首页 > 最新文献

Oncogenesis最新文献

英文 中文
FLT4 activation promotes acute lymphoid leukemia survival through stabilization of MDM2/MDMX and inactivation of p53. FLT4激活通过稳定MDM2/MDMX和p53失活来促进急性淋巴细胞白血病的存活。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-05-02 DOI: 10.1038/s41389-025-00552-7
Djazia Haferssas, Marion Dubuissez, Jonatan Barrera-Chimal, Clémence Messmer, El Bachir Affar, Bruno Larrivée, Xue-Song Liu, Casimiro Gerarduzzi

Aberrant Receptor Tyrosine Kinase (RTK) signaling allows cancer cells to modulate survival, proliferation, and death, leading to tumorigenesis and chemoresistance. In leukemia, the RTK FMS-Related Tyrosine Kinase 4 (FLT4) (also known as VEGFR3, Vascular Endothelial Growth Factor Receptor- 3) is deregulated and correlates with cancer progression. However, the underlying consequences of its deregulation remain to be determined. Moreover, chemotherapy treatment requires that cancer cells retain a wild-type p53 to respond to DNA damage by tumor-suppressing activities, i.e. apoptosis. p53 activity is predominantly limited by its two major negative regulators, MDM2 and MDMX, which inactivate p53 by promoting its degradation and/or cytoplasmic localization. In this study, we have shown that activation of FLT4 by either overexpression or binding of its ligand, VEGFC, increases MDM2/MDMX stability, inactivates p53, and leads to resistance to DNA-damaging therapies. Moreover, we found that MDMX Ser-314 phosphorylation, a consensus sequence of CDK4/6, increases MDMX stability, which subsequently affects MDM2 and p53 degradation and could be reversed by the CDK4/6 inhibitor Palbociclib. More importantly, leukemic cells treated with Palbociclib were more susceptible to DNA-damaging induction of apoptosis and had reduced cell proliferation. Leukemic cells overexpressing FLT4 displayed accelerated proliferation when injected into NOD-SCID mice as compared to wild-type cells. Altogether, our research proposes an innovative way to reactivate p53 in leukemia through the pharmacological inhibition of FLT4 signaling, which could serve as a potential treatment option. Schematic representation of FLT4-mediated MDM2/MDMX complex stabilization and suppression of p53 activity. VEGFC triggers FLT4 activation, leading to CDK4/6 activation, which phosphorylates MDMX on Ser-314. As a result, MDMX levels increase and bind to MDM2, stabilizing the MDM2/MDMX complex. This complex binds to p53, facilitating its suppression by reducing its transcriptional activity or enhancing its export to the cytoplasm for proteasomal degradation. Consequently, p53 inactivation promotes their survival, proliferation, and resistance to chemotherapy-induced apoptosis. The figure was created in BioRender.com.

异常的受体酪氨酸激酶(RTK)信号允许癌细胞调节生存、增殖和死亡,导致肿瘤发生和化疗耐药。在白血病中,RTK fms相关酪氨酸激酶4 (FLT4)(也称为VEGFR3,血管内皮生长因子受体- 3)被解除调控并与癌症进展相关。然而,放松管制的潜在后果仍有待确定。此外,化疗需要癌细胞保留野生型p53,以通过肿瘤抑制活性(即凋亡)对DNA损伤做出反应。p53的活性主要受其两个主要负调节因子MDM2和MDMX的限制,它们通过促进p53的降解和/或细胞质定位而使p53失活。在这项研究中,我们已经证明,通过过表达或结合其配体VEGFC激活FLT4,可以增加MDM2/MDMX的稳定性,使p53失活,并导致对dna损伤治疗的抵抗。此外,我们发现MDMX Ser-314磷酸化(CDK4/6的共识序列)增加了MDMX的稳定性,从而影响MDM2和p53的降解,并且可以被CDK4/6抑制剂Palbociclib逆转。更重要的是,帕博西尼治疗的白血病细胞更容易受到dna损伤诱导凋亡,细胞增殖减少。与野生型细胞相比,将过表达FLT4的白血病细胞注射到NOD-SCID小鼠体内时,其增殖速度加快。总之,我们的研究提出了一种创新的方法,通过药物抑制FLT4信号来重新激活白血病中的p53,这可能是一种潜在的治疗选择。flt4介导的MDM2/MDMX复合物稳定和p53活性抑制的示意图。VEGFC触发FLT4激活,导致CDK4/6激活,从而磷酸化Ser-314上的MDMX。结果,MDMX水平增加并与MDM2结合,稳定MDM2/MDMX复合物。该复合物与p53结合,通过降低其转录活性或增强其向细胞质出口以进行蛋白酶体降解,从而促进其抑制。因此,p53的失活促进了它们的存活、增殖和对化疗诱导的细胞凋亡的抵抗。这个图是在BioRender.com中创建的。
{"title":"FLT4 activation promotes acute lymphoid leukemia survival through stabilization of MDM2/MDMX and inactivation of p53.","authors":"Djazia Haferssas, Marion Dubuissez, Jonatan Barrera-Chimal, Clémence Messmer, El Bachir Affar, Bruno Larrivée, Xue-Song Liu, Casimiro Gerarduzzi","doi":"10.1038/s41389-025-00552-7","DOIUrl":"https://doi.org/10.1038/s41389-025-00552-7","url":null,"abstract":"<p><p>Aberrant Receptor Tyrosine Kinase (RTK) signaling allows cancer cells to modulate survival, proliferation, and death, leading to tumorigenesis and chemoresistance. In leukemia, the RTK FMS-Related Tyrosine Kinase 4 (FLT4) (also known as VEGFR3, Vascular Endothelial Growth Factor Receptor- 3) is deregulated and correlates with cancer progression. However, the underlying consequences of its deregulation remain to be determined. Moreover, chemotherapy treatment requires that cancer cells retain a wild-type p53 to respond to DNA damage by tumor-suppressing activities, i.e. apoptosis. p53 activity is predominantly limited by its two major negative regulators, MDM2 and MDMX, which inactivate p53 by promoting its degradation and/or cytoplasmic localization. In this study, we have shown that activation of FLT4 by either overexpression or binding of its ligand, VEGFC, increases MDM2/MDMX stability, inactivates p53, and leads to resistance to DNA-damaging therapies. Moreover, we found that MDMX Ser-314 phosphorylation, a consensus sequence of CDK4/6, increases MDMX stability, which subsequently affects MDM2 and p53 degradation and could be reversed by the CDK4/6 inhibitor Palbociclib. More importantly, leukemic cells treated with Palbociclib were more susceptible to DNA-damaging induction of apoptosis and had reduced cell proliferation. Leukemic cells overexpressing FLT4 displayed accelerated proliferation when injected into NOD-SCID mice as compared to wild-type cells. Altogether, our research proposes an innovative way to reactivate p53 in leukemia through the pharmacological inhibition of FLT4 signaling, which could serve as a potential treatment option. Schematic representation of FLT4-mediated MDM2/MDMX complex stabilization and suppression of p53 activity. VEGFC triggers FLT4 activation, leading to CDK4/6 activation, which phosphorylates MDMX on Ser-314. As a result, MDMX levels increase and bind to MDM2, stabilizing the MDM2/MDMX complex. This complex binds to p53, facilitating its suppression by reducing its transcriptional activity or enhancing its export to the cytoplasm for proteasomal degradation. Consequently, p53 inactivation promotes their survival, proliferation, and resistance to chemotherapy-induced apoptosis. The figure was created in BioRender.com.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"14"},"PeriodicalIF":5.9,"publicationDate":"2025-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12048674/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144037157","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
MYCN and KAT2A form a feedforward loop to drive an oncogenic transcriptional program in neuroblastoma. 在神经母细胞瘤中,MYCN和KAT2A形成一个前馈回路来驱动致癌转录程序。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-24 DOI: 10.1038/s41389-025-00557-2
Zhihui Liu, Jason J Hong, Xiyuan Zhang, Carly M Sayers, Wendy Fang, Man Xu, Sydney Loria, Sakereh Maskal, Haiyan Lei, Haitao Wu, Rolf Swenson, Jordan L Meier, Jack F Shern, Carol J Thiele

The oncoprotein MYCN drives malignancy in various cancer types, including neuroblastoma (NB). However, our understanding of the mechanisms underlying its transcriptional activity and oncogenic function, as well as effective strategies to target it, remains limited. We discovered that MYCN interacts with the transcriptional coactivator KAT2A, and this interaction significantly contributes to MYCN's activity in NB. Our genome-wide analyses indicate MYCN recruits KAT2A to bind to DNA, thereby transcriptionally regulating genes associated with ribosome biogenesis and RNA processing. Moreover, we identified that MYCN directly activates KAT2A transcription, while KAT2A acetylates MYCN, increasing MYCN protein stability. Consequently, MYCN and KAT2A establish a feedforward loop that effectively regulates global gene expression, governing the malignant NB phenotype. Treatment of NB cells with a KAT2A Proteolysis Targeting Chimera (PROTAC) degrader reduces MYCN protein levels, antagonizes MYCN-mediated gene transcription regulation and suppresses cell proliferation. This study highlights the potential of transcriptional cofactors as viable targets for developing anti-MYCN therapies.

癌蛋白MYCN驱动各种癌症类型的恶性肿瘤,包括神经母细胞瘤(NB)。然而,我们对其转录活性和致癌功能的机制以及针对它的有效策略的理解仍然有限。我们发现MYCN与转录辅激活子KAT2A相互作用,这种相互作用显著地促进了MYCN在NB中的活性。我们的全基因组分析表明,MYCN招募KAT2A与DNA结合,从而转录调节与核糖体生物发生和RNA加工相关的基因。此外,我们发现MYCN直接激活KAT2A转录,而KAT2A乙酰化MYCN,增加MYCN蛋白的稳定性。因此,MYCN和KAT2A建立了一个前馈回路,有效地调节了全球基因表达,控制了恶性NB表型。用KAT2A蛋白水解靶向嵌合体(PROTAC)降解剂处理NB细胞可降低MYCN蛋白水平,拮抗MYCN介导的基因转录调控并抑制细胞增殖。这项研究强调了转录辅助因子作为开发抗mycn疗法的可行靶点的潜力。
{"title":"MYCN and KAT2A form a feedforward loop to drive an oncogenic transcriptional program in neuroblastoma.","authors":"Zhihui Liu, Jason J Hong, Xiyuan Zhang, Carly M Sayers, Wendy Fang, Man Xu, Sydney Loria, Sakereh Maskal, Haiyan Lei, Haitao Wu, Rolf Swenson, Jordan L Meier, Jack F Shern, Carol J Thiele","doi":"10.1038/s41389-025-00557-2","DOIUrl":"https://doi.org/10.1038/s41389-025-00557-2","url":null,"abstract":"<p><p>The oncoprotein MYCN drives malignancy in various cancer types, including neuroblastoma (NB). However, our understanding of the mechanisms underlying its transcriptional activity and oncogenic function, as well as effective strategies to target it, remains limited. We discovered that MYCN interacts with the transcriptional coactivator KAT2A, and this interaction significantly contributes to MYCN's activity in NB. Our genome-wide analyses indicate MYCN recruits KAT2A to bind to DNA, thereby transcriptionally regulating genes associated with ribosome biogenesis and RNA processing. Moreover, we identified that MYCN directly activates KAT2A transcription, while KAT2A acetylates MYCN, increasing MYCN protein stability. Consequently, MYCN and KAT2A establish a feedforward loop that effectively regulates global gene expression, governing the malignant NB phenotype. Treatment of NB cells with a KAT2A Proteolysis Targeting Chimera (PROTAC) degrader reduces MYCN protein levels, antagonizes MYCN-mediated gene transcription regulation and suppresses cell proliferation. This study highlights the potential of transcriptional cofactors as viable targets for developing anti-MYCN therapies.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"13"},"PeriodicalIF":5.9,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12022051/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144017455","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
EPHA5 promotes cell proliferation and inhibits apoptosis in Follicular Thyroid Cancer via the STAT3 signaling pathway. EPHA5通过STAT3信号通路促进滤泡性甲状腺癌细胞增殖和抑制细胞凋亡。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-22 DOI: 10.1038/s41389-025-00556-3
Guorong Wang, Gaoran Xu, Yanan Fan, Guangzhi Wang, Jingchao Xu, Ning Zhang, Junzhu Chen, Huanjie Chen, Zhoufan Li, Xianwang Cao, Yongfu Zhao

Follicular thyroid carcinoma (FTC) is a common endocrine malignancy characterized by a higher propensity for invasion and metastasis compared to papillary thyroid carcinoma (PTC). Ephrin type A receptor 5 (EPHA5) is a crucial receptor tyrosine kinase involved in orchestrating diverse physiological processes, including apoptosis and proliferation. However, the mechanism of EPHA5 in FTC remains unclear. This study identified significant overexpression of EPHA5 in FTC. In vitro experiments showed that increased expression of EPHA5 promotes proliferation and inhibits apoptosis in FTC. Furthermore, EPHA5 activates the STAT3 signaling pathway. To explore the interaction between EPHA5 and the STAT3 signaling pathway, we used SH-4-54 (a STAT3-specific inhibitor). Interestingly, the influence of EPHA5 on proliferation and apoptosis was reduced upon combination with SH-4-54. In summary, this study unveils the involvement of the EPHA5-STAT3 signaling pathway in FTC and implies that the function of EPHA5 in FTC may partly depend on the STAT3 signaling pathway.

滤泡性甲状腺癌(FTC)是一种常见的内分泌恶性肿瘤,与乳头状甲状腺癌(PTC)相比,其侵袭和转移的倾向更高。Ephrin type A receptor 5 (EPHA5)是一种重要的酪氨酸激酶受体,参与多种生理过程,包括细胞凋亡和增殖。然而,EPHA5在FTC中的作用机制尚不清楚。本研究发现EPHA5在FTC中显著过表达。体外实验表明,EPHA5表达增加可促进FTC细胞增殖,抑制凋亡。此外,EPHA5激活STAT3信号通路。为了探索EPHA5与STAT3信号通路之间的相互作用,我们使用了SH-4-54(一种STAT3特异性抑制剂)。有趣的是,与SH-4-54联合后,EPHA5对细胞增殖和凋亡的影响减弱。综上所述,本研究揭示了EPHA5-STAT3信号通路参与FTC,提示EPHA5在FTC中的功能可能部分依赖于STAT3信号通路。
{"title":"EPHA5 promotes cell proliferation and inhibits apoptosis in Follicular Thyroid Cancer via the STAT3 signaling pathway.","authors":"Guorong Wang, Gaoran Xu, Yanan Fan, Guangzhi Wang, Jingchao Xu, Ning Zhang, Junzhu Chen, Huanjie Chen, Zhoufan Li, Xianwang Cao, Yongfu Zhao","doi":"10.1038/s41389-025-00556-3","DOIUrl":"https://doi.org/10.1038/s41389-025-00556-3","url":null,"abstract":"<p><p>Follicular thyroid carcinoma (FTC) is a common endocrine malignancy characterized by a higher propensity for invasion and metastasis compared to papillary thyroid carcinoma (PTC). Ephrin type A receptor 5 (EPHA5) is a crucial receptor tyrosine kinase involved in orchestrating diverse physiological processes, including apoptosis and proliferation. However, the mechanism of EPHA5 in FTC remains unclear. This study identified significant overexpression of EPHA5 in FTC. In vitro experiments showed that increased expression of EPHA5 promotes proliferation and inhibits apoptosis in FTC. Furthermore, EPHA5 activates the STAT3 signaling pathway. To explore the interaction between EPHA5 and the STAT3 signaling pathway, we used SH-4-54 (a STAT3-specific inhibitor). Interestingly, the influence of EPHA5 on proliferation and apoptosis was reduced upon combination with SH-4-54. In summary, this study unveils the involvement of the EPHA5-STAT3 signaling pathway in FTC and implies that the function of EPHA5 in FTC may partly depend on the STAT3 signaling pathway.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"12"},"PeriodicalIF":5.9,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12015243/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144014538","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
O-GlcNAcylation of FBP1 promotes pancreatic cancer progression by facilitating its Lys48-linked polyubiquitination in hypoxic environments. FBP1的o - glcn酰化通过促进其在缺氧环境中与lys48相关的多泛素化而促进胰腺癌的进展。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-22 DOI: 10.1038/s41389-025-00555-4
Yi Zhu, Xiaoman He, Xiaojing Ma, Yan Zhang, Wei Feng

Fructose-1,6-bisphosphatase 1 (FBP1), a rate-limiting enzyme in gluconeogenesis, is important for cancer progression. The post-translational regulation of FBP1 in hypoxic environments is still unclear. Here, we report that FBP1 is down-regulated, and a low expression level of FBP1 predicts a poor prognosis in pancreatic cancer. A hypoxic environment makes FBP1 more prone to degradation, and this effect can be reversed by inhibiting global O-GlcNAcylation signalling. O-linked N-acetylglucosamine transferase (OGT) interacts with FBP1 and induces its O-GlcNAcylation at serine 47 residue (FBP1-S47) to modulate its protein function in pancreatic cancer cells. O-GlcNAcylation of FBP1-S47 promotes FBP1 degradation and also influences the expression of canonical HIF-1α target genes involved in glucose metabolism, resulting in an increase in glucose uptake and lactate secretion in pancreatic cancer cells. In addition, O-GlcNAcylation of FBP1-S47 facilitates FBP1 K48-linked polyubiquitination at lysine 51 residue (FBP1-K51), in which GlcNAc moiety can serve as a prerequisite for an FBP1 ubiquitin ligase. FBP1 (K51) K48-linked polyubiquitination mediated protein degradation can also promote cancer progression, similarly to the O-GlcNAcylation of FBP1-S47. Our data uncover a mechanism whereby FBP1 can be regulated by a protein O-GlcNAcylation-polyubiquitination axis, paving the way to cancer cell metabolic reprogramming.

果糖-1,6-二磷酸酶1 (FBP1)是糖异生中的限速酶,对癌症的进展很重要。FBP1在缺氧环境下的翻译后调控尚不清楚。在这里,我们报道FBP1是下调的,FBP1的低表达水平预示着胰腺癌的不良预后。缺氧环境使FBP1更容易降解,这种效应可以通过抑制全局o - glcn酰化信号传导来逆转。O-linked N-acetylglucosamine transferase (OGT)与FBP1相互作用,诱导其丝氨酸47残基上的o - glcn酰化(FBP1- s47)调节其在胰腺癌细胞中的蛋白功能。FBP1- s47的o - glcn酰化促进FBP1降解,并影响参与葡萄糖代谢的典型HIF-1α靶基因的表达,导致胰腺癌细胞中葡萄糖摄取和乳酸分泌增加。此外,FBP1- s47的o - glcn酰化促进了FBP1 k48连接的赖氨酸51残基多泛素化(FBP1- k51),其中GlcNAc片段可以作为FBP1泛素连接酶的先决条件。FBP1 (K51) k48关联的多泛素化介导的蛋白降解也可以促进癌症进展,类似于FBP1- s47的o - glcn酰化。我们的数据揭示了FBP1可以通过蛋白o - glcn酰化-多泛素化轴调节的机制,为癌细胞代谢重编程铺平了道路。
{"title":"O-GlcNAcylation of FBP1 promotes pancreatic cancer progression by facilitating its Lys48-linked polyubiquitination in hypoxic environments.","authors":"Yi Zhu, Xiaoman He, Xiaojing Ma, Yan Zhang, Wei Feng","doi":"10.1038/s41389-025-00555-4","DOIUrl":"https://doi.org/10.1038/s41389-025-00555-4","url":null,"abstract":"<p><p>Fructose-1,6-bisphosphatase 1 (FBP1), a rate-limiting enzyme in gluconeogenesis, is important for cancer progression. The post-translational regulation of FBP1 in hypoxic environments is still unclear. Here, we report that FBP1 is down-regulated, and a low expression level of FBP1 predicts a poor prognosis in pancreatic cancer. A hypoxic environment makes FBP1 more prone to degradation, and this effect can be reversed by inhibiting global O-GlcNAcylation signalling. O-linked N-acetylglucosamine transferase (OGT) interacts with FBP1 and induces its O-GlcNAcylation at serine 47 residue (FBP1-S47) to modulate its protein function in pancreatic cancer cells. O-GlcNAcylation of FBP1-S47 promotes FBP1 degradation and also influences the expression of canonical HIF-1α target genes involved in glucose metabolism, resulting in an increase in glucose uptake and lactate secretion in pancreatic cancer cells. In addition, O-GlcNAcylation of FBP1-S47 facilitates FBP1 K48-linked polyubiquitination at lysine 51 residue (FBP1-K51), in which GlcNAc moiety can serve as a prerequisite for an FBP1 ubiquitin ligase. FBP1 (K51) K48-linked polyubiquitination mediated protein degradation can also promote cancer progression, similarly to the O-GlcNAcylation of FBP1-S47. Our data uncover a mechanism whereby FBP1 can be regulated by a protein O-GlcNAcylation-polyubiquitination axis, paving the way to cancer cell metabolic reprogramming.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"11"},"PeriodicalIF":5.9,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12015445/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144035474","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
Chasing non-existent "microRNAs" in cancer. 在癌症中寻找不存在的“microrna”。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-18 DOI: 10.1038/s41389-025-00550-9
Ayla Orang, Nicholas I Warnock, Melodie Migault, B Kate Dredge, Andrew G Bert, Julie M Bracken, Philip A Gregory, Katherine A Pillman, Gregory J Goodall, Cameron P Bracken

MicroRNAs (miRNAs) are important regulators of gene expression whose dysregulation is widely linked to tumourigenesis, tumour progression and Epithelial-Mesenchymal Transition (EMT), a developmental process that promotes metastasis when inappropriately activated. However, controversy has emerged regarding how many functional miRNAs are encoded in the genome, and to what extent non-regulatory products of RNA degradation have been mis-identified as miRNAs. Central to miRNA function is their capacity to associate with an Argonaute (AGO) protein and form an RNA-Induced Silencing Complex (RISC), which mediates target mRNA suppression. We report that numerous "miRNAs" previously reported in EMT and cancer contexts, are not incorporated into RISC and are not capable of endogenously silencing target genes, despite the fact that hundreds of publications in the cancer field describe their roles. Apparent function can be driven through the expression of artificial miRNA mimics which is not necessarily reflective of any endogenous gene regulatory function. We present biochemical and bioinformatic criteria that can be used to distinguish functional miRNAs from mistakenly annotated RNA fragments.

MicroRNAs (miRNAs)是基因表达的重要调控因子,其失调与肿瘤发生、肿瘤进展和上皮-间质转化(EMT)广泛相关,EMT是一种不适当激活时促进转移的发育过程。然而,关于基因组中编码了多少功能性mirna,以及RNA降解的非调节性产物在多大程度上被错误地识别为mirna,已经出现了争议。miRNA功能的核心是它们与Argonaute (AGO)蛋白结合并形成rna诱导沉默复合体(RISC)的能力,该复合体介导靶mRNA的抑制。我们报告了先前在EMT和癌症背景下报道的许多“mirna”,没有纳入RISC,并且不能内源性沉默靶基因,尽管癌症领域的数百篇出版物描述了它们的作用。表观功能可以通过人工miRNA模拟物的表达来驱动,而这并不一定反映任何内源基因的调控功能。我们提出了生化和生物信息学标准,可用于区分功能性mirna和错误注释的RNA片段。
{"title":"Chasing non-existent \"microRNAs\" in cancer.","authors":"Ayla Orang, Nicholas I Warnock, Melodie Migault, B Kate Dredge, Andrew G Bert, Julie M Bracken, Philip A Gregory, Katherine A Pillman, Gregory J Goodall, Cameron P Bracken","doi":"10.1038/s41389-025-00550-9","DOIUrl":"https://doi.org/10.1038/s41389-025-00550-9","url":null,"abstract":"<p><p>MicroRNAs (miRNAs) are important regulators of gene expression whose dysregulation is widely linked to tumourigenesis, tumour progression and Epithelial-Mesenchymal Transition (EMT), a developmental process that promotes metastasis when inappropriately activated. However, controversy has emerged regarding how many functional miRNAs are encoded in the genome, and to what extent non-regulatory products of RNA degradation have been mis-identified as miRNAs. Central to miRNA function is their capacity to associate with an Argonaute (AGO) protein and form an RNA-Induced Silencing Complex (RISC), which mediates target mRNA suppression. We report that numerous \"miRNAs\" previously reported in EMT and cancer contexts, are not incorporated into RISC and are not capable of endogenously silencing target genes, despite the fact that hundreds of publications in the cancer field describe their roles. Apparent function can be driven through the expression of artificial miRNA mimics which is not necessarily reflective of any endogenous gene regulatory function. We present biochemical and bioinformatic criteria that can be used to distinguish functional miRNAs from mistakenly annotated RNA fragments.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"10"},"PeriodicalIF":5.9,"publicationDate":"2025-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008284/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144022071","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
Myeloma mesenchymal stem cells’ bioenergetics afford a novel selective therapeutic target 骨髓瘤间充质干细胞的生物能为新型选择性治疗提供了靶点
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-11 DOI: 10.1038/s41389-025-00554-5
Oded Komemi, Elina Orbuch, Osnat Jarchowsky-Dolberg, Yaron Shraga Brin, Shelly Tartakover-Matalon, Metsada Pasmanik-Chor, Michael Lishner, Liat Drucker

Bone-marrow mesenchymal stem cells (BM-MSCs) rely on glycolysis, yet their trafficked mitochondria benefit recipient cells’ bioenergetics in regenerative and cancerous settings, most relevant to BM-resident multiple myeloma (MM) cells. Fission/fusion dynamics regulate mitochondria function. Proteomics demonstrates excessive mitochondrial processes in BM-MSCs from MM patients compared to normal donors (ND). Thus, we aimed to characterize BM-MSCs (ND, MM) mitochondrial fitness, bioenergetics and dynamics with a focus on therapeutics. MM-MSCs displayed compromised mitochondria evidenced by decreased mitochondrial membrane potential (ΔΨm) and elevated proton leak. This was accompanied by stimulation of stress-coping mechanisms: spare respiratory capacity (SRC), mitochondrial fusion and UPRmt. Interfering with BM-MSCs mitochondrial dynamics equilibrium demonstrated their significance to bioenergetics and fitness according to the source. While ND-MSCs depended on fission, reducing MM-MSCs fusion attenuated glycolysis, OXPHOS and mtROS. Interestingly, optimization of mtROS levels is central to ΔΨm preservation in MM-MSCs only. MM-MSCs also demonstrated STAT3 activation, which regulates their OXPHOS and SRC. Targeting MM-MSC’ SRC with Venetoclax diminished their pro-MM support and sensitized co-cultured MM cells to Bortezomib. Overall, MM-MSCs distinct mitochondrial bioenergetics are integral to their robustness. Repurposing Venetoclax as anti-SRC treatment in combination with conventional anti-MM drugs presents a potential selective way to target MM-MSCs conferred drug resistance.

骨髓间充质干细胞(BM-MSCs)依赖于糖酵解,但其运输的线粒体在再生和癌变环境中有益于受体细胞的生物能量学,这与BM-MSCs中的多发性骨髓瘤(MM)细胞最为相关。裂变/融合动力学调节线粒体功能。蛋白质组学显示,与正常供体(ND)相比,来自MM患者的BM-MSCs中线粒体过程过多。因此,我们旨在表征BM-MSCs (ND, MM)线粒体适应度,生物能量学和动力学,重点是治疗方法。MM-MSCs显示线粒体受损,线粒体膜电位降低(ΔΨm)和质子泄漏升高。这伴随着应激应对机制的刺激:备用呼吸能力(SRC)、线粒体融合和UPRmt。干扰骨髓间充质干细胞线粒体动力学平衡对生物能量学和适应度具有重要意义。而ND-MSCs依赖于裂变,减少MM-MSCs融合会减弱糖酵解、OXPHOS和mtROS。有趣的是,仅在MM-MSCs中,mtROS水平的优化是ΔΨm保存的核心。MM-MSCs也表现出STAT3激活,其调控OXPHOS和SRC。Venetoclax靶向MM- msc SRC,降低了MM细胞的前支持,并使共培养的MM细胞对硼替佐米敏感。总的来说,MM-MSCs独特的线粒体生物能量学是其稳健性的组成部分。将Venetoclax与常规抗mm药物联合用于抗src治疗,为靶向MM-MSCs产生耐药性提供了一种潜在的选择性方法。
{"title":"Myeloma mesenchymal stem cells’ bioenergetics afford a novel selective therapeutic target","authors":"Oded Komemi, Elina Orbuch, Osnat Jarchowsky-Dolberg, Yaron Shraga Brin, Shelly Tartakover-Matalon, Metsada Pasmanik-Chor, Michael Lishner, Liat Drucker","doi":"10.1038/s41389-025-00554-5","DOIUrl":"https://doi.org/10.1038/s41389-025-00554-5","url":null,"abstract":"<p>Bone-marrow mesenchymal stem cells (BM-MSCs) rely on glycolysis, yet their trafficked mitochondria benefit recipient cells’ bioenergetics in regenerative and cancerous settings, most relevant to BM-resident multiple myeloma (MM) cells. Fission/fusion dynamics regulate mitochondria function. Proteomics demonstrates excessive mitochondrial processes in BM-MSCs from MM patients compared to normal donors (ND). Thus, we aimed to characterize BM-MSCs (ND, MM) mitochondrial fitness, bioenergetics and dynamics with a focus on therapeutics. MM-MSCs displayed compromised mitochondria evidenced by decreased mitochondrial membrane potential (ΔΨm) and elevated proton leak. This was accompanied by stimulation of stress-coping mechanisms: spare respiratory capacity (SRC), mitochondrial fusion and UPR<sup>mt</sup>. Interfering with BM-MSCs mitochondrial dynamics equilibrium demonstrated their significance to bioenergetics and fitness according to the source. While ND-MSCs depended on fission, reducing MM-MSCs fusion attenuated glycolysis, OXPHOS and mtROS. Interestingly, optimization of mtROS levels is central to ΔΨm preservation in MM-MSCs only. MM-MSCs also demonstrated STAT3 activation, which regulates their OXPHOS and SRC. Targeting MM-MSC’ SRC with Venetoclax diminished their pro-MM support and sensitized co-cultured MM cells to Bortezomib. Overall, MM-MSCs distinct mitochondrial bioenergetics are integral to their robustness. Repurposing Venetoclax as anti-SRC treatment in combination with conventional anti-MM drugs presents a potential selective way to target MM-MSCs conferred drug resistance.</p><figure></figure>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":""},"PeriodicalIF":6.2,"publicationDate":"2025-04-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143832549","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
A HOTAIR-associated super-enhancer orchestrates glioblastoma malignancy via MEST. hotair相关的超级增强子通过MEST协调胶质母细胞瘤恶性。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-07 DOI: 10.1038/s41389-025-00551-8
Peng Li, Yang Yang, Chunpeng Luan, Wenbin Wang, Yuan Jiang, Zhenhao Zhao, Bo Wang, Yuting Zhao, Yunlong Bai, Man Liu, Zhongfang Zhao, Lei Zhang, Yuyang Qian, Jiandang Shi

Glioblastoma (GBM) is one of the most malignant primary brain tumors, and factors governing its progression are not fully characterized. Recent research suggests that the long non-coding RNA (lncRNA) HOTAIR and super-enhancers (SEs) contribute significantly to GBM progression. Here, we performed TCGA data analysis revealing that high HOTAIR expression in GBM is associated with poor prognosis. Conversely, HOTAIR knock-down (KD) decreased proliferation, colony formation, and invasion of GBM cells. Furthermore, RNA-seq analysis identified DEGs in GBM cells related to cell growth and adhesion. Using an integrated approach, we also identify MEST as a HOTAIR-associated SE target gene. Intriguingly, MEST suppression in GBM cells phenocopied HOTAIR KD, as evidenced by reduced cell proliferation and invasion, whereas MEST overexpression counteracted effects of HOTAIR depletion. Moreover, 3 C technique-based PCR confirmed reduced interaction between HOTAIR-associated SEs and target genes after HOTAIR KD. This study reveals a novel regulatory mechanism governing GBM, offering promising directions for clinical interventions.

胶质母细胞瘤(GBM)是最恶性的原发性脑肿瘤之一,控制其进展的因素尚未完全确定。最近的研究表明,长链非编码RNA (lncRNA) HOTAIR和超级增强子(se)在GBM的进展中起着重要作用。本研究通过TCGA数据分析发现,HOTAIR在GBM中的高表达与不良预后相关。相反,HOTAIR敲除(KD)可降低GBM细胞的增殖、集落形成和侵袭。此外,RNA-seq分析鉴定了GBM细胞中与细胞生长和粘附相关的DEGs。通过综合方法,我们还确定了MEST是hotair相关的SE靶基因。有趣的是,在GBM细胞中,MEST抑制表型HOTAIR KD,证明了细胞增殖和侵袭减少,而MEST过表达抵消了HOTAIR耗损的影响。此外,基于3c技术的PCR证实HOTAIR KD后HOTAIR相关SEs与靶基因的相互作用减少。本研究揭示了一种新的GBM调控机制,为临床干预提供了有希望的方向。
{"title":"A HOTAIR-associated super-enhancer orchestrates glioblastoma malignancy via MEST.","authors":"Peng Li, Yang Yang, Chunpeng Luan, Wenbin Wang, Yuan Jiang, Zhenhao Zhao, Bo Wang, Yuting Zhao, Yunlong Bai, Man Liu, Zhongfang Zhao, Lei Zhang, Yuyang Qian, Jiandang Shi","doi":"10.1038/s41389-025-00551-8","DOIUrl":"10.1038/s41389-025-00551-8","url":null,"abstract":"<p><p>Glioblastoma (GBM) is one of the most malignant primary brain tumors, and factors governing its progression are not fully characterized. Recent research suggests that the long non-coding RNA (lncRNA) HOTAIR and super-enhancers (SEs) contribute significantly to GBM progression. Here, we performed TCGA data analysis revealing that high HOTAIR expression in GBM is associated with poor prognosis. Conversely, HOTAIR knock-down (KD) decreased proliferation, colony formation, and invasion of GBM cells. Furthermore, RNA-seq analysis identified DEGs in GBM cells related to cell growth and adhesion. Using an integrated approach, we also identify MEST as a HOTAIR-associated SE target gene. Intriguingly, MEST suppression in GBM cells phenocopied HOTAIR KD, as evidenced by reduced cell proliferation and invasion, whereas MEST overexpression counteracted effects of HOTAIR depletion. Moreover, 3 C technique-based PCR confirmed reduced interaction between HOTAIR-associated SEs and target genes after HOTAIR KD. This study reveals a novel regulatory mechanism governing GBM, offering promising directions for clinical interventions.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"8"},"PeriodicalIF":5.9,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11976998/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143803857","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
FLT3 inhibition upregulates OCT4/NANOG to promote maintenance and TKI resistance of FLT3-ITD+ acute myeloid leukemia. FLT3抑制上调OCT4/NANOG,促进FLT3- itd +急性髓系白血病的维持和TKI耐药。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-03-29 DOI: 10.1038/s41389-025-00553-6
Qi Zhou, Zijian Li, Pingping Zhao, Yongyu Guan, Huiyuan Chu, Yaming Xi

Up to 30% of acute myeloid leukemia (AML) patients face unfavorable outcomes due to the FMS-like receptor tyrosine kinase-3 (FLT3) internal tandem duplication (ITD) mutation. Although FLT3 inhibitors show encouraging outcomes in treatment, they fail to eliminate leukemia stem cells, the origin of persistent and resistant lesions. Exploration of the mechanism in FLT3-ITD+ AML maintenance and chemoresistance is crucial for the development of novel therapeutic approaches. The manifestation of pluripotency transcription factors (TFs) and their link to clinical outcomes have been documented in various tumors. This study investigates the correlation between core pluripotency TF and treatment in AML. We discovered that FLT3 inhibition induced upregulation of OCT4 and NANOG in FLT3-ITD+ AML cells. Subsequently, we demonstrated that downregulation of OCT4 or NANOG inhibited cell growth, promoted apoptosis, and induced G0/G1 cell cycle phase arrest in FLT3-ITD+ AML cells. Knockdown of OCT and NANOG inhibited tumor growth in a mouse tumor model. OCT4 promotes the malignant biological behavior of FLT3-ITD+ AML by enhancing the abnormal FLT3 signaling pathway through transcriptional activation of NANOG. Importantly, downregulation of OCT4 or NANOG increased responsiveness to FLT3-tyrosine kinase inhibitor (TKI) (Gilteritinib), implying that OCT4 and NANOG may contribute to TKI resistance in FLT3-ITD+ AML. Our study verifies the involvement of OCT4/NANOG in regulating TKI sensitivity and targeting them may improve the cytotoxicity of FLT3-TKIs in FLT3-ITD+ AML.

高达30%的急性髓性白血病(AML)患者由于fms样受体酪氨酸激酶-3 (FLT3)内部串联重复(ITD)突变而面临不良结局。尽管FLT3抑制剂在治疗中显示出令人鼓舞的结果,但它们不能消除白血病干细胞,这是持续和耐药病变的起源。探索FLT3-ITD+ AML维持和化疗耐药的机制对于开发新的治疗方法至关重要。多能性转录因子(TFs)的表现及其与临床结果的联系已在各种肿瘤中得到证实。本研究探讨核心多能性TF与AML治疗之间的相关性。我们发现FLT3抑制可诱导FLT3- itd + AML细胞中OCT4和NANOG的上调。随后,我们证明了在FLT3-ITD+ AML细胞中,下调OCT4或NANOG抑制细胞生长,促进细胞凋亡,并诱导G0/G1细胞周期期阻滞。在小鼠肿瘤模型中,敲低OCT和NANOG抑制肿瘤生长。OCT4通过转录激活NANOG,增强异常FLT3信号通路,促进FLT3- itd + AML的恶性生物学行为。重要的是,OCT4或NANOG的下调增加了对flt3 -酪氨酸激酶抑制剂(TKI) (Gilteritinib)的反应性,这意味着OCT4和NANOG可能有助于FLT3-ITD+ AML的TKI耐药。我们的研究证实OCT4/NANOG参与调节TKI敏感性,靶向它们可能改善FLT3-TKIs在FLT3-ITD+ AML中的细胞毒性。
{"title":"FLT3 inhibition upregulates OCT4/NANOG to promote maintenance and TKI resistance of FLT3-ITD<sup>+</sup> acute myeloid leukemia.","authors":"Qi Zhou, Zijian Li, Pingping Zhao, Yongyu Guan, Huiyuan Chu, Yaming Xi","doi":"10.1038/s41389-025-00553-6","DOIUrl":"10.1038/s41389-025-00553-6","url":null,"abstract":"<p><p>Up to 30% of acute myeloid leukemia (AML) patients face unfavorable outcomes due to the FMS-like receptor tyrosine kinase-3 (FLT3) internal tandem duplication (ITD) mutation. Although FLT3 inhibitors show encouraging outcomes in treatment, they fail to eliminate leukemia stem cells, the origin of persistent and resistant lesions. Exploration of the mechanism in FLT3-ITD<sup>+</sup> AML maintenance and chemoresistance is crucial for the development of novel therapeutic approaches. The manifestation of pluripotency transcription factors (TFs) and their link to clinical outcomes have been documented in various tumors. This study investigates the correlation between core pluripotency TF and treatment in AML. We discovered that FLT3 inhibition induced upregulation of OCT4 and NANOG in FLT3-ITD<sup>+</sup> AML cells. Subsequently, we demonstrated that downregulation of OCT4 or NANOG inhibited cell growth, promoted apoptosis, and induced G0/G1 cell cycle phase arrest in FLT3-ITD<sup>+</sup> AML cells. Knockdown of OCT and NANOG inhibited tumor growth in a mouse tumor model. OCT4 promotes the malignant biological behavior of FLT3-ITD<sup>+</sup> AML by enhancing the abnormal FLT3 signaling pathway through transcriptional activation of NANOG. Importantly, downregulation of OCT4 or NANOG increased responsiveness to FLT3-tyrosine kinase inhibitor (TKI) (Gilteritinib), implying that OCT4 and NANOG may contribute to TKI resistance in FLT3-ITD<sup>+</sup> AML. Our study verifies the involvement of OCT4/NANOG in regulating TKI sensitivity and targeting them may improve the cytotoxicity of FLT3-TKIs in FLT3-ITD<sup>+</sup> AML.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"7"},"PeriodicalIF":5.9,"publicationDate":"2025-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11954930/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143743412","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
Tumor cell-derived ISG15 promotes fibroblast recruitment in oral squamous cell carcinoma via CD11a-dependent glycolytic reprogramming. 肿瘤细胞来源的ISG15通过cd11a依赖性糖酵解重编程促进口腔鳞状细胞癌的成纤维细胞募集。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-03-11 DOI: 10.1038/s41389-025-00549-2
Ssu-Han Wang, Yu-Lin Chen, Shih-Han Huang, Yu-Ke Fu, Su-Fang Lin, Shih Sheng Jiang, Shu-Chen Liu, Jenn-Ren Hsiao, Jang-Yang Chang, Ya-Wen Chen

Cancer-associated fibroblast (CAF) recruitment and activation within the tumor microenvironment (TME) are increasingly acknowledged as drivers of oral squamous cell carcinoma (OSCC) tumor growth and metastasis. Therefore, the mechanisms underlying tumor cell and fibroblast crosstalk warrant further investigation. We discovered that ectopic interferon-stimulated gene 15 (ISG15) expression, which is a promising and novel oncoprotein biomarker elevated in a variety of cancers, enhanced OSCC growth and elevated collagen and α-smooth muscle actin (α-SMA) expression in ISG15-expressing tumors. Analysis of immunohistochemistry revealed high ISG15 expression in human oral tissues correlated with high expression of α-SMA and fibroblast activation protein (FAP). Fibroblast migration and recruitment by ISG15-expressing OSCC cells were confirmed by in vitro and in vivo experiments. Exogenous ISG15 induced fibroblast migration, morphological changes, and vimentin expression. Enrichment of glycolysis pathway genes, as well as increased glycolysis-related gene expression, glucose uptake, and lactate production were observed in ISG15-treated fibroblasts. Lactate release and fibroblast migration were blocked by a competitive inhibitor of glucose metabolism. Furthermore, the knockdown of integrin αL (ITGAL)/CD11a, a subunit of ISG15 receptor lymphocyte functional-associated antigen-1 (LFA-1), in immortalized fibroblasts diminished extracellular ISG15-mediated glycolysis and migration. Our findings suggest that ISG15 derived from OSCC cells interacts with fibroblasts through the LFA-1 receptor, leading to glycolytic reprogramming and promotion of fibroblast migration into the TME.

肿瘤微环境(TME)内癌症相关成纤维细胞(CAF)的募集和激活越来越被认为是口腔鳞状细胞癌(OSCC)肿瘤生长和转移的驱动因素。因此,肿瘤细胞和成纤维细胞串扰的机制值得进一步研究。我们发现异位干扰素刺激基因15 (ISG15)的表达,这是一种在多种癌症中有前景的新型癌蛋白生物标志物,在表达ISG15的肿瘤中促进OSCC生长,提高胶原和α-平滑肌肌动蛋白(α-SMA)的表达。免疫组化分析显示,ISG15在人口腔组织中的高表达与α-SMA和成纤维细胞活化蛋白(FAP)的高表达相关。体外和体内实验证实了isg15表达的OSCC细胞对成纤维细胞的迁移和募集。外源性ISG15诱导成纤维细胞迁移、形态改变和波形蛋白表达。在isg15处理的成纤维细胞中,糖酵解途径基因丰富,糖酵解相关基因表达增加,葡萄糖摄取和乳酸生成增加。乳酸释放和成纤维细胞迁移被竞争性葡萄糖代谢抑制剂阻断。此外,ISG15受体淋巴细胞功能相关抗原-1 (LFA-1)的亚基整合素αL (ITGAL)/CD11a在永活成纤维细胞中的表达下调会减少ISG15介导的细胞外糖酵解和迁移。我们的研究结果表明,来源于OSCC细胞的ISG15通过LFA-1受体与成纤维细胞相互作用,导致糖酵解重编程并促进成纤维细胞向TME迁移。
{"title":"Tumor cell-derived ISG15 promotes fibroblast recruitment in oral squamous cell carcinoma via CD11a-dependent glycolytic reprogramming.","authors":"Ssu-Han Wang, Yu-Lin Chen, Shih-Han Huang, Yu-Ke Fu, Su-Fang Lin, Shih Sheng Jiang, Shu-Chen Liu, Jenn-Ren Hsiao, Jang-Yang Chang, Ya-Wen Chen","doi":"10.1038/s41389-025-00549-2","DOIUrl":"10.1038/s41389-025-00549-2","url":null,"abstract":"<p><p>Cancer-associated fibroblast (CAF) recruitment and activation within the tumor microenvironment (TME) are increasingly acknowledged as drivers of oral squamous cell carcinoma (OSCC) tumor growth and metastasis. Therefore, the mechanisms underlying tumor cell and fibroblast crosstalk warrant further investigation. We discovered that ectopic interferon-stimulated gene 15 (ISG15) expression, which is a promising and novel oncoprotein biomarker elevated in a variety of cancers, enhanced OSCC growth and elevated collagen and α-smooth muscle actin (α-SMA) expression in ISG15-expressing tumors. Analysis of immunohistochemistry revealed high ISG15 expression in human oral tissues correlated with high expression of α-SMA and fibroblast activation protein (FAP). Fibroblast migration and recruitment by ISG15-expressing OSCC cells were confirmed by in vitro and in vivo experiments. Exogenous ISG15 induced fibroblast migration, morphological changes, and vimentin expression. Enrichment of glycolysis pathway genes, as well as increased glycolysis-related gene expression, glucose uptake, and lactate production were observed in ISG15-treated fibroblasts. Lactate release and fibroblast migration were blocked by a competitive inhibitor of glucose metabolism. Furthermore, the knockdown of integrin αL (ITGAL)/CD11a, a subunit of ISG15 receptor lymphocyte functional-associated antigen-1 (LFA-1), in immortalized fibroblasts diminished extracellular ISG15-mediated glycolysis and migration. Our findings suggest that ISG15 derived from OSCC cells interacts with fibroblasts through the LFA-1 receptor, leading to glycolytic reprogramming and promotion of fibroblast migration into the TME.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"6"},"PeriodicalIF":5.9,"publicationDate":"2025-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11897235/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143605866","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
CENPF (+) cancer cells promote malignant progression of early-stage TP53 mutant lung adenocarcinoma. CENPF(+)癌细胞会促进早期 TP53 突变肺腺癌的恶性发展。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2025-03-05 DOI: 10.1038/s41389-025-00546-5
Yanlu Xiong, Jie Lei, Miaomiao Wen, Yongfu Ma, Jinbo Zhao, Yahui Tian, Zitong Wan, Xiaoyan Li, Jianfei Zhu, Wenchen Wang, Xiaohong Ji, Ying Sun, Jie Yang, Jiao Zhang, Shaowei Xin, Yang Liu, Lintao Jia, Yong Han, Tao Jiang

The prevention and precise treatment of early-stage lung adenocarcinoma (LUAD) characterized by small nodules (stage IA) remains a significant challenge for clinicians, which is due largely to the limited understanding of the oncogenic mechanisms spanning from preneoplasia to invasive adenocarcinoma. Our study highlights the pivotal role of cancer cells exhibiting high expression of centromere protein F (CENPF), driven by TP53 mutations, which become increasingly prevalent during the transition from preneoplasia to invasive LUAD. Biologically, cancer cells (CENPF+) exhibited robust proliferative and stem-like capabilities, thereby propelling the malignant progression of early-stage LUAD. Clinically, autoantibodies against CENPF in the serum and elevated cancer cells (CENPF+) in tissue correlated positively with the progression of early-stage LUAD, especially those in stage IA. Our findings suggest that cancer cells (CENPF+) play a central role in orchestrating the malignant evolution of LUAD and hold potential as a novel biomarker for early-stage detection and management of the disease.

以小结节(IA期)为特征的早期肺腺癌(LUAD)的预防和精确治疗仍然是临床医生面临的一个重大挑战,这在很大程度上是由于对从瘤前病变到浸润性腺癌的致癌机制的了解有限。我们的研究强调了由TP53突变驱动的着丝粒蛋白F (CENPF)高表达的癌细胞的关键作用,这在从瘤前病变到侵袭性LUAD的转变过程中变得越来越普遍。在生物学上,癌细胞(CENPF+)表现出强大的增殖和干细胞样能力,从而推动早期LUAD的恶性进展。临床中,血清中抗CENPF自身抗体和组织中升高的癌细胞(CENPF+)与早期LUAD的进展呈正相关,尤其是IA期。我们的研究结果表明,癌细胞(CENPF+)在协调LUAD的恶性进化中起着核心作用,并具有作为早期检测和治疗该疾病的新型生物标志物的潜力。
{"title":"CENPF (+) cancer cells promote malignant progression of early-stage TP53 mutant lung adenocarcinoma.","authors":"Yanlu Xiong, Jie Lei, Miaomiao Wen, Yongfu Ma, Jinbo Zhao, Yahui Tian, Zitong Wan, Xiaoyan Li, Jianfei Zhu, Wenchen Wang, Xiaohong Ji, Ying Sun, Jie Yang, Jiao Zhang, Shaowei Xin, Yang Liu, Lintao Jia, Yong Han, Tao Jiang","doi":"10.1038/s41389-025-00546-5","DOIUrl":"10.1038/s41389-025-00546-5","url":null,"abstract":"<p><p>The prevention and precise treatment of early-stage lung adenocarcinoma (LUAD) characterized by small nodules (stage IA) remains a significant challenge for clinicians, which is due largely to the limited understanding of the oncogenic mechanisms spanning from preneoplasia to invasive adenocarcinoma. Our study highlights the pivotal role of cancer cells exhibiting high expression of centromere protein F (CENPF), driven by TP53 mutations, which become increasingly prevalent during the transition from preneoplasia to invasive LUAD. Biologically, cancer cells (CENPF+) exhibited robust proliferative and stem-like capabilities, thereby propelling the malignant progression of early-stage LUAD. Clinically, autoantibodies against CENPF in the serum and elevated cancer cells (CENPF+) in tissue correlated positively with the progression of early-stage LUAD, especially those in stage IA. Our findings suggest that cancer cells (CENPF+) play a central role in orchestrating the malignant evolution of LUAD and hold potential as a novel biomarker for early-stage detection and management of the disease.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"14 1","pages":"5"},"PeriodicalIF":5.9,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11882812/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143567034","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
期刊
Oncogenesis
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