首页 > 最新文献

Phytomedicine最新文献

英文 中文
Modified Shenqi Dihuang Decoction inhibits prostate cancer metastasis by disrupting TCA cycle energy metabolism via NF-kB/p65-mediated OGDH regulation.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-20 DOI: 10.1016/j.phymed.2025.156405
Tongtong Zhang, Jixiang Yuan, Xiran Ju, Jielong Zhou, Xinyu Zhai, Chuanmin Chu, Mingyue Tan, Guanqun Ju, Jianyi Gu, Dongliang Xu

Background: Prostate cancer (PCa) is a significant malignancy in men, particularly challenging in the metastatic stage due to poor prognosis and limited treatment efficacy. Traditional Chinese Medicine, particularly Modified Shenqi Dihuang Decoction (MSDD), has demonstrated promise in inhibiting PCa metastasis, although its mechanisms remain unclear.

Methods: The efficacy of MSDD was evaluated using migration assays and a nude mouse model. Metabolomics was employed to identify the biological processes affected by MSDD. Systematic pharmacology, bioinformatics, and molecular dynamics were utilized to determine direct action targets of MSDD. Additionally, luciferase reporter assays, ChIP-qPCR, and gene editing were applied to elucidate the pharmacological mechanisms.

Results: MSDD effectively inhibited prostate metastasis both in vivo and in vitro, without significant adverse events reported. Metabolomics and molecular biology experiments indicated that MSDD transcriptionally represses OGDH, affecting energy metabolism associated with the tricarboxylic acid cycle (TCA) in PCa. The active components of MSDD were found to potentially bind to the transcription factor RELA (NF-kB-p65), and further experiments demonstrated that RELA regulates OGDH transcription. Further experiments revealed that the anti-metastatic effects of MSDD are RELA-dependent, indicating the crucial role of the NF-kB/OGDH axis in this process.

Conclusions: These findings support the clinical use of MSDD in metastatic PCa, emphasizing its potential to address current treatment gaps. The identified NF-kB/OGDH-dependent mechanism not only underpins MSDD's anti-metastatic effects but also reflects OGDH as a potential therapeutic target. Further research into the role of TCA in PCa progression is imperative.

{"title":"Modified Shenqi Dihuang Decoction inhibits prostate cancer metastasis by disrupting TCA cycle energy metabolism via NF-kB/p65-mediated OGDH regulation.","authors":"Tongtong Zhang, Jixiang Yuan, Xiran Ju, Jielong Zhou, Xinyu Zhai, Chuanmin Chu, Mingyue Tan, Guanqun Ju, Jianyi Gu, Dongliang Xu","doi":"10.1016/j.phymed.2025.156405","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156405","url":null,"abstract":"<p><strong>Background: </strong>Prostate cancer (PCa) is a significant malignancy in men, particularly challenging in the metastatic stage due to poor prognosis and limited treatment efficacy. Traditional Chinese Medicine, particularly Modified Shenqi Dihuang Decoction (MSDD), has demonstrated promise in inhibiting PCa metastasis, although its mechanisms remain unclear.</p><p><strong>Methods: </strong>The efficacy of MSDD was evaluated using migration assays and a nude mouse model. Metabolomics was employed to identify the biological processes affected by MSDD. Systematic pharmacology, bioinformatics, and molecular dynamics were utilized to determine direct action targets of MSDD. Additionally, luciferase reporter assays, ChIP-qPCR, and gene editing were applied to elucidate the pharmacological mechanisms.</p><p><strong>Results: </strong>MSDD effectively inhibited prostate metastasis both in vivo and in vitro, without significant adverse events reported. Metabolomics and molecular biology experiments indicated that MSDD transcriptionally represses OGDH, affecting energy metabolism associated with the tricarboxylic acid cycle (TCA) in PCa. The active components of MSDD were found to potentially bind to the transcription factor RELA (NF-kB-p65), and further experiments demonstrated that RELA regulates OGDH transcription. Further experiments revealed that the anti-metastatic effects of MSDD are RELA-dependent, indicating the crucial role of the NF-kB/OGDH axis in this process.</p><p><strong>Conclusions: </strong>These findings support the clinical use of MSDD in metastatic PCa, emphasizing its potential to address current treatment gaps. The identified NF-kB/OGDH-dependent mechanism not only underpins MSDD's anti-metastatic effects but also reflects OGDH as a potential therapeutic target. Further research into the role of TCA in PCa progression is imperative.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156405"},"PeriodicalIF":6.7,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029337","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
Syringin inhibits the crosstalk between macrophages and fibroblast-like synoviocytes to treat rheumatoid arthritis via PDE4.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-18 DOI: 10.1016/j.phymed.2025.156401
Shan Cong, Ning Wang, Huan Pei, Zixuan Li, Yan Meng, Saimire Maimaitituersun, Xue Zhao, Rong Wan, Qianqian Wan, Li Luo, Yuhong Bian, Weibo Wen, Huantian Cui

Background: Syringin (SRG) is well-known for its anti-inflammatory effects. However, its pharmacological mechanisms against rheumatoid arthritis (RA) are not fully understood.

Materials and methods: We assessed the anti-RA effects of SRG using a collagen-induced arthritis (CIA) rat model. And, we employed single-cell RNA sequencing (scRNA-seq) to analyze the changes in cell types and gene expression in the synovial tissues. Building on these observations, we investigated the effects of SRG on M1 macrophage polarization and RA-fibroblast-like synoviocytes (FLS) proliferation.

Results: Our findings highlighted the anti-RA effects of SRG on CIA rat. scRNA-seq of rat synovial tissues revealed significant changes in M1 and RA-FLS. Specifically, SRG decreased the levels of inflammatory factors in the supernatants of LPS and IFN-γ induced THP-1 cells and downregulated M1-polarized markers in these cells. Further analysis indicated that SRG's regulation of phosphodiesterase 4 (PDE4) and its associated factors was crucial for its anti-M1 polarization effects. Besides, we found that SRG inhibited the activation of FLS in vivo but showed no direct effects on RA-FLS in vitro. However, in RA-FLS, co-cultured with supernatant from SRG-treated M1-polarized THP-1 cells exhibited lower ability of cell proliferation and activation as compared to co-cultured with supernatant from M1-polarized THP-1 cells.

Conclusion: By integrating scRNA-seq analysis with in vivo and in vitro validations, our study revealed that SRG achieved its anti-RA effects by blocking the interaction between macrophages and RA-FLS, with PDE4 playing a central role. This study may provide a novel research paradigm in studying the multi-cell regulatory mechanisms of natural compounds.

{"title":"Syringin inhibits the crosstalk between macrophages and fibroblast-like synoviocytes to treat rheumatoid arthritis via PDE4.","authors":"Shan Cong, Ning Wang, Huan Pei, Zixuan Li, Yan Meng, Saimire Maimaitituersun, Xue Zhao, Rong Wan, Qianqian Wan, Li Luo, Yuhong Bian, Weibo Wen, Huantian Cui","doi":"10.1016/j.phymed.2025.156401","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156401","url":null,"abstract":"<p><strong>Background: </strong>Syringin (SRG) is well-known for its anti-inflammatory effects. However, its pharmacological mechanisms against rheumatoid arthritis (RA) are not fully understood.</p><p><strong>Materials and methods: </strong>We assessed the anti-RA effects of SRG using a collagen-induced arthritis (CIA) rat model. And, we employed single-cell RNA sequencing (scRNA-seq) to analyze the changes in cell types and gene expression in the synovial tissues. Building on these observations, we investigated the effects of SRG on M1 macrophage polarization and RA-fibroblast-like synoviocytes (FLS) proliferation.</p><p><strong>Results: </strong>Our findings highlighted the anti-RA effects of SRG on CIA rat. scRNA-seq of rat synovial tissues revealed significant changes in M1 and RA-FLS. Specifically, SRG decreased the levels of inflammatory factors in the supernatants of LPS and IFN-γ induced THP-1 cells and downregulated M1-polarized markers in these cells. Further analysis indicated that SRG's regulation of phosphodiesterase 4 (PDE4) and its associated factors was crucial for its anti-M1 polarization effects. Besides, we found that SRG inhibited the activation of FLS in vivo but showed no direct effects on RA-FLS in vitro. However, in RA-FLS, co-cultured with supernatant from SRG-treated M1-polarized THP-1 cells exhibited lower ability of cell proliferation and activation as compared to co-cultured with supernatant from M1-polarized THP-1 cells.</p><p><strong>Conclusion: </strong>By integrating scRNA-seq analysis with in vivo and in vitro validations, our study revealed that SRG achieved its anti-RA effects by blocking the interaction between macrophages and RA-FLS, with PDE4 playing a central role. This study may provide a novel research paradigm in studying the multi-cell regulatory mechanisms of natural compounds.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156401"},"PeriodicalIF":6.7,"publicationDate":"2025-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143024368","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
Astragaloside IV inhibits retinal pigment epithelial cell senescence and reduces IL-1β mRNA stability by targeting FTO-mediated m6A methylation.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-18 DOI: 10.1016/j.phymed.2025.156408
Si-Wei Wang, Ping Li, Shi-Yu Liu, De-Lian Huang, Si-Jia Zhang, Xi-Xi Zeng, Tian Lan, Kai-Li Mao, Yuan Gao, Yi-Fan Cheng, Qing Shen, Ye-Ping Ruan, Zhu-Jun Mao

Background: Resistance to senescence in retinal pigment epithelial (RPE) cells can delay the progression of age-related macular degeneration (AMD). However, the mechanisms underlying RPE cell senescence remain inadequately understood, and effective therapeutic strategies are lacking. While astragaloside IV (Ast) has demonstrated anti-aging properties, its specific effects on RPE cell senescence and potential mechanisms are not yet fully clarified.

Purpose: This study aimed to explore the impacts of Ast on RPE cell senescence and to uncover the molecular mechanisms involved.

Methods: The therapeutic efficacy of Ast was assessed using sodium iodate (NaIO3)-induced adult retinal pigment epithelial cell line 19 (ARPE-19) cell models and an AMD mouse model. To investigate the mechanisms by which Ast mitigated RPE cell senescence, RNA sequencing (RNA-seq), drug affinity responsive target stability-mass spectrometry (DARTS-MS), cellular thermal shift assay (CETSA), reverse transcription quantitative PCR (RT-qPCR), as well as western blotting were conducted.

Results: Ast significantly inhibited NaIO3-treated ARPE-19 cell senescence and protected against NaIO3-induced AMD in mice. RNA-seq analysis revealed that Ast significantly attenuated inflammation-related signaling pathways and reduced the mRNA levels of interleukin-1 beta (IL-1β). Specifically, Ast decreased the stability of IL-1β mRNA while enhancing its N6-methyladenosine (m6A) methylation. Furthermore, Ast directly interacted with fat mass and obesity-associated protein (FTO). Knockdown or pharmacological inhibition of FTO mitigated the senescence and IL-1β expression in NaIO3-treated ARPE-19 cells. FTO was essential for Ast to inhibit cellular senescence and IL-1β expression. Additionally, inhibition or knockdown of FTO conferred also provided resistance to AMD in the murine model.

Conclusion: Our results indicated that Ast significantly attenuated RPE cell senescence and showed anti-AMD properties. FTO was demonstrated to be a promising therapeutic target for AMD treatment. These findings may provide a deeper understanding of the molecular mechanisms underlying RPE cell senescence in AMD and offer potential strategies for its prevention and management.

{"title":"Astragaloside IV inhibits retinal pigment epithelial cell senescence and reduces IL-1β mRNA stability by targeting FTO-mediated m<sup>6</sup>A methylation.","authors":"Si-Wei Wang, Ping Li, Shi-Yu Liu, De-Lian Huang, Si-Jia Zhang, Xi-Xi Zeng, Tian Lan, Kai-Li Mao, Yuan Gao, Yi-Fan Cheng, Qing Shen, Ye-Ping Ruan, Zhu-Jun Mao","doi":"10.1016/j.phymed.2025.156408","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156408","url":null,"abstract":"<p><strong>Background: </strong>Resistance to senescence in retinal pigment epithelial (RPE) cells can delay the progression of age-related macular degeneration (AMD). However, the mechanisms underlying RPE cell senescence remain inadequately understood, and effective therapeutic strategies are lacking. While astragaloside IV (Ast) has demonstrated anti-aging properties, its specific effects on RPE cell senescence and potential mechanisms are not yet fully clarified.</p><p><strong>Purpose: </strong>This study aimed to explore the impacts of Ast on RPE cell senescence and to uncover the molecular mechanisms involved.</p><p><strong>Methods: </strong>The therapeutic efficacy of Ast was assessed using sodium iodate (NaIO<sub>3</sub>)-induced adult retinal pigment epithelial cell line 19 (ARPE-19) cell models and an AMD mouse model. To investigate the mechanisms by which Ast mitigated RPE cell senescence, RNA sequencing (RNA-seq), drug affinity responsive target stability-mass spectrometry (DARTS-MS), cellular thermal shift assay (CETSA), reverse transcription quantitative PCR (RT-qPCR), as well as western blotting were conducted.</p><p><strong>Results: </strong>Ast significantly inhibited NaIO<sub>3</sub>-treated ARPE-19 cell senescence and protected against NaIO<sub>3</sub>-induced AMD in mice. RNA-seq analysis revealed that Ast significantly attenuated inflammation-related signaling pathways and reduced the mRNA levels of interleukin-1 beta (IL-1β). Specifically, Ast decreased the stability of IL-1β mRNA while enhancing its N6-methyladenosine (m<sup>6</sup>A) methylation. Furthermore, Ast directly interacted with fat mass and obesity-associated protein (FTO). Knockdown or pharmacological inhibition of FTO mitigated the senescence and IL-1β expression in NaIO<sub>3</sub>-treated ARPE-19 cells. FTO was essential for Ast to inhibit cellular senescence and IL-1β expression. Additionally, inhibition or knockdown of FTO conferred also provided resistance to AMD in the murine model.</p><p><strong>Conclusion: </strong>Our results indicated that Ast significantly attenuated RPE cell senescence and showed anti-AMD properties. FTO was demonstrated to be a promising therapeutic target for AMD treatment. These findings may provide a deeper understanding of the molecular mechanisms underlying RPE cell senescence in AMD and offer potential strategies for its prevention and management.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156408"},"PeriodicalIF":6.7,"publicationDate":"2025-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029332","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
β-Sitosterol modulates osteogenic and adipogenic balance in BMSCs to suppress osteoporosis via regulating mTOR-IMP1-Adipoq axis.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-17 DOI: 10.1016/j.phymed.2025.156400
Hao Li, Ying Gong, Yanna Wang, Wanyu Sang, Changyuan Wang, Yukun Zhang, Hanrui Zhang, Peixuan Liu, Mozhen Liu, Huijun Sun

Background: Osteoporosis (OP) is a prevalent global health concern, impacting millions of individuals, especially the elderly. The etiology of senile OP is associated with the imbalance of osteogenic and adipogenic differentiation in the bone marrow mesenchymal stem cells (BMSCs). The imbalance of BMSCs differentiation fate will leading to bone loss and lipids accumulation. β-sitosterol, a naturally occurring phytosterol which is abundant in plants and has a similar structure to cholesterol, demonstrates diverse bioactivities, including lipid-lowering effect and osteogenesis-inducing effects. These effects indicate that β-sitosterol might have anti-OP effects. Nevertheless, the precise mechanism underlying β-sitosterol's anti-osteoporotic efficacy via modulating BMSCs differentiation fate remains obscure.

Purpose: This study endeavors to elucidate whether β-sitosterol has the potential to augment the osteogenic differentiation of BMSCs while mitigating their adipogenic differentiation, thereby exerting an anti-OP effect; and to reveal its molecular mechanisms of action.

Methods: In this study, a dosage form HP-β-cyclodextrin-coated β-sitosterol was developed for intragastric administration in mice to enhancing its bioavailability. Subsequently by using an integrative approach encompassing bioinformatics, computer molecular simulations, high-throughput sequencing, and in vitro/vivo as well as in-tube experiments, we investigated the anti-osteoporotic and bone healing effects of β-sitosterol and delineated its underlying mechanisms.

Results: Our findings demonstrate that β-sitosterol exhibits anti-osteoporotic and bone healing effects both in vitro and in vivo by modulating the osteogenic and adipogenic differentiation of BMSCs. Mechanistically, these effects are mediated through the direct inhibition of mTOR's kinase activity independent of mediating autophagy, leading to the suppression of the mTOR-IMP1-Adipoq axis in BMSCs.

Conclusion: These results unveil β-sitosterol as a promising therapeutic agent for OP, shedding light on its underlying mechanisms. This research contributes potential candidates for diagnostic and therapeutic interventions in the realm of OP.

{"title":"β-Sitosterol modulates osteogenic and adipogenic balance in BMSCs to suppress osteoporosis via regulating mTOR-IMP1-Adipoq axis.","authors":"Hao Li, Ying Gong, Yanna Wang, Wanyu Sang, Changyuan Wang, Yukun Zhang, Hanrui Zhang, Peixuan Liu, Mozhen Liu, Huijun Sun","doi":"10.1016/j.phymed.2025.156400","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156400","url":null,"abstract":"<p><strong>Background: </strong>Osteoporosis (OP) is a prevalent global health concern, impacting millions of individuals, especially the elderly. The etiology of senile OP is associated with the imbalance of osteogenic and adipogenic differentiation in the bone marrow mesenchymal stem cells (BMSCs). The imbalance of BMSCs differentiation fate will leading to bone loss and lipids accumulation. β-sitosterol, a naturally occurring phytosterol which is abundant in plants and has a similar structure to cholesterol, demonstrates diverse bioactivities, including lipid-lowering effect and osteogenesis-inducing effects. These effects indicate that β-sitosterol might have anti-OP effects. Nevertheless, the precise mechanism underlying β-sitosterol's anti-osteoporotic efficacy via modulating BMSCs differentiation fate remains obscure.</p><p><strong>Purpose: </strong>This study endeavors to elucidate whether β-sitosterol has the potential to augment the osteogenic differentiation of BMSCs while mitigating their adipogenic differentiation, thereby exerting an anti-OP effect; and to reveal its molecular mechanisms of action.</p><p><strong>Methods: </strong>In this study, a dosage form HP-β-cyclodextrin-coated β-sitosterol was developed for intragastric administration in mice to enhancing its bioavailability. Subsequently by using an integrative approach encompassing bioinformatics, computer molecular simulations, high-throughput sequencing, and in vitro/vivo as well as in-tube experiments, we investigated the anti-osteoporotic and bone healing effects of β-sitosterol and delineated its underlying mechanisms.</p><p><strong>Results: </strong>Our findings demonstrate that β-sitosterol exhibits anti-osteoporotic and bone healing effects both in vitro and in vivo by modulating the osteogenic and adipogenic differentiation of BMSCs. Mechanistically, these effects are mediated through the direct inhibition of mTOR's kinase activity independent of mediating autophagy, leading to the suppression of the mTOR-IMP1-Adipoq axis in BMSCs.</p><p><strong>Conclusion: </strong>These results unveil β-sitosterol as a promising therapeutic agent for OP, shedding light on its underlying mechanisms. This research contributes potential candidates for diagnostic and therapeutic interventions in the realm of OP.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156400"},"PeriodicalIF":6.7,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029370","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
Exploring the therapeutic potential of Abelmoschi Corolla in psoriasis: Mechanisms of action and inflammatory pathway disruption.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-17 DOI: 10.1016/j.phymed.2025.156379
Baoquan Qu, Guanglu Li, Ning Zhao, Ruonan Li, Huike Ma, Haoyue Zhu, Ping Li, Jingxia Zhao

Background: Psoriasis is a prevalent chronic inflammatory skin condition for which existing treatments often fall short of fully addressing patient needs. Abelmoschi Corolla (AC), a traditional Chinese medicine, and its ethanol extract, huangkui capsule, are well established for the treatment of chronic kidney diseases. The therapeutic mechanisms of AC include anti-inflammatory effects and immune modulation, which align with psoriasis treatment strategies. Nevertheless, the potential of AC as a therapeutic agent for psoriasis remains unexplored.

Purpose: This study aimed to evaluate the efficacy of AC in treating psoriasis and, if effective, to elucidate the underlying mechanisms by which AC exerts its therapeutic effects.

Methods: To assess the therapeutic potential of AC, an imiquimod-induced psoriasis-like mouse model was utilized. Bioinformatics and machine learning approaches were employed to predict the targets and mechanisms of AC in psoriasis. Further validation was performed using targeted metabolite quantification, immunohistochemistry, polymerase chain reaction, and western blotting.

Results: AC treatment significantly improved psoriasis-like skin lesions, as indicated by enhancements in appearance, PASI scores, a reduction in epidermal hyperproliferation, and decreased immune cell infiltration. Bioinformatics and machine learning analyses identified arginase 1 (ARG1) as a key target of AC in psoriasis. Experimental validation demonstrated that AC reduced ARG1 expression, arginine metabolism, and polyamine production by upregulating PP6 expression and inhibiting C/EBP-β activation in psoriatic keratinocytes, resulting in the suppression of dendritic cell infiltration and a reduction in the expression of inflammatory cytokines, including IL-23, IL-6, IL-1β, IL-17A, and TNF-α.

Conclusions: AC disrupted the inflammatory pathways associated with psoriasis and alleviated imiquimod-induced psoriasis-like skin inflammation by inhibiting ARG1 overexpression and arginine metabolism in psoriatic keratinocytes. These findings suggest that AC has significant potential as a therapeutic agent for psoriasis and warrants further research and development.

{"title":"Exploring the therapeutic potential of Abelmoschi Corolla in psoriasis: Mechanisms of action and inflammatory pathway disruption.","authors":"Baoquan Qu, Guanglu Li, Ning Zhao, Ruonan Li, Huike Ma, Haoyue Zhu, Ping Li, Jingxia Zhao","doi":"10.1016/j.phymed.2025.156379","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156379","url":null,"abstract":"<p><strong>Background: </strong>Psoriasis is a prevalent chronic inflammatory skin condition for which existing treatments often fall short of fully addressing patient needs. Abelmoschi Corolla (AC), a traditional Chinese medicine, and its ethanol extract, huangkui capsule, are well established for the treatment of chronic kidney diseases. The therapeutic mechanisms of AC include anti-inflammatory effects and immune modulation, which align with psoriasis treatment strategies. Nevertheless, the potential of AC as a therapeutic agent for psoriasis remains unexplored.</p><p><strong>Purpose: </strong>This study aimed to evaluate the efficacy of AC in treating psoriasis and, if effective, to elucidate the underlying mechanisms by which AC exerts its therapeutic effects.</p><p><strong>Methods: </strong>To assess the therapeutic potential of AC, an imiquimod-induced psoriasis-like mouse model was utilized. Bioinformatics and machine learning approaches were employed to predict the targets and mechanisms of AC in psoriasis. Further validation was performed using targeted metabolite quantification, immunohistochemistry, polymerase chain reaction, and western blotting.</p><p><strong>Results: </strong>AC treatment significantly improved psoriasis-like skin lesions, as indicated by enhancements in appearance, PASI scores, a reduction in epidermal hyperproliferation, and decreased immune cell infiltration. Bioinformatics and machine learning analyses identified arginase 1 (ARG1) as a key target of AC in psoriasis. Experimental validation demonstrated that AC reduced ARG1 expression, arginine metabolism, and polyamine production by upregulating PP6 expression and inhibiting C/EBP-β activation in psoriatic keratinocytes, resulting in the suppression of dendritic cell infiltration and a reduction in the expression of inflammatory cytokines, including IL-23, IL-6, IL-1β, IL-17A, and TNF-α.</p><p><strong>Conclusions: </strong>AC disrupted the inflammatory pathways associated with psoriasis and alleviated imiquimod-induced psoriasis-like skin inflammation by inhibiting ARG1 overexpression and arginine metabolism in psoriatic keratinocytes. These findings suggest that AC has significant potential as a therapeutic agent for psoriasis and warrants further research and development.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156379"},"PeriodicalIF":6.7,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143041199","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
Qige Decoction attenuated non-alcoholic fatty liver disease through regulating SIRT6-PPARα-mediated fatty acid oxidation.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-17 DOI: 10.1016/j.phymed.2025.156395
Simin Fan, Wei Chen, Yanfang Li, Kaixin Guo, Hui Tang, Jintong Ye, Zunming Zhou, Meiao Tan, Haoyang Wei, Xiwen Huang, Keer Huang, Xuehong Ke

Background: Sirtuin 6 (SIRT6), a potential therapeutic target for non-alcoholic fatty liver disease (NAFLD), has been shown to regulate fatty acid oxidation (FAO) by interacting with peroxisome proliferator-activated receptor α (PPARα). However, the impact of SIRT6-PPARα pathway on NAFLD phenotype has not yet been reported. Qige decoction (QG), a traditional Chinese medicine (TCM) formula, is widely applied to treat disorders of glycolipid metabolism. Our previous experiments showed that QG reduced hepatic steatosis and provided preliminary evidence that QG may promote FAO. However, a thorough understanding of molecular mechanisms by which QG regulates FAO requires further investigation.

Purpose: To investigate the role of SIRT6-PPARα signalling pathway on NAFLD phenotype and explore the mechanism by which QG improves NAFLD and its relationship with FAO regulated by SIRT6-PPARα signalling pathway.

Methods: In vivo study, NAFLD mice induced by high fat diet (HFD) were divided into two parts. The first part involved four groups: control (CON), model (MOD), PPARα agonist (WY-14,643, WY), and SIRT6 inhibitor (OSS-128,167, OS) groups. The second part involved five groups: CON group, MOD group, positive drug (POS) group, low dose QG (QGL) group, and high dose QG (QGH) group. Widely-targeted lipidomic were performed by UHPLC-QTOF/MS to analyse differential lipids (DELs) in the liver, while differentially expressed genes (DEGs) were analysed by transcriptome analysis on the Illumina sequencing platform. In vitro study, co-immunoprecipitation and dual luciferase assay were employed to further identify the molecular mechanisms of SIRT6-PPARα interaction. The lentiviral vector, TG assay, and acetyl-CoA assay were used to clarify the indispensable role of the SIRT6-PPARα signalling pathway on QG amelioration of lipid accumulation in vitro.

Results: Down-regulation of SIRT6 inhibited PPARα-mediated FAO and aggravated lipid accumulation in hepatocytes both in vivo and in vitro. SIRT6 bound to PPARα in HepG2 cells; however, SIRT6 activation of the PPARα promoter was not detected. Along with QG reduced hepatocyte lipid accumulation, SIRT6-PPARα signalling pathway was upregulated in vivo and in vitro. However, the alleviating effect of QG on lipid accumulation was blocked by SIRT6 silencing in vitro.

Conclusion: This study verified that SIRT6-PPARα signalling pathway inhibition exacerbated NAFLD dyslipidaemia and hepatic steatosis. In addition, this study provided the first in-depth analysis of the molecular mechanisms by which QG ameliorates NFALD, involving promotion of FAO through activation of the SIRT6-PPARα signalling pathway. Our study offers significant insights for the clinical application of QG.

{"title":"Qige Decoction attenuated non-alcoholic fatty liver disease through regulating SIRT6-PPARα-mediated fatty acid oxidation.","authors":"Simin Fan, Wei Chen, Yanfang Li, Kaixin Guo, Hui Tang, Jintong Ye, Zunming Zhou, Meiao Tan, Haoyang Wei, Xiwen Huang, Keer Huang, Xuehong Ke","doi":"10.1016/j.phymed.2025.156395","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156395","url":null,"abstract":"<p><strong>Background: </strong>Sirtuin 6 (SIRT6), a potential therapeutic target for non-alcoholic fatty liver disease (NAFLD), has been shown to regulate fatty acid oxidation (FAO) by interacting with peroxisome proliferator-activated receptor α (PPARα). However, the impact of SIRT6-PPARα pathway on NAFLD phenotype has not yet been reported. Qige decoction (QG), a traditional Chinese medicine (TCM) formula, is widely applied to treat disorders of glycolipid metabolism. Our previous experiments showed that QG reduced hepatic steatosis and provided preliminary evidence that QG may promote FAO. However, a thorough understanding of molecular mechanisms by which QG regulates FAO requires further investigation.</p><p><strong>Purpose: </strong>To investigate the role of SIRT6-PPARα signalling pathway on NAFLD phenotype and explore the mechanism by which QG improves NAFLD and its relationship with FAO regulated by SIRT6-PPARα signalling pathway.</p><p><strong>Methods: </strong>In vivo study, NAFLD mice induced by high fat diet (HFD) were divided into two parts. The first part involved four groups: control (CON), model (MOD), PPARα agonist (WY-14,643, WY), and SIRT6 inhibitor (OSS-128,167, OS) groups. The second part involved five groups: CON group, MOD group, positive drug (POS) group, low dose QG (QGL) group, and high dose QG (QGH) group. Widely-targeted lipidomic were performed by UHPLC-QTOF/MS to analyse differential lipids (DELs) in the liver, while differentially expressed genes (DEGs) were analysed by transcriptome analysis on the Illumina sequencing platform. In vitro study, co-immunoprecipitation and dual luciferase assay were employed to further identify the molecular mechanisms of SIRT6-PPARα interaction. The lentiviral vector, TG assay, and acetyl-CoA assay were used to clarify the indispensable role of the SIRT6-PPARα signalling pathway on QG amelioration of lipid accumulation in vitro.</p><p><strong>Results: </strong>Down-regulation of SIRT6 inhibited PPARα-mediated FAO and aggravated lipid accumulation in hepatocytes both in vivo and in vitro. SIRT6 bound to PPARα in HepG2 cells; however, SIRT6 activation of the PPARα promoter was not detected. Along with QG reduced hepatocyte lipid accumulation, SIRT6-PPARα signalling pathway was upregulated in vivo and in vitro. However, the alleviating effect of QG on lipid accumulation was blocked by SIRT6 silencing in vitro.</p><p><strong>Conclusion: </strong>This study verified that SIRT6-PPARα signalling pathway inhibition exacerbated NAFLD dyslipidaemia and hepatic steatosis. In addition, this study provided the first in-depth analysis of the molecular mechanisms by which QG ameliorates NFALD, involving promotion of FAO through activation of the SIRT6-PPARα signalling pathway. Our study offers significant insights for the clinical application of QG.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156395"},"PeriodicalIF":6.7,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143041231","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
Puerarin mitigates cognitive decline and white matter injury via CD36-Mediated microglial phagocytosis in chronic cerebral hypoperfusion. 葛根素通过cd36介导的小胶质细胞吞噬减轻慢性脑灌注不足患者的认知能力下降和白质损伤。
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-16 DOI: 10.1016/j.phymed.2025.156396
Qinghua Luo, Fang Li, Xu Liu, Tengfeng Yan, Li Yang, Wenping Zhu, Heqing Zheng, Yan Li, Jianglong Tu, Xingen Zhu

Background: Chronic cerebral hypoperfusion (CCH) contributes significantly to white matter injury (WMI) and cognitive impairment, often leading to vascular dementia (VaD). Inefficient clearance of myelin debris by microglia impedes white matter repair, making microglia-mediated myelin clearance a promising therapeutic strategy for WMI. Puerarin (Pu), an isoflavonoid monomer from Pueraria lobata, is known for its neuroprotective, anti-inflammatory, and immunoregulatory properties. However, its effects and underlying mechanisms in counteracting CCH-induced damage remain unclear. In this study, we aimed to investigate the therapeutic effects and underlying mechanisms of puerarin in a CCH mouse model.

Methods: Right unilateral common carotid artery occlusion (rUCCAO) was used to model CCH in C57BL/6J mice. Puerarin (400 mg/kg/day) was administered intraperitoneally for 10 consecutive days starting immediately post-surgery. Cognitive function was assessed by the Morris Water Maze (MWM) test. WMI, remyelination, neuroinflammation, and microglial phagocytosis were evaluated by western blotting, immunofluorescence staining, RT-PCR, or flow cytometry both in vivo and in vitro.

Results: Puerarin treatment significantly improved cognitive performance and mitigated WMI in rUCCAO mice. These effects were associated with enhanced microglial phagocytosis and remyelination, reduced neuroinflammation, and increased CD36 expression. Additionally, puerarin also increased the levels of IL-10 and phosphorylated STAT3 (p-STAT3) in brain tissues. Notably, IL-10 neutralization reversed these benefits effects by reducing microglial myelin debris uptake, downregulating STAT3 phosphorylation and CD36 expression.

Conclusions: Our findings demonstrate that puerarin has significant therapeutic potential in treating CCH-related cognitive impairments and WMI by modulating CD36-mediated microglial myelin clearance through the IL-10/STAT3 pathway. However, our study was reliant on preclinical animal models, further studies are needed to explore applicability in human subjects.

背景:慢性脑灌注不足(CCH)是脑白质损伤(WMI)和认知障碍的重要因素,常导致血管性痴呆(VaD)。小胶质细胞对髓磷脂碎片的低效清除阻碍了白质的修复,这使得小胶质细胞介导的髓磷脂清除成为治疗WMI的一种有希望的治疗策略。葛根素(Pu)是一种来自葛根的类黄酮单体,以其神经保护、抗炎和免疫调节特性而闻名。然而,其在对抗cch诱导的损伤中的作用和潜在机制尚不清楚。在本研究中,我们旨在探讨葛根素对CCH小鼠模型的治疗作用及其机制。方法:采用右侧单侧颈总动脉闭塞(rUCCAO)模型建立C57BL/6J小鼠颈总动脉闭塞模型。术后立即腹腔注射葛根素(400mg /kg/天),连续10天。采用Morris水迷宫(Morris Water Maze, MWM)测试认知功能。采用western blotting、免疫荧光染色、RT-PCR或流式细胞术在体内和体外评估WMI、髓鞘再生、神经炎症和小胶质细胞吞噬。结果:葛根素治疗可显著改善rUCCAO小鼠的认知能力,减轻WMI。这些作用与增强的小胶质细胞吞噬和髓鞘再生、减少的神经炎症和增加的CD36表达有关。此外,葛根素还增加了脑组织中IL-10和磷酸化STAT3 (p-STAT3)的水平。值得注意的是,IL-10中和通过减少小胶质髓鞘碎片摄取、下调STAT3磷酸化和CD36表达逆转了这些益处。结论:我们的研究结果表明,葛根素通过IL-10/STAT3通路调节cd36介导的小胶质髓鞘清除,在治疗cch相关认知障碍和WMI方面具有显著的治疗潜力。然而,我们的研究依赖于临床前动物模型,需要进一步的研究来探索在人类受试者中的适用性。
{"title":"Puerarin mitigates cognitive decline and white matter injury via CD36-Mediated microglial phagocytosis in chronic cerebral hypoperfusion.","authors":"Qinghua Luo, Fang Li, Xu Liu, Tengfeng Yan, Li Yang, Wenping Zhu, Heqing Zheng, Yan Li, Jianglong Tu, Xingen Zhu","doi":"10.1016/j.phymed.2025.156396","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156396","url":null,"abstract":"<p><strong>Background: </strong>Chronic cerebral hypoperfusion (CCH) contributes significantly to white matter injury (WMI) and cognitive impairment, often leading to vascular dementia (VaD). Inefficient clearance of myelin debris by microglia impedes white matter repair, making microglia-mediated myelin clearance a promising therapeutic strategy for WMI. Puerarin (Pu), an isoflavonoid monomer from Pueraria lobata, is known for its neuroprotective, anti-inflammatory, and immunoregulatory properties. However, its effects and underlying mechanisms in counteracting CCH-induced damage remain unclear. In this study, we aimed to investigate the therapeutic effects and underlying mechanisms of puerarin in a CCH mouse model.</p><p><strong>Methods: </strong>Right unilateral common carotid artery occlusion (rUCCAO) was used to model CCH in C57BL/6J mice. Puerarin (400 mg/kg/day) was administered intraperitoneally for 10 consecutive days starting immediately post-surgery. Cognitive function was assessed by the Morris Water Maze (MWM) test. WMI, remyelination, neuroinflammation, and microglial phagocytosis were evaluated by western blotting, immunofluorescence staining, RT-PCR, or flow cytometry both in vivo and in vitro.</p><p><strong>Results: </strong>Puerarin treatment significantly improved cognitive performance and mitigated WMI in rUCCAO mice. These effects were associated with enhanced microglial phagocytosis and remyelination, reduced neuroinflammation, and increased CD36 expression. Additionally, puerarin also increased the levels of IL-10 and phosphorylated STAT3 (p-STAT3) in brain tissues. Notably, IL-10 neutralization reversed these benefits effects by reducing microglial myelin debris uptake, downregulating STAT3 phosphorylation and CD36 expression.</p><p><strong>Conclusions: </strong>Our findings demonstrate that puerarin has significant therapeutic potential in treating CCH-related cognitive impairments and WMI by modulating CD36-mediated microglial myelin clearance through the IL-10/STAT3 pathway. However, our study was reliant on preclinical animal models, further studies are needed to explore applicability in human subjects.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156396"},"PeriodicalIF":6.7,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010250","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
Tiaogeng decoction improves mild cognitive impairment in menopausal APP/PS1 mice through the ERs/NF-κ b/AQP1 signaling pathway.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-16 DOI: 10.1016/j.phymed.2025.156391
Xuan-Ling Li, Zhi-Heng Lin, Si-Ru Chen, Shuang Ni, Guang-Yao Lin, Wei Wang, Jing-Yu Lin, Qian Zhao, Chao Cong, Lian-Wei Xu
<p><p>People with mild cognitive impairment (MCI) carry a considerable risk of developing dementia. Studies have shown that female sex hormones have long-lasting neuroprotective and anti-aging properties, and the increased risk of MCI and AD is associated with the lack of estrogen during menopause. Previous studies have shown that Tiao Geng Decoction (TGD) may have antioxidant and anti apoptotic properties, which may prevent neurodegenerative diseases. However, whether TGD is effective in improving mild cognitive impairment due to postmenopausal estrogen deficiency and its potential pharmacological mechanisms remain unclear. The aim of this study was to investigate the possible pharmacological mechanisms of TGD in preventing postmenopausal MCI. We utilized RNA-seq technology to screen for differentially expressed genes (DEGs) and enrichment pathways in the hippocampal tissue of different groups of mice. Additionally, we adopted single-cell sequencing technology to study the cell types of Alzheimer's disease (AD) group and Normal Control (NC) group, the differential marker genes of each cell subgroup, and the GO enrichment analysis of each cell type. Both RNA sequencing and single-cell sequencing results showed a significant correlation between TGD and NF-κb pathway in improving mild cognitive impairment in postmenopausal women. The experimental verification results showed that the spatial learning and memory abilities of APP/PS1 model mice were weakened after ovariectomy, and the reproductive cycle on vaginal smears was in the interphase of diestrus. The levels of serum E2, and P-tau181 in mice were significantly down regulated, while the levels of brain tissue homogenate A β 42, IL-1 β, and IL-18 were significantly up-regulated, indicating successful modeling. Combining Western blotting, RT-qPCR, and transmission electron microscopy analyses, it was found that the low estrogen environment induced by oophorectomy can activate the NF-κb signaling pathway, activate the expression of NLRP3 inflammasome and A β secretase BACE1, and induce neuroinflammatory damage in hippocampal astrocytes. These results conform to the modeling characteristics of MCI. After TGD intervention, the spatial learning and memory abilities of MCI mice were significantly improved. The pharmacological validation results indicated that high concentration doses of TGD had a more significant effect on MCI. Subsequently, we used high concentration TGD (0.32 g/ml) as the traditional Chinese medicine group for further validation, protein blotting and RT-qPCR results indicated that TGD can effectively stimulate the secretion of ER α and ER β, inhibit the NF-κb pathway, downregulate BACE1, and inhibit the expression of NLRP3 inflammasome related proteins. In addition, the immunofluorescence results of hippocampal astrocytes showed that TGD can effectively facilitate the expression of AQP1 and significantly lower the sedimentation of A β compared with the model group. Our research sugg
{"title":"Tiaogeng decoction improves mild cognitive impairment in menopausal APP/PS1 mice through the ERs/NF-κ b/AQP1 signaling pathway.","authors":"Xuan-Ling Li, Zhi-Heng Lin, Si-Ru Chen, Shuang Ni, Guang-Yao Lin, Wei Wang, Jing-Yu Lin, Qian Zhao, Chao Cong, Lian-Wei Xu","doi":"10.1016/j.phymed.2025.156391","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156391","url":null,"abstract":"&lt;p&gt;&lt;p&gt;People with mild cognitive impairment (MCI) carry a considerable risk of developing dementia. Studies have shown that female sex hormones have long-lasting neuroprotective and anti-aging properties, and the increased risk of MCI and AD is associated with the lack of estrogen during menopause. Previous studies have shown that Tiao Geng Decoction (TGD) may have antioxidant and anti apoptotic properties, which may prevent neurodegenerative diseases. However, whether TGD is effective in improving mild cognitive impairment due to postmenopausal estrogen deficiency and its potential pharmacological mechanisms remain unclear. The aim of this study was to investigate the possible pharmacological mechanisms of TGD in preventing postmenopausal MCI. We utilized RNA-seq technology to screen for differentially expressed genes (DEGs) and enrichment pathways in the hippocampal tissue of different groups of mice. Additionally, we adopted single-cell sequencing technology to study the cell types of Alzheimer's disease (AD) group and Normal Control (NC) group, the differential marker genes of each cell subgroup, and the GO enrichment analysis of each cell type. Both RNA sequencing and single-cell sequencing results showed a significant correlation between TGD and NF-κb pathway in improving mild cognitive impairment in postmenopausal women. The experimental verification results showed that the spatial learning and memory abilities of APP/PS1 model mice were weakened after ovariectomy, and the reproductive cycle on vaginal smears was in the interphase of diestrus. The levels of serum E2, and P-tau181 in mice were significantly down regulated, while the levels of brain tissue homogenate A β 42, IL-1 β, and IL-18 were significantly up-regulated, indicating successful modeling. Combining Western blotting, RT-qPCR, and transmission electron microscopy analyses, it was found that the low estrogen environment induced by oophorectomy can activate the NF-κb signaling pathway, activate the expression of NLRP3 inflammasome and A β secretase BACE1, and induce neuroinflammatory damage in hippocampal astrocytes. These results conform to the modeling characteristics of MCI. After TGD intervention, the spatial learning and memory abilities of MCI mice were significantly improved. The pharmacological validation results indicated that high concentration doses of TGD had a more significant effect on MCI. Subsequently, we used high concentration TGD (0.32 g/ml) as the traditional Chinese medicine group for further validation, protein blotting and RT-qPCR results indicated that TGD can effectively stimulate the secretion of ER α and ER β, inhibit the NF-κb pathway, downregulate BACE1, and inhibit the expression of NLRP3 inflammasome related proteins. In addition, the immunofluorescence results of hippocampal astrocytes showed that TGD can effectively facilitate the expression of AQP1 and significantly lower the sedimentation of A β compared with the model group. Our research sugg","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156391"},"PeriodicalIF":6.7,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029363","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 metabolomics and mass spectrometry imaging analysis reveal the efficacy and mechanism of Huangkui capsule on type 2 diabetic nephropathy.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-16 DOI: 10.1016/j.phymed.2025.156397
Jinwei Han, Ping Li, Hui Sun, Ying Zheng, Chang Liu, Xiangmei Chen, Shihan Guan, Fengting Yin, Xijun Wang

Background: Huangkui capsule (HKC), a Chinese patent medicine, is clinically used for treating diabetic nephropathy. However, the core disease-specific biomarkers and targets of type 2 diabetic nephropathy (T2DN) and the therapeutic mechanism of HKC are not fully elucidated.

Purpose: This study aimed to investigate the therapeutic effects and underlying molecular mechanisms of HKC for T2DN.

Study design: The db/db mouse model was used to evaluate the efficacy of HKC for T2DN, and the core pathways regulated by HKC were studied to determine its kidney protective mechanism.

Methods: High-throughput UPLC-MS/MS and multivariate analysis were employed to analyze the serum and kidney metabolic profiles of db/db mice, identifying potential core biomarkers of T2DN. Atmospheric pressure matrix-assisted laser desorption/ionization mass spectrometry imaging was used to locate in situ spatial distribution of core biomarkers and drug active ingredients in kidney tissues. Biochemical indicators, histopathology, immunohistochemistry, immunofluorescence, molecular docking, and western blotting were combined to reveal therapeutic effects, pathways, and targets of HKC.

Results: HKC substantially improved pathological characteristics, kidney function, oxidative stress, inflammation, and lipid metabolism indicators of T2DN. Twelve core disease-specific biomarker that significantly influenced clustering were identified and its unique spatial distribution information in the kidneys was revealed. 3-dehydrosphinganine, retinyl ester, and 9-cis-retinoic acid (9cRA) could serve as novel disease-specific biomarkers for T2DN. Based on newly discovered biomarkers, quercetin, myricetin, and isorhamnetin were found to act on key enzymes SPT, ALDH1A1, AOX, LRAT, and DGAT1 in retinol and sphingolipid metabolism pathways. Western blotting showed that HKC ameliorated T2DN by targeting these enzymes, upregulating 9cRA and retinyl ester, downregulating 3-dehydrosphinganine, increasing TGF-β signal transduction, inhibiting the expression of the immune fibrosis proteins OX-8, Col-I and α-SMA, inhibiting Th17 cell development and ceramide synthesis, reducing IL-1β, TNF-α, MDA, TC, LDL-C, and TG levels, and increaseing SOD activity.

Conclusions: HKC exerts significant therapeutic effects on T2DN. HKC corrects the metabolic disorder of sphingolipids and retinol, and improves T2DN by regulating the activities of SPT, ALDH1A1, AOX, LRAT, and DGAT1. This study provides valuable ideas and new mechanistic insights for the treatment of T2DN with HKC.

{"title":"Integrated metabolomics and mass spectrometry imaging analysis reveal the efficacy and mechanism of Huangkui capsule on type 2 diabetic nephropathy.","authors":"Jinwei Han, Ping Li, Hui Sun, Ying Zheng, Chang Liu, Xiangmei Chen, Shihan Guan, Fengting Yin, Xijun Wang","doi":"10.1016/j.phymed.2025.156397","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156397","url":null,"abstract":"<p><strong>Background: </strong>Huangkui capsule (HKC), a Chinese patent medicine, is clinically used for treating diabetic nephropathy. However, the core disease-specific biomarkers and targets of type 2 diabetic nephropathy (T2DN) and the therapeutic mechanism of HKC are not fully elucidated.</p><p><strong>Purpose: </strong>This study aimed to investigate the therapeutic effects and underlying molecular mechanisms of HKC for T2DN.</p><p><strong>Study design: </strong>The db/db mouse model was used to evaluate the efficacy of HKC for T2DN, and the core pathways regulated by HKC were studied to determine its kidney protective mechanism.</p><p><strong>Methods: </strong>High-throughput UPLC-MS/MS and multivariate analysis were employed to analyze the serum and kidney metabolic profiles of db/db mice, identifying potential core biomarkers of T2DN. Atmospheric pressure matrix-assisted laser desorption/ionization mass spectrometry imaging was used to locate in situ spatial distribution of core biomarkers and drug active ingredients in kidney tissues. Biochemical indicators, histopathology, immunohistochemistry, immunofluorescence, molecular docking, and western blotting were combined to reveal therapeutic effects, pathways, and targets of HKC.</p><p><strong>Results: </strong>HKC substantially improved pathological characteristics, kidney function, oxidative stress, inflammation, and lipid metabolism indicators of T2DN. Twelve core disease-specific biomarker that significantly influenced clustering were identified and its unique spatial distribution information in the kidneys was revealed. 3-dehydrosphinganine, retinyl ester, and 9-cis-retinoic acid (9cRA) could serve as novel disease-specific biomarkers for T2DN. Based on newly discovered biomarkers, quercetin, myricetin, and isorhamnetin were found to act on key enzymes SPT, ALDH1A1, AOX, LRAT, and DGAT1 in retinol and sphingolipid metabolism pathways. Western blotting showed that HKC ameliorated T2DN by targeting these enzymes, upregulating 9cRA and retinyl ester, downregulating 3-dehydrosphinganine, increasing TGF-β signal transduction, inhibiting the expression of the immune fibrosis proteins OX-8, Col-I and α-SMA, inhibiting Th17 cell development and ceramide synthesis, reducing IL-1β, TNF-α, MDA, TC, LDL-C, and TG levels, and increaseing SOD activity.</p><p><strong>Conclusions: </strong>HKC exerts significant therapeutic effects on T2DN. HKC corrects the metabolic disorder of sphingolipids and retinol, and improves T2DN by regulating the activities of SPT, ALDH1A1, AOX, LRAT, and DGAT1. This study provides valuable ideas and new mechanistic insights for the treatment of T2DN with HKC.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156397"},"PeriodicalIF":6.7,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143041222","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
Regulation of inflammatory responses: Harnessing the Ruan Mai Jian targeting of EphA2/ephrinA1 pathway to enhance atherosclerosis amelioration.
IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-01-15 DOI: 10.1016/j.phymed.2025.156398
Xue Zhao, Hanyu Zhang, Jiayi Wang, Lu Zhang, Shuang Gao, Yun Gu, Te Liu, Wenting Du

Background: Atherosclerosis is a major contributor to global cardiovascular morbidity and mortality, driven by the chronic inflammatory proliferation of vascular smooth muscle cells (VSMCs), which destabilizes atherosclerotic plaques. The EphA2/ephrinA1 signaling pathway plays a critical role in modulating VSMC inflammatory responses, making it an attractive therapeutic target. However, the clinical application of EphA2 inhibitors remains limited due to safety concerns. Ruan Mai Jian (RMJ), a traditional Chinese herbal medicine, has demonstrated potential efficacy in treating atherosclerosis, though its precise mechanisms remain insufficiently characterized. To date, no study has investigated a Chinese medicine compound capable of regulating atherosclerotic inflammatory responses via the EphA2/ephrinA1 pathway. This study aims to determine whether RMJ treats atherosclerosis both in vivo and in vitro by modulating the EphA2/ephrinA1 pathway, while evaluating its potential hepatic and renal toxicity.

Study design: A combination of in vivo (ApoE-/- murine model) and in vitro studies was employed to investigate the effects of RMJ on atherosclerotic progression, inflammatory markers, and VSMC function.

Methods: ApoE-/- mice were fed a high-fat diet to induce atherosclerosis and subsequently treated with RMJ at varying doses. Serum lipid levels, inflammatory cytokines (TNF-α, IL-6, IL-1β), and plaque morphology were analyzed. Immunohistochemical and Western blot analyses were performed to assess the modulation of the EphA2/ephrinA1 pathway. VSMC proliferation and migration assays were conducted to evaluate the effects of RMJ on cellular behavior in vitro.

Results: RMJ treatment significantly attenuated serum lipid levels, reduced systemic inflammation, and stabilized atherosclerotic plaques by increasing collagen content and decreasing lipid deposition. RMJ downregulated EphA2 expression and upregulated ephrinA1, effectively inhibiting VSMC proliferation and migration through suppression of the AKT1/ERK1/2 signaling cascade. Importantly, no hepatic or renal toxicity was observed in treated mice, indicating a favorable safety profile.

Conclusion: RMJ demonstrates significant therapeutic potential for the treatment of atherosclerosis, primarily through modulation of the EphA2/ephrinA1 signaling pathway, resulting in reduced inflammation and VSMC proliferation. Its efficacy, combined with the absence of hepatotoxicity or nephrotoxicity, highlights RMJ as a promising candidate for further investigation as a novel therapeutic agent for atherosclerotic cardiovascular disease.

{"title":"Regulation of inflammatory responses: Harnessing the Ruan Mai Jian targeting of EphA2/ephrinA1 pathway to enhance atherosclerosis amelioration.","authors":"Xue Zhao, Hanyu Zhang, Jiayi Wang, Lu Zhang, Shuang Gao, Yun Gu, Te Liu, Wenting Du","doi":"10.1016/j.phymed.2025.156398","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156398","url":null,"abstract":"<p><strong>Background: </strong>Atherosclerosis is a major contributor to global cardiovascular morbidity and mortality, driven by the chronic inflammatory proliferation of vascular smooth muscle cells (VSMCs), which destabilizes atherosclerotic plaques. The EphA2/ephrinA1 signaling pathway plays a critical role in modulating VSMC inflammatory responses, making it an attractive therapeutic target. However, the clinical application of EphA2 inhibitors remains limited due to safety concerns. Ruan Mai Jian (RMJ), a traditional Chinese herbal medicine, has demonstrated potential efficacy in treating atherosclerosis, though its precise mechanisms remain insufficiently characterized. To date, no study has investigated a Chinese medicine compound capable of regulating atherosclerotic inflammatory responses via the EphA2/ephrinA1 pathway. This study aims to determine whether RMJ treats atherosclerosis both in vivo and in vitro by modulating the EphA2/ephrinA1 pathway, while evaluating its potential hepatic and renal toxicity.</p><p><strong>Study design: </strong>A combination of in vivo (ApoE-/- murine model) and in vitro studies was employed to investigate the effects of RMJ on atherosclerotic progression, inflammatory markers, and VSMC function.</p><p><strong>Methods: </strong>ApoE-/- mice were fed a high-fat diet to induce atherosclerosis and subsequently treated with RMJ at varying doses. Serum lipid levels, inflammatory cytokines (TNF-α, IL-6, IL-1β), and plaque morphology were analyzed. Immunohistochemical and Western blot analyses were performed to assess the modulation of the EphA2/ephrinA1 pathway. VSMC proliferation and migration assays were conducted to evaluate the effects of RMJ on cellular behavior in vitro.</p><p><strong>Results: </strong>RMJ treatment significantly attenuated serum lipid levels, reduced systemic inflammation, and stabilized atherosclerotic plaques by increasing collagen content and decreasing lipid deposition. RMJ downregulated EphA2 expression and upregulated ephrinA1, effectively inhibiting VSMC proliferation and migration through suppression of the AKT1/ERK1/2 signaling cascade. Importantly, no hepatic or renal toxicity was observed in treated mice, indicating a favorable safety profile.</p><p><strong>Conclusion: </strong>RMJ demonstrates significant therapeutic potential for the treatment of atherosclerosis, primarily through modulation of the EphA2/ephrinA1 signaling pathway, resulting in reduced inflammation and VSMC proliferation. Its efficacy, combined with the absence of hepatotoxicity or nephrotoxicity, highlights RMJ as a promising candidate for further investigation as a novel therapeutic agent for atherosclerotic cardiovascular disease.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"138 ","pages":"156398"},"PeriodicalIF":6.7,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143024361","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