Translational Oncology最新文献

筛选
英文 中文
Cell-free nucleic acid fragmentomics: A non-invasive window into cellular epigenomes 无细胞核酸片段组学:了解细胞表观基因组的非侵入性窗口
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-22 DOI: 10.1016/j.tranon.2024.102085
{"title":"Cell-free nucleic acid fragmentomics: A non-invasive window into cellular epigenomes","authors":"","doi":"10.1016/j.tranon.2024.102085","DOIUrl":"10.1016/j.tranon.2024.102085","url":null,"abstract":"<div><p>Clinical genomic profiling of cell-free nucleic acids (e.g. cell-free DNA or cfDNA) from blood and other body fluids has ushered in a new era in non-invasive diagnostics and treatment monitoring strategies for health conditions and diseases such as cancer. Genomic analysis of cfDNAs not only identifies disease-associated mutations, but emerging findings suggest that structural, topological, and fragmentation characteristics of cfDNAs reveal crucial information about the location of source tissues, their epigenomes, and other clinically relevant characteristics, leading to the burgeoning field of fragmentomics. The field has seen rapid developments in computational and genomics methodologies for conducting large-scale studies on health conditions and diseases – that have led to fundamental, mechanistic discoveries as well as translational applications. Several recent studies have shown the clinical utilities of the cfDNA fragmentomics technique which has the potential to be effective for early disease diagnosis, determining treatment outcomes, and risk-free continuous patient monitoring in a non-invasive manner. In this article, we outline recent developments in computational genomic methodologies and analysis strategies, as well as the emerging insights from cfNA fragmentomics. We conclude by highlighting the current challenges and opportunities.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002122/pdfft?md5=95e4c1fe568908ffdbd9b3e80da04a42&pid=1-s2.0-S1936523324002122-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142040325","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
Chitinase 3-like-1 (CHI3L1) in the pathogenesis of epidermal growth factor receptor mutant non-small cell lung cancer 几丁质酶 3-like-1 (CHI3L1) 在表皮生长因子受体突变型非小细胞肺癌发病机制中的作用
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-22 DOI: 10.1016/j.tranon.2024.102108
{"title":"Chitinase 3-like-1 (CHI3L1) in the pathogenesis of epidermal growth factor receptor mutant non-small cell lung cancer","authors":"","doi":"10.1016/j.tranon.2024.102108","DOIUrl":"10.1016/j.tranon.2024.102108","url":null,"abstract":"<div><p>Non-small cell lung cancer (NSCLC) accounts for 85 % of all lung cancers. In NSCLC, 10–20 % of Caucasian patients and 30–50 % of Asian patients have tumors with activating mutations in the Epidermal Growth Factor Receptor (<em>EGFR</em>). A high percentage of these patients exhibit favorable responses to treatment with tyrosine kinase inhibitors (TKI). Unfortunately, a majority of these patients develop therapeutic resistance with progression free survival lasting 9–18 months. The mechanisms that underlie the tumorigenic effects of <em>EGFR</em> and the ability of NSCLC to develop resistance to TKI therapies, however, are poorly understood. Here we demonstrate that CHI3L1 is produced by EGFR activation of normal epithelial cells, transformed epithelial cells with wild type <em>EGFR</em> and cells with cancer-associated, activating <em>EGFR</em> mutations. We also demonstrate that CHI3L1 auto-induces itself and feeds back to stimulate EGFR and its ligands via a STAT3-dependent mechanism(s). Highly specific antibodies against CHI3L1 (anti-CHI3L1/FRG) and TKI, individually and in combination, abrogated the effects of EGFR activation on CHI3L1 and the ability of CHI3L1 to stimulate the EGFR axis. Anti-CHI3L1 also interacted with osimertinib to reverse TKI therapeutic resistance and induce tumor cell death and inhibit pulmonary metastasis while stimulating tumor suppressor genes including <em>KEAP1</em>. CHI3L1 is a downstream target of EGFR that feeds back to stimulate and activate the EGFR axis. Anti-CHI3L1 is an exciting potential therapeutic for <em>EGFR</em> mutant NSCLC, alone and in combination with osimertinib or other TKIs.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002353/pdfft?md5=d5be8da9a21b66cf7af4e49dd467b900&pid=1-s2.0-S1936523324002353-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142044715","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
Multifaceted role of GCN2 in tumor adaptation and therapeutic targeting GCN2 在肿瘤适应和靶向治疗中的多方面作用
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-22 DOI: 10.1016/j.tranon.2024.102096
{"title":"Multifaceted role of GCN2 in tumor adaptation and therapeutic targeting","authors":"","doi":"10.1016/j.tranon.2024.102096","DOIUrl":"10.1016/j.tranon.2024.102096","url":null,"abstract":"<div><p>Tumor cells voraciously consume nutrients from their environment to facilitate rapid proliferation, necessitating effective strategies to manage nutrient scarcity during tumor growth and progression. A pivotal regulatory mechanism in this context is the Integrated Stress Response (ISR), which ensures cellular homeostasis under conditions such as endoplasmic reticulum stress, the unfolded protein response, and nutrient deprivation. Within the ISR framework, the kinase GCN2 is critical, orchestrating a myriad of cellular processes including the inhibition of protein synthesis, the enhancement of amino acid transport, autophagy initiation, and angiogenesis. These processes collectively enable tumor survival and adaptation under nutrient-limited conditions. Furthermore, GCN2-mediated pathways may induce apoptosis, a property exploited by specific therapeutic agents. Leveraging extensive datasets from TCGA, GEO, and GTEx projects, we conducted a pan-cancer analysis to investigate the prognostic significance of GCN2 expression across diverse cancer types. Our analysis indicates that GCN2 expression significantly varies and correlates with both adverse and favorable prognoses depending on the type of cancer, illustrating its complex role in tumorigenesis. Importantly, GCN2 also modulates the tumor immune microenvironment, influencing immune checkpoint expression and the functionality of immune cells, thereby affecting immunotherapy outcomes. This study highlights the potential of targeting GCN2 with specific inhibitors, as evidenced by their efficacy in preclinical models to augment treatment responses and combat resistance in oncology. These findings advocate for a deeper exploration of GCN2′s multifaceted roles, which could pave the way for novel targeted therapies in cancer treatment, aiming to improve clinical outcomes.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002237/pdfft?md5=2572faeffc39b56bc5979d76706dd5a0&pid=1-s2.0-S1936523324002237-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142044714","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
GTF2H5 Identified as a crucial synthetic lethal target to counteract chemoresistance in colorectal cancer GTF2H5 被确定为对抗结直肠癌化疗耐药性的关键合成致死靶点
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-21 DOI: 10.1016/j.tranon.2024.102097
{"title":"GTF2H5 Identified as a crucial synthetic lethal target to counteract chemoresistance in colorectal cancer","authors":"","doi":"10.1016/j.tranon.2024.102097","DOIUrl":"10.1016/j.tranon.2024.102097","url":null,"abstract":"<div><h3>Background</h3><p>Synthetic lethality (SL) emerges as a novel concept being explored to combat cancer progression and resistance to conventional therapy. Despite the efficacy of chemotherapy in select cases of colorectal cancer (CRC), a substantial proportion of patients encounter challenges, leading to an adverse prognosis of CRC patients. CRC-related SL genes offer a potential avenue for identifying therapeutic targets.</p></div><div><h3>Methods</h3><p>CRC-related SL genes were obtained from the SynLethDB database. The bulk RNA sequencing data, mutation data, and clinical information for treated and untreated CRC patients were enrolled from the UCSC and GEO databases. The Tumor Immunology Single Cell Center database served as the repository for collecting and analyzing single-cell RNA sequencing data. The synergistic killing effect of SL genes and chemotherapeutic drugs on resistant cells was experimentally verified.</p></div><div><h3>Results</h3><p>In the present study, pivotal SL genes associated with chemoresistance identified by using WGCNA and CRC patients categorized into two groups based on these genes. Variations between the groups were most pronounced in pathways associated with extracellular matrix remodeling. Further by integrating mutation data, five potential SL genes were discerned, which were highly expressed in the presence of TP53 or KRAS mutations, leading to a severely poor prognosis. Subsequent time series analysis revealed that the expression of GTF2H5 was gradually elevated at different stages of the transition from sensitive to resistant in CRC cells. Finally, it was preliminarily verified by experiments that GTF2H5 may play a key role in driving the drug-resistant transition within CRC cells.</p></div><div><h3>Conclusions</h3><p>The identification of SL genes that collaboratively interact with chemotherapeutic agents could provide new insights into solving the issue of chemotherapy resistance in CRC patients. And GTF2H5 wields a fundamental influence in inducing chemoresistance in CRC, which provided a potential therapeutic target for CRC.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002249/pdfft?md5=237ce71eb6d2bcb8d019aea27a8c26ea&pid=1-s2.0-S1936523324002249-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142021137","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
SUGT1 regulates the progression of ovarian cancer through the AKT/PI3K/mTOR signaling pathway SUGT1 通过 AKT/PI3K/mTOR 信号通路调控卵巢癌的进展
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-20 DOI: 10.1016/j.tranon.2024.102088
{"title":"SUGT1 regulates the progression of ovarian cancer through the AKT/PI3K/mTOR signaling pathway","authors":"","doi":"10.1016/j.tranon.2024.102088","DOIUrl":"10.1016/j.tranon.2024.102088","url":null,"abstract":"<div><p>This study investigates the expression and functional roles of SUGT1 in ovarian cancer, utilizing data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) projects. Our analyses reveal that SUGT1 is significantly upregulated in ovarian cancer tissues compared to normal controls. We further explore the prognostic value of SUGT1, where elevated expression correlates with poorer patient outcomes, particularly in ovarian cancer. The functional implications of SUGT1 in cancer biology were assessed through in vitro and in vivo experiments. Gene Set Enrichment Analysis (GSEA) indicates a significant association between high SUGT1 expression and the activation of glycolytic pathways, suggesting a potential role in metabolic reprogramming. Inhibition of SUGT1 via siRNA in ovarian cancer cell lines results in decreased proliferation and increased apoptosis, along with reduced migration and invasion capabilities. Additionally, our study identifies the transcription factor ELF1 as a significant regulator of SUGT1 expression. Through promoter analysis and chromatin immunoprecipitation, we demonstrate that ELF1 directly binds to the SUGT1 promoter, enhancing its transcription. This regulatory mechanism underscores the importance of transcriptional control in cancer metabolism, providing insights into potential therapeutic targets. Our findings establish SUGT1 as a crucial player in the oncogenic processes of ovarian cancer, influencing both metabolic pathways and transcriptional regulation. This highlights its potential as a biomarker and therapeutic target in managing ovarian cancer.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002158/pdfft?md5=70bd937397fc1cb6c0648c0a80883489&pid=1-s2.0-S1936523324002158-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142012033","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
IGFBP1 promotes the proliferation and migration of lung adenocarcinoma cells through the PPARα pathway IGFBP1 通过 PPARα 途径促进肺腺癌细胞的增殖和迁移
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-20 DOI: 10.1016/j.tranon.2024.102095
{"title":"IGFBP1 promotes the proliferation and migration of lung adenocarcinoma cells through the PPARα pathway","authors":"","doi":"10.1016/j.tranon.2024.102095","DOIUrl":"10.1016/j.tranon.2024.102095","url":null,"abstract":"<div><h3>Background</h3><p>The immune status is closely linked to cancer progression, metastasis, and prognosis. Lipid metabolism, crucial for reshaping immune status, plays a key role in regulating the advancement of lung adenocarcinoma (LUAD) and deserves further investigation.</p></div><div><h3>Methods</h3><p>This study classifies LUAD patients into different immune subtypes based on lipid metabolism-related genes and compares the clinical characteristics among these subtypes. Single-multi COX analysis screens out key genes related to prognosis, and a risk feature and prognostic model are constructed. Cell cloning, scratch, transwell, western blotting and flow cytometry cell cycle analysis to detect the function of key genes. A subcutaneous tumor animal model is used to investigate the in vivo function and molecular mechanisms of key genes.</p></div><div><h3>Results</h3><p>LUAD patients are classified into three immune subtypes, among which C3 subtype has lower immune status and higher frequency of gene mutations, and show lower immunoreactivity in immunotherapy. COX analysis identified a prognostic model for four lipid metabolism factors (IGFBP1, NR0B2, PPARA, and POMC). IGFBP1, a core gene in this model, is highly expressed in the C3 subtype. Functionally, knocking down IGFBP1 significantly inhibits tumor cell cloning, scratch, and migration abilities, and downregulates the expression of cell cycle and EMT-related proteins. Knocking down IGFBP1 significantly inhibits tumor burden (<em>P</em> &lt; 0.05). Mechanistically, knocking down IGFBP1 inhibits the activation of PPARα to regulate tumor cell growth.</p></div><div><h3>Conclusions</h3><p>This study found that lipid metabolism genes are closely related to LUAD, and IGFBP1 may be a key gene in regulating tumor growth and development.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002225/pdfft?md5=21aee5bf589efd3f0954a52c78daa173&pid=1-s2.0-S1936523324002225-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142012035","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
Niraparib plays synergistic antitumor effects with NRT in a mouse ovarian cancer model with HRP 尼拉帕利与 NRT 在小鼠卵巢癌 HRP 模型中发挥协同抗肿瘤作用
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-19 DOI: 10.1016/j.tranon.2024.102094
{"title":"Niraparib plays synergistic antitumor effects with NRT in a mouse ovarian cancer model with HRP","authors":"","doi":"10.1016/j.tranon.2024.102094","DOIUrl":"10.1016/j.tranon.2024.102094","url":null,"abstract":"<div><h3>Objective</h3><p>PARPi offers less clinical benefit for HRP patients compared to HRD patients. PARPi has an immunomodulatory function. NRT therapy targets tumor neoantigens without off-target immune toxicity. We explored the synergy between Niraparib and NRT in enhancing antitumor activity in an HRP ovarian cancer mouse model.</p></div><div><h3>Methods</h3><p>In the C57BL/6 mouse ID8 ovarian cancer model, the effect of Niraparib on reshaping TIME was evaluated by immune cell infiltration analysis of transcriptomic data. The antitumor effects of Niraparib, NRT, and their combined use were systematically evaluated. To corroborate alterations in TILs, TAMs, and chemokine profiles within the TIME, we employed immunofluorescence imaging and transcriptome sequencing analysis.</p></div><div><h3>Results</h3><p>Niraparib increased the M1-TAMs and activated CD8+ T cells in tumor tissues of C57BL/6 mice with ID8 ovarian cancer. GSEA showed that gene set associated with immature DC and INFα, cytokines and chemokines were significantly enriched in immune feature, KEGG and GO gene sets, meanwhile CCL5, CXCL9 and CXCL10 play dominant roles together. In the animal trials, combined group had a tumor growth delay compared with Niraparib group (<em>P</em> &lt; 0.01) and control group (<em>P</em> &lt; 0.001), and longer survival compared with the single agent group (<em>P</em>&lt;0.01) .</p></div><div><h3>Conclusions</h3><p>Niraparib could exert immune-reshaping effects, then acts synergistic antitumor effects with NRT in HRP ovarian cancer model. Our findings provide new ideas and rationale for combined immunotherapy in HRP ovarian cancer.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002213/pdfft?md5=c354a6db85573a59d3400ba8746245d0&pid=1-s2.0-S1936523324002213-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142006909","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
A mini-review-cancer energy reprogramming on drug resistance and immune response 微型综述--癌症能量重编程对耐药性和免疫反应的影响
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-19 DOI: 10.1016/j.tranon.2024.102099
{"title":"A mini-review-cancer energy reprogramming on drug resistance and immune response","authors":"","doi":"10.1016/j.tranon.2024.102099","DOIUrl":"10.1016/j.tranon.2024.102099","url":null,"abstract":"<div><p>With the growing interest to harness cancer metabolism and energy reprogramming, this mini review aimed to explain the metabolic programming revealing the mechanisms regarding the treatment resistance. This mini review summarized the prominent cancer metabolic reprogramming on macromolecules. In addition, metabolic reprogramming explaining immune response and treatment resistance as well as energy reprogramming mechanisms are briefly discussed. Finally, some prospects in MR for reversing cancer drug resistance are highlighted.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002262/pdfft?md5=54251ac5cee84255953d80cc80c4ee04&pid=1-s2.0-S1936523324002262-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142006908","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
Unlocking mitochondrial dysfunction-associated senescence (MiDAS) with NAD+ – A Boolean model of mitochondrial dynamics and cell cycle control 用 NAD+ 解锁线粒体功能障碍相关衰老 (MiDAS) - 线粒体动力学和细胞周期控制的布尔模型
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-19 DOI: 10.1016/j.tranon.2024.102084
{"title":"Unlocking mitochondrial dysfunction-associated senescence (MiDAS) with NAD+ – A Boolean model of mitochondrial dynamics and cell cycle control","authors":"","doi":"10.1016/j.tranon.2024.102084","DOIUrl":"10.1016/j.tranon.2024.102084","url":null,"abstract":"<div><p>The steady accumulation of senescent cells with aging creates tissue environments that aid cancer evolution. Aging cell states are highly heterogeneous. 'Deep senescent' cells rely on healthy mitochondria to fuel a strong proinflammatory secretome, including cytokines, growth and transforming signals. Yet, the physiological triggers of senescence such as reactive oxygen species (ROS) can also trigger mitochondrial dysfunction, and sufficient energy deficit to alter their secretome and cause chronic oxidative stress – a state termed Mitochondrial Dysfunction-Associated Senescence (MiDAS). Here, we offer a mechanistic hypothesis for the molecular processes leading to MiDAS, along with testable predictions. To do this we have built a Boolean regulatory network model that qualitatively captures key aspects of mitochondrial dynamics during cell cycle progression (hyper-fusion at the G1/S boundary, fission in mitosis), apoptosis (fission and dysfunction) and glucose starvation (reversible hyper-fusion), as well as MiDAS in response to <em>SIRT3</em> knockdown or oxidative stress. Our model reaffirms the protective role of NAD<sup>+</sup> and external pyruvate. We offer testable predictions about the growth factor- and glucose-dependence of MiDAS and its reversibility at different stages of reactive oxygen species (ROS)-induced senescence. Our model provides mechanistic insights into the distinct stages of DNA-damage induced senescence, the relationship between senescence and epithelial-to-mesenchymal transition in cancer and offers a foundation for building multiscale models of tissue aging.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002110/pdfft?md5=d565099af9e04f30f0523ebdf544d8ad&pid=1-s2.0-S1936523324002110-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142006907","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
Oxazine drug-seed induces paraptosis and apoptosis through reactive oxygen species/JNK pathway in human breast cancer cells 恶嗪药籽通过活性氧/JNK 通路诱导人类乳腺癌细胞发生副凋亡和细胞凋亡
IF 5 2区 医学
Translational Oncology Pub Date : 2024-08-18 DOI: 10.1016/j.tranon.2024.102101
{"title":"Oxazine drug-seed induces paraptosis and apoptosis through reactive oxygen species/JNK pathway in human breast cancer cells","authors":"","doi":"10.1016/j.tranon.2024.102101","DOIUrl":"10.1016/j.tranon.2024.102101","url":null,"abstract":"<div><p>Small molecule-driven JNK activation has been found to induce apoptosis and paraptosis in cancer cells. Herein pharmacological effects of synthetic oxazine (4<em>aS</em>, 7<em>aS</em>)-3-((4-(4‑chloro-2-fluorophenyl)piperazin-1-yl)methyl)-4-phenyl-4, 4<em>a</em>, 5, 6, 7, 7<em>a</em>-hexahydrocyclopenta[<em>e</em>] [1,2]oxazine (FPPO; BSO-07) on JNK-driven apoptosis and paraptosis has been demonstrated in human breast cancer (BC) MDA-MB231 and MCF-7 cells respectively. BSO-07 imparted significant cytotoxicity in BC cells, induced activation of JNK, and increased intracellular reactive oxygen species (ROS) levels. It also enhanced the expression of apoptosis-associated proteins like PARP, Bax, and phosphorylated p53, while decreasing the levels of Bcl-2, Bcl-xL, and Survivin. Furthermore, the drug altered the expression of proteins linked to paraptosis, such as ATF4 and CHOP. Treatment with N-acetyl-cysteine (antioxidant) or SP600125 (JNK inhibitor) partly reversed the effects of BSO-07 on apoptosis and paraptosis. Advanced <em>in silico</em> bioinformatics, cheminformatics, density Fourier transform and molecular electrostatic potential analysis further demonstrated that BSO-07 induced apoptosis and paraptosis <em>via</em> the ROS/JNK pathway in human BC cells.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002286/pdfft?md5=b575e682403aa934402019a386ccf958&pid=1-s2.0-S1936523324002286-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142001969","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