International Journal of Biological Sciences最新文献

筛选
英文 中文
Exosomal SLC16A1-AS1-induced M2 macrophages polarization facilitates hepatocellular carcinoma progression. 外泌体 SLC16A1-AS1 诱导的 M2 巨噬细胞极化促进了肝细胞癌的进展。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-12 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.94440
Yuhang Hu, Yang Li, Hewei Xiong, Ya Zhang, Fan Wang, Wenfeng Zhuo, Zhu Zeng, Yong Zhao, Hongda Wang, Ping Hu, Shengbo Han, Yan Huang, Guozheng Lv, Gang Zhao
{"title":"Exosomal SLC16A1-AS1-induced M2 macrophages polarization facilitates hepatocellular carcinoma progression.","authors":"Yuhang Hu, Yang Li, Hewei Xiong, Ya Zhang, Fan Wang, Wenfeng Zhuo, Zhu Zeng, Yong Zhao, Hongda Wang, Ping Hu, Shengbo Han, Yan Huang, Guozheng Lv, Gang Zhao","doi":"10.7150/ijbs.94440","DOIUrl":"https://doi.org/10.7150/ijbs.94440","url":null,"abstract":"<p><p>Macrophages are the most abundant alternative immune cells in the tumor microenvironment (TME). The cross-talk between macrophages and tumor cells provides an important shelter for the occurrence and development of tumors. As an important information transfer medium, exosomes play an important role in intercellular communication. Nonetheless, how exosomal lncRNAs coordinate the communication between tumor cells and immune cells in hepatocellular carcinoma (HCC) is incompletely understood. We found that HCC exosomes-derived antisense RNA of SLC16A1(SLC16A1-AS1) promoted the malignant progression of HCC by regulating macrophage M2-type polarization. Mechanistically, the HCC exosomal SLC16A1-AS1 enhanced mRNA stabilization of SLC16A1 in macrophage by promoting the interaction between 3' untranslated regions (3'UTR) of SLC16A1 mRNA and heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1). As a lactate transporter, SLC16A1 accelerated lactate influx and then activated c-Raf/ERK signaling to induce M2 polarization of macrophages. Reciprocally, M2 macrophages secreted IL-6 to activate STAT3 and then induce METTL3 transcription in HCC cells, which increasing m6A methylation and stabilization of SLC16A1-AS1. In turn, the reciprocal SLC16A1-AS1/IL-6 signaling between HCC cells and M2 macrophages promoted the proliferation, invasion and glycolysis of HCC cells. Our study highlights that exosomal SLC16A1-AS1 acts as a signaling message that induces lactate-mediated M2 polarization of macrophages, and implies that SLC16A1-AS1 might be an applicable target for therapeutic treatment of HCC.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379075/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
RUNX1-PDIA5 Axis Promotes Malignant Progression of Glioblastoma by Regulating CCAR1 Protein Expression. RUNX1-PDIA5轴通过调控CCAR1蛋白表达促进胶质母细胞瘤恶性进展
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-12 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.92595
Qiankun Ji, Zewei Tu, Junzhe Liu, Zhihong Zhou, Fengze Li, Xingen Zhu, Kai Huang
{"title":"RUNX1-PDIA5 Axis Promotes Malignant Progression of Glioblastoma by Regulating CCAR1 Protein Expression.","authors":"Qiankun Ji, Zewei Tu, Junzhe Liu, Zhihong Zhou, Fengze Li, Xingen Zhu, Kai Huang","doi":"10.7150/ijbs.92595","DOIUrl":"https://doi.org/10.7150/ijbs.92595","url":null,"abstract":"<p><p>PDIA5 is responsible for modification of disulfide bonds of proteins. However, its impact on the malignant progression of glioblastoma multiforme (GBM) remains unknown. We analyzed the expression and prognostic significance of PDIA5 in cohorts of GBM and clinical samples. The PDIA5 protein was significantly overexpressed in GBM tissues, and higher expression of PDIA5 was statistically associated with a worse prognosis in patients with GBM. Transcriptional data from PDIA5 knockdown GBM cells revealed that downstream regulatory genes of PDIA5 were enriched in malignant regulatory pathways and PDIA5 enhanced the proliferative and invasive abilities of GBM cells. By constructing a PDIA5 CXXC motif mutant plasmid, we found CCAR1 was the vital downstream factor of PDIA5 in regulating GBM malignancy <i>in vitro</i> and <i>in vivo</i>. Additionally, RUNX1 bound to the promoter region of PDIA5 and regulated gene transcription, leading to activation of the PDIA5/CCAR1 regulatory axis in GBM. The RUNX1/PDIA5/CCAR1 axis significantly influenced the malignant behavior of GBM cells. In conclusion, this study comprehensively elucidates the crucial role of PDIA5 in the malignant progression of GBM. Downregulating PDIA5 can mitigate the malignant biological behavior of GBM both <i>in vitro</i> and <i>in vivo</i>, potentially improving the efficacy of treatment for clinical patients with GBM.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379074/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142157180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ruxolitinib-based senomorphic therapy mitigates cardiomyocyte senescence in septic cardiomyopathy by inhibiting the JAK2/STAT3 signaling pathway. 基于Ruxolitinib的衰老疗法通过抑制JAK2/STAT3信号通路减轻脓毒症心肌病中心肌细胞的衰老。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-12 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.96489
Boshen Yang, Taixi Li, Zhixiang Wang, Yuankang Zhu, Kaifan Niu, Sien Hu, Zhiqi Lin, Xinjie Zheng, Xian Jin, Chengxing Shen
{"title":"Ruxolitinib-based senomorphic therapy mitigates cardiomyocyte senescence in septic cardiomyopathy by inhibiting the JAK2/STAT3 signaling pathway.","authors":"Boshen Yang, Taixi Li, Zhixiang Wang, Yuankang Zhu, Kaifan Niu, Sien Hu, Zhiqi Lin, Xinjie Zheng, Xian Jin, Chengxing Shen","doi":"10.7150/ijbs.96489","DOIUrl":"https://doi.org/10.7150/ijbs.96489","url":null,"abstract":"<p><p><b>Background:</b> Cellular senescence has emerged as a pivotal focus in cardiovascular research. This study investigates the previously unrecognized role of cellular senescence in septic cardiomyopathy (SCM) and evaluates senomorphic therapy using ruxolitinib (Rux) as a potential treatment option. <b>Methods:</b> We employed lipopolysaccharide (LPS)-induced neonatal rat cardiomyocytes (NRCMs) and two mouse models-LPS-induced and cecal ligation and puncture (CLP)-induced SCM models-to assess Rux's effects. RNA sequencing, western blotting (WB), quantitative polymerase chain reaction (qPCR), immunofluorescence, immunohistochemistry, senescence-associated β-galactosidase (SA-β-gal) assay, and other techniques were utilized to investigate underlying mechanisms. <b>Results:</b> Senescence-associated secretory phenotype (SASP) and cellular senescence markers were markedly elevated in LPS-induced NRCMs and SCM animal models, confirmed by the SA-β-gal assay. Rux treatment attenuated SASP <i>in vitro</i> and <i>in vivo</i>, alongside downregulation of senescence markers. Moreover, Rux-based senomorphic therapy mitigated mitochondrial-mediated apoptosis, improved cardiac function in SCM mice, restored the balance of antioxidant system, and reduced reactive oxygen species (ROS) levels. Rux treatment restored mitochondrial membrane potential, mitigated mitochondrial morphological damage, and upregulated mitochondrial complex-related gene expression, thereby enhancing mitochondrial function. Additionally, Rux treatment ameliorated SCM-induced mitochondrial dynamic dysfunction and endoplasmic reticulum stress. Mechanistically, Rux inhibited JAK2-STAT3 signaling activation both <i>in vitro</i> and <i>in vivo</i>. Notably, low-dose Rux and ABT263 showed comparable efficacy in mitigating SCM. <b>Conclusions:</b> This study highlighted the potential significance of cellular senescence in SCM pathogenesis and suggested Rux-based senomorphic therapy as a promising therapeutic approach for SCM.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379065/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153952","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhancing mitophagy by ligustilide through BNIP3-LC3 interaction attenuates oxidative stress-induced neuronal apoptosis in spinal cord injury. 利格列奈通过 BNIP3-LC3 相互作用增强有丝分裂,可减轻氧化应激诱导的脊髓损伤神经细胞凋亡。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-12 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.98051
Hui Yao, Chaoyang Cai, Weijun Huang, Caizhen Zhong, Tianlun Zhao, Jiawei Di, Juliang Tang, Depeng Wu, Mao Pang, Lei He, Limin Rong, Bin Liu
{"title":"Enhancing mitophagy by ligustilide through BNIP3-LC3 interaction attenuates oxidative stress-induced neuronal apoptosis in spinal cord injury.","authors":"Hui Yao, Chaoyang Cai, Weijun Huang, Caizhen Zhong, Tianlun Zhao, Jiawei Di, Juliang Tang, Depeng Wu, Mao Pang, Lei He, Limin Rong, Bin Liu","doi":"10.7150/ijbs.98051","DOIUrl":"https://doi.org/10.7150/ijbs.98051","url":null,"abstract":"<p><p>Mitophagy selectively eliminates damaged or dysfunctional mitochondria, playing a crucial role in maintaining mitochondrial quality control. However, it remains unclear whether mitophagy can be fully activated and how it evolves after SCI. Our RNA-seq analysis of animal samples from sham and 1, 3, 5, and 7 days post-SCI indicated that mitophagy was indeed inhibited during the acute and subacute early stages. <i>In vitro</i> experiments showed that this inhibition was closely related to excessive production of reactive oxygen species (ROS) and the downregulation of BNIP3. Excessive ROS led to the blockage of mitophagy flux, accompanied by further mitochondrial dysfunction and increased neuronal apoptosis. Fortunately, ligustilide (LIG) was found to have the ability to reverse the oxidative stress-induced downregulation of BNIP3 and enhance mitophagy through BNIP3-LC3 interaction, alleviating mitochondrial dysfunction and ultimately reducing neuronal apoptosis. Further animal experiments demonstrated that LIG alleviated oxidative stress and mitophagy inhibition, rescued neuronal apoptosis, and promoted tissue repair, ultimately leading to improved motor function. In summary, this study elucidated the state of mitophagy inhibition following SCI and its potential mechanisms, and confirmed the effects of LIG-enhanced mitophagy through BNIP3-LC3, providing new therapeutic targets and strategies for repairing SCI.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379069/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153908","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Interaction Between the PERK/ATF4 Branch of the Endoplasmic Reticulum Stress and Mitochondrial One-Carbon Metabolism Regulates Neuronal Survival After Intracerebral Hemorrhage. 内质网应激的 PERK/ATF4 分支与线粒体一碳代谢之间的相互作用可调控脑出血后神经元的存活。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-06 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.93787
Yikui Liu, Fengzhen Cui, Aoqian Xu, Baofeng Wang, Yuxiao Ma, Qixiang Zhang, Qingfang Sun, Yongtao Zheng, Yuxiao Xue, Yuhao Sun, Liuguan Bian
{"title":"Interaction Between the PERK/ATF4 Branch of the Endoplasmic Reticulum Stress and Mitochondrial One-Carbon Metabolism Regulates Neuronal Survival After Intracerebral Hemorrhage.","authors":"Yikui Liu, Fengzhen Cui, Aoqian Xu, Baofeng Wang, Yuxiao Ma, Qixiang Zhang, Qingfang Sun, Yongtao Zheng, Yuxiao Xue, Yuhao Sun, Liuguan Bian","doi":"10.7150/ijbs.93787","DOIUrl":"https://doi.org/10.7150/ijbs.93787","url":null,"abstract":"<p><p>Recent investigations have revealed that oxidative stress can lead to neuronal damage and disrupt mitochondrial and endoplasmic reticulum functions after intracerebral hemorrhage (ICH). However, there is limited evidence elucidating their role in maintaining neuronal homeostasis. Metabolomics analysis, RNA sequencing, and CUT&Tag-seq were performed to investigate the mechanism underlying the interaction between the PERK/ATF4 branch of the endoplasmic reticulum stress (ERS) and mitochondrial one-carbon (1C) metabolism during neuronal resistance to oxidative stress. The association between mitochondrial 1C metabolism and the PERK/ATF4 branch of the ERS after ICH was investigated using transcription factor motif analysis and co-immunoprecipitation. The findings revealed interactions between the GRP78/PERK/ATF4 and mitochondrial 1C metabolism, which are important in preserving neuronal homeostasis after ICH. ATF4 is an upstream transcription factor that directly regulates the expression of 1C metabolism genes. Additionally, the GRP78/PERK/ATF4 forms a negative regulatory loop with MTHFD2 because of the interaction between GRP78 and MTHFD2. This study presents evidence of disrupted 1C metabolism and the occurrence of ERS in neurons post-ICH. Supplementing exogenous NADPH or interfering with the PERK/ATF4 could reduce symptoms related to neuronal injuries, suggesting new therapeutic prospects for ICH.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379068/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142157179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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 m-TORC1 inhibitor Sirolimus increases the effectiveness of Photodynamic therapy in the treatment of cutaneous Squamous Cell Carcinoma, impairing NRF2 antioxidant signaling. m-TORC1抑制剂西罗莫司可提高光动力疗法治疗皮肤鳞状细胞癌的疗效,同时损害NRF2抗氧化信号传导。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-06 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.94883
Jimena Nicolás-Morala, Marta Mascaraque-Checa, María Gallego-Rentero, Andrea Barahona, Edgar Abarca-Lachen, Elisa Carrasco, Yolanda Gilaberte, Salvador González, Ángeles Juarranz
{"title":"The m-TORC1 inhibitor Sirolimus increases the effectiveness of Photodynamic therapy in the treatment of cutaneous Squamous Cell Carcinoma, impairing NRF2 antioxidant signaling.","authors":"Jimena Nicolás-Morala, Marta Mascaraque-Checa, María Gallego-Rentero, Andrea Barahona, Edgar Abarca-Lachen, Elisa Carrasco, Yolanda Gilaberte, Salvador González, Ángeles Juarranz","doi":"10.7150/ijbs.94883","DOIUrl":"https://doi.org/10.7150/ijbs.94883","url":null,"abstract":"<p><p>Squamous Cell Carcinoma (SCC) is a subtype of Non-Melanoma Skin Cancer, the most common group of malignancies worldwide. Photodynamic therapy (PDT) is a non-invasive treatment approved for specific subtypes of SCC. Some malignancies resist PDT, forming more aggressive tumors and multiple relapses. Thus, new approaches aimed at optimizing the response to PDT are needed. The mTORC1 inhibitor rapamycin, also known as Sirolimus (SRL), interferes with protein synthesis and cell metabolism. The use of SRL as an immunosuppressant is associated to lower rates of SCC in kidney-transplanted patients, which are frequently affected by this pathology. We have evaluated SRL pre-treatment efficacy to enhance the damage induced by PDT with Methyl 5-aminolevulinate in two different cutaneous SCC established cell lines (SCC13 and A431) <i>in vitro</i> and therapy sensitization in PDT-resistant cell lines. We tested for the first time the SRL + PDT combination in a SKH-1 mouse model of photocarcinogenesis, diminishing the frequency of lesions and restraining tumor growth. Molecular studies revealed that protoporphyrin IX and reactive oxygen species production induced by PDT were promoted by SRL pre-treatment. Lastly, SRL modifies the expression and intracellular location of NRF2, interfering with the downstream antioxidant response modulated by NQO1 and HO-1. In conclusion, we propose SRL as a potential adjuvant to enhance PDT efficacy for SCC treatment.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379070/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IL-6 signaling accelerates iron overload by upregulating DMT1 in endothelial cells to promote aortic dissection. IL-6 信号通过上调内皮细胞中的 DMT1 加速铁超载,从而促进主动脉夹层。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-06 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.99511
Qiang Xie, Jianji Wang, Runqiao Li, Hao Liu, Yongliang Zhong, Qinfeng Xu, Yipeng Ge, Chengnan Li, Lizhong Sun, Junming Zhu
{"title":"IL-6 signaling accelerates iron overload by upregulating DMT1 in endothelial cells to promote aortic dissection.","authors":"Qiang Xie, Jianji Wang, Runqiao Li, Hao Liu, Yongliang Zhong, Qinfeng Xu, Yipeng Ge, Chengnan Li, Lizhong Sun, Junming Zhu","doi":"10.7150/ijbs.99511","DOIUrl":"https://doi.org/10.7150/ijbs.99511","url":null,"abstract":"<p><p>Aortic dissection (AD), caused by tearing of the intima and avulsion of the aortic media, is a severe threat to patient life and organ function. Iron is closely related to dissection formation and organ injury, but the mechanism of iron ion transport disorder in endothelial cells (ECs) remains unclear. We identified the characteristic EC of dissection with iron overload by single-cell RNA sequencing data. After intersecting iron homeostasis and differentially expressed genes, it was found that hypoxia-inducible factor-1α (HIF-1α) and divalent metal transporter 1 (DMT1) are key genes for iron ion disorder. Subsequently, IL-6R was identified as an essential reason for the JAK-STAT activation, a classical iron regulation pathway, through further intersection and validation. In <i>in vivo</i> and <i>in vitro</i>, both high IL-6 receptor expression and elevated IL-6 levels promote JAK1-STAT3 phosphorylation, leading to increased HIF-1α protein levels. Elevated HIF-1α binds explicitly to the 5'-UTR sequence of the DMT1 gene and transcriptionally promotes DMT1 expression, thereby increasing Fe<sup>2+</sup> accumulation and endoplasmic reticulum stress (ERS). Blocking IL-6R and free iron with deferoxamine and tocilizumab significantly prolonged survival and reduced aortic and organ damage in dissection mice. A comparison of perioperative data between AD patients and others revealed that high free iron, IL-6, and ERS levels are characteristics of AD patients and are correlated with prognosis. In conclusion, activated IL-6/JAK1/STAT3 signaling axis up-regulates DMT1 expression by increasing HIF-1α, thereby increasing intracellular Fe<sup>2+</sup> accumulation and tissue injury, which suggests a potential therapeutic target for AD.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379073/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153939","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
m6A modification enhances the stability of CDC25A promotes tumorigenicity of esophagogastric junction adenocarcinoma via cell cycle. m6A 修饰会增强 CDC25A 的稳定性,并通过细胞周期促进食管胃交界处腺癌的致瘤性。
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-06 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.98535
Yongbo Pan, Huolun Feng, Jianlong Zhou, Wenxing Zhang, Yongfeng Liu, Jiabin Zheng, Junjiang Wang, Shan Gao, Yong Li
{"title":"m<sup>6</sup>A modification enhances the stability of <i>CDC25A</i> promotes tumorigenicity of esophagogastric junction adenocarcinoma via cell cycle.","authors":"Yongbo Pan, Huolun Feng, Jianlong Zhou, Wenxing Zhang, Yongfeng Liu, Jiabin Zheng, Junjiang Wang, Shan Gao, Yong Li","doi":"10.7150/ijbs.98535","DOIUrl":"https://doi.org/10.7150/ijbs.98535","url":null,"abstract":"<p><p><i>N</i>6-Methyladenosine (m<sup>6</sup>A) modification and its regulators play critical roles in human cancers, but their functions and regulatory mechanisms in adenocarcinoma of the esophagogastric junction (AEG) remain unclear. Here, we identified that IGF2BP3 is the most significantly up-regulated m<sup>6</sup>A regulator in AEG tumors versus paired normal adjacent tissues from the expression profile of m<sup>6</sup>A regulators in a large cohort of AEG patients. Silencing IGF2BP3 inhibits AEG progression <i>in vitro</i> and <i>in vivo</i>. By profiling transcriptome-wide targets of IGF2BP3 and the m<sup>6</sup>A methylome in AEG, we found that IGF2BP3-mediated stabilization and enhanced expression of m<sup>6</sup>A-modified targets, including targets of the cell cycle pathway, such as <i>CDC25A</i>, <i>CDK4</i>, and <i>E2F1</i>, are critical for AEG progression. Mechanistically, the increased m<sup>6</sup>A modification of <i>CDC25A</i> accelerates the G1-S transition. Clinically, up-regulated IGF2BP3, METTL3, and CDC25A show a strong positive correlation in TCGA pan-cancer, including AEG. In conclusion, our study highlights the role of post-transcriptional regulation in modulating AEG tumor progression and elucidates the functional importance of the m<sup>6</sup>A/IGF2BP3/CDC25A axis in AEG cells.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379066/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153942","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PPARγ Agonist Pioglitazone Prevents Hypoxia-induced Cardiac Dysfunction by Reprogramming Glucose Metabolism. PPARγ激动剂吡格列酮通过重编程葡萄糖代谢防止缺氧诱导的心脏功能障碍
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-06 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.98387
Yijin Wang, Ru Zhang, Qian Chen, Zhangwen Lei, Caiyu Shi, Yifei Pang, Shan'an Zhang, Linjie He, Longtao Xu, Jinliang Xing, Haitao Guo
{"title":"PPARγ Agonist Pioglitazone Prevents Hypoxia-induced Cardiac Dysfunction by Reprogramming Glucose Metabolism.","authors":"Yijin Wang, Ru Zhang, Qian Chen, Zhangwen Lei, Caiyu Shi, Yifei Pang, Shan'an Zhang, Linjie He, Longtao Xu, Jinliang Xing, Haitao Guo","doi":"10.7150/ijbs.98387","DOIUrl":"https://doi.org/10.7150/ijbs.98387","url":null,"abstract":"<p><p>The heart relies on various defense mechanisms, including metabolic plasticity, to maintain its normal structure and function under high-altitude hypoxia. Pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ), sensitizes insulin, which in turn regulates blood glucose levels. However, its preventive effects against hypoxia-induced cardiac dysfunction at high altitudes have not been reported. In this study, pioglitazone effectively prevented cardiac dysfunction in hypoxic mice for 4 weeks, independent of its effects on insulin sensitivity. <i>In vitro</i> experiments demonstrated that pioglitazone enhanced the contractility of primary cardiomyocytes and reduced the risk of QT interval prolongation under hypoxic conditions. Additionally, pioglitazone promoted cardiac glucose metabolic reprogramming by increasing glycolytic capacity; enhancing glucose oxidation, electron transfer, and oxidative phosphorylation processes; and reducing mitochondrial reactive ROS production, which ultimately maintained mitochondrial membrane potential and ATP production in cardiomyocytes under hypoxic conditions. Notably, as a PPARγ agonist, pioglitazone promoted hypoxia-inducible factor 1α (HIF-1α) expression in hypoxic myocardium. Moreover, KC7F2, a HIF-1α inhibitor, disrupted the reprogramming of cardiac glucose metabolism and reduced cardiac function in pioglitazone-treated mice under hypoxic conditions. In conclusion, pioglitazone effectively prevented high-altitude hypoxia-induced cardiac dysfunction by reprogramming cardiac glucose metabolism.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379067/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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 PI3K/Akt-Nrf2 Signaling Pathway and Mitophagy Synergistically Mediate Hydroxytyrosol to Alleviate Intestinal Oxidative Damage. PI3K/Akt-Nrf2信号通路和丝裂噬协同介导羟基酪醇减轻肠道氧化损伤
IF 8.2 2区 生物学
International Journal of Biological Sciences Pub Date : 2024-08-06 eCollection Date: 2024-01-01 DOI: 10.7150/ijbs.97263
Xiaobin Wen, Shanlong Tang, Fan Wan, Ruqing Zhong, Liang Chen, Hongfu Zhang
{"title":"The PI3K/Akt-Nrf2 Signaling Pathway and Mitophagy Synergistically Mediate Hydroxytyrosol to Alleviate Intestinal Oxidative Damage.","authors":"Xiaobin Wen, Shanlong Tang, Fan Wan, Ruqing Zhong, Liang Chen, Hongfu Zhang","doi":"10.7150/ijbs.97263","DOIUrl":"https://doi.org/10.7150/ijbs.97263","url":null,"abstract":"<p><p>Oxidative stress is a major pathogenic factor in many intestinal diseases, such as inflammatory bowel disease (IBD) and colorectal cancer (CRC). The Nrf2 signaling pathway and mitophagy can reduce reactive oxygen species (ROS) and alleviate oxidative stress, but their relationship is unclear. Hydroxytyrosol (HT), a polyphenolic compound abundant in olive oil, has strong antioxidant activity and may help treat these diseases. We used pigs as a model to investigate HT's effect on intestinal oxidative damage and its mechanisms. Diquat (DQ) induced oxidative stress and impaired intestinal barrier function, which HT mitigated. Mechanistic studies in IPEC-J2 cells showed that HT protected against oxidative damage by activating the PI3K/Akt-Nrf2 signaling pathway and promoting mitophagy. Our study highlighted the synergistic relationship between Nrf2 and mitophagy in mediating HT's antioxidant effects. Inhibition studies confirmed that disrupting either pathway compromised HT's protective effects. Maintaining redox balance through Nrf2 and mitophagy is important for eliminating excess ROS. Nrf2 increases antioxidant enzymes to clear existing ROS, while mitophagy removes damaged mitochondria and reduces ROS generation. This study demonstrates that these pathways collaboratively modulate the antioxidant effects of HT, with neither being dispensable. Targeting Nrf2 and mitophagy could be a promising strategy for treating oxidative stress-related intestinal diseases, with HT as a potential treatment.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11379072/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153956","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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