Cell Biology and Toxicology最新文献

筛选
英文 中文
NMF typing and machine learning algorithm-based exploration of preeclampsia-related mechanisms on ferroptosis signature genes. NMF分型和基于机器学习算法的子痫前期相关机制对铁下垂特征基因的探索。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-21 DOI: 10.1007/s10565-024-09963-5
Xuemin Liu, Di Zhang, Hui Qiu
{"title":"NMF typing and machine learning algorithm-based exploration of preeclampsia-related mechanisms on ferroptosis signature genes.","authors":"Xuemin Liu, Di Zhang, Hui Qiu","doi":"10.1007/s10565-024-09963-5","DOIUrl":"10.1007/s10565-024-09963-5","url":null,"abstract":"<p><strong>Background: </strong>Globally, pre-eclampsia (PE) poses a major threat to the health and survival of pregnant women and fetuses, contributing significantly to morbidity and mortality. Recent studies suggest a pathological link between PE and ferroptosis. We aim to utilize non-negative matrix factorization (NMF) clustering and machine learning algorithms to pinpoint disease-specific genes related to the process of ferroptosis in PE and investigate likely underlying biochemistry mechanisms.</p><p><strong>Methods: </strong>The acquisition of four microarray datasets from the Gene Expression Omnibus (GEO) repository, the integration of these datasets, and the elimination of batch effects formed the core procedure. Genes related to ferroptosis in PE (DE-FRG) were identified. NMF clustering was performed on DE-FRG for unsupervised analysis, generating a heatmap for clustering validation via principal component analysis. Immunocyte infiltration differences between different subtypes were compared to elucidate the impact of ferroptosis on immune infiltration in the placental tissue of PE patients. The application of weighted gene co-expression network analysis (WGCNA) revealed important module genes linked to sample subtypes and disease status. The screening of PE feature genes involved employing SVM, RF, GLM, and XGB machine learning algorithms, and their predictive performance was validated using various analyses and an external dataset. The iRegulon tool was utilized to predict upstream transcription factors associated with ferroptosis feature genes, from which differentially expressed transcription factors were screened to construct a \"Transcription Factor-FRG-ferroptosis\" regulatory network. Finally, in vitro (cultured cells) and in vivo (rat) models were utilized to evaluate the regulatory mechanisms of ferroptosis in normal and PE placental tissues.</p><p><strong>Results: </strong>Differential analysis of the four merged GEO datasets identified 41 DE-FRGs. NMF clustering based on DE-FRGs revealed two PE subtypes. Immunocyte infiltration analysis indicated significant differences in immune levels between these subtypes. Further WGCNA analysis identified module genes associated with PE and these two subtypes. Subsequently, we developed an integrated machine learning model incorporating five FRGs and validated its predictive efficacy using various analyses and an external validation dataset. Finally, based on the transcription factor ARID3A and ferroptosis feature genes EPHB3 and PAPPA2, we constructed a \"Transcription Factor-FRG-ferroptosis\" regulatory network, with in vitro and in vivo experiments confirming that ARID3A promotes the progression of PE and ferroptosis by activating the expression of EPHB3 and PAPPA2.</p><p><strong>Conclusion: </strong>This analytical journey illuminated a critical regulatory nexus in PE, underscoring the central influence of ARID3A on PE through ferroptosis-mediated pathways.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"14"},"PeriodicalIF":5.3,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662041/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871491","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
METTL3, m6A modification, and EGR1: interplay affecting myocardial I/R injury outcomes. METTL3、m6A修饰和EGR1:影响心肌I/R损伤结局的相互作用
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-21 DOI: 10.1007/s10565-024-09937-7
Chen Huang, Xun Zhang, Shi-Xiong Wu, Qing Chang, Zhi-Kun Zheng, Jing Xu
{"title":"METTL3, m6A modification, and EGR1: interplay affecting myocardial I/R injury outcomes.","authors":"Chen Huang, Xun Zhang, Shi-Xiong Wu, Qing Chang, Zhi-Kun Zheng, Jing Xu","doi":"10.1007/s10565-024-09937-7","DOIUrl":"10.1007/s10565-024-09937-7","url":null,"abstract":"<p><p>The occurrence of severe myocardial ischemia/reperfusion (I/R) injury is associated with the clinical application of reestablishment technique for heart disease, and understanding its underlying mechanisms is currently an urgent issue. Prior investigations have demonstrated the potential enhancement of MIRI through EGR1 suppression, although the precise underlying regulatory pathways require further elucidation. The core focus of this investigation is to examine the molecular pathways through EGR1 regulates mitophagy-mediated myocardial cell pyroptosis and its impact on MIRI. Cardiomyocyte hypoxia/reoxygenation (H/R) injury models and mouse models of myocardial I/R injury were used to investigate the involvement of EGR1 in regulating mitophagy-mediated myocardial cell pyroptosis in myocardial I/R injury. The research outcomes demonstrated that under H/R conditions, EGR1 expression was upregulated and inhibited the JAK2/STAT3 pathway, leading to enhanced mitophagy and disrupted mitochondrial fusion/fission dynamics, ultimately resulting in myocardial cell pyroptosis. Further research revealed that the upregulation of EGR1 expression was mediated by methyltransferase like 3 (METTL3)-mediated m6A modification of EGR1 mRNA and depended on the binding of insulin like growth factor 2 mrna binding protein 2 (IGF2BP2) to the N6-methyladenosine (m6A) modification site to enhance mRNA stability. In vivo animal experiments confirmed that METTL3 upregulated EGR1 expression through IGF2BP2 and suppressed activation of the janus kinase 2 (JAK2) /signal transducer and activator of transcription 3 (STAT3) pathway, thereby inhibiting mitophagy, disrupting mitochondrial dynamics, promoting myocardial cell pyroptosis, and exacerbating I/R injury.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"7"},"PeriodicalIF":5.3,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662061/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871487","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SS18-SSX drives TYK2 expression to activate STAT3/Bcl2 axis, facilitating apoptosis evasion and advancing synovial sarcoma progression. SS18-SSX驱动TYK2表达激活STAT3/Bcl2轴,促进细胞凋亡逃逸,促进滑膜肉瘤进展。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-21 DOI: 10.1007/s10565-024-09952-8
Wenjing Qin, Changliang Peng, Xianhe Yang, Alan Jiang, Nanshan Zhong, Yunyun Liu, Xiaochun Zhang, Angela C Hirbe, Mei Ma, Xin Yue
{"title":"SS18-SSX drives TYK2 expression to activate STAT3/Bcl2 axis, facilitating apoptosis evasion and advancing synovial sarcoma progression.","authors":"Wenjing Qin, Changliang Peng, Xianhe Yang, Alan Jiang, Nanshan Zhong, Yunyun Liu, Xiaochun Zhang, Angela C Hirbe, Mei Ma, Xin Yue","doi":"10.1007/s10565-024-09952-8","DOIUrl":"10.1007/s10565-024-09952-8","url":null,"abstract":"<p><p>Synovial sarcoma (SS) is a rare soft tissue sarcoma characterized by high-grade malignancy and poor prognosis. Preliminary research indicates that apoptosis evasion is a key factor in SS progression, primarily attributed to the overexpression of anti-apoptotic genes. However, the mechanisms underlying this phenomenon are still not fully understood. This study aims to investigate the factors responsible for apoptosis evasion, evaluate their potential as targets for anti-apoptotic interventions, and analyze their mechanisms in detail. Our findings reveal that tyrosine kinase 2 (TYK2) is upregulated in highly malignant SS. Through in vitro as well as in vivo functional analyses, we have demonstrated that, TYK2 significantly accelerates SS cells progression. Mechanistically, TYK2 activates STAT3, which promotes the expression of BCL2, an anti-apoptotic gene. Inhibition of STAT3 activation using specific inhibitors can disrupt the TYK2-enhanced expression of Bcl2, indicating that the TYK2/STAT3/Bcl2 axis is a key regulatory pathway mediating apoptosis evasion in SS. Furthermore, our investigation into the upstream regulation of TYK2 reveals that the fusion protein SS18-SSX enhances the transcriptional activity of TYK2 by binding to the promoter region of the TYK2 gene, thereby increasing its expression levels. Thus, the TYK2/STAT3/Bcl2 axis is a crucial mechanism through which SS18-SSX mediates apoptosis evasion in SS cells. In conclusion, our findings contribute to understanding how SS18-SSX-driven TYK2 expression mediates apoptosis evasion mechanisms and propose targeting TYK2 as a strategy to induce apoptosis in SS.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"8"},"PeriodicalIF":5.3,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662063/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871494","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
eEF2K alleviates doxorubicin-induced cardiotoxicity by inhibiting GSK3β and improving autophagy dysfunction. eEF2K通过抑制GSK3β和改善自噬功能障碍来减轻阿霉素诱导的心脏毒性。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-21 DOI: 10.1007/s10565-024-09966-2
Junjie Guan, Hongwei Mo, Vicheth Virak, Runze Guo, Dongdong Que, Wenjie Yu, Xuwei Zhang, Jing Yan, Yuxi Wang, Yashu Yang, Bowen Rui, Guanlin Huang, Deshu Chen, Chongbin Zhong, Pingzhen Yang
{"title":"eEF2K alleviates doxorubicin-induced cardiotoxicity by inhibiting GSK3β and improving autophagy dysfunction.","authors":"Junjie Guan, Hongwei Mo, Vicheth Virak, Runze Guo, Dongdong Que, Wenjie Yu, Xuwei Zhang, Jing Yan, Yuxi Wang, Yashu Yang, Bowen Rui, Guanlin Huang, Deshu Chen, Chongbin Zhong, Pingzhen Yang","doi":"10.1007/s10565-024-09966-2","DOIUrl":"10.1007/s10565-024-09966-2","url":null,"abstract":"<p><p>Doxorubicin-induced cardiotoxicity (DIC) poses a threat to the health and prognosis of cancer patients. It is important to find a safe and effective method for the prevention and treatment of DIC. eEF2K, which is a highly conserved α-kinase, is thought to be a therapeutic target for several human diseases. Nonetheless, it is still uncertain if eEF2K contributes to the cardiotoxic effects caused by doxorubicin (DOX). Our research revealed that eEF2K expression decreased in the DIC. eEF2K was overexpressed through adeno-associated virus in vivo and adenovirus in vitro, which presented alleviative cardiomyocyte death and cell atrophy induced by DOX. Autophagy dysfunction is one of important mechanisms in DIC. As a result, autophagic function was evaluated using Transmission electron microscopy in vivo, as well as LysoSensor and mRFPGFP-LC3 puncta in vitro. eEF2K overexpression improves DOX-induced autophagy blockade. In addition, eEF2K knockdown aggravated autophagy blockade and cardiomyocyte injury in DIC model. eEF2K also phosphorylated and inhibited GSK3β in DIC model. AR-A014418 (ARi), known for selectively inhibiting GSK3β, countered the effects of eEF2K knockdown, which aggravated autophagy blockade in the DIC. In conclusion, this study proposes that eEF2K alleviates DIC by inhibiting GSK3β and improving autophagy dysfunction. eEF2K is a promising therapeutic target against DIC.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"15"},"PeriodicalIF":5.3,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11663172/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting protein tyrosine phosphatase non-receptor type 6 (PTPN6) as a therapeutic strategy in acute myeloid leukemia. 靶向蛋白酪氨酸磷酸酶非受体6型(PTPN6)作为急性髓系白血病的治疗策略。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-21 DOI: 10.1007/s10565-024-09965-3
Xiaoou Wang, Zhenggang Li, Jing Shen, Lin Liu
{"title":"Targeting protein tyrosine phosphatase non-receptor type 6 (PTPN6) as a therapeutic strategy in acute myeloid leukemia.","authors":"Xiaoou Wang, Zhenggang Li, Jing Shen, Lin Liu","doi":"10.1007/s10565-024-09965-3","DOIUrl":"10.1007/s10565-024-09965-3","url":null,"abstract":"<p><p>Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by the clonal expansion of myeloid progenitor cells. Despite advancements in treatment, the prognosis for AML patients remains poor, highlighting the need for novel therapeutic targets. Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6), also known as SHP-1, is a critical regulator of hematopoietic cell signaling and has been implicated in various leukemias. This study investigates the therapeutic potential of targeting PTPN6 in AML. We employed both in vitro and in vivo models to evaluate the effects of PTPN6 inhibition on AML cell proliferation, apoptosis, and differentiation. Our results demonstrate that PTPN6 inhibition leads to a significant reduction in AML cell viability, induces apoptosis, and promotes differentiation of leukemic cells into mature myeloid cells. Mechanistic studies revealed that PTPN6 inhibition disrupts key signaling pathways involved in AML pathogenesis, including the JAK/STAT and PI3K/AKT pathways. Furthermore, the combination of PTPN6 inhibitors with standard chemotherapeutic agents exhibited a synergistic effect, enhancing the overall therapeutic efficacy. These findings suggest that PTPN6 is a promising therapeutic target in AML and warrants further investigation into the development of PTPN6 inhibitors for clinical application in AML treatment.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"11"},"PeriodicalIF":5.3,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662038/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exercise training alleviates neuronal apoptosis and re-establishes mitochondrial quality control after cerebral ischemia by increasing SIRT3 expression. 运动训练通过提高SIRT3表达,减轻脑缺血后神经元凋亡,重建线粒体质量控制。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-21 DOI: 10.1007/s10565-024-09957-3
Wenwen Wu, Zengyu Wei, Zhiyun Wu, Jianmin Chen, Ji Liu, Manli Chen, Jinjin Yuan, Zhijian Zheng, Zijun Zhao, Qiang Lin, Nan Liu, Hongbin Chen
{"title":"Exercise training alleviates neuronal apoptosis and re-establishes mitochondrial quality control after cerebral ischemia by increasing SIRT3 expression.","authors":"Wenwen Wu, Zengyu Wei, Zhiyun Wu, Jianmin Chen, Ji Liu, Manli Chen, Jinjin Yuan, Zhijian Zheng, Zijun Zhao, Qiang Lin, Nan Liu, Hongbin Chen","doi":"10.1007/s10565-024-09957-3","DOIUrl":"10.1007/s10565-024-09957-3","url":null,"abstract":"<p><p>Existing evidence indicates that exercise training can enhance neural function by regulating mitochondrial quality control (MQC), which can be impaired by cerebral ischemia, and that sirtuin-3 (SIRT3), a protein localized in mitochondria, is crucial in maintaining mitochondrial functions. However, the relationship among exercise training, SIRT3, and MQC after cerebral ischemia remains obscure. This study attempted to elucidate the relationship among exercise training, SIRT3 and MQC after cerebral ischemia in rats. Male adult SD rats received tMCAO after the transfection of adeno-associated virus encoding either sirtuin-3 (AAV-SIRT3) or SIRT3 knockdown (AAV-sh-SIRT3) into the ipsilateral striata and cortex. Subsequently, the animals were randomly selected for exercise training. The index changes were measured by transmission electron microscopy, Western blot analysis, nuclear magnetic resonance imaging, TUNEL staining, and immunofluorescence staining, etc. The results revealed that after cerebral ischemia, exercise training increased SIRT3 expression, significantly improved neural function, alleviated infarct volume and neuronal apoptosis, maintained the mitochondrial structural integrity, and re-established MQC. The latter promoted mitochondrial biogenesis, balanced mitochondrial fission/fusion, and enhanced mitophagy. These favorable benefits were reversed after SIRT3 interference. In addition, a cellular OGD/R model showed that the increased SIRT3 expression alleviates neuronal apoptosis and re-establishes mitochondrial quality control by activating the β-catenin pathway. These findings suggest that exercise training may optimize mitochondrial quality control by increasing the expression of SIRT3, thereby improving neural functions after cerebral ischemia, which illuminates the mechanism underlying the exercise training-conferred neural benefits and indicates SIRT3 as a therapeutic strategy for brain ischemia.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"10"},"PeriodicalIF":5.3,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662049/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871558","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
miR-21-loaded bone marrow mesenchymal stem cell-derived exosomes inhibit pyroptosis by targeting MALT1 to repair chemotherapy-induced premature ovarian insufficiency. 负载mir -21的骨髓间充质干细胞衍生的外泌体通过靶向MALT1修复化疗诱导的卵巢早衰来抑制焦亡。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-20 DOI: 10.1007/s10565-024-09946-6
Lichao Tang, Yutao Yang, Mingxin Yang, Jiaxin Xie, Aiping Zhuo, Yanhong Wu, Mengli Mao, Youhong Zheng, Xiafei Fu
{"title":"miR-21-loaded bone marrow mesenchymal stem cell-derived exosomes inhibit pyroptosis by targeting MALT1 to repair chemotherapy-induced premature ovarian insufficiency.","authors":"Lichao Tang, Yutao Yang, Mingxin Yang, Jiaxin Xie, Aiping Zhuo, Yanhong Wu, Mengli Mao, Youhong Zheng, Xiafei Fu","doi":"10.1007/s10565-024-09946-6","DOIUrl":"10.1007/s10565-024-09946-6","url":null,"abstract":"<p><p>Chemotherapy is essential for treating malignant tumors, but it can cause premature ovarian insufficiency (POI). Recent studies suggest that exosomes enriched with miR-21 (miR-21-Exo) may help mitigate POI, though the underlying mechanisms remain largely unexplored. This research investigates how miR-21-Exo influences chemotherapy-induced POI using an experimental model where KGN cells are exposed to cisplatin. We assessed the impact of miR-21 on cellular activity and generated miR-21 overexpressing bone marrow mesenchymal stem cells (miR-21-BMSC) via lentiviral modification. Isolated miR-21-Exo was analyzed for its effects on cellular function. Bioinformatics identified Mucosa-Associated Lymphoid Tissue Lymphoma Translocation Protein 1 (MALT1) as a target of miR-21. We confirmed that miR-21-Exo regulates MALT1 and the NF-κB signaling pathway to prevent cell pyroptosis. Further studies in a rat model demonstrated the therapeutic potential and safety of miR-21-Exo. Overall, our findings highlight a novel strategy for addressing chemotherapy-induced POI by modulating MALT1 and the NF-κB pathway, offering significant therapeutic implications.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"3"},"PeriodicalIF":5.3,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662076/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871489","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
JAG1 mediates apoptosis in herpes simplex keratitis by suppressing autophagy via ROS/JAG1/NOTCH1/pULK1 signaling pathway. JAG1通过ROS/JAG1/NOTCH1/pULK1信号通路抑制自噬,介导单纯疱疹性角膜炎细胞凋亡。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-20 DOI: 10.1007/s10565-024-09968-0
Jingyao Chang, Yao Yao, Xinghong Sun, Wenzhe Wang, Haochen Qian, Yumeilan Liu, Chunyan Xue, Wei Ye, Feng Jiang
{"title":"JAG1 mediates apoptosis in herpes simplex keratitis by suppressing autophagy via ROS/JAG1/NOTCH1/pULK1 signaling pathway.","authors":"Jingyao Chang, Yao Yao, Xinghong Sun, Wenzhe Wang, Haochen Qian, Yumeilan Liu, Chunyan Xue, Wei Ye, Feng Jiang","doi":"10.1007/s10565-024-09968-0","DOIUrl":"10.1007/s10565-024-09968-0","url":null,"abstract":"<p><p>Herpes simplex keratitis (HSK), an ocular disease resulted from herpes simplex virus type 1 (HSV-1) infection, leads to the majority of infectious corneal blindness worldwide. The apoptosis of corneal epithelial cells (CECs) resulted from HSV-1 disrupts the epithelial barrier and exacerbates the infection; however, there is no definitive cure for HSK. Jagged1 (JAG1), one of the primary functional ligands for NOTCH receptors, plays a crucial role in regulating apoptosis and autophagy; however, its role in HSK is unclear. Our transcriptome analysis showed JAG1 was significantly upregulated in HSV-1-infected human CECs. We aimed to explore JAG1's role in regulating apoptosis in HSV-1-infected human CECs and in HSK mice. HSV-1 infection induced apoptosis and reactive oxygen species (ROS) generation in CECs. HSV-1 also activated the JAG1/NOTCH1 signaling pathway. The ROS scavenger N-acetylcysteine significantly mitigated these effects. Additionally, inhibiting the JAG1/NOTCH1 pathway with short hairpin RNA against JAG1 or a NOTCH1 inhibitor (N-[N-{3,5-difuorophenacetyl}-1-alanyl]-S-phenylglycine t-butyl ester [DAPT]) alleviated HSV-1-induced CEC apoptosis. Transmission electron microscopy and western blotting revealed that HSV-1 infection suppressed ULK1-mediated autophagy in CECs, while DAPT treatment enhanced autophagy by suppressing ULK1 phosphorylation. The activation of autophagy by rapamycin treatment markedly reduced ROS levels and apoptosis in HSV-1-infected CECs, revealing a synergistic effect between the suppressed autophagy and increased ROS levels, ultimately leading to apoptosis. Thus, HSV-1 induces CEC apoptosis by suppressing autophagy through ROS/JAG1/NOTCH1/pULK1 signaling pathway in vitro and in vivo, providing potential therapeutic targets for HSK.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"1"},"PeriodicalIF":5.3,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662045/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142863520","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nrf2/NRF1 signaling activation and crosstalk amplify mitochondrial biogenesis in the treatment of triptolide-induced cardiotoxicity using calycosin. Nrf2/NRF1信号激活和串扰在使用毛蕊异黄酮治疗雷公藤甲素诱导的心脏毒性中放大线粒体生物发生。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-20 DOI: 10.1007/s10565-024-09969-z
Xiao-Ming Qi, Wei-Zheng Zhang, Yu-Qin Zuo, Yuan-Biao Qiao, Yuan-Lin Zhang, Jin-Hong Ren, Qing-Shan Li
{"title":"Nrf2/NRF1 signaling activation and crosstalk amplify mitochondrial biogenesis in the treatment of triptolide-induced cardiotoxicity using calycosin.","authors":"Xiao-Ming Qi, Wei-Zheng Zhang, Yu-Qin Zuo, Yuan-Biao Qiao, Yuan-Lin Zhang, Jin-Hong Ren, Qing-Shan Li","doi":"10.1007/s10565-024-09969-z","DOIUrl":"10.1007/s10565-024-09969-z","url":null,"abstract":"<p><p>Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates both oxidative stress and mitochondrial biogenesis. Our previous study reported the cardioprotection of calycosin against triptolide toxicity through promoting mitochondrial biogenesis by activating nuclear respiratory factor 1 (NRF1), a coregulatory effect contributed by Nrf2 was not fully elucidated. This work aimed at investigating the involvement of Nrf2 in mitochondrial protection and elucidating Nrf2/NRF1 signaling crosstalk on amplifying the detoxification of calycosin. Results indicated that calycosin inhibited cardiomyocytes apoptosis and F-actin depolymerization following triptolide exposure. Cardiac contraction was improved by calycosin through increasing both fractional shortening (FS%) and ejection fraction (EF%). This enhanced contractile capacity of heart was benefited from mitochondrial protection reflected by ultrastructure improvement, augment in mitochondrial mass and ATP production. NRF1 overexpression in cardiomyocytes increased mitochondrial mass and DNA copy number, whereas NRF1 knockdown mitigated calycosin-mediated enhancement in mitochondrial mass. For nuclear Nrf2, it was upregulated by calycosin in a way of disrupting Nrf2-Keap1 (Kelch-like ECH associated protein 1) interaction, followed by inhibiting ubiquitination and degradation. The involvement of Nrf2 in mitochondrial protection was validated by the results that both Nrf2 knockdown and Nrf2 inhibitor blocked the calycosin effects on mitochondrial biogenesis and respiration. In the case of calycosin treatment, its effect on NRF1 and Nrf2 upregulations were respectively blocked by PGCα/Nrf2 and NRF1 knockdown, indicative of the mutual regulation between Nrf2 and NRF1. Accordingly, calycosin activated Nrf2/NRF1 and the signaling crosstalk, leading to mitochondrial biogenesis amplification, which would become a novel mechanism of calycosin against triptolide-induced cardiotoxicity.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"2"},"PeriodicalIF":5.3,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662064/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871492","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The mechanism of sevoflurane affecting ovarian cancer cell proliferation and migration by regulating RNA methylase TRDMT1 to activate the β-catenin pathway. 七氟醚通过调控RNA甲基化酶TRDMT1激活β-catenin通路影响卵巢癌细胞增殖和迁移的机制。
IF 5.3 2区 医学
Cell Biology and Toxicology Pub Date : 2024-12-04 DOI: 10.1007/s10565-024-09941-x
Xiaochen Huang, Xuewei Lao, Chengyan He, Jia Wang, Ying Pan
{"title":"The mechanism of sevoflurane affecting ovarian cancer cell proliferation and migration by regulating RNA methylase TRDMT1 to activate the β-catenin pathway.","authors":"Xiaochen Huang, Xuewei Lao, Chengyan He, Jia Wang, Ying Pan","doi":"10.1007/s10565-024-09941-x","DOIUrl":"10.1007/s10565-024-09941-x","url":null,"abstract":"<p><strong>Objective: </strong>Sevoflurane (Sevo), a commonly used inhalant anesthetic clinically, is associated with a worsened cancer prognosis, and we investigated its effect on RNA methylase tRNA aspartic acid methyltransferase 1 (TRDMT1) expression and ovarian cancer (OC) cell malignant phenotypes.</p><p><strong>Methods: </strong>Human OC cells (OVCAR3/SKOV3) were pretreated with 3.6% Sevo and cultured under normal conditions for 48 h, with their viability assessed. After 2-h Sevo treatment or interference plasmid transfections to down-regulate TRDMT1/adenomatous polyposis coli (APC), changes in TRDMT1, APC and β-catenin expression, cell proliferative activity, cycle, apoptosis, migration, invasion, and 5-methylcytosine (m5C) methylation potential modification sites were evaluated. Additionally, APC mRNA m5C methylation level and stability, the binding of APC mRNA with TRDMT1, the binding intensity of APC and β-catenin, and β-catenin nuclear translocation were detected Lastly, Cyclin D1, cellular-myelocytomatosis viral oncogene (C-myc) and β-catenin protein levels, and ki67-positive rate were assessed.</p><p><strong>Results: </strong>Sevo treatment boosted cell cycle, proliferation, migration and invasion, suppressed apoptosis and APC expression, and up-regulated C-myc, β-catenin, TRDMT1 and Cyclin D1 levels. Silencing TRDMT1 or β-catenin partially averted Sevo-mediated promotion effects on cell malignant biological behaviors. Lowly-expressed APC annulled the effect of silencing TRDMT1 and promoted cell malignant behaviors. Sevo enhanced APC mRNA m5C modification and degradation and activated the APC/β-catenin pathway by increasing TRDMT1, thus encouraging OC growth in vivo.</p><p><strong>Conclusions: </strong>Sevo stimulated APC m5C modification and curbed its expression by up-regulating TRDMT1, which in turn activated the β-catenin pathway to stimulate OC cell cycle, invasion, proliferation, and migration and to suppress apoptosis.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"40 1","pages":"108"},"PeriodicalIF":5.3,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11618209/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142766572","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信