首页 > 最新文献

Toxicology and applied pharmacology最新文献

英文 中文
EGFR tyrosine kinase inhibitor ZZC4 overcomes acquired resistance to gefitinib.
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-23 DOI: 10.1016/j.taap.2025.117280
Mawusse K I Attiogbe, Ting-Ting Huang, Hongyi Zhao, Hongying Wang, Lei Cao, Ping-Ping Yan, Sanqi Zhang, Yongxiao Cao

Despite the tremendous progress of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) development, acquired resistance mechanisms have limited their efficacy in treating non-small cell lung cancer (NSCLC). To overcome these limitations, novel EGFR-TKIs are needed. In our previous study, we presented ZZC4 as a potent EGFR-TKI. In this study, we developed NSCLC cells resistant to EGFR-TKI gefitinib and osimertinib and assessed the effect and mechanism of action of ZZC4 on those cells. HCC827 cells were cultured with gefitinib in a concentration-escalation manner to achieve HCC827 gefitinib-resistant (HCC827-GR) cells after 6 months of treatment. Then, the effect of ZZC4 was assessed at the cellular and animal levels. To understand ZZC4's mechanism of action, the proteome alteration induced by ZZC4 on the resistant cell line was compared to the parental HCC827 cells using comparative proteomics. The result showed that gefitinib's IC50 on HCC827 was 533 nM, approximately 80 times its IC50 on normal cells (7.6 nM), confirming its resistance to HCC827 cells. The obtained resistant cells were treated with ZZC4, which potently suppressed the resistant cells' proliferation with an IC50 of 0.1 nM. In tumor-bearing mice, ZZC4 also suppressed the growth of HCC827-GR cell tumors with an inhibition ratio of 82 % at ZZC4 4 mg/kg. Further, the proteomic analysis revealed that ZZC4 inhibited HCC827-GR cell growth by upregulating CDKN1B and downregulating CCNA2 and CHEK1. In conclusion, ZZC4 overcomes resistance to gefitinib by altering the cell cycle pathway.

{"title":"EGFR tyrosine kinase inhibitor ZZC4 overcomes acquired resistance to gefitinib.","authors":"Mawusse K I Attiogbe, Ting-Ting Huang, Hongyi Zhao, Hongying Wang, Lei Cao, Ping-Ping Yan, Sanqi Zhang, Yongxiao Cao","doi":"10.1016/j.taap.2025.117280","DOIUrl":"https://doi.org/10.1016/j.taap.2025.117280","url":null,"abstract":"<p><p>Despite the tremendous progress of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) development, acquired resistance mechanisms have limited their efficacy in treating non-small cell lung cancer (NSCLC). To overcome these limitations, novel EGFR-TKIs are needed. In our previous study, we presented ZZC4 as a potent EGFR-TKI. In this study, we developed NSCLC cells resistant to EGFR-TKI gefitinib and osimertinib and assessed the effect and mechanism of action of ZZC4 on those cells. HCC827 cells were cultured with gefitinib in a concentration-escalation manner to achieve HCC827 gefitinib-resistant (HCC827-GR) cells after 6 months of treatment. Then, the effect of ZZC4 was assessed at the cellular and animal levels. To understand ZZC4's mechanism of action, the proteome alteration induced by ZZC4 on the resistant cell line was compared to the parental HCC827 cells using comparative proteomics. The result showed that gefitinib's IC<sub>50</sub> on HCC827 was 533 nM, approximately 80 times its IC<sub>50</sub> on normal cells (7.6 nM), confirming its resistance to HCC827 cells. The obtained resistant cells were treated with ZZC4, which potently suppressed the resistant cells' proliferation with an IC<sub>50</sub> of 0.1 nM. In tumor-bearing mice, ZZC4 also suppressed the growth of HCC827-GR cell tumors with an inhibition ratio of 82 % at ZZC4 4 mg/kg. Further, the proteomic analysis revealed that ZZC4 inhibited HCC827-GR cell growth by upregulating CDKN1B and downregulating CCNA2 and CHEK1. In conclusion, ZZC4 overcomes resistance to gefitinib by altering the cell cycle pathway.</p>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":" ","pages":"117280"},"PeriodicalIF":3.3,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143504330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Misrepresentation of mouse genotypes: A call for precision
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-22 DOI: 10.1016/j.taap.2025.117276
Wei Li
{"title":"Misrepresentation of mouse genotypes: A call for precision","authors":"Wei Li","doi":"10.1016/j.taap.2025.117276","DOIUrl":"10.1016/j.taap.2025.117276","url":null,"abstract":"","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117276"},"PeriodicalIF":3.3,"publicationDate":"2025-02-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143479843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Analysis of accumulation of DNA double-strand breaks in mouse tissues by pulsed-field gel electrophoresis
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-20 DOI: 10.1016/j.taap.2025.117278
Takeshi Terabayashi , Takako Sasaki , Toshimasa Ishizaki , Tadashi Tomo , Yoshihiro Nishida , Katsuhiro Hanada
Double-strand DNA breaks (DSBs) are among the most cytotoxic DNA lesions, which can lead to chromosomal instability and eventually cell death. The substances that can potentially induce DSB formation are thus regarded as genotoxic. To date, many genotoxicity tests for detecting DNA breaks have been designed. However, there are limited options available for measuring the accumulation of DSBs in vivo. In this study, we aimed to evaluate a method of detecting the DSBs formed by the direct action of genotoxic substances using pulsed-field gel electrophoresis (PFGE). This approach has the advantage of making it easier to distinguish between DSBs and single-strand DNA breaks (SSBs) induced by the direct action of genotoxic substances. To confirm the detection of DSBs using PFGE, we investigated their accumulation after treatment with cis-diamminedichloroplatinum(II) (cisplatin) or γ-rays in mouse organs. The results revealed the successful detection of cisplatin-induced DSB formation in mouse kidney and thymus and γ-ray-induced DSB formation in all organs. We also discuss the advantages of PFGE-based detection of DSBs in vivo.
{"title":"Analysis of accumulation of DNA double-strand breaks in mouse tissues by pulsed-field gel electrophoresis","authors":"Takeshi Terabayashi ,&nbsp;Takako Sasaki ,&nbsp;Toshimasa Ishizaki ,&nbsp;Tadashi Tomo ,&nbsp;Yoshihiro Nishida ,&nbsp;Katsuhiro Hanada","doi":"10.1016/j.taap.2025.117278","DOIUrl":"10.1016/j.taap.2025.117278","url":null,"abstract":"<div><div>Double-strand DNA breaks (DSBs) are among the most cytotoxic DNA lesions, which can lead to chromosomal instability and eventually cell death. The substances that can potentially induce DSB formation are thus regarded as genotoxic. To date, many genotoxicity tests for detecting DNA breaks have been designed. However, there are limited options available for measuring the accumulation of DSBs in vivo. In this study, we aimed to evaluate a method of detecting the DSBs formed by the direct action of genotoxic substances using pulsed-field gel electrophoresis (PFGE). This approach has the advantage of making it easier to distinguish between DSBs and single-strand DNA breaks (SSBs) induced by the direct action of genotoxic substances. To confirm the detection of DSBs using PFGE, we investigated their accumulation after treatment with <em>cis</em>-diamminedichloroplatinum(II) (cisplatin) or γ-rays in mouse organs. The results revealed the successful detection of cisplatin-induced DSB formation in mouse kidney and thymus and γ-ray-induced DSB formation in all organs. We also discuss the advantages of PFGE-based detection of DSBs in vivo.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117278"},"PeriodicalIF":3.3,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143473909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Hepatotoxicity from long-term administration of hepatoprotective low doses of oleanolic acid in mice
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-19 DOI: 10.1016/j.taap.2025.117277
Yasha Xu , Xi Yang , Yuanfu Lu , Jie Liu
Oleanolic acid is a triterpenoid existed in many medicinal herbs/plants. Oleanolic acid at low doses are hepatoprotective but at high doses produce cholestasis. This study examined hepatotoxicity potential of low doses of oleanolic acid after log-term administration. Male Kunming mice were orally given oleanolic acid at 100, 200 and 300 μmol/kg daily for 14 weeks. Body weights were monitored, and liver injury was determined via blood biochemistry. Histopathology was examined via H&E, Masson, and Sirius red stains. Oleanolic acid accumulation in plasma and liver was determined by LC-MS and hepatic gene expression by qPCR. Oleanolic acid at low doses did not affect animal body weights, but increased liver index. Serum alanine aminotransferase and alkaline phosphatase were increased, while total bilirubin was unchanged. Chronic oleanolic acid produced hepatocyte degeneration, spot necrosis, and fibrosis. Plasma oleanolic acid was increased more than that in the liver. Oleanolic acid increased hepatic expression of Nrf2, Nqo1, Gclc and Mgst1; Expression of bile acid synthesis genes (Cyp7a1, Cyp8b1, Cyp27a1, Cyp7b1, FXR, SHP) was also suppressed at higher doses. The expression of TGF-β1 and Smad3 was increased, while Smad7 decreased, suggesting the progression to liver fibrosis. High dose of oleanolic acid was less effective in producing these changes, probably due to increased liver injury. Overall, oral administration of low doses of oleanolic acid for 14 weeks produced liver injury and fibrosis. These harmful effects were associated with increased oleanolic acid in plasma and liver, and the disruptions of bile acid metabolism, the Nrf2 and TGF-β/Smad signaling pathways.
{"title":"Hepatotoxicity from long-term administration of hepatoprotective low doses of oleanolic acid in mice","authors":"Yasha Xu ,&nbsp;Xi Yang ,&nbsp;Yuanfu Lu ,&nbsp;Jie Liu","doi":"10.1016/j.taap.2025.117277","DOIUrl":"10.1016/j.taap.2025.117277","url":null,"abstract":"<div><div>Oleanolic acid is a triterpenoid existed in many medicinal herbs/plants. Oleanolic acid at low doses are hepatoprotective but at high doses produce cholestasis. This study examined hepatotoxicity potential of low doses of oleanolic acid after log-term administration. Male Kunming mice were orally given oleanolic acid at 100, 200 and 300 μmol/kg daily for 14 weeks. Body weights were monitored, and liver injury was determined via blood biochemistry. Histopathology was examined via H&amp;E, Masson, and Sirius red stains. Oleanolic acid accumulation in plasma and liver was determined by LC-MS and hepatic gene expression by qPCR. Oleanolic acid at low doses did not affect animal body weights, but increased liver index. Serum alanine aminotransferase and alkaline phosphatase were increased, while total bilirubin was unchanged. Chronic oleanolic acid produced hepatocyte degeneration, spot necrosis, and fibrosis. Plasma oleanolic acid was increased more than that in the liver. Oleanolic acid increased hepatic expression of Nrf2, Nqo1, Gclc and Mgst1; Expression of bile acid synthesis genes (Cyp7a1, Cyp8b1, Cyp27a1, Cyp7b1, FXR, SHP) was also suppressed at higher doses. The expression of TGF-β1 and Smad3 was increased, while Smad7 decreased, suggesting the progression to liver fibrosis. High dose of oleanolic acid was less effective in producing these changes, probably due to increased liver injury. Overall, oral administration of low doses of oleanolic acid for 14 weeks produced liver injury and fibrosis. These harmful effects were associated with increased oleanolic acid in plasma and liver, and the disruptions of bile acid metabolism, the Nrf2 and TGF-β/Smad signaling pathways.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117277"},"PeriodicalIF":3.3,"publicationDate":"2025-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143454308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Afzelechin alleviates deltamethrin induced hepatic dysfunction via regulating TLR4/MyD88, HMGB1/RAGE and NF-κB pathway
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-17 DOI: 10.1016/j.taap.2025.117275
Fuad M. Alzahrani , Khalid J. Alzahrani , Khalaf F. Alsharif , Muhammad Faisal Hayat , Ahmed Al-Emam
Deltamethrin (DMN) is a type-II pyrethroid that has been documented to instigate numerous organ toxicities. Afzelechin (ALN) is a plant based polyphenolic compound that exhibits marvelous biological properties. The present research was conducted to assess the alleviative potential of ALN against DMN induced hepatic dysregulations. Thirty-six male albino (Sprague Dawley) rats were apportioned into four random groups including the control, DMN (5mgkg−1), DMN (5mgkg−1) + ALN (2mgkg−1), and ALN (2mgkg−1) alone administrated group. ALN protected hepatic tissues against DMN induced oxidative stress, inflammation and apoptosis. ALN supplementation donwregulated the gene expression of receptor for advanced glycation end products (RAGE), high mobility group box1 (HMGB1), tumor necrosis factor- α (TNF-α), Myeloid differentiation primary response 88 (MyD88), nuclear factor- kappa B (NF-κB), interleukin-6 (IL-6), toll-like receptor 4 (TLR4), cyclooxygenase-2 (COX-2), and interleukin-1β (IL-1β). Besides, ALN administration reduced the levels of reactive oxygen species (ROS) and malondialdehyde while increasing the activities of glutathione peroxidase (GPx), catalase (CAT), glutathione reductase (GSR), heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and glutathione (GSH). The levels of hepatic function markers including GGT, ALT, ALP, and AST were lowered while the concentrations of albumin and total proteins were promoted following the ALN treatment. The levels of Bax, Caspase-9 and Caspase-3 were suppressed while the levels of Bcl-2 were escalated after ALN therapy. Moreover, ALN treatment remarkably mitigated DMN induced histological impairments. These findings highlight the hepatoprotective efficacy of ALN against DMN induced liver toxicity.
{"title":"Afzelechin alleviates deltamethrin induced hepatic dysfunction via regulating TLR4/MyD88, HMGB1/RAGE and NF-κB pathway","authors":"Fuad M. Alzahrani ,&nbsp;Khalid J. Alzahrani ,&nbsp;Khalaf F. Alsharif ,&nbsp;Muhammad Faisal Hayat ,&nbsp;Ahmed Al-Emam","doi":"10.1016/j.taap.2025.117275","DOIUrl":"10.1016/j.taap.2025.117275","url":null,"abstract":"<div><div>Deltamethrin (DMN) is a type-II pyrethroid that has been documented to instigate numerous organ toxicities. Afzelechin (ALN) is a plant based polyphenolic compound that exhibits marvelous biological properties. The present research was conducted to assess the alleviative potential of ALN against DMN induced hepatic dysregulations. Thirty-six male albino (Sprague Dawley) rats were apportioned into four random groups including the control, DMN (5mgkg<sup>−1</sup>), DMN (5mgkg<sup>−1</sup>) + ALN (2mgkg<sup>−1</sup>), and ALN (2mgkg<sup>−1</sup>) alone administrated group. ALN protected hepatic tissues against DMN induced oxidative stress, inflammation and apoptosis. ALN supplementation donwregulated the gene expression of <em>receptor for advanced glycation end products (RAGE), high mobility group box1 (HMGB1), tumor necrosis factor- α (TNF-α), Myeloid differentiation primary response 88 (MyD88), nuclear factor- kappa B (NF-κB), interleukin-6 (IL-6), toll-like receptor 4 (TLR4), cyclooxygenase-2 (COX-2)</em>, and <em>interleukin-1β (IL-1β)</em>. Besides, ALN administration reduced the levels of reactive oxygen species (ROS) and malondialdehyde while increasing the activities of glutathione peroxidase (GPx), catalase (CAT), glutathione reductase (GSR), heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and glutathione (GSH). The levels of hepatic function markers including GGT, ALT, ALP, and AST were lowered while the concentrations of albumin and total proteins were promoted following the ALN treatment. The levels of Bax, Caspase-9 and Caspase-3 were suppressed while the levels of Bcl-2 were escalated after ALN therapy. Moreover, ALN treatment remarkably mitigated DMN induced histological impairments. These findings highlight the hepatoprotective efficacy of ALN against DMN induced liver toxicity.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117275"},"PeriodicalIF":3.3,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143459405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Cyanidin-3-O-glucoside inhibits the malignant progression of colorectal cancer by regulating Kruppel-like factor 4-mediated ERK/p38 signaling pathway
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-17 DOI: 10.1016/j.taap.2025.117268
Jian Chang, Geqiong Xiao

Background

Cyanidin-3-O-glucoside (Cy3g) is a natural anthocyanin, showing favorable anti-cancer efficacy in colorectal cancer (CRC). However, its specific mechanism in CRC remains largely unexplored.

Objective

This study aimed to investigate the underlying mechanisms of Cy3g on CRC.

Methods

Cell viability of human CRC cell lines (SW620, HT29, LS174T, and HCT116) and normal colon fibroblast cell line (CCD-18Co) treated with Cy3g was detected by CCK-8. Effects of Cy3g on malignant characteristics of SW620 cells were determined by CCK-8, EdU, colony formation, wound healing, Transwell, and flow cytometry assays. To further elucidate Cy3g's mechanism in CRC, KLF4 expression was detected by RT-qPCR, and expression of the extracellular signal-related kinase (ERK) and p38 was examined by western blotting. The effects and mechanisms of Cy3g on CRC progression were further validated in a xenograft mouse model.

Results

Cy3g significantly inhibited the cell viability of human CRC cell lines but rarely affected the cell viability of normal colon fibroblast. Cy3g dose-dependently inhibited proliferation, migration, and invasion and promoted apoptosis of SW620 cells. Moreover, Cy3g upregulated KLF4 expression and inactivated the ERK/p38 pathway in a concentration-dependent manner. KLF4 knockdown reversed the inhibitory effects of Cy3g on the malignant characteristics of SW620 and expression of ERK and p38. Animal experiments further validated that Cy3g inhibited tumor growth without altering body weight, activated KLF4, and suppressed the ERK/p38 pathway in CRC model mice.

Conclusion

Cy3g inhibits CRC progression by suppressing the KLF4-mediated ERK/p38 pathway, offering new insights into CRC prevention and treatment.
{"title":"Cyanidin-3-O-glucoside inhibits the malignant progression of colorectal cancer by regulating Kruppel-like factor 4-mediated ERK/p38 signaling pathway","authors":"Jian Chang,&nbsp;Geqiong Xiao","doi":"10.1016/j.taap.2025.117268","DOIUrl":"10.1016/j.taap.2025.117268","url":null,"abstract":"<div><h3>Background</h3><div>Cyanidin-3-<em>O</em>-glucoside (Cy3g) is a natural anthocyanin, showing favorable anti-cancer efficacy in colorectal cancer (CRC). However, its specific mechanism in CRC remains largely unexplored.</div></div><div><h3>Objective</h3><div>This study aimed to investigate the underlying mechanisms of Cy3g on CRC.</div></div><div><h3>Methods</h3><div>Cell viability of human CRC cell lines (SW620, HT29, LS174T, and HCT116) and normal colon fibroblast cell line (CCD-18Co) treated with Cy3g was detected by CCK-8. Effects of Cy3g on malignant characteristics of SW620 cells were determined by CCK-8, EdU, colony formation, wound healing, Transwell, and flow cytometry assays. To further elucidate Cy3g's mechanism in CRC, KLF4 expression was detected by RT-qPCR, and expression of the extracellular signal-related kinase (ERK) and p38 was examined by western blotting. The effects and mechanisms of Cy3g on CRC progression were further validated in a xenograft mouse model.</div></div><div><h3>Results</h3><div>Cy3g significantly inhibited the cell viability of human CRC cell lines but rarely affected the cell viability of normal colon fibroblast. Cy3g dose-dependently inhibited proliferation, migration, and invasion and promoted apoptosis of SW620 cells. Moreover, Cy3g upregulated KLF4 expression and inactivated the ERK/p38 pathway in a concentration-dependent manner. KLF4 knockdown reversed the inhibitory effects of Cy3g on the malignant characteristics of SW620 and expression of ERK and p38. Animal experiments further validated that Cy3g inhibited tumor growth without altering body weight, activated KLF4, and suppressed the ERK/p38 pathway in CRC model mice.</div></div><div><h3>Conclusion</h3><div>Cy3g inhibits CRC progression by suppressing the KLF4-mediated ERK/p38 pathway, offering new insights into CRC prevention and treatment.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117268"},"PeriodicalIF":3.3,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143445800","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Isonardosinone attenuates osteoclastogenesis and OVX-induced bone loss via the MAPK/NF-κB pathway
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-15 DOI: 10.1016/j.taap.2025.117267
Guangwei Wen , Haishan Li , Jiasheng Yang , Bin Mai , Tengpeng Zhou , GuoYe Mo , Yongxian Li , Yiyi Lai
Osteoporosis is a globally prevalent metabolic bone disease that manifests itself as a decrease in bone mineral density and deterioration of bone structure, which reduces overall bone strength and increases fracture risk. However, the effect of anti-inflammatory isonardosinone (the active ingredient in Nardostachys chinensis) on osteoclastogenesis is unknown. We first predicted the main pathways and targets of ISO action in osteoporosis by network pharmacology. CCK-8 was used to test whether ISO affects cell proliferation of BMMs (osteoclast precursor cells) and to determine the safe action concentration. TRAcP and F-actin staining were used to characterise the inhibitory effect on osteoclast differentiation. RT-PCR and WB were used to examine changes in the relative expression of genes and proteins generated by osteoclasts under isopinacolone treatment, and we examined its effects on the RANKL-activated MAPK and NF-κB signaling pathways. An ovariectomy-induced osteoporosis model was constructed to assess the in vivo therapeutic effects of ISO. CCK-8 results showed that ISO had no cytotoxic or proliferative effects on BMMs at concentrations below 30 μM; TRAcP staining showed that ISO suppressed osteoclastogenesis in a concentration- and time-gradient-dependent manner; and F-actin staining showed that ISO suppressed osteoblast skeleton formation and expansion; RT-PCR and Western Blot assays showed that ISO suppressed the expression of CTSK, NFATC1, MMP9, C-Fos, and ACP5, inhibited the phosphorylation of JNK, P38, and ERK, and reversed the degradation of IκB-α, especially within 15 min. The in vivo results indicated that ISO has therapeutic effects on osteoporosis by improving bone microstructure to rescue bone loss. Taken together, these results lead to the conclusion that ISO is an attractive drug development strategy for the treatment of osteoporosis by effectively suppressing osteoclastogenesis through the MAPK/NF-κB signaling pathway, thereby reversing the bone loss associated with ovariectomy in vivo.
{"title":"Isonardosinone attenuates osteoclastogenesis and OVX-induced bone loss via the MAPK/NF-κB pathway","authors":"Guangwei Wen ,&nbsp;Haishan Li ,&nbsp;Jiasheng Yang ,&nbsp;Bin Mai ,&nbsp;Tengpeng Zhou ,&nbsp;GuoYe Mo ,&nbsp;Yongxian Li ,&nbsp;Yiyi Lai","doi":"10.1016/j.taap.2025.117267","DOIUrl":"10.1016/j.taap.2025.117267","url":null,"abstract":"<div><div>Osteoporosis is a globally prevalent metabolic bone disease that manifests itself as a decrease in bone mineral density and deterioration of bone structure, which reduces overall bone strength and increases fracture risk. However, the effect of anti-inflammatory isonardosinone (the active ingredient in <em>Nardostachys chinensis</em>) on osteoclastogenesis is unknown. We first predicted the main pathways and targets of ISO action in osteoporosis by network pharmacology. CCK-8 was used to test whether ISO affects cell proliferation of BMMs (osteoclast precursor cells) and to determine the safe action concentration. TRAcP and F-actin staining were used to characterise the inhibitory effect on osteoclast differentiation. RT-PCR and WB were used to examine changes in the relative expression of genes and proteins generated by osteoclasts under isopinacolone treatment, and we examined its effects on the RANKL-activated MAPK and NF-κB signaling pathways. An ovariectomy-induced osteoporosis model was constructed to assess the <em>in vivo</em> therapeutic effects of ISO. CCK-8 results showed that ISO had no cytotoxic or proliferative effects on BMMs at concentrations below 30 μM; TRAcP staining showed that ISO suppressed osteoclastogenesis in a concentration- and time-gradient-dependent manner; and F-actin staining showed that ISO suppressed osteoblast skeleton formation and expansion; RT-PCR and Western Blot assays showed that ISO suppressed the expression of CTSK, NFATC1, MMP9, C-Fos, and ACP5, inhibited the phosphorylation of JNK, P38, and ERK, and reversed the degradation of IκB-α, especially within 15 min. The <em>in vivo</em> results indicated that ISO has therapeutic effects on osteoporosis by improving bone microstructure to rescue bone loss. Taken together, these results lead to the conclusion that ISO is an attractive drug development strategy for the treatment of osteoporosis by effectively suppressing osteoclastogenesis through the MAPK/NF-κB signaling pathway, thereby reversing the bone loss associated with ovariectomy <em>in vivo</em>.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117267"},"PeriodicalIF":3.3,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143427685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Magnolin ameliorates acetaminophen-induced liver injury in mice via modulating the MAPK pathway and lipid metabolism
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-12 DOI: 10.1016/j.taap.2025.117264
Ting Yao , Youhe Wu , Liyun Fu, Lanjun Li
Acetaminophen (APAP)-induced liver injury (AILI) represents a common yet potentially severe type of drug-induced liver injury with limited available effective therapeutic methods. Magnolin possesses excellent anti-inflammatory and antioxidant properties for treating various diseases. However, its effects against AILI and the fundamental mechanisms still lack comprehensive exploration. This research endeavors to assess magnolin's hepatoprotective properties against AILI. The AILI model was established in male C57BL/6 mice via intraperitoneal injection of 300 mg/kg APAP and in the HepG2 cell line by treating it with 20 mM APAP. The levels of oxidation, liver damage and inflammation were assessed. Transcriptomics and metabolomics were utilized to explore the mechanism underlying magnolin treatment in AILI. We found that 5 mg/kg magnolin effectively mitigated the elevated serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT), along with inflammatory factor (IL-6, and TNF-α) levels in vivo. Meanwhile, magnolin relieved oxidative stress by increasing superoxide dismutase activity and reducing malondialdehyde along with oxidized glutathione/reduced glutathione (GSSG/GSH). 6 μM magnolin increased cell viability and reduced the lipid peroxidation in vitro. Furthermore, 5 mg/kg magnolin altered the expression of 413 genes and the levels of 70 metabolites compared with Control group, which were enriched in lipid metabolism, inflammatory responses, and the MAPK signaling pathway. However, 10 mg/kg magnolin tended to exacerbate liver damage. Overall, 5 mg/kg magnolin effectively protects against AILI by modulating inflammatory responses and the MAPK pathway, whereas 10 mg/kg worsens liver injury, underscoring the need for dose optimization. These findings offer a fresh perspective and novel therapy for AILI.
{"title":"Magnolin ameliorates acetaminophen-induced liver injury in mice via modulating the MAPK pathway and lipid metabolism","authors":"Ting Yao ,&nbsp;Youhe Wu ,&nbsp;Liyun Fu,&nbsp;Lanjun Li","doi":"10.1016/j.taap.2025.117264","DOIUrl":"10.1016/j.taap.2025.117264","url":null,"abstract":"<div><div>Acetaminophen (APAP)-induced liver injury (AILI) represents a common yet potentially severe type of drug-induced liver injury with limited available effective therapeutic methods. Magnolin possesses excellent anti-inflammatory and antioxidant properties for treating various diseases. However, its effects against AILI and the fundamental mechanisms still lack comprehensive exploration. This research endeavors to assess magnolin's hepatoprotective properties against AILI. The AILI model was established in male C57BL/6 mice via intraperitoneal injection of 300 mg/kg APAP and in the HepG2 cell line by treating it with 20 mM APAP. The levels of oxidation, liver damage and inflammation were assessed. Transcriptomics and metabolomics were utilized to explore the mechanism underlying magnolin treatment in AILI. We found that 5 mg/kg magnolin effectively mitigated the elevated serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT), along with inflammatory factor (IL-6, and TNF-α) levels in vivo. Meanwhile, magnolin relieved oxidative stress by increasing superoxide dismutase activity and reducing malondialdehyde along with oxidized glutathione/reduced glutathione (GSSG/GSH). 6 μM magnolin increased cell viability and reduced the lipid peroxidation in vitro. Furthermore, 5 mg/kg magnolin altered the expression of 413 genes and the levels of 70 metabolites compared with Control group, which were enriched in lipid metabolism, inflammatory responses, and the MAPK signaling pathway. However, 10 mg/kg magnolin tended to exacerbate liver damage. Overall, 5 mg/kg magnolin effectively protects against AILI by modulating inflammatory responses and the MAPK pathway, whereas 10 mg/kg worsens liver injury, underscoring the need for dose optimization. These findings offer a fresh perspective and novel therapy for AILI.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117264"},"PeriodicalIF":3.3,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143419367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Targeting high mobility group protein B2 exerts antiproliferative effects in hypoxic pulmonary hypertension by modulating miR-21
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-12 DOI: 10.1016/j.taap.2025.117265
Pan Wang , Xu Zhang , Mengge Yao , Jiakang Li , Xiaozhen Wei , Zhihuang Qiu , Liangwan Chen , Li Zhang

Objective

Pulmonary hypertension (PH) is characterized by excessive vascular cell proliferation, leading to vascular remodeling. In this study, we aimed to investigate the molecular mechanisms underlying the regulation of vascular cell proliferation in the context of HMGB2 and its potential involvement in the pathogenesis of PH.

Methods

Animals and pulmonary vascular smooth muscle cells (PASMCs) were exposed to hypoxia. Pathological changes in pulmonary vessels were detected by HE and Masson staining. The effect of HMGB2 on cell proliferation was detected by siRNA transfections and recombinant protein treatment. miR-21 inhibitor and mimics were applied, and TPM1 expression was detected. HMGB2−/− mice were applied to observe the possible preventive effect of HMGB2 in PH development.

Results

HMGB2 expression was increased in hypoxic rats and PASMCs. Silencing ZDHHC5 reduced HMGB2 expression and cell proliferation. Cell proliferation was inhibited by knocking down HMGB2 and promoted by its over-expression. Hypoxia-induced miR-21 upregulation and TPM1 downregulation were mediated by HMGB2. 8-Br-cGMP suppressed HMGB2-induced PASMC proliferation and increased SOX2 expression by activating the cGMP/PKG signaling pathway. HMGB2−/− attenuated pulmonary vascular remodeling and fibrosis in hypoxia induced PH mice.

Conclusions

HMGB2 promotes PASMC proliferation through the cGMP/PKG-SOX2-miR-21-TPM1 pathway, which provides a new theoretical basis and possible targets for the pathogenesis and clinical prevention of PH.
{"title":"Targeting high mobility group protein B2 exerts antiproliferative effects in hypoxic pulmonary hypertension by modulating miR-21","authors":"Pan Wang ,&nbsp;Xu Zhang ,&nbsp;Mengge Yao ,&nbsp;Jiakang Li ,&nbsp;Xiaozhen Wei ,&nbsp;Zhihuang Qiu ,&nbsp;Liangwan Chen ,&nbsp;Li Zhang","doi":"10.1016/j.taap.2025.117265","DOIUrl":"10.1016/j.taap.2025.117265","url":null,"abstract":"<div><h3>Objective</h3><div>Pulmonary hypertension (PH) is characterized by excessive vascular cell proliferation, leading to vascular remodeling. In this study, we aimed to investigate the molecular mechanisms underlying the regulation of vascular cell proliferation in the context of HMGB2 and its potential involvement in the pathogenesis of PH.</div></div><div><h3>Methods</h3><div>Animals and pulmonary vascular smooth muscle cells (PASMCs) were exposed to hypoxia. Pathological changes in pulmonary vessels were detected by HE and Masson staining. The effect of HMGB2 on cell proliferation was detected by siRNA transfections and recombinant protein treatment. miR-21 inhibitor and mimics were applied, and TPM1 expression was detected. HMGB2<sup>−/−</sup> mice were applied to observe the possible preventive effect of HMGB2 in PH development.</div></div><div><h3>Results</h3><div>HMGB2 expression was increased in hypoxic rats and PASMCs. Silencing ZDHHC5 reduced HMGB2 expression and cell proliferation. Cell proliferation was inhibited by knocking down HMGB2 and promoted by its over-expression. Hypoxia-induced miR-21 upregulation and TPM1 downregulation were mediated by HMGB2. 8-Br-cGMP suppressed HMGB2-induced PASMC proliferation and increased SOX2 expression by activating the cGMP/PKG signaling pathway. HMGB2<sup>−/−</sup> attenuated pulmonary vascular remodeling and fibrosis in hypoxia induced PH mice.</div></div><div><h3>Conclusions</h3><div>HMGB2 promotes PASMC proliferation through the cGMP/PKG-SOX2-miR-21-TPM1 pathway, which provides a new theoretical basis and possible targets for the pathogenesis and clinical prevention of PH.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117265"},"PeriodicalIF":3.3,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143419366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Sodium taurocholate co-transporting polypeptide deficiency attenuates acetaminophen-induced hepatotoxicity via regulating expression of drug metabolism enzymes in mice
IF 3.3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-11 DOI: 10.1016/j.taap.2025.117266
Fangji Yang , Lina Wu , Wenxiong Xu , Yuankai Wu , Shu Zhu , Yuzhen Zhang , Yutian Chong , Liang Peng
Acetaminophen (APAP) overdose can induce liver injury and is generally accompanied by disruption of bile acid homeostasis. Physiologically, sodium taurocholate co-transporting polypeptide (NTCP) participates in the uptake of bile acids from portal blood into hepatocytes to maintain enterohepatic recirculation but its role in APAP-induced hepatotoxicity is unclear. Wild-type (WT) C57BL/6J and NTCP knockout (KO) mice were injected with 400 mg/kg APAP and liver injury was evaluated by serum biochemical markers and histologic evaluation. RNA-seq analysis was performed to evaluate the liver gene expression profiles in APAP-treated mice. Compared with WT mice, the exposure to APAP overdose caused liver dysfunction, oxidative stress, inflammation and cell death, which were ameliorated by NTCP deficiency. APAP detoxification, metabolism, and elimination were significantly accelerated by the upregulation of UDP-glucuronosyltransferase (Ugt1a1, Ugt1a6 and Ugt1a9), sulfotransferase (Sult1a1 and Sult2a1) and bile acid efflux transporters (Abcc2/3/4) in NTCP KO mice compared with WT mice. Interestingly, APAP-induced hepatotoxicity was ameliorated using Irbesartan and Ezetimibe (NTCP inhibitors). In conclusion, NTCP deficiency attenuates APAP-induced hepatotoxicity by enhancing the metabolism and elimination of APAP. NTCP inhibitors protect against APAP-induced hepatotoxicity and thus are a potential therapeutic option.
{"title":"Sodium taurocholate co-transporting polypeptide deficiency attenuates acetaminophen-induced hepatotoxicity via regulating expression of drug metabolism enzymes in mice","authors":"Fangji Yang ,&nbsp;Lina Wu ,&nbsp;Wenxiong Xu ,&nbsp;Yuankai Wu ,&nbsp;Shu Zhu ,&nbsp;Yuzhen Zhang ,&nbsp;Yutian Chong ,&nbsp;Liang Peng","doi":"10.1016/j.taap.2025.117266","DOIUrl":"10.1016/j.taap.2025.117266","url":null,"abstract":"<div><div>Acetaminophen (APAP) overdose can induce liver injury and is generally accompanied by disruption of bile acid homeostasis. Physiologically, sodium taurocholate <em>co</em>-transporting polypeptide (NTCP) participates in the uptake of bile acids from portal blood into hepatocytes to maintain enterohepatic recirculation but its role in APAP-induced hepatotoxicity is unclear. Wild-type (WT) C57BL/6J and NTCP knockout (KO) mice were injected with 400<!--> <!-->mg/kg APAP and liver injury was evaluated by serum biochemical markers and histologic evaluation. RNA-seq analysis was performed to evaluate the liver gene expression profiles in APAP-treated mice. Compared with WT mice, the exposure to APAP overdose caused liver dysfunction, oxidative stress, inflammation and cell death, which were ameliorated by NTCP deficiency. APAP detoxification, metabolism, and elimination were significantly accelerated by the upregulation of UDP-glucuronosyltransferase (Ugt1a1, Ugt1a6 and Ugt1a9), sulfotransferase (Sult1a1 and Sult2a1) and bile acid efflux transporters (Abcc2/3/4) in NTCP KO mice compared with WT mice. Interestingly, APAP-induced hepatotoxicity was ameliorated using Irbesartan and Ezetimibe (NTCP inhibitors). In conclusion, NTCP deficiency attenuates APAP-induced hepatotoxicity by enhancing the metabolism and elimination of APAP. NTCP inhibitors protect against APAP-induced hepatotoxicity and thus are a potential therapeutic option.</div></div>","PeriodicalId":23174,"journal":{"name":"Toxicology and applied pharmacology","volume":"497 ","pages":"Article 117266"},"PeriodicalIF":3.3,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143415341","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Toxicology and applied pharmacology
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