1 deacetylation. This review will provide the latest and comprehensive knowledge on the therapeutic benefits and mechanisms of natural flavonoids in neuroinflammation, and the natural flavonoids might be developed into food supplements or lead compounds for neuroinflammation-associated brain disorders.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Ziwei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junren Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Cheng Peng, ; Dan Li,
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Cheng Peng, ; Dan Li,
| |
Collapse
|
47
|
Yu M, Li Y, Tan X, Hu Q. Steroid Receptor Coactivator-Interacting Protein (SIP) Alleviates Ischemic Cerebral Infarction Damage Through the NF-κB Pathway. Horm Metab Res 2022; 54:704-710. [PMID: 36055280 DOI: 10.1055/a-1913-8088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Ischemic stroke leads to high mortality and disability rates in humans. Cerebral ischemic injury has a severe complex pathophysiological mechanism. The abnormal release of inflammatory cytokines will cause brain tissue damage and destroy the blood-brain barrier integrity, which aggravates the process of brain injury. Therefore, attenuating the level of inflammatory response is critical for the therapy of cerebral ischemia injury. This study examined the rule of SIP treatment to support neuron protective effect after cerebral injury in an animal model of middle cerebral artery occlusion (MCAO). After ischemia/reperfusion, neurological function, neuroglia cells activation, infarction volume, brain water content, brain tissue apoptosis ratio, and inflammatory response were assessed, and quantitative PCR and western blot were also detected, respectively. Treatment of SIP ameliorated neurological dysfunction, brain infarction, brain edema, and brain cell apoptosis after MCAO operation. Overexpression SIP also suppressed pro-inflammatory cytokines release. Furthermore, the protective effect of SIP on brain injury occurs through reduced neuroglia cells activation through downregulation of the NF-κB pathway. In summary, the present work indicated that SIP prevents ischemic cerebral infarction-induced inflammation and apoptosis by blocking inflammasome activation via NF-κB signaling pathway. Those results suggest that SIP treatment is an attractive strategy for prevention of ischemic cerebral infarction.
Collapse
Affiliation(s)
- Min Yu
- Department of Internal Neurology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Yan Li
- Department of Internal Neurology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Xianpei Tan
- Department of Internal Neurology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Qiao Hu
- Department of Internal Neurology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| |
Collapse
|
48
|
SU LJ, REN YC, CHEN Z, MA HF, ZHENG F, LI F, ZHANG YY, GONG SS, KOU JP. Ginsenoside Rb1 improves brain, lung, and intestinal barrier damage in middle cerebral artery occlusion/reperfusion (MCAO/R) micevia the PPARγ signaling pathway. Chin J Nat Med 2022; 20:561-571. [DOI: 10.1016/s1875-5364(22)60204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Indexed: 11/28/2022]
|
49
|
Polyphenols for the Treatment of Ischemic Stroke: New Applications and Insights. Molecules 2022; 27:molecules27134181. [PMID: 35807426 PMCID: PMC9268254 DOI: 10.3390/molecules27134181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Currently, the main therapeutic strategy involves the use of intravenous thrombolysis to restore cerebral blood flow to prevent the transition of the penumbra to the infarct core. However, due to various limitations and complications, including the narrow time window in which this approach is effective, less than 10% of patients benefit from such therapy. Thus, there is an urgent need for alternative therapeutic strategies, with neuroprotection against the ischemic cascade response after IS being one of the most promising options. In the past few decades, polyphenolic compounds have shown great potential in animal models of IS because of their high biocompatibility and ability to target multiple ischemic cascade signaling pathways, although low bioavailability is an issue that limits the applications of several polyphenols. Here, we review the pathophysiological changes following cerebral ischemia and summarize the research progress regarding the applications of polyphenolic compounds in the treatment of IS over the past 5 years. Furthermore, we discuss several potential strategies for improving the bioavailability of polyphenolic compounds as well as some essential issues that remain to be addressed for the translation of the related therapies to the clinic.
Collapse
|
50
|
Protective Effects of Nuciferine in Middle Cerebral Artery Occlusion Rats Based on Transcriptomics. Brain Sci 2022; 12:brainsci12050572. [PMID: 35624959 PMCID: PMC9139097 DOI: 10.3390/brainsci12050572] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Middle cerebral artery occlusion (MCAO), with the characteristics of high morbidity, high recurrence rate, high mortality, and disability rate, is a typical manifestation of ischemic stroke and has become a hot research topic in the clinical field. The protective effects of nuciferine on brain injury MCAO rats were investigated and its mechanisms of actions were revealed. The MCAO rats were established by the suture method. The pathological staining of the rat brain was processed and observed, the pharmacodynamics assay of nuciferine were studied, and the gene expression regulation by nuciferine was detected by transcriptome technology. The results showed that nuciferine significantly alleviated brain damage in MCAO rats, and the transcriptomic results suggested that nuciferine could exert therapeutic effects through the regulation of lipid metabolism, including arachidonic acid metabolism, sphingolipid metabolism, the PPAR signaling pathway and other related pathways. This finding provided new perspectives on the treatment of MCAO with nuciferine and facilitates the development of novel drugs for this disease.
Collapse
|