首页 > 最新文献

Molecular Carcinogenesis最新文献

英文 中文
HMGB1 Assists in Overcoming Cisplatin Resistance in Chemoresistant Human Ovarian Cancer Cells. HMGB1协助克服化疗耐药人卵巢癌细胞的顺铂耐药性。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-09 DOI: 10.1002/mc.70074
Van Huynh, Guliang Wang, Anirban Mukherjee, Karen M Vasquez

Cisplatin is one of the most effective chemotherapeutic agents used in the treatment of ovarian cancer. However, the frequent development of cisplatin resistance remains a significant limitation, leading to therapeutic failure and poor patient outcomes. Cisplatin cytotoxicity is attributed to the generation of toxic DNA lesions, which can be recognized and processed by a variety of proteins, including the high mobility group box 1 (HMGB1) protein. HMGB1 is a multifunctional protein, which is involved in chromatin remodeling and multiple DNA damage repair pathways. In this study, we investigated the role of HMGB1 in modulating cisplatin sensitivity in human ovarian cancer cells. Using cisplatin-sensitive and cisplatin-resistant human ovarian cancer cell lines, we employed siRNA-mediated HMGB1 knockdown to assess its impact on the cellular responses to cisplatin treatment. In clonogenic survival assays, HMGB1 depletion resulted in a significant reduction in colony formation in cisplatin-resistant cells upon cisplatin exposure, compared with nontargeting siRNA treated cells. Additionally, HMGB1 inhibition significantly enhanced cisplatin-induced apoptosis in the cisplatin-resistant cells. Mechanistically, HMGB1-depleted cells exhibited altered DNA damage responses via modulation of ATM/CHK2 and ATR/CHK1 activity following cisplatin treatment. Notably, DNA immunoblot and modified alkaline comet assay results demonstrated that HMGB1 depletion stimulated cisplatin-DNA adduct formation and impaired the removal of cisplatin-DNA adducts, particularly in the cisplatin-resistant cells. Collectively, these findings uncover novel functions of HMGB1 in mediating cisplatin sensitivity, emphasizing its potential as a therapeutic target to overcome cisplatin resistance in ovarian cancer.

顺铂是卵巢癌治疗中最有效的化疗药物之一。然而,顺铂耐药的频繁发展仍然是一个重大限制,导致治疗失败和患者预后不良。顺铂的细胞毒性归因于毒性DNA病变的产生,这种病变可以被多种蛋白质识别和处理,包括高迁移率群盒1 (HMGB1)蛋白。HMGB1是一种多功能蛋白,参与染色质重塑和多种DNA损伤修复途径。在这项研究中,我们研究了HMGB1在调节人卵巢癌细胞顺铂敏感性中的作用。使用顺铂敏感和顺铂耐药的人卵巢癌细胞系,我们采用sirna介导的HMGB1敲低来评估其对顺铂治疗的细胞反应的影响。在克隆生存试验中,与非靶向siRNA处理的细胞相比,HMGB1缺失导致顺铂耐药细胞在顺铂暴露后菌落形成显著减少。此外,HMGB1抑制显著增强顺铂诱导的顺铂耐药细胞凋亡。在机制上,hmgb1缺失的细胞在顺铂治疗后通过调节ATM/CHK2和ATR/CHK1活性表现出改变的DNA损伤反应。值得注意的是,DNA免疫印迹和修饰碱性彗星试验结果表明,HMGB1缺失刺激顺铂-DNA加合物的形成,并损害顺铂-DNA加合物的去除,特别是在顺铂耐药细胞中。总之,这些发现揭示了HMGB1介导顺铂敏感性的新功能,强调了其作为克服卵巢癌顺铂耐药的治疗靶点的潜力。
{"title":"HMGB1 Assists in Overcoming Cisplatin Resistance in Chemoresistant Human Ovarian Cancer Cells.","authors":"Van Huynh, Guliang Wang, Anirban Mukherjee, Karen M Vasquez","doi":"10.1002/mc.70074","DOIUrl":"https://doi.org/10.1002/mc.70074","url":null,"abstract":"<p><p>Cisplatin is one of the most effective chemotherapeutic agents used in the treatment of ovarian cancer. However, the frequent development of cisplatin resistance remains a significant limitation, leading to therapeutic failure and poor patient outcomes. Cisplatin cytotoxicity is attributed to the generation of toxic DNA lesions, which can be recognized and processed by a variety of proteins, including the high mobility group box 1 (HMGB1) protein. HMGB1 is a multifunctional protein, which is involved in chromatin remodeling and multiple DNA damage repair pathways. In this study, we investigated the role of HMGB1 in modulating cisplatin sensitivity in human ovarian cancer cells. Using cisplatin-sensitive and cisplatin-resistant human ovarian cancer cell lines, we employed siRNA-mediated HMGB1 knockdown to assess its impact on the cellular responses to cisplatin treatment. In clonogenic survival assays, HMGB1 depletion resulted in a significant reduction in colony formation in cisplatin-resistant cells upon cisplatin exposure, compared with nontargeting siRNA treated cells. Additionally, HMGB1 inhibition significantly enhanced cisplatin-induced apoptosis in the cisplatin-resistant cells. Mechanistically, HMGB1-depleted cells exhibited altered DNA damage responses via modulation of ATM/CHK2 and ATR/CHK1 activity following cisplatin treatment. Notably, DNA immunoblot and modified alkaline comet assay results demonstrated that HMGB1 depletion stimulated cisplatin-DNA adduct formation and impaired the removal of cisplatin-DNA adducts, particularly in the cisplatin-resistant cells. Collectively, these findings uncover novel functions of HMGB1 in mediating cisplatin sensitivity, emphasizing its potential as a therapeutic target to overcome cisplatin resistance in ovarian cancer.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145934427","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
Insights Into the Crosstalk of HIF and Autophagy: Regulation Mechanisms and Therapeutic Potentials in Cancer. HIF和自噬的串扰:肿瘤的调控机制和治疗潜力。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1002/mc.70081
Chaolei Chen, Jiaqi Lin, Qiwei Liang, Kangmiao Guo, Xingyao Li, Feng Qiu, Ning Kang, Qiang Zhang

Hypoxic and nutrient-deprived conditions are characteristics of the solid tumors' microenvironment (TME), where hypoxia-inducible factor (HIF) and autophagy serve as the central modulators in cancer cells. Herein, we synthesize decades of research regarding HIF and autophagy in cancer, highlighting their regulatory roles in modulating progression, such as HIF-1α-BNIP3/BNIP3L-Beclin-1 complex signaling and HIF-mTOR-ULK1 axis. Given the paucity of comprehensive syntheses regarding this intricate interplay, we systematically deconstruct recent findings to map the molecular landscape connecting HIF-1 to autophagic flux and cell death. Specifically, we explore the metabolic bridges linking HIF and autophagy, including glycolysis and reactive oxygen species (ROS). Furthermore, by refining the content of dual-target molecules, we propose strategies for co-targeting HIF and autophagy, aiming to catalyze the development of novel therapeutic interventions.

缺氧和营养剥夺是实体肿瘤微环境(TME)的特征,其中缺氧诱导因子(HIF)和自噬是癌细胞的中心调节因子。在此,我们综合了几十年来关于HIF和自噬在癌症中的研究,强调了它们在调节进展中的调节作用,如HIF-1α- bnip3 /BNIP3L-Beclin-1复合物信号和HIF- mtor - ulk1轴。鉴于缺乏关于这种复杂相互作用的综合合成,我们系统地解构了最近的发现,以绘制连接HIF-1与自噬通量和细胞死亡的分子景观。具体来说,我们探讨了连接HIF和自噬的代谢桥梁,包括糖酵解和活性氧(ROS)。此外,通过完善双靶点分子的含量,我们提出了HIF和自噬共靶向的策略,旨在促进新的治疗干预措施的发展。
{"title":"Insights Into the Crosstalk of HIF and Autophagy: Regulation Mechanisms and Therapeutic Potentials in Cancer.","authors":"Chaolei Chen, Jiaqi Lin, Qiwei Liang, Kangmiao Guo, Xingyao Li, Feng Qiu, Ning Kang, Qiang Zhang","doi":"10.1002/mc.70081","DOIUrl":"https://doi.org/10.1002/mc.70081","url":null,"abstract":"<p><p>Hypoxic and nutrient-deprived conditions are characteristics of the solid tumors' microenvironment (TME), where hypoxia-inducible factor (HIF) and autophagy serve as the central modulators in cancer cells. Herein, we synthesize decades of research regarding HIF and autophagy in cancer, highlighting their regulatory roles in modulating progression, such as HIF-1α-BNIP3/BNIP3L-Beclin-1 complex signaling and HIF-mTOR-ULK1 axis. Given the paucity of comprehensive syntheses regarding this intricate interplay, we systematically deconstruct recent findings to map the molecular landscape connecting HIF-1 to autophagic flux and cell death. Specifically, we explore the metabolic bridges linking HIF and autophagy, including glycolysis and reactive oxygen species (ROS). Furthermore, by refining the content of dual-target molecules, we propose strategies for co-targeting HIF and autophagy, aiming to catalyze the development of novel therapeutic interventions.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145912473","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
Lung Cancer Cells Secrete Glutamine to Accumulate Tumor-Associated Macrophages. 肺癌细胞分泌谷氨酰胺积聚肿瘤相关巨噬细胞。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1002/mc.70077
Joshua P Reddy, Rebecca A Clague, Beatriz P Peixoto, Sara Bernstein, Emma E Lauth, Clara Y Takanohashi, Sophie C Kim, Hiromi I Wettersten

Tumor-associated macrophages (TAMs) are critical contributors to cancer progression and are often recruited by cancer cells to support a pro-tumorigenic microenvironment. Integrin αvβ3 is a known driver and marker of cancer stem-like properties, including tumor initiation, in various epithelial cancers. We have previously shown a positive correlation between αvβ3 expression and TAM infiltration across multiple tumor types; however, the mechanistic link remains undefined. Here, we demonstrated that integrin αvβ3 expression in non-small cell lung cancer (NSCLC) cells is both necessary and sufficient to drive TAM accumulation. In orthotopic murine and human NSCLC models, ectopic integrin αvβ3 expression increased TAM infiltration independently of T cells, whereas genetic deletion of integrin β3 significantly reduced TAM numbers and tumor burden. Mechanistically, integrin αvβ3 promotes glutamine secretion from NSCLC cells, which enhances the survival and/or differentiation of bone marrow-derived macrophages. Functionally, TAMs are essential for the elevated tumor-initiating capacity of αvβ3+ cancer cells, as macrophage depletion abolished this effect. Together, our findings uncover a novel mechanism by which NSCLC cells remodel the tumor microenvironment via αvβ3-mediated glutamine secretion, promoting TAM enrichment and tumor initiation. Targeting this axis may offer therapeutic benefits in αvβ3-expressing cancers.

肿瘤相关巨噬细胞(tam)是癌症进展的关键贡献者,通常被癌细胞招募以支持促肿瘤微环境。整合素αvβ3是一种已知的驱动因子和肿瘤干细胞样特性的标记物,包括各种上皮癌的肿瘤起始。我们之前已经证明αvβ3的表达与TAM浸润在多种肿瘤类型之间呈正相关;然而,机制上的联系仍未明确。在这里,我们证明了整合素αvβ3在非小细胞肺癌(NSCLC)细胞中的表达是驱动TAM积累的必要和充分条件。在原位小鼠和人非小细胞肺癌模型中,异位整合素αvβ3的表达独立于T细胞增加了TAM浸润,而整合素β3的基因缺失显著减少了TAM数量和肿瘤负荷。机制上,整合素αvβ3促进NSCLC细胞分泌谷氨酰胺,从而增强骨髓源性巨噬细胞的存活和/或分化。在功能上,tam对αvβ3+癌细胞的肿瘤启动能力的提高是必不可少的,因为巨噬细胞的消耗消除了这种作用。总之,我们的研究结果揭示了NSCLC细胞通过αvβ3介导的谷氨酰胺分泌重塑肿瘤微环境,促进TAM富集和肿瘤发生的新机制。靶向这条轴可能对αvβ3表达的癌症提供治疗益处。
{"title":"Lung Cancer Cells Secrete Glutamine to Accumulate Tumor-Associated Macrophages.","authors":"Joshua P Reddy, Rebecca A Clague, Beatriz P Peixoto, Sara Bernstein, Emma E Lauth, Clara Y Takanohashi, Sophie C Kim, Hiromi I Wettersten","doi":"10.1002/mc.70077","DOIUrl":"10.1002/mc.70077","url":null,"abstract":"<p><p>Tumor-associated macrophages (TAMs) are critical contributors to cancer progression and are often recruited by cancer cells to support a pro-tumorigenic microenvironment. Integrin αvβ3 is a known driver and marker of cancer stem-like properties, including tumor initiation, in various epithelial cancers. We have previously shown a positive correlation between αvβ3 expression and TAM infiltration across multiple tumor types; however, the mechanistic link remains undefined. Here, we demonstrated that integrin αvβ3 expression in non-small cell lung cancer (NSCLC) cells is both necessary and sufficient to drive TAM accumulation. In orthotopic murine and human NSCLC models, ectopic integrin αvβ3 expression increased TAM infiltration independently of T cells, whereas genetic deletion of integrin β3 significantly reduced TAM numbers and tumor burden. Mechanistically, integrin αvβ3 promotes glutamine secretion from NSCLC cells, which enhances the survival and/or differentiation of bone marrow-derived macrophages. Functionally, TAMs are essential for the elevated tumor-initiating capacity of αvβ3+ cancer cells, as macrophage depletion abolished this effect. Together, our findings uncover a novel mechanism by which NSCLC cells remodel the tumor microenvironment via αvβ3-mediated glutamine secretion, promoting TAM enrichment and tumor initiation. Targeting this axis may offer therapeutic benefits in αvβ3-expressing cancers.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12799098/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145912429","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
NOP2 Promotes Glycolysis and Tumor Development in Larynx Cancer by Stabilizing TPI1 mRNA Through N5-Methylcytosine Modification. NOP2通过n5 -甲基胞嘧啶修饰稳定TPI1 mRNA,促进喉癌糖酵解和肿瘤发展。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1002/mc.70079
Gan Wang, Zhiling Chen

Larynx cancer, a malignant tumor originating from the epithelial cells of the larynx, remains a significant clinical challenge. Although both N5-methylcytosine (m5C) modification and glycolysis are critically implicated in cancer progression, their functional interplay in larynx cancer is not well defined. This study aims to elucidate the mechanism through which m5C modification influences larynx cancer progression via glycolysis. We performed bioinformatics analysis on the GSE59102 data set to identify m5C-related differentially expressed genes (DEGs) between larynx cancer and normal tissues. Functional assays, including CCK-8, EdU staining, glucose uptake, lactate production, and extracellular acidification rate (ECAR) measurements, were conducted to assess cell viability, proliferation, and glycolysis in larynx cancer cell lines (AMC-HN-8 and TU686). The underlying mechanism was further investigated using methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays. In vivo validation was obtained through xenograft tumor models and immunohistochemistry. Our results demonstrated that the m5C methyltransferase NOP2 was significantly upregulated in larynx cancer. Knockdown of NOP2 inhibited cell viability, proliferation, and glycolysis in larynx cancer cells, and attenuated tumor growth in nude mice. Mechanistically, NOP2 silencing reduced the m5C modification on TPI1 mRNA, thereby decreasing its stability. Furthermore, overexpression of TPI1 rescued the impaired glycolysis in larynx cancer cells caused by NOP2 knockdown. In summary, this study reveals that NOP2 facilitates larynx cancer progression by enhancing glycolysis through m5C-mediated stabilization of TPI1 mRNA. Our findings uncover the NOP2/m5C/TPI1 axis as a novel therapeutic target and provide new insights into RNA methylation-driven metabolic reprogramming in larynx cancer.

喉癌是一种起源于喉部上皮细胞的恶性肿瘤,至今仍是一个重大的临床挑战。尽管n5 -甲基胞嘧啶(m5C)修饰和糖酵解都与癌症进展密切相关,但它们在喉癌中的功能相互作用尚未明确。本研究旨在阐明m5C修饰通过糖酵解影响喉癌进展的机制。我们对GSE59102数据集进行生物信息学分析,以确定喉癌组织与正常组织之间m5c相关的差异表达基因(DEGs)。功能测定,包括CCK-8、EdU染色、葡萄糖摄取、乳酸生成和细胞外酸化率(ECAR)测量,用于评估喉癌细胞系(AMC-HN-8和TU686)的细胞活力、增殖和糖酵解。利用甲基化RNA免疫沉淀(MeRIP)、RNA免疫沉淀(RIP)和双荧光素酶报告基因检测进一步研究了潜在的机制。通过异种移植肿瘤模型和免疫组织化学获得体内验证。我们的研究结果表明,m5C甲基转移酶NOP2在喉癌中显著上调。敲低NOP2抑制喉癌细胞活力、增殖和糖酵解,并减弱裸鼠肿瘤生长。从机制上讲,NOP2沉默减少了m5C对TPI1 mRNA的修饰,从而降低了其稳定性。此外,TPI1的过表达挽救了NOP2敲低引起的喉癌细胞糖酵解受损。综上所述,本研究表明NOP2通过m5c介导的TPI1 mRNA的稳定来促进糖酵解,从而促进喉癌的进展。我们的研究结果揭示了NOP2/m5C/TPI1轴作为一个新的治疗靶点,并为RNA甲基化驱动的喉癌代谢重编程提供了新的见解。
{"title":"NOP2 Promotes Glycolysis and Tumor Development in Larynx Cancer by Stabilizing TPI1 mRNA Through N<sup>5</sup>-Methylcytosine Modification.","authors":"Gan Wang, Zhiling Chen","doi":"10.1002/mc.70079","DOIUrl":"https://doi.org/10.1002/mc.70079","url":null,"abstract":"<p><p>Larynx cancer, a malignant tumor originating from the epithelial cells of the larynx, remains a significant clinical challenge. Although both N<sup>5</sup>-methylcytosine (m<sup>5</sup>C) modification and glycolysis are critically implicated in cancer progression, their functional interplay in larynx cancer is not well defined. This study aims to elucidate the mechanism through which m<sup>5</sup>C modification influences larynx cancer progression via glycolysis. We performed bioinformatics analysis on the GSE59102 data set to identify m<sup>5</sup>C-related differentially expressed genes (DEGs) between larynx cancer and normal tissues. Functional assays, including CCK-8, EdU staining, glucose uptake, lactate production, and extracellular acidification rate (ECAR) measurements, were conducted to assess cell viability, proliferation, and glycolysis in larynx cancer cell lines (AMC-HN-8 and TU686). The underlying mechanism was further investigated using methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays. In vivo validation was obtained through xenograft tumor models and immunohistochemistry. Our results demonstrated that the m<sup>5</sup>C methyltransferase NOP2 was significantly upregulated in larynx cancer. Knockdown of NOP2 inhibited cell viability, proliferation, and glycolysis in larynx cancer cells, and attenuated tumor growth in nude mice. Mechanistically, NOP2 silencing reduced the m<sup>5</sup>C modification on TPI1 mRNA, thereby decreasing its stability. Furthermore, overexpression of TPI1 rescued the impaired glycolysis in larynx cancer cells caused by NOP2 knockdown. In summary, this study reveals that NOP2 facilitates larynx cancer progression by enhancing glycolysis through m<sup>5</sup>C-mediated stabilization of TPI1 mRNA. Our findings uncover the NOP2/m<sup>5</sup>C/TPI1 axis as a novel therapeutic target and provide new insights into RNA methylation-driven metabolic reprogramming in larynx cancer.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145912439","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
Highly Expressed Fatty Acid-Binding Protein 6 Mediates Lipid Metabolism Remodeling in Tumor Cells via Intracellular Bile Acid Transport to Promote Pancreatic Cancer Metastasis. 高表达的脂肪酸结合蛋白6通过细胞内胆汁酸转运介导肿瘤细胞脂质代谢重塑促进胰腺癌转移
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-06 DOI: 10.1002/mc.70076
Jiepeng Jia, Chao Bai, Wen Zhang, Jiale Xu, Tao Hu

Pancreatic cancer (PC) endangers patients' lives and health, and the current diagnosis and treatment situation is not optimistic. Bile acids level was reported to be involved in PC progression, but how they regulate PC progression at the molecular level remains unclear. There is an urgent need to conduct in-depth research. Clinical samples from 58 PC patients and PC cells, including PANC-1 cells and CFAC-1 cells, served as the main research objects. RT-qPCR, IHC, and western blot were used to detect the levels of related molecules. Bile acids, lactate, triglycerides, and cholesterol levels were measured by commercial kits and the lipid levels were evaluated by Oil Red O assay. CCK-8, EdU, and Transwell assays were employed to detect malignant features of PC cells. In PC clinical samples, fatty acid-binding protein 6 (FABP6) expression and endogenous bile acid levels were abnormally elevated. Besides, FABP6 overexpression could accelerate tumor growth and metastasis and facilitated lipid metabolism reprogramming in PC mice. In addition, in PC cells, FABP6 overexpression promoted cellular lipid metabolism by enhancing intracellular bile acid transport, which promoted the malignant characteristics of PC cells. As expected, FABP6 silencing achieved opposite results. Moreover, FABP6 overexpression affected lipid metabolism reprogramming and promoted PC cell malignant features by strengthening intracellular bile acid transport to activate NR1H2/3. FABP6 overexpression promoted lipid metabolism reprogramming by enhancing intracellular bile acid transport to activate NR1H2/3, thereby accelerating PC progression. These findings offered new insights into PC molecular mechanisms and potential therapeutic targets.

胰腺癌危及患者的生命和健康,目前的诊治情况不容乐观。据报道,胆汁酸水平与PC进展有关,但它们如何在分子水平上调节PC进展尚不清楚。迫切需要进行深入的研究。以58例PC患者临床样本和PC细胞为主要研究对象,包括PANC-1细胞和CFAC-1细胞。RT-qPCR、免疫组化、western blot检测相关分子水平。胆汁酸、乳酸、甘油三酯和胆固醇水平用商用试剂盒测定,脂质水平用Oil Red O法测定。CCK-8、EdU和Transwell检测PC细胞的恶性特征。PC临床标本中,脂肪酸结合蛋白6 (FABP6)表达及内源性胆汁酸水平异常升高。此外,FABP6过表达可促进PC小鼠肿瘤生长和转移,促进脂质代谢重编程。此外,在PC细胞中,FABP6过表达通过增强细胞内胆汁酸转运来促进细胞脂质代谢,从而促进PC细胞的恶性特征。正如预期的那样,FABP6沉默取得了相反的结果。此外,FABP6过表达通过加强细胞内胆汁酸转运激活NR1H2/3,影响脂质代谢重编程,促进PC细胞恶性特征。FABP6过表达通过增强细胞内胆汁酸转运激活NR1H2/3,促进脂质代谢重编程,从而加速PC的进展。这些发现为PC分子机制和潜在的治疗靶点提供了新的见解。
{"title":"Highly Expressed Fatty Acid-Binding Protein 6 Mediates Lipid Metabolism Remodeling in Tumor Cells via Intracellular Bile Acid Transport to Promote Pancreatic Cancer Metastasis.","authors":"Jiepeng Jia, Chao Bai, Wen Zhang, Jiale Xu, Tao Hu","doi":"10.1002/mc.70076","DOIUrl":"https://doi.org/10.1002/mc.70076","url":null,"abstract":"<p><p>Pancreatic cancer (PC) endangers patients' lives and health, and the current diagnosis and treatment situation is not optimistic. Bile acids level was reported to be involved in PC progression, but how they regulate PC progression at the molecular level remains unclear. There is an urgent need to conduct in-depth research. Clinical samples from 58 PC patients and PC cells, including PANC-1 cells and CFAC-1 cells, served as the main research objects. RT-qPCR, IHC, and western blot were used to detect the levels of related molecules. Bile acids, lactate, triglycerides, and cholesterol levels were measured by commercial kits and the lipid levels were evaluated by Oil Red O assay. CCK-8, EdU, and Transwell assays were employed to detect malignant features of PC cells. In PC clinical samples, fatty acid-binding protein 6 (FABP6) expression and endogenous bile acid levels were abnormally elevated. Besides, FABP6 overexpression could accelerate tumor growth and metastasis and facilitated lipid metabolism reprogramming in PC mice. In addition, in PC cells, FABP6 overexpression promoted cellular lipid metabolism by enhancing intracellular bile acid transport, which promoted the malignant characteristics of PC cells. As expected, FABP6 silencing achieved opposite results. Moreover, FABP6 overexpression affected lipid metabolism reprogramming and promoted PC cell malignant features by strengthening intracellular bile acid transport to activate NR1H2/3. FABP6 overexpression promoted lipid metabolism reprogramming by enhancing intracellular bile acid transport to activate NR1H2/3, thereby accelerating PC progression. These findings offered new insights into PC molecular mechanisms and potential therapeutic targets.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145912451","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
Combination of Simvastatin and Metformin Reduces Triple-Negative Breast Cancer Tumor Growth Through AKT/AMPK/ACC Signaling Axis. 辛伐他汀联合二甲双胍通过AKT/AMPK/ACC信号轴抑制三阴性乳腺癌肿瘤生长
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 Epub Date: 2025-10-21 DOI: 10.1002/mc.70056
Santosh Kumar Maurya, Shashank Kumar

Chemotherapy continues to be the standard of care for metastatic malignancies, such as triple-negative breast cancer (TNBC). Although the treatment strategy increases survival rates marginally, it frequently leads to the development of resistant disease and side effects. It is imperative to develop an alternate chemotherapy formulation with better efficacy and lesser adverse effects in TNBC patients. Cell viability and cholesterol level were measured using spectrophotometer and fluorometric assays. The 4T1 syngeneic BALB/c female mice were used as an in vivo metastatic TNBC model. Simvastatin (Sim) and Metformin (Met) were administered in combination (3.5-7.0 and 175-350 μg/g body weight, respectively) and alone (Sim 7.0 μg/g/day, or Met 350 μg/g/day) orally over an 8-week period, and the standard Anticancer drug docetaxel (Doc) was administered at a dose of 24 μg/g body weight through IP injection every 3 weeks. Phosphorylation levels of protein and histopathology of tumors were studied by immunoblot and H & E staining methods, respectively. We report that the viability of TNBC cells is significantly and synergistically reduced by Sim and Met co-treatment, with negligible adverse effects on normal breast cell line. Sim Met combination down regulates phosphorylation at specific sites of AKT (Ser-473/Thr-308) and AMPKα (Ser-485/491) and up regulates ACC phosphorylation (Ser-79), which in turn minimizes the cellular cholesterol synthesis in the TNBC model. Further study demonstrated that the combination significantly reduced tumor formation effectively than docetaxel. Study confirmed that the combination of Sim and Met is a promising chemotherapeutic approach for metastatic TNBC.

化疗仍然是转移性恶性肿瘤的标准治疗方法,如三阴性乳腺癌(TNBC)。虽然这种治疗策略略微提高了生存率,但它经常导致耐药疾病和副作用的发展。开发一种疗效更好、不良反应更小的替代化疗方案是TNBC患者的当务之急。用分光光度计和荧光法测定细胞活力和胆固醇水平。采用4T1同基因BALB/c雌性小鼠作为体内转移性TNBC模型。采用辛伐他汀(Sim)和二甲双胍(Met)联合口服(分别为3.5 ~ 7.0 μg/g体重和175 ~ 350 μg/g体重)和单用(Sim 7.0 μg/g/天或Met 350 μg/天),疗程8周,标准抗癌药物多西他赛(Doc)每3周通过IP注射,剂量为24 μg/g体重。分别用免疫印迹法和H、E染色法研究蛋白磷酸化水平和肿瘤组织病理学。我们报告说,Sim和Met联合处理显著地协同降低了TNBC细胞的活力,对正常乳腺细胞系的不良影响可以忽略不计。Sim - Met组合下调AKT (Ser-473/Thr-308)和AMPKα (Ser-485/491)特定位点的磷酸化,上调ACC磷酸化(Ser-79),从而在TNBC模型中最小化细胞胆固醇合成。进一步的研究表明,联合用药比多西紫杉醇更有效地减少肿瘤的形成。研究证实,Sim和Met联合治疗转移性TNBC是一种很有前途的化疗方法。
{"title":"Combination of Simvastatin and Metformin Reduces Triple-Negative Breast Cancer Tumor Growth Through AKT/AMPK/ACC Signaling Axis.","authors":"Santosh Kumar Maurya, Shashank Kumar","doi":"10.1002/mc.70056","DOIUrl":"10.1002/mc.70056","url":null,"abstract":"<p><p>Chemotherapy continues to be the standard of care for metastatic malignancies, such as triple-negative breast cancer (TNBC). Although the treatment strategy increases survival rates marginally, it frequently leads to the development of resistant disease and side effects. It is imperative to develop an alternate chemotherapy formulation with better efficacy and lesser adverse effects in TNBC patients. Cell viability and cholesterol level were measured using spectrophotometer and fluorometric assays. The 4T1 syngeneic BALB/c female mice were used as an in vivo metastatic TNBC model. Simvastatin (Sim) and Metformin (Met) were administered in combination (3.5-7.0 and 175-350 μg/g body weight, respectively) and alone (Sim 7.0 μg/g/day, or Met 350 μg/g/day) orally over an 8-week period, and the standard Anticancer drug docetaxel (Doc) was administered at a dose of 24 μg/g body weight through IP injection every 3 weeks. Phosphorylation levels of protein and histopathology of tumors were studied by immunoblot and H & E staining methods, respectively. We report that the viability of TNBC cells is significantly and synergistically reduced by Sim and Met co-treatment, with negligible adverse effects on normal breast cell line. Sim Met combination down regulates phosphorylation at specific sites of AKT (Ser-473/Thr-308) and AMPKα (Ser-485/491) and up regulates ACC phosphorylation (Ser-79), which in turn minimizes the cellular cholesterol synthesis in the TNBC model. Further study demonstrated that the combination significantly reduced tumor formation effectively than docetaxel. Study confirmed that the combination of Sim and Met is a promising chemotherapeutic approach for metastatic TNBC.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":"69-80"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145346327","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
USP2 Promotes Nasopharyngeal Carcinoma Progression by Activating TRAF2/NF-κB Signaling and Stabilizing MMP2. USP2通过激活TRAF2/NF-κB信号和稳定MMP2促进鼻咽癌进展。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 Epub Date: 2025-10-27 DOI: 10.1002/mc.70054
Tingting Li, Xiujuan Huang, Weiwei Xu, Junjun Qi, Lili Liu, Ping Chen, Yingchun Cao

Metastasis remains the primary determinant of poor prognosis in nasopharyngeal carcinoma (NPC). While dysregulated ubiquitination drives cancer progression, the functional contributions of deubiquitinating enzymes (DUBs) to NPC dissemination are poorly defined. Here, we investigated USP2, a DUB implicated in oncogenesis, as a potential regulator of NPC migration and invasion. In our study, bioinformatics analysis of the GEO data set GSE200792 identified USP2 as a metastasis-associated gene with elevated m6A methylation and mRNA levels in metastatic NPC. Validation employed qPCR, Western blot, and immunohistochemistry in clinical samples and NPC cell lines. Functional assays included CCK-8, flow cytometry, Transwell, wound healing, and mechanistic studies such as cyclohexane chase, co-immunoprecipitation, ubiquitination assays were performed under USP2 knockdown/overexpression and NF-κB inhibition. Our results showed that USP2 was significantly upregulated in metastatic NPC tissues and cell lines. USP2 knockdown suppressed proliferation, migration, and invasion, induced apoptosis, and attenuated NF-κB activation by reducing nuclear p65 and TRAF2/MMP2 expression. Conversely, USP2 overexpression enhanced malignancy, which was reversed by NF-κB inhibition. Critically, USP2 directly bound MMP2, extended its protein half-life, and reduced K48-linked polyubiquitination. In conclusion, USP2 drives NPC migration and invasion by activating TRAF2-dependent NF-κB signaling and directly mediating K48-linked deubiquitination of MMP2, dually enhancing MMP2 expression. Targeting the USP2-MMP2 axis may offer a novel therapeutic strategy to impede NPC dissemination, addressing an unmet clinical need in advanced disease.

转移仍然是鼻咽癌(NPC)预后不良的主要决定因素。虽然失调的泛素化驱动癌症进展,但去泛素化酶(DUBs)对NPC传播的功能贡献尚不明确。在这里,我们研究了USP2,一个与肿瘤发生有关的DUB,作为NPC迁移和侵袭的潜在调节因子。在我们的研究中,对GEO数据集GSE200792的生物信息学分析发现,USP2是转移性鼻咽癌中m6A甲基化和mRNA水平升高的转移相关基因。在临床样本和鼻咽癌细胞系中采用qPCR、Western blot和免疫组织化学进行验证。功能测试包括CCK-8、流式细胞术、Transwell、伤口愈合,以及机制研究,如环己烷追踪、共免疫沉淀、泛素化测试,在USP2敲低/过表达和NF-κB抑制下进行。我们的研究结果表明,USP2在转移性鼻咽癌组织和细胞系中显著上调。USP2敲低可抑制细胞增殖、迁移和侵袭,诱导细胞凋亡,并通过降低核p65和TRAF2/MMP2的表达减弱NF-κB的活化。相反,USP2过表达可增强恶性肿瘤,这可通过NF-κB抑制而逆转。关键是,USP2直接结合MMP2,延长其蛋白质半衰期,并减少k48连接的多泛素化。综上所述,USP2通过激活traf2依赖的NF-κB信号和直接介导k48相关的MMP2去泛素化,双重增强MMP2的表达,从而驱动NPC的迁移和侵袭。靶向USP2-MMP2轴可能提供一种新的治疗策略来阻止鼻咽癌的传播,解决了晚期疾病未满足的临床需求。
{"title":"USP2 Promotes Nasopharyngeal Carcinoma Progression by Activating TRAF2/NF-κB Signaling and Stabilizing MMP2.","authors":"Tingting Li, Xiujuan Huang, Weiwei Xu, Junjun Qi, Lili Liu, Ping Chen, Yingchun Cao","doi":"10.1002/mc.70054","DOIUrl":"10.1002/mc.70054","url":null,"abstract":"<p><p>Metastasis remains the primary determinant of poor prognosis in nasopharyngeal carcinoma (NPC). While dysregulated ubiquitination drives cancer progression, the functional contributions of deubiquitinating enzymes (DUBs) to NPC dissemination are poorly defined. Here, we investigated USP2, a DUB implicated in oncogenesis, as a potential regulator of NPC migration and invasion. In our study, bioinformatics analysis of the GEO data set GSE200792 identified USP2 as a metastasis-associated gene with elevated m<sup>6</sup>A methylation and mRNA levels in metastatic NPC. Validation employed qPCR, Western blot, and immunohistochemistry in clinical samples and NPC cell lines. Functional assays included CCK-8, flow cytometry, Transwell, wound healing, and mechanistic studies such as cyclohexane chase, co-immunoprecipitation, ubiquitination assays were performed under USP2 knockdown/overexpression and NF-κB inhibition. Our results showed that USP2 was significantly upregulated in metastatic NPC tissues and cell lines. USP2 knockdown suppressed proliferation, migration, and invasion, induced apoptosis, and attenuated NF-κB activation by reducing nuclear p65 and TRAF2/MMP2 expression. Conversely, USP2 overexpression enhanced malignancy, which was reversed by NF-κB inhibition. Critically, USP2 directly bound MMP2, extended its protein half-life, and reduced K48-linked polyubiquitination. In conclusion, USP2 drives NPC migration and invasion by activating TRAF2-dependent NF-κB signaling and directly mediating K48-linked deubiquitination of MMP2, dually enhancing MMP2 expression. Targeting the USP2-MMP2 axis may offer a novel therapeutic strategy to impede NPC dissemination, addressing an unmet clinical need in advanced disease.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":"94-105"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145377999","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
NSUN2 Promotes Prostate Cancer Cell Proliferation and Migration Through m5C Modifications of YES1. NSUN2通过m5C修饰YES1促进前列腺癌细胞增殖和迁移
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 Epub Date: 2025-11-02 DOI: 10.1002/mc.70060
Tianming Chen, Xiaokang Yang, Shuai Su, Delin Wang

Prostate cancer (PCa) is one of the most common genitourinary malignancies in men worldwide. As a 5-methylcytosine (m5C) methyltransferase, NSUN2 has been implicated in regulating PCa progression. This study aimed to investigate the role of NSUN2 in PCa and elucidate its underlying mechanisms. The biological behaviors of PCa cells were assessed using Cell Counting Kit-8, EdU incorporation, and Transwell assays. The expression levels of relevant RNAs were determined via quantitative real-time PCR. The interaction between NSUN2 and YES proto-oncogene 1 (YES1) was examined through methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays. Results showed that NSUN2 was elevated in PCa, and its downregulation suppressed cell viability, proliferation, migration, and invasion. Mechanistically, NSUN2 interacted with YES1 and stabilized its mRNA by promoting m5C modification on YES1. The oncogenic role of NSUN2 was further confirmed in xenograft models in vivo. In conclusion, our study demonstrated that NSUN2 facilitated malignant proliferation and migration of PCa cells by enhancing YES1 mRNA stability via m5C modification. These findings suggested that both NSUN2 and YES1 may serve as potential therapeutic targets for PCa, offering new strategies for treatment.

前列腺癌(PCa)是全球男性最常见的泌尿生殖系统恶性肿瘤之一。作为一种5-甲基胞嘧啶(m5C)甲基转移酶,NSUN2参与调节前列腺癌的进展。本研究旨在探讨NSUN2在前列腺癌中的作用并阐明其潜在机制。采用细胞计数试剂盒-8、EdU掺入和Transwell法评估PCa细胞的生物学行为。通过实时荧光定量PCR检测相关rna的表达水平。通过甲基化RNA免疫沉淀(MeRIP)、RNA免疫沉淀(RIP)和双荧光素酶报告基因检测检测NSUN2和YES原癌基因1 (YES1)之间的相互作用。结果表明,NSUN2在PCa中表达升高,其下调抑制了细胞活力、增殖、迁移和侵袭。机制上,NSUN2与YES1相互作用,并通过促进m5C修饰YES1来稳定其mRNA。在体内异种移植瘤模型中进一步证实了NSUN2的致癌作用。总之,我们的研究表明,NSUN2通过m5C修饰增强YES1 mRNA的稳定性,促进了PCa细胞的恶性增殖和迁移。这些发现表明,NSUN2和YES1可能是PCa的潜在治疗靶点,为治疗PCa提供了新的策略。
{"title":"NSUN2 Promotes Prostate Cancer Cell Proliferation and Migration Through m5C Modifications of YES1.","authors":"Tianming Chen, Xiaokang Yang, Shuai Su, Delin Wang","doi":"10.1002/mc.70060","DOIUrl":"10.1002/mc.70060","url":null,"abstract":"<p><p>Prostate cancer (PCa) is one of the most common genitourinary malignancies in men worldwide. As a 5-methylcytosine (m5C) methyltransferase, NSUN2 has been implicated in regulating PCa progression. This study aimed to investigate the role of NSUN2 in PCa and elucidate its underlying mechanisms. The biological behaviors of PCa cells were assessed using Cell Counting Kit-8, EdU incorporation, and Transwell assays. The expression levels of relevant RNAs were determined via quantitative real-time PCR. The interaction between NSUN2 and YES proto-oncogene 1 (YES1) was examined through methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays. Results showed that NSUN2 was elevated in PCa, and its downregulation suppressed cell viability, proliferation, migration, and invasion. Mechanistically, NSUN2 interacted with YES1 and stabilized its mRNA by promoting m5C modification on YES1. The oncogenic role of NSUN2 was further confirmed in xenograft models in vivo. In conclusion, our study demonstrated that NSUN2 facilitated malignant proliferation and migration of PCa cells by enhancing YES1 mRNA stability via m5C modification. These findings suggested that both NSUN2 and YES1 may serve as potential therapeutic targets for PCa, offering new strategies for treatment.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":"138-145"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145427034","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
TOP2A, Stabilized by IGF2BP3 in an m6A-Dependent Manner, Drives Macrophage Recruitment and M2 Polarization in Hepatocellular Carcinoma by YAP1-Mediated CCL2 Activation. TOP2A被IGF2BP3以依赖m6a的方式稳定,通过yap1介导的CCL2激活在肝癌中驱动巨噬细胞募集和M2极化。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 Epub Date: 2025-11-02 DOI: 10.1002/mc.70059
Lu Li, Chenxu Du, Nai Sun, Xingguo Xiao, Kunkun Li, Huili Wu, Jianzhuang Gong

M2-like tumor-associated macrophages (TAMs) exert immunosuppressive and protumorigenic functions in hepatocellular carcinoma (HCC). In this study, we explored the function and mechanism of Type IIA topoisomerase (TOP2A) in TAM recruitment and M2 polarization in the HCC microenvironment. The IGF2BP3-TOP2A relationship was confirmed by RIP, MeRIP, luciferase, and mRNA stability assays. Coculture experiments using a transwell system were performed to analyze the impact on the migration, CD206+ cell population, and M2-related marker expression in THP-1-differentiated macrophages (THP-1-M0). Xenograft models were constructed to evaluate TOP2A's role in tumor growth. Expression analysis was performed by quantitative PCR (qPCR), immunoblotting, and immunohistochemical staining. Increased TOP2A expression was associated with advanced tumor stage and worse outcomes in HCC. IGF2BP3 was upregulated and positively correlated with TOP2A expression in HCC samples. TOP2A depletion reduced THP-1-M0 migration and M2 polarization in vitro and attenuated xenograft growth by suppressing TAM infiltration and M2 polarization in vivo. Mechanistically, IGF2BP3 recognized METTL3-catalyzed m6A sites to increase the stability and expression of TOP2A mRNA. TOP2A re-expression abolished IGF2BP3 knockdown-driven suppression of THP-1-M0 migration and M2 polarization. Moreover, TOP2A depletion decreased CCL2 production and YAP1 activation. CCL2 reconstruction or the Hippo pathway inhibitor XMU-MP-1 reversed TOP2A knockdown-driven suppression of THP-1-M0 migration and M2 polarization. Our findings identify the IGF2BP3/TOP2A axis as a master regulator of TAM recruitment and polarization in HCC via IGF2BP3-m6A-dependent TOP2A stabilization to facilitate YAP1-mediated CCL2 upregulation, providing novel strategies to overcome immunosuppression and combat HCC.

m2样肿瘤相关巨噬细胞(tam)在肝细胞癌(HCC)中发挥免疫抑制和致瘤功能。在本研究中,我们探讨了IIA型拓扑异构酶(TOP2A)在HCC微环境中TAM募集和M2极化中的功能和机制。通过RIP、MeRIP、荧光素酶和mRNA稳定性分析证实了IGF2BP3-TOP2A的关系。利用transwell系统进行共培养实验,分析对thp -1分化巨噬细胞(THP-1-M0)迁移、CD206+细胞群和m2相关标志物表达的影响。构建异种移植物模型来评估TOP2A在肿瘤生长中的作用。通过定量PCR (qPCR)、免疫印迹和免疫组织化学染色进行表达分析。在HCC中,TOP2A表达增加与肿瘤分期晚期和预后较差相关。HCC样本中IGF2BP3表达上调,与TOP2A表达呈正相关。TOP2A耗竭减少THP-1-M0在体外的迁移和M2极化,并通过抑制体内TAM浸润和M2极化来减弱异种移植物的生长。从机制上讲,IGF2BP3识别mettl3催化的m6A位点,从而增加TOP2A mRNA的稳定性和表达。TOP2A的重新表达消除了IGF2BP3敲低驱动的THP-1-M0迁移和M2极化的抑制。此外,TOP2A的缺失降低了CCL2的产生和YAP1的激活。CCL2重建或Hippo通路抑制剂XMU-MP-1逆转了TOP2A敲低驱动的THP-1-M0迁移和M2极化的抑制。我们的研究发现IGF2BP3/TOP2A轴是HCC中TAM募集和极化的主要调节因子,通过IGF2BP3- m6a依赖的TOP2A稳定促进yap1介导的CCL2上调,为克服免疫抑制和对抗HCC提供了新的策略。
{"title":"TOP2A, Stabilized by IGF2BP3 in an m6A-Dependent Manner, Drives Macrophage Recruitment and M2 Polarization in Hepatocellular Carcinoma by YAP1-Mediated CCL2 Activation.","authors":"Lu Li, Chenxu Du, Nai Sun, Xingguo Xiao, Kunkun Li, Huili Wu, Jianzhuang Gong","doi":"10.1002/mc.70059","DOIUrl":"10.1002/mc.70059","url":null,"abstract":"<p><p>M2-like tumor-associated macrophages (TAMs) exert immunosuppressive and protumorigenic functions in hepatocellular carcinoma (HCC). In this study, we explored the function and mechanism of Type IIA topoisomerase (TOP2A) in TAM recruitment and M2 polarization in the HCC microenvironment. The IGF2BP3-TOP2A relationship was confirmed by RIP, MeRIP, luciferase, and mRNA stability assays. Coculture experiments using a transwell system were performed to analyze the impact on the migration, CD206<sup>+</sup> cell population, and M2-related marker expression in THP-1-differentiated macrophages (THP-1-M0). Xenograft models were constructed to evaluate TOP2A's role in tumor growth. Expression analysis was performed by quantitative PCR (qPCR), immunoblotting, and immunohistochemical staining. Increased TOP2A expression was associated with advanced tumor stage and worse outcomes in HCC. IGF2BP3 was upregulated and positively correlated with TOP2A expression in HCC samples. TOP2A depletion reduced THP-1-M0 migration and M2 polarization in vitro and attenuated xenograft growth by suppressing TAM infiltration and M2 polarization in vivo. Mechanistically, IGF2BP3 recognized METTL3-catalyzed m6A sites to increase the stability and expression of TOP2A mRNA. TOP2A re-expression abolished IGF2BP3 knockdown-driven suppression of THP-1-M0 migration and M2 polarization. Moreover, TOP2A depletion decreased CCL2 production and YAP1 activation. CCL2 reconstruction or the Hippo pathway inhibitor XMU-MP-1 reversed TOP2A knockdown-driven suppression of THP-1-M0 migration and M2 polarization. Our findings identify the IGF2BP3/TOP2A axis as a master regulator of TAM recruitment and polarization in HCC via IGF2BP3-m6A-dependent TOP2A stabilization to facilitate YAP1-mediated CCL2 upregulation, providing novel strategies to overcome immunosuppression and combat HCC.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":"123-137"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145427043","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
AN02, a Naovel Curcumin Derivative, Orchestrates APC-SMAD4-Mediated CTLA-4 Degradation for Ovarian Cancer Therapy. AN02是一种新型姜黄素衍生物,可协调apc - smad4介导的CTLA-4降解,用于卵巢癌治疗。
IF 3.2 2区 医学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 Epub Date: 2025-10-21 DOI: 10.1002/mc.70048
Hairong Jin, Mengjie Zhang, Mengna Shi, Pin Miao, Min-Jie Zhang

Curcumin is a natural polyphenolic compound extracted from the rhizomes of Curcuma longa, exhibiting a wide range of biological activities, including anti-inflammatory, antioxidant, antitumor, antibacterial, antiviral, and neuroprotective effects. However, its low oral absorption rate and poor bioavailability limit its clinical application. To address this issue, this study synthesized a novel curcumin derivative, AN02, which significantly improves the absorption rate and bioavailability while enhancing its antitumor activity. This study focused on the antitumor mechanism of AN02 in ovarian cancer, particularly its ability to inhibit ovarian cancer cell proliferation, invasion, and migration by regulating the APC (Adenomatous Polyposis Coli)-SMAD4 (SMAD family member 4)-CTLA-4 (Cytotoxic T-Lymphocyte-Associated Protein 4) molecular axis. Experimental results demonstrated that AN02 significantly inhibited ovarian cancer cell proliferation at very low concentrations, with its half-maximal inhibitory concentration (IC50) significantly lower than that of curcumin. Additionally, AN02 exerted its antitumor effects by activating the APC-SMAD4 molecular axis and inhibiting the CTLA-4 molecular axis. Silencing CTLA-4 inhibits the proliferation and immune escape of ovarian cancer. Further molecular mechanism studies revealed that APC directly regulates the SMAD4-CTLA-4 molecular axis, while SMAD4 forms a chaperone relationship with CTLA-4 and promotes CTLA-4 degradation through the K48-dependent ubiquitination pathway, thereby suppressing the malignant phenotype of ovarian cancer cells. These findings not only reveal the antitumor mechanism of AN02 but also provide new insights for the treatment of ovarian cancer. Animal experiments also demonstrated that AN02 significantly inhibits the proliferation of subcutaneous xenograft tumors in mice. As a novel curcumin derivative, AN02 exhibits significant antitumor activity and inhibits ovarian cancer progression by regulating the APC-SMAD4-CTLA-4 molecular axis. This study lays an important theoretical foundation for the development of novel antitumor drugs based on AN02, with significant clinical application potential. However, the long-term toxicity and safety of AN02 require further investigation to establish safe dosage standards for clinical use. Future studies will focus on exploring combination therapy strategies of AN02 in cisplatin-resistant ovarian cancer to provide new directions for precision treatment of ovarian cancer.

姜黄素是从姜黄根茎中提取的一种天然多酚类化合物,具有抗炎、抗氧化、抗肿瘤、抗菌、抗病毒和神经保护等多种生物活性。但口服吸收率低,生物利用度差,限制了其临床应用。为了解决这一问题,本研究合成了一种新的姜黄素衍生物AN02,该衍生物显著提高了姜黄素的吸收率和生物利用度,同时增强了其抗肿瘤活性。本研究重点研究了AN02在卵巢癌中的抗肿瘤机制,特别是其通过调节APC (Adenomatous Polyposis Coli)-SMAD4 (SMAD家族成员4)-CTLA-4 (Cytotoxic T-Lymphocyte-Associated Protein 4)分子轴抑制卵巢癌细胞增殖、侵袭和迁移的能力。实验结果表明,AN02在极低浓度下显著抑制卵巢癌细胞增殖,其半最大抑制浓度(IC50)显著低于姜黄素。此外,AN02通过激活APC-SMAD4分子轴和抑制CTLA-4分子轴发挥抗肿瘤作用。沉默CTLA-4抑制卵巢癌的增殖和免疫逃逸。进一步的分子机制研究发现,APC直接调控SMAD4-CTLA-4分子轴,而SMAD4与CTLA-4形成伴侣关系,通过k48依赖的泛素化途径促进CTLA-4降解,从而抑制卵巢癌细胞的恶性表型。这些发现不仅揭示了AN02的抗肿瘤机制,也为卵巢癌的治疗提供了新的思路。动物实验也表明,AN02显著抑制小鼠皮下异种移植肿瘤的增殖。AN02是一种新型姜黄素衍生物,具有显著的抗肿瘤活性,通过调节APC-SMAD4-CTLA-4分子轴抑制卵巢癌进展。本研究为开发基于AN02的新型抗肿瘤药物奠定了重要的理论基础,具有重要的临床应用潜力。然而,AN02的长期毒性和安全性需要进一步研究,以建立临床使用的安全剂量标准。未来的研究将重点探索AN02在顺铂耐药卵巢癌中的联合治疗策略,为卵巢癌的精准治疗提供新的方向。
{"title":"AN02, a Naovel Curcumin Derivative, Orchestrates APC-SMAD4-Mediated CTLA-4 Degradation for Ovarian Cancer Therapy.","authors":"Hairong Jin, Mengjie Zhang, Mengna Shi, Pin Miao, Min-Jie Zhang","doi":"10.1002/mc.70048","DOIUrl":"10.1002/mc.70048","url":null,"abstract":"<p><p>Curcumin is a natural polyphenolic compound extracted from the rhizomes of Curcuma longa, exhibiting a wide range of biological activities, including anti-inflammatory, antioxidant, antitumor, antibacterial, antiviral, and neuroprotective effects. However, its low oral absorption rate and poor bioavailability limit its clinical application. To address this issue, this study synthesized a novel curcumin derivative, AN02, which significantly improves the absorption rate and bioavailability while enhancing its antitumor activity. This study focused on the antitumor mechanism of AN02 in ovarian cancer, particularly its ability to inhibit ovarian cancer cell proliferation, invasion, and migration by regulating the APC (Adenomatous Polyposis Coli)-SMAD4 (SMAD family member 4)-CTLA-4 (Cytotoxic T-Lymphocyte-Associated Protein 4) molecular axis. Experimental results demonstrated that AN02 significantly inhibited ovarian cancer cell proliferation at very low concentrations, with its half-maximal inhibitory concentration (IC<sub>50</sub>) significantly lower than that of curcumin. Additionally, AN02 exerted its antitumor effects by activating the APC-SMAD4 molecular axis and inhibiting the CTLA-4 molecular axis. Silencing CTLA-4 inhibits the proliferation and immune escape of ovarian cancer. Further molecular mechanism studies revealed that APC directly regulates the SMAD4-CTLA-4 molecular axis, while SMAD4 forms a chaperone relationship with CTLA-4 and promotes CTLA-4 degradation through the K48-dependent ubiquitination pathway, thereby suppressing the malignant phenotype of ovarian cancer cells. These findings not only reveal the antitumor mechanism of AN02 but also provide new insights for the treatment of ovarian cancer. Animal experiments also demonstrated that AN02 significantly inhibits the proliferation of subcutaneous xenograft tumors in mice. As a novel curcumin derivative, AN02 exhibits significant antitumor activity and inhibits ovarian cancer progression by regulating the APC-SMAD4-CTLA-4 molecular axis. This study lays an important theoretical foundation for the development of novel antitumor drugs based on AN02, with significant clinical application potential. However, the long-term toxicity and safety of AN02 require further investigation to establish safe dosage standards for clinical use. Future studies will focus on exploring combination therapy strategies of AN02 in cisplatin-resistant ovarian cancer to provide new directions for precision treatment of ovarian cancer.</p>","PeriodicalId":19003,"journal":{"name":"Molecular Carcinogenesis","volume":" ","pages":"18-38"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145346262","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 Carcinogenesis
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