首页 > 最新文献

Molecular Cancer Research最新文献

英文 中文
Canonical and Nuclear mTOR Specify Distinct Transcriptional Programs in Androgen-Dependent Prostate Cancer Cells. 典型和核mTOR在雄激素依赖性前列腺癌症细胞中指定了不同的转录程序。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 DOI: 10.1158/1541-7786.MCR-23-0087
Yonghong Chen, Lingwei Han, Catherine Rosa Dufour, Anthony Alfonso, Vincent Giguère

mTOR is a serine/threonine kinase that controls prostate cancer cell growth in part by regulating gene programs associated with metabolic and cell proliferation pathways. mTOR-mediated control of gene expression can be achieved via phosphorylation of transcription factors, leading to changes in their cellular localization and activities. mTOR also directly associates with chromatin in complex with transcriptional regulators, including the androgen receptor (AR). Nuclear mTOR (nmTOR) has been previously shown to act as a transcriptional integrator of the androgen signaling pathway in association with the chromatin remodeling machinery, AR, and FOXA1. However, the contribution of cytoplasmic mTOR (cmTOR) and nmTOR and the role played by FOXA1 in this process remains to be explored. Herein, we engineered cells expressing mTOR tagged with nuclear localization and export signals dictating mTOR localization. Transcriptome profiling in AR-positive prostate cancer cells revealed that nmTOR generally downregulates a subset of the androgen response pathway independently of its kinase activity, while cmTOR upregulates a cell cycle-related gene signature in a kinase-dependent manner. Biochemical and genome-wide transcriptomic analyses demonstrate that nmTOR functionally interacts with AR and FOXA1. Ablation of FOXA1 reprograms the nmTOR cistrome and transcriptome of androgen responsive prostate cancer cells. This works highlights a transcriptional regulatory pathway in which direct interactions between nmTOR, AR and FOXA1 dictate a combinatorial role for these factors in the control of specific gene programs in prostate cancer cells.

Implications: The finding that canonical and nuclear mTOR signaling pathways control distinct gene programs opens therapeutic opportunities to modulate mTOR activity in prostate cancer cells.

mTOR是一种丝氨酸/苏氨酸激酶,部分通过调节与代谢和细胞增殖途径相关的基因程序来控制前列腺癌症(PCa)细胞生长。mTOR介导的基因表达控制可以通过转录因子的磷酸化实现,从而改变其细胞定位和活性。mTOR还与染色质直接结合,与转录调节因子形成复合体,包括雄激素受体(AR)。核mTOR(nmTOR)先前已被证明是雄激素信号通路的转录整合因子,与染色质重塑机制AR和FOXA1相关。然而,细胞质mTOR(cmTOR)和nmTOR的贡献以及FOXA1在这一过程中所起的作用仍有待探索。在此,我们设计了表达mTOR的细胞,用核定位标记并输出指示mTOR定位的信号。AR阳性PCa细胞的转录组分析显示,nmTOR通常下调雄激素反应途径的一个子集,而不依赖于其激酶活性,而cmTOR以激酶依赖的方式上调细胞周期相关的基因标记。生化和全基因组转录组学分析表明,nmTOR在功能上与AR和FOXA1相互作用。FOXA1的消融重新编程雄激素反应性PCa细胞的nmTOR池组和转录组。这项工作强调了一种转录调控途径,其中nmTOR、AR和FOXA1之间的直接相互作用决定了这些因子在PCa细胞中控制特定基因程序中的组合作用。意义:经典和核mTOR信号通路控制不同基因程序的发现为调节前列腺癌细胞中mTOR活性提供了治疗机会。
{"title":"Canonical and Nuclear mTOR Specify Distinct Transcriptional Programs in Androgen-Dependent Prostate Cancer Cells.","authors":"Yonghong Chen, Lingwei Han, Catherine Rosa Dufour, Anthony Alfonso, Vincent Giguère","doi":"10.1158/1541-7786.MCR-23-0087","DOIUrl":"10.1158/1541-7786.MCR-23-0087","url":null,"abstract":"<p><p>mTOR is a serine/threonine kinase that controls prostate cancer cell growth in part by regulating gene programs associated with metabolic and cell proliferation pathways. mTOR-mediated control of gene expression can be achieved via phosphorylation of transcription factors, leading to changes in their cellular localization and activities. mTOR also directly associates with chromatin in complex with transcriptional regulators, including the androgen receptor (AR). Nuclear mTOR (nmTOR) has been previously shown to act as a transcriptional integrator of the androgen signaling pathway in association with the chromatin remodeling machinery, AR, and FOXA1. However, the contribution of cytoplasmic mTOR (cmTOR) and nmTOR and the role played by FOXA1 in this process remains to be explored. Herein, we engineered cells expressing mTOR tagged with nuclear localization and export signals dictating mTOR localization. Transcriptome profiling in AR-positive prostate cancer cells revealed that nmTOR generally downregulates a subset of the androgen response pathway independently of its kinase activity, while cmTOR upregulates a cell cycle-related gene signature in a kinase-dependent manner. Biochemical and genome-wide transcriptomic analyses demonstrate that nmTOR functionally interacts with AR and FOXA1. Ablation of FOXA1 reprograms the nmTOR cistrome and transcriptome of androgen responsive prostate cancer cells. This works highlights a transcriptional regulatory pathway in which direct interactions between nmTOR, AR and FOXA1 dictate a combinatorial role for these factors in the control of specific gene programs in prostate cancer cells.</p><p><strong>Implications: </strong>The finding that canonical and nuclear mTOR signaling pathways control distinct gene programs opens therapeutic opportunities to modulate mTOR activity in prostate cancer cells.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"113-124"},"PeriodicalIF":4.1,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"54230316","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
Spatial Transcriptomics Suggests That Alterations Occur in the Preneoplastic Breast Microenvironment of BRCA1/2 Mutation Carriers. 空间转录组学表明,BRCA1/2突变携带者的肿瘤前乳房微环境发生了改变。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 DOI: 10.1158/1541-7786.MCR-23-0489
Anthony Caputo, Kavya Vipparthi, Peter Bazeley, Erinn Downs-Kelly, Patrick McIntire, Lauren A Duckworth, Ying Ni, Bo Hu, Ruth A Keri, Mihriban Karaayvaz

Breast cancer is the most common cancer in females, affecting one in every eight women and accounting for the majority of cancer-related deaths in women worldwide. Germline mutations in the BRCA1 and BRCA2 genes are significant risk factors for specific subtypes of breast cancer. BRCA1 mutations are associated with basal-like breast cancers, whereas BRCA2 mutations are associated with luminal-like disease. Defects in mammary epithelial cell differentiation have been previously recognized in germline BRCA1/2 mutation carriers even before cancer incidence. However, the underlying mechanism is largely unknown. Here, we employ spatial transcriptomics to investigate defects in mammary epithelial cell differentiation accompanied by distinct microenvironmental alterations in preneoplastic breast tissues from BRCA1/2 mutation carriers and normal breast tissues from noncarrier controls. We uncovered spatially defined receptor-ligand interactions in these tissues for the investigation of autocrine and paracrine signaling. We discovered that β1-integrin-mediated autocrine signaling in BRCA2-deficient mammary epithelial cells may differ from BRCA1-deficient mammary epithelial cells. In addition, we found that the epithelial-to-stromal paracrine signaling in the breast tissues of BRCA1/2 mutation carriers is greater than in control tissues. More integrin-ligand pairs were differentially correlated in BRCA1/2-mutant breast tissues than noncarrier breast tissues with more integrin receptor-expressing stromal cells.

Implications: These results suggest alterations in the communication between mammary epithelial cells and the microenvironment in BRCA1 and BRCA2 mutation carriers, laying the foundation for designing innovative breast cancer chemo-prevention strategies for high-risk patients.

癌症是女性最常见的癌症,每八名女性中就有一人患有乳腺癌,占全世界女性癌症相关死亡的大多数。BRCA1和BRCA2基因的种系突变是癌症特定亚型的重要危险因素。BRCA1突变与基底样乳腺癌相关,而BRCA2突变与管腔样疾病相关。甚至在癌症发病之前,就已经在种系BRCA1/2突变携带者中发现了乳腺上皮细胞分化缺陷。然而,其根本机制在很大程度上是未知的。在这里,我们使用空间转录组学来研究BRCA1/2突变携带者的癌前乳腺组织和非携带者对照的正常乳腺组织中乳腺上皮细胞分化的缺陷,并伴有不同的微环境变化。我们揭示了这些组织中空间定义的受体-配体相互作用,用于研究自分泌和旁分泌信号传导。我们发现BRCA2缺陷的乳腺上皮细胞中β1-整合素介导的自分泌信号可能与BRCA1缺陷的乳腺表皮细胞不同。此外,我们发现BRCA1/2突变携带者乳腺组织中上皮-基质旁分泌信号传导大于对照组织。与具有更多整合素受体表达基质细胞的非载体乳腺组织相比,BRCA1/2毛乳腺组织中更多的整合素配体对存在差异相关性。意义:这些结果表明,BRCA1和BRCA2突变携带者的乳腺上皮细胞与微环境之间的通讯发生了变化,为为高危患者设计创新的乳腺癌症化疗预防策略奠定了基础。
{"title":"Spatial Transcriptomics Suggests That Alterations Occur in the Preneoplastic Breast Microenvironment of BRCA1/2 Mutation Carriers.","authors":"Anthony Caputo, Kavya Vipparthi, Peter Bazeley, Erinn Downs-Kelly, Patrick McIntire, Lauren A Duckworth, Ying Ni, Bo Hu, Ruth A Keri, Mihriban Karaayvaz","doi":"10.1158/1541-7786.MCR-23-0489","DOIUrl":"10.1158/1541-7786.MCR-23-0489","url":null,"abstract":"<p><p>Breast cancer is the most common cancer in females, affecting one in every eight women and accounting for the majority of cancer-related deaths in women worldwide. Germline mutations in the BRCA1 and BRCA2 genes are significant risk factors for specific subtypes of breast cancer. BRCA1 mutations are associated with basal-like breast cancers, whereas BRCA2 mutations are associated with luminal-like disease. Defects in mammary epithelial cell differentiation have been previously recognized in germline BRCA1/2 mutation carriers even before cancer incidence. However, the underlying mechanism is largely unknown. Here, we employ spatial transcriptomics to investigate defects in mammary epithelial cell differentiation accompanied by distinct microenvironmental alterations in preneoplastic breast tissues from BRCA1/2 mutation carriers and normal breast tissues from noncarrier controls. We uncovered spatially defined receptor-ligand interactions in these tissues for the investigation of autocrine and paracrine signaling. We discovered that β1-integrin-mediated autocrine signaling in BRCA2-deficient mammary epithelial cells may differ from BRCA1-deficient mammary epithelial cells. In addition, we found that the epithelial-to-stromal paracrine signaling in the breast tissues of BRCA1/2 mutation carriers is greater than in control tissues. More integrin-ligand pairs were differentially correlated in BRCA1/2-mutant breast tissues than noncarrier breast tissues with more integrin receptor-expressing stromal cells.</p><p><strong>Implications: </strong>These results suggest alterations in the communication between mammary epithelial cells and the microenvironment in BRCA1 and BRCA2 mutation carriers, laying the foundation for designing innovative breast cancer chemo-prevention strategies for high-risk patients.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"169-180"},"PeriodicalIF":4.1,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10872731/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50158434","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
Exosomal lncRNA NEAT1 Inhibits NK-Cell Activity to Promote Multiple Myeloma Cell Immune Escape via an EZH2/PBX1 Axis. 外泌体lncRNA NEAT1通过EZH2/PBX1轴抑制NK细胞活性以促进多发性骨髓瘤细胞免疫逃逸。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 DOI: 10.1158/1541-7786.MCR-23-0282
Qing-Ming Wang, Guang-Yu Lian, Su-Mei Sheng, Jing Xu, Long-Long Ye, Chao Min, Shu-Fang Guo

Exosomal long noncoding RNAs (lncRNA) derived from cancer cells are implicated in various processes, including cancer cell proliferation, metastasis, and immunomodulation. We investigated the role and underlying mechanism of exosome-transmitted lncRNA NEAT1 in the immune escape of multiple myeloma cells from natural killer (NK) cells. Multiple myeloma cells and samples from patients with multiple myeloma were obtained. The effects of multiple myeloma cell-derived exosomes (multiple myeloma exosomes) and exosomal NEAT1 on the functions of NK cells were evaluated using EdU staining, CCK-8, flow cytometry, and ELISA. Chromatin and RNA immunoprecipitation were performed to identify interactions between NEAT1, enhancer of Zeste Homolog 2 (EZH2), and pre-B-cell leukemia transcription factor 1 (PBX1). A xenograft tumor model was constructed to verify the effects of exosomal NEAT1 on tumor growth. qRT-PCR, Western blot analysis, and IHC were conducted to detect related genes. NEAT1 levels were upregulated in multiple myeloma tumor tissues, multiple myeloma cells, and multiple myeloma exosomes. Multiple myeloma exosomes suppressed cell proliferation, promoted apoptosis, reduced natural killer group 2, member D (NKG2D)-positive cells, and the production of TNFα) and interferon-gamma (IFN-γ) in NK cells, whereas NEAT1-silenced exosomes had little effect. NEAT1 silenced PBX1 by recruiting EZH2. PBX1 knockdown abrogated the effects of NEAT1-silenced exosomes on NK and multiple myeloma cells. NEAT1-silenced exosomes inhibited tumor growth in mice, decreased Ki67 and PD-L1, and increased NKG2D, TNFα, and IFNγ in tumor tissues. In summary, multiple myeloma cell-derived exosomal NEAT1 suppressed NK-cell activity by downregulating PBX1, promoting multiple myeloma cell immune escape. This study suggests a potential strategy for treating multiple myeloma.

Implications: This study reveals that exosomal NEAT1 regulates EZH2/PBX1 axis to inhibit NK-cell activity, thereby promoting multiple myeloma cell immune escape, which offers a novel therapeutic potential for multiple myeloma.

来源于癌症细胞的外显体长非编码RNA(lncRNA)参与各种过程,包括癌症细胞增殖、转移和免疫调节。我们研究了外泌体传输的lncRNA NEAT1在多发性骨髓瘤(MM)细胞从自然杀伤(NK)细胞免疫逃逸中的作用和潜在机制。获得MM患者的MM细胞和样本。利用EdU染色、CCK-8、流式细胞术和ELISA评估MM细胞来源的外泌体(MM外泌体)和外泌体NEAT1对NK细胞功能的影响。染色质和RNA免疫沉淀用于鉴定NEAT1、Zeste同源物2的增强子(EZH2)和前B细胞白血病转录因子1(PBX1)之间的相互作用。构建异种移植物肿瘤模型以验证外泌体NEAT1对肿瘤生长的影响。用qRT-PCR、westernblot和免疫组织化学方法检测相关基因。NEAT1水平在MM肿瘤组织、MM细胞和MM外泌体中上调。MM外泌体抑制细胞增殖,促进细胞凋亡,减少自然杀伤组2成员D(NKG2D)阳性细胞,以及NK细胞中肿瘤坏死因子α(TNF-α)和干扰素γ(IFN-γ)的产生,而NEAT1沉默的外泌体几乎没有作用。NEAT1通过招募EZH2使PBX1沉默。PBX1敲除消除了NEAT1沉默的外泌体对NK和MM细胞的影响。NEAT1沉默的外泌体抑制小鼠的肿瘤生长,降低Ki67和PD-L1,并增加肿瘤组织中的NKG2D、TNF-α和IFN-γ。总之,MM细胞衍生的外泌体NEAT1通过下调PBX1抑制NK细胞活性,促进MM细胞免疫逃逸。本研究提出了一种治疗MM的潜在策略。提示:本研究揭示了外泌体NEAT1调节EZH2/PBX1轴抑制NK细胞活性,从而促进多发性骨髓瘤细胞免疫逃逸,为MM的治疗提供了新的潜力。
{"title":"Exosomal lncRNA NEAT1 Inhibits NK-Cell Activity to Promote Multiple Myeloma Cell Immune Escape via an EZH2/PBX1 Axis.","authors":"Qing-Ming Wang, Guang-Yu Lian, Su-Mei Sheng, Jing Xu, Long-Long Ye, Chao Min, Shu-Fang Guo","doi":"10.1158/1541-7786.MCR-23-0282","DOIUrl":"10.1158/1541-7786.MCR-23-0282","url":null,"abstract":"<p><p>Exosomal long noncoding RNAs (lncRNA) derived from cancer cells are implicated in various processes, including cancer cell proliferation, metastasis, and immunomodulation. We investigated the role and underlying mechanism of exosome-transmitted lncRNA NEAT1 in the immune escape of multiple myeloma cells from natural killer (NK) cells. Multiple myeloma cells and samples from patients with multiple myeloma were obtained. The effects of multiple myeloma cell-derived exosomes (multiple myeloma exosomes) and exosomal NEAT1 on the functions of NK cells were evaluated using EdU staining, CCK-8, flow cytometry, and ELISA. Chromatin and RNA immunoprecipitation were performed to identify interactions between NEAT1, enhancer of Zeste Homolog 2 (EZH2), and pre-B-cell leukemia transcription factor 1 (PBX1). A xenograft tumor model was constructed to verify the effects of exosomal NEAT1 on tumor growth. qRT-PCR, Western blot analysis, and IHC were conducted to detect related genes. NEAT1 levels were upregulated in multiple myeloma tumor tissues, multiple myeloma cells, and multiple myeloma exosomes. Multiple myeloma exosomes suppressed cell proliferation, promoted apoptosis, reduced natural killer group 2, member D (NKG2D)-positive cells, and the production of TNFα) and interferon-gamma (IFN-γ) in NK cells, whereas NEAT1-silenced exosomes had little effect. NEAT1 silenced PBX1 by recruiting EZH2. PBX1 knockdown abrogated the effects of NEAT1-silenced exosomes on NK and multiple myeloma cells. NEAT1-silenced exosomes inhibited tumor growth in mice, decreased Ki67 and PD-L1, and increased NKG2D, TNFα, and IFNγ in tumor tissues. In summary, multiple myeloma cell-derived exosomal NEAT1 suppressed NK-cell activity by downregulating PBX1, promoting multiple myeloma cell immune escape. This study suggests a potential strategy for treating multiple myeloma.</p><p><strong>Implications: </strong>This study reveals that exosomal NEAT1 regulates EZH2/PBX1 axis to inhibit NK-cell activity, thereby promoting multiple myeloma cell immune escape, which offers a novel therapeutic potential for multiple myeloma.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"125-136"},"PeriodicalIF":4.1,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"54230317","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
ORAOV1, CCND1, and MIR548K Are the Driver Oncogenes of the 11q13 Amplicon in Squamous Cell Carcinoma. ORAOV1、CCND1和MIR548K是鳞状细胞癌11q13扩增子的驱动致癌基因。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 DOI: 10.1158/1541-7786.MCR-23-0746
Céline I Mahieu, Andrew G Mancini, Ellee P Vikram, Vicente Planells-Palop, Nancy M Joseph, Aaron D Tward

11q13 amplification is a frequent event in human cancer and in particular in squamous cell carcinomas (SCC). Despite almost invariably spanning 10 genes, it is unclear which genetic components of the amplicon are the key driver events in SCC. A combination of computational, in vitro, ex vivo, and in vivo models leveraging efficient primary human keratinocyte genome editing by Cas9-RNP electroporation, identified ORAOV1, CCND1, and MIR548K as the critical drivers of the amplicon in head and neck SCC. CCND1 amplification drives the cell cycle in a CDK4/6/RB1-independent fashion and may confer a novel dependency on RRM2. MIR548K contributes to epithelial-mesenchymal transition. Finally, we identify ORAOV1 as an oncogene that acts likely via its ability to modulate reactive oxygen species. Thus, the 11q13 amplicon drives SCC through at least three independent genetic elements and suggests therapeutic targets for this morbid and lethal disease.

Implications: This work demonstrates novel mechanisms and ways to target these mechanisms underlying the most common amplification in squamous cell carcinoma, one of the most prevalent and deadly forms of human cancer.

11q13扩增是人类癌症,特别是鳞状细胞癌(SCC)中的常见事件。尽管几乎总是跨越10个基因,但尚不清楚扩增子的哪些遗传成分是SCC的关键驱动因素。计算、体外、离体和体内模型的结合利用了通过Cas9 RNP电穿孔进行的有效的原代人类角质形成细胞基因组编辑,确定ORAOV1、CCND1和MIR548K是头颈部SCC中扩增子的关键驱动因素。CCND1扩增以CDK4/6/RB1独立的方式驱动细胞周期,并且可以赋予对RRM2的新的依赖性。MIR548K有助于上皮-间质转化。最后,我们确定ORAOV1是一种致癌基因,其作用可能是通过其调节活性氧的能力。因此,11q13扩增子通过至少三个独立的遗传元件驱动SCC,并提出了这种病态和致命疾病的治疗靶点。意义:这项工作展示了新的机制和方法来靶向鳞状细胞癌中最常见扩增的这些机制,鳞状细胞癌是人类癌症最普遍和致命的形式之一。
{"title":"ORAOV1, CCND1, and MIR548K Are the Driver Oncogenes of the 11q13 Amplicon in Squamous Cell Carcinoma.","authors":"Céline I Mahieu, Andrew G Mancini, Ellee P Vikram, Vicente Planells-Palop, Nancy M Joseph, Aaron D Tward","doi":"10.1158/1541-7786.MCR-23-0746","DOIUrl":"10.1158/1541-7786.MCR-23-0746","url":null,"abstract":"<p><p>11q13 amplification is a frequent event in human cancer and in particular in squamous cell carcinomas (SCC). Despite almost invariably spanning 10 genes, it is unclear which genetic components of the amplicon are the key driver events in SCC. A combination of computational, in vitro, ex vivo, and in vivo models leveraging efficient primary human keratinocyte genome editing by Cas9-RNP electroporation, identified ORAOV1, CCND1, and MIR548K as the critical drivers of the amplicon in head and neck SCC. CCND1 amplification drives the cell cycle in a CDK4/6/RB1-independent fashion and may confer a novel dependency on RRM2. MIR548K contributes to epithelial-mesenchymal transition. Finally, we identify ORAOV1 as an oncogene that acts likely via its ability to modulate reactive oxygen species. Thus, the 11q13 amplicon drives SCC through at least three independent genetic elements and suggests therapeutic targets for this morbid and lethal disease.</p><p><strong>Implications: </strong>This work demonstrates novel mechanisms and ways to target these mechanisms underlying the most common amplification in squamous cell carcinoma, one of the most prevalent and deadly forms of human cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"152-168"},"PeriodicalIF":4.1,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10831340/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71484220","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
SOX10 Loss Sensitizes Melanoma Cells to Cytokine-Mediated Inflammatory Cell Death. SOX10缺失使黑色素瘤细胞对细胞因子介导的炎症细胞死亡敏感。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 DOI: 10.1158/1541-7786.MCR-23-0290
Sheera R Rosenbaum, Signe Caksa, Casey D Stefanski, Isabella V Trachtenberg, Haley P Wilson, Nicole A Wilski, Connor A Ott, Timothy J Purwin, Jelan I Haj, Danielle Pomante, Daniel Kotas, Inna Chervoneva, Claudia Capparelli, Andrew E Aplin

The transcription factor, SOX10, plays an important role in the differentiation of neural crest precursors to the melanocytic lineage. Malignant transformation of melanocytes leads to the development of melanoma, and SOX10 promotes melanoma cell proliferation and tumor formation. SOX10 expression in melanomas is heterogeneous, and loss of SOX10 causes a phenotypic switch toward an invasive, mesenchymal-like cell state and therapy resistance; hence, strategies to target SOX10-deficient cells are an active area of investigation. The impact of cell state and SOX10 expression on antitumor immunity is not well understood but will likely have important implications for immunotherapeutic interventions. To this end, we tested whether SOX10 status affects the response to CD8+ T cell-mediated killing and T cell-secreted cytokines, TNFα and IFNγ, which are critical effectors in the cytotoxic killing of cancer cells. We observed that genetic ablation of SOX10 rendered melanoma cells more sensitive to CD8+ T cell-mediated killing and cell death induction by either TNFα or IFNγ. Cytokine-mediated cell death in SOX10-deficient cells was associated with features of caspase-dependent pyroptosis, an inflammatory form of cell death that has the potential to increase immune responses.

Implications: These data support a role for SOX10 expression altering the response to T cell-mediated cell death and contribute to a broader understanding of the interaction between immune cells and melanoma cells.

转录因子SOX10在神经嵴前体向黑素细胞谱系的分化中起着重要作用。黑色素细胞的恶性转化导致黑色素瘤的发展,SOX10促进黑色素瘤细胞增殖和肿瘤形成。SOX10在黑色素瘤中的表达是异质性的,SOX10的缺失导致表型转变为侵袭性间充质样细胞状态和治疗耐药性;因此,靶向SOX10缺陷细胞的策略是一个活跃的研究领域。细胞状态和SOX10表达对抗肿瘤免疫的影响尚不清楚,但可能对免疫治疗干预具有重要意义。为此,我们测试了SOX10状态是否影响对CD8+T细胞介导的杀伤和T细胞分泌的细胞因子TNFα和IFNγ的反应,这些细胞因子是癌症细胞毒性杀伤的关键效应物。我们观察到SOX10的基因消融使黑色素瘤细胞对TNFα或IFNγ介导的CD8+T细胞介导的杀伤和细胞死亡诱导更敏感。SOX10缺陷细胞中细胞因子介导的细胞死亡与胱天蛋白酶依赖性焦下垂的特征有关,这是一种具有增加免疫反应潜力的细胞死亡的炎症形式。含义:这些数据支持SOX10表达改变T细胞介导的细胞死亡反应的作用,并有助于更广泛地理解免疫细胞和黑色素瘤细胞之间的相互作用。
{"title":"SOX10 Loss Sensitizes Melanoma Cells to Cytokine-Mediated Inflammatory Cell Death.","authors":"Sheera R Rosenbaum, Signe Caksa, Casey D Stefanski, Isabella V Trachtenberg, Haley P Wilson, Nicole A Wilski, Connor A Ott, Timothy J Purwin, Jelan I Haj, Danielle Pomante, Daniel Kotas, Inna Chervoneva, Claudia Capparelli, Andrew E Aplin","doi":"10.1158/1541-7786.MCR-23-0290","DOIUrl":"10.1158/1541-7786.MCR-23-0290","url":null,"abstract":"<p><p>The transcription factor, SOX10, plays an important role in the differentiation of neural crest precursors to the melanocytic lineage. Malignant transformation of melanocytes leads to the development of melanoma, and SOX10 promotes melanoma cell proliferation and tumor formation. SOX10 expression in melanomas is heterogeneous, and loss of SOX10 causes a phenotypic switch toward an invasive, mesenchymal-like cell state and therapy resistance; hence, strategies to target SOX10-deficient cells are an active area of investigation. The impact of cell state and SOX10 expression on antitumor immunity is not well understood but will likely have important implications for immunotherapeutic interventions. To this end, we tested whether SOX10 status affects the response to CD8+ T cell-mediated killing and T cell-secreted cytokines, TNFα and IFNγ, which are critical effectors in the cytotoxic killing of cancer cells. We observed that genetic ablation of SOX10 rendered melanoma cells more sensitive to CD8+ T cell-mediated killing and cell death induction by either TNFα or IFNγ. Cytokine-mediated cell death in SOX10-deficient cells was associated with features of caspase-dependent pyroptosis, an inflammatory form of cell death that has the potential to increase immune responses.</p><p><strong>Implications: </strong>These data support a role for SOX10 expression altering the response to T cell-mediated cell death and contribute to a broader understanding of the interaction between immune cells and melanoma cells.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"209-220"},"PeriodicalIF":4.1,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10842433/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41237159","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
PKCδ Regulates Chromatin Remodeling and DNA Repair through SIRT6. PKC通过SIRT6调节染色质重塑和DNA修复。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 DOI: 10.1158/1541-7786.MCR-23-0493
Trisiani Affandi, Ami Haas, Angela M Ohm, Gregory M Wright, Joshua C Black, Mary E Reyland

Irradiation (IR) is a highly effective cancer therapy; however, IR damage to tumor-adjacent healthy tissues can result in significant comorbidities and potentially limit the course of therapy. We have previously shown that protein kinase C delta (PKCδ) is required for IR-induced apoptosis and that inhibition of PKCδ activity provides radioprotection in vivo. Here we show that PKCδ regulates histone modification, chromatin accessibility, and double-stranded break (DSB) repair through a mechanism that requires Sirtuin 6 (SIRT6). Overexpression of PKCδ promotes genomic instability and increases DNA damage and apoptosis. Conversely, depletion of PKCδ increases DNA repair via nonhomologous end joining (NHEJ) and homologous recombination (HR) as evidenced by increased formation of DNA damage foci, increased expression of DNA repair proteins, and increased repair of NHEJ and HR fluorescent reporter constructs. Nuclease sensitivity indicates that PKCδ depletion is associated with more open chromatin, while overexpression of PKCδ reduces chromatin accessibility. Epiproteome analysis reveals increased chromatin associated H3K36me2 in PKCδ-depleted cells which is accompanied by chromatin disassociation of KDM2A. We identify SIRT6 as a downstream mediator of PKCδ. PKCδ-depleted cells have increased SIRT6 expression, and depletion of SIRT6 reverses changes in chromatin accessibility, histone modification and DSB repair in PKCδ-depleted cells. Furthermore, depletion of SIRT6 reverses radioprotection in PKCδ-depleted cells. Our studies describe a novel pathway whereby PKCδ orchestrates SIRT6-dependent changes in chromatin accessibility to regulate DNA repair, and define a mechanism for regulation of radiation-induced apoptosis by PKCδ.

Implications: PKCδ controls sensitivity to irradiation by regulating DNA repair.

辐射(IR)是一种高效的癌症治疗方法,然而,IR对肿瘤邻近健康组织的损伤可能导致显著的并发症,并可能限制治疗过程。我们之前已经表明,蛋白激酶Cδ(PKC?)是IR诱导的细胞凋亡所必需的,并且PKC?活性的抑制在体内提供了放射性保护。在这里,我们发现PKC通过一种需要SIRT6的机制调节组蛋白修饰、染色质可及性和双链断裂(DSB)修复。PKC的过度表达促进了基因组的不稳定性,并增加了DNA损伤和细胞凋亡。相反,PKC的缺失通过非同源末端连接(NHEJ)和同源重组(HR)增加了DNA修复,这可以通过增加DNA损伤灶的形成、增加DNA修复蛋白的表达以及增加NHEJ和HR荧光报告构建体的修复来证明。核酸酶敏感性表明PKC的缺失与更开放的染色质有关,而PKC的过表达降低了染色质的可及性。表观蛋白质组分析显示,PKC缺失细胞中染色质相关的H3K36me2增加,伴有KDM2A的染色质解离。我们确定SIRT6是PKC的下游介质。PKCğ-缺失的细胞增加了SIRT6的表达,SIRT6的缺失逆转了PKC abl-缺失细胞中染色质可及性、组蛋白修饰和DSB修复的变化。此外,SIRT6的缺失可逆转PKC缺失细胞的放射保护作用。我们的研究描述了一种新的途径,PKC通过该途径协调染色质可及性的SIRT6依赖性变化来调节DNA修复,并确定了PKC调节辐射诱导的细胞凋亡的机制。意义:PKC通过调节DNA修复来控制对辐射的敏感性。
{"title":"PKCδ Regulates Chromatin Remodeling and DNA Repair through SIRT6.","authors":"Trisiani Affandi, Ami Haas, Angela M Ohm, Gregory M Wright, Joshua C Black, Mary E Reyland","doi":"10.1158/1541-7786.MCR-23-0493","DOIUrl":"10.1158/1541-7786.MCR-23-0493","url":null,"abstract":"<p><p>Irradiation (IR) is a highly effective cancer therapy; however, IR damage to tumor-adjacent healthy tissues can result in significant comorbidities and potentially limit the course of therapy. We have previously shown that protein kinase C delta (PKCδ) is required for IR-induced apoptosis and that inhibition of PKCδ activity provides radioprotection in vivo. Here we show that PKCδ regulates histone modification, chromatin accessibility, and double-stranded break (DSB) repair through a mechanism that requires Sirtuin 6 (SIRT6). Overexpression of PKCδ promotes genomic instability and increases DNA damage and apoptosis. Conversely, depletion of PKCδ increases DNA repair via nonhomologous end joining (NHEJ) and homologous recombination (HR) as evidenced by increased formation of DNA damage foci, increased expression of DNA repair proteins, and increased repair of NHEJ and HR fluorescent reporter constructs. Nuclease sensitivity indicates that PKCδ depletion is associated with more open chromatin, while overexpression of PKCδ reduces chromatin accessibility. Epiproteome analysis reveals increased chromatin associated H3K36me2 in PKCδ-depleted cells which is accompanied by chromatin disassociation of KDM2A. We identify SIRT6 as a downstream mediator of PKCδ. PKCδ-depleted cells have increased SIRT6 expression, and depletion of SIRT6 reverses changes in chromatin accessibility, histone modification and DSB repair in PKCδ-depleted cells. Furthermore, depletion of SIRT6 reverses radioprotection in PKCδ-depleted cells. Our studies describe a novel pathway whereby PKCδ orchestrates SIRT6-dependent changes in chromatin accessibility to regulate DNA repair, and define a mechanism for regulation of radiation-induced apoptosis by PKCδ.</p><p><strong>Implications: </strong>PKCδ controls sensitivity to irradiation by regulating DNA repair.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"181-196"},"PeriodicalIF":4.1,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10872792/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"54230318","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
LRRC75A-AS1 drives the epithelial-mesenchymal transition in cervical cancer by binding IGF2BP1 and inhibiting SYVN1-mediated NLRP3 ubiquitination. LRRC75A-AS1 通过结合 IGF2BP1 和抑制 SYVN1 介导的 NLRP3 泛素化,推动宫颈癌的上皮-间质转化。
IF 5.2 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-01-05 DOI: 10.1158/1541-7786.MCR-23-0478
Hongying Sui, Caixia Shi, Zhipeng Yan, Jinyang Chen, Lin Man, Fang Wang

Cervical cancer severely affects women's health with increased incidence and poor survival for patients with metastasis. Our study aims to investigate the mechanism by which lncRNA LRRC75A-AS1 regulates the epithelial-mesenchymal transition (EMT) of cervical cancer through modulating m6A and ubiquitination modification. In this study, tumor tissues were collected from patients to analyze the expression of LRRC75A-AS1 and SYVN1. Migratory and invasive capacities of HeLa and CaSki cells were evaluated with wound healing and transwell assays. CCK-8 and EdU incorporation assays were employed to examine cell proliferation. The interaction between LRRC75A-AS1, IGF2BP1, SYVN1, and NLRP3 was evaluated through RNA immunoprecipitation, RNA pull-down, FISH, and Co-IP assays, respectively. MeRIP-qPCR was applied to analyze the m6A modification of SYVN1 mRNA. A subcutaneous tumor model of cervical cancer was established. We showed LRRC75A-AS1 was upregulated in tumor tissues, and LRRC75A-AS1 enhanced EMT through activating NLRP3/IL-1β/Smad2/3 signaling in cervical cancer. Furthermore, LRRC75A-AS1 inhibited SYVN1-mediated NLRP3 ubiquitination by destabilizing SYVN1 mRNA. LRRC75A-AS1 competitively bound to IGF2BP1 protein and subsequently impaired the m6A modification of SYVN1 mRNA and its stability. Knockdown of LRRC75A-AS1 repressed EMT and tumor growth via inhibiting NLRP3/IL-1β/Smad2/3 signaling in mice. In conclusion, LRRC75A-AS1 competitively binds to IGF2BP1 protein to destabilize SYVN1 mRNA, subsequently suppresses SYVN1-mediated NLRP3 ubiquitination degradation and activates IL-1β/Smad2/3 signaling, thus promoting EMT in cervical cancer. Implications: LRRC75A-AS1 promotes cervical cancer progression, and this study suggests LRRC75A-AS1 as a new therapeutic target for cervical cancer.

宫颈癌严重影响妇女健康,发病率增加,转移患者生存率低。我们的研究旨在探讨lncRNA LRRC75A-AS1通过调节m6A和泛素化修饰调控宫颈癌上皮-间质转化(EMT)的机制。本研究收集了患者的肿瘤组织,以分析 LRRC75A-AS1 和 SYVN1 的表达。通过伤口愈合和透孔试验评估了 HeLa 和 CaSki 细胞的迁移和侵袭能力。CCK-8和EdU掺入试验用于检测细胞增殖。LRRC75A-AS1、IGF2BP1、SYVN1 和 NLRP3 之间的相互作用分别通过 RNA 免疫沉淀、RNA 下拉、FISH 和 Co-IP 试验进行了评估。MeRIP-qPCR 被用于分析 SYVN1 mRNA 的 m6A 修饰。建立了宫颈癌皮下肿瘤模型。我们发现 LRRC75A-AS1 在肿瘤组织中上调,并且 LRRC75A-AS1 通过激活 NLRP3/IL-1β/Smad2/3 信号增强了宫颈癌的 EMT。此外,LRRC75A-AS1 还通过破坏 SYVN1 mRNA 的稳定性来抑制 SYVN1 介导的 NLRP3 泛素化。LRRC75A-AS1 与 IGF2BP1 蛋白竞争性结合,进而损害了 SYVN1 mRNA 的 m6A 修饰及其稳定性。通过抑制 NLRP3/IL-1β/Smad2/3 信号传导,敲除 LRRC75A-AS1 可抑制小鼠的 EMT 和肿瘤生长。总之,RRC75A-AS1能竞争性地与IGF2BP1蛋白结合,从而破坏SYVN1 mRNA的稳定性,随后抑制SYVN1介导的NLRP3泛素化降解,激活IL-1β/Smad2/3信号传导,从而促进宫颈癌的EMT。影响:LRRC75A-AS1会促进宫颈癌的进展,本研究表明LRRC75A-AS1是宫颈癌的一个新的治疗靶点。
{"title":"LRRC75A-AS1 drives the epithelial-mesenchymal transition in cervical cancer by binding IGF2BP1 and inhibiting SYVN1-mediated NLRP3 ubiquitination.","authors":"Hongying Sui, Caixia Shi, Zhipeng Yan, Jinyang Chen, Lin Man, Fang Wang","doi":"10.1158/1541-7786.MCR-23-0478","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-23-0478","url":null,"abstract":"<p><p>Cervical cancer severely affects women's health with increased incidence and poor survival for patients with metastasis. Our study aims to investigate the mechanism by which lncRNA LRRC75A-AS1 regulates the epithelial-mesenchymal transition (EMT) of cervical cancer through modulating m6A and ubiquitination modification. In this study, tumor tissues were collected from patients to analyze the expression of LRRC75A-AS1 and SYVN1. Migratory and invasive capacities of HeLa and CaSki cells were evaluated with wound healing and transwell assays. CCK-8 and EdU incorporation assays were employed to examine cell proliferation. The interaction between LRRC75A-AS1, IGF2BP1, SYVN1, and NLRP3 was evaluated through RNA immunoprecipitation, RNA pull-down, FISH, and Co-IP assays, respectively. MeRIP-qPCR was applied to analyze the m6A modification of SYVN1 mRNA. A subcutaneous tumor model of cervical cancer was established. We showed LRRC75A-AS1 was upregulated in tumor tissues, and LRRC75A-AS1 enhanced EMT through activating NLRP3/IL-1β/Smad2/3 signaling in cervical cancer. Furthermore, LRRC75A-AS1 inhibited SYVN1-mediated NLRP3 ubiquitination by destabilizing SYVN1 mRNA. LRRC75A-AS1 competitively bound to IGF2BP1 protein and subsequently impaired the m6A modification of SYVN1 mRNA and its stability. Knockdown of LRRC75A-AS1 repressed EMT and tumor growth via inhibiting NLRP3/IL-1β/Smad2/3 signaling in mice. In conclusion, LRRC75A-AS1 competitively binds to IGF2BP1 protein to destabilize SYVN1 mRNA, subsequently suppresses SYVN1-mediated NLRP3 ubiquitination degradation and activates IL-1β/Smad2/3 signaling, thus promoting EMT in cervical cancer. Implications: LRRC75A-AS1 promotes cervical cancer progression, and this study suggests LRRC75A-AS1 as a new therapeutic target for cervical cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":5.2,"publicationDate":"2024-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139098293","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
SP4 Facilitates Esophageal Squamous Cell Carcinoma Progression by Activating PHF14 Transcription and Wnt/Β-Catenin Signaling. SP4通过激活PHF14转录和Wnt/β-儿茶素信号传导促进食管鳞状细胞癌的进展。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-01-02 DOI: 10.1158/1541-7786.MCR-22-0835
Li Wei, Chaowei Deng, Bo Zhang, Guanghui Wang, Yan Meng, Hao Qin

Specificity protein 4 transcription factor (SP4), a member of the Sp/Krüppel-like family (KLF), could bind to GT and GC box promoters, and plays an essential role in transcriptional activating. Despite SP4 having been detected to be highly expressed in a variety of human tumors, its biological effect and underlying molecular mechanism in esophageal squamous cell carcinoma (ESCC) remains unclear. Our research discovered that high SP4 expression is detected in primary ESCC specimens and cell lines and is strongly associated with the ESCC tumor grade and poor prognosis. In vitro, knockdown of SP4 suppressed cell proliferation and cell-cycle progression and promoted apoptosis, whereas overexpression of SP4 did the opposite. In vivo, inhibiting SP4 expression in ESCC cells suppresses tumor growth. Subsequently, we demonstrated that SP4 acts as the transcriptional upstream of PHF14, which binds to PHF14 promoter region, thus promoting PHF14 transcription. PHF14 was also significantly expressed in patient tissues and various ESCC cell lines and its expression promoted cell proliferation and inhibited apoptosis. Moreover, knockdown of SP4 inhibited the Wnt/β-catenin signaling pathway, whereas overexpression of PHF14 eliminated the effects of SP4 knockdown in ESCC cells. These results demonstrate that SP4 activates the Wnt/β-catenin signaling pathway by driving PHF14 transcription, thereby promoting ESCC progression, which indicates that SP4 might act as a prospective prognostic indicator or therapeutic target for patients with ESCC.

Implications: This study identified SP4/PH14 axis as a new mechanism to promote the progression of ESCC, which may serve as a novel therapeutic target for patients with ESCC.

特异性蛋白4转录因子(SP4)是Sp/Krüppel样家族(KLF)的一员,可与GT和GC盒启动子结合,在转录激活中发挥重要作用。尽管SP4已被检测在多种人类肿瘤中高表达,但其在食管鳞状细胞癌(ESCC)中的生物学作用和潜在的分子机制尚不清楚。我们的研究发现,在原发性ESCC标本和细胞系中检测到高SP4表达,并且与ESCC肿瘤分级和不良预后密切相关。在体外,敲低SP4抑制细胞增殖和细胞周期进展并促进细胞凋亡,而过表达SP4则相反。在体内,抑制ESCC细胞中SP4的表达抑制肿瘤生长。随后,我们证明SP4作为PHF14的转录上游,与PHF14启动子区结合,从而促进PHF14转录。PHF14也在患者组织和各种ESCC细胞系中显著表达,其表达促进细胞增殖并抑制细胞凋亡。此外,敲低SP4可抑制Wnt/β-catenin信号通路,而过表达PHF14可消除ESCC细胞中SP4敲低的影响。这些结果表明,SP4通过驱动PHF14转录激活Wnt/β-catenin信号通路,从而促进ESCC的进展,这表明SP4可能是ESCC患者的前瞻性预后指标或治疗靶点。意义:本研究确定SP4/PH14轴是促进ESCC进展的一种新机制,可作为ESCC患者的新治疗靶点。
{"title":"SP4 Facilitates Esophageal Squamous Cell Carcinoma Progression by Activating PHF14 Transcription and Wnt/Β-Catenin Signaling.","authors":"Li Wei, Chaowei Deng, Bo Zhang, Guanghui Wang, Yan Meng, Hao Qin","doi":"10.1158/1541-7786.MCR-22-0835","DOIUrl":"10.1158/1541-7786.MCR-22-0835","url":null,"abstract":"<p><p>Specificity protein 4 transcription factor (SP4), a member of the Sp/Krüppel-like family (KLF), could bind to GT and GC box promoters, and plays an essential role in transcriptional activating. Despite SP4 having been detected to be highly expressed in a variety of human tumors, its biological effect and underlying molecular mechanism in esophageal squamous cell carcinoma (ESCC) remains unclear. Our research discovered that high SP4 expression is detected in primary ESCC specimens and cell lines and is strongly associated with the ESCC tumor grade and poor prognosis. In vitro, knockdown of SP4 suppressed cell proliferation and cell-cycle progression and promoted apoptosis, whereas overexpression of SP4 did the opposite. In vivo, inhibiting SP4 expression in ESCC cells suppresses tumor growth. Subsequently, we demonstrated that SP4 acts as the transcriptional upstream of PHF14, which binds to PHF14 promoter region, thus promoting PHF14 transcription. PHF14 was also significantly expressed in patient tissues and various ESCC cell lines and its expression promoted cell proliferation and inhibited apoptosis. Moreover, knockdown of SP4 inhibited the Wnt/β-catenin signaling pathway, whereas overexpression of PHF14 eliminated the effects of SP4 knockdown in ESCC cells. These results demonstrate that SP4 activates the Wnt/β-catenin signaling pathway by driving PHF14 transcription, thereby promoting ESCC progression, which indicates that SP4 might act as a prospective prognostic indicator or therapeutic target for patients with ESCC.</p><p><strong>Implications: </strong>This study identified SP4/PH14 axis as a new mechanism to promote the progression of ESCC, which may serve as a novel therapeutic target for patients with ESCC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"55-69"},"PeriodicalIF":4.1,"publicationDate":"2024-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10758695/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41143506","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
RNF185 Control of COL3A1 Expression Limits Prostate Cancer Migration and Metastatic Potential. RNF185对COL3A1表达的控制限制了前列腺癌症的迁移和转移潜力。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-01-02 DOI: 10.1158/1541-7786.MCR-23-0512
Benjamin Van Espen, Htoo Zarni Oo, Colin Collins, Ladan Fazli, Alfredo Molinolo, Kevin Yip, Rabi Murad, Martin Gleave, Ze'ev A Ronai

RNF185 is a RING finger domain-containing ubiquitin ligase implicated in ER-associated degradation. Prostate tumor patient data analysis revealed a negative correlation between RNF185 expression and prostate cancer progression and metastasis. Likewise, several prostate cancer cell lines exhibited greater migration and invasion capabilities in culture upon RNF185 depletion. Subcutaneous inoculation of mouse prostate cancer MPC3 cells stably expressing short hairpin RNA against RNF185 into mice resulted in larger tumors and more frequent lung metastases. RNA-sequencing and Ingenuity Pathway Analysis identified wound-healing and cellular movement among the most significant pathways upregulated in RNF185-depleted lines, compared with control prostate cancer cells. Gene Set Enrichment Analyses performed in samples from patients harboring low RNF185 expression and in RNF185-depleted lines confirmed the deregulation of genes implicated in epithelial-to-mesenchymal transition. Among those, COL3A1 was identified as the primary mediator of RNF185's ability to impact migration phenotypes. Correspondingly, enhanced migration and metastasis of RNF185 knockdown (KD) prostate cancer cells were attenuated upon co-inhibition of COL3A1. Our results identify RNF185 as a gatekeeper of prostate cancer metastasis, partly via its control of COL3A1 availability.

Implications: RNF185 is identified as an important regulator of prostate cancer migration and metastasis, in part due to its regulation of COL3A1. Both RNF185 and COL3A1 may serve as novel markers for prostate tumors.

RNF185是一个含有泛素连接酶的环指结构域,与ER相关的降解有关。前列腺肿瘤患者数据分析显示RNF185表达与前列腺癌症进展和转移呈负相关。同样,几种前列腺癌症细胞系在RNF185缺失时在培养中表现出更大的迁移和侵袭能力。将稳定表达针对RNF185的shRNA的小鼠前列腺癌症MPC3细胞皮下接种到小鼠中导致更大的肿瘤和更频繁的肺转移。RNA序列和摄入途径分析确定,与对照前列腺癌症细胞相比,在RNF185缺失的细胞中,伤口愈合和细胞运动是上调的最重要途径。对携带低RNF185表达的患者的样本和RNF185缺失的品系进行的基因集富集分析证实了EMT相关基因的失调。其中,COL3A1被鉴定为RNF185影响迁移表型能力的主要介质。相应地,在COL3A1的共同抑制下,RNF185KD前列腺癌症细胞增强的迁移和转移减弱。我们的研究结果确定RNF185是前列腺癌症转移的看门人,部分是通过其对COL3A1可用性的控制。意义:通过控制COL3A1的表达,RNF185被鉴定为前列腺癌症转移的新标志物。
{"title":"RNF185 Control of COL3A1 Expression Limits Prostate Cancer Migration and Metastatic Potential.","authors":"Benjamin Van Espen, Htoo Zarni Oo, Colin Collins, Ladan Fazli, Alfredo Molinolo, Kevin Yip, Rabi Murad, Martin Gleave, Ze'ev A Ronai","doi":"10.1158/1541-7786.MCR-23-0512","DOIUrl":"10.1158/1541-7786.MCR-23-0512","url":null,"abstract":"<p><p>RNF185 is a RING finger domain-containing ubiquitin ligase implicated in ER-associated degradation. Prostate tumor patient data analysis revealed a negative correlation between RNF185 expression and prostate cancer progression and metastasis. Likewise, several prostate cancer cell lines exhibited greater migration and invasion capabilities in culture upon RNF185 depletion. Subcutaneous inoculation of mouse prostate cancer MPC3 cells stably expressing short hairpin RNA against RNF185 into mice resulted in larger tumors and more frequent lung metastases. RNA-sequencing and Ingenuity Pathway Analysis identified wound-healing and cellular movement among the most significant pathways upregulated in RNF185-depleted lines, compared with control prostate cancer cells. Gene Set Enrichment Analyses performed in samples from patients harboring low RNF185 expression and in RNF185-depleted lines confirmed the deregulation of genes implicated in epithelial-to-mesenchymal transition. Among those, COL3A1 was identified as the primary mediator of RNF185's ability to impact migration phenotypes. Correspondingly, enhanced migration and metastasis of RNF185 knockdown (KD) prostate cancer cells were attenuated upon co-inhibition of COL3A1. Our results identify RNF185 as a gatekeeper of prostate cancer metastasis, partly via its control of COL3A1 availability.</p><p><strong>Implications: </strong>RNF185 is identified as an important regulator of prostate cancer migration and metastasis, in part due to its regulation of COL3A1. Both RNF185 and COL3A1 may serve as novel markers for prostate tumors.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"41-54"},"PeriodicalIF":4.1,"publicationDate":"2024-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10841372/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41207069","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
PRL2 Phosphatase Promotes Oncogenic KIT Signaling in Leukemia Cells through Modulating CBL Phosphorylation. PRL2磷酸酶通过调节CBL磷酸化促进白血病细胞中致癌KIT信号传导。
IF 4.1 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-01-02 DOI: 10.1158/1541-7786.MCR-23-0115
Hongxia Chen, Yunpeng Bai, Michihiro Kobayashi, Shiyu Xiao, Sergio Barajas, Wenjie Cai, Sisi Chen, Jinmin Miao, Frederick Nguele Meke, Chonghua Yao, Yuxia Yang, Katherine Strube, Odelia Satchivi, Jianmin Sun, Lars Rönnstrand, James M Croop, H Scott Boswell, Yuzhi Jia, Huiping Liu, Loretta S Li, Jessica K Altman, Elizabeth A Eklund, Madina Sukhanova, Peng Ji, Wei Tong, Hamid Band, Danny T Huang, Leonidas C Platanias, Zhong-Yin Zhang, Yan Liu

Receptor tyrosine kinase KIT is frequently activated in acute myeloid leukemia (AML). While high PRL2 (PTP4A2) expression is correlated with activation of SCF/KIT signaling in AML, the underlying mechanisms are not fully understood. We discovered that inhibition of PRL2 significantly reduces the burden of oncogenic KIT-driven leukemia and extends leukemic mice survival. PRL2 enhances oncogenic KIT signaling in leukemia cells, promoting their proliferation and survival. We found that PRL2 dephosphorylates CBL at tyrosine 371 and inhibits its activity toward KIT, leading to decreased KIT ubiquitination and enhanced AKT and ERK signaling in leukemia cells.

Implications: Our studies uncover a novel mechanism that fine-tunes oncogenic KIT signaling in leukemia cells and will likely identify PRL2 as a novel therapeutic target in AML with KIT mutations.

受体酪氨酸激酶KIT在急性髓细胞白血病(AML)中经常被激活。虽然PRL2(PTP4A2)的高表达与AML中SCF/KIT信号的激活相关,但其潜在机制尚不完全清楚。我们发现PRL2的抑制显著降低了致癌KIT驱动的白血病的负担,并延长了白血病小鼠的生存期。PRL2增强白血病细胞中致癌KIT信号传导,促进其增殖和存活。我们发现PRL2在酪氨酸371处使CBL去磷酸化,并抑制其对KIT的活性,导致白血病细胞中KIT泛素化减少,AKT和ERK信号增强。启示:我们的研究揭示了一种新的机制,可以微调白血病细胞中的致癌KIT信号,并可能将PRL2确定为具有KIT突变的AML的新治疗靶点。
{"title":"PRL2 Phosphatase Promotes Oncogenic KIT Signaling in Leukemia Cells through Modulating CBL Phosphorylation.","authors":"Hongxia Chen, Yunpeng Bai, Michihiro Kobayashi, Shiyu Xiao, Sergio Barajas, Wenjie Cai, Sisi Chen, Jinmin Miao, Frederick Nguele Meke, Chonghua Yao, Yuxia Yang, Katherine Strube, Odelia Satchivi, Jianmin Sun, Lars Rönnstrand, James M Croop, H Scott Boswell, Yuzhi Jia, Huiping Liu, Loretta S Li, Jessica K Altman, Elizabeth A Eklund, Madina Sukhanova, Peng Ji, Wei Tong, Hamid Band, Danny T Huang, Leonidas C Platanias, Zhong-Yin Zhang, Yan Liu","doi":"10.1158/1541-7786.MCR-23-0115","DOIUrl":"10.1158/1541-7786.MCR-23-0115","url":null,"abstract":"<p><p>Receptor tyrosine kinase KIT is frequently activated in acute myeloid leukemia (AML). While high PRL2 (PTP4A2) expression is correlated with activation of SCF/KIT signaling in AML, the underlying mechanisms are not fully understood. We discovered that inhibition of PRL2 significantly reduces the burden of oncogenic KIT-driven leukemia and extends leukemic mice survival. PRL2 enhances oncogenic KIT signaling in leukemia cells, promoting their proliferation and survival. We found that PRL2 dephosphorylates CBL at tyrosine 371 and inhibits its activity toward KIT, leading to decreased KIT ubiquitination and enhanced AKT and ERK signaling in leukemia cells.</p><p><strong>Implications: </strong>Our studies uncover a novel mechanism that fine-tunes oncogenic KIT signaling in leukemia cells and will likely identify PRL2 as a novel therapeutic target in AML with KIT mutations.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"94-103"},"PeriodicalIF":4.1,"publicationDate":"2024-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10841656/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41128364","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
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