首页 > 最新文献

Molecular Cancer Research最新文献

英文 中文
The Complex Regulation of Cytokinesis upon Abscission Checkpoint Activation. 脱落检查点激活时对细胞分裂的复杂调控。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-02 DOI: 10.1158/1541-7786.MCR-24-0365
Paulius Gibieža, Vilma Petrikaitė

Cytokinetic abscission is a crucial process that guides the separation of daughter cells at the end of each cell division. This process involves the cleavage of the intercellular bridge, which connects the newly formed daughter cells. Over the years, researchers have identified several cellular contributors and intracellular processes that influence the spatial and temporal distribution of the cytoskeleton during cytokinetic abscission. This review presents the most important scientific discoveries that allow activation of the abscission checkpoint, ensuring a smooth and successful separation of a single cell into two cells during cell division. Here, we describe different factors, such as abscission checkpoint, ICB tension, nuclear pore defects, DNA replication stress, chromosomal stability, and midbody proteins, which play a role in the regulation and correct timing of cytokinetic abscission. Furthermore, we explore the downsides associated with the dysregulation of abscission, including its negative impact on cells and the potential to induce tumor formation in humans. Finally, we propose a novel factor for improving cancer therapy and give future perspectives in this research field.

细胞分裂是每次细胞分裂结束时引导子细胞分离的关键过程。这一过程涉及连接新形成子细胞的细胞间桥的裂解。多年来,研究人员已经确定了细胞分裂过程中影响细胞骨架空间和时间分布的几个细胞贡献者和细胞内过程。本综述介绍了最重要的科学发现,这些发现使细胞脱落检查点得以激活,确保细胞分裂过程中单细胞顺利、成功地分离成两个细胞。在此,我们将介绍不同的因素,如脱落检查点、ICB张力、核孔缺陷、DNA复制应激、染色体稳定性和中体蛋白,它们在细胞运动性脱落的调控和正确时间安排方面发挥着作用。此外,我们还探讨了与脱落失调相关的弊端,包括对细胞的负面影响和诱发人类肿瘤形成的可能性。最后,我们提出了改善癌症治疗的新因素,并展望了这一研究领域的未来前景。
{"title":"The Complex Regulation of Cytokinesis upon Abscission Checkpoint Activation.","authors":"Paulius Gibieža, Vilma Petrikaitė","doi":"10.1158/1541-7786.MCR-24-0365","DOIUrl":"10.1158/1541-7786.MCR-24-0365","url":null,"abstract":"<p><p>Cytokinetic abscission is a crucial process that guides the separation of daughter cells at the end of each cell division. This process involves the cleavage of the intercellular bridge, which connects the newly formed daughter cells. Over the years, researchers have identified several cellular contributors and intracellular processes that influence the spatial and temporal distribution of the cytoskeleton during cytokinetic abscission. This review presents the most important scientific discoveries that allow activation of the abscission checkpoint, ensuring a smooth and successful separation of a single cell into two cells during cell division. Here, we describe different factors, such as abscission checkpoint, ICB tension, nuclear pore defects, DNA replication stress, chromosomal stability, and midbody proteins, which play a role in the regulation and correct timing of cytokinetic abscission. Furthermore, we explore the downsides associated with the dysregulation of abscission, including its negative impact on cells and the potential to induce tumor formation in humans. Finally, we propose a novel factor for improving cancer therapy and give future perspectives in this research field.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141971526","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
Ubiquitin Ligase TRIM22 Inhibits Ovarian Cancer Malignancy via TCF4 Degradation. 泛素连接酶TRIM22通过TCF4降解抑制卵巢癌恶变
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-02 DOI: 10.1158/1541-7786.MCR-23-0962
Tao Tao, Yongqi Zhang, Chunyan Guan, Shuxiang Wang, Xiaoli Liu, Min Wang

Ovarian cancer is one of the most common malignancies in women. Tripartite motif-containing protein 22 (TRIM22) plays an important role in the initiation and progression of malignant tumors. Similarly, the transcription factor 4 (TCF4) is an essential factor involved in the initiation and progression of many tumors. However, it is still unclear whether TRIM22 can affect TCF4 in ovarian cancer. Therefore, this study aims to investigate the mechanism related to TRIM22 and TCF4 in ovarian cancer. TRIM22 protein and mRNA levels were analyzed in samples from clinical and cell lines. The effects of TRIM22 knockdown and overexpression on cell proliferation, colony formation, migration, invasion, and related biomarkers were evaluated. In addition, the role of ubiquitination-mediated degradation of TCF4 was investigated by qRT-PCR and Western blotting. The association between TRIM22 and TCF4 was evaluated by Western blotting, coimmunoprecipitation, proliferation, colony formation, invasion, migration, and related biomarkers. The results showed that the expression of TRIM22 was minimal in ovarian cancer tissues. Furthermore, upregulation of TRIM22 significantly inhibited ovarian cancer cell proliferation, colony formation, migration, and invasion. In addition, TRIM22 was observed to regulate the degradation of TCF4 through the ubiquitination pathway. TCF4 can reverse the effects of TRIM22 on proliferation, colony formation, migration, and invasion in ovarian cancer cells. TRIM22-mediated ubiquitination of TCF4 at K48 is facilitated by the RING domain. Implications: In conclusion, ubiquitination of TCF4 protein in ovarian cancer is regulated by TRIM22, which has the potential to limit the proliferation, migration, and invasion of ovarian cancer.

卵巢癌(OC)是女性最常见的恶性肿瘤之一。含三方基序蛋白 22(TRIM22)在恶性肿瘤的发生和发展过程中起着重要作用。同样,转录因子 4(TCF4)也是参与许多肿瘤发生和发展的重要因子。然而,TRIM22是否会影响OC中的TCF4仍不清楚。因此,本研究旨在探讨TRIM22与TCF4在OC中的相关机制。本研究分析了临床样本和细胞系样本中的TRIM22蛋白和mRNA水平。评估了TRIM22敲除和过表达对细胞增殖、集落形成、迁移、侵袭和相关生物标志物的影响。此外,还通过qRT-PCR和Western印迹法研究了泛素化介导的TCF4降解的作用。通过Western印迹、共免疫沉淀、增殖、集落形成、侵袭、迁移及相关生物标记评估了TRIM22与TCF4之间的关联。结果显示,TRIM22在OC组织中的表达量极少。此外,上调 TRIM22 能显著抑制 OC 细胞的增殖、集落形成、迁移和侵袭。此外,还观察到TRIM22通过泛素化途径调控TCF4的降解。TCF4 可以逆转 TRIM22 对 OC 细胞增殖、集落形成、迁移和侵袭的影响。TRIM22介导的TCF4在K48处的泛素化是由RING结构域促进的。意义:总之,TCF4 蛋白在 OC 中的泛素化是由 TRIM22 调节的,它有可能限制 OC 的增殖、迁移和侵袭。
{"title":"Ubiquitin Ligase TRIM22 Inhibits Ovarian Cancer Malignancy via TCF4 Degradation.","authors":"Tao Tao, Yongqi Zhang, Chunyan Guan, Shuxiang Wang, Xiaoli Liu, Min Wang","doi":"10.1158/1541-7786.MCR-23-0962","DOIUrl":"10.1158/1541-7786.MCR-23-0962","url":null,"abstract":"<p><p>Ovarian cancer is one of the most common malignancies in women. Tripartite motif-containing protein 22 (TRIM22) plays an important role in the initiation and progression of malignant tumors. Similarly, the transcription factor 4 (TCF4) is an essential factor involved in the initiation and progression of many tumors. However, it is still unclear whether TRIM22 can affect TCF4 in ovarian cancer. Therefore, this study aims to investigate the mechanism related to TRIM22 and TCF4 in ovarian cancer. TRIM22 protein and mRNA levels were analyzed in samples from clinical and cell lines. The effects of TRIM22 knockdown and overexpression on cell proliferation, colony formation, migration, invasion, and related biomarkers were evaluated. In addition, the role of ubiquitination-mediated degradation of TCF4 was investigated by qRT-PCR and Western blotting. The association between TRIM22 and TCF4 was evaluated by Western blotting, coimmunoprecipitation, proliferation, colony formation, invasion, migration, and related biomarkers. The results showed that the expression of TRIM22 was minimal in ovarian cancer tissues. Furthermore, upregulation of TRIM22 significantly inhibited ovarian cancer cell proliferation, colony formation, migration, and invasion. In addition, TRIM22 was observed to regulate the degradation of TCF4 through the ubiquitination pathway. TCF4 can reverse the effects of TRIM22 on proliferation, colony formation, migration, and invasion in ovarian cancer cells. TRIM22-mediated ubiquitination of TCF4 at K48 is facilitated by the RING domain. Implications: In conclusion, ubiquitination of TCF4 protein in ovarian cancer is regulated by TRIM22, which has the potential to limit the proliferation, migration, and invasion of ovarian cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141262339","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
RNA-Binding Protein Lin28B Promotes Chronic Myeloid Leukemia Blast Crisis by Transcriptionally Upregulating miR-181d. RNA结合蛋白Lin28B通过转录上调miR-181d促进慢性髓性白血病爆发危象。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-02 DOI: 10.1158/1541-7786.MCR-23-0928
Minran Zhou, Xiaolin Yin, Lu Zhang, Zelong Cui, Xinwen Jiang, Qingli Ji, Sai Ma, Chunyan Chen

The blast crisis (BC) of chronic myeloid leukemia (CML) has poor efficacy against existing treatments and extremely short survival. However, the molecular mechanism of CML-chronic phase (CP) transformation to CML-BC is not yet fully understood. Here, we show that Lin28B, an RNA-binding protein, acted as an activator enhancing the transformation to CML-BC by mediating excessive cell proliferation. The level of Lin28B expression was apparently elevated in patients with CML-BC compared with newly diagnosed patients with CML-CP. The overexpression of Lin28B promoted the proliferation of leukemia cells. Mechanistically, we identified Lin28B as a DNA-binding protein by binding to the promoter region of miR-181d and upregulating its expression, which inhibited the expression of programmed cell death 4 (PDCD4) by binding to the PDCD4 3'UTR region, thereby enhancing the proliferation of CML cells. Overall, the "Lin28B-miR-181d-PDCD4" regulatory axis promoted CML blast crisis. Implications: Our findings highlight the oncogenic role of Lin28B in CML blast crisis, acting as a DNA-binding protein that transcriptionally upregulates miR-181d expression.

慢性髓性白血病(CML)的出血危象(BC)对现有疗法疗效不佳,存活期极短。然而,CML-慢性期(CP)向CML-BC转化的分子机制尚未完全明了。在这里,我们发现 RNA 结合蛋白 Lin28B 可通过介导细胞过度增殖,激活向 CML-BC 的转化。与新诊断的 CML-CP 患者相比,Lin28B 在 CML-BC 患者中的表达水平明显升高。Lin28B的过表达促进了白血病细胞的增殖。从机理上讲,我们发现Lin28B是一种DNA结合蛋白,它与miR-181d的启动子区域结合并上调其表达,而miR-181d通过与PDCD4 3'UTR区域结合抑制了程序性细胞死亡4(PDCD4)的表达,从而促进了CML细胞的增殖。总之,"Lin28B-miR-181d-PDCD4 "调控轴促进了CML爆破危机。意义:我们的研究结果突显了Lin28B在CML爆裂危象中的致癌作用,它是一种DNA结合蛋白,可转录上调miR-181d的表达。
{"title":"RNA-Binding Protein Lin28B Promotes Chronic Myeloid Leukemia Blast Crisis by Transcriptionally Upregulating miR-181d.","authors":"Minran Zhou, Xiaolin Yin, Lu Zhang, Zelong Cui, Xinwen Jiang, Qingli Ji, Sai Ma, Chunyan Chen","doi":"10.1158/1541-7786.MCR-23-0928","DOIUrl":"10.1158/1541-7786.MCR-23-0928","url":null,"abstract":"<p><p>The blast crisis (BC) of chronic myeloid leukemia (CML) has poor efficacy against existing treatments and extremely short survival. However, the molecular mechanism of CML-chronic phase (CP) transformation to CML-BC is not yet fully understood. Here, we show that Lin28B, an RNA-binding protein, acted as an activator enhancing the transformation to CML-BC by mediating excessive cell proliferation. The level of Lin28B expression was apparently elevated in patients with CML-BC compared with newly diagnosed patients with CML-CP. The overexpression of Lin28B promoted the proliferation of leukemia cells. Mechanistically, we identified Lin28B as a DNA-binding protein by binding to the promoter region of miR-181d and upregulating its expression, which inhibited the expression of programmed cell death 4 (PDCD4) by binding to the PDCD4 3'UTR region, thereby enhancing the proliferation of CML cells. Overall, the \"Lin28B-miR-181d-PDCD4\" regulatory axis promoted CML blast crisis. Implications: Our findings highlight the oncogenic role of Lin28B in CML blast crisis, acting as a DNA-binding protein that transcriptionally upregulates miR-181d expression.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141284297","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
Lactate Induces Tumor Progression via LAR Motif-Dependent Yin-Yang 1 Degradation. 乳酸通过依赖于 LAR motif 的阴阳 1 降解诱导肿瘤进展。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-02 DOI: 10.1158/1541-7786.MCR-23-0583
Shujuan Du, Xiaoting Chen, Xiao Han, Yuyan Wang, Dan Yu, Ying Li, Caixia Zhu, Yin Tong, Shujun Gao, Junwen Wang, Fang Wei, Qiliang Cai

The metabolic reprogramming of aerobic glycolysis contributes to tumorigenesis. High plasma lactate is a critical regulator in the development of many human malignancies; however, the underlying molecular mechanisms of cancer progression in response to lactate (LA) remain elusive. Here, we show that the reduction of Yin-Yang 1 (YY1) expression correlated with high LA commonly occurs in various cancer cell types, including B-lymphoma and cervical cancer. Mechanistically, LA induces YY1 nuclear export and degradation via HSP70-mediated autophagy adjacent to mitochondria in a histidine (His)-rich LA-responsive (LAR) motif-dependent manner. The mutation of the LAR motif blocks LA-mediated YY1 cytoplasmic accumulation and in turn enhances cell apoptosis. Furthermore, low expression of YY1 promotes colony formation, invasion, angiogenesis, and growth of cancer cells in response to LA in vitro and in vivo using a murine xenograft model. Taken together, our findings reveal a key LAR element and may serve as therapeutic target for intervening cancer progression. Implications: We have shown that lactate can induce YY1 degradation via its His-rich LAR motif and low expression of YY1 promotes cancer cell progression in response to lactate, leading to better prediction of YY1 targeting therapy.

有氧糖酵解的代谢重编程有助于肿瘤发生。高血浆乳酸是许多人类恶性肿瘤发展过程中的一个关键调节因子;然而,癌症进展对乳酸(LA)反应的潜在分子机制仍然难以捉摸。在这里,我们发现阴阳1(YY1)表达的减少与高LA相关,这通常发生在各种癌症细胞类型中,包括B淋巴瘤和宫颈癌。从机理上讲,LA 通过 HSP70 介导的线粒体附近的自噬,以富含组氨酸的 LAR(LA 响应)基序依赖的方式诱导 YY1 核输出和降解。LAR 基因突变会阻止 LA 介导的 YY1 胞质积累,进而促进细胞凋亡。此外,YY1 的低表达在体外和体内小鼠异种移植模型中促进了癌细胞对 LA 的集落形成、侵袭、血管生成和生长。综上所述,我们的研究结果揭示了一个关键的乳酸反应元件,它可作为干预癌症进展的治疗靶点。意义:我们的研究表明,乳酸可通过富含组氨酸的LAR基序诱导YY1降解,而YY1的低表达可促进癌细胞对乳酸的反应,从而更好地预测YY1靶向疗法。
{"title":"Lactate Induces Tumor Progression via LAR Motif-Dependent Yin-Yang 1 Degradation.","authors":"Shujuan Du, Xiaoting Chen, Xiao Han, Yuyan Wang, Dan Yu, Ying Li, Caixia Zhu, Yin Tong, Shujun Gao, Junwen Wang, Fang Wei, Qiliang Cai","doi":"10.1158/1541-7786.MCR-23-0583","DOIUrl":"10.1158/1541-7786.MCR-23-0583","url":null,"abstract":"<p><p>The metabolic reprogramming of aerobic glycolysis contributes to tumorigenesis. High plasma lactate is a critical regulator in the development of many human malignancies; however, the underlying molecular mechanisms of cancer progression in response to lactate (LA) remain elusive. Here, we show that the reduction of Yin-Yang 1 (YY1) expression correlated with high LA commonly occurs in various cancer cell types, including B-lymphoma and cervical cancer. Mechanistically, LA induces YY1 nuclear export and degradation via HSP70-mediated autophagy adjacent to mitochondria in a histidine (His)-rich LA-responsive (LAR) motif-dependent manner. The mutation of the LAR motif blocks LA-mediated YY1 cytoplasmic accumulation and in turn enhances cell apoptosis. Furthermore, low expression of YY1 promotes colony formation, invasion, angiogenesis, and growth of cancer cells in response to LA in vitro and in vivo using a murine xenograft model. Taken together, our findings reveal a key LAR element and may serve as therapeutic target for intervening cancer progression. Implications: We have shown that lactate can induce YY1 degradation via its His-rich LAR motif and low expression of YY1 promotes cancer cell progression in response to lactate, leading to better prediction of YY1 targeting therapy.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141420015","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
Characterization of Wnt Signaling Pathway Aberrations in Metastatic Prostate Cancer. 转移性前列腺癌中 Wnt 信号通路畸变的特征。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-02 DOI: 10.1158/1541-7786.MCR-24-0395
Sharon H Choi, Elizabeth Pan, Andrew Elliott, Himisha Beltran, Justine Panian, Christina Jamieson, Aditya Bagrodia, Brent Rose, Daniel Herchenhorn, Elisabeth Heath, Chadi Nabhan, Emmanuel S Antonarakis, Rana R McKay

Wnt (wingless-type) signaling pathway (WSP) alterations have been identified in patients with prostate cancer and are implicated in disease progression and hormonal resistance. In this study, we utilized a multi-institutional dataset to characterize molecular alterations in the canonical and noncanonical WSPs in prostate cancer. Patients with prostate cancer who underwent tissue-based genomic sequencing were investigated. Tumors with somatic activating mutations in CTNNB1 or RSPO2 or inactivating mutations in either APC or RNF43 were characterized as having aberrant canonical Wnt signaling (WSP-activated). Overall survival analyses were restricted to microsatellite-stable (MSS) tumors lacking RNF43 G659fs* mutations. We also investigated noncanonical WSP by evaluation of ROR1, ROR2, and WNT5 in WSP-activated versus WSP wild-type (WSP-WT) tumors. Of 4,138 prostate cancer samples, 3,684 were MSS. Among MSS tumors, 42.4% were from metastatic sites, of which 19.1% were WSP activated, and 57.6% were from the prostate, of which 10.1% were WSP activated. WSP-activated tumors were more prevalent in metastatic sites than in primary prostate cancer. WSP-activated prostate cancer exhibited more SPOP mutations and higher expression of canonical WSP activators than WSP-WT tumors. ROR1 gene expression was elevated in WSP-activated tumors from both primary and metastatic sites. M2 macrophages predominated the tumor microenvironment in WSP-activated tumors. There was no significant difference in overall survival between patients with WSP-activated and WSP-WT prostate cancer. WSP-activated prostate cancer demonstrated a more immunosuppressed tumor microenvironment and a pronounced upregulation of ROR1 gene expression, underscoring its potential involvement in the crosstalk between canonical and noncanonical WSPs. Implications: Our findings may provide a rationale for developing novel therapeutic strategies targeting Wnt-activated prostate cancer.

在前列腺癌(PCa)患者中发现了 Wnt 信号通路(WSP)的改变,这些改变与疾病进展和激素抗性有关。我们利用一个多机构数据集来描述 PCa 中规范和非规范 WSP 的分子改变。我们对接受组织基因组测序的 PCa 患者进行了调查。CTNNB1 或 RSPO2 发生体细胞活化突变,或 APC 或 RNF43 发生失活突变的肿瘤被定性为具有异常规范 Wnt 信号转导(WSP 激活)。总生存期(OS)分析仅限于缺乏 RNF43 G659fs* 突变的微卫星稳定(MSS)肿瘤。我们还通过评估WSP激活与WSP野生型(WSP-WT)肿瘤中的ROR1、ROR2和WNT5,研究了非经典WSP。在 4,138 份 PCa 样本中,3,684 份为 MSS。在MSS肿瘤中,42.4%来自转移部位,其中19.1%为WSP激活肿瘤;57.6%来自前列腺,其中10.1%为WSP激活肿瘤。与原发性PCa相比,WSP激活的肿瘤在转移部位更为常见。与WSP-WT肿瘤相比,WSP激活型PCa表现出更多的SPOP突变和更高的典型WSP激活剂表达。在原发和转移部位的 WSP 激活肿瘤中,ROR1 基因表达均升高。在WSP激活的肿瘤中,M2巨噬细胞在肿瘤微环境中占主导地位。WSP激活型和WSP-WT型PCa患者的OS无明显差异。WSP激活的PCa表现出更强的免疫抑制肿瘤微环境和明显的ROR1基因表达上调,这强调了它可能参与了典型和非典型Wnt信号通路之间的串扰。影响:我们的研究结果可为开发针对 Wnt 激活型 PCa 的新型治疗策略提供依据。
{"title":"Characterization of Wnt Signaling Pathway Aberrations in Metastatic Prostate Cancer.","authors":"Sharon H Choi, Elizabeth Pan, Andrew Elliott, Himisha Beltran, Justine Panian, Christina Jamieson, Aditya Bagrodia, Brent Rose, Daniel Herchenhorn, Elisabeth Heath, Chadi Nabhan, Emmanuel S Antonarakis, Rana R McKay","doi":"10.1158/1541-7786.MCR-24-0395","DOIUrl":"10.1158/1541-7786.MCR-24-0395","url":null,"abstract":"<p><p>Wnt (wingless-type) signaling pathway (WSP) alterations have been identified in patients with prostate cancer and are implicated in disease progression and hormonal resistance. In this study, we utilized a multi-institutional dataset to characterize molecular alterations in the canonical and noncanonical WSPs in prostate cancer. Patients with prostate cancer who underwent tissue-based genomic sequencing were investigated. Tumors with somatic activating mutations in CTNNB1 or RSPO2 or inactivating mutations in either APC or RNF43 were characterized as having aberrant canonical Wnt signaling (WSP-activated). Overall survival analyses were restricted to microsatellite-stable (MSS) tumors lacking RNF43 G659fs* mutations. We also investigated noncanonical WSP by evaluation of ROR1, ROR2, and WNT5 in WSP-activated versus WSP wild-type (WSP-WT) tumors. Of 4,138 prostate cancer samples, 3,684 were MSS. Among MSS tumors, 42.4% were from metastatic sites, of which 19.1% were WSP activated, and 57.6% were from the prostate, of which 10.1% were WSP activated. WSP-activated tumors were more prevalent in metastatic sites than in primary prostate cancer. WSP-activated prostate cancer exhibited more SPOP mutations and higher expression of canonical WSP activators than WSP-WT tumors. ROR1 gene expression was elevated in WSP-activated tumors from both primary and metastatic sites. M2 macrophages predominated the tumor microenvironment in WSP-activated tumors. There was no significant difference in overall survival between patients with WSP-activated and WSP-WT prostate cancer. WSP-activated prostate cancer demonstrated a more immunosuppressed tumor microenvironment and a pronounced upregulation of ROR1 gene expression, underscoring its potential involvement in the crosstalk between canonical and noncanonical WSPs. Implications: Our findings may provide a rationale for developing novel therapeutic strategies targeting Wnt-activated prostate cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141443118","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
NAPRT Silencing in FH-Deficient Renal Cell Carcinoma Confers Therapeutic Vulnerabilities via NAD+ Depletion. FH缺陷肾细胞癌中的NAPRT沉默通过NAD+耗竭产生治疗脆弱性。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-02 DOI: 10.1158/1541-7786.MCR-23-1003
Katelyn J Noronha, Karlie N Lucas, Sateja Paradkar, Joseph Edmonds, Sam Friedman, Matthew A Murray, Samantha Liu, Dipti P Sajed, Chana Sachs, Josh Spurrier, Mitch Raponi, Jiayu Liang, Hao Zeng, Ranjini K Sundaram, Brian Shuch, Juan C Vasquez, Ranjit S Bindra

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by loss of function mutations in fumarate hydratase (FH) and results in an aggressive subtype of renal cell carcinoma with limited treatment options. Loss of FH leads to accumulation of fumarate, an oncometabolite that disrupts multiple cellular processes and drives tumor progression. High levels of fumarate inhibit alpha ketoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) enzymes, and can lead to global DNA hypermethylation. Here, we report patterns of hypermethylation in FH-mutant cell lines and tumor samples are associated with the silencing of nicotinate phosphoribosyl transferase (NAPRT), a rate-limiting enzyme in the Preiss-Handler pathway of NAD+ biosynthesis, in a subset of HLRCC cases. NAPRT is hypermethylated at a CpG island in the promoter in cell line models and patient samples, resulting in loss of NAPRT expression. We find that FH-deficient RCC models with loss of NAPRT expression, as well as other oncometabolite-producing cancer models that silence NAPRT, are extremely sensitive to nicotinamide phosphoribosyl transferase inhibitors (NAMPTi). NAPRT silencing was also associated with synergistic tumor cell killing with PARP inhibitors and NAMPTis, which was associated with effects on PAR-mediated DNA repair. Overall, our findings indicate that NAPRT silencing can be targeted in oncometabolite-producing cancers and elucidates how oncometabolite-associated hypermethylation can impact diverse cellular processes and lead to therapeutically relevant vulnerabilities in cancer cells. Implications: NAPRT is a novel biomarker for targeting NAD+ metabolism in FH-deficient HLRCCs with NAMPTis alone and targeting DNA repair processes with the combination of NAMPTis and PARP inhibitors.

遗传性乳糜尿和肾细胞癌(HLRCC)是由富马酸氢化酶(FH)功能缺失突变引起的,是一种侵袭性肾细胞癌亚型,治疗方案有限。富马酸氢化酶的缺失会导致富马酸盐的积累,而富马酸盐是一种副代谢产物,会破坏多种细胞过程并推动肿瘤进展。高浓度的富马酸会抑制α-酮戊二酸依赖性二氧酶,包括十-十一转位(TET)酶,并可导致全局性DNA高甲基化。在这里,我们报告了 FH 突变细胞系和肿瘤样本中的高甲基化模式与烟酸磷酸核糖转移酶(NAPRT)的沉默有关,NAPRT 是 HLRCC 亚群病例中 NAD+ 生物合成的 Preiss-Handler 途径中的一个限速酶。在细胞系模型和患者样本中,NAPRT 在启动子的一个 CpG 岛处发生了高甲基化,导致 NAPRT 表达缺失。我们发现,NAPRT表达缺失的FH缺陷RCC模型以及其他沉默了NAPRT的产生肿瘤甲胎蛋白的癌症模型对烟酰胺磷酸核糖转移酶抑制剂(NAMPTis)极为敏感。NAPRT 沉默还与多(ADP)核糖聚合酶抑制剂(PARPis)和 NAMPTis 协同杀伤肿瘤细胞有关,这与 PAR 介导的 DNA 修复效应有关。总之,我们的研究结果表明,NAPRT-沉默可作为产生肿瘤代谢物的癌症的靶点,并阐明了肿瘤代谢物相关的高甲基化如何影响多种细胞过程,并导致癌细胞中与治疗相关的脆弱性。意义:NAPRT 是一种新的生物标志物,可用于单独使用 NAMPTis 靶向 FH 缺失的 HLRCC 中的 NAD+ 代谢,以及结合使用 NAMPTis 和 PARPis 靶向 DNA 修复过程。
{"title":"NAPRT Silencing in FH-Deficient Renal Cell Carcinoma Confers Therapeutic Vulnerabilities via NAD+ Depletion.","authors":"Katelyn J Noronha, Karlie N Lucas, Sateja Paradkar, Joseph Edmonds, Sam Friedman, Matthew A Murray, Samantha Liu, Dipti P Sajed, Chana Sachs, Josh Spurrier, Mitch Raponi, Jiayu Liang, Hao Zeng, Ranjini K Sundaram, Brian Shuch, Juan C Vasquez, Ranjit S Bindra","doi":"10.1158/1541-7786.MCR-23-1003","DOIUrl":"10.1158/1541-7786.MCR-23-1003","url":null,"abstract":"<p><p>Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by loss of function mutations in fumarate hydratase (FH) and results in an aggressive subtype of renal cell carcinoma with limited treatment options. Loss of FH leads to accumulation of fumarate, an oncometabolite that disrupts multiple cellular processes and drives tumor progression. High levels of fumarate inhibit alpha ketoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) enzymes, and can lead to global DNA hypermethylation. Here, we report patterns of hypermethylation in FH-mutant cell lines and tumor samples are associated with the silencing of nicotinate phosphoribosyl transferase (NAPRT), a rate-limiting enzyme in the Preiss-Handler pathway of NAD+ biosynthesis, in a subset of HLRCC cases. NAPRT is hypermethylated at a CpG island in the promoter in cell line models and patient samples, resulting in loss of NAPRT expression. We find that FH-deficient RCC models with loss of NAPRT expression, as well as other oncometabolite-producing cancer models that silence NAPRT, are extremely sensitive to nicotinamide phosphoribosyl transferase inhibitors (NAMPTi). NAPRT silencing was also associated with synergistic tumor cell killing with PARP inhibitors and NAMPTis, which was associated with effects on PAR-mediated DNA repair. Overall, our findings indicate that NAPRT silencing can be targeted in oncometabolite-producing cancers and elucidates how oncometabolite-associated hypermethylation can impact diverse cellular processes and lead to therapeutically relevant vulnerabilities in cancer cells. Implications: NAPRT is a novel biomarker for targeting NAD+ metabolism in FH-deficient HLRCCs with NAMPTis alone and targeting DNA repair processes with the combination of NAMPTis and PARP inhibitors.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11445649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469683","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
Gram-negative microflora dysbiosis facilitates tumor progression and immune evasion by activating CCL3/CCL5-CCR1-MAPK-PD-L1 pathway in esophageal squamous cell carcinoma. 革兰氏阴性微生物菌群失调通过激活食管鳞状细胞癌中的 CCL3/CCL5-CCR1-MAPK-PD-L1 通路促进肿瘤进展和免疫逃避
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1541-7786.MCR-24-0451
Huiqin Yang, Jiahao Cai, Xiao Long Huang, Cheng Zhan, Chunlai Lu, Jie Gu, Teng Ma, Hongyu Zhang, Tao Cheng, Fengkai Xu, Di Ge

Gram-negative micro-flora dysbiosis occurs in multiple digestive tumors and is found to be the dominant micro-flora in esophageal squamous cell carcinoma (ESCC) micro-environment. The continuous stimulation of G- bacterium metabolites may cause tumorigenesis and reshape the micro-immune environment in ESCC. However, the mechanism of G- bacilli causing immune evasion in ESCC remains underexplored. We identified CC Chemokine receptor 1 (CCR1) as a tumor-indicating gene in ESCC. Interestingly, expression levels of CCR1 and PD-L1 were mutually up regulated after G- bacilli metabolites lipopolysaccharide (LPS) stimulation. Firstly, we found CCR1 high expression level to be associated with poor overall survival in ESCC. Importantly, we found that high level expression of CCR1 up-regulated PD-L1 expression by activating MAPK phosphorylation in ESCC and induced tumor malignant behavior. Finally, we found that T cells exhaustion and cytotoxicity suppression were associated with CCR1 expression in ESCC, which were decreased after CCR1 inhibiting. Our work identifies CCR1 as a potential immune check point regulator of PD-L1 and may cause T cell exhaustion and cytotoxicity suppression in ESCC micro-environment and highlights the potential value of CCR1 as therapeutic target of immunotherapy. Implications: The esophageal microbial environment and its metabolites significantly affect the outcome of immunotherapy for ESCC.

革兰氏阴性微生物菌群失调发生在多种消化系统肿瘤中,并被发现是食管鳞状细胞癌(ESCC)微环境中的主要微生物菌群。G- 菌代谢产物的持续刺激可能会导致肿瘤发生并重塑 ESCC 的微免疫环境。然而,G-杆菌导致 ESCC 免疫逃避的机制仍未得到充分探索。我们发现CC趋化因子受体1(CCR1)是ESCC的肿瘤提示基因。有趣的是,G-杆菌代谢物脂多糖(LPS)刺激后,CCR1和PD-L1的表达水平相互上调。首先,我们发现CCR1的高表达水平与ESCC的总生存率低有关。重要的是,我们发现 CCR1 的高水平表达通过激活 ESCC 中的 MAPK 磷酸化而上调 PD-L1 的表达,并诱导肿瘤的恶性行为。最后,我们发现 ESCC 中 T 细胞衰竭和细胞毒性抑制与 CCR1 的表达有关,而抑制 CCR1 后,T 细胞衰竭和细胞毒性抑制均会降低。我们的研究发现 CCR1 是 PD-L1 的潜在免疫检查点调节因子,可能导致 ESCC 微环境中的 T 细胞衰竭和细胞毒性抑制,并强调了 CCR1 作为免疫疗法治疗靶点的潜在价值。影响:食管微生物环境及其代谢产物对 ESCC 的免疫治疗结果有重要影响。
{"title":"Gram-negative microflora dysbiosis facilitates tumor progression and immune evasion by activating CCL3/CCL5-CCR1-MAPK-PD-L1 pathway in esophageal squamous cell carcinoma.","authors":"Huiqin Yang, Jiahao Cai, Xiao Long Huang, Cheng Zhan, Chunlai Lu, Jie Gu, Teng Ma, Hongyu Zhang, Tao Cheng, Fengkai Xu, Di Ge","doi":"10.1158/1541-7786.MCR-24-0451","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-24-0451","url":null,"abstract":"<p><p>Gram-negative micro-flora dysbiosis occurs in multiple digestive tumors and is found to be the dominant micro-flora in esophageal squamous cell carcinoma (ESCC) micro-environment. The continuous stimulation of G- bacterium metabolites may cause tumorigenesis and reshape the micro-immune environment in ESCC. However, the mechanism of G- bacilli causing immune evasion in ESCC remains underexplored. We identified CC Chemokine receptor 1 (CCR1) as a tumor-indicating gene in ESCC. Interestingly, expression levels of CCR1 and PD-L1 were mutually up regulated after G- bacilli metabolites lipopolysaccharide (LPS) stimulation. Firstly, we found CCR1 high expression level to be associated with poor overall survival in ESCC. Importantly, we found that high level expression of CCR1 up-regulated PD-L1 expression by activating MAPK phosphorylation in ESCC and induced tumor malignant behavior. Finally, we found that T cells exhaustion and cytotoxicity suppression were associated with CCR1 expression in ESCC, which were decreased after CCR1 inhibiting. Our work identifies CCR1 as a potential immune check point regulator of PD-L1 and may cause T cell exhaustion and cytotoxicity suppression in ESCC micro-environment and highlights the potential value of CCR1 as therapeutic target of immunotherapy. Implications: The esophageal microbial environment and its metabolites significantly affect the outcome of immunotherapy for ESCC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142350768","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
Defining splicing factor requirements for androgen receptor variant synthesis in advanced prostate cancer. 确定晚期前列腺癌中雄激素受体变体合成的剪接因子要求。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-30 DOI: 10.1158/1541-7786.MCR-23-0958
Laura Walker, Ruaridh Duncan, Beth Adamson, Hannah Kendall, Nicholas Brittain, Sara Luzzi, Dominic Jones, Lewis Chaytor, Samantha Peel, Claire Crafter, Daniel J O'Neill, Luke Gaughan

Resistance to androgen receptor (AR)-targeted therapies represent a major challenge in prostate cancer (PC). A key mechanism of treatment resistance in patients who progress to castrate-resistant PC (CRPC) is the generation of alternatively spliced androgen receptor variants (AR-Vs). Unlike full-length AR (FL-AR) isoforms, AR-Vs are constitutively active and refractory to current receptor-targeting agents hence drive tumour progression. Identifying regulators of AR-V synthesis may therefore provide new therapeutic opportunities in combination with conventional AR-targeting agents. Our understanding of AR transcript splicing, and the factors that control the synthesis of AR-Vs, remains limited. While candidate-based approaches have identified a small number of AR-V splicing regulators, an unbiased analysis of splicing factors important for AR-V generation is required to fill an important knowledge gap and furnish the field with novel and tractable targets for PC treatment. To that end, we conducted a bespoke CRISPR screen to profile splicing factor requirements for AR-V synthesis. MFAP1 and CWC22 were shown to be required for the generation of AR-V mRNA transcripts and their depletion resulted in reduced AR-V protein abundance and cell proliferation in several CRPC models. Global transcriptomic analysis of MFAP1-depleted cells revealed both AR-dependent and -independent transcriptional impact, including genes associated with DDR. As such, MFAP1 downregulation sensitised PC cells to ionising radiation suggesting therapeutically targeting AR-V splicing could provide novel cellular vulnerabilities which can be exploited in CRPC. Implications: We have utilised a CRISPR screening approach to identify key regulators of pathogenic AR splicing in prostate cancer.

雄激素受体(AR)靶向疗法的耐药性是前列腺癌(PC)的一大挑战。进展为阉割耐药 PC(CRPC)的患者产生耐药性的一个关键机制是产生了交替剪接的雄激素受体变体(AR-Vs)。与全长AR(FL-AR)异构体不同,AR-Vs具有组成性活性,对目前的受体靶向药物具有耐药性,因此会推动肿瘤进展。因此,确定AR-V合成的调控因子可为结合传统的AR靶向药物提供新的治疗机会。我们对 AR 转录本剪接以及控制 AR-Vs 合成的因素的了解仍然有限。虽然基于候选的方法已经确定了少量的 AR-V 剪接调节因子,但还需要对对 AR-V 生成很重要的剪接因子进行无偏见的分析,以填补这一重要的知识空白,并为 PC 治疗领域提供新颖、可行的靶点。为此,我们进行了一次定制的 CRISPR 筛选,以确定 AR-V 合成所需的剪接因子。结果表明,MFAP1和CWC22是AR-V mRNA转录本生成所必需的,而且在几种CRPC模型中,它们的缺失会导致AR-V蛋白丰度降低和细胞增殖减少。对去除了 MFAP1 的细胞进行的全局转录组学分析显示了依赖 AR 和不依赖 AR 的转录影响,包括与 DDR 相关的基因。因此,MFAP1的下调使PC细胞对电离辐射敏感,这表明针对AR-V剪接的治疗可提供新的细胞脆弱性,可在CRPC中加以利用。意义:我们利用 CRISPR 筛选方法确定了前列腺癌中致病性 AR 剪接的关键调控因子。
{"title":"Defining splicing factor requirements for androgen receptor variant synthesis in advanced prostate cancer.","authors":"Laura Walker, Ruaridh Duncan, Beth Adamson, Hannah Kendall, Nicholas Brittain, Sara Luzzi, Dominic Jones, Lewis Chaytor, Samantha Peel, Claire Crafter, Daniel J O'Neill, Luke Gaughan","doi":"10.1158/1541-7786.MCR-23-0958","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-23-0958","url":null,"abstract":"<p><p>Resistance to androgen receptor (AR)-targeted therapies represent a major challenge in prostate cancer (PC). A key mechanism of treatment resistance in patients who progress to castrate-resistant PC (CRPC) is the generation of alternatively spliced androgen receptor variants (AR-Vs). Unlike full-length AR (FL-AR) isoforms, AR-Vs are constitutively active and refractory to current receptor-targeting agents hence drive tumour progression. Identifying regulators of AR-V synthesis may therefore provide new therapeutic opportunities in combination with conventional AR-targeting agents. Our understanding of AR transcript splicing, and the factors that control the synthesis of AR-Vs, remains limited. While candidate-based approaches have identified a small number of AR-V splicing regulators, an unbiased analysis of splicing factors important for AR-V generation is required to fill an important knowledge gap and furnish the field with novel and tractable targets for PC treatment. To that end, we conducted a bespoke CRISPR screen to profile splicing factor requirements for AR-V synthesis. MFAP1 and CWC22 were shown to be required for the generation of AR-V mRNA transcripts and their depletion resulted in reduced AR-V protein abundance and cell proliferation in several CRPC models. Global transcriptomic analysis of MFAP1-depleted cells revealed both AR-dependent and -independent transcriptional impact, including genes associated with DDR. As such, MFAP1 downregulation sensitised PC cells to ionising radiation suggesting therapeutically targeting AR-V splicing could provide novel cellular vulnerabilities which can be exploited in CRPC. Implications: We have utilised a CRISPR screening approach to identify key regulators of pathogenic AR splicing in prostate cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142350767","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
c-Myc, AKT, Hsc70, and the T-Box Transcription Factor TBX3 Form an Important Oncogenic Signaling Axis in Breast Cancer c-Myc、AKT、Hsc70 和 T-Box 转录因子 TBX3 在乳腺癌中形成了一个重要的致癌信号轴
IF 5.2 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-12 DOI: 10.1158/1541-7786.mcr-23-1031
Stephanie M. Ncube, ArulJothi Nagarajan, Dirk Lang, Musalula Sinkala, Carly A. Burmeister, Karabo Serala, Jonathan Blackburn, Sharon Prince
Breast cancer is the second leading cause of death in women globally, and it remains a health burden due to poor therapy response, cancer cell drug resistance, and the debilitating side effects associated with most therapies. One approach to addressing the need to improve breast cancer therapies has been to elucidate the mechanism(s) underpinning this disease to identify key drivers that can be targeted in molecular therapies. The T-box transcription factor, TBX3, is upregulated in breast cancer, in which it contributes to important oncogenic processes, and it has been validated as a potential therapeutic target. Here, we investigated the molecular mechanisms that upregulate TBX3 in breast cancer, and we show that it involves transcriptional activation by c-Myc, post-translational modification by AKT1 and AKT3, and interaction with the molecular chaperone Hsc70. Together, the results from this study provide evidence that c-Myc, AKT, Hsc70, and TBX3 form part of an important oncogenic pathway in breast cancer and thus reveal versatile ways of interfering with the oncogenic activity of TBX3 for the treatment of this neoplasm. Implications: Targeting the c-Myc/AKT/TBX3/Hsc70 signaling axis may be an effective treatment strategy for TBX3-driven breast cancer.
乳腺癌是全球女性的第二大死因,由于治疗反应不佳、癌细胞耐药性以及大多数疗法带来的副作用,乳腺癌仍然是一种健康负担。为满足改善乳腺癌疗法的需要,一种方法是阐明这种疾病的发病机制,以确定分子疗法中可针对的关键驱动因素。T-盒转录因子TBX3在乳腺癌中上调,它在乳腺癌的重要致癌过程中起着重要作用,而且它已被证实是一个潜在的治疗靶点。在此,我们研究了乳腺癌中 TBX3 上调的分子机制,结果表明它涉及 c-Myc 的转录激活、AKT1 和 AKT3 的翻译后修饰以及与分子伴侣 Hsc70 的相互作用。总之,这项研究的结果提供了证据,证明 c-Myc、AKT、Hsc70 和 TBX3 构成了乳腺癌重要致癌途径的一部分,从而揭示了干扰 TBX3 致癌活性以治疗这种肿瘤的多种方法。影响:靶向c-Myc/AKT/TBX3/Hsc70信号轴可能是治疗TBX3驱动的乳腺癌的有效策略。
{"title":"c-Myc, AKT, Hsc70, and the T-Box Transcription Factor TBX3 Form an Important Oncogenic Signaling Axis in Breast Cancer","authors":"Stephanie M. Ncube, ArulJothi Nagarajan, Dirk Lang, Musalula Sinkala, Carly A. Burmeister, Karabo Serala, Jonathan Blackburn, Sharon Prince","doi":"10.1158/1541-7786.mcr-23-1031","DOIUrl":"https://doi.org/10.1158/1541-7786.mcr-23-1031","url":null,"abstract":"Breast cancer is the second leading cause of death in women globally, and it remains a health burden due to poor therapy response, cancer cell drug resistance, and the debilitating side effects associated with most therapies. One approach to addressing the need to improve breast cancer therapies has been to elucidate the mechanism(s) underpinning this disease to identify key drivers that can be targeted in molecular therapies. The T-box transcription factor, TBX3, is upregulated in breast cancer, in which it contributes to important oncogenic processes, and it has been validated as a potential therapeutic target. Here, we investigated the molecular mechanisms that upregulate TBX3 in breast cancer, and we show that it involves transcriptional activation by c-Myc, post-translational modification by AKT1 and AKT3, and interaction with the molecular chaperone Hsc70. Together, the results from this study provide evidence that c-Myc, AKT, Hsc70, and TBX3 form part of an important oncogenic pathway in breast cancer and thus reveal versatile ways of interfering with the oncogenic activity of TBX3 for the treatment of this neoplasm. Implications: Targeting the c-Myc/AKT/TBX3/Hsc70 signaling axis may be an effective treatment strategy for TBX3-driven breast cancer.","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142203090","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
Intrinsic Epigenetic State of Primary Osteosarcoma Drives Metastasis. 原发性骨肉瘤的内在表观遗传学状态推动了转移。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-04 DOI: 10.1158/1541-7786.MCR-23-0055
Irtisha Singh, Nino Rainusso, Lyazat Kurenbekova, Bikesh K Nirala, Juan Dou, Abhinaya Muruganandham, Jason T Yustein

Osteosarcoma is the most common primary malignant bone tumor affecting the pediatric population with a high potential to metastasize. However, insights into the molecular features enabling its metastatic potential are limited. We mapped the active chromatin landscapes of osteosarcoma tumors by integrating histone H3 lysine-acetylated chromatin state (n = 13), chromatin accessibility profiles (n = 11), and gene expression (n = 13) to understand the differences in their active chromatin profiles and their impact on molecular mechanisms driving the malignant phenotypes. Primary osteosarcoma tumors from patients with metastasis (primary met) have a distinct active chromatin landscape compared with those without metastasis (localized). This difference shapes the transcriptional profile of osteosarcoma. We identified novel candidate genes, including PPP1R1B, PREX1, and IGF2BP1, that exhibit increased chromatin activity in primary met. Loss of PREX1 in primary met osteosarcoma cells significantly diminishes osteosarcoma proliferation, invasion, migration, and colony formation capacity. Differential chromatin activity in primary met is associated with genes regulating cytoskeleton organization, cellular adhesion, and extracellular matrix, suggesting their role in facilitating osteosarcoma metastasis. Chromatin profiling of tumors from metastatic lung lesions shows increased chromatin activity in genes involved in cell migration and Wnt pathway. These data demonstrate that metastatic potential is intrinsically present in primary met tumors, with cellular chromatin profiles further adapting for successful dissemination, migration, and colonization at the distal site. Implications: Our study demonstrates that metastatic potential is intrinsic to primary metastatic osteosarcoma tumors, with chromatin profiles further adapting for successful dissemination, migration, and colonization at the distal metastatic site.

骨肉瘤(Osteosarcoma,OS)是影响儿童群体的最常见的原发性恶性骨肿瘤,具有很高的转移潜力。然而,人们对导致其转移潜力的分子特征的了解还很有限。我们通过整合组蛋白H3赖氨酸乙酰化染色质状态(13例)、染色质可及性图谱(11例)和基因表达(13例),绘制了OS肿瘤的活性染色质图谱,以了解其活性染色质图谱的差异及其对驱动恶性表型的分子机制的影响。有转移(原发转移)患者的原发性OS肿瘤与无转移(局部转移)患者的原发性OS肿瘤相比,具有不同的活性染色质图谱。这种差异决定了 OS 的转录谱。我们发现了新的候选基因,包括 PPP1R1B、PREX1 和 IGF2BP1,这些基因在原发性 met 中表现出更高的染色质活性。原代met OS细胞中PREX1的缺失会显著降低OS的增殖、侵袭、迁移和集落形成能力。原发性met的染色质活性差异与调控细胞骨架组织、细胞粘附和细胞外基质的基因有关,这表明它们在促进OS转移中的作用。对转移性肺部病变的肿瘤进行染色质图谱分析表明,参与细胞迁移和Wnt通路的基因的染色质活性增加。这些数据表明,原发性转移性肿瘤本身就具有转移潜能,细胞染色质图谱会进一步调整,以便在远端部位成功扩散、迁移和定植。影响:我们的研究表明,转移潜能是原发性转移性骨肉瘤肿瘤的固有特性,染色质图谱会进一步适应远端转移部位的成功扩散、迁移和定植。
{"title":"Intrinsic Epigenetic State of Primary Osteosarcoma Drives Metastasis.","authors":"Irtisha Singh, Nino Rainusso, Lyazat Kurenbekova, Bikesh K Nirala, Juan Dou, Abhinaya Muruganandham, Jason T Yustein","doi":"10.1158/1541-7786.MCR-23-0055","DOIUrl":"10.1158/1541-7786.MCR-23-0055","url":null,"abstract":"<p><p>Osteosarcoma is the most common primary malignant bone tumor affecting the pediatric population with a high potential to metastasize. However, insights into the molecular features enabling its metastatic potential are limited. We mapped the active chromatin landscapes of osteosarcoma tumors by integrating histone H3 lysine-acetylated chromatin state (n = 13), chromatin accessibility profiles (n = 11), and gene expression (n = 13) to understand the differences in their active chromatin profiles and their impact on molecular mechanisms driving the malignant phenotypes. Primary osteosarcoma tumors from patients with metastasis (primary met) have a distinct active chromatin landscape compared with those without metastasis (localized). This difference shapes the transcriptional profile of osteosarcoma. We identified novel candidate genes, including PPP1R1B, PREX1, and IGF2BP1, that exhibit increased chromatin activity in primary met. Loss of PREX1 in primary met osteosarcoma cells significantly diminishes osteosarcoma proliferation, invasion, migration, and colony formation capacity. Differential chromatin activity in primary met is associated with genes regulating cytoskeleton organization, cellular adhesion, and extracellular matrix, suggesting their role in facilitating osteosarcoma metastasis. Chromatin profiling of tumors from metastatic lung lesions shows increased chromatin activity in genes involved in cell migration and Wnt pathway. These data demonstrate that metastatic potential is intrinsically present in primary met tumors, with cellular chromatin profiles further adapting for successful dissemination, migration, and colonization at the distal site. Implications: Our study demonstrates that metastatic potential is intrinsic to primary metastatic osteosarcoma tumors, with chromatin profiles further adapting for successful dissemination, migration, and colonization at the distal metastatic site.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141262338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
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