首页 > 最新文献

Journal of Biochemical and Molecular Toxicology最新文献

英文 中文
Sarsasapogenin Inhibits HCT116 and Caco-2 Cell Malignancy and Tumor Growth in a Xenograft Mouse Model of Colorectal Cancer by Inactivating MAPK Signaling
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-26 DOI: 10.1002/jbt.70189
Ping Pan, Zhen Zhang, Yu Xu, Fangfang Li, Qingle Yang, Bing Liang

Colorectal cancer (CRC) is a prevalent malignancy globally and holds the third position in terms of cancer-related mortality in the United States. The study aimed to explore the impact of sarsasapogenin (Sar), a natural component, on CRC cell behavior and the related mechanism. Caco-2 and HCT116 cells were treated with 0–40 μM Sar or 5-fluorouracil (5-FU) to compare their cytotoxicity. Then, the optimal concentration of Sar was identified for subsequent experiments, and CRC cells in the control group were treated with dimethyl sulfoxide (DMSO). Cell counting kit-8 assays, colony-forming assays, and flow cytometry analyses were carried out to measure cell viability, proliferation, and apoptosis, respectively. Cell migration and invasion were evaluated by Transwell assays. HCT116 cells were inoculated into nude mice to induce tumorigenesis, and oral gavage of Sar was performed when tumor volume reached 50–100 mm3. Immunohistochemistry was performed to measure Ki67, E-cadherin, Vimentin, and N-cadherin expression in mouse tumor tissues. Western blot analysis was performed to assess protein levels of factors related to apoptosis, epithelial-mesenchymal transition (EMT) and mitogen-activated protein kinase (MAPK) pathway in CRC cells or mouse tumor tissues. Results showed that Sar repressed CRC cell viability in a dose-dependent manner, and the IC50 of Sar is 9.53 and 9.69 μM in HCT116 cells and Caco-2 cells. The number of CRC cell colonies was significantly decreased by Sar compared with that in DMSO group (HCT116: 52 vs. 162; Caco-2: 46 vs. 146), while cell apoptotic rate was increased by Sar (20.41% and 20.78%) compared to that in response to DMSO treatment (5.26% and 5.65%). Sar led to significant upregulation of Bax and cleaved caspase-3 protein levels while reducing Bcl-2 protein level. The number of migrated cells was reduced by Sar treatment in comparison to those in the context of DMSO treatment (HCT116: 65 vs. 223; Caco-2: 32 vs. 168). The same inhibitory impact of Sar was found on the number of invaded cells (p < 0.001). E-cadherin level was noticeably elevated while N-cadherin and vimentin levels were prominently lessened in Sar-treated CRC cells. For animal experiments, the size, growth rate, and weight of tumors were all repressed by Sar (p < 0.001). Ki67 expression was reduced and the EMT process was obstructed in mouse tumors of the Sar group (p < 0.001). Sar inhibited the activation of MAPK signaling both in CRC cells and mouse tumors (p < 0.001). In conclusion, Sar represses HCT116 and Caco-2 cell proliferation, migration, invasion, and xenograft tumor growth while promoting CRC cell apoptosis by inactivating the MAPK signaling.

{"title":"Sarsasapogenin Inhibits HCT116 and Caco-2 Cell Malignancy and Tumor Growth in a Xenograft Mouse Model of Colorectal Cancer by Inactivating MAPK Signaling","authors":"Ping Pan,&nbsp;Zhen Zhang,&nbsp;Yu Xu,&nbsp;Fangfang Li,&nbsp;Qingle Yang,&nbsp;Bing Liang","doi":"10.1002/jbt.70189","DOIUrl":"https://doi.org/10.1002/jbt.70189","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 <p>Colorectal cancer (CRC) is a prevalent malignancy globally and holds the third position in terms of cancer-related mortality in the United States. The study aimed to explore the impact of sarsasapogenin (Sar), a natural component, on CRC cell behavior and the related mechanism. Caco-2 and HCT116 cells were treated with 0–40 μM Sar or 5-fluorouracil (5-FU) to compare their cytotoxicity. Then, the optimal concentration of Sar was identified for subsequent experiments, and CRC cells in the control group were treated with dimethyl sulfoxide (DMSO). Cell counting kit-8 assays, colony-forming assays, and flow cytometry analyses were carried out to measure cell viability, proliferation, and apoptosis, respectively. Cell migration and invasion were evaluated by Transwell assays. HCT116 cells were inoculated into nude mice to induce tumorigenesis, and oral gavage of Sar was performed when tumor volume reached 50–100 mm<sup>3</sup>. Immunohistochemistry was performed to measure Ki67, E-cadherin, Vimentin, and N-cadherin expression in mouse tumor tissues. Western blot analysis was performed to assess protein levels of factors related to apoptosis, epithelial-mesenchymal transition (EMT) and mitogen-activated protein kinase (MAPK) pathway in CRC cells or mouse tumor tissues. Results showed that Sar repressed CRC cell viability in a dose-dependent manner, and the IC50 of Sar is 9.53 and 9.69 μM in HCT116 cells and Caco-2 cells. The number of CRC cell colonies was significantly decreased by Sar compared with that in DMSO group (HCT116: 52 vs. 162; Caco-2: 46 vs. 146), while cell apoptotic rate was increased by Sar (20.41% and 20.78%) compared to that in response to DMSO treatment (5.26% and 5.65%). Sar led to significant upregulation of Bax and cleaved caspase-3 protein levels while reducing Bcl-2 protein level. The number of migrated cells was reduced by Sar treatment in comparison to those in the context of DMSO treatment (HCT116: 65 vs. 223; Caco-2: 32 vs. 168). The same inhibitory impact of Sar was found on the number of invaded cells (<i>p </i>&lt; 0.001). E-cadherin level was noticeably elevated while N-cadherin and vimentin levels were prominently lessened in Sar-treated CRC cells. For animal experiments, the size, growth rate, and weight of tumors were all repressed by Sar (<i>p </i>&lt; 0.001). Ki67 expression was reduced and the EMT process was obstructed in mouse tumors of the Sar group (<i>p </i>&lt; 0.001). Sar inhibited the activation of MAPK signaling both in CRC cells and mouse tumors (<i>p </i>&lt; 0.001). In conclusion, Sar represses HCT116 and Caco-2 cell proliferation, migration, invasion, and xenograft tumor growth while promoting CRC cell apoptosis by inactivating the MAPK signaling.</p>\u0000 </section>\u0000 </div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143497372","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
Mitochondrial Quality Control and Melatonin: A Strategy Against Myocardial Injury
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-26 DOI: 10.1002/jbt.70194
Nima Ghavamikia, Faranak Mehrnoosh, Farshad Zare, Payam Ali-khiavi, Ali Sinehsepehr, Yasaman Ghodsi Boushehri, Milad Vahedinezhad, Elham Abdollahi, Ahmed Hjazi, Siamak Aminnezhad, Hossein Saffarfar, Sina Hamzehzadeh, Mehrdad Nourizadeh, Sepideh KarkonShayan

Melatonin exhibits various biological functions, including regulation of circadian and endocrine rhythms, anti-inflammatory, and antioxidant effects. Aging and damaged mitochondria are major sources of oxidative stress (OS), and mitochondrial quality control (MQC) is crucial for maintaining normal mitochondrial function. Myocardial ischemia-reperfusion injury is a major complication that can arise during reperfusion therapy for coronary heart disease. However, effective intervention strategies are currently lacking. Mitochondrial dysfunction and OS are considered central mechanisms of myocardial reperfusion injury, with mitochondrial-targeted interventions being a potential treatment direction. Recent studies have shown that melatonin improves mitochondrial structure and function through multiple pathways. This review discusses the mechanisms by which melatonin ameliorates myocardial ischemia-reperfusion injury, focusing on MQC, and explores its potential applications in the prevention and treatment of myocardial ischemia-reperfusion injury.

{"title":"Mitochondrial Quality Control and Melatonin: A Strategy Against Myocardial Injury","authors":"Nima Ghavamikia,&nbsp;Faranak Mehrnoosh,&nbsp;Farshad Zare,&nbsp;Payam Ali-khiavi,&nbsp;Ali Sinehsepehr,&nbsp;Yasaman Ghodsi Boushehri,&nbsp;Milad Vahedinezhad,&nbsp;Elham Abdollahi,&nbsp;Ahmed Hjazi,&nbsp;Siamak Aminnezhad,&nbsp;Hossein Saffarfar,&nbsp;Sina Hamzehzadeh,&nbsp;Mehrdad Nourizadeh,&nbsp;Sepideh KarkonShayan","doi":"10.1002/jbt.70194","DOIUrl":"https://doi.org/10.1002/jbt.70194","url":null,"abstract":"<div>\u0000 \u0000 <p>Melatonin exhibits various biological functions, including regulation of circadian and endocrine rhythms, anti-inflammatory, and antioxidant effects. Aging and damaged mitochondria are major sources of oxidative stress (OS), and mitochondrial quality control (MQC) is crucial for maintaining normal mitochondrial function. Myocardial ischemia-reperfusion injury is a major complication that can arise during reperfusion therapy for coronary heart disease. However, effective intervention strategies are currently lacking. Mitochondrial dysfunction and OS are considered central mechanisms of myocardial reperfusion injury, with mitochondrial-targeted interventions being a potential treatment direction. Recent studies have shown that melatonin improves mitochondrial structure and function through multiple pathways. This review discusses the mechanisms by which melatonin ameliorates myocardial ischemia-reperfusion injury, focusing on MQC, and explores its potential applications in the prevention and treatment of myocardial ischemia-reperfusion injury.</p>\u0000 </div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143497370","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
Curcumin Restrains TGF-β2-Induced Proliferation, Migration, Invasion and EMT in Lens Epithelial Cells by Regulating FGF7/ZEB1 Axis
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-26 DOI: 10.1002/jbt.70191
Jing He, Ping Xie, Yangjun Ou

Posterior capsular opacification (PCO) is the most common complication after cataract surgery characterized by hyperproliferation, migration and epithelial-mesenchymal transition (EMT) in residual lens epithelial cells (LECs). Curcumin is a polyphenol compound possessing diverse pharmacological properties. Here, we investigated the functions and its potential mechanisms of curcumin in PCO using transforming growth factor beta2 (TGF-β2)-treated LECs. Cell phenotypes were analyzed using MTT, 5-thynyl-2′-deoxyuridine (EdU), transwell, and scratch assays, respectively. Levels of FGF7 (Fibroblast Growth Factor 7), ZEB1 (Zinc finger E-box binding homeobox 1), and EMT-related proteins were detected by qRT-PCR and western blot analysis. The protein interaction between FGF7 and ZEB1 was validated using Co-immunoprecipitation assay. Curcumin treatment weakened TGF-β2-induced proliferation, migration, invasion and EMT progression in LECs. The expression of FGF7 was boosted by curcumin in LECs. Functionally, FGF7 deficiency suppressed TGF-β2-induced proliferation, migration, invasion and EMT progression in LECs, and could reverse the suppressing action of curcumin on TGF-β2-induced LEC dysfunction. Mechanistically, FGF7 directly interacted with ZEB1, and curcumin could regulate ZEB1 expression via FGF7. Moreover, ZEB1 overexpression could abolish the protective effects of curcumin or FGF7 deficiency on LECs under TGF-β2 stimulation. In conclusion, curcumin protected LECs against TGF-β2-induced enhancement on the proliferation, migration, invasion and EMT process by regulating FGF7/ZEB1 axis, suggesting a new insight into the application of curcumin in PCO therapy.

{"title":"Curcumin Restrains TGF-β2-Induced Proliferation, Migration, Invasion and EMT in Lens Epithelial Cells by Regulating FGF7/ZEB1 Axis","authors":"Jing He,&nbsp;Ping Xie,&nbsp;Yangjun Ou","doi":"10.1002/jbt.70191","DOIUrl":"https://doi.org/10.1002/jbt.70191","url":null,"abstract":"<div>\u0000 \u0000 <p>Posterior capsular opacification (PCO) is the most common complication after cataract surgery characterized by hyperproliferation, migration and epithelial-mesenchymal transition (EMT) in residual lens epithelial cells (LECs). Curcumin is a polyphenol compound possessing diverse pharmacological properties. Here, we investigated the functions and its potential mechanisms of curcumin in PCO using transforming growth factor beta2 (TGF-β2)-treated LECs. Cell phenotypes were analyzed using MTT, 5-thynyl-2′-deoxyuridine (EdU), transwell, and scratch assays, respectively. Levels of FGF7 (Fibroblast Growth Factor 7), ZEB1 (Zinc finger E-box binding homeobox 1), and EMT-related proteins were detected by qRT-PCR and western blot analysis. The protein interaction between FGF7 and ZEB1 was validated using Co-immunoprecipitation assay. Curcumin treatment weakened TGF-β2-induced proliferation, migration, invasion and EMT progression in LECs. The expression of FGF7 was boosted by curcumin in LECs. Functionally, FGF7 deficiency suppressed TGF-β2-induced proliferation, migration, invasion and EMT progression in LECs, and could reverse the suppressing action of curcumin on TGF-β2-induced LEC dysfunction. Mechanistically, FGF7 directly interacted with ZEB1, and curcumin could regulate ZEB1 expression via FGF7. Moreover, ZEB1 overexpression could abolish the protective effects of curcumin or FGF7 deficiency on LECs under TGF-β2 stimulation. In conclusion, curcumin protected LECs against TGF-β2-induced enhancement on the proliferation, migration, invasion and EMT process by regulating FGF7/ZEB1 axis, suggesting a new insight into the application of curcumin in PCO therapy.</p>\u0000 </div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143497369","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
Protective Effects of Galangin Against Cyclophosphamide-Induced Cardiotoxicity via Suppressing NF-κB and Improving Mitochondrial Biogenesis 高良姜素通过抑制 NF-κB 和改善线粒体生物生成对环磷酰胺诱导的心脏毒性具有保护作用
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-25 DOI: 10.1002/jbt.70193
Manar Ali Elsayed, Doaa A. Radwan, Hanem Mohamed Rabah, Hemat El-Sayed El-Horany, Nahla Anas Nasef, Rehab E. Abo El Gheit, Marwa N. Emam, Rasha Osama Elesawy, Walaa Elseady, Alia Mahmoud

Cyclophosphamide (CYP) is an effective chemotherapeutic and immunosuppressive agent; however, its clinical application is limited by a variety of toxic side effects. Mitochondrial dysfunction has been associated with the pathogenesis of chemotherapy-induced cardiotoxicity. This work aimed to evaluate the possible protective effect of galangin (Gal) on CYP-induced cardiotoxicity, pointing to its ability to promote mitochondrial biogenesis. Thirty two male rats were allocated equally into four groups: control; Gal-treated; CYP-treated; and Gal + CYP-treated groups. Markers of cardiac injury, oxidative/antioxidant status, inflammation, apoptosis, and mitochondrial function were assessed in addition to histopathological and electrocardiographic (ECG) evaluation. The current results revealed that Gal treatment significantly attenuated the cardiac injury and retrieved the alterations in cardiac histopathology and ECG changes. Also, it restored redox balance, as evidenced by the alleviation of malondialdehyde (MDA) levels and increased glutathione peroxidase (GPx) activity. Gal activated the sirtuin (SIRT) 1/nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated signaling pathway, as indicated by upregulation of SIRT1, Nrf2, SIRT3, and mitochondrial transcription factor (TFAM), in addition to increased levels of superoxide dismutase 2 (SOD)2 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), together with increased activity of citrate synthase (CS), pointing to improved mitochondrial function. It ameliorated the inflammation and apoptosis-associated markers supported by biochemical and immunostaining data. Our study provided novel insights elucidating the mitigative potential of against CYP-induced cardiac oxidative damage, inflammation, apoptosis, and mitochondrial dysfunction by upregulating the SIRT1/Nrf2/SIRT3/PGC-1α/TFAM survival pathway.

{"title":"Protective Effects of Galangin Against Cyclophosphamide-Induced Cardiotoxicity via Suppressing NF-κB and Improving Mitochondrial Biogenesis","authors":"Manar Ali Elsayed,&nbsp;Doaa A. Radwan,&nbsp;Hanem Mohamed Rabah,&nbsp;Hemat El-Sayed El-Horany,&nbsp;Nahla Anas Nasef,&nbsp;Rehab E. Abo El Gheit,&nbsp;Marwa N. Emam,&nbsp;Rasha Osama Elesawy,&nbsp;Walaa Elseady,&nbsp;Alia Mahmoud","doi":"10.1002/jbt.70193","DOIUrl":"https://doi.org/10.1002/jbt.70193","url":null,"abstract":"<div>\u0000 \u0000 <p>Cyclophosphamide (CYP) is an effective chemotherapeutic and immunosuppressive agent; however, its clinical application is limited by a variety of toxic side effects. Mitochondrial dysfunction has been associated with the pathogenesis of chemotherapy-induced cardiotoxicity. This work aimed to evaluate the possible protective effect of galangin (Gal) on CYP-induced cardiotoxicity, pointing to its ability to promote mitochondrial biogenesis. Thirty two male rats were allocated equally into four groups: control; Gal-treated; CYP-treated; and Gal + CYP-treated groups. Markers of cardiac injury, oxidative/antioxidant status, inflammation, apoptosis, and mitochondrial function were assessed in addition to histopathological and electrocardiographic (ECG) evaluation. The current results revealed that Gal treatment significantly attenuated the cardiac injury and retrieved the alterations in cardiac histopathology and ECG changes. Also, it restored redox balance, as evidenced by the alleviation of malondialdehyde (MDA) levels and increased glutathione peroxidase (GPx) activity. Gal activated the sirtuin (SIRT) 1/nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated signaling pathway, as indicated by upregulation of SIRT1, Nrf2, SIRT3, and mitochondrial transcription factor (TFAM), in addition to increased levels of superoxide dismutase 2 (SOD)2 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), together with increased activity of citrate synthase (CS), pointing to improved mitochondrial function. It ameliorated the inflammation and apoptosis-associated markers supported by biochemical and immunostaining data. Our study provided novel insights elucidating the mitigative potential of against CYP-induced cardiac oxidative damage, inflammation, apoptosis, and mitochondrial dysfunction by upregulating the SIRT1/Nrf2/SIRT3/PGC-1α/TFAM survival pathway.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143489914","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
Unveiling Novel Targets in Lung Tumors for Enhanced Radiotherapy Efficacy: A Comprehensive Review
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-23 DOI: 10.1002/jbt.70180
Faris Anad Muhammad, Ayat Hussein Adhab, Morug Salih Mahdi, Vicky Jain, Subbulakshmi Ganesan, Deepak Bhanot, K. Satyam Naidu, Sharnjeet Kaur, Aseel Salah Mansoor, Usama Kadem Radi, Nasr Saadoun Abd, Muthena Kariem

Radiotherapy is a cornerstone of lung cancer management, though its efficacy is frequently undermined by intrinsic and acquired radioresistance. This review examines the complexity of lung tumors, highlighting their potential as a reservoir of novel targets for radiosensitization. Ionizing radiation (IR) primarily exerts its effects through oxidative damage and DNA double-strand breaks (DSBs). Lung cancer cells, however, develop mutations that enhance DNA damage response (DDR) and suppress cell death pathways. Additionally, interactions between tumor cells and tumor microenvironment (TME) components—including immune cells, stromal cells, and molecular mediators such as cytokines, chemokines, and growth factors—contribute to resistance against IR. Understanding these intricate relationships reveals potential targets to improve radiotherapy outcomes. Promising targets include DDR pathways, immunosuppressive cells and molecules, hypoxia, proangiogenic mediators, and other key signaling pathways. This review discusses emerging strategies, such as combining radiotherapy with immunomodulators, hypoxia and proangiogenic inhibitors, DDR-targeting agents, and other innovative approaches. By offering a comprehensive analysis of the lung TME, this review underscores opportunities to enhance radiotherapy effectiveness through targeted radiosensitization strategies.

{"title":"Unveiling Novel Targets in Lung Tumors for Enhanced Radiotherapy Efficacy: A Comprehensive Review","authors":"Faris Anad Muhammad,&nbsp;Ayat Hussein Adhab,&nbsp;Morug Salih Mahdi,&nbsp;Vicky Jain,&nbsp;Subbulakshmi Ganesan,&nbsp;Deepak Bhanot,&nbsp;K. Satyam Naidu,&nbsp;Sharnjeet Kaur,&nbsp;Aseel Salah Mansoor,&nbsp;Usama Kadem Radi,&nbsp;Nasr Saadoun Abd,&nbsp;Muthena Kariem","doi":"10.1002/jbt.70180","DOIUrl":"https://doi.org/10.1002/jbt.70180","url":null,"abstract":"<div>\u0000 \u0000 <p>Radiotherapy is a cornerstone of lung cancer management, though its efficacy is frequently undermined by intrinsic and acquired radioresistance. This review examines the complexity of lung tumors, highlighting their potential as a reservoir of novel targets for radiosensitization. Ionizing radiation (IR) primarily exerts its effects through oxidative damage and DNA double-strand breaks (DSBs). Lung cancer cells, however, develop mutations that enhance DNA damage response (DDR) and suppress cell death pathways. Additionally, interactions between tumor cells and tumor microenvironment (TME) components—including immune cells, stromal cells, and molecular mediators such as cytokines, chemokines, and growth factors—contribute to resistance against IR. Understanding these intricate relationships reveals potential targets to improve radiotherapy outcomes. Promising targets include DDR pathways, immunosuppressive cells and molecules, hypoxia, proangiogenic mediators, and other key signaling pathways. This review discusses emerging strategies, such as combining radiotherapy with immunomodulators, hypoxia and proangiogenic inhibitors, DDR-targeting agents, and other innovative approaches. By offering a comprehensive analysis of the lung TME, this review underscores opportunities to enhance radiotherapy effectiveness through targeted radiosensitization strategies.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143475583","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
Low-Power Blue LED Modulates NF-κB and Proinflammatory Cytokines in Doxorubicin-Treated MDA-MB-231 Cells
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-23 DOI: 10.1002/jbt.70192
Adilson Fonseca Teixeira, Bruno Ricardo Barreto Pires, Carolina Panis, Andréa Monte-Alto-Costa, Adenilson de Souza da Fonseca, Andre Luiz Mencalha

Doxorubicin is a crucial chemotherapy used in the treatment of triple-negative breast cancer (TNBC) patients, but elevated doxorubicin doses may induce therapeutic resistance. To overcome this limitation, we have previously established a photodynamic therapeutic (PDT)-like strategy that irradiates doxorubicin-treated cells with a low-power nonionizing blue LED device. This combined treatment increases the production of reactive oxygen species to promote cell death, consequently enabling reduced doxorubicin dosages. Yet, precisely determining the molecular mechanisms that drive this outcome is still required for advancing such PDT-like approach. Here, we aimed to correlate the expression of the inflammatory markers NF-κB, IL-8, IL-6, and IL-1β with the survival of TNBC cells submitted to our PDT-like protocol. Our results show that NF-κB/p65 nuclear levels were enhanced in MDA-MB-231 cells treated with doxorubicin and blue LED. Moreover, this PDT-like strategy increased IL-6 mRNA levels in MDA-MB-231 cells. IL-1β and IL-8 mRNA were upregulated in samples incubated with doxorubicin regardless of concomitant irradiation with blue LED. These results show that our PDT-like protocol is effective in elevating inflammatory signals, shedding light on the molecular mechanisms that underlie the efficacy of this innovative anticancer therapeutic approach.

{"title":"Low-Power Blue LED Modulates NF-κB and Proinflammatory Cytokines in Doxorubicin-Treated MDA-MB-231 Cells","authors":"Adilson Fonseca Teixeira,&nbsp;Bruno Ricardo Barreto Pires,&nbsp;Carolina Panis,&nbsp;Andréa Monte-Alto-Costa,&nbsp;Adenilson de Souza da Fonseca,&nbsp;Andre Luiz Mencalha","doi":"10.1002/jbt.70192","DOIUrl":"https://doi.org/10.1002/jbt.70192","url":null,"abstract":"<div>\u0000 \u0000 <p>Doxorubicin is a crucial chemotherapy used in the treatment of triple-negative breast cancer (TNBC) patients, but elevated doxorubicin doses may induce therapeutic resistance. To overcome this limitation, we have previously established a photodynamic therapeutic (PDT)-like strategy that irradiates doxorubicin-treated cells with a low-power nonionizing blue LED device. This combined treatment increases the production of reactive oxygen species to promote cell death, consequently enabling reduced doxorubicin dosages. Yet, precisely determining the molecular mechanisms that drive this outcome is still required for advancing such PDT-like approach. Here, we aimed to correlate the expression of the inflammatory markers NF-κB, IL-8, IL-6, and IL-1β with the survival of TNBC cells submitted to our PDT-like protocol. Our results show that NF-κB/p65 nuclear levels were enhanced in MDA-MB-231 cells treated with doxorubicin and blue LED. Moreover, this PDT-like strategy increased IL-6 mRNA levels in MDA-MB-231 cells. IL-1β and IL-8 mRNA were upregulated in samples incubated with doxorubicin regardless of concomitant irradiation with blue LED. These results show that our PDT-like protocol is effective in elevating inflammatory signals, shedding light on the molecular mechanisms that underlie the efficacy of this innovative anticancer therapeutic approach.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143475587","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
Methyl Donor Ameliorates CCl4-Induced Nephrotoxicity by Inhibiting Oxidative Stress, Inflammation, and Fibrosis Through the Attenuation of Kidney Injury Molecule 1 and Neutrophil Gelatinase-Associated Lipocalin Expression
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-23 DOI: 10.1002/jbt.70188
Nirmal Manhar, Sumeet Kumar Singh, Poonam Yadav, Manish Bishnolia, Amit Khurana, Jasvinder Singh Bhatti, Umashanker Navik

Carbon tetrachloride (CCl4), a volatile organic compound, is harmful to multi-organs, including the liver, lungs, muscles, and kidneys. Methyl donors, such as methionine, choline, betaine, and folic acid, are vital to one-carbon metabolism and have great potential to alleviate oxidative stress and inflammation, thus mitigating disease onset. Hence, the current study aims to examine the therapeutic effect of methyl donors against CCl4-induced nephrotoxicity. Nephrotoxicity was developed in male Sprague Dawley rats using CCl4 at a dose of 1 mL/kg (4-week model induction) twice a week via the intraperitoneal route. Thereafter, methyl donor treatments through oral gavage were given for the next 6 weeks with a continuation of CCl4 administration. Biochemical, oxidative stress parameters, histopathological, and qRT-PCR analyses were done at the completion of the 10-week. Biochemical analyses revealed that CCl4 induces nephrotoxicity, as evidenced by increased urea and creatinine levels and decreased albumin levels. These detrimental effects were significantly ameliorated by methyl donor treatment. Moreover, CCl4 decreased the antioxidant enzyme activity (superoxide dismutase; SOD and catalase; CAT) while increasing oxidative stress markers (malondialdehyde; MDA and nitrite). Methyl donor treatment effectively mitigated these oxidative changes. Histopathological analysis demonstrated the nephroprotective effect of methyl donors against CCl4-induced nephrotoxicity, showing reduced tissue damage and protection of renal architecture. At the molecular level, methyl donor treatment alleviated the CCl4-induced increase in kidney injury biomarkers (Kidney injury molecule 1; KIM-1 and Neutrophil gelatinase-associated lipocalin; NGAL), as well as inflammatory (IL-6 and TNF-α) and fibrosis-related genes (Acta-2 and TGF-β). In conclusion, our findings suggest that methyl donors possess anti-inflammatory and anti-fibrotic properties. They protect against CCl4-induced oxidative damage to renal cells, likely due to their reactive oxygen species scavenging capabilities and their ability to restore key early renal injury biomarkers (KIM-1 and NGAL). Methyl donors hold great promise as a cutting-edge therapy approach for preventing CCl4-induced nephrotoxicity.

{"title":"Methyl Donor Ameliorates CCl4-Induced Nephrotoxicity by Inhibiting Oxidative Stress, Inflammation, and Fibrosis Through the Attenuation of Kidney Injury Molecule 1 and Neutrophil Gelatinase-Associated Lipocalin Expression","authors":"Nirmal Manhar,&nbsp;Sumeet Kumar Singh,&nbsp;Poonam Yadav,&nbsp;Manish Bishnolia,&nbsp;Amit Khurana,&nbsp;Jasvinder Singh Bhatti,&nbsp;Umashanker Navik","doi":"10.1002/jbt.70188","DOIUrl":"https://doi.org/10.1002/jbt.70188","url":null,"abstract":"<div>\u0000 \u0000 <p>Carbon tetrachloride (CCl<sub>4</sub>), a volatile organic compound, is harmful to multi-organs, including the liver, lungs, muscles, and kidneys. Methyl donors, such as methionine, choline, betaine, and folic acid, are vital to one-carbon metabolism and have great potential to alleviate oxidative stress and inflammation, thus mitigating disease onset. Hence, the current study aims to examine the therapeutic effect of methyl donors against CCl<sub>4</sub>-induced nephrotoxicity. Nephrotoxicity was developed in male Sprague Dawley rats using CCl<sub>4</sub> at a dose of 1 mL/kg (4-week model induction) twice a week via the intraperitoneal route. Thereafter, methyl donor treatments through oral gavage were given for the next 6 weeks with a continuation of CCl<sub>4</sub> administration. Biochemical, oxidative stress parameters, histopathological, and qRT-PCR analyses were done at the completion of the 10-week. Biochemical analyses revealed that CCl<sub>4</sub> induces nephrotoxicity, as evidenced by increased urea and creatinine levels and decreased albumin levels. These detrimental effects were significantly ameliorated by methyl donor treatment. Moreover, CCl<sub>4</sub> decreased the antioxidant enzyme activity (superoxide dismutase; SOD and catalase; CAT) while increasing oxidative stress markers (malondialdehyde; MDA and nitrite). Methyl donor treatment effectively mitigated these oxidative changes. Histopathological analysis demonstrated the nephroprotective effect of methyl donors against CCl<sub>4</sub>-induced nephrotoxicity, showing reduced tissue damage and protection of renal architecture. At the molecular level, methyl donor treatment alleviated the CCl<sub>4</sub>-induced increase in kidney injury biomarkers (Kidney injury molecule 1; KIM-1 and Neutrophil gelatinase-associated lipocalin; NGAL), as well as inflammatory (IL-6 and TNF-α) and fibrosis-related genes (Acta-2 and TGF-β). In conclusion, our findings suggest that methyl donors possess anti-inflammatory and anti-fibrotic properties. They protect against CCl<sub>4</sub>-induced oxidative damage to renal cells, likely due to their reactive oxygen species scavenging capabilities and their ability to restore key early renal injury biomarkers (KIM-1 and NGAL). Methyl donors hold great promise as a cutting-edge therapy approach for preventing CCl<sub>4</sub>-induced nephrotoxicity.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143475590","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
METTL3-Mediated m6A Methylation Stabilizes IFI27 to Drive Esophageal Squamous Cell Carcinoma Progression Through an IGF2BP2-Dependent Mechanism
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-23 DOI: 10.1002/jbt.70167
Xinhua Zhang, Yu Bai, Linlin Shang, Yinghao Wang, Wenjian Yao, Sen Wu

Dysregulation of m6A modification has emerged as a vital factor in the development of esophageal squamous cell carcinoma (ESCC). Here, we sought to explore the critical role of m6A methylation mediated by the m6A methyltransferase METTL3 in ESCC. Protein expression analysis was performed by immunohistochemistry and immunoblot assays. The mRNA levels of METTL3 and IFI27 were detected by quantitative PCR. Cell sphere formation potential, migration, invasiveness, apoptosis, proliferation and viability were assessed by standard sphere formation, wound healing, transwell, flow cytometry, EdU and CCK-8 assays, respectively. The impact of METTL3 or IGF2BP2 on IFI27 mRNA was evaluated by methylated RNA immunoprecipitation (MeRIP), RIP or mRNA stability analysis. Xenograft assays were used to detect the in vivo function of METTL3. Elevated levels of METTL3 were observed in ESCC tumors and cells, and these increased levels were associated with the declined prognosis of ESCC. MELLT3 depletion impeded ESCC cell growth, invasiveness, migration, and sphere formation, and induced cell apoptosis in vitro. Elevated IFI27 expression was positively correlated with METTL3 levels in ESCC. Moreover, METTL3 mediated m6A methylation of IFI27 mRNA to stabilize the mRNA. The m6A reader IGF2BP2 also affected m6A methylation and expression of IFI27 mRNA. Additionally, IFI27 re-expression had a counteracting impact on the effects of METTL3 deficiency on in vitro ESCC cell behaviors and in vivo KYSE30 xenograft growth. Our findings demonstrate that METTL3-mediated IFI27 mRNA m6A methylation drives ESCC development through an IGF2BP2-dependent mechanism. Blocking the METTL3/IFI27 axis may be effective for preventing ESCC.

{"title":"METTL3-Mediated m6A Methylation Stabilizes IFI27 to Drive Esophageal Squamous Cell Carcinoma Progression Through an IGF2BP2-Dependent Mechanism","authors":"Xinhua Zhang,&nbsp;Yu Bai,&nbsp;Linlin Shang,&nbsp;Yinghao Wang,&nbsp;Wenjian Yao,&nbsp;Sen Wu","doi":"10.1002/jbt.70167","DOIUrl":"https://doi.org/10.1002/jbt.70167","url":null,"abstract":"<div>\u0000 \u0000 <p>Dysregulation of m6A modification has emerged as a vital factor in the development of esophageal squamous cell carcinoma (ESCC). Here, we sought to explore the critical role of m6A methylation mediated by the m6A methyltransferase METTL3 in ESCC. Protein expression analysis was performed by immunohistochemistry and immunoblot assays. The mRNA levels of METTL3 and IFI27 were detected by quantitative PCR. Cell sphere formation potential, migration, invasiveness, apoptosis, proliferation and viability were assessed by standard sphere formation, wound healing, transwell, flow cytometry, EdU and CCK-8 assays, respectively. The impact of METTL3 or IGF2BP2 on IFI27 mRNA was evaluated by methylated RNA immunoprecipitation (MeRIP), RIP or mRNA stability analysis. Xenograft assays were used to detect the in vivo function of METTL3. Elevated levels of METTL3 were observed in ESCC tumors and cells, and these increased levels were associated with the declined prognosis of ESCC. MELLT3 depletion impeded ESCC cell growth, invasiveness, migration, and sphere formation, and induced cell apoptosis in vitro. Elevated IFI27 expression was positively correlated with METTL3 levels in ESCC. Moreover, METTL3 mediated m6A methylation of IFI27 mRNA to stabilize the mRNA. The m6A reader IGF2BP2 also affected m6A methylation and expression of IFI27 mRNA. Additionally, IFI27 re-expression had a counteracting impact on the effects of METTL3 deficiency on in vitro ESCC cell behaviors and in vivo KYSE30 xenograft growth. Our findings demonstrate that METTL3-mediated IFI27 mRNA m6A methylation drives ESCC development through an IGF2BP2-dependent mechanism. Blocking the METTL3/IFI27 axis may be effective for preventing ESCC.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143475591","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
Assessment of the Cytotoxicity Mechanism of Diazinon on HFFF2 Cells: A Bioinformatic and Experimental Study
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-23 DOI: 10.1002/jbt.70146
Mehdi Sarailoo, Vahid Asghariazar, Sina Seifimansour, Mahtab Kadkhodayi, Erfan Zare, Parnia Vajdi, Mehdi Asghari Vostakolaei

Pesticide exposure can cause many skin diseases such as hypopigmentation and contact dermatitis, but the underlying mechanisms remain unclear. Furthermore, Organophosphate pesticides (OPs) including Diazinon (DZN) can affect cellular pathways like ATPase, leading to mitochondrial energy deficit and even apoptosis in the cell's functions. Following cell exposure to the OP pesticide DZN through treatment, we evaluated alteration in gene expression and DNA damage. Bioinformatic analysis was performed based on the AutoDock, Protein Data Bank, STRING, Way2Drug, and Comparative Toxicogenomics databases and tools. The MTT assay, wound healing, DAPI staining, flow cytometry, and real-time PCR were applied in the current study. The results showed that the viability and migration capacity of HFFF2 cells decreased, and the apoptosis rate increased in the DZN-treated group. These findings revealed that DZN regulated the expression of the apoptotic genes in DZN cells.

接触杀虫剂可导致色素沉着和接触性皮炎等多种皮肤病,但其潜在机制仍不清楚。此外,包括二嗪农(DZN)在内的有机磷农药(OPs)会影响 ATPase 等细胞通路,导致线粒体能量不足,甚至导致细胞功能凋亡。细胞暴露于 OP 杀虫剂 DZN 处理后,我们评估了基因表达和 DNA 损伤的变化。我们利用 AutoDock、蛋白质数据库、STRING、Way2Drug 和比较毒物基因组学数据库和工具进行了生物信息学分析。本研究采用了 MTT 试验、伤口愈合、DAPI 染色、流式细胞术和实时 PCR 技术。结果显示,DZN处理组HFFF2细胞的活力和迁移能力下降,凋亡率上升。这些研究结果表明,DZN调节了DZN细胞中凋亡基因的表达。
{"title":"Assessment of the Cytotoxicity Mechanism of Diazinon on HFFF2 Cells: A Bioinformatic and Experimental Study","authors":"Mehdi Sarailoo,&nbsp;Vahid Asghariazar,&nbsp;Sina Seifimansour,&nbsp;Mahtab Kadkhodayi,&nbsp;Erfan Zare,&nbsp;Parnia Vajdi,&nbsp;Mehdi Asghari Vostakolaei","doi":"10.1002/jbt.70146","DOIUrl":"https://doi.org/10.1002/jbt.70146","url":null,"abstract":"<div>\u0000 \u0000 <p>Pesticide exposure can cause many skin diseases such as hypopigmentation and contact dermatitis, but the underlying mechanisms remain unclear. Furthermore, Organophosphate pesticides (OPs) including Diazinon (DZN) can affect cellular pathways like ATPase, leading to mitochondrial energy deficit and even apoptosis in the cell's functions. Following cell exposure to the OP pesticide DZN through treatment, we evaluated alteration in gene expression and DNA damage. Bioinformatic analysis was performed based on the AutoDock, Protein Data Bank, STRING, Way2Drug, and Comparative Toxicogenomics databases and tools. The MTT assay, wound healing, DAPI staining, flow cytometry, and real-time PCR were applied in the current study. The results showed that the viability and migration capacity of HFFF2 cells decreased, and the apoptosis rate increased in the DZN-treated group. These findings revealed that DZN regulated the expression of the apoptotic genes in DZN cells.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143475588","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
Mechanism of the microRNA-373-3p/LATS2 Axis in the Prognosis and Metastasis of Thyroid Cancer Patients
IF 3.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-02-23 DOI: 10.1002/jbt.70181
Yingchao Gu, Hongbing Liu, Ming Shi, Fei Pu

This study focused on the role of the microRNA (miR)-373-3p/LATS2 axis in the prognosis and metastasis of thyroid cancer patients. miR-373-3p and LATS2 expression were assessed in thyroid cancer tissues and cells. The relationship between miR-373-3p and clinicopathological characteristics of patients with thyroid cancer and the impact of miR-373-3p and LATS2 expression levels on the survival and prognosis of thyroid cancer patients were analyzed. The targeting relationship between miR-373-3p and LATS2 was predicted and verified, and their impact on the malignant cell phenotype was assessed. Compared with adjacent normal tissues and normal human thyroid cells, miR-373-3p was highly expressed, while LATS2 was expressed at low levels in thyroid cancer tissues and cells (both p < 0.001). miR-373-3p expression was independent of age (p = 0.201) and gender (p = 0.516), and it was correlated with lymph node metastasis and TNM stage of thyroid cancer (both p < 0.001). Moreover, high miR-373-3p expression was associated with poor patient prognosis (p = 0.034). Interference with miR-373-3p or overexpression of LATS2 repressed KMH-2 cell malignant phenotypes (all p < 0.05). miR-373-3p targeted and suppressed LATS2 expression. Interference with miR-373-3p blocked its inhibition on LATS2, thereby repressing thyroid cancer progression and metastasis.

{"title":"Mechanism of the microRNA-373-3p/LATS2 Axis in the Prognosis and Metastasis of Thyroid Cancer Patients","authors":"Yingchao Gu,&nbsp;Hongbing Liu,&nbsp;Ming Shi,&nbsp;Fei Pu","doi":"10.1002/jbt.70181","DOIUrl":"https://doi.org/10.1002/jbt.70181","url":null,"abstract":"<div>\u0000 \u0000 <p>This study focused on the role of the microRNA (miR)-373-3p/LATS2 axis in the prognosis and metastasis of thyroid cancer patients. miR-373-3p and LATS2 expression were assessed in thyroid cancer tissues and cells. The relationship between miR-373-3p and clinicopathological characteristics of patients with thyroid cancer and the impact of miR-373-3p and LATS2 expression levels on the survival and prognosis of thyroid cancer patients were analyzed. The targeting relationship between miR-373-3p and LATS2 was predicted and verified, and their impact on the malignant cell phenotype was assessed. Compared with adjacent normal tissues and normal human thyroid cells, miR-373-3p was highly expressed, while LATS2 was expressed at low levels in thyroid cancer tissues and cells (both <i>p</i> &lt; 0.001). miR-373-3p expression was independent of age (<i>p</i> = 0.201) and gender (<i>p</i> = 0.516), and it was correlated with lymph node metastasis and TNM stage of thyroid cancer (both <i>p</i> &lt; 0.001). Moreover, high miR-373-3p expression was associated with poor patient prognosis (<i>p</i> = 0.034). Interference with miR-373-3p or overexpression of LATS2 repressed KMH-2 cell malignant phenotypes (all <i>p</i> &lt; 0.05). miR-373-3p targeted and suppressed LATS2 expression. Interference with miR-373-3p blocked its inhibition on LATS2, thereby repressing thyroid cancer progression and metastasis.</p></div>","PeriodicalId":15151,"journal":{"name":"Journal of Biochemical and Molecular Toxicology","volume":"39 3","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143475582","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
Journal of Biochemical and Molecular Toxicology
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