首页 > 最新文献

Molecular Cancer Research最新文献

英文 中文
Tumor Cholesterol Synthesis, Statin Use, and Lethal Prostate Cancer. 肿瘤胆固醇合成,他汀类药物的使用和致死性前列腺癌。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-12-02 DOI: 10.1158/1541-7786.MCR-24-0864
Sinead Flanagan, Rosina T Lis, Ying Huang, Lina Jehane, Jane B Vaselkiv, Emma H Allott, Stephen P Finn, Tamara L Lotan, Michelangelo Fiorentino, Massimo Loda, Gwo-Shu Mary Lee, Goutam Chakraborty, Philip W Kantoff, Lorelei A Mucci, Konrad H Stopsack

Prostate tumor cells produce cholesterol de novo, and statin therapy targets the initial rate-limiting enzyme in this process, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR). The extent to which the expression of HMGCR in prostate tumors could influence progression and predict the potential antineoplastic effects of statins remains unclear. In a prospective cohort study of 1,098 men diagnosed with primary prostate cancer from 1982 to 2009 in the Health Professionals Follow-up Study and Physicians' Health Study, 16% of prostate tumors showed strong HMGCR staining intensity, and 31% showed no staining. HMGCR expression was higher in tumors with PTEN loss but did not differ by statin use or long-term dietary cholesterol or saturated fat intake. Participants were followed for lethal events (distant metastases or prostate cancer-related death) over up to 32 years, and 96 lethal events occurred in those without metastases at diagnosis. Strong HMGCR expression was associated with higher rates of lethal prostate cancer (HR, 2.2; 95% confidence interval, 1.3-3.7), adjusting for age at diagnosis and Gleason score but without a linear dose response. In vitro, in the LNCaP human prostate cancer cell line, atorvastatin affected tumor cell viability in cells with experimentally lowered HMGCR expression. This study corroborates that high cholesterol synthesis in prostate tumor cells is associated with PTEN loss, aggressive tumor characteristics, and a greater risk of lethality.

Implications: High expression of HMGCR, the first rate-limiting enzyme of cholesterol synthesis, is a feature of prostate tumors that are more likely to progress to metastatic disease or death from prostate cancer.

前列腺肿瘤细胞重新产生胆固醇,而他汀类药物治疗针对这一过程中的初始限速酶,3-羟基-3-甲基-戊二酰辅酶A还原酶(HMGCR)。前列腺肿瘤中HMGCR的表达在多大程度上影响肿瘤进展并预测他汀类药物潜在的抗肿瘤作用尚不清楚。在一项来自卫生专业人员随访研究和医生健康研究的1982-2009年诊断为原发性前列腺癌的1098名男性的前瞻性队列研究中,16%的前列腺肿瘤显示强烈的HMGCR染色强度,31%没有染色。HMGCR在PTEN缺失的肿瘤中表达较高,但与他汀类药物使用或长期膳食胆固醇或饱和脂肪摄入无关。研究人员对参与者进行了长达32年的致命事件(远处转移或前列腺癌相关死亡)随访,在诊断时没有转移的患者中发生了96起致命事件。高HMGCR表达与较高的致死性前列腺癌发生率相关(风险比2.2,95%可信区间1.3-3.7),校正了诊断年龄和Gleason评分,但没有线性剂量反应。在LNCaP人前列腺癌细胞系中,阿托伐他汀通过实验降低HMGCR表达影响肿瘤细胞活力。本研究证实,前列腺肿瘤细胞中的高胆固醇合成与PTEN缺失、侵袭性肿瘤特征和更大的致死风险相关。结论:高表达的HMGCR(第一限速胆固醇合成酶)是前列腺肿瘤更容易发展为转移性疾病或死于前列腺癌的一个特征。
{"title":"Tumor Cholesterol Synthesis, Statin Use, and Lethal Prostate Cancer.","authors":"Sinead Flanagan, Rosina T Lis, Ying Huang, Lina Jehane, Jane B Vaselkiv, Emma H Allott, Stephen P Finn, Tamara L Lotan, Michelangelo Fiorentino, Massimo Loda, Gwo-Shu Mary Lee, Goutam Chakraborty, Philip W Kantoff, Lorelei A Mucci, Konrad H Stopsack","doi":"10.1158/1541-7786.MCR-24-0864","DOIUrl":"10.1158/1541-7786.MCR-24-0864","url":null,"abstract":"<p><p>Prostate tumor cells produce cholesterol de novo, and statin therapy targets the initial rate-limiting enzyme in this process, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR). The extent to which the expression of HMGCR in prostate tumors could influence progression and predict the potential antineoplastic effects of statins remains unclear. In a prospective cohort study of 1,098 men diagnosed with primary prostate cancer from 1982 to 2009 in the Health Professionals Follow-up Study and Physicians' Health Study, 16% of prostate tumors showed strong HMGCR staining intensity, and 31% showed no staining. HMGCR expression was higher in tumors with PTEN loss but did not differ by statin use or long-term dietary cholesterol or saturated fat intake. Participants were followed for lethal events (distant metastases or prostate cancer-related death) over up to 32 years, and 96 lethal events occurred in those without metastases at diagnosis. Strong HMGCR expression was associated with higher rates of lethal prostate cancer (HR, 2.2; 95% confidence interval, 1.3-3.7), adjusting for age at diagnosis and Gleason score but without a linear dose response. In vitro, in the LNCaP human prostate cancer cell line, atorvastatin affected tumor cell viability in cells with experimentally lowered HMGCR expression. This study corroborates that high cholesterol synthesis in prostate tumor cells is associated with PTEN loss, aggressive tumor characteristics, and a greater risk of lethality.</p><p><strong>Implications: </strong>High expression of HMGCR, the first rate-limiting enzyme of cholesterol synthesis, is a feature of prostate tumors that are more likely to progress to metastatic disease or death from prostate cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"1025-1033"},"PeriodicalIF":4.7,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12434729/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144784869","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
MTHFD2-mediated ubiquitination and degradation of FOXO1 promote tongue squamous cell carcinoma progression. mthfd2介导的泛素化和fox01降解促进舌鳞状细胞癌的进展。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-19 DOI: 10.1158/1541-7786.MCR-25-0724
Aikebaier Tuohuti, Lucheng Fang, Zehua Lin, Sa Wu, Weisong Cai, Xiaoping Ming, Zhe Chen, Qiang Xie, Qiuyang Fu, Xiaoqi Sun, Xiong Chen

Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), a pivotal enzyme in one-carbon metabolism, is significantly overexpressed in tongue squamous cell carcinoma (TSCC) and correlates with poor patient prognosis. Functional studies demonstrate that MTHFD2 drives TSCC proliferation and migration in vitro and in vivo, while its inhibition suppresses tumor progression. Mechanistically, MTHFD2 orchestrates a dual post-translational modification cascade: through its enzymatic activity, it simultaneously induces FOXO1 hypermethylation and ubiquitination, ultimately triggering ubiquitin-proteasome degradation of this tumor suppressor. This methylation-primed degradation axis is clinically validated by the inverse MTHFD2-high/FOXO1-low expression pattern in TSCC specimens, which predicts adverse outcomes. Critically, pharmacological inhibition of MTHFD2 (e.g., DS18561882) blocks FOXO1 degradation, establishing the MTHFD2-FOXO1 axis as a promising therapeutic target for TSCC through its novel metabolic-epigenetic regulatory mechanism. Implications: This study identifies the MTHFD2-FOXO1 axis as a druggable metabolic-epigenetic pathway in TSCC, providing both a prognostic marker and a therapeutic target.

亚甲基四氢叶酸脱氢酶2 (MTHFD2)是一种单碳代谢的关键酶,在舌鳞癌(TSCC)中显著过表达,并与患者预后不良相关。功能研究表明,MTHFD2在体外和体内均可促进TSCC的增殖和迁移,而其抑制作用可抑制肿瘤进展。从机制上讲,MTHFD2协调了一个双重翻译后修饰级联:通过其酶活性,它同时诱导FOXO1超甲基化和泛素化,最终触发该肿瘤抑制因子的泛素蛋白酶体降解。这种甲基化引发的降解轴在TSCC标本中得到了mthfd2高/ fox01低表达模式的临床验证,这预示着不良后果。重要的是,药物抑制MTHFD2(如DS18561882)可阻断FOXO1的降解,通过其新的代谢-表观遗传调控机制,使MTHFD2-FOXO1轴成为TSCC的一个有希望的治疗靶点。意义:本研究确定MTHFD2-FOXO1轴是TSCC中可药物代谢-表观遗传通路,提供了预后标志物和治疗靶点。
{"title":"MTHFD2-mediated ubiquitination and degradation of FOXO1 promote tongue squamous cell carcinoma progression.","authors":"Aikebaier Tuohuti, Lucheng Fang, Zehua Lin, Sa Wu, Weisong Cai, Xiaoping Ming, Zhe Chen, Qiang Xie, Qiuyang Fu, Xiaoqi Sun, Xiong Chen","doi":"10.1158/1541-7786.MCR-25-0724","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-25-0724","url":null,"abstract":"<p><p>Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), a pivotal enzyme in one-carbon metabolism, is significantly overexpressed in tongue squamous cell carcinoma (TSCC) and correlates with poor patient prognosis. Functional studies demonstrate that MTHFD2 drives TSCC proliferation and migration in vitro and in vivo, while its inhibition suppresses tumor progression. Mechanistically, MTHFD2 orchestrates a dual post-translational modification cascade: through its enzymatic activity, it simultaneously induces FOXO1 hypermethylation and ubiquitination, ultimately triggering ubiquitin-proteasome degradation of this tumor suppressor. This methylation-primed degradation axis is clinically validated by the inverse MTHFD2-high/FOXO1-low expression pattern in TSCC specimens, which predicts adverse outcomes. Critically, pharmacological inhibition of MTHFD2 (e.g., DS18561882) blocks FOXO1 degradation, establishing the MTHFD2-FOXO1 axis as a promising therapeutic target for TSCC through its novel metabolic-epigenetic regulatory mechanism. Implications: This study identifies the MTHFD2-FOXO1 axis as a druggable metabolic-epigenetic pathway in TSCC, providing both a prognostic marker and a therapeutic target.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145550202","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
BCOR mutations deregulate cell cycle and hypoxic adaptation pathways in retinoblastoma. BCOR突变解除了视网膜母细胞瘤细胞周期和缺氧适应途径的调控。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-05 DOI: 10.1158/1541-7786.MCR-24-1078
Michelle G Zhang, Jeffim N Kuznetsoff, Natalie C Cetta, Sebastian Salazar, Renata L Volonterio, Stefan Kurtenbach, Zelia M Correa, Daniel Pelaez, J William Harbour

Retinoblastoma (RB) is the most common pediatric eye cancer. Most cases of RB are initiated by bi-allelic mutational inactivation of the RB1 gene, yet most RB tumors harbor additional genomic aberrations that may promote tumor progression. After RB1, the gene that is most commonly mutated gene in RB is BCOR, which is mutated in approximately 20% of RB tumors and is associated with a more aggressive tumor phenotype and worse patient outcomes. Despite its importance, little is known about the role of BCOR in RB. Here, we interrogated BCOR in low passage RB cell lines using mass spectrometry, chromatin immunoprecipitation sequencing, and RNA sequencing. We show that the BCOR protein interacts with members of the ncPRC1.1 Polycomb Repressive Complex and localizes at gene loci with traditionally activating and repressing chromatin markers. Loss of BCOR downregulates the expression of genes associated with cell cycle regulation and upregulates genes associated with hypoxic adaptation. We conclude that BCOR mutations slow cell proliferation and drive hypoxic adaptation in RB via epigenetic mechanisms that may be amenable to targeted therapy. Implications: This study reveals that BCOR may play a noncanonical, multi-faceted role in retinoblastoma with implications in cell cycle, differentiation, and hypoxic adaptation, ultimately shedding insight into its molecular framework for future therapeutic strategies.

视网膜母细胞瘤(RB)是最常见的儿童眼癌。大多数RB病例是由RB1基因的双等位基因突变失活引发的,然而大多数RB肿瘤含有可能促进肿瘤进展的额外基因组畸变。在RB1之后,RB中最常见的突变基因是BCOR,在大约20%的RB肿瘤中发生突变,并与更具侵袭性的肿瘤表型和更差的患者预后相关。尽管它很重要,但人们对BCOR在RB中的作用知之甚少。在这里,我们使用质谱、染色质免疫沉淀测序和RNA测序对低传代RB细胞系的BCOR进行了检测。我们发现BCOR蛋白与ncPRC1.1多梳抑制复合体的成员相互作用,并定位于传统上激活和抑制染色质标记的基因位点。BCOR缺失会下调与细胞周期调控相关的基因的表达,上调与缺氧适应相关的基因的表达。我们得出结论,BCOR突变通过表观遗传机制减缓RB细胞增殖并驱动缺氧适应,这可能适合靶向治疗。本研究揭示了BCOR可能在视网膜母细胞瘤中发挥非规范的、多方面的作用,包括细胞周期、分化和缺氧适应,最终揭示了其分子框架,为未来的治疗策略提供了新的思路。
{"title":"BCOR mutations deregulate cell cycle and hypoxic adaptation pathways in retinoblastoma.","authors":"Michelle G Zhang, Jeffim N Kuznetsoff, Natalie C Cetta, Sebastian Salazar, Renata L Volonterio, Stefan Kurtenbach, Zelia M Correa, Daniel Pelaez, J William Harbour","doi":"10.1158/1541-7786.MCR-24-1078","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-24-1078","url":null,"abstract":"<p><p>Retinoblastoma (RB) is the most common pediatric eye cancer. Most cases of RB are initiated by bi-allelic mutational inactivation of the RB1 gene, yet most RB tumors harbor additional genomic aberrations that may promote tumor progression. After RB1, the gene that is most commonly mutated gene in RB is BCOR, which is mutated in approximately 20% of RB tumors and is associated with a more aggressive tumor phenotype and worse patient outcomes. Despite its importance, little is known about the role of BCOR in RB. Here, we interrogated BCOR in low passage RB cell lines using mass spectrometry, chromatin immunoprecipitation sequencing, and RNA sequencing. We show that the BCOR protein interacts with members of the ncPRC1.1 Polycomb Repressive Complex and localizes at gene loci with traditionally activating and repressing chromatin markers. Loss of BCOR downregulates the expression of genes associated with cell cycle regulation and upregulates genes associated with hypoxic adaptation. We conclude that BCOR mutations slow cell proliferation and drive hypoxic adaptation in RB via epigenetic mechanisms that may be amenable to targeted therapy. Implications: This study reveals that BCOR may play a noncanonical, multi-faceted role in retinoblastoma with implications in cell cycle, differentiation, and hypoxic adaptation, ultimately shedding insight into its molecular framework for future therapeutic strategies.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145452448","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Synergistic DBNDD1-GDF15 signaling activates the NF-κB pathway to promote colorectal cancer progression. 协同DBNDD1-GDF15信号激活NF-κB通路促进结直肠癌进展。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-03 DOI: 10.1158/1541-7786.MCR-25-0153
Xiaomin Qi, Caijuan Bai, Li Dong, Anqi Wang, Chaojun Wei, Yonghong Li, Mengyuan Zhao, Chongge You

Colorectal cancer (CRC) is a highly lethal gastrointestinal tract malignancy whose pathogenesis and molecular drivers are not fully understood. This study focused on searching for genes that are differentially expressed in cancer versus normal mucosa, with the goal of identifying molecular patterns of expression that are mechanistically linked to CRC pathogenesis. We analyzed 585 CRC samples and 329 normal samples from the Gene Expression Omnibus (GEO) database, creating a weighted gene co-expression network (WGCNA) across 24,069 genes. Through this approach, five modules associated with CRC were identified, which were enriched in MAPK signaling and cholesterol metabolism pathways. Using LASSO regression, we selected 13 hub genes (ABCB5, AOC1, ARHGAP44, CACNG3, DBNDD1, GAS7, GTF2IRD1, PRSS22, SCN4A, TTC22, DLX6, PDK4, and SLC13A2) from these modules. Survival analysis indicated that higher expression of DBNDD1 correlated with worse overall survival in CRC patients. Machine learning validation confirmed the stability of these genetic markers. Experimental validation demonstrated increased levels of DBNDD1 and GDF15 in CRC, promoting constant NF-κB activation via DBNDD1-dependent GDF15 induction. Knocking down DBNDD1 inhibited cell proliferation, migration, and invasion in vitro (DLD1/HCT116 cells), alongside decreased GDF15 expression and reduced p-NF-κB p65/p-IκB signaling. Additionally, DBNDD1 knockdown resulted in reduced tumor growth in vivo, highlighting that DBNDD1-GDF15-NF-κB signaling pathway drives CRC pathogenesis. Implications: This study highlights the crucial role of the DBNDD1/GDF15/NF-κB signaling pathway in CRC development, positioning DBNDD1 as a promising target for precision medicine strategies aimed at enhancing patient outcomes.

结直肠癌(CRC)是一种高致死率的胃肠道恶性肿瘤,其发病机制和分子驱动因素尚不完全清楚。本研究的重点是寻找癌症与正常粘膜中差异表达的基因,目的是确定与结直肠癌发病机制相关的分子表达模式。我们分析了基因表达Omnibus (Gene Expression Omnibus, GEO)数据库中的585例结直肠癌样本和329例正常样本,创建了一个包含24069个基因的加权基因共表达网络(WGCNA)。通过这种方法,我们发现了5个与CRC相关的模块,它们在MAPK信号通路和胆固醇代谢途径中富集。利用LASSO回归,我们从这些模块中选择了13个枢纽基因(ABCB5、AOC1、ARHGAP44、CACNG3、DBNDD1、GAS7、GTF2IRD1、PRSS22、SCN4A、TTC22、DLX6、PDK4和SLC13A2)。生存分析表明,在结直肠癌患者中,DBNDD1的高表达与较差的总生存相关。机器学习验证证实了这些遗传标记的稳定性。实验验证表明,结直肠癌中DBNDD1和GDF15水平升高,通过DBNDD1依赖性GDF15诱导促进NF-κB的持续活化。敲低DBNDD1可抑制体外细胞(DLD1/HCT116细胞)的增殖、迁移和侵袭,同时降低GDF15表达和p-NF-κB p65/p- i -κB信号。此外,DBNDD1敲低导致体内肿瘤生长减少,强调DBNDD1- gdf15 - nf -κB信号通路驱动CRC发病。本研究强调了DBNDD1/GDF15/NF-κB信号通路在结直肠癌发展中的关键作用,将DBNDD1定位为旨在提高患者预后的精准医疗策略的有希望的靶点。
{"title":"Synergistic DBNDD1-GDF15 signaling activates the NF-κB pathway to promote colorectal cancer progression.","authors":"Xiaomin Qi, Caijuan Bai, Li Dong, Anqi Wang, Chaojun Wei, Yonghong Li, Mengyuan Zhao, Chongge You","doi":"10.1158/1541-7786.MCR-25-0153","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-25-0153","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a highly lethal gastrointestinal tract malignancy whose pathogenesis and molecular drivers are not fully understood. This study focused on searching for genes that are differentially expressed in cancer versus normal mucosa, with the goal of identifying molecular patterns of expression that are mechanistically linked to CRC pathogenesis. We analyzed 585 CRC samples and 329 normal samples from the Gene Expression Omnibus (GEO) database, creating a weighted gene co-expression network (WGCNA) across 24,069 genes. Through this approach, five modules associated with CRC were identified, which were enriched in MAPK signaling and cholesterol metabolism pathways. Using LASSO regression, we selected 13 hub genes (ABCB5, AOC1, ARHGAP44, CACNG3, DBNDD1, GAS7, GTF2IRD1, PRSS22, SCN4A, TTC22, DLX6, PDK4, and SLC13A2) from these modules. Survival analysis indicated that higher expression of DBNDD1 correlated with worse overall survival in CRC patients. Machine learning validation confirmed the stability of these genetic markers. Experimental validation demonstrated increased levels of DBNDD1 and GDF15 in CRC, promoting constant NF-κB activation via DBNDD1-dependent GDF15 induction. Knocking down DBNDD1 inhibited cell proliferation, migration, and invasion in vitro (DLD1/HCT116 cells), alongside decreased GDF15 expression and reduced p-NF-κB p65/p-IκB signaling. Additionally, DBNDD1 knockdown resulted in reduced tumor growth in vivo, highlighting that DBNDD1-GDF15-NF-κB signaling pathway drives CRC pathogenesis. Implications: This study highlights the crucial role of the DBNDD1/GDF15/NF-κB signaling pathway in CRC development, positioning DBNDD1 as a promising target for precision medicine strategies aimed at enhancing patient outcomes.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145438425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
β-Catenin-Cohesin Ring-CEGRs/ALCDs Axis Activation Contributes to the Development of Hepatoblastoma and Fibrolamellar HCC. β-连环蛋白-内聚素环- cegrs / alcd轴的激活有助于肝母细胞瘤和纤维层性肝细胞癌的发展。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-03 DOI: 10.1158/1541-7786.MCR-25-0236
Yasmeen Fleifil, Ruhi Gulati, Katherine Jennings, Ina Kycia, Alex Bondoc, Gregory Tiao, James Geller, Matthew Weirauch, Khashayar Vakili, Lubov Timchenko, Nikolai Timchenko

The pediatric and adolescent liver cancers, hepatoblastoma (HBL) and fibrolamellar hepatocellular carcinoma (FLC), respectively, are dangerous diseases requiring aggressive surgery, when feasible, and nontargeted toxic chemotherapy for a chance of cure due to insufficient knowledge of underlying molecular mechanisms. We have previously reported the essential role of ph-S675-β-catenin in the reorganization of genomic structure in HBL and FLC by oncogenic activation via chromosomal regions called cancer-enhancing genomic regions or aggressive liver cancer domains (CEGR/ALCD). In FLC, the fusion DNAJB1-PKAc (J-PKAc) oncoprotein phosphorylates β-catenin at Ser675, triggering such CEGRs/ALCDs-mediated activation of oncogenes. In this study, we found that all members of the cohesin ring-CTCF, Rad21, SMC1, SMC3, and STAG1-and β-catenin-TCF4 are bound to CEGRs/ALCDs of oncogenes in HBL and FLC, as well as many other cancers, and that this binding increases transcription. Examination of a large cohort of HBL and FLC samples revealed that cohesin ring expression is dramatically elevated in the majority. The cohesin ring, as well as the ph-S675-β-catenin-TCF4-p300 complex, is detected on both the promoter and intron-located CEGRs/ALCDs of NRF2 and Thy1, correlating with increased transcription. This suggests that the cohesin ring creates the DNA loop for oncogene activation. The inhibition of the cohesin ring by JQ1 reduces the proliferation of HBL and FLC cells in culture, as well as cells expressing the FLC-specific J-PKAc fusion oncogene.

Implications: These studies provide evidence that J-PKAc-β-catenin and the cohesin ring cooperate in oncogenic activation for both HBL and FLC.

儿童和青少年肝癌肝母细胞瘤(HBL)和纤维层状肝细胞癌(FLC)是危险的疾病,由于对潜在的分子机制了解不足,需要在可行的情况下进行积极的手术和非靶向毒性化疗以获得治愈的机会。我们之前报道了ph-S675-β-catenin在HBL和FLC基因组结构重组中的重要作用,通过称为癌症增强基因组区域或侵袭性肝癌结构域(cegr / alcd)的染色体区域进行致癌激活。在FLC中,融合的j - ppkac癌蛋白磷酸化β-catenin的Ser675位点,触发这种CEGRs/ alcd介导的癌基因激活。在本文中,我们发现黏结环的所有成员- CTCF, Rad21, SMC1, SMC3和STAG1 -和β-catenin-TCF4在HBL和FLC以及许多其他癌症中与癌基因的cegr / alcd结合,并且这种结合增加了转录。对大量HBL和FLC样本的检查显示,大多数黏结蛋白环的表达显著升高。内聚环以及ph-S675-β-catenin-TCF4-p300复合物在NRF2和Thy1的启动子和内含子位置的cegr / alcd上都被检测到,这与转录增加有关。这表明内聚环为致癌基因激活创造了dna环。JQ1对内聚环的抑制抑制了培养的HBL和FLC细胞以及表达FLC特异性DNAJB1-PKAc融合癌基因的细胞的增殖。意义:这些研究提供了j - ppkac -β-catenin和内聚蛋白环共同参与HBL和FLC的致癌激活的证据。
{"title":"β-Catenin-Cohesin Ring-CEGRs/ALCDs Axis Activation Contributes to the Development of Hepatoblastoma and Fibrolamellar HCC.","authors":"Yasmeen Fleifil, Ruhi Gulati, Katherine Jennings, Ina Kycia, Alex Bondoc, Gregory Tiao, James Geller, Matthew Weirauch, Khashayar Vakili, Lubov Timchenko, Nikolai Timchenko","doi":"10.1158/1541-7786.MCR-25-0236","DOIUrl":"10.1158/1541-7786.MCR-25-0236","url":null,"abstract":"<p><p>The pediatric and adolescent liver cancers, hepatoblastoma (HBL) and fibrolamellar hepatocellular carcinoma (FLC), respectively, are dangerous diseases requiring aggressive surgery, when feasible, and nontargeted toxic chemotherapy for a chance of cure due to insufficient knowledge of underlying molecular mechanisms. We have previously reported the essential role of ph-S675-β-catenin in the reorganization of genomic structure in HBL and FLC by oncogenic activation via chromosomal regions called cancer-enhancing genomic regions or aggressive liver cancer domains (CEGR/ALCD). In FLC, the fusion DNAJB1-PKAc (J-PKAc) oncoprotein phosphorylates β-catenin at Ser675, triggering such CEGRs/ALCDs-mediated activation of oncogenes. In this study, we found that all members of the cohesin ring-CTCF, Rad21, SMC1, SMC3, and STAG1-and β-catenin-TCF4 are bound to CEGRs/ALCDs of oncogenes in HBL and FLC, as well as many other cancers, and that this binding increases transcription. Examination of a large cohort of HBL and FLC samples revealed that cohesin ring expression is dramatically elevated in the majority. The cohesin ring, as well as the ph-S675-β-catenin-TCF4-p300 complex, is detected on both the promoter and intron-located CEGRs/ALCDs of NRF2 and Thy1, correlating with increased transcription. This suggests that the cohesin ring creates the DNA loop for oncogene activation. The inhibition of the cohesin ring by JQ1 reduces the proliferation of HBL and FLC cells in culture, as well as cells expressing the FLC-specific J-PKAc fusion oncogene.</p><p><strong>Implications: </strong>These studies provide evidence that J-PKAc-β-catenin and the cohesin ring cooperate in oncogenic activation for both HBL and FLC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"901-912"},"PeriodicalIF":4.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144691066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Genomic Signatures of Poor Prognosis in Merkel Cell Carcinoma: A Single-Institution Prospective Study. 默克尔细胞癌预后不良的基因组特征:一项单机构前瞻性研究。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-03 DOI: 10.1158/1541-7786.MCR-24-1138
Joshua D Smith, Apurva D Bhangale, Wenjin Gu, Collin Brummel, Elizabeth Gensterblum-Miller, Scott McLean, Paul W Harms, Kelly L Harms, Christopher K Bichakjian, Matthew E Spector, Ryan E Mills, J Chad Brenner

Merkel cell carcinoma (MCC) is an aggressive disease with poor survival outcomes and increasing incidence. There is a clear and present need for enhanced understanding of cellular mechanisms of tumorigenesis, validation of robust genetic signatures predictive of aggressive disease, and novel informatics tools to simplify analysis of Merkel cell polyomavirus (MCPyV)-host genome interactions. Genomic DNA was harvested from 54 MCC tumors for exome sequencing and in-depth genetic profiling of a 226-gene panel. We further developed a robust informatics package (MCPyViewer) optimized for MCPyV integration site analysis with graphical output to simplify usability for end users. Finally, we assessed the prognostic impact of specific genetic signatures on MCC-specific survival in our cohort. Our study included 54 patients (n = 44 MCPyV positive), 11 (20.4%) of whom had died of MCC at last follow-up. Human genes altered at high frequency included LRP1B (n = 10, 18.5%), FAT1 (n = 9, 16.7%), KMT2D (n = 9, 16.7%), and RB1 (n = 7, 13.0%). In 36 of 44 (81.8%) MCPyV-positive tumors, we identified viral integration into the human genome with a median of two events per tumor. In six tumors, MCPyV integrated into Catalogue of Somatic Mutations in Cancer tier 1 or tier 2 cancer-related human genes.

Implications: A combined genomics score incorporating tumor mutational burden and copy-number variation was strongly prognostic of MCC-specific survival controlling for lymph node metastases and tumor MCPyV status; thus, our study adds critical understanding to prognostic markers and tumorigenic mechanisms in MCC.

默克尔细胞癌(MCC)是一种侵袭性疾病,生存率差,发病率增加。目前显然需要加强对肿瘤发生的细胞机制的理解,验证预测侵袭性疾病的强大遗传特征,以及新的信息学工具来简化默克尔细胞多瘤病毒(MCPyV)-宿主基因组相互作用的分析。从54个MCC肿瘤中收集基因组DNA,用于外显子组测序和226个基因面板的深入遗传分析。我们进一步开发了一个强大的信息包(MCPyViewer),优化了MCPyV集成站点分析和图形输出,以简化最终用户的可用性。最后,我们在我们的队列中评估了特定遗传特征对mcc特异性生存的预后影响。本研究纳入54例mcpyv阳性患者(n = 44),其中11例(20.4%)在最后随访时死于MCC。高频改变的人类基因包括LRP1B (n = 10, 18.5%)、FAT1 (n = 9, 16.7%)、KMT2D (n = 9, 16.7%)和RB1 (n = 7, 13.0%)。在44个mcpyv阳性肿瘤中的36个(81.8%)中,我们发现病毒整合到人类基因组中,每个肿瘤中位数为两个事件。在6种肿瘤中,MCPyV整合到COSMIC 1级或2级癌症相关人类基因中。意义:结合肿瘤突变负担和拷贝数变异的基因组学综合评分对控制淋巴结转移和肿瘤MCPyV状态的MCC特异性生存具有很强的预后作用,因此,我们的研究为MCC的预后标志物和致瘤机制提供了重要的理解。
{"title":"Genomic Signatures of Poor Prognosis in Merkel Cell Carcinoma: A Single-Institution Prospective Study.","authors":"Joshua D Smith, Apurva D Bhangale, Wenjin Gu, Collin Brummel, Elizabeth Gensterblum-Miller, Scott McLean, Paul W Harms, Kelly L Harms, Christopher K Bichakjian, Matthew E Spector, Ryan E Mills, J Chad Brenner","doi":"10.1158/1541-7786.MCR-24-1138","DOIUrl":"10.1158/1541-7786.MCR-24-1138","url":null,"abstract":"<p><p>Merkel cell carcinoma (MCC) is an aggressive disease with poor survival outcomes and increasing incidence. There is a clear and present need for enhanced understanding of cellular mechanisms of tumorigenesis, validation of robust genetic signatures predictive of aggressive disease, and novel informatics tools to simplify analysis of Merkel cell polyomavirus (MCPyV)-host genome interactions. Genomic DNA was harvested from 54 MCC tumors for exome sequencing and in-depth genetic profiling of a 226-gene panel. We further developed a robust informatics package (MCPyViewer) optimized for MCPyV integration site analysis with graphical output to simplify usability for end users. Finally, we assessed the prognostic impact of specific genetic signatures on MCC-specific survival in our cohort. Our study included 54 patients (n = 44 MCPyV positive), 11 (20.4%) of whom had died of MCC at last follow-up. Human genes altered at high frequency included LRP1B (n = 10, 18.5%), FAT1 (n = 9, 16.7%), KMT2D (n = 9, 16.7%), and RB1 (n = 7, 13.0%). In 36 of 44 (81.8%) MCPyV-positive tumors, we identified viral integration into the human genome with a median of two events per tumor. In six tumors, MCPyV integrated into Catalogue of Somatic Mutations in Cancer tier 1 or tier 2 cancer-related human genes.</p><p><strong>Implications: </strong>A combined genomics score incorporating tumor mutational burden and copy-number variation was strongly prognostic of MCC-specific survival controlling for lymph node metastases and tumor MCPyV status; thus, our study adds critical understanding to prognostic markers and tumorigenic mechanisms in MCC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"923-935"},"PeriodicalIF":4.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12336863/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144675321","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
Differential Control of Growth and Identity by HNF4α Isoforms in Pancreatic Ductal Adenocarcinoma. 胰腺导管腺癌中HNF4α亚型对生长和特性的差异控制。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-03 DOI: 10.1158/1541-7786.MCR-25-0175
Pengshu Fang, Emily R Wilson, Sydney N Larsen, Walter A Orellana, Margaret A Hall, Chris Stubben, Acramul Haque Kabir, Kajsa Affolter, Richard A Moffitt, Xiaoyang Zhang, Eric L Snyder

Although transcriptomic studies have stratified pancreatic ductal adenocarcinoma (PDAC) into clinically relevant subtypes, classic or basal-like, further research is needed to identify the transcriptional regulators of each subtype. Previous studies identified HNF4α as a key regulator of the classic subtype. Still, the distinct contributions of its isoforms (P1 and P2), which display dichotomous functions in normal development and gastrointestinal malignancies, remain unexplored. In this study, we show that HNF4α-positive human PDAC tumors exhibit uniform expression of P2 isoforms but variable expression of P1 isoforms. To dissect the roles of each isoform in PDAC, we performed functional, transcriptomic, and epigenetic analyses after exogenous expression in HNF4α-negative models or CRISPRi-mediated knockdown of endogenous isoforms. We demonstrated that P1 isoforms are less compatible with growth and stronger transcriptional regulators than P2. Despite both isoforms sharing a common DNA-binding domain, P1 isoforms displayed stronger binding at HNF4α target genes, resulting in increased transcriptional activity. These findings provide a detailed characterization of HNF4α P1 and P2 isoforms and their distinct roles in PDAC biology.

Implications: HNF4α isoforms exhibit heterogeneous expression in PDAC and have distinct effects on proliferation and gene expression, including markers of clinically relevant molecular subtypes.

尽管转录组学研究已经将胰腺导管腺癌(pancreatic ductal adencarcinoma, PDAC)分为临床相关亚型,经典亚型或基底样亚型,但需要进一步的研究来确定每种亚型的转录调节因子。先前的研究发现HNF4α是经典亚型的关键调节因子。尽管如此,其同种异构体(P1和P2)在正常发育和胃肠道恶性肿瘤中表现出两种功能,其独特的贡献仍未被探索。在这里,我们发现hnf4 α阳性的人类PDAC肿瘤表现出p2亚型的统一表达,而P1亚型的表达则不同。为了剖析每个异构体在PDAC中的作用,我们在hnf4 α-阴性模型中外源表达或crispr介导的内源异构体敲除后进行了功能、转录组学和表观遗传学分析。我们证明P1同工型与生长的相容性较差,并且比P2具有更强的转录调节因子。尽管这两种亚型共享一个共同的dna结合域,但P1亚型在HNF4α靶基因上表现出更强的结合,导致转录活性增加。这些发现提供了HNF4α P1和P2亚型的详细特征及其在PDAC生物学中的独特作用。含义:HNF4α亚型在PDAC中表现出异质表达,对增殖和基因表达有明显影响,包括临床相关分子亚型的标记。
{"title":"Differential Control of Growth and Identity by HNF4α Isoforms in Pancreatic Ductal Adenocarcinoma.","authors":"Pengshu Fang, Emily R Wilson, Sydney N Larsen, Walter A Orellana, Margaret A Hall, Chris Stubben, Acramul Haque Kabir, Kajsa Affolter, Richard A Moffitt, Xiaoyang Zhang, Eric L Snyder","doi":"10.1158/1541-7786.MCR-25-0175","DOIUrl":"10.1158/1541-7786.MCR-25-0175","url":null,"abstract":"<p><p>Although transcriptomic studies have stratified pancreatic ductal adenocarcinoma (PDAC) into clinically relevant subtypes, classic or basal-like, further research is needed to identify the transcriptional regulators of each subtype. Previous studies identified HNF4α as a key regulator of the classic subtype. Still, the distinct contributions of its isoforms (P1 and P2), which display dichotomous functions in normal development and gastrointestinal malignancies, remain unexplored. In this study, we show that HNF4α-positive human PDAC tumors exhibit uniform expression of P2 isoforms but variable expression of P1 isoforms. To dissect the roles of each isoform in PDAC, we performed functional, transcriptomic, and epigenetic analyses after exogenous expression in HNF4α-negative models or CRISPRi-mediated knockdown of endogenous isoforms. We demonstrated that P1 isoforms are less compatible with growth and stronger transcriptional regulators than P2. Despite both isoforms sharing a common DNA-binding domain, P1 isoforms displayed stronger binding at HNF4α target genes, resulting in increased transcriptional activity. These findings provide a detailed characterization of HNF4α P1 and P2 isoforms and their distinct roles in PDAC biology.</p><p><strong>Implications: </strong>HNF4α isoforms exhibit heterogeneous expression in PDAC and have distinct effects on proliferation and gene expression, including markers of clinically relevant molecular subtypes.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"936-952"},"PeriodicalIF":4.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12333154/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144643034","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 LINC00519/hsa-miR-22-3p/MECOM Axis Accelerates Intrahepatic Cholangiocarcinoma Progression through PI3K/AKT Signaling. LINC00519/hsa-miR-22-3p/MECOM轴通过PI3K/AKT信号通路加速肝内胆管癌进展
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-03 DOI: 10.1158/1541-7786.MCR-25-0207
Zhuxin Gu, Yanjun Sun, Fajing Chen, Weiwei Gu, Xiaohua Lu, Suming Zhao, Qinan Geng, Yang Yang

Intrahepatic cholangiocarcinoma (ICC) is the second most common liver cancer. LINC00519 plays a prominent role in the progression of numerous cancers. To explore the molecular mechanism of LINC00519 in ICC, the expressions of LINC00519, hsa-miR-22-3p, and MECOM in ICC were assessed using the ENCORI database and qRT-PCR. The biological functions of LINC00519 in ICC were examined using a clone formation experiment, Transwell analysis, flow cytometry, and Western blot. Meanwhile, the mechanism of LINC00519 in ICC was determined by a dual-luciferase reporter assay. Results showed that LINC00519 and MECOM were highly expressed in ICC, whereas hsa-miR-22-3p was decreased. Functionally, silencing LINC00519 weakened ICC cell proliferation and migration and induced cell apoptosis. Also, LINC00519 knockdown repressed the PI3K/AKT (protein kinase B) pathway. Mechanistically, LINC00519 acted as a competitive endogenous RNA to target MECOM by sponging hsa-miR-22-3p. Meanwhile, rescue assays further proved that low LINC00519 expression restrained ICC cell proliferation and migration and accelerated apoptosis through the PI3K/AKT pathway by miR-22-3p/MECOM. In conclusion, this research revealed a novel LINC00519/hsa-miR-22-3p/MECOM regulatory axis and PI3K/AKT pathway that modulated ICC progression.

Implications: This study deepens the understanding of the noncoding RNA regulatory network in ICC and provides potential targets for the diagnosis and targeted therapy of ICC.

肝内胆管癌(ICC)是第二常见的肝癌。LINC00519在许多癌症的进展中起着重要作用。为探讨LINC00519在ICC中的分子机制,采用ENCORI数据库和实时荧光定量PCR检测LINC00519、hsa-miR-22-3p和MECOM在ICC中的表达。采用克隆形成实验、Transwell分析、流式细胞术和Western blot检测LINC00519在ICC中的生物学功能。同时,采用双荧光素酶报告基因法确定了LINC00519在ICC中的作用机制。结果显示,LINC00519和MECOM在ICC中高表达,而hsa-miR-22-3p表达降低。在功能上,沉默LINC00519可减弱ICC细胞的增殖、迁移和诱导细胞凋亡。此外,LINC00519敲低抑制PI3K/AKT通路。机制上,LINC00519作为竞争性内源性RNA,通过海绵吸附hsa-miR-22-3p靶向MECOM。同时,救援实验进一步证明,LINC00519低表达抑制ICC细胞增殖和迁移,并通过miR-22-3p/MECOM通过PI3K/AKT通路加速凋亡。总之,本研究揭示了一种新的LINC00519/hsa-miR-22-3p/MECOM调控轴和PI3K/AKT通路调节ICC进展。意义:本研究加深了对ICC非编码RNA调控网络的认识,为ICC的诊断和靶向治疗提供了潜在靶点。
{"title":"The LINC00519/hsa-miR-22-3p/MECOM Axis Accelerates Intrahepatic Cholangiocarcinoma Progression through PI3K/AKT Signaling.","authors":"Zhuxin Gu, Yanjun Sun, Fajing Chen, Weiwei Gu, Xiaohua Lu, Suming Zhao, Qinan Geng, Yang Yang","doi":"10.1158/1541-7786.MCR-25-0207","DOIUrl":"10.1158/1541-7786.MCR-25-0207","url":null,"abstract":"<p><p>Intrahepatic cholangiocarcinoma (ICC) is the second most common liver cancer. LINC00519 plays a prominent role in the progression of numerous cancers. To explore the molecular mechanism of LINC00519 in ICC, the expressions of LINC00519, hsa-miR-22-3p, and MECOM in ICC were assessed using the ENCORI database and qRT-PCR. The biological functions of LINC00519 in ICC were examined using a clone formation experiment, Transwell analysis, flow cytometry, and Western blot. Meanwhile, the mechanism of LINC00519 in ICC was determined by a dual-luciferase reporter assay. Results showed that LINC00519 and MECOM were highly expressed in ICC, whereas hsa-miR-22-3p was decreased. Functionally, silencing LINC00519 weakened ICC cell proliferation and migration and induced cell apoptosis. Also, LINC00519 knockdown repressed the PI3K/AKT (protein kinase B) pathway. Mechanistically, LINC00519 acted as a competitive endogenous RNA to target MECOM by sponging hsa-miR-22-3p. Meanwhile, rescue assays further proved that low LINC00519 expression restrained ICC cell proliferation and migration and accelerated apoptosis through the PI3K/AKT pathway by miR-22-3p/MECOM. In conclusion, this research revealed a novel LINC00519/hsa-miR-22-3p/MECOM regulatory axis and PI3K/AKT pathway that modulated ICC progression.</p><p><strong>Implications: </strong>This study deepens the understanding of the noncoding RNA regulatory network in ICC and provides potential targets for the diagnosis and targeted therapy of ICC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"913-922"},"PeriodicalIF":4.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580760/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144760600","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
MACC1-AS1: An Emerging Star in the Field of Tumor Biology with Bright Futures in Medicine. MACC1-AS1:肿瘤生物学领域的一颗新星,医学前景光明。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-11-03 DOI: 10.1158/1541-7786.MCR-25-0170
Li Dong, Weidong Li, Wei Zhu, Yamin Sun

Long noncoding RNAs act as modulators, with significant influence on a wide array of biological functions. They form an extensive communication network between genes and contribute to the pathophysiology of various human diseases, especially cancer. A growing body of research has demonstrated that long noncoding RNAs, acting either as promoters or inhibitors of oncogenesis, are intricately linked to the initiation and progression of cancer. Metastasis-associated colon cancer 1 antisense RNA1 (MACC1-AS1) is a newly identified long noncoding RNA that is abnormally expressed in various types of human tumors. Poor clinical characteristics, such as larger tumor size, advanced tumor stage, lymph node metastasis, and a lower overall survival rate, are linked to the overexpression of MACC1-AS1. MACC1-AS1 exerts a complex regulatory function: It acts as a competitive RNA, interacts with multiple proteins, and influences diverse pathways, leading to tumor development. It is essential to note the decreased efficacy of conventional chemotherapy drugs, which diminishes the efficacy of cancer treatment. Ongoing research has been highlighting the multifaceted functions of MACC1-AS1, and thus, it is required to unravel its exact molecular mechanisms. In this overarching review, we explore the significance of MACC1-AS1 as a potential cancer treatment target and biomarker. This study can potentially play an important role in the advancement of the field and confirm its potential clinical applicability.

长链非编码rna作为调制剂,对多种生物功能具有重要影响。它们在基因之间形成了广泛的通讯网络,并对各种人类疾病,特别是癌症的病理生理有贡献。越来越多的研究表明,lncrna作为肿瘤发生的启动子或抑制剂,与癌症的发生和发展有着复杂的联系。MACC1-AS1是一种新发现的LncRNA,在各种类型的人类肿瘤中异常表达。肿瘤较大、肿瘤分期较晚、淋巴结转移、总生存率较低等不良临床特征与MACC1-AS1过表达有关。MACC1-AS1发挥复杂的调控功能:它作为竞争性RNA,与多种蛋白质相互作用,影响多种途径,导致肿瘤的发展。必须注意的是,传统化疗药物的疗效下降,这降低了癌症治疗的疗效。正在进行的研究已经强调了MACC1-AS1的多方面功能,因此,需要揭示其确切的分子机制。在这篇综述中,我们探讨了MACC1-AS1作为潜在的癌症治疗靶点和生物标志物的意义。本研究可能在该领域的发展中发挥重要作用,并证实其潜在的临床适用性。
{"title":"MACC1-AS1: An Emerging Star in the Field of Tumor Biology with Bright Futures in Medicine.","authors":"Li Dong, Weidong Li, Wei Zhu, Yamin Sun","doi":"10.1158/1541-7786.MCR-25-0170","DOIUrl":"10.1158/1541-7786.MCR-25-0170","url":null,"abstract":"<p><p>Long noncoding RNAs act as modulators, with significant influence on a wide array of biological functions. They form an extensive communication network between genes and contribute to the pathophysiology of various human diseases, especially cancer. A growing body of research has demonstrated that long noncoding RNAs, acting either as promoters or inhibitors of oncogenesis, are intricately linked to the initiation and progression of cancer. Metastasis-associated colon cancer 1 antisense RNA1 (MACC1-AS1) is a newly identified long noncoding RNA that is abnormally expressed in various types of human tumors. Poor clinical characteristics, such as larger tumor size, advanced tumor stage, lymph node metastasis, and a lower overall survival rate, are linked to the overexpression of MACC1-AS1. MACC1-AS1 exerts a complex regulatory function: It acts as a competitive RNA, interacts with multiple proteins, and influences diverse pathways, leading to tumor development. It is essential to note the decreased efficacy of conventional chemotherapy drugs, which diminishes the efficacy of cancer treatment. Ongoing research has been highlighting the multifaceted functions of MACC1-AS1, and thus, it is required to unravel its exact molecular mechanisms. In this overarching review, we explore the significance of MACC1-AS1 as a potential cancer treatment target and biomarker. This study can potentially play an important role in the advancement of the field and confirm its potential clinical applicability.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"895-900"},"PeriodicalIF":4.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144962317","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting LAMP2A enhances SELENBP1 expression and suppresses malignant behaviors in HNSCC. 靶向LAMP2A可增强SELENBP1的表达,抑制HNSCC的恶性行为。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-30 DOI: 10.1158/1541-7786.MCR-25-0388
Hongwei Cao, Dongsheng Xing, Hanbing Yu, Feifei Jiang, Li Qi, Kunrong Wang, Yan Wang, Aihui Yan

Lysosome-Associated Membrane Protein Type 2A (LAMP2A) serves as the critical rate-limiting component of chaperone-mediated autophagy (CMA), governing substrate translocation into lysosomes. Accumulating studies indicate that LAMP2A downregulation leads to CMA impairment in multiple cancer malignancies. In this study, we found that LAMP2A is significantly upregulated in head and neck squamous cell carcinoma (HNSCC) compared to normal tissues. Cell functional studies performed on FaDu and CAL-27 cells showed that downregulation of LAMP2A inhibited cell proliferation and stemness, and induced cell apoptosis. Since CMA specifically targets proteins containing a pentapeptide motif (KFERQ-like motif) in a LAMP2A-dependent manner, we further employed an integrated proteomic-interactomic approach combined with KFERQ motif analysis. This comprehensive strategy identified Selenium Binding Protein 1 (SELENBP1) as a novel putative CMA substrate in HNSCC. Subsequent validation confirmed that knockdown of CMA receptor LAMP2A significantly increased SELENBP1 protein levels both in vitro and in vivo. CO-IP assays confirmed that SELENBP1 interacts with the CMA chaperone protein heat shock cognate 71 kDa protein (HSPA8) in a KFERQ motif ("EKVIQ")-dependent manner. Overexpression of SELENBP1 attenuated HNSCC cell proliferation and viability. Most importantly, silencing of SELENBP1 partially rescued the tumor-suppressive phenotypes induced by LAMP2A knockdown, suggesting that SELENBP1 mediated the effects of LAMP2A knockdown on HNSCC. This study provides insights into the role of the LAMP2A-CMA-SELENBP1 axis in the development of novel therapies for HNSCC. Implications: This study provides a novel insight into the role of CMA during the pathogenesis of HNSCC.

溶酶体相关膜蛋白2A型(LAMP2A)是伴侣介导的自噬(CMA)的关键限速成分,控制底物转运到溶酶体。越来越多的研究表明,在多种恶性肿瘤中,LAMP2A下调可导致CMA损伤。在本研究中,我们发现与正常组织相比,头颈部鳞状细胞癌(HNSCC)中LAMP2A的表达明显上调。对FaDu和CAL-27细胞进行的细胞功能研究表明,下调LAMP2A可抑制细胞增殖和干性,诱导细胞凋亡。由于CMA以lamp2a依赖的方式特异性靶向含有五肽基序(KFERQ样基序)的蛋白质,因此我们进一步采用了结合KFERQ基序分析的综合蛋白质组学-相互作用组学方法。该综合策略确定硒结合蛋白1 (SELENBP1)是HNSCC中一种新的假定的CMA底物。随后的验证证实,CMA受体LAMP2A的敲除显著增加了硒bp1蛋白在体外和体内的水平。CO-IP实验证实,SELENBP1以KFERQ motif(“EKVIQ”)依赖的方式与CMA伴侣蛋白热休克同源蛋白71kda蛋白(HSPA8)相互作用。过表达SELENBP1可减弱HNSCC细胞的增殖和活力。最重要的是,SELENBP1的沉默部分挽救了LAMP2A敲低诱导的肿瘤抑制表型,这表明SELENBP1介导了LAMP2A敲低对HNSCC的影响。这项研究提供了对LAMP2A-CMA-SELENBP1轴在HNSCC新疗法开发中的作用的见解。意义:本研究为CMA在HNSCC发病机制中的作用提供了新的见解。
{"title":"Targeting LAMP2A enhances SELENBP1 expression and suppresses malignant behaviors in HNSCC.","authors":"Hongwei Cao, Dongsheng Xing, Hanbing Yu, Feifei Jiang, Li Qi, Kunrong Wang, Yan Wang, Aihui Yan","doi":"10.1158/1541-7786.MCR-25-0388","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-25-0388","url":null,"abstract":"<p><p>Lysosome-Associated Membrane Protein Type 2A (LAMP2A) serves as the critical rate-limiting component of chaperone-mediated autophagy (CMA), governing substrate translocation into lysosomes. Accumulating studies indicate that LAMP2A downregulation leads to CMA impairment in multiple cancer malignancies. In this study, we found that LAMP2A is significantly upregulated in head and neck squamous cell carcinoma (HNSCC) compared to normal tissues. Cell functional studies performed on FaDu and CAL-27 cells showed that downregulation of LAMP2A inhibited cell proliferation and stemness, and induced cell apoptosis. Since CMA specifically targets proteins containing a pentapeptide motif (KFERQ-like motif) in a LAMP2A-dependent manner, we further employed an integrated proteomic-interactomic approach combined with KFERQ motif analysis. This comprehensive strategy identified Selenium Binding Protein 1 (SELENBP1) as a novel putative CMA substrate in HNSCC. Subsequent validation confirmed that knockdown of CMA receptor LAMP2A significantly increased SELENBP1 protein levels both in vitro and in vivo. CO-IP assays confirmed that SELENBP1 interacts with the CMA chaperone protein heat shock cognate 71 kDa protein (HSPA8) in a KFERQ motif (\"EKVIQ\")-dependent manner. Overexpression of SELENBP1 attenuated HNSCC cell proliferation and viability. Most importantly, silencing of SELENBP1 partially rescued the tumor-suppressive phenotypes induced by LAMP2A knockdown, suggesting that SELENBP1 mediated the effects of LAMP2A knockdown on HNSCC. This study provides insights into the role of the LAMP2A-CMA-SELENBP1 axis in the development of novel therapies for HNSCC. Implications: This study provides a novel insight into the role of CMA during the pathogenesis of HNSCC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145401166","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Molecular Cancer Research
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:604180095
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