1
|
Wang Y, Cheng D, He J, Liu S, Wang X, Wang M. Magnolol protects C6 glioma cells against neurotoxicity of FB1 via modulating PI3K/Akt and mitochondria-associated apoptosis signaling pathways. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 372:126017. [PMID: 40057167 DOI: 10.1016/j.envpol.2025.126017] [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: 11/22/2024] [Revised: 02/14/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Fumonisin B1 (FB1) is a contaminant commonly occurring in crops and food. Mycotoxin contamination, including FB1, has been progressively shown to be an important risk factor in mediating neurotoxicity and neurodegenerative diseases. Studies have found that magnolol (MAG) exhibits favorable pharmacological effects in the central nervous system. However, the protective effects of MAG against FB1-induced neurotoxicity and the molecular pathways involved have not been fully elucidated. Our study aimed to investigate the neuroprotective effects of MAG on FB1-exposed C6 cells and to identify the underlying mechanisms. A model of FB1-induced cytotoxicity in C6 glial cells was established. C6 cells were treated with MAG (40, 80 and 160 μM) in the presence/absence of FB1 (15 μM) and then assessed for cell viability, cellular and mitochondrial morphology and oxidative stress. The mechanism of action of MAG was revealed using a variety of means including RNA-seq, qRT-PCR, Western blot, immunofluorescence, scanning electron microscopy analysis and agonist validation experiments. Our results indicated that MAG significantly alleviated AFB1-induced C6 astroglial cytotoxicity, as evidenced by elevated cell viability and restoration of overall cellular and mitochondrial morphology. Meanwhile, MAG also alleviated oxidative stress in FB1-exposed C6 cells, with 80 μM MAG showing the best effect. Transcriptome analysis showed that PI3K/Akt and apoptosis involved in it might be the key pathway for MAG to treat FB1 neurotoxicity. MAG suppressed FB1-induced mitochondria-dependent apoptosis in C6 cells, primarily manifested by reduced apoptosis rate and reversal of apoptosis-associated protein expression. It was verified that MAG restored the expression of p-PI3K and p-Akt in FB1-treated cells and reversed the downstream effectors IKKα and NF-κB via measurement of related protein levels. The rescue experiment using Akt pathway activator (SC79) was further confirmed that activation of the PI3K/Akt signaling pathway is an effective strategy for MAG to mitigate FB1-induced cytotoxicity in C6 astroglial cells.
Collapse
Affiliation(s)
- Yingjie Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Dai Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Jingjing He
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Sijia Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xinlu Wang
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Meng Wang
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
2
|
Fan X, Zhang Z, Gao W, Pan Q, Luo K, He B, Pu Y. An Engineered Butyrate-Derived Polymer Nanoplatform as a Mucosa-Healing Enhancer Potentiates the Therapeutic Effect of Magnolol in Inflammatory Bowel Disease. ACS NANO 2024; 18:229-244. [PMID: 38112525 DOI: 10.1021/acsnano.3c05732] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Colonic epithelial damage and dysregulated immune response are crucial factors in the progression and exacerbation of inflammatory bowel disease (IBD). Nanoenabled targeted drug delivery to the inflamed intestinal mucosa has shown promise in inducing and maintaining colitis remission, while minimizing side effects. Inspired by the excellent antioxidative and anti-inflammatory efficacy of naturally derived magnolol (Mag) and gut homeostasis regulation of microbiota-derived butyrate, we developed a pH/redox dual-responsive butyrate-rich polymer nanoparticle (PSBA) as an oral Mag delivery system for combinational therapy of IBD. PSBA showed a high butyrate content of 22% and effectively encapsulated Mag. The Mag-loaded nanoparticles (PSBA@Mag) demonstrated colonic pH and reduction-responsive drug release, ensuring efficient retention and adhesion in the colon of colitis mice. PSBA@Mag not only normalized the level of reactive oxygen species and inflammatory effectors in inflamed colonic mucosa but also restored the epithelial barrier function in both ulcerative colitis and Crohn's disease mouse models. Importantly, PSBA promoted the migration and healing ability of intestinal epithelial cells in vitro and in vivo, sensitizing the therapeutic efficacy of Mag in animal models. Moreover, transcriptomics and metabolism analyses revealed that PSBA@Mag mitigated inflammation by suppressing the production of pro-inflammatory cytokines and chemokines and restoring the lipid metabolism. Additionally, this nanomedicine modulated the gut microbiota by inhibiting pathogenic Proteus and Escherichia-Shigella and promoting the proliferation of beneficial probiotics, including Lachnoclostridium, Lachnospiraceae_NK4A136_group and norank_f_Ruminococcaceae. Overall, our findings highlight the potential of butyrate-functionalized polymethacrylates as versatile and effective nanoplatforms for colonic drug delivery and mucosa repair in combating IBD and other gastrointestinal disorders.
Collapse
Affiliation(s)
- Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhuangzhuang Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
3
|
Li Z, Zheng Y, Liu K, Liang Y, Lu J, Li Q, Zhao B, Liu X, Li X. Lignans as multi-targeted natural products in neurodegenerative diseases and depression: Recent perspectives. Phytother Res 2023; 37:5599-5621. [PMID: 37669911 DOI: 10.1002/ptr.8003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/09/2023] [Accepted: 08/19/2023] [Indexed: 09/07/2023]
Abstract
As the global population ages, the treatment of neurodegenerative diseases is becoming more and more important. There is an urgent need to discover novel drugs that are effective in treating neurological diseases. In recent years, natural products and their biological activities have gained widespread attention. Lignans are a class of metabolites extensively present in Chinese herbal medicine and possess good pharmacological effects. Latest studies have demonstrated their neuroprotective pharmacological activity in preventing acute/chronic neurodegenerative diseases and depression. In this review, the pharmacological effects of these disorders, the pharmacokinetics, safety, and clinical trials of lignans were summarized according to the scientific literature. These results proved that lignans mainly exert antioxidant and anti-inflammatory activities. Anti-apoptosis, regulation of nervous system functions, and modulation of synaptic signals are also potential effects. Despite the substantial evidence of the neuroprotective potential of lignans, it is not sufficient to support their use in the clinical management. Our study suggests that lignans can be used as prospective agents for the treatment of neurodegenerative diseases and depression, with a view to informing their further development and utilization.
Collapse
Affiliation(s)
- Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Youdan Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Cui D, Chen Y, Ye B, Guo W, Wang D, He J. Natural products for the treatment of neurodegenerative diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155101. [PMID: 37778246 DOI: 10.1016/j.phymed.2023.155101] [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: 01/29/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Neurodegenerative diseases are among the most common diseases in older adults worldwide. Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases, and are accompanied by cerebral cortical atrophy, neuronal loss, protein accumulation, and excessive accumulation of metal ions. Natural products exhibit outstanding performance in improving cerebral circulatory disorders, promoting cerebral haematoma absorption, repairing damaged nerve tissue, and improving damaged nerve function. In recent years, studies have shown that neuroinflammatory mechanisms and signalling pathways closely related to the occurrence and development of neurological diseases include microglial activation, nuclear factor-κB (NF-κB) pathway, mitogen activated protein kinases (MAPK) pathway, reactive oxygen pathway, nucleotide binding oligomerisation domain-like receptor protein3 (NLRP3) inflammasomes, toll-like receptor4 (TLR4) pathway, nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway, phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, and intestinal flora. Therefore, this study considered the mechanism of neurological diseases as the starting point to review the mechanism of action of natural products in the prevention and treatment of AD and PD in recent years to provide a theoretical basis for clinical prevention and treatment. AIM Natural products are a promising source of novel lead structures that have long been used to treat various nervous system diseases. METHODOLOGY This review collected literature on neurological diseases and natural products from 2012 to 2022, which were mainly searched through databases such as ScienceDirect, Springer, PubMed, SciFinder, China National Knowledge Infrastructure (CNKI), Wanfang, Google Scholar, and Baidu Academic. The following keywords were searched: neurological disorders, natural products, signalling pathway, mechanism of action. RESULTS This review summarises the pathogenesis of degenerative neurological diseases, recent findings on natural products used in neurodegenerative diseases, and the molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Donghan Cui
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China
| | - Yajuan Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bengui Ye
- 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 610041, China; Medical College of Tibet University, Lasa 850002, China
| | - Wenhao Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China.
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, Department of Medicine, McMaster University, HSC 4N71, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada.
| | - Jun He
- 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 610041, China.
| |
Collapse
|
5
|
Fan Q, Du E, Chen F, Tao W, Zhao N, Huang S, Guo W, Huang J, Wei J. Maternal Magnolol Supplementation during Pregnancy and Lactation Promotes Antioxidant Capacity, Improves Gut Health, and Alters Gut Microbiota and Metabolites of Weanling Piglets. Metabolites 2023; 13:797. [PMID: 37512505 PMCID: PMC10383630 DOI: 10.3390/metabo13070797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Maternal nutrition exerts a profound effect on the postnatal performance of offspring, especially during the weaning period. The multifunctional bioactive component magnolol (MAG) has shown promise as a dietary supplement. This study aimed to explore the effects of maternal MAG supplementation on the antioxidant capacity, gut health, gut microbiome, and metabolome composition of weanling piglets. Fifty pregnant sows were randomly divided into two equally sized groups, the control group and the group supplemented with 100 g/t MAG during the gestation and lactation periods, and 7 days postweaning, the pups were euthanized. The microbiome and metabolome features of weanling piglet colons were compared. Our results revealed that maternal MAG supplementation modified the serum redox status of weanling piglets by decreasing malondialdehyde concentration and increasing superoxide dismutase activity and total antioxidant capacity. Moreover, the decreased indicators of diarrhea were accompanied by improved gut barrier function, in which serum diamine oxidase concentration was decreased, and expressions of zona occludens-1, claudin-1, and intestinal alkaline phosphatase were increased in the colon of weanling piglets from sows supplemented with MAG. Further analysis of the gut microbiota indicated that maternal MAG supplementation significantly increased the relative abundance of beneficial bacteria in the colon of weanling piglets, including Faecalibacterium prausnitzii and Oscillospira. Metabolome analysis identified 540 differential metabolites in the colon of piglets from MAG-fed dams, of which glycerophospholipid classes were highly correlated with progeny gut health and key beneficial bacteria. Our findings indicated that maternal MAG supplementation can improve the oxidative status and gut health of weanling piglets, possibly due to alterations in the gut microbiota and metabolites.
Collapse
Affiliation(s)
- Qiwen Fan
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Encun Du
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Fang Chen
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Wenjing Tao
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Na Zhao
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Shaowen Huang
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Wanzheng Guo
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Jing Huang
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| | - Jintao Wei
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan 430064, China
| |
Collapse
|
6
|
Pang X, Mao L, Ye D, Wang W, Yang H, Fan X, Yang Y, Su Z, Ma T, Sun M, Liu Y. Synthesis, anti-aging and mechanism of magnolol derivatives. Front Chem 2023; 11:1180375. [PMID: 37288076 PMCID: PMC10242077 DOI: 10.3389/fchem.2023.1180375] [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: 03/06/2023] [Accepted: 04/27/2023] [Indexed: 06/09/2023] Open
Abstract
Magnolol (M), a hydroquinone containing an allyl side chain, is one of the major active components of Houpoea officinalis for antioxidation and anti-aging. To enhance the antioxidant activity of magnolol, the different sites of magnolol were structurally modified in this experiment, and a total of 12 magnolol derivatives were obtained. Based on the preliminary exploration of the anti-aging effect of magnolol derivatives in a Caenorhabditis elegans (C. elegans) model. Our results indicate that the active groups of magnolol exerting anti-aging effects were allyl groups and hydroxyl on the phenyl. Meanwhile, the anti-aging effect of the novel magnolol derivative M27 was found to be significantly superior to that of magnolol. To investigate the effect of M27 on senescence and the potential mechanism of action, we investigated the effect of M27 on senescence in C. elegans. In this study, we investigated the effect of M27 on C. elegans physiology by examining body length, body curvature and pharyngeal pumping frequency. The effect of M27 on stress resistance in C. elegans was explored by acute stress experiments. The mechanism of M27 anti-aging was investigated by measuring ROS content, DAF-16 nuclear translocation, sod-3 expression, and lifespan of transgenic nematodes. Our results indicate that M27 prolonged the lifespan of C. elegans. Meanwhile, M27 improved the healthy lifespan of C. elegans by improving pharyngeal pumping ability and reducing lipofuscin accumulation in C. elegans. M27 increased resistance to high temperature and oxidative stress in C. elegans by reducing ROS. M27 induced DAF-16 translocation from cytoplasm to nucleus in transgenic TJ356 nematodes and upregulated the expression of sod-3 (a gene downstream of DAF-16) in CF1553 nematodes. Furthermore, M27 did not extend the lifespan of daf-16, age-1, daf-2, and hsp-16.2 mutants. This work suggests that M27 may ameliorate aging and extend lifespan in C. elegans through the IIS pathway.
Collapse
Affiliation(s)
- Xinxin Pang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Li Mao
- Beijing Tide Pharmaceutical Co, Ltd., Beijing Econnomi Technological Development Area (BDA), Beijing, China
| | - Danyang Ye
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenqi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongliu Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuping Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhijun Su
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Mingqian Sun
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
ElShebiney S, Elgohary R, El-Shamarka M, Mowaad N, Abulseoud OA. Natural Polyphenols-Resveratrol, Quercetin, Magnolol, and β-Catechin-Block Certain Aspects of Heroin Addiction and Modulate Striatal IL-6 and TNF-α. TOXICS 2023; 11:379. [PMID: 37112606 PMCID: PMC10145039 DOI: 10.3390/toxics11040379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 06/19/2023]
Abstract
We have examined the effects of four different polyphenols in attenuating heroin addiction using a conditioned place preference (CPP) paradigm. Adult male Sprague Dawley rats received heroin (alternating with saline) in escalating doses starting from 10 mg/kg, i.p. up to 80 mg/kg/d for 14 consecutive days. The rats were treated with distilled water (1 mL), quercetin (50 mg/kg/d), β-catechin (100 mg/kg/d), resveratrol (30 mg/kg/d), or magnolol (50 mg/kg/d) through oral gavage for 7 consecutive days, 30 min before heroin administration, starting on day 8. Heroin withdrawal manifestations were assessed 24 h post last heroin administration following the administration of naloxone (1 mg/kg i.p). Heroin CPP reinstatement was tested following a single dose of heroin (10 mg/kg i.p.) administration. Striatal interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) were quantified (ELISA) after naloxone-precipitated heroin withdrawal. Compared to the vehicle, the heroin-administered rats spent significantly more time in the heroin-paired chamber (p < 0.0001). Concomitant administration of resveratrol and quercetin prevented the acquisition of heroin CPP, while resveratrol, quercetin, and magnolol blocked heroin-triggered reinstatement. Magnolol, quercetin, and β-catechin blocked naloxone-precipitated heroin withdrawal and increased striatal IL-6 concentration (p < 0.01). Resveratrol administration was associated with significantly higher withdrawal scores compared to those of the control animals (p < 0.0001). The results of this study show that different polyphenols target specific behavioral domains of heroin addiction in a CPP model and modulate the increase in striatal inflammatory cytokines TNF-α and IL-6 observed during naloxone-precipitated heroin withdrawal. Further research is needed to study the clinical utility of polyphenols and to investigate the intriguing finding that resveratrol enhances, rather than attenuates naloxone-precipitated heroin withdrawal.
Collapse
Affiliation(s)
- Shaimaa ElShebiney
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Rania Elgohary
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Marwa El-Shamarka
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Noha Mowaad
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Osama A. Abulseoud
- Department of Psychiatry and Psychology, Mayo Clinic, Phoenix, AZ 85001, USA
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ 85001, USA
| |
Collapse
|
8
|
Peng W, Zhou N, Song Z, Zhang H, He X. Magnolol as a Protective Antioxidant Alleviates Rotenone-Induced Oxidative Stress and Liver Damage through MAPK/mTOR/Nrf2 in Broilers. Metabolites 2023; 13:metabo13010084. [PMID: 36677009 PMCID: PMC9867015 DOI: 10.3390/metabo13010084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
This study aimed to investigate the protective effects and molecular mechanism of magnolol supplementation on rotenone-induced oxidative stress in broilers. Two hundred and eighty-eight old male AA broilers were randomly divided into four groups: the CON group: basic diet with sunflower oil injection; the ROT group: basic diet with 24 mg/kg BW rotenone; the MAG + ROT group: basic diet with 300 mg/kg magnolol and rotenone injection; and the MAG group: basic diet with 300 mg/kg magnolol and sunflower oil injection. At 21−27 days of age, the broilers in each group were intraperitoneally injected with rotenone (24 mg/kg BW) or the same volume of sunflower oil. The results showed that magnolol reversed the decrease in ADG post-injection and FBW via rotenone induction. Compared to the ROT group, MAG + ROT group enhanced the average daily gain post injection (p < 0.05). Magnolol supplement could improve the activity and mRNA expression of rotenone-suppressed antioxidant enzymes such as GSH and GSH-PX (p < 0.05). Similarly, the MDA content as an oxidative damage marker was significantly reduced after magnolol addition (p < 0.05). The hepatocyte apoptosis and the mRNA expression of apoptosis-related signaling pathway in the ROT group increased, but magnolol supplementation inhibited rotenone-induced apoptosis through the Nrf2 signaling pathway. Through RNA transcriptome analysis, there were 277 differential genes expressions (DEGs) among the CON group with ROT group, and 748 DEGs were found between the ROT group and the MAG + ROT group. KEGG pathway enrichment found that magnolol relieved rotenone-induced energy metabolism disorder and oxidative damage through signaling pathways such as MAPK and mTOR. In conclusion, magnolol attenuates rotenone-induced hepatic injury and oxidative stress of broilers, presumably by restoring hepatic antioxidant function via the MAPK/mTOR/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Weishi Peng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha 410128, China
| | - Nanxuan Zhou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha 410128, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha 410128, China
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha 410128, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha 410128, China
- Correspondence: ; Tel.: +86-138-7496-1391
| |
Collapse
|
9
|
Dai X, Xie L, Liu K, Liang Y, Cao Y, Lu J, Wang X, Zhang X, Li X. The Neuropharmacological Effects of Magnolol and Honokiol: A Review of Signal Pathways and Molecular Mechanisms. Curr Mol Pharmacol 2023; 16:161-177. [PMID: 35196977 DOI: 10.2174/1874467215666220223141101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 11/22/2022]
Abstract
Magnolol and honokiol are natural lignans with good physiological effects. As the main active substances derived from Magnolia officinalis, their pharmacological activities have attracted extensive attention. It is reported that both of them can cross the blood-brain barrier (BBB) and exert neuroprotective effects through a variety of mechanisms. This suggests that these two ingredients can be used as effective therapeutic compounds to treat a wide range of neurological diseases. This article provides a review of the mechanisms involved in the therapeutic effects of magnolol and honokiol in combating diseases, such as cerebral ischemia, neuroinflammation, Alzheimer's disease, and brain tumors, as well as psychiatric disorders, such as anxiety and depression. Although magnolol and honokiol have the pharmacological effects described above, their clinical potential remains untapped. More research is needed to improve the bioavailability of magnolol and honokiol and perform experiments to examine the therapeutic potential of magnolol and honokiol.
Collapse
Affiliation(s)
- Xiaolin Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Kai Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Youdan Liang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yi Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jing Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xian Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xumin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| |
Collapse
|
10
|
Zhu S, Liu F, Zhang R, Xiong Z, Zhang Q, Hao L, Chen S. Neuroprotective Potency of Neolignans in Magnolia officinalis Cortex Against Brain Disorders. Front Pharmacol 2022; 13:857449. [PMID: 35784755 PMCID: PMC9244706 DOI: 10.3389/fphar.2022.857449] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/20/2022] [Indexed: 12/03/2022] Open
Abstract
In recent years, neurological diseases including Alzheimer’s disease, Parkinson’s disease and stroke are one of the main causes of death in the world. At the same time, the incidence of psychiatric disorders including depression and anxiety has been increasing. Accumulating elderly and stressed people suffer from these brain disorders, which is undoubtedly a huge burden on the modern aging society. Neolignans, the main active ingredients in Magnolia officinalis cortex, were reported to have neuroprotective effects. In addition, the key bioactive ingredients of neolignans, magnolol (1) and honokiol (2), were proved to prevent and treat neurological diseases and psychiatric disorders by protecting nerve cells and brain microvascular endothelial cells (BMECs). Furthermore, neolignans played a role in protecting nerve cells via regulation of neuronal function, suppression of neurotoxicity, etc. This review summarizes the neuroprotective effect, primary mechanisms of the leading neolignans and provides new prospects for the treatment of brain disorders in the future.
Collapse
Affiliation(s)
- Shun Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fang Liu, ; Shiyin Chen,
| | - Ruiyuan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zongxiang Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Hao
- Huarun Sanjiu (ya’an) Pharmaceutical Group Co., LTD., Ya’an, China
| | - Shiyin Chen
- Department of Orthopedics of Traditional Chinese Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Fang Liu, ; Shiyin Chen,
| |
Collapse
|
11
|
Li D, Luo F, Guo T, Han S, Wang H, Lin Q. Targeting NF-κB pathway by dietary lignans in inflammation: expanding roles of gut microbiota and metabolites. Crit Rev Food Sci Nutr 2022; 63:5967-5983. [PMID: 35068283 DOI: 10.1080/10408398.2022.2026871] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammation is a major factor affecting human health. Nuclear factor-kappa B (NF-κB) plays a vital role in the development of inflammation, and the promoters of most inflammatory cytokine genes have NF-κB-binding sites. Targeting NF-κB could be an exciting route for the prevention and treatment of inflammatory diseases. As important constituents of natural plants, lignans are proved to have numerous biological functions. There are growing pieces of evidence demonstrate that lignans have the potential anti-inflammatory activities. In this work, the type, structure and source of lignans and the influence on mitigating the inflammation are systematically summarized. This review focuses on the targeting NF-κB signaling pathway in the inflammatory response by different lignans and their molecular mechanisms. Lignans also regulate gut microflora and change gut microbial metabolites, which exert novel pathway to prevent NF-κB activation. Taken together, lignans target NF-κB with various mechanisms to inhibit inflammatory cytokine expressions in the inflammatory response. It will provide a scientific theoretical basis for further research on the anti-inflammatory effects of lignans and the development of functional foods.
Collapse
Affiliation(s)
- Dan Li
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Tianyi Guo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Shuai Han
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Hanqing Wang
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Qinlu Lin
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
12
|
Liu Y, Zhu T, Li J, Bao Y, Cheng B, Chen S, Du J, Hu S. Magnolol Hybrid Nanofibrous Mat with Antibacterial, Anti-Inflammatory, and Microvascularized Properties for Wound Treatment. Biomacromolecules 2022; 23:1124-1137. [DOI: 10.1021/acs.biomac.1c01430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yonghang Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P. R. China
| | - Tonghe Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P. R. China
| | - Jun Li
- Department of Orthopedics, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 301 Yanchang Road, Shanghai 200072, P. R. China
| | - Yiming Bao
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P. R. China
| | - Biao Cheng
- Department of Orthopedics, Shanghai Tenth People’s Hospital Affiliated to Tongji University, 301 Yanchang Road, Shanghai 200072, P. R. China
| | - Sihao Chen
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P. R. China
| | - Juan Du
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P. R. China
| | - Shaowei Hu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Biomedical Sciences of Fudan University, NHC Key Laboratory of Hearing Medicine of Fudan University, Fudan University, 83 Fenyang Road, Shanghai 200031, P. R. China
| |
Collapse
|
13
|
Chtioui W, Balmas V, Delogu G, Migheli Q, Oufensou S. Bioprospecting Phenols as Inhibitors of Trichothecene-Producing Fusarium: Sustainable Approaches to the Management of Wheat Pathogens. Toxins (Basel) 2022; 14:72. [PMID: 35202101 PMCID: PMC8875213 DOI: 10.3390/toxins14020072] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Fusarium spp. are ubiquitous fungi able to cause Fusarium head blight and Fusarium foot and root rot on wheat. Among relevant pathogenic species, Fusarium graminearum and Fusarium culmorum cause significant yield and quality loss and result in contamination of the grain with mycotoxins, mainly type B trichothecenes, which are a major health concern for humans and animals. Phenolic compounds of natural origin are being increasingly explored as fungicides on those pathogens. This review summarizes recent research activities related to the antifungal and anti-mycotoxigenic activity of natural phenolic compounds against Fusarium, including studies into the mechanisms of action of major exogenous phenolic inhibitors, their structure-activity interaction, and the combined effect of these compounds with other natural products or with conventional fungicides in mycotoxin modulation. The role of high-throughput analysis tools to decipher key signaling molecules able to modulate the production of mycotoxins and the development of sustainable formulations enhancing potential inhibitors' efficacy are also discussed.
Collapse
Affiliation(s)
- Wiem Chtioui
- Dipartimento di Agraria, Università degli Studi di Sassari, Via E. De Nicola 9, 07100 Sassari, Italy; (W.C.); (V.B.); (Q.M.)
| | - Virgilio Balmas
- Dipartimento di Agraria, Università degli Studi di Sassari, Via E. De Nicola 9, 07100 Sassari, Italy; (W.C.); (V.B.); (Q.M.)
| | - Giovanna Delogu
- Istituto CNR di Chimica Biomolecolare, Traversa La Crucca 3, 07100 Sassari, Italy;
| | - Quirico Migheli
- Dipartimento di Agraria, Università degli Studi di Sassari, Via E. De Nicola 9, 07100 Sassari, Italy; (W.C.); (V.B.); (Q.M.)
- Nucleo di Ricerca sulla Desertificazione, Università degli Studi di Sassari, Via E. De Nicola 9, 07100 Sassari, Italy
| | - Safa Oufensou
- Dipartimento di Agraria, Università degli Studi di Sassari, Via E. De Nicola 9, 07100 Sassari, Italy; (W.C.); (V.B.); (Q.M.)
- Nucleo di Ricerca sulla Desertificazione, Università degli Studi di Sassari, Via E. De Nicola 9, 07100 Sassari, Italy
| |
Collapse
|
14
|
Wan Y, Wang J, Yang B, Huang C, Tang X, Yi H, Liu Y, Wang S. Effects and mechanisms of CTRP3 overexpression in secondary brain injury following intracerebral hemorrhage in rats. Exp Ther Med 2021; 23:35. [PMID: 34849150 PMCID: PMC8613529 DOI: 10.3892/etm.2021.10957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/15/2021] [Indexed: 12/27/2022] Open
Abstract
C1q/TNF-related protein-3 (CTRP3) is a novel adipokine that serves an important role in oxidative stress, anti-apoptosis, anti-inflammation and immune regulation. The aim of the present study was to investigate the protective role of CTRP3 against intracerebral hemorrhage (ICH)-induced brain injury. A model of autologous arterial blood-induced ICH was constructed in rats. Intracerebral infusion of a lentivirus carrying the CTRP3 gene was used to induce CTRP3 overexpression in the brain. The effects and mechanisms of CTRP3 overexpression on brain injury were investigated by detecting brain edema, blood-brain barrier (BBB) integrity, neurological function and inflammatory-associated factors 3 days after ICH. The present results demonstrated that CTRP3 overexpression ameliorated ICH-induced neurological dysfunction, decreased brain edema, maintained BBB integrity and attenuated inflammation. The protective effect of CTRP3 overexpression was associated with increased activation of silent information regulator 1 (SIRT1). In conclusion, the present study demonstrated that CTRP3 overexpression protected against ICH-induced brain injury in rats, potentially via activating the SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Yu Wan
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Jieqiong Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Bo Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Conggai Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Xiaoqin Tang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Hong Yi
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Yun Liu
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| | - Shaohua Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China.,Medical Experiment Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuang 646000, P.R. China
| |
Collapse
|
15
|
Fu M, Luo F, Wang E, Jiang Y, Liu S, Peng J, Liu B. Magnolol Attenuates Right Ventricular Hypertrophy and Fibrosis in Hypoxia-Induced Pulmonary Arterial Hypertensive Rats Through Inhibition of the JAK2/STAT3 Signaling Pathway. Front Pharmacol 2021; 12:755077. [PMID: 34764873 PMCID: PMC8576411 DOI: 10.3389/fphar.2021.755077] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/11/2021] [Indexed: 01/13/2023] Open
Abstract
Right ventricular (RV) remodeling is one of the essential pathological features in pulmonary arterial hypertension (PAH). RV hypertrophy or fibrosis are the leading causes of RV remodeling. Magnolol (6, 6′, 7, 12-tetramethoxy-2,2′-dimethyl-1-β-berbaman, C18H18O2) is a compound isolated from Magnolia Officinalis. It possesses multiple pharmacological activities, such as anti-oxidation and anti-inflammation. This study aims to evaluate the effects and underlying mechanisms of magnolol on RV remodeling in hypoxia-induced PAH. In vivo, male Sprague Dawley rats were exposed to 10% O2 for 4 weeks to establish an RV remodeling model, which showed hypertrophic and fibrotic features (increases of Fulton index, cellular size, hypertrophic and fibrotic marker expression), accompanied by an elevation in phosphorylation levels of JAK2 and STAT3; these changes were attenuated by treating with magnolol. In vitro, the cultured H9c2 cells or cardiac fibroblasts were exposed to 3% O2 for 48 h to induce hypertrophy or fibrosis, which showed hypertrophic (increases in cellular size as well as the expression of ANP and BNP) or fibrotic features (increases in the expression of collagen Ⅰ, collagen Ⅲ, and α-SMA). Administration of magnolol and TG-101348 or JSI-124 (both JAK2 selective inhibitors) could prevent myocardial hypertrophy and fibrosis, accompanied by the decrease in the phosphorylation level of JAK2 and STAT3. Based on these observations, we conclude that magnolol can attenuate RV hypertrophy and fibrosis in hypoxia-induced PAH rats through a mechanism involving inhibition of the JAK2/STAT3 signaling pathway. Magnolol may possess the potential clinical value for PAH therapy.
Collapse
Affiliation(s)
- Minyi Fu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, China
| | - Fangmei Luo
- Department of Pharmacy, Hunan Children's Hospital, Changsha, China
| | - Eli Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Bin Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Bi R, Fang Z, You M, He Q, Hu B. Microglia Phenotype and Intracerebral Hemorrhage: A Balance of Yin and Yang. Front Cell Neurosci 2021; 15:765205. [PMID: 34720885 PMCID: PMC8549831 DOI: 10.3389/fncel.2021.765205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Intracerebral hemorrhage (ICH) features extremely high rates of morbidity and mortality, with no specific and effective therapy. And local inflammation caused by the over-activated immune cells seriously damages the recovery of neurological function after ICH. Fortunately, immune intervention to microglia has provided new methods and ideas for ICH treatment. Microglia, as the resident immune cells in the brain, play vital roles in both tissue damage and repair processes after ICH. The perihematomal activated microglia not only arouse acute inflammatory responses, oxidative stress, excitotoxicity, and cytotoxicity to cause neuron death, but also show another phenotype that inhibit inflammation, clear hematoma and promote tissue regeneration. The proportion of microglia phenotypes determines the progression of brain tissue damage or repair after ICH. Therefore, microglia may be a promising and imperative therapeutic target for ICH. In this review, we discuss the dual functions of microglia in the brain after an ICH from immunological perspective, elaborate on the activation mechanism of perihematomal microglia, and summarize related therapeutic drugs researches.
Collapse
Affiliation(s)
- Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Fang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingfeng You
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quanwei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Magnolol upregulates CHRM1 to attenuate Amyloid-β-triggered neuronal injury through regulating the cAMP/PKA/CREB pathway. J Nat Med 2021; 76:188-199. [PMID: 34705126 DOI: 10.1007/s11418-021-01574-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by neuronal degeneration and hyperphosphorylated Tau. Magnolol is an active component isolated from Magnolia officinalis with potential neuroprotection activity. However, the function and mechanism of magnolol in AD progression is largely uncertain. In present study, the biomarkers related to AD and magnolol were predicted by bioinformatics analyses. The key biomarker levels were predicted by GSE5281 and GSE36980 using AlzData. Cell viability was detected by CCK-8 assay. mRNA and protein levels were examined by qRT-PCR and western blotting assays. Cell apoptosis was investigated by caspase-3 activity and flow cytometry analyses. The cAMP/PKA/CREB signaling was evaluated by ELISA and western blotting analyses. The results showed that CHRM1 was a key biomarker for magnolol against AD progression. Magnolol attenuated Aβ-induced viability inhibition, Tau hyperphosphorylation and apoptosis in SH-SY5Y cells by upregulating CHRM1. In addition, the cAMP signaling might be a potential pathway of CHRM1 in AD. Magnolol contributed to activation of the cAMP/PKA/CREB pathway through enhancing CHRM1 level. Inactivation of the cAMP/PKA/CREB signaling reversed the suppressive effect of magnolol on Tau hyperphosphorylation and apoptosis in Aβ-treated SH-SY5Y cells. As a conclusion, magnolol mitigated Aβ-induced Tau hyperphosphorylation and neuron apoptosis by upregulating CHRM1 and activating the cAMP/PKA/CREB pathway.
Collapse
|
18
|
Chen W, Jiang L, Hu Y, Fang G, Yang B, Li J, Liang N, Wu L, Hussain Z. Nanomedicines, an emerging therapeutic regimen for treatment of ischemic cerebral stroke: A review. J Control Release 2021; 340:342-360. [PMID: 34695522 DOI: 10.1016/j.jconrel.2021.10.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022]
Abstract
Owing to its intricate pathophysiology, cerebral stroke is a serious medical condition caused by interruption or obstruction of blood supply (blockage of vasculature) to the brain tissues which results in diminished supply of essential nutrients and oxygen (hypoxia) and ultimate necrosis of neuronal tissues. A prompt risks assessment and immediate rational therapeutic plan with proficient neuroprotection play critically important role in the effective management of this neuronal emergency. Various conventional medications are being used for treatment of acute ischemic cerebral stroke but fibrinolytic agents, alone or in combination with other agents are considered the mainstay. These clot-busting agents effectively restore blood supply (reperfusion) to ischemic regions of the brain; however, their clinical significance is hampered due to various factors such as short plasma half-life, limited distribution to brain tissues due to the presence of highly efficient physiological barrier, blood brain barrier (BBB), and lacking of target-specific delivery to the ischemic brain regions. To alleviate these issues, various types of nanomedicines such as polymeric nanoparticles (NPs), liposomes, nanoemulsion, micelles and dendrimers have been designed and evaluated. The implication of these newer therapies (nanomedicines) have revolutionized the therapeutic outcomes by improving the plasma half-life, permeation across BBB, efficient distribution to ischemic cerebral tissues and neuroprotection. Furthermore, the adaptation of some diverse techniques including PEGylation, tethering of targeting ligands on the surfaces of nanomedicines, and pH responsive features have also been pondered. The implication of these emerging adaptations have shown remarkable potential in maximizing the targeting efficiency of drugs to ischemic brain tissues, simultaneous delivery of drugs and imaging agents (for early prognosis as well as monitoring of therapy), and therapeutic outcomes such as long-term neuroprotection.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Lingfei Jiang
- Graduate College, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Yueqiang Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China.
| | - Gang Fang
- Guangxi Zhuang and Yao Medicine Engineering Technology Research Center, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Bilin Yang
- Graduate College, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Junhong Li
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Ni Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Lin Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China.
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
19
|
Fan Q, Chen F, Zhang W, Du E, Zhao N, Huang S, Guo W, Yan X, Chen M, Wei J. Maternal magnolol supplementation alters placental morphology, promotes placental angiogenesis during mid-gestation and improves offspring growth in a pregnant mouse model. Reprod Biol 2021; 21:100567. [PMID: 34653815 DOI: 10.1016/j.repbio.2021.100567] [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: 05/18/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
The placenta is the organ that determines the growth of the fetus and the outcome of pregnancy. Magnolol is a multifunctional polyphenol with antioxidant, anti-inflammatory, anticancer and neuroprotective functions. However, there is less knowledge of the effects or complications in the placenta and the mechanism underlying the effect of magnolol when used during pregnancy. The aim of this study was to explore the effects of maternal magnolol supplementation on pregnancy outcomes and placental alterations in a pregnant mouse model. A total of 128 pregnant mice were randomly divided into 4 groups supplemented with 0, 40, 80 and 160 μM magnolol from gestational day 0 (GD0) to delivery. Our results revealed that the number of large-for-gestation-age fetuses on GD13 and the weaning weight of offspring were increased in the magnolol treatment groups. Moreover, maternal magnolol supplementation increased superoxide dismutase (SOD), decreased malondialdehyde (MDA) in maternal serum, and promoted the expression of heme oxygenase-1 (HO-1) in the placenta. Furthermore, magnolol significantly increased the area of the junctional zone and decidua in the placentas and increased the expression of interferon-γ (INF-γ), tumor necrosis factor-α (TNF-α), chemokine (CC Motif) Ligand 3 (CCL3), chemokine (CXC motif) ligand 10 (CXCL10), insulin-like growth factor-1 (IGF-1) and T-box transcription factor 21 (T-bet) in the placenta during GD13 in pregnant mice, while suppressor of cytokine signaling 1 (SOCS1) was reduced. Moreover, the ratio of blood space in the labyrinth area, hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) were all increased in the magnolol treatment groups on GD13. Taken together, these results indicate that magnolol can improve the growth of offspring, which might be due to the alteration of placental morphology and the promotion of placental angiogenesis during mid-gestation.
Collapse
Affiliation(s)
- Qiwen Fan
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Fang Chen
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Wei Zhang
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Encun Du
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Na Zhao
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Shaowen Huang
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Wanzheng Guo
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China
| | - Xianghua Yan
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Mingxin Chen
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China.
| | - Jintao Wei
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan, 430064, Hubei, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, 430064, Hubei, China.
| |
Collapse
|
20
|
Zhang X, Wang J, Sui A, Zhang N, Lv Q, Liu Z. Antinociceptive Effect of Magnolol in a Neuropathic Pain Model of Mouse. J Pain Res 2021; 14:2083-2093. [PMID: 34267552 PMCID: PMC8275150 DOI: 10.2147/jpr.s317204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Neuropathic pain remains a clinical challenge with limited effective treatments. Previous studies have found that magnolol (Mag), an ingredient existing in some herbs, showed neuroprotective effect. However, it remains unclear whether Mag can alleviate neuropathic pain. METHODS Chronic constriction injury (CCI) is used as the neuropathic pain model. Mice were randomly divided into 5 groups: Sham, CCI, CCI + 5, 10, 30 mg/kg Mag groups. Thermal and mechanical paw withdrawal threshold were performed at baseline and on the 3rd, 5th, 7th, 14th days post-surgery. Lumbar spinal cord and blood samples were collected on the 14th day. Blood lipid profile, kidney and liver functions, as well as the activation of microglia were evaluated, along with the related signal pathway examined using multiple methods including immunohistochemistry, RT-PCR and Western blot. RESULTS Mag alleviated thermal and mechanical hypersensitivity in CCI mice. CCI activated microglia and upregulated the expression of P2Y12, while Mag inhibited microglial activation, and downregulated the expression of P2Y12. Mag also blocked the activation of p38 mitogen-activated protein kinase (MAPK) and other pain-related cytokines such as IL-6, TNF-α and IL-1β. CONCLUSION The findings indicate that Mag has antinociceptive effect on neuropathic pain, probably mediated through P2Y12 receptors and p38 MAPK mediated pathways. With its relatively safe profile, Mag may be a potential therapeutic agent for neuropathic pain.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Juntao Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Aihua Sui
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Nannan Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Qiulan Lv
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Zhenfang Liu
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| |
Collapse
|
21
|
Shen J, Cao MS, Zhou T, Chen Y, Liang J, Song Y, Xue C, Cao MH, Ke K. PGE1 triggers Nrf2/HO-1 signal pathway to resist hemin-induced toxicity in mouse cortical neurons. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:634. [PMID: 33987332 PMCID: PMC8106031 DOI: 10.21037/atm-20-5839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background Prostaglandin E1 (PGE1) exerts various pharmacological effects such as membrane stabilization, anti-inflammatory functions, vasodilation, and platelet aggregation inhibition. We have previously demonstrated that PGE1 has a beneficial impact on patients suffering from intracerebral hemorrhage (ICH). The related mechanism underlying PGE1’s beneficial effect on ICH treatment needs further exploration. Methods The present study elucidates the mechanism of PGE1 on ICH treatment using a neuronal apoptosis model in vitro. The mouse primary cortical neurons were pretreated with different concentrations of PGE1, followed by the treatment with hemin, the main catabolite in whole blood, to mimic the clinical ICH. Results Comparing with the vehicle-treated group, PGE1 prevented cultured cortical neurons from the accumulation of inhibited intracellular levels of reactive oxygen species (ROS), amelioration of mitochondrial membrane potential, and hemin-induced apoptosis. The reduction of ROS and apoptosis were associated with the up-regulation of Heme oxygenase-1 (HO-1) expression. Knockdown of nuclear transcription factor erythroid 2-related factor (Nrf2) by siRNA attenuated the upregulation of HO-1 as well as the protective effect of PGE1. Conclusions Our work suggests that the Nrf2/HO-1 molecular pathway may play a crucial role in treating ICH patients with PGE1 and may represent novel molecular targets, resulting in discovering new drugs for ICH treatment.
Collapse
Affiliation(s)
- Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Mao-Sheng Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Tingting Zhou
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Ying Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jingjing Liang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Song
- Department of Neurology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Chengbin Xue
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mao-Hong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
22
|
Lin Y, Li Y, Zeng Y, Tian B, Qu X, Yuan Q, Song Y. Pharmacology, Toxicity, Bioavailability, and Formulation of Magnolol: An Update. Front Pharmacol 2021; 12:632767. [PMID: 33815113 PMCID: PMC8010308 DOI: 10.3389/fphar.2021.632767] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Magnolol (MG) is one of the primary active components of Magnoliae officinalis cortex, which has been widely used in traditional Chinese and Japanese herbal medicine and possesses a wide range of pharmacological activities. In recent years, attention has been drawn to this component due to its potential as an anti-inflammatory and antitumor drug. To summarize the new biological and pharmacological data on MG, we screened the literature from January 2011 to October 2020. In this review, we provide an actualization of already known anti-inflammatory, cardiovascular protection, antiangiogenesis, antidiabetes, hypoglycemic, antioxidation, neuroprotection, gastrointestinal protection, and antibacterial activities of MG. Besides, results from studies on antitumor activity are presented. We also summarized the molecular mechanisms, toxicity, bioavailability, and formulations of MG. Therefore, we provide a valid cognition of MG.
Collapse
Affiliation(s)
- Yiping Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanlian Zeng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Tian
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolan Qu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianghua Yuan
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Song
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
23
|
Qiu J, Zhang L, Hong J, Ni X, Li J, Li G, Zhang G. Magnolol inhibits sodium currents in freshly isolated mouse dorsal root ganglion neurons. Clin Exp Pharmacol Physiol 2021; 48:347-354. [PMID: 33064853 DOI: 10.1111/1440-1681.13422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022]
Abstract
The voltage-gated sodium channel (VGSC) currents in dorsal root ganglion (DRG) neurons contain mainly TTX-sensitive (TTX-S) and TTX-resistant (TTX-R) Na+ currents. Magnolol (Mag), a hydroxylated biphenyl compound isolated from the bark of Magnolia officinalis, has been well documented to exhibit analgesic effects, but its mechanism is not yet fully understood. The aim of the present study was to investigate whether the antinociceptive effects of Mag is through inhibition of Na+ currents. Na+ currents in freshly isolated mouse DRG neurons were recorded with the whole cell patch clamp technique. Results showed that Mag inhibited TTX-S and TTX-R Na+ currents in a concentration-dependent manner. The IC50 values for block of TTX-S and TTX-R Na+ currents were 9.4 and 7.0 μmol/L, respectively. Therefore, TTX-R Na+ current was more susceptible to Mag than TTX-S Na+ current. For TTX-S Na+ channel, 10 μmol/L Mag shifted the steady state inactivation curve toward more negative by 9.8 mV, without affecting the activation curve. For TTX-R Na+ channel, 7 μmol/L Mag shifted the steady state activation and inactivation curves toward more positive and negative potentials by 6.5 and 11.7 mV, respectively. In addition, Mag significantly postponed recovery of TTX-S and TTX-R Na+ currents from inactivation, and produced frequency dependent blocks of both subtypes of Na+ currents. These results suggest that the inhibitory effects of Mag on Na+ channels may contribute to its analgesic effect.
Collapse
Affiliation(s)
- Jie Qiu
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lulu Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiangru Hong
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiao Ni
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research of Southwest, Medical University, Luzhou, China
| | - Jun Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research of Southwest, Medical University, Luzhou, China
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research of Southwest, Medical University, Luzhou, China
| | - Guangqin Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
24
|
Fu X, Zeng H, Zhao J, Zhou G, Zhou H, Zhuang J, Xu C, Li J, Peng Y, Cao Y, Li Y, Chen H, Wang L, Yan F, Chen G. Inhibition of Dectin-1 Ameliorates Neuroinflammation by Regulating Microglia/Macrophage Phenotype After Intracerebral Hemorrhage in Mice. Transl Stroke Res 2021; 12:1018-1034. [PMID: 33539006 DOI: 10.1007/s12975-021-00889-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/03/2020] [Accepted: 01/10/2021] [Indexed: 01/28/2023]
Abstract
Polarization of microglia/macrophages toward the pro-inflammatory phenotype is an important contributor to neuroinflammation after intracerebral hemorrhage (ICH). Dectin-1 is a pattern recognition receptor that has been reported to play a key role in regulating neuroinflammation in ischemic stroke and spinal cord injury. However, the role and mechanism of action of Dectin-1 after ICH remains unclear. In this study, we investigated the effect of Dectin-1 on modulating the microglia/macrophage phenotype and neuroinflammation and the possible underlying mechanism after ICH. We found that Dectin-1 expression increased after ICH, and was mainly localized in microglia/macrophages. Neutrophil infiltration and microglia/macrophage polarization toward the pro-inflammatory phenotype increased after ICH. However, treatment with a Dectin-1 inhibitor reversed these phenomena and induced a shift the anti-inflammatory phenotype in microglia/macrophages; this resulted in alleviation of neurological dysfunction and facilitated hematoma clearance after ICH. We also found that Dectin-1 crosstalks with the downstream pro-inflammatory pathway, Card9/NF-κB, by activating spleen tyrosine kinase (Syk) both in vivo and in vitro. In conclusion, our data suggest that Dectin-1 is involved in the microglia/macrophage polarization and functional recovery after ICH, and that this mechanism, at least in part, may contribute to the involvement of the Syk/Card9/NF-kB pathway.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hanhai Zeng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jikuang Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.,Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Guoyang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yang Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yin Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Huaijun Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| |
Collapse
|
25
|
Zeng Z, Gong X, Hu Z. L-3-n-butylphthalide attenuates inflammation response and brain edema in rat intracerebral hemorrhage model. Aging (Albany NY) 2020; 12:11768-11780. [PMID: 32564011 PMCID: PMC7343495 DOI: 10.18632/aging.103342] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
L-3-n-butylphthalide(NBP), a compound found in Apium graveolens Linn seed extracts, has a therapeutic effect on acute ischemic stroke. The pathological inflammatory pathways and consequent brain edema in intracerebral hemorrhage (ICH) share some similar characteristics with ischemic stroke. We hypothesized that NBP has anti-inflammatory and therapeutic effects on rats with ICH. ICH was induced by an infusion of bacterial collagenase type IV into the unilateral striatum of anesthetized rats. The therapeutic effect of NBP was measured by assessing neurological function, brain water content, blood-brain barrier permeability, and expression of tumor necrosis factor-alpha (TNF-α) and matrix metalloproteinase-9 (MMP-9) around the hematoma 48 hours after surgery. Magnetic resonance imaging was performed 4 and 48 hours after ICH induction, and ICH-induced injured area volumes were measured using T2-weighted images. The NBP treatment group performed better in the neurological function test than the vehicle group. Moreover, in comparison with the vehicle group, NBP group showed a lower expanded hematoma volume, brain water content, blood-brain barrier permeability, and TNF-α/ MMP-9 expression level. Our results indicate that NBP attenuates inflammation and brain edema in rat ICH model. Therefore, our findings also provide a potential therapeutic strategy for the treatment of ICH with NBP.
Collapse
Affiliation(s)
- Zhou Zeng
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xiyu Gong
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
26
|
Chen J, Tao C, Huang X, Chen Z, Wang L, Li X, Ma M, Wu Z. CT2-3, a novel magnolol analogue suppresses NSCLC cells through triggering cell cycle arrest and apoptosis. Bioorg Med Chem 2020; 28:115352. [PMID: 32044229 DOI: 10.1016/j.bmc.2020.115352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/09/2020] [Accepted: 01/28/2020] [Indexed: 12/21/2022]
Abstract
Magnolol, a major bioactive component found in Magnolia officinalis with anti-inflammation and anti-oxidation activities as well as minimized cytotoxic effects. Although magnolol has a wide range of clinical applications, the anti-tumor activity of magnolol is not efficient. Herein, we reported the synthesis and anti-cancer activities of three novel magnolol analogues CT2-1, CT2-2, CT2-3, among which CT2-3 revealed more efficient anti-non-small cell lung cancer (NSCLC) activity than magnolol. Our data showed that CT2-3 could significantly inhibit the proliferation of human NSCLC cells in a dose-dependent manner. In addition, we revealed CT2-3 could induce cell cycle arrest through down-regulating mRNA expression of CDK4, CDK6 and cyclin D1. Moreover, we verified that CT2-3 could cause ROS generation, leading to apoptosis of human NSCLC cells. Further more, we also provided strong evidences that CT2-3 down-regulates the expression of c-Myc and topoisomerases, and contributes to the apoptosis of human NSCLC cells. Taken together, the current study is the first to report a promising new chemotherapeutic drug candidate CT2-3 that can efficiently eliminate human NSCLC cells through triggering cell cycle arrest as well as ROS-mediated and c-Myc/topoisomerases-mediated apoptosis.
Collapse
Affiliation(s)
- Jian Chen
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China; The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China
| | - Cheng Tao
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China; The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China
| | - Xiaofei Huang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zide Chen
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Li Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xinping Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China
| | - Min Ma
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China; The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China.
| | - Zhengzhi Wu
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China; The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China.
| |
Collapse
|