首页 > 最新文献

Phytomedicine最新文献

英文 中文
A comprehensive and systematic review on Curcumin as a promising candidate for the inhibition of melanoma growth: From pre-clinical evidence to molecular mechanisms of action. 姜黄素作为抑制黑色素瘤生长的有望候选药物的全面系统综述:从临床前证据到分子作用机制。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-07 DOI: 10.1016/j.phymed.2024.156073
Qi-Hao Guo, Ling-Yan Jian, Yihan Hu, Shu Wang

Background: Melanoma, a highly malignant skin tumor, can develop systemic metastases during the early stage. Several studies of melanoma animal models indicate that curcumin, a natural plant extract, inhibits melanoma growth through various mechanisms. To evaluate the relationships among different experimental conditions, curcumin itself, its derivatives, and special formulations, it is necessary to conduct a systematic review and meta-analysis.

Purpose: This meta-analysis aims to evaluate the potential of Curcumin as a drug for inhibiting the growth of melanoma and to determine the optimal dosage range and treatment duration for Curcumin administration.

Methods: A systematic search of studies published from inception to December 2023 was conducted across six databases (PubMed, Web of Science, Embase, China National Knowledge Infrastructure, Wanfang Data, and VIP). Methodological quality was assessed using SYRCLE's RoB tool. Study heterogeneity was assessed using Cochran's Q test and I2 statistics. Publication bias risk was evaluated using a funnel plot. All analyses were performed using R (version 4.3.3). Additionally, three-dimensional effect analysis and machine learning techniques were utilized to determine the optimal dosage range and treatment duration for Curcumin administration.

Results: Forty studies involving 989 animals were included. The results demonstrated that, relative to the control group, administration of Curcumin resulted in a significant reduction in tumor volume. [SMD=-3.44; 95 % CI (-4.25, -2.63); P<0.01; I2 = 79 %] and tumor weight [SMD=-1.93; 95 % CI (-2.41, -1.45); P<0.01; I2 = 75 %]. Additionally, Curcumin demonstrated a significant capacity to decrease the number of lung tumor nodules and microangiogenesis, as well as to extend survival time, in animal models. The results from three-dimensional effect analysis and machine learning emphasize that the optimal dosage range for Curcumin is 25-50 mg/kg, with an intervention duration of 10-20 days.

Conclusion: Curcumin can inhibit the growth of melanoma, and the dose-response relationship is not linear. However, further large-scale animal and clinical studies are required to confirm these conclusions.

背景:黑色素瘤是一种高度恶性的皮肤肿瘤,可在早期发生全身转移。多项黑色素瘤动物模型研究表明,姜黄素(一种天然植物提取物)可通过多种机制抑制黑色素瘤的生长。为了评估不同实验条件、姜黄素本身、其衍生物和特殊制剂之间的关系,有必要进行系统回顾和荟萃分析。目的:本荟萃分析旨在评估姜黄素作为抑制黑色素瘤生长药物的潜力,并确定姜黄素的最佳剂量范围和给药疗程:方法:在六个数据库(PubMed、Web of Science、Embase、中国国家知识基础设施、万方数据和VIP)中对从开始到2023年12月发表的研究进行了系统检索。采用 SYRCLE 的 RoB 工具对方法学质量进行评估。研究异质性采用 Cochran's Q 检验和 I2 统计量进行评估。使用漏斗图评估发表偏倚风险。所有分析均使用 R(4.3.3 版)进行。此外,还利用三维效应分析和机器学习技术来确定姜黄素的最佳剂量范围和治疗时间:结果:共纳入 40 项研究,涉及 989 只动物。结果表明,与对照组相比,姜黄素能显著减少肿瘤体积。[SMD=-3.44; 95 % CI (-4.25, -2.63); P2 = 79 %]和肿瘤重量[SMD=-1.93; 95 % CI (-2.41, -1.45); P2 = 75 %]。此外,姜黄素在动物模型中还能显著减少肺部肿瘤结节数量和微血管生成,并延长生存时间。三维效应分析和机器学习的结果强调,姜黄素的最佳剂量范围为 25-50 毫克/千克,干预时间为 10-20 天:结论:姜黄素可以抑制黑色素瘤的生长,其剂量-反应关系不是线性的。结论:姜黄素可以抑制黑色素瘤的生长,剂量反应关系不是线性的,但还需要进一步的大规模动物和临床研究来证实这些结论。
{"title":"A comprehensive and systematic review on Curcumin as a promising candidate for the inhibition of melanoma growth: From pre-clinical evidence to molecular mechanisms of action.","authors":"Qi-Hao Guo, Ling-Yan Jian, Yihan Hu, Shu Wang","doi":"10.1016/j.phymed.2024.156073","DOIUrl":"https://doi.org/10.1016/j.phymed.2024.156073","url":null,"abstract":"<p><strong>Background: </strong>Melanoma, a highly malignant skin tumor, can develop systemic metastases during the early stage. Several studies of melanoma animal models indicate that curcumin, a natural plant extract, inhibits melanoma growth through various mechanisms. To evaluate the relationships among different experimental conditions, curcumin itself, its derivatives, and special formulations, it is necessary to conduct a systematic review and meta-analysis.</p><p><strong>Purpose: </strong>This meta-analysis aims to evaluate the potential of Curcumin as a drug for inhibiting the growth of melanoma and to determine the optimal dosage range and treatment duration for Curcumin administration.</p><p><strong>Methods: </strong>A systematic search of studies published from inception to December 2023 was conducted across six databases (PubMed, Web of Science, Embase, China National Knowledge Infrastructure, Wanfang Data, and VIP). Methodological quality was assessed using SYRCLE's RoB tool. Study heterogeneity was assessed using Cochran's Q test and I<sup>2</sup> statistics. Publication bias risk was evaluated using a funnel plot. All analyses were performed using R (version 4.3.3). Additionally, three-dimensional effect analysis and machine learning techniques were utilized to determine the optimal dosage range and treatment duration for Curcumin administration.</p><p><strong>Results: </strong>Forty studies involving 989 animals were included. The results demonstrated that, relative to the control group, administration of Curcumin resulted in a significant reduction in tumor volume. [SMD=-3.44; 95 % CI (-4.25, -2.63); P<0.01; I<sup>2</sup> = 79 %] and tumor weight [SMD=-1.93; 95 % CI (-2.41, -1.45); P<0.01; I<sup>2</sup> = 75 %]. Additionally, Curcumin demonstrated a significant capacity to decrease the number of lung tumor nodules and microangiogenesis, as well as to extend survival time, in animal models. The results from three-dimensional effect analysis and machine learning emphasize that the optimal dosage range for Curcumin is 25-50 mg/kg, with an intervention duration of 10-20 days.</p><p><strong>Conclusion: </strong>Curcumin can inhibit the growth of melanoma, and the dose-response relationship is not linear. However, further large-scale animal and clinical studies are required to confirm these conclusions.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"156073"},"PeriodicalIF":6.7,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142606007","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Taohe Chengqi decoction improves diabetic cognitive dysfunction by alleviating neural stem cell senescence through HIF1α-driven metabolic signaling 洮河承气汤通过HIF1α驱动的代谢信号传导缓解神经干细胞衰老,从而改善糖尿病认知功能障碍。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-05 DOI: 10.1016/j.phymed.2024.156219
Tingting Qin , Zhangxu He , Hozeifa Mohamed Hassan , Qiqi Wang , Le Shi , Yun Yu , Yang Zhou , Wenzhou Zhang , Ziqiao Yuan

Objective

Type 2 diabetes mellitus (T2DM) is characterized by numerous long-term complications, in which progressive cognitive decline represents a significant risk factor for dementia and other neurodegenerative disorders. Taohe Chengqi decoction (THCQ) is a common traditional Chinese formula for treating T2DM; however, the neuroprotective effect of THCQ on diabetes-associated cognitive dysfunction remains unclear. Hence, the present study investigated the therapeutic effects of THCQ on cognitive impairment associated with T2DM and elucidated the underlying mechanisms.

Methods

A stable high-fat diet (HFD) and streptozotocin (STZ)-induced T2DM mouse model was established and received intragastrical THCQ administration. Blood and tissue samples were investigated for biochemical parameters and neuropathology, whereas hippocampal tissue underwent transcriptome analyses and the role of neural stem cell (NSC) senescence was detected both in vivo and in vitro. Network pharmacology analysis and subsequent primary NSC experiments were conducted to explore the involvement of the HIF1α signaling pathway in THCQ-mediated hippocampal NSC senescence. Furthermore, a lentivirus vector overexpressing HIF1α was used to verify the THCQ potential therapeutic effects on HIF1α/PDKs metabolic signaling that influenced NSC senescence.

Results

THCQ alleviated cognitive dysfunction and metabolic abnormalities in HFD/STZ mice, and relieved hippocampal neurodegeneration. Transcriptome analyses and validation experiments revealed THCQ-induced neuroprotective effects by targeting high glucose-mediated hippocampal neuropathy and NSC senescence. Bioinformatic analysis indicated that HIF1α signaling played a significant role in THCQ therapeutic outcomes; while HIF1α overexpression impaired the effects of THCQ on high glucose-induced metabolic disorders and NSC senescence.

Conclusion

The present study demonstrated that THCQ improved diabetic cognitive dysfunction and hippocampal neurogenesis, the effects of which were mainly attributed to the restoration of metabolic homeostasis and inhibition of NSC senescence through HIF1α signaling. Our results provide novel insights into the therapeutic framework for diabetic neuropathy and indicate that THCQ might be a promising candidate for the management of T2DM-related cognitive disorders.
目的:2 型糖尿病(T2DM)有许多长期并发症,其中认知能力的逐渐下降是导致痴呆和其他神经退行性疾病的重要风险因素。洮河承气汤(THCQ)是治疗 T2DM 的常用传统中药配方;然而,THCQ 对糖尿病相关认知功能障碍的神经保护作用仍不清楚。因此,本研究探讨了THCQ对T2DM相关认知功能障碍的治疗作用,并阐明了其潜在机制:方法:建立稳定的高脂饮食(HFD)和链脲佐菌素(STZ)诱导的 T2DM 小鼠模型,并在胃内给予 THCQ。对血液和组织样本进行了生化参数和神经病理学调查,同时对海马组织进行了转录组分析,并在体内和体外检测了神经干细胞(NSC)衰老的作用。研究人员进行了网络药理学分析和随后的原代神经干细胞实验,以探索HIF1α信号通路在THCQ介导的海马神经干细胞衰老中的参与情况。此外,还使用过表达HIF1α的慢病毒载体来验证THCQ对影响NSC衰老的HIF1α/PDKs代谢信号的潜在治疗作用:结果:THCQ减轻了HFD/STZ小鼠的认知功能障碍和代谢异常,缓解了海马神经退行性变。转录组分析和验证实验显示,THCQ通过靶向高糖介导的海马神经病变和NSC衰老诱导神经保护作用。生物信息学分析表明,HIF1α信号传导在THCQ的治疗结果中发挥了重要作用;而HIF1α过表达则削弱了THCQ对高糖诱导的代谢紊乱和NSC衰老的作用:本研究表明,THCQ可改善糖尿病认知功能障碍和海马神经发生,其作用主要归因于通过HIF1α信号传导恢复代谢平衡和抑制NSC衰老。我们的研究结果为糖尿病神经病变的治疗框架提供了新的见解,并表明THCQ可能是治疗T2DM相关认知障碍的一种有前途的候选药物。
{"title":"Taohe Chengqi decoction improves diabetic cognitive dysfunction by alleviating neural stem cell senescence through HIF1α-driven metabolic signaling","authors":"Tingting Qin ,&nbsp;Zhangxu He ,&nbsp;Hozeifa Mohamed Hassan ,&nbsp;Qiqi Wang ,&nbsp;Le Shi ,&nbsp;Yun Yu ,&nbsp;Yang Zhou ,&nbsp;Wenzhou Zhang ,&nbsp;Ziqiao Yuan","doi":"10.1016/j.phymed.2024.156219","DOIUrl":"10.1016/j.phymed.2024.156219","url":null,"abstract":"<div><h3>Objective</h3><div>Type 2 diabetes mellitus (T2DM) is characterized by numerous long-term complications, in which progressive cognitive decline represents a significant risk factor for dementia and other neurodegenerative disorders. Taohe Chengqi decoction (THCQ) is a common traditional Chinese formula for treating T2DM; however, the neuroprotective effect of THCQ on diabetes-associated cognitive dysfunction remains unclear. Hence, the present study investigated the therapeutic effects of THCQ on cognitive impairment associated with T2DM and elucidated the underlying mechanisms.</div></div><div><h3>Methods</h3><div>A stable high-fat diet (HFD) and streptozotocin (STZ)-induced T2DM mouse model was established and received intragastrical THCQ administration. Blood and tissue samples were investigated for biochemical parameters and neuropathology, whereas hippocampal tissue underwent transcriptome analyses and the role of neural stem cell (NSC) senescence was detected both <em>in vivo</em> and <em>in vitro</em>. Network pharmacology analysis and subsequent primary NSC experiments were conducted to explore the involvement of the HIF1α signaling pathway in THCQ-mediated hippocampal NSC senescence. Furthermore, a lentivirus vector overexpressing HIF1α was used to verify the THCQ potential therapeutic effects on HIF1α/PDKs metabolic signaling that influenced NSC senescence.</div></div><div><h3>Results</h3><div>THCQ alleviated cognitive dysfunction and metabolic abnormalities in HFD/STZ mice, and relieved hippocampal neurodegeneration. Transcriptome analyses and validation experiments revealed THCQ-induced neuroprotective effects by targeting high glucose-mediated hippocampal neuropathy and NSC senescence. Bioinformatic analysis indicated that HIF1α signaling played a significant role in THCQ therapeutic outcomes; while HIF1α overexpression impaired the effects of THCQ on high glucose-induced metabolic disorders and NSC senescence.</div></div><div><h3>Conclusion</h3><div>The present study demonstrated that THCQ improved diabetic cognitive dysfunction and hippocampal neurogenesis, the effects of which were mainly attributed to the restoration of metabolic homeostasis and inhibition of NSC senescence through HIF1α signaling. Our results provide novel insights into the therapeutic framework for diabetic neuropathy and indicate that THCQ might be a promising candidate for the management of T2DM-related cognitive disorders.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"Article 156219"},"PeriodicalIF":6.7,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626470","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Icariin ameliorates Coxsackievirus B3-induced viral myocarditis by modulating the S100 calcium binding protein A6/β-catenin/c-Myc signaling pathway. 淫羊藿苷通过调节S100钙结合蛋白A6/β-catenin/c-Myc信号通路改善柯萨奇病毒B3诱导的病毒性心肌炎
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-04 DOI: 10.1016/j.phymed.2024.156214
Huizhen Tian, Qigang Pan, Jianfeng Wu, Juanjuan Liao, Yuwei Wan, Ke Pei, Qiong Liu, Lingbing Zeng, Yanli Cao, Qiaofa Shi, Nanzhen Kuang, LiJuan Sun, Xiaomin Yu, Xiaotian Huang

Background: Coxsackievirus B3 (CVB3) is a leading cause of viral myocarditis and is currently lacking specific pharmacological treatments, highlighting the critical need for therapeutic development. Icariin (ICA), a prenylated flavonol glycoside, was previously found to exhibit several pharmacological effects, but its potential to combat CVB3 remains uninvestigated.

Purpose: This study aimed to elucidate the anti-CVB3 efficacy of ICA and elucidate its molecular mechanisms.

Methods: CVB3-infected HeLa cells, H9C2 cells and neonate rat ventricular cardiomyocytes (NRVCs) were selected as in vitro models, and were treated with ICA at 1 and 10 μM. Additionally, BALB/c mice that were infected with CVB3 via intraperitoneal injection were chosen as in vivo model and were treated with ICA or ribavirin over 3 days. The effect of ICA against CVB3 was determined by Cell Counting Kit-8 (CCK-8) assay, western blot, real-time fluorescence quantitative PCR (RT-qPCR), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) and flow cytometry.

Results: In this study, it was found that ICA is capable of reducing CVB3 viral load both in vitro and in vivo. Mechanistic studies suggested that ICA prevents cardiomyocyte apoptosis by attenuating the S100 calcium binding protein A6 (S100A6)/β-catenin/c-Myc signaling pathway. Additionally, ICA inhibits the secretion of proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1β) and CXC motif chemokine ligand 2 (CXCL2) in heart tissue, thereby mitigating CVB3-induced myocarditis. Moreover, ICA also regulates the immune response of CD4+ T, CD8+ T and Treg cells by changing the cells numbers in spleen tissue. Lastly, ICA can reduce the load of other enteroviruses (such as CVA6, CVA16 and EV71) in rhabdomyosarcoma (RD) cells as well.

Conclusion: Our findings indicate that ICA provides significant protection against CVB3 infection by modulating the S100A6/β-catenin/c-Myc signaling pathway, suggesting its potential use as a novel drug against CVB3 infection in clinical application.

背景:柯萨奇病毒B3(CVB3)是病毒性心肌炎的主要病因,目前缺乏特效药物治疗,这凸显了治疗开发的迫切需要。目的:本研究旨在阐明ICA的抗CVB3功效并阐明其分子机制:方法:选择CVB3感染的HeLa细胞、H9C2细胞和新生大鼠心室心肌细胞(NRVCs)作为体外模型,用1和10 μM的ICA处理。此外,还选择经腹腔注射感染 CVB3 的 BALB/c 小鼠作为体内模型,用 ICA 或利巴韦林治疗 3 天。通过细胞计数试剂盒-8(CCK-8)检测、Western印迹、实时荧光定量 PCR(RT-qPCR)、末端脱氧核苷酸转移酶 dUTP 缺口标记(TUNEL)检测、苏木精和伊红(H&E)染色、免疫组织化学(IHC)和流式细胞术测定 ICA 对 CVB3 的作用:结果:本研究发现,ICA 在体外和体内均能降低 CVB3 病毒载量。机理研究表明,ICA可通过抑制S100钙结合蛋白A6(S100A6)/β-catenin/c-Myc信号通路防止心肌细胞凋亡。此外,ICA 还能抑制心脏组织中促炎细胞因子肿瘤坏死因子α(TNF-α)、白细胞介素-1β(IL-1β)和 CXC motif 趋化因子配体 2(CXCL2)的分泌,从而减轻 CVB3 引起的心肌炎。此外,ICA 还能通过改变脾脏组织中 CD4+ T、CD8+ T 和 Treg 细胞的数量来调节其免疫反应。最后,ICA 还能减少横纹肌肉瘤(RD)细胞中其他肠道病毒(如 CVA6、CVA16 和 EV71)的载量:我们的研究结果表明,ICA通过调节S100A6/β-catenin/c-Myc信号通路,对CVB3感染具有显著的保护作用,这表明它有可能作为一种新型药物用于临床抗CVB3感染。
{"title":"Icariin ameliorates Coxsackievirus B3-induced viral myocarditis by modulating the S100 calcium binding protein A6/β-catenin/c-Myc signaling pathway.","authors":"Huizhen Tian, Qigang Pan, Jianfeng Wu, Juanjuan Liao, Yuwei Wan, Ke Pei, Qiong Liu, Lingbing Zeng, Yanli Cao, Qiaofa Shi, Nanzhen Kuang, LiJuan Sun, Xiaomin Yu, Xiaotian Huang","doi":"10.1016/j.phymed.2024.156214","DOIUrl":"https://doi.org/10.1016/j.phymed.2024.156214","url":null,"abstract":"<p><strong>Background: </strong>Coxsackievirus B3 (CVB3) is a leading cause of viral myocarditis and is currently lacking specific pharmacological treatments, highlighting the critical need for therapeutic development. Icariin (ICA), a prenylated flavonol glycoside, was previously found to exhibit several pharmacological effects, but its potential to combat CVB3 remains uninvestigated.</p><p><strong>Purpose: </strong>This study aimed to elucidate the anti-CVB3 efficacy of ICA and elucidate its molecular mechanisms.</p><p><strong>Methods: </strong>CVB3-infected HeLa cells, H9C2 cells and neonate rat ventricular cardiomyocytes (NRVCs) were selected as in vitro models, and were treated with ICA at 1 and 10 μM. Additionally, BALB/c mice that were infected with CVB3 via intraperitoneal injection were chosen as in vivo model and were treated with ICA or ribavirin over 3 days. The effect of ICA against CVB3 was determined by Cell Counting Kit-8 (CCK-8) assay, western blot, real-time fluorescence quantitative PCR (RT-qPCR), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) and flow cytometry.</p><p><strong>Results: </strong>In this study, it was found that ICA is capable of reducing CVB3 viral load both in vitro and in vivo. Mechanistic studies suggested that ICA prevents cardiomyocyte apoptosis by attenuating the S100 calcium binding protein A6 (S100A6)/β-catenin/c-Myc signaling pathway. Additionally, ICA inhibits the secretion of proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1β) and CXC motif chemokine ligand 2 (CXCL2) in heart tissue, thereby mitigating CVB3-induced myocarditis. Moreover, ICA also regulates the immune response of CD4<sup>+</sup> T, CD8<sup>+</sup> T and Treg cells by changing the cells numbers in spleen tissue. Lastly, ICA can reduce the load of other enteroviruses (such as CVA6, CVA16 and EV71) in rhabdomyosarcoma (RD) cells as well.</p><p><strong>Conclusion: </strong>Our findings indicate that ICA provides significant protection against CVB3 infection by modulating the S100A6/β-catenin/c-Myc signaling pathway, suggesting its potential use as a novel drug against CVB3 infection in clinical application.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"156214"},"PeriodicalIF":6.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrated gut microbiome and UHPLC-MS metabolomics to reveal the prevention mechanism of pidanjiangtang granules on IGT Rats. 综合肠道微生物组学和超高效液相色谱-质谱代谢组学揭示皮丹江唐颗粒对IGT大鼠的防治机制
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-03 DOI: 10.1016/j.phymed.2024.156201
Yu Xie, Zirong Li, Yue Fan, Xinyi Liu, Ran Yi, Yaoyao Gan, Zixuan Yang, Shangjian Liu

Introduction: Pidanjiangtang (PDJT) is a traditional Chinese medicine formula empirically used to treat impaired glucose tolerance (IGT) based on the "Pidan" theory from the classic ancient book Nei Jing. However, the mechanism of PDJT intervention for IGT remains to be studied.

Objective: This study aims to explore the mechanism of PDJT granules intervention in IGT by integrating gut microbiome and UHPLC-MS untargeted metabolomics.

Materials and methods: The IGT model was established in 6-week-old male Sprague-Dawley (SD) rats by feeding them a high-fat diet and using an STZ injection. The low, medium, and high doses of PDJT were used for six weeks. metformin (Glucophage) was used as the positive control drug. The efficacy of PDJT was evaluated using fasting blood glucose (FBG), blood glucose maximum (BGmax), blood lipid, and inflammatory factor levels. Finally, 16S rDNA gut microbiome sequencing with metabolomics analysis was used to explore the pharmacological mechanism of PDJT intervention in IGT.

Results: PDJT could reverse the phenotype of IGT rats, reduce blood glucose levels, improve lipid metabolism disorder, and reduce inflammatory response. Gut microbiome analysis found that PDJT can improve gut microbiota composition and abundance of three phyla (Firmicutes, Bacteroidota, Desulfobacterota) and four genera (unclassified_f__Lachnospiraceae, Ruminococcus, Allobaculum, Desulfovibrio), which play an important role in the process of PDJT intervention on glucose metabolism and lipid metabolism in IGT rats. UHPLC-MS untargeted metabolomics showed that PDJT could regulate the levels of 258 metabolites in lipid metabolism pathways, inflammatory response pathways, fat and protein digestion, and absorption. The combined analysis of the two omics showed that improving the body's metabolism by gut microbes may be the possible mechanism of PDJT in treating IGT. Thus, this study provides a new method to integrate gut microbiome and UHPLC-MS untargeted metabolomics to evaluate the pharmacodynamics and mechanism of PDJT intervention in IGT, providing valuable ideas and insights for future research on the treatment of IGT with traditional Chinese medicine.

简介皮丹降糖汤(PDJT)是根据经典古书《内经》中的 "皮丹 "理论,经验性地用于治疗糖耐量受损(IGT)的传统中药方剂。然而,PDJT干预IGT的机制仍有待研究:本研究旨在通过整合肠道微生物组和 UHPLC-MS 非靶向代谢组学,探讨 PDJT 颗粒干预 IGT 的机制:以6周龄雄性Sprague-Dawley(SD)大鼠为研究对象,采用高脂饮食和注射STZ的方法建立IGT模型。以二甲双胍(Glucophage)为阳性对照药,分别使用低、中、高剂量 PDJT 六周。通过空腹血糖(FBG)、血糖最高值(BGmax)、血脂和炎症因子水平来评估 PDJT 的疗效。最后,通过 16S rDNA 肠道微生物组测序和代谢组学分析,探讨了 PDJT 干预 IGT 的药理机制:结果:PDJT能逆转IGT大鼠的表型,降低血糖水平,改善脂质代谢紊乱,减轻炎症反应。肠道微生物组分析发现,PDJT能改善肠道微生物组的组成和丰度,其中3个门(Firmicutes、Bacteroidota、Desulfobacterota)和4个属(unclassified__f__Lachnospiraceae、Ruminococcus、Allobaculum、Desulfovibrio)在PDJT干预IGT大鼠糖代谢和脂代谢的过程中发挥了重要作用。超高效液相色谱-质谱(UHPLC-MS)非靶向代谢组学研究表明,PDJT可调节脂质代谢通路、炎症反应通路、脂肪和蛋白质消化吸收通路中258种代谢物的水平。这两项全息研究的综合分析表明,通过肠道微生物改善机体代谢可能是 PDJT 治疗 IGT 的可能机制。因此,本研究提供了一种整合肠道微生物组和UHPLC-MS非靶向代谢组学的新方法来评估PDJT干预IGT的药效学和机制,为今后中药治疗IGT的研究提供了宝贵的思路和见解。
{"title":"Integrated gut microbiome and UHPLC-MS metabolomics to reveal the prevention mechanism of pidanjiangtang granules on IGT Rats.","authors":"Yu Xie, Zirong Li, Yue Fan, Xinyi Liu, Ran Yi, Yaoyao Gan, Zixuan Yang, Shangjian Liu","doi":"10.1016/j.phymed.2024.156201","DOIUrl":"https://doi.org/10.1016/j.phymed.2024.156201","url":null,"abstract":"<p><strong>Introduction: </strong>Pidanjiangtang (PDJT) is a traditional Chinese medicine formula empirically used to treat impaired glucose tolerance (IGT) based on the \"Pidan\" theory from the classic ancient book Nei Jing. However, the mechanism of PDJT intervention for IGT remains to be studied.</p><p><strong>Objective: </strong>This study aims to explore the mechanism of PDJT granules intervention in IGT by integrating gut microbiome and UHPLC-MS untargeted metabolomics.</p><p><strong>Materials and methods: </strong>The IGT model was established in 6-week-old male Sprague-Dawley (SD) rats by feeding them a high-fat diet and using an STZ injection. The low, medium, and high doses of PDJT were used for six weeks. metformin (Glucophage) was used as the positive control drug. The efficacy of PDJT was evaluated using fasting blood glucose (FBG), blood glucose maximum (BGmax), blood lipid, and inflammatory factor levels. Finally, 16S rDNA gut microbiome sequencing with metabolomics analysis was used to explore the pharmacological mechanism of PDJT intervention in IGT.</p><p><strong>Results: </strong>PDJT could reverse the phenotype of IGT rats, reduce blood glucose levels, improve lipid metabolism disorder, and reduce inflammatory response. Gut microbiome analysis found that PDJT can improve gut microbiota composition and abundance of three phyla (Firmicutes, Bacteroidota, Desulfobacterota) and four genera (unclassified_f__Lachnospiraceae, Ruminococcus, Allobaculum, Desulfovibrio), which play an important role in the process of PDJT intervention on glucose metabolism and lipid metabolism in IGT rats. UHPLC-MS untargeted metabolomics showed that PDJT could regulate the levels of 258 metabolites in lipid metabolism pathways, inflammatory response pathways, fat and protein digestion, and absorption. The combined analysis of the two omics showed that improving the body's metabolism by gut microbes may be the possible mechanism of PDJT in treating IGT. Thus, this study provides a new method to integrate gut microbiome and UHPLC-MS untargeted metabolomics to evaluate the pharmacodynamics and mechanism of PDJT intervention in IGT, providing valuable ideas and insights for future research on the treatment of IGT with traditional Chinese medicine.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"156201"},"PeriodicalIF":6.7,"publicationDate":"2024-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Damascenone inhibits osteoclastogenesis by epigenetically modulating Nrf2-mediated ROS scavenge and counteracts OVX-induced osteoporosis. 大马士酮通过表观遗传调节 Nrf2 介导的 ROS 清除抑制破骨细胞生成,并对抗 OVX 诱导的骨质疏松症。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-03 DOI: 10.1016/j.phymed.2024.156205
Qingliang Ma, Yinuo Xiong, Zhiwei Jie, Changzhen Li, Congyu Wang, Jingwen Cai, Yuchen Zhang, Jinghang Li, Yunhao You, Mingzheng Chang, Dapeng Zhang, Cheng Qiu, Yuhua Li, Xinyu Liu, Lianlei Wang

Background: Bone formation and resorption regulate bone homeostasis. Excessive osteoclastogenesis enhances bone resorption and causes osteoporosis. Although medicines targeting osteoclast have been developed, these drugs have several side effects. Natural compounds have advantages in safety and efficiency, making them potential candidates for osteoporosis treatment.

Purpose: This study aims to elucidate the role of damascenone (Dama) in osteoclastogenesis and osteoporosis.

Study design and methods: To demonstrate the effect of Dama on osteoclast differentiation and function, we performed multiple in vitro experiments including TRAP staining, F-actin staining, bone slice resorption assay, real-time PCR, and western bolt. Further, ROS detection, network pharmacology, microscale thermophoresis assay, and ChIP assay were conducted to elucidate the underlying molecular mechanism. Finally, the in vivo effects of Dama were verified using an ovariectomy induced osteoporosis mice model.

Results: Dama inhibited RANKL-induced osteoclast differentiation and bone resorptive function in vitro. The expression of osteoclast-related genes and activation of MAPKs and NF-κB signaling in osteoclast were also attenuated by Dama. Meanwhile, Dama reduced intracellular ROS level via up-regulating Nrf2 expression. Network pharmacology demonstrated that HDAC2 is the potential direct target of Dama. Dama inhibited HDAC2 function and increased H3K27ac level of Nrf2, which induced Nrf2 expression and activated ROS scavenging enzymes. Inhibiting NRF2 or activating HDAC2 attenuated the effect of Dama on osteoclastogenesis. Finally, Dama injection suppressed in vivo osteoclastogenesis and ameliorated bone loss induced by OVX.

Conclusion: Dama attenuates osteoclastogenesis by epigenetically modulating Nrf2 expression and ROS scavenge. This study provides evidence for Dama being a potential treatment for osteoporosis.

背景:骨形成和骨吸收调节骨平衡。过度的破骨细胞生成会促进骨吸收,导致骨质疏松症。虽然针对破骨细胞的药物已经开发出来,但这些药物有一些副作用。目的:本研究旨在阐明大马士酮(Dama)在破骨细胞生成和骨质疏松症中的作用:为了证明达玛对破骨细胞分化和功能的影响,我们进行了多项体外实验,包括TRAP染色、F-肌动蛋白染色、骨片吸收试验、实时PCR和Western bolt。此外,我们还进行了 ROS 检测、网络药理学、微尺度热电泳分析和 ChIP 分析,以阐明其潜在的分子机制。最后,利用卵巢切除诱导的骨质疏松症小鼠模型验证了 Dama 的体内效应:结果:Dama在体外抑制了RANKL诱导的破骨细胞分化和骨吸收功能。结果:Dama抑制了RANKL诱导的破骨细胞体外分化和骨吸收功能,破骨细胞相关基因的表达以及破骨细胞中MAPKs和NF-κB信号的激活也受到Dama的抑制。同时,Dama 通过上调 Nrf2 的表达降低了细胞内 ROS 的水平。网络药理学证明,HDAC2是Dama的潜在直接靶点。Dama抑制了HDAC2的功能,提高了Nrf2的H3K27ac水平,从而诱导了Nrf2的表达,激活了ROS清除酶。抑制 NRF2 或激活 HDAC2 可减轻 Dama 对破骨细胞生成的影响。最后,Dama注射液抑制了体内破骨细胞的生成,并改善了OVX诱导的骨质流失:结论:Dama通过表观遗传调节Nrf2的表达和ROS的清除来抑制破骨细胞的生成。这项研究为达玛作为一种潜在的骨质疏松症治疗方法提供了证据。
{"title":"Damascenone inhibits osteoclastogenesis by epigenetically modulating Nrf2-mediated ROS scavenge and counteracts OVX-induced osteoporosis.","authors":"Qingliang Ma, Yinuo Xiong, Zhiwei Jie, Changzhen Li, Congyu Wang, Jingwen Cai, Yuchen Zhang, Jinghang Li, Yunhao You, Mingzheng Chang, Dapeng Zhang, Cheng Qiu, Yuhua Li, Xinyu Liu, Lianlei Wang","doi":"10.1016/j.phymed.2024.156205","DOIUrl":"https://doi.org/10.1016/j.phymed.2024.156205","url":null,"abstract":"<p><strong>Background: </strong>Bone formation and resorption regulate bone homeostasis. Excessive osteoclastogenesis enhances bone resorption and causes osteoporosis. Although medicines targeting osteoclast have been developed, these drugs have several side effects. Natural compounds have advantages in safety and efficiency, making them potential candidates for osteoporosis treatment.</p><p><strong>Purpose: </strong>This study aims to elucidate the role of damascenone (Dama) in osteoclastogenesis and osteoporosis.</p><p><strong>Study design and methods: </strong>To demonstrate the effect of Dama on osteoclast differentiation and function, we performed multiple in vitro experiments including TRAP staining, F-actin staining, bone slice resorption assay, real-time PCR, and western bolt. Further, ROS detection, network pharmacology, microscale thermophoresis assay, and ChIP assay were conducted to elucidate the underlying molecular mechanism. Finally, the in vivo effects of Dama were verified using an ovariectomy induced osteoporosis mice model.</p><p><strong>Results: </strong>Dama inhibited RANKL-induced osteoclast differentiation and bone resorptive function in vitro. The expression of osteoclast-related genes and activation of MAPKs and NF-κB signaling in osteoclast were also attenuated by Dama. Meanwhile, Dama reduced intracellular ROS level via up-regulating Nrf2 expression. Network pharmacology demonstrated that HDAC2 is the potential direct target of Dama. Dama inhibited HDAC2 function and increased H3K27ac level of Nrf2, which induced Nrf2 expression and activated ROS scavenging enzymes. Inhibiting NRF2 or activating HDAC2 attenuated the effect of Dama on osteoclastogenesis. Finally, Dama injection suppressed in vivo osteoclastogenesis and ameliorated bone loss induced by OVX.</p><p><strong>Conclusion: </strong>Dama attenuates osteoclastogenesis by epigenetically modulating Nrf2 expression and ROS scavenge. This study provides evidence for Dama being a potential treatment for osteoporosis.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"156205"},"PeriodicalIF":6.7,"publicationDate":"2024-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142639573","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cycloastragenol reduces microglial NLRP3 inflammasome activation in Parkinson's disease models by promoting autophagy and reducing Scrib-driven ROS 环黄芪醇通过促进自噬和减少 Scrib 驱动的 ROS,减少帕金森病模型中小胶质细胞 NLRP3 炎性体的激活。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-03 DOI: 10.1016/j.phymed.2024.156210
Linjuan Feng , Hsuan Lo , Jiahao Zheng , Weipin Weng , Yixin Sun , Xiaodong Pan

Background

In Parkinson's disease (PD), microglial autophagy is crucial for the maintenance of cellular redox homeostasis. Meanwhile, cycloastragenol (CAG), a triterpenoid saponin and the principal active component of Astragalus, reduces the activation of NLRP3 inflammasomes. Nevertheless, the specific molecular mechanisms underlying the CAG-mitigated microglial neuroinflammation remains obscure in PD.

Purpose

This study explored the role of CAG in the activation of microglial NLRP3 inflammasome and the mechanisms underlying its therapeutic potential for PD treatment.

Study design

The effect of CAG was assessed in α-Syn-induced primary microglia and PD models.

Methods

AAV1/2-hsyn-SNCA (A53T) was stereo-injected into the striatum of mice to induce PD models and CAG was orally administered. The mice underwent quantitative 4D proteomics analysis and behavioral assessments. The primary microglia and neuron cultures were analyzed by western blotting, immunofluorescence, transmission electron microscopy, etc.

Results

CAG reduced phagocytosis-induced reactive oxygen species (ROS) by suppressing the microglial Scribble (Scrib) and p22phox expression. Concurrently, CAG enhanced autophagy, promoted α-Syn clearance, and reduced mitochondrial damage. These synergistic effects downregulated NLRP3 inflammasome activation, in turn reducing gasdermin D cleavage, caspase-1 activation, and the release of interleukin-1β and interleukin-18. Further investigation revealed that CAG shielded neurons from α-Syn toxicity, thus attenuating behavioral impairments observed in the mouse PD model.

Conclusion

CAG mitigates neuroinflammation by inhibiting ROS-induced NLRP3 inflammasome activation in microglia via promoting microglial autophagy and reducing the activity of Scrib-associated nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which signifies a promising alternative approach to PD management.
背景:在帕金森病(PD)中,小胶质细胞自噬是维持细胞氧化还原平衡的关键。同时,黄芪的主要活性成分环黄芪皂苷(CAG)可降低NLRP3炎性体的活化。目的:本研究探讨了CAG在激活小胶质细胞NLRP3炎症小体中的作用及其治疗PD的潜在机制:研究设计:在α-Syn诱导的原发性小胶质细胞和PD模型中评估CAG的作用:方法:将 AAV1/2-hsyn-SNCA (A53T) 立体注射到小鼠纹状体以诱导 PD 模型,并口服 CAG。对小鼠进行定量 4D 蛋白质组学分析和行为评估。原代小胶质细胞和神经元培养物通过 Western 印迹、免疫荧光、透射电子显微镜等进行分析。结果:CAG 通过抑制小胶质细胞 Scribble (Scrib) 和 p22phox 的表达,减少了吞噬诱导的活性氧(ROS)。同时,CAG 还能增强自噬、促进 α-Syn 清除并减少线粒体损伤。这些协同作用降低了 NLRP3 炎性体的激活,进而减少了 gasdermin D 的裂解、caspase-1 的激活以及白细胞介素-1β 和白细胞介素-18 的释放。进一步研究发现,CAG能保护神经元免受α-Syn毒性的影响,从而减轻在小鼠帕金森病模型中观察到的行为障碍:结论:CAG通过促进小胶质细胞自噬和降低与Scrib相关的烟酰胺腺嘌呤二核苷酸磷酸(NADPH)氧化酶的活性,抑制ROS诱导的小胶质细胞NLRP3炎性体的激活,从而缓解神经炎症,是治疗帕金森病的一种很有前景的替代方法。
{"title":"Cycloastragenol reduces microglial NLRP3 inflammasome activation in Parkinson's disease models by promoting autophagy and reducing Scrib-driven ROS","authors":"Linjuan Feng ,&nbsp;Hsuan Lo ,&nbsp;Jiahao Zheng ,&nbsp;Weipin Weng ,&nbsp;Yixin Sun ,&nbsp;Xiaodong Pan","doi":"10.1016/j.phymed.2024.156210","DOIUrl":"10.1016/j.phymed.2024.156210","url":null,"abstract":"<div><h3>Background</h3><div>In Parkinson's disease (PD), microglial autophagy is crucial for the maintenance of cellular redox homeostasis. Meanwhile, cycloastragenol (CAG), a triterpenoid saponin and the principal active component of Astragalus, reduces the activation of NLRP3 inflammasomes. Nevertheless, the specific molecular mechanisms underlying the CAG-mitigated microglial neuroinflammation remains obscure in PD.</div></div><div><h3>Purpose</h3><div>This study explored the role of CAG in the activation of microglial NLRP3 inflammasome and the mechanisms underlying its therapeutic potential for PD treatment.</div></div><div><h3>Study design</h3><div>The effect of CAG was assessed in α-Syn-induced primary microglia and PD models.</div></div><div><h3>Methods</h3><div>AAV1/2-hsyn-SNCA (A53T) was stereo-injected into the striatum of mice to induce PD models and CAG was orally administered. The mice underwent quantitative 4D proteomics analysis and behavioral assessments. The primary microglia and neuron cultures were analyzed by western blotting, immunofluorescence, transmission electron microscopy, etc.</div></div><div><h3>Results</h3><div>CAG reduced phagocytosis-induced reactive oxygen species (ROS) by suppressing the microglial Scribble (Scrib) and p22<sup>phox</sup> expression. Concurrently, CAG enhanced autophagy, promoted α-Syn clearance, and reduced mitochondrial damage. These synergistic effects downregulated NLRP3 inflammasome activation, in turn reducing gasdermin D cleavage, caspase-1 activation, and the release of interleukin-1β and interleukin-18. Further investigation revealed that CAG shielded neurons from α-Syn toxicity, thus attenuating behavioral impairments observed in the mouse PD model.</div></div><div><h3>Conclusion</h3><div>CAG mitigates neuroinflammation by inhibiting ROS-induced NLRP3 inflammasome activation in microglia via promoting microglial autophagy and reducing the activity of Scrib-associated nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which signifies a promising alternative approach to PD management.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"Article 156210"},"PeriodicalIF":6.7,"publicationDate":"2024-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626295","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabolic pathways, pharmacokinetic, and brain neurochemicals effects of capsaicin: Comprehensively insights from in vivo studies 辣椒素的代谢途径、药代动力学和脑神经化学效应:体内研究的全面见解。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-02 DOI: 10.1016/j.phymed.2024.156212
Lulu Guo , Wu Fan , Die Li , Zhilin Hao , Pingping Liu , Chang Liu , Kun Cui , Wenjuan Zhang , Xingyu Liu , Qidong Zhang , Jian Mao , Jianping Xie

Background

Capsaicin (CAP), a prominent component of chili pepper known for its potent agonistic effects on TRPV1, has attracted significant attention for its diverse physiological effects. Nevertheless, there remains a paucity of data concerning its in vivo distribution, metabolism, pharmacodynamic properties, and influence on the metabolic profile of the brain.

Methods

Stable isotope tracing, in vitro enzyme incubation, microdialysis coupled with UHPLC-MS/MS techniques were employed to investigate the in vivo metabolic pathways, distribution, and pharmacokinetic properties of CAP, and the potential biases in metabolic pathways was elucidate through molecular docking. Furthermore, the effect of CAP on brain metabolic profiles was assessed using untargeted metabolomics, and spatial visualization analysis was conducted through mass spectrometry imaging.

Results

CAP was distributed predominantly in the kidneys, with lower content in the liver, heart, lungs, brain, and spleen following peripheral administration, and the absorption half-life in the body was about 20 min. CAP primarily underwent alkyl terminal dehydrogenation, hydroxylation, and macrocyclization metabolic pathways under the action of CYP2C9, CYP2C19 and CYP2D6, resulting in at least four metabolites. Among them, the hydroxylation products were main metabolites and the dehydrogenation product 16,17-dihydrocapsaicin could interact with the key binding sites Leu515 and Thr550 of TRPV1 like CAP. CAP quickly diffused to various brain regions and the metabolic characteristics in the striatum were relatively different from that in the blood. The distribution of CAP in the brain primarily triggered the release of neurotransmitters in areas associated with reward, cognition, and memory. Both acute and chronic exposure to CAP elevated amino acid levels in cortical regions, while producing contrasting effects on nucleotide metabolites.

Conclusion

This study offers an initial in-depth analysis of the distribution patterns, metabolic pathways and pharmacodynamic properties of CAP in the body and brain. These findings established a basis for further studies on CAP's pharmacology properties and its influence on the central nervous system.
背景:辣椒素(CAP)是辣椒的一种主要成分,以其对TRPV1的强效激动作用而闻名。然而,有关其体内分布、代谢、药效学特性以及对大脑代谢特征的影响的数据仍然很少:方法:采用稳定同位素示踪、体外酶孵育、微透析和超高效液相色谱-质谱/质谱(UHPLC-MS/MS)技术研究了CAP的体内代谢途径、分布和药代动力学特性,并通过分子对接阐明了代谢途径的潜在偏差。此外,还利用非靶向代谢组学评估了 CAP 对大脑代谢谱的影响,并通过质谱成像进行了空间可视化分析:结果:CAP主要分布在肾脏,外周给药后在肝、心、肺、脑和脾中的含量较低,在体内的吸收半衰期约为20分钟。在 CYP2C9、CYP2C19 和 CYP2D6 的作用下,CAP 主要经过烷基末端脱氢、羟化和大环化代谢途径,产生至少四种代谢物。其中,羟化产物是主要的代谢产物,脱氢产物 16,17-二氢辣椒素能像 CAP 一样与 TRPV1 的关键结合位点 Leu515 和 Thr550 发生相互作用。CAP 很快扩散到大脑各区域,在纹状体中的代谢特征与血液中的代谢特征相对不同。CAP 在大脑中的分布主要触发与奖赏、认知和记忆相关区域的神经递质释放。急性和慢性接触 CAP 都会提高大脑皮层区域的氨基酸水平,同时对核苷酸代谢物产生截然不同的影响:本研究初步深入分析了 CAP 在体内和大脑中的分布模式、代谢途径和药效学特性。这些发现为进一步研究 CAP 的药理特性及其对中枢神经系统的影响奠定了基础。
{"title":"Metabolic pathways, pharmacokinetic, and brain neurochemicals effects of capsaicin: Comprehensively insights from in vivo studies","authors":"Lulu Guo ,&nbsp;Wu Fan ,&nbsp;Die Li ,&nbsp;Zhilin Hao ,&nbsp;Pingping Liu ,&nbsp;Chang Liu ,&nbsp;Kun Cui ,&nbsp;Wenjuan Zhang ,&nbsp;Xingyu Liu ,&nbsp;Qidong Zhang ,&nbsp;Jian Mao ,&nbsp;Jianping Xie","doi":"10.1016/j.phymed.2024.156212","DOIUrl":"10.1016/j.phymed.2024.156212","url":null,"abstract":"<div><h3>Background</h3><div>Capsaicin (CAP), a prominent component of chili pepper known for its potent agonistic effects on TRPV1, has attracted significant attention for its diverse physiological effects. Nevertheless, there remains a paucity of data concerning its <em>in vivo</em> distribution, metabolism, pharmacodynamic properties, and influence on the metabolic profile of the brain.</div></div><div><h3>Methods</h3><div>Stable isotope tracing, <em>in vitro</em> enzyme incubation, microdialysis coupled with UHPLC-MS/MS techniques were employed to investigate the <em>in vivo</em> metabolic pathways, distribution, and pharmacokinetic properties of CAP, and the potential biases in metabolic pathways was elucidate through molecular docking. Furthermore, the effect of CAP on brain metabolic profiles was assessed using untargeted metabolomics, and spatial visualization analysis was conducted through mass spectrometry imaging.</div></div><div><h3>Results</h3><div>CAP was distributed predominantly in the kidneys, with lower content in the liver, heart, lungs, brain, and spleen following peripheral administration, and the absorption half-life in the body was about 20 min. CAP primarily underwent alkyl terminal dehydrogenation, hydroxylation, and macrocyclization metabolic pathways under the action of CYP2C9, CYP2C19 and CYP2D6, resulting in at least four metabolites. Among them, the hydroxylation products were main metabolites and the dehydrogenation product 16,17-dihydrocapsaicin could interact with the key binding sites Leu515 and Thr550 of TRPV1 like CAP. CAP quickly diffused to various brain regions and the metabolic characteristics in the striatum were relatively different from that in the blood. The distribution of CAP in the brain primarily triggered the release of neurotransmitters in areas associated with reward, cognition, and memory. Both acute and chronic exposure to CAP elevated amino acid levels in cortical regions, while producing contrasting effects on nucleotide metabolites.</div></div><div><h3>Conclusion</h3><div>This study offers an initial in-depth analysis of the distribution patterns, metabolic pathways and pharmacodynamic properties of CAP in the body and brain. These findings established a basis for further studies on CAP's pharmacology properties and its influence on the central nervous system.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"Article 156212"},"PeriodicalIF":6.7,"publicationDate":"2024-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Yi-Qi-Jian-Pi-Xiao-Yu formula inhibits cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated ferritinophagy 益气健脾解毒汤通过 STING-NCOA4 介导的嗜铁蛋白抑制顺铂诱导的急性肾损伤
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-31 DOI: 10.1016/j.phymed.2024.156189
Ji Zhu , Aini Yuan , Yifei Le , Xiaohui Chen , Jianan Guo , Jing Liu , Hang Chen , Cai-Yi Wang , Dezhao Lu , Keda Lu
<div><h3>Background</h3><div>The kidneys are the primary excretory organs for platinum drugs, making them susceptible to damage from these drugs. Cisplatin-induced acute kidney injury (CIAKI) is the most common side effect observed in patients undergoing clinical cisplatin treatment. A traditional Chinese medicinal preparation, the Yi-Qi-Jian-Pi-Xiao-Yu formula (YQJPXY), which is a modified formulation of the classical Chinese medicine formula Buyang Huanwu Decoction, has long been used in the treatment of clinical kidney diseases. It is expected to be used to ameliorate cisplatin-induced acute kidney injury. However, the mechanism of this YQJPXY for the treatment of cisplatin-induced acute kidney injury remains unclear.</div></div><div><h3>Purpose</h3><div>The objective of this study is to examine the impact of the YQJPXY on the inhibition of ferroptosis in cisplatin-induced acute kidney injury and to elucidate the underlying mechanisms.</div></div><div><h3>Methods</h3><div>The active components of YQJPXY were analysed using UPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of YQJPXY on CIAKI and ferroptosis in mice subjected to acute cisplatin treatment and in mice receiving cisplatin treatment after STING expression was inhibited using the STING inhibitor C176. The renoprotective effect of YQJPXY on cisplatin-treated mice was evaluated by measuring tissue damage, inflammation and pro-fibrosis. In addition, we employed network pharmacology and molecular docking methodologies to analyse the principal regulatory targets of YQJPXY. Furthermore, the expression of key proteins and markers of ferroptosis and iron metabolism, as well as the levels of key indicators related to STING-associated ferritinophagy, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes.</div></div><div><h3>Results</h3><div>The results demonstrated that YQJPXY reduced the levels of indicators of injury, inflammation and pro-fibrosis in CIAKI mice, with renoprotective effects. Network pharmacological analyses revealed that ferroptosis might be the main biological process regulated by YQJPXY. Furthermore, molecular docking results indicated that STING might be a potential regulatory target of YQJPXY. Furthermore, YQJPXY treatment resulted in a significant reduction in MDA and 4-HNE levels, as well as the inhibition of ferroptosis and improvement in iron metabolic processes. Concomitantly, YQJPXY exhibited a robust protective effect on ferroptosis and iron metabolism homeostasis, as evidenced by its inhibitory action on ferritinophagy. Validation experiments utilising the cisplatin inhibitor C176 demonstrated that YQJPXY inhibits cisplatin-induced ferroptosis in kidney via STING-mediated ferritinophagy.</div></div><div><h3>Conclusion</h3><div>These suggest that YQJPXY alleviates cisplatin-induced acute kidney injury through suppressing ferroptosis v
背景:肾脏是铂类药物的主要排泄器官,因此很容易受到这些药物的损害。顺铂诱发的急性肾损伤(CIAKI)是临床上接受顺铂治疗的患者最常见的副作用。传统中药制剂 "益气活血汤"(YQJPXY)是经典中药方剂 "步阳黄五煎 "的改良配方,长期以来一直被用于临床肾脏疾病的治疗。它有望用于改善顺铂引起的急性肾损伤。目的:本研究旨在探讨 YQJPXY 对抑制顺铂诱导的急性肾损伤中铁细胞生成的影响,并阐明其潜在机制:方法:采用 UPLC-MS/MS 分析 YQJPXY 的活性成分。方法:采用 UPLC-MS/MS 分析了 YQJPXY 的活性成分,并对 YQJPXY 对急性顺铂治疗小鼠和 STING 抑制剂 C176 抑制 STING 表达后接受顺铂治疗的小鼠的 CIAKI 和铁变态反应的影响和调控机制进行了全面研究。我们通过测量组织损伤、炎症和纤维化原,评估了 YQJPXY 对顺铂治疗小鼠的肾脏保护作用。此外,我们还采用网络药理学和分子对接方法分析了 YQJPXY 的主要调控靶点。此外,我们还通过免疫印迹、免疫组织化学、免疫沉淀、实时定量 PCR(qPCR)和特异性探针检测了铁变态反应和铁代谢的关键蛋白和标记物的表达,以及与 STING 相关的嗜铁蛋白相关的关键指标的水平:结果表明:YQJPXY能降低CIAKI小鼠损伤、炎症和纤维化的指标水平,具有肾保护作用。网络药理学分析表明,铁突变可能是 YQJPXY 调节的主要生物过程。此外,分子对接结果表明 STING 可能是 YQJPXY 的潜在调控靶点。此外,YQJPXY 还能显著降低 MDA 和 4-HNE 水平,抑制铁变态反应,改善铁代谢过程。同时,YQJPXY 对嗜铁蛋白的抑制作用也证明了它对铁变态反应和铁代谢平衡具有强大的保护作用。利用顺铂抑制剂 C176 进行的验证实验表明,YQJPXY 可通过 STING 介导的噬铁蛋白作用抑制顺铂诱导的肾脏嗜铁细胞增多:这些研究表明,YQJPXY 可通过 STING-NCOA4 介导的嗜铁蛋白作用抑制顺铂诱导的急性肾损伤,从而减轻顺铂诱导的急性肾损伤。
{"title":"Yi-Qi-Jian-Pi-Xiao-Yu formula inhibits cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated ferritinophagy","authors":"Ji Zhu ,&nbsp;Aini Yuan ,&nbsp;Yifei Le ,&nbsp;Xiaohui Chen ,&nbsp;Jianan Guo ,&nbsp;Jing Liu ,&nbsp;Hang Chen ,&nbsp;Cai-Yi Wang ,&nbsp;Dezhao Lu ,&nbsp;Keda Lu","doi":"10.1016/j.phymed.2024.156189","DOIUrl":"10.1016/j.phymed.2024.156189","url":null,"abstract":"&lt;div&gt;&lt;h3&gt;Background&lt;/h3&gt;&lt;div&gt;The kidneys are the primary excretory organs for platinum drugs, making them susceptible to damage from these drugs. Cisplatin-induced acute kidney injury (CIAKI) is the most common side effect observed in patients undergoing clinical cisplatin treatment. A traditional Chinese medicinal preparation, the Yi-Qi-Jian-Pi-Xiao-Yu formula (YQJPXY), which is a modified formulation of the classical Chinese medicine formula Buyang Huanwu Decoction, has long been used in the treatment of clinical kidney diseases. It is expected to be used to ameliorate cisplatin-induced acute kidney injury. However, the mechanism of this YQJPXY for the treatment of cisplatin-induced acute kidney injury remains unclear.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Purpose&lt;/h3&gt;&lt;div&gt;The objective of this study is to examine the impact of the YQJPXY on the inhibition of ferroptosis in cisplatin-induced acute kidney injury and to elucidate the underlying mechanisms.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Methods&lt;/h3&gt;&lt;div&gt;The active components of YQJPXY were analysed using UPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of YQJPXY on CIAKI and ferroptosis in mice subjected to acute cisplatin treatment and in mice receiving cisplatin treatment after STING expression was inhibited using the STING inhibitor C176. The renoprotective effect of YQJPXY on cisplatin-treated mice was evaluated by measuring tissue damage, inflammation and pro-fibrosis. In addition, we employed network pharmacology and molecular docking methodologies to analyse the principal regulatory targets of YQJPXY. Furthermore, the expression of key proteins and markers of ferroptosis and iron metabolism, as well as the levels of key indicators related to STING-associated ferritinophagy, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Results&lt;/h3&gt;&lt;div&gt;The results demonstrated that YQJPXY reduced the levels of indicators of injury, inflammation and pro-fibrosis in CIAKI mice, with renoprotective effects. Network pharmacological analyses revealed that ferroptosis might be the main biological process regulated by YQJPXY. Furthermore, molecular docking results indicated that STING might be a potential regulatory target of YQJPXY. Furthermore, YQJPXY treatment resulted in a significant reduction in MDA and 4-HNE levels, as well as the inhibition of ferroptosis and improvement in iron metabolic processes. Concomitantly, YQJPXY exhibited a robust protective effect on ferroptosis and iron metabolism homeostasis, as evidenced by its inhibitory action on ferritinophagy. Validation experiments utilising the cisplatin inhibitor C176 demonstrated that YQJPXY inhibits cisplatin-induced ferroptosis in kidney via STING-mediated ferritinophagy.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Conclusion&lt;/h3&gt;&lt;div&gt;These suggest that YQJPXY alleviates cisplatin-induced acute kidney injury through suppressing ferroptosis v","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"Article 156189"},"PeriodicalIF":6.7,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142606135","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Avicularin inhibits ferroptosis and improves cognitive impairments in Alzheimer's disease by modulating the NOX4/Nrf2 axis 阿维菌素通过调节NOX4/Nrf2轴抑制铁蛋白沉积并改善阿尔茨海默病的认知障碍
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-31 DOI: 10.1016/j.phymed.2024.156209
Zixiang Li , Yingying Lu , Yongqi Zhen , Wenke Jin , Xuelan Ma , Ziyue Yuan , Bo Liu , Xian-Li Zhou , Lan Zhang

Background

Alzheimer's disease (AD) is a widespread neurodegenerative disorder for which effective therapies remain elusive, primarily due to the complexity of its underlying pathophysiology. In recent years, natural products have gained attention for their therapeutic potential in AD, owing to their multi-targeted actions and low toxicity profiles. Avicularin (Avi), a flavonoid derived from the peels of Zanthoxylum bungeanum Maxim., has shown promise as an anti-AD agent. However, the specific mechanisms by which Avi mitigates oxidative stress and inhibits ferroptosis in AD models remain insufficiently understood. Further investigation is required to elucidate its therapeutic potential in these pathways.

Purpose

Therefore, this study aims to elucidate the neuroprotective effects of Avi in AD by investigating its impact on the NOX4/Nrf2 signaling pathway, as well as its role in modulating oxidative stress and ferroptosis.

Methods

In this study, an in vitro H2O2-induced oxidative stress model in SH-SY5Y cells was utilized to evaluate the pharmacological efficacy and underlying mechanisms of Avi. Molecular docking, cellular thermal shift assay and bio-layer interferometry assays were conducted to identify potential molecular targets of Avi. Additionally, in vivo models, including scopolamine (SCOP)-induced and APP/PS1 transgenic mice, were employed to assess the cognitive effects of Avi and further explore its associated molecular mechanisms.

Results

Our study demonstrates that Avi effectively attenuates H2O2-induced toxicity in SH-SY5Y cells by reducing apoptosis and enhancing cellular antioxidant defenses. This neuroprotective effect is mediated through the inhibition of NOX4 and the promotion of Nrf2 nuclear translocation. Furthermore, Avi improves cognitive function and mitigates ferroptosis in both SCOP-induced and APP/PS1 transgenic mouse models of AD.

Conclusion

Avi emerges as an effective neuroprotective agent against AD, offering a promising therapeutic approach by targeting the NOX4/Nrf2 signaling axis to alleviate oxidative stress and ferroptosis.
背景:阿尔茨海默病(AD)是一种广泛存在的神经退行性疾病,主要由于其潜在病理生理学的复杂性,有效的治疗方法仍然难以找到。近年来,天然产物因其多靶点作用和低毒性特征,在老年痴呆症的治疗潜力方面备受关注。从Zanthoxylum bungeanum Maxim.果皮中提取的黄酮类化合物Avicularin(Avi)已显示出作为抗AD药物的前景。然而,人们对阿维减轻氧化应激和抑制AD模型中铁细胞凋亡的具体机制仍不甚了解。目的:因此,本研究旨在通过研究阿维对 NOX4/Nrf2 信号通路的影响及其在调节氧化应激和铁氧化过程中的作用,阐明阿维对 AD 的神经保护作用:本研究利用体外 H2O2- 诱导的 SH-SY5Y 细胞氧化应激模型来评估阿维的药理作用和内在机制。通过分子对接、细胞热转移测定和生物层干涉测定,确定了阿维的潜在分子靶点。此外,我们还采用了体内模型,包括东莨菪碱(SCOP)诱导的小鼠和APP/PS1转基因小鼠,来评估阿维对认知的影响,并进一步探索其相关的分子机制:我们的研究表明,阿维通过减少细胞凋亡和增强细胞抗氧化防御能力,有效减轻了H2O2诱导的SH-SY5Y细胞毒性。这种神经保护作用是通过抑制 NOX4 和促进 Nrf2 核转位来实现的。此外,阿维还能改善 SCOP 诱导的 AD 模型和 APP/PS1 转基因小鼠模型的认知功能,减轻铁突变:结论:阿维是一种有效的神经保护剂,可通过靶向 NOX4/Nrf2 信号轴来缓解氧化应激和铁蛋白沉积,从而提供一种有前景的治疗方法。
{"title":"Avicularin inhibits ferroptosis and improves cognitive impairments in Alzheimer's disease by modulating the NOX4/Nrf2 axis","authors":"Zixiang Li ,&nbsp;Yingying Lu ,&nbsp;Yongqi Zhen ,&nbsp;Wenke Jin ,&nbsp;Xuelan Ma ,&nbsp;Ziyue Yuan ,&nbsp;Bo Liu ,&nbsp;Xian-Li Zhou ,&nbsp;Lan Zhang","doi":"10.1016/j.phymed.2024.156209","DOIUrl":"10.1016/j.phymed.2024.156209","url":null,"abstract":"<div><h3>Background</h3><div>Alzheimer's disease (AD) is a widespread neurodegenerative disorder for which effective therapies remain elusive, primarily due to the complexity of its underlying pathophysiology. In recent years, natural products have gained attention for their therapeutic potential in AD, owing to their multi-targeted actions and low toxicity profiles. Avicularin (Avi), a flavonoid derived from the peels of <em>Zanthoxylum bungeanum</em> Maxim., has shown promise as an anti-AD agent. However, the specific mechanisms by which Avi mitigates oxidative stress and inhibits ferroptosis in AD models remain insufficiently understood. Further investigation is required to elucidate its therapeutic potential in these pathways.</div></div><div><h3>Purpose</h3><div>Therefore, this study aims to elucidate the neuroprotective effects of Avi in AD by investigating its impact on the NOX4/Nrf2 signaling pathway, as well as its role in modulating oxidative stress and ferroptosis.</div></div><div><h3>Methods</h3><div>In this study, an <em>in vitro</em> H<sub>2</sub>O<sub>2</sub>-induced oxidative stress model in SH-SY5Y cells was utilized to evaluate the pharmacological efficacy and underlying mechanisms of Avi. Molecular docking, cellular thermal shift assay and bio-layer interferometry assays were conducted to identify potential molecular targets of Avi. Additionally, <em>in vivo</em> models, including scopolamine (SCOP)-induced and APP/PS1 transgenic mice, were employed to assess the cognitive effects of Avi and further explore its associated molecular mechanisms.</div></div><div><h3>Results</h3><div>Our study demonstrates that Avi effectively attenuates H<sub>2</sub>O<sub>2</sub>-induced toxicity in SH-SY5Y cells by reducing apoptosis and enhancing cellular antioxidant defenses. This neuroprotective effect is mediated through the inhibition of NOX4 and the promotion of Nrf2 nuclear translocation. Furthermore, Avi improves cognitive function and mitigates ferroptosis in both SCOP-induced and APP/PS1 transgenic mouse models of AD.</div></div><div><h3>Conclusion</h3><div>Avi emerges as an effective neuroprotective agent against AD, offering a promising therapeutic approach by targeting the NOX4/Nrf2 signaling axis to alleviate oxidative stress and ferroptosis.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"Article 156209"},"PeriodicalIF":6.7,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142606109","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lutein, a versatile carotenoid: Insight on neuroprotective potential and recent advances 叶黄素--一种多功能类胡萝卜素:对神经保护潜力的见解和最新进展。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-31 DOI: 10.1016/j.phymed.2024.156185
Megha Jayakanthan , Janani Manochkumar , Thomas Efferth , Siva Ramamoorthy

Background

Neurodegenerative diseases (NDDs) are a diverse group of neurological disorders with progressive neuronal loss at specific brain regions, leading to impaired cognitive functioning, loss of neuroplasticity, severe neurological impairment, and dementia. The incidence of neurodegenerative diseases is increasing at an alarming rate with current treatments struggling to barely prolong the inevitable. The desperation to discover a therapeutic agent to treat neurodegenerative diseases and to aid in the process of healthy recovery has opened a gateway into natural pigments.

Hypothesis

The xanthophyll pigment lutein may bear the potential as a therapeutic agent against NDDs.

Results

Lutein plays an important role in brain development, cognitive functioning, and improving neuroplasticity. In vitro and in vivo studies revealed the neuroprotective properties of lutein against NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and cerebral ischemia. The neuroprotective effect of lutein is evidenced by the reduction of free radicals and the simultaneous strengthening of the endogenous antioxidant systems by activating the NRF-2/ERK/AKT pathway. Further, it effectively suppressed mitochondrial aberrations, excitotoxicity, overaccumulation of metals, and its resultant complications. The immunomodulatory activity of lutein prevents neuroinflammation by hindering NF-κB nuclear translocation, regulation of NIK/IKK, PI3K/AKT, MAPK/ERK, JNK pathways, and ICAM-1 downregulation. Lutein also rescued the dysregulated cholinergic system and resolved memory defects. Along with its neuroprotective properties, lutein also improved neuroplasticity by enabling neurogenesis through increased GAP-43, NCAM, and BDNF levels.

Conclusion

Lutein exhibits strong neuroprotective activities against various NDDs. Though the investigations are in the exploratory phase, this review presents the consolidation of scattered evidence of the neuroprotective properties of lutein and urges its further exploration in clinical studies.
背景:神经退行性疾病(NDDs)是一类神经系统疾病,其特征是特定脑区的神经元逐渐丧失,导致认知功能受损、神经可塑性丧失、严重神经功能损伤和痴呆。神经退行性疾病的发病率正以惊人的速度增长,而目前的治疗方法只能勉强延长不可避免的病程。人们迫切希望找到一种治疗剂来治疗神经退行性疾病并帮助健康恢复,这就为天然色素打开了大门:假设:叶黄素可能具有治疗神经退行性疾病的潜力:叶黄素在大脑发育、认知功能和改善神经可塑性方面发挥着重要作用。体外和体内研究显示,叶黄素对阿尔茨海默病、帕金森病、亨廷顿病和脑缺血等 NDD 具有神经保护作用。叶黄素的神经保护作用体现在通过激活 NRF-2/ERK/AKT 通路减少自由基并同时加强内源性抗氧化系统。此外,它还能有效抑制线粒体畸变、兴奋性毒性、金属过度积累及其导致的并发症。叶黄素的免疫调节活性通过阻碍 NF-κB 核转位、调节 NIK/IKK、PI3K/AKT、MAPK/ERK、JNK 通路和下调 ICAM-1 来防止神经炎症。叶黄素还能挽救失调的胆碱能系统,解决记忆缺陷问题。叶黄素除了具有神经保护特性外,还能通过提高 GAP-43、NCAM 和 BDNF 水平促进神经发生,从而改善神经可塑性:叶黄素对各种 NDD 具有很强的神经保护作用。尽管研究还处于探索阶段,但本综述整合了叶黄素神经保护特性的零散证据,并敦促在临床研究中进一步探索叶黄素的神经保护特性。
{"title":"Lutein, a versatile carotenoid: Insight on neuroprotective potential and recent advances","authors":"Megha Jayakanthan ,&nbsp;Janani Manochkumar ,&nbsp;Thomas Efferth ,&nbsp;Siva Ramamoorthy","doi":"10.1016/j.phymed.2024.156185","DOIUrl":"10.1016/j.phymed.2024.156185","url":null,"abstract":"<div><h3>Background</h3><div>Neurodegenerative diseases (NDDs) are a diverse group of neurological disorders with progressive neuronal loss at specific brain regions, leading to impaired cognitive functioning, loss of neuroplasticity, severe neurological impairment, and dementia. The incidence of neurodegenerative diseases is increasing at an alarming rate with current treatments struggling to barely prolong the inevitable. The desperation to discover a therapeutic agent to treat neurodegenerative diseases and to aid in the process of healthy recovery has opened a gateway into natural pigments.</div></div><div><h3>Hypothesis</h3><div>The xanthophyll pigment lutein may bear the potential as a therapeutic agent against NDDs.</div></div><div><h3>Results</h3><div>Lutein plays an important role in brain development, cognitive functioning, and improving neuroplasticity. <em>In vitro</em> and <em>in vivo</em> studies revealed the neuroprotective properties of lutein against NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and cerebral ischemia. The neuroprotective effect of lutein is evidenced by the reduction of free radicals and the simultaneous strengthening of the endogenous antioxidant systems by activating the NRF-2/ERK/AKT pathway. Further, it effectively suppressed mitochondrial aberrations, excitotoxicity, overaccumulation of metals, and its resultant complications. The immunomodulatory activity of lutein prevents neuroinflammation by hindering NF-κB nuclear translocation, regulation of NIK/IKK, PI3K/AKT, MAPK/ERK, JNK pathways, and ICAM-1 downregulation. Lutein also rescued the dysregulated cholinergic system and resolved memory defects. Along with its neuroprotective properties, lutein also improved neuroplasticity by enabling neurogenesis through increased GAP-43, NCAM, and BDNF levels.</div></div><div><h3>Conclusion</h3><div>Lutein exhibits strong neuroprotective activities against various NDDs. Though the investigations are in the exploratory phase, this review presents the consolidation of scattered evidence of the neuroprotective properties of lutein and urges its further exploration in clinical studies.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"Article 156185"},"PeriodicalIF":6.7,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626433","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Phytomedicine
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1