首页 > 最新文献

Oncogene最新文献

英文 中文
Correction: Editorial Expression of Concern: Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer 更正:编辑表达的关切:通过去甲基化或基因转移重新表达 caspase-8,对死亡受体或药物诱导的细胞凋亡敏感。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-23 DOI: 10.1038/s41388-024-03204-z
Simone Fulda, Martin U. Kuüfer, Eric Meyer, Frans van Valen, Barbara Dockhorn-Dworniczak, Klaus-Michael Debatin
{"title":"Correction: Editorial Expression of Concern: Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer","authors":"Simone Fulda, Martin U. Kuüfer, Eric Meyer, Frans van Valen, Barbara Dockhorn-Dworniczak, Klaus-Michael Debatin","doi":"10.1038/s41388-024-03204-z","DOIUrl":"10.1038/s41388-024-03204-z","url":null,"abstract":"","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"43 46","pages":"3417-3417"},"PeriodicalIF":6.9,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41388-024-03204-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505250","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
USP10 drives cancer stemness and enables super-competitor signalling in colorectal cancer. USP10 在结直肠癌中驱动癌症干性并实现超级竞争信号传导。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-23 DOI: 10.1038/s41388-024-03141-x
Michaela Reissland, Oliver Hartmann, Saskia Tauch, Jeroen M Bugter, Cristian Prieto-Garcia, Clemens Schulte, Sinah Loebbert, Daniel Solvie, Eliya Bitman-Lotan, Ashwin Narain, Anne-Claire Jacomin, Christina Schuelein-Voelk, Carmina T Fuss, Nikolett Pahor, Carsten Ade, Viktoria Buck, Michael Potente, Vivian Li, Gerti Beliu, Armin Wiegering, Tom Grossmann, Martin Eilers, Elmar Wolf, Hans Maric, Mathias Rosenfeldt, Madelon M Maurice, Ivan Dikic, Peter Gallant, Amir Orian, Markus E Diefenbacher

The contribution of deubiquitylating enzymes (DUBs) to β-Catenin stabilization in intestinal stem cells and colorectal cancer (CRC) is poorly understood. Here, and by using an unbiassed screen, we discovered that the DUB USP10 stabilizes β-Catenin specifically in APC-truncated CRC in vitro and in vivo. Mechanistic studies, including in vitro binding together with computational modelling, revealed that USP10 binding to β-Catenin is mediated via the unstructured N-terminus of USP10 and is outcompeted by intact APC, favouring β-catenin degradation. However, in APC-truncated cancer cells USP10 binds to β-catenin, increasing its stability which is critical for maintaining an undifferentiated tumour identity. Elimination of USP10 reduces the expression of WNT and stem cell signatures and induces the expression of differentiation genes. Remarkably, silencing of USP10 in murine and patient-derived CRC organoids established that it is essential for NOTUM signalling and the APC super competitor-phenotype, reducing tumorigenic properties of APC-truncated CRC. These findings are clinically relevant as patient-derived organoids are highly dependent on USP10, and abundance of USP10 correlates with poorer prognosis of CRC patients. Our findings reveal, therefore, a role for USP10 in CRC cell identity, stemness, and tumorigenic growth by stabilising β-Catenin, leading to aberrant WNT signalling and degradation resistant tumours. Thus, USP10 emerges as a unique therapeutic target in APC truncated CRC.

人们对去泛素化酶(DUBs)在肠干细胞和结直肠癌(CRC)中稳定β-Catenin的作用知之甚少。在这里,通过无偏见筛选,我们发现DUB USP10能在体外和体内特异性地稳定APC截断的CRC中的β-Catenin。包括体外结合和计算建模在内的机理研究显示,USP10 与 β-Catenin 的结合是通过 USP10 的非结构化 N 端介导的,并被完整的 APC 竞争,从而有利于 β-catenin 降解。然而,在APC截断的癌细胞中,USP10会与β-catenin结合,增加其稳定性,这对维持未分化肿瘤特性至关重要。消除 USP10 会减少 WNT 和干细胞特征的表达,并诱导分化基因的表达。值得注意的是,在小鼠和患者来源的 CRC 有机体中沉默 USP10 证实,它对 NOTUM 信号和 APC 超级竞争者表型至关重要,可降低 APC 截断 CRC 的致瘤特性。这些发现与临床相关,因为患者衍生的器官组织高度依赖于 USP10,而 USP10 的丰度与 CRC 患者较差的预后相关。因此,我们的研究结果揭示了 USP10 在 CRC 细胞特性、干性和肿瘤性生长中的作用,它通过稳定 β-Catenin,导致 WNT 信号异常和抗降解肿瘤。因此,USP10 成为 APC 截断型 CRC 的独特治疗靶点。
{"title":"USP10 drives cancer stemness and enables super-competitor signalling in colorectal cancer.","authors":"Michaela Reissland, Oliver Hartmann, Saskia Tauch, Jeroen M Bugter, Cristian Prieto-Garcia, Clemens Schulte, Sinah Loebbert, Daniel Solvie, Eliya Bitman-Lotan, Ashwin Narain, Anne-Claire Jacomin, Christina Schuelein-Voelk, Carmina T Fuss, Nikolett Pahor, Carsten Ade, Viktoria Buck, Michael Potente, Vivian Li, Gerti Beliu, Armin Wiegering, Tom Grossmann, Martin Eilers, Elmar Wolf, Hans Maric, Mathias Rosenfeldt, Madelon M Maurice, Ivan Dikic, Peter Gallant, Amir Orian, Markus E Diefenbacher","doi":"10.1038/s41388-024-03141-x","DOIUrl":"https://doi.org/10.1038/s41388-024-03141-x","url":null,"abstract":"<p><p>The contribution of deubiquitylating enzymes (DUBs) to β-Catenin stabilization in intestinal stem cells and colorectal cancer (CRC) is poorly understood. Here, and by using an unbiassed screen, we discovered that the DUB USP10 stabilizes β-Catenin specifically in APC-truncated CRC in vitro and in vivo. Mechanistic studies, including in vitro binding together with computational modelling, revealed that USP10 binding to β-Catenin is mediated via the unstructured N-terminus of USP10 and is outcompeted by intact APC, favouring β-catenin degradation. However, in APC-truncated cancer cells USP10 binds to β-catenin, increasing its stability which is critical for maintaining an undifferentiated tumour identity. Elimination of USP10 reduces the expression of WNT and stem cell signatures and induces the expression of differentiation genes. Remarkably, silencing of USP10 in murine and patient-derived CRC organoids established that it is essential for NOTUM signalling and the APC super competitor-phenotype, reducing tumorigenic properties of APC-truncated CRC. These findings are clinically relevant as patient-derived organoids are highly dependent on USP10, and abundance of USP10 correlates with poorer prognosis of CRC patients. Our findings reveal, therefore, a role for USP10 in CRC cell identity, stemness, and tumorigenic growth by stabilising β-Catenin, leading to aberrant WNT signalling and degradation resistant tumours. Thus, USP10 emerges as a unique therapeutic target in APC truncated CRC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
TMSB4Y restrains sphingomyelin synthesis via de novo purine synthesis to exert a tumor suppressor function in male esophageal squamous cell carcinoma. TMSB4Y 通过新生嘌呤合成抑制鞘磷脂合成,从而在男性食管鳞状细胞癌中发挥肿瘤抑制功能。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-23 DOI: 10.1038/s41388-024-03193-z
Tongyang Gong, Wanyuan Sun, Xukun Li, Jiahui Cai, Ning Zhao, Minyi Lu, Juan Xu, Zhihua Liu, Hongyan Chen

Y chromosome genes play a vital role in sex difference of cancer. The dysregulation and functional implications of Y chromosome genes in esophageal squamous cell carcinoma (ESCC) remains elusive. Here, we analyze the Y chromosome gene signature and identify TMSB4Y as an emerging prognostic predictor in male ESCC. Functional analyses show that TMSB4Y inhibits the proliferation, invasion and metastasis of male ESCC cells. Mechanistically, we demonstrate that TMSB4Y interacts with PAICS, wherein TMSB4Y disrupts the formation of the PAICS octamer to inhibit purine de novo synthesis, leading to a decrease in the AMP/ATP ratio, subsequently impeding AMPK phosphorylation. Furthermore, we uncover a regulatory cascade orchestrated by the TMSB4Y/PAICS-AMPK axis, which exerts a suppressive effect on sphingomyelin metabolism by inhibiting the expression of sphingomyelin synthases (SMSs). Notably, Malabaricone C, an inhibitor of SMS1 and SMS2, effectively suppresses male ESCC cell proliferation and xenograft tumor growth. Collectively, these findings reveal the regulation of sphingomyelin metabolism by TMSB4Y/PAICS-AMPK axis and underscore the potential of targeting SMSs as a promising therapeutic approach for the treatment of male ESCC.

Y 染色体基因在癌症的性别差异中起着至关重要的作用。Y染色体基因在食管鳞状细胞癌(ESCC)中的失调和功能性影响仍未被发现。在这里,我们分析了 Y 染色体基因特征,并发现 TMSB4Y 是男性 ESCC 的一个新兴预后预测因子。功能分析显示,TMSB4Y 能抑制男性 ESCC 细胞的增殖、侵袭和转移。从机理上讲,我们证明了 TMSB4Y 与 PAICS 相互作用,TMSB4Y 会破坏 PAICS 八聚体的形成,从而抑制嘌呤的新合成,导致 AMP/ATP 比率下降,进而阻碍 AMPK 磷酸化。此外,我们还发现了一个由 TMSB4Y/PAICS-AMPK 轴协调的调控级联,它通过抑制鞘磷脂合成酶(SMSs)的表达,对鞘磷脂代谢产生抑制作用。值得注意的是,SMS1 和 SMS2 的抑制剂 Malabaricone C 能有效抑制男性 ESCC 细胞的增殖和异种移植肿瘤的生长。总之,这些发现揭示了TMSB4Y/PAICS-AMPK轴对鞘磷脂代谢的调控,并强调了靶向SMSs作为治疗男性ESCC的一种有前景的治疗方法的潜力。
{"title":"TMSB4Y restrains sphingomyelin synthesis via de novo purine synthesis to exert a tumor suppressor function in male esophageal squamous cell carcinoma.","authors":"Tongyang Gong, Wanyuan Sun, Xukun Li, Jiahui Cai, Ning Zhao, Minyi Lu, Juan Xu, Zhihua Liu, Hongyan Chen","doi":"10.1038/s41388-024-03193-z","DOIUrl":"https://doi.org/10.1038/s41388-024-03193-z","url":null,"abstract":"<p><p>Y chromosome genes play a vital role in sex difference of cancer. The dysregulation and functional implications of Y chromosome genes in esophageal squamous cell carcinoma (ESCC) remains elusive. Here, we analyze the Y chromosome gene signature and identify TMSB4Y as an emerging prognostic predictor in male ESCC. Functional analyses show that TMSB4Y inhibits the proliferation, invasion and metastasis of male ESCC cells. Mechanistically, we demonstrate that TMSB4Y interacts with PAICS, wherein TMSB4Y disrupts the formation of the PAICS octamer to inhibit purine de novo synthesis, leading to a decrease in the AMP/ATP ratio, subsequently impeding AMPK phosphorylation. Furthermore, we uncover a regulatory cascade orchestrated by the TMSB4Y/PAICS-AMPK axis, which exerts a suppressive effect on sphingomyelin metabolism by inhibiting the expression of sphingomyelin synthases (SMSs). Notably, Malabaricone C, an inhibitor of SMS1 and SMS2, effectively suppresses male ESCC cell proliferation and xenograft tumor growth. Collectively, these findings reveal the regulation of sphingomyelin metabolism by TMSB4Y/PAICS-AMPK axis and underscore the potential of targeting SMSs as a promising therapeutic approach for the treatment of male ESCC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505254","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
PTPRZ1 dephosphorylates and stabilizes RNF26 to reduce the efficacy of TKIs and PD-1 blockade in ccRCC. PTPRZ1 可使 RNF26 去磷酸化并使其稳定,从而降低 TKIs 和 PD-1 阻断剂在 ccRCC 中的疗效。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-23 DOI: 10.1038/s41388-024-03198-8
Yongkang Ma, Wei Li, Xinlin Liu, Weilin Peng, Bei Qing, Shangqing Ren, Wentao Liu, Xiaobing Chen

Clear cell renal cell carcinoma (ccRCC), the most common subtype of renal cell carcinoma, often exhibits resistance to tyrosine kinase inhibitors (TKIs) when used as monotherapy. However, the integration of PD-1 blockade with TKIs has significantly improved patient survival, making it a leading therapeutic strategy for ccRCC. Despite these advancements, the efficacy of this combined therapy remains suboptimal, necessitating a deeper understanding of the underlying regulatory mechanisms. Through comprehensive analyses, including mass spectrometry, RNA sequencing, lipidomic profiling, immunohistochemical staining, and ex vivo experiments, we explored the interaction between PTPRZ1 and RNF26 and its impact on ccRCC cell behavior. Our results revealed a unique interaction where PTPRZ1 stabilized RNF26 protein expression by dephosphorylating it at the Y432 site. The modulation of RNF26 levels by PTPRZ1 was found to be mediated through the proteasome pathway. Additionally, PTPRZ1, via its interaction with RNF26, activated the TNF/NF-κB signaling pathway, thereby promoting cell proliferation, angiogenesis, and lipid metabolism in ccRCC cells. Importantly, inhibiting PTPRZ1 enhanced the sensitivity of ccRCC to TKIs and PD-1 blockade, an effect that was attenuated when RNF26 was simultaneously knocked down. These findings highlight the critical role of the PTPRZ1-RNF26 axis in ccRCC and suggest that combining PTPRZ1 inhibitors with current TKIs and PD-1 blockade therapies could significantly improve treatment outcomes for ccRCC patients.

透明细胞肾细胞癌(ccRCC)是肾细胞癌中最常见的亚型,在单药治疗时通常会对酪氨酸激酶抑制剂(TKIs)产生耐药性。然而,将 PD-1 阻断与 TKIs 结合使用可显著提高患者生存率,使其成为 ccRCC 的主要治疗策略。尽管取得了这些进展,但这种联合疗法的疗效仍不理想,因此有必要深入了解其潜在的调控机制。通过质谱分析、RNA测序、脂质组分析、免疫组化染色和体内外实验等综合分析,我们探索了PTPRZ1和RNF26之间的相互作用及其对ccRCC细胞行为的影响。我们的结果发现了一种独特的相互作用,即 PTPRZ1 通过在 Y432 位点使 RNF26 蛋白去磷酸化,从而稳定 RNF26 蛋白的表达。PTPRZ1对RNF26水平的调节是通过蛋白酶体途径介导的。此外,PTPRZ1通过与RNF26相互作用,激活了TNF/NF-κB信号通路,从而促进了ccRCC细胞的增殖、血管生成和脂质代谢。重要的是,抑制 PTPRZ1 会增强 ccRCC 对 TKIs 和 PD-1 阻断的敏感性,而同时敲除 RNF26 会减弱这种效应。这些发现凸显了PTPRZ1-RNF26轴在ccRCC中的关键作用,并表明将PTPRZ1抑制剂与目前的TKIs和PD-1阻断疗法相结合可显著改善ccRCC患者的治疗效果。
{"title":"PTPRZ1 dephosphorylates and stabilizes RNF26 to reduce the efficacy of TKIs and PD-1 blockade in ccRCC.","authors":"Yongkang Ma, Wei Li, Xinlin Liu, Weilin Peng, Bei Qing, Shangqing Ren, Wentao Liu, Xiaobing Chen","doi":"10.1038/s41388-024-03198-8","DOIUrl":"https://doi.org/10.1038/s41388-024-03198-8","url":null,"abstract":"<p><p>Clear cell renal cell carcinoma (ccRCC), the most common subtype of renal cell carcinoma, often exhibits resistance to tyrosine kinase inhibitors (TKIs) when used as monotherapy. However, the integration of PD-1 blockade with TKIs has significantly improved patient survival, making it a leading therapeutic strategy for ccRCC. Despite these advancements, the efficacy of this combined therapy remains suboptimal, necessitating a deeper understanding of the underlying regulatory mechanisms. Through comprehensive analyses, including mass spectrometry, RNA sequencing, lipidomic profiling, immunohistochemical staining, and ex vivo experiments, we explored the interaction between PTPRZ1 and RNF26 and its impact on ccRCC cell behavior. Our results revealed a unique interaction where PTPRZ1 stabilized RNF26 protein expression by dephosphorylating it at the Y432 site. The modulation of RNF26 levels by PTPRZ1 was found to be mediated through the proteasome pathway. Additionally, PTPRZ1, via its interaction with RNF26, activated the TNF/NF-κB signaling pathway, thereby promoting cell proliferation, angiogenesis, and lipid metabolism in ccRCC cells. Importantly, inhibiting PTPRZ1 enhanced the sensitivity of ccRCC to TKIs and PD-1 blockade, an effect that was attenuated when RNF26 was simultaneously knocked down. These findings highlight the critical role of the PTPRZ1-RNF26 axis in ccRCC and suggest that combining PTPRZ1 inhibitors with current TKIs and PD-1 blockade therapies could significantly improve treatment outcomes for ccRCC patients.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
KRAS-mediated upregulation of CIP2A promotes suppression of PP2A-B56α to initiate pancreatic cancer development. KRAS 介导的 CIP2A 上调促进了 PP2A-B56α 的抑制,从而启动了胰腺癌的发展。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-23 DOI: 10.1038/s41388-024-03196-w
Samantha L Tinsley, Ella Rose D Chianis, Rebecca A Shelley, Gaganpreet K Mall, Alisha Dhiman, Garima Baral, Harish Kothandaraman, Mary C Thoma, Isabel A English, Colin J Daniel, Luis Carlos Sanjuan Acosta, Luis Solorio, Nadia Atallah Lanman, Marina Pasca di Magliano, Goutham Narla, Emily C Dykhuizen, Rosalie C Sears, Brittany L Allen-Petersen

Oncogenic mutations in KRAS are present in ~95% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) and are considered the initiating event of pancreatic intraepithelial neoplasia (PanIN) precursor lesions. While it is well established that KRAS mutations drive the activation of oncogenic kinase cascades during pancreatic oncogenesis, the effects of oncogenic KRAS signaling on regulation of phosphatases during this process is not fully appreciated. Protein Phosphatase 2A (PP2A) has been implicated in suppressing KRAS-driven cellular transformation and low PP2A activity is observed in PDAC cells compared to non-transformed cells, suggesting that suppression of PP2A activity is an important step in the overall development of PDAC. In the current study, we demonstrate that KRASG12D induces the expression of an endogenous inhibitor of PP2A activity, Cancerous Inhibitor of PP2A (CIP2A), and phosphorylation of the PP2A substrate, c-MYC. Consistent with these findings, KRASG12D sequestered the specific PP2A subunit responsible for c-MYC degradation, B56α, away from the active PP2A holoenzyme in a CIP2A-dependent manner. During PDAC initiation in vivo, knockout of B56α promoted KRASG12D tumorigenesis by accelerating acinar-to-ductal metaplasia (ADM) and the formation of PanIN lesions. The process of ADM was attenuated ex vivo in response to pharmacological re-activation of PP2A utilizing direct small molecule activators of PP2A (SMAPs). Together, our results suggest that suppression of PP2A-B56α through KRAS signaling can promote the MYC-driven initiation of pancreatic tumorigenesis.

在确诊为胰腺导管腺癌(PDAC)的患者中,约 95% 存在 KRAS 的致癌突变,这被认为是胰腺上皮内瘤变(PanIN)前驱病变的起始事件。KRAS突变在胰腺肿瘤发生过程中驱动激活致癌激酶级联,这一点已得到公认,但在这一过程中,致癌KRAS信号对磷酸酶调控的影响尚未得到充分认识。蛋白磷酸酶 2A(PP2A)与抑制 KRAS 驱动的细胞转化有关,与未转化细胞相比,PDAC 细胞中的 PP2A 活性较低,这表明 PP2A 活性的抑制是 PDAC 整体发展过程中的一个重要步骤。在目前的研究中,我们证明了 KRASG12D 会诱导 PP2A 活性的内源性抑制剂--PP2A 癌症抑制剂(CIP2A)的表达,以及 PP2A 底物 c-MYC 的磷酸化。与这些发现一致的是,KRASG12D以一种依赖于CIP2A的方式将负责c-MYC降解的特定PP2A亚基B56α从活性PP2A全酶中分离出来。在体内PDAC的发病过程中,B56α的敲除通过加速渐变(ADM)和PanIN病变的形成,促进了KRASG12D的肿瘤发生。利用 PP2A 的直接小分子激活剂(SMAPs)对 PP2A 进行药理再激活后,体内 ADM 的进程会减弱。我们的研究结果表明,通过 KRAS 信号抑制 PP2A-B56α 可促进 MYC 驱动的胰腺肿瘤发生。
{"title":"KRAS-mediated upregulation of CIP2A promotes suppression of PP2A-B56α to initiate pancreatic cancer development.","authors":"Samantha L Tinsley, Ella Rose D Chianis, Rebecca A Shelley, Gaganpreet K Mall, Alisha Dhiman, Garima Baral, Harish Kothandaraman, Mary C Thoma, Isabel A English, Colin J Daniel, Luis Carlos Sanjuan Acosta, Luis Solorio, Nadia Atallah Lanman, Marina Pasca di Magliano, Goutham Narla, Emily C Dykhuizen, Rosalie C Sears, Brittany L Allen-Petersen","doi":"10.1038/s41388-024-03196-w","DOIUrl":"10.1038/s41388-024-03196-w","url":null,"abstract":"<p><p>Oncogenic mutations in KRAS are present in ~95% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) and are considered the initiating event of pancreatic intraepithelial neoplasia (PanIN) precursor lesions. While it is well established that KRAS mutations drive the activation of oncogenic kinase cascades during pancreatic oncogenesis, the effects of oncogenic KRAS signaling on regulation of phosphatases during this process is not fully appreciated. Protein Phosphatase 2A (PP2A) has been implicated in suppressing KRAS-driven cellular transformation and low PP2A activity is observed in PDAC cells compared to non-transformed cells, suggesting that suppression of PP2A activity is an important step in the overall development of PDAC. In the current study, we demonstrate that KRAS<sup>G12D</sup> induces the expression of an endogenous inhibitor of PP2A activity, Cancerous Inhibitor of PP2A (CIP2A), and phosphorylation of the PP2A substrate, c-MYC. Consistent with these findings, KRAS<sup>G12D</sup> sequestered the specific PP2A subunit responsible for c-MYC degradation, B56α, away from the active PP2A holoenzyme in a CIP2A-dependent manner. During PDAC initiation in vivo, knockout of B56α promoted KRAS<sup>G12D</sup> tumorigenesis by accelerating acinar-to-ductal metaplasia (ADM) and the formation of PanIN lesions. The process of ADM was attenuated ex vivo in response to pharmacological re-activation of PP2A utilizing direct small molecule activators of PP2A (SMAPs). Together, our results suggest that suppression of PP2A-B56α through KRAS signaling can promote the MYC-driven initiation of pancreatic tumorigenesis.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Zinc ions activate AKT and promote prostate cancer cell proliferation via disrupting AKT intramolecular interaction. 锌离子通过破坏 AKT 分子内相互作用,激活 AKT 并促进前列腺癌细胞增殖。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-23 DOI: 10.1038/s41388-024-03195-x
Kangjunjie Wang, Min Chen, Shukun Yan, Ying Han, Huairui Yuan, Qiuli Liu, Dayun Lu, Long Li, Kaihua Wang, Fen Liu, Qianqian Li, Dakui Luo, Jun Jiang, Hu Zhou, Yong Chen, Jun Qin, Daming Gao

Prostate is a zinc rich organ and the physiological function of the abundant zinc ions is relatively less understood. AKT kinase is a pivotal regulator downstream of cytokines, growth factors and other extracellular stimuli, and the attachment of its PH domain to PtdIns-3,4,5-P3 (PIP3) and the subsequent phosphorylation of its kinase domain by PDPK1 are considered important for its activation. Herein, we report a regulatory mechanism of AKT kinase by zinc ions. Mechanistically, zinc ions directly bind to AKT and facilitate AKT activation through disrupting the interaction between PH and kinase domains within AKT molecule. Consistently, AKT1-H89A/E91A mutant (zinc-binding-deficient) fails to respond to zinc ions and exhibits strong interaction between PH and kinase domains, and it is less oncogenic in orthotopic xenograft model of prostate cancer. On the other hand, the AKT1-W80L mutant with minimum intra-molecular interaction between PH and kinase domains shows strong tumor promoting capacity although it could not be further stimulated by zinc ions. Overall, this study reveals a distinctive regulatory mechanism of AKT activation and implies a tumor promoting role of the zinc ions in prostate cancer.

前列腺是一个富含锌的器官,但人们对其中丰富的锌离子的生理功能了解相对较少。AKT激酶是细胞因子、生长因子和其他细胞外刺激下游的关键调节因子,其PH结构域与PtdIns-3,4,5-P3 (PIP3)的连接以及随后PDPK1对其激酶结构域的磷酸化被认为是其活化的重要因素。在此,我们报告了锌离子对 AKT 激酶的调控机制。从机制上讲,锌离子直接与 AKT 结合,并通过破坏 AKT 分子内 PH 与激酶结构域之间的相互作用促进 AKT 的活化。一致的是,AKT1-H89A/E91A突变体(锌结合缺陷型)不能对锌离子做出反应,并且在PH和激酶结构域之间表现出强烈的相互作用,在前列腺癌的正位异种移植模型中致癌性较低。另一方面,AKT1-W80L突变体的PH和激酶结构域之间的分子内相互作用最小,虽然不能受到锌离子的进一步刺激,但却表现出很强的肿瘤促进能力。总之,这项研究揭示了 AKT 激活的独特调控机制,并暗示锌离子在前列腺癌中的促瘤作用。
{"title":"Zinc ions activate AKT and promote prostate cancer cell proliferation via disrupting AKT intramolecular interaction.","authors":"Kangjunjie Wang, Min Chen, Shukun Yan, Ying Han, Huairui Yuan, Qiuli Liu, Dayun Lu, Long Li, Kaihua Wang, Fen Liu, Qianqian Li, Dakui Luo, Jun Jiang, Hu Zhou, Yong Chen, Jun Qin, Daming Gao","doi":"10.1038/s41388-024-03195-x","DOIUrl":"https://doi.org/10.1038/s41388-024-03195-x","url":null,"abstract":"<p><p>Prostate is a zinc rich organ and the physiological function of the abundant zinc ions is relatively less understood. AKT kinase is a pivotal regulator downstream of cytokines, growth factors and other extracellular stimuli, and the attachment of its PH domain to PtdIns-3,4,5-P3 (PIP3) and the subsequent phosphorylation of its kinase domain by PDPK1 are considered important for its activation. Herein, we report a regulatory mechanism of AKT kinase by zinc ions. Mechanistically, zinc ions directly bind to AKT and facilitate AKT activation through disrupting the interaction between PH and kinase domains within AKT molecule. Consistently, AKT1-H89A/E91A mutant (zinc-binding-deficient) fails to respond to zinc ions and exhibits strong interaction between PH and kinase domains, and it is less oncogenic in orthotopic xenograft model of prostate cancer. On the other hand, the AKT1-W80L mutant with minimum intra-molecular interaction between PH and kinase domains shows strong tumor promoting capacity although it could not be further stimulated by zinc ions. Overall, this study reveals a distinctive regulatory mechanism of AKT activation and implies a tumor promoting role of the zinc ions in prostate cancer.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142505256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
IL-17RA/CTSK axis mediates H. pylori-induced castration-resistant prostate cancer growth. IL-17RA/CTSK轴介导幽门螺杆菌诱导的耐阉割前列腺癌生长
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 DOI: 10.1038/s41388-024-03169-z
Guowen Lin, Feng Tian, Qiwei Yu, Xiaoling Weng, Nanhui Yu, Feng Zhang, Chen Yi, Jian Ye, Dingwei Ye

In this investigation, we explored the molecular dynamics guiding the progression of castration-resistant prostate cancer (CRPC) influenced by Helicobacter pylori (H. pylori)-mediated M2 polarization of macrophages through the IL-17RA/CTSK/EMT axis. An 830-patient clinical trial categorized subjects into hormone-sensitive prostate cancer (HSPC) and CRPC groups. H. pylori infection, evaluated by ELISA, exhibited a higher incidence in CRPC patients, impacting overall survival (OS) and progression-free survival. In-depth in vitro and in vivo experiments, including 16S rDNA sequencing, immunohistochemical tests, and transcriptome analysis, unveiled that H. pylori promotes CRPC growth and metastasis by upregulating IL-17RA and CTSK, leading to enhanced EMT. Notably, M2 macrophages emerged as pivotal immune cells influencing CRPC progression. This study uncovers a novel pathway wherein H. pylori enrichment exacerbates CRPC by inducing macrophage M2 polarization, IL-17RA/CTSK expression, and EMT activation, shedding light on a previously unrecognized mechanism contributing to the growth and metastasis of CRPC.

在这项研究中,我们探讨了幽门螺杆菌(H. pylori)通过IL-17RA/CTSK/EMT轴介导的巨噬细胞M2极化对去势抵抗性前列腺癌(CRPC)进展的分子动力学影响。一项有 830 名患者参加的临床试验将受试者分为激素敏感性前列腺癌 (HSPC) 组和 CRPC 组。通过酶联免疫吸附试验(ELISA)评估,幽门螺杆菌感染在CRPC患者中的发病率较高,影响总生存期(OS)和无进展生存期。深入的体外和体内实验,包括16S rDNA测序、免疫组化检测和转录组分析,揭示了幽门螺杆菌通过上调IL-17RA和CTSK促进CRPC生长和转移,导致EMT增强。值得注意的是,M2巨噬细胞成为影响CRPC进展的关键免疫细胞。这项研究发现了一种新的途径,即幽门螺杆菌富集通过诱导巨噬细胞M2极化、IL-17RA/CTSK表达和EMT活化来加剧CRPC,从而揭示了一种以前未被认识到的导致CRPC生长和转移的机制。
{"title":"IL-17RA/CTSK axis mediates H. pylori-induced castration-resistant prostate cancer growth.","authors":"Guowen Lin, Feng Tian, Qiwei Yu, Xiaoling Weng, Nanhui Yu, Feng Zhang, Chen Yi, Jian Ye, Dingwei Ye","doi":"10.1038/s41388-024-03169-z","DOIUrl":"https://doi.org/10.1038/s41388-024-03169-z","url":null,"abstract":"<p><p>In this investigation, we explored the molecular dynamics guiding the progression of castration-resistant prostate cancer (CRPC) influenced by Helicobacter pylori (H. pylori)-mediated M2 polarization of macrophages through the IL-17RA/CTSK/EMT axis. An 830-patient clinical trial categorized subjects into hormone-sensitive prostate cancer (HSPC) and CRPC groups. H. pylori infection, evaluated by ELISA, exhibited a higher incidence in CRPC patients, impacting overall survival (OS) and progression-free survival. In-depth in vitro and in vivo experiments, including 16S rDNA sequencing, immunohistochemical tests, and transcriptome analysis, unveiled that H. pylori promotes CRPC growth and metastasis by upregulating IL-17RA and CTSK, leading to enhanced EMT. Notably, M2 macrophages emerged as pivotal immune cells influencing CRPC progression. This study uncovers a novel pathway wherein H. pylori enrichment exacerbates CRPC by inducing macrophage M2 polarization, IL-17RA/CTSK expression, and EMT activation, shedding light on a previously unrecognized mechanism contributing to the growth and metastasis of CRPC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142471603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
MAZ promotes tumor proliferation and immune evasion in lung adenocarcinoma. MAZ 可促进肺腺癌的肿瘤增殖和免疫逃避。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 DOI: 10.1038/s41388-024-03194-y
Yan Chen, Xinsheng Zhu, Jue Wang, Junjie Hu, Jing Zhang, Xun Zhang, Lu Han, Huansha Yu, Haiyang Hu, Ke Fei, Peng Zhang, Lele Zhang

Lung adenocarcinoma (LUAD) is the most dominant histological subtype of lung cancer and one of the most lethal malignancies. The identification of novel therapeutic targets is required for the treatment of LUAD. Here, we showed that MYC-associated zinc-finger protein (MAZ) is upregulated in LUAD tissues. MAZ expression levels are inversely correlated with patient survival. Silencing of MAZ decreased tumor proliferation and the expression of pro-tumorigenic chemokines and Galectin-9 (Gal-9), an immune checkpoint molecule. The pro-tumorigenic chemokines and Gal-9 induce immune suppression by recruitment of myeloid cells and inhibition of T cell activation, respectively. Mechanistically, MAZ transcriptionally regulates KRAS expression and activates its downstream AKT-NF-κB signaling pathway, which is crucial for tumor progression and immune evasion. Additionally, in vivo animal models and bioinformatic analyses indicated that MAZ suppression could enhance the efficacy of immune checkpoint blockade (ICB) therapy for LUAD. Overall, our results suggest that MAZ plays an important role in regulating cell proliferation and immune evasion via KRAS/AKT/NF-κB signaling in LUAD. Our findings offer a candidate molecular target for LUAD therapy, with implications for improving the efficacy of ICB therapy.

肺腺癌(LUAD)是肺癌中最主要的组织学亚型,也是最致命的恶性肿瘤之一。治疗肺腺癌需要找到新的治疗靶点。在这里,我们发现MYC相关锌指蛋白(MAZ)在LUAD组织中上调。MAZ的表达水平与患者的存活率成反比。沉默MAZ可减少肿瘤增殖,降低促致癌趋化因子和免疫检查点分子Galectin-9(Gal-9)的表达。促致癌趋化因子和Gal-9分别通过招募髓系细胞和抑制T细胞活化来诱导免疫抑制。从机理上讲,MAZ 可转录调节 KRAS 的表达,并激活其下游的 AKT-NF-κB 信号通路,这对肿瘤进展和免疫逃避至关重要。此外,体内动物模型和生物信息学分析表明,抑制MAZ可提高免疫检查点阻断疗法(ICB)治疗LUAD的疗效。总之,我们的研究结果表明,在LUAD中,MAZ通过KRAS/AKT/NF-κB信号在调节细胞增殖和免疫逃避方面发挥着重要作用。我们的研究结果为LUAD的治疗提供了一个候选分子靶点,对提高ICB疗法的疗效具有重要意义。
{"title":"MAZ promotes tumor proliferation and immune evasion in lung adenocarcinoma.","authors":"Yan Chen, Xinsheng Zhu, Jue Wang, Junjie Hu, Jing Zhang, Xun Zhang, Lu Han, Huansha Yu, Haiyang Hu, Ke Fei, Peng Zhang, Lele Zhang","doi":"10.1038/s41388-024-03194-y","DOIUrl":"https://doi.org/10.1038/s41388-024-03194-y","url":null,"abstract":"<p><p>Lung adenocarcinoma (LUAD) is the most dominant histological subtype of lung cancer and one of the most lethal malignancies. The identification of novel therapeutic targets is required for the treatment of LUAD. Here, we showed that MYC-associated zinc-finger protein (MAZ) is upregulated in LUAD tissues. MAZ expression levels are inversely correlated with patient survival. Silencing of MAZ decreased tumor proliferation and the expression of pro-tumorigenic chemokines and Galectin-9 (Gal-9), an immune checkpoint molecule. The pro-tumorigenic chemokines and Gal-9 induce immune suppression by recruitment of myeloid cells and inhibition of T cell activation, respectively. Mechanistically, MAZ transcriptionally regulates KRAS expression and activates its downstream AKT-NF-κB signaling pathway, which is crucial for tumor progression and immune evasion. Additionally, in vivo animal models and bioinformatic analyses indicated that MAZ suppression could enhance the efficacy of immune checkpoint blockade (ICB) therapy for LUAD. Overall, our results suggest that MAZ plays an important role in regulating cell proliferation and immune evasion via KRAS/AKT/NF-κB signaling in LUAD. Our findings offer a candidate molecular target for LUAD therapy, with implications for improving the efficacy of ICB therapy.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142471604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Obesity-induced extracellular vesicles proteins drive the endometrial cancer pathogenesis: therapeutic potential of HO-3867 and Metformin. 肥胖诱导的细胞外囊泡蛋白驱动子宫内膜癌发病机制:HO-3867 和二甲双胍的治疗潜力
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-16 DOI: 10.1038/s41388-024-03182-2
Takahiko Sakaue, Kalpana Deepa Priya Dorayappan, Roman Zingarelli, Wafa Khadraoui, Muralidharan Anbalagan, John Wallbillich, Balazs Bognar, Ross Wanner, Casey Cosgrove, Adrian Suarez, Hironori Koga, G Larry Maxwell, David M O'Malley, David E Cohn, Karuppaiyah Selvendiran

Endometrial cancer (EC) is the leading gynecologic malignancy in the United States with obesity implicated in 57% of cases. This research investigates the molecular complexities of extracellular vesicles (EV) secretion as carriers of oncogenic protein and their involvement in obesity-mediated EC. An understanding of these mechanisms is pivotal for unraveling pathways relevant to obesity-associated EC, thereby guiding the development of innovative prevention and treatment strategies. Our exploration revealed a significant increase in EV secretion carrying oncogenic proteins (TMEM205, STAT5, and FAS) in adipose and uterine tissues/serum samples from obese EC patients compared to control (without cancer). We identified alterations in EV-regulating proteins (Rab7, Rab11, and Rab27a) in obesity-mediated EC patients, adipose/uterine tissues, and serum samples. Through a 24-week analysis of the effects of a 45% kcal high-fat diet (HFD) on mice, we observed increased body weight, increased adipose tissue, enlarged uterine horns, and increased inflammation in the HFD group. This correlated with elevated levels of EV secretion and increased expression of oncogenic proteins TMEM205, FAS, and STAT5 and downregulation of the tumor suppressor gene PIAS3 in adipose and uterine tissues. Furthermore, our study confirmed that adipocyte derived EV increased EC cell proliferation, migration and xenograft tumor growth. Additionally, we identified that the small molecule inhibitors (HO-3867) or Metformin inhibited EV secretion in vitro and in vivo, demonstrating significant inhibition of high glucose or adipocyte-mediated EC cell proliferation and a reduction in body weight and adipose tissue accumulation when administered to HFD mice. Moreover, HO-3867 or Metformin treatment inhibited HFD induced hyperplasia (precursor of EC) by altering the expression of EV-regulated proteins and decreasing oncogenic protein expression levels. This study provides critical insights into the mechanisms underpinning obesity-mediated EV secretion with oncogenic protein expression, shedding light on their role in EC pathogenesis. Additionally, it offers pre-clinical evidence supporting the initiation of novel studies for EV-targeted therapies aimed at preventing obesity-mediated EC.

子宫内膜癌(EC)是美国最主要的妇科恶性肿瘤,其中 57% 的病例与肥胖有关。这项研究调查了细胞外囊泡(EV)分泌作为致癌蛋白载体的分子复杂性及其在肥胖介导的子宫内膜癌中的参与。了解这些机制对于揭示肥胖相关EC的相关途径至关重要,从而指导创新性预防和治疗策略的开发。我们的研究发现,与对照组(未患癌症)相比,肥胖EC患者的脂肪和子宫组织/血清样本中携带致癌蛋白(TMEM205、STAT5和FAS)的EV分泌明显增加。我们在肥胖引发的EC患者、脂肪/子宫组织和血清样本中发现了EV调节蛋白(Rab7、Rab11和Rab27a)的变化。通过对 45% 千卡高脂饮食(HFD)对小鼠影响的 24 周分析,我们观察到 HFD 组小鼠体重增加、脂肪组织增加、子宫角增大、炎症加重。这与脂肪组织和子宫组织中 EV 分泌水平升高、致癌蛋白 TMEM205、FAS 和 STAT5 表达增加以及抑癌基因 PIAS3 下调有关。此外,我们的研究证实,脂肪细胞衍生的 EV 增加了 EC 细胞的增殖、迁移和异种移植肿瘤的生长。此外,我们发现小分子抑制剂(HO-3867)或二甲双胍可抑制体外和体内的EV分泌,对高血糖或脂肪细胞介导的EC细胞增殖有显著抑制作用,并可减少高血糖小鼠的体重和脂肪组织堆积。此外,HO-3867 或二甲双胍通过改变 EV 调控蛋白的表达和降低致癌蛋白的表达水平,抑制了 HFD 诱导的增生(EC 的前体)。这项研究深入揭示了肥胖介导的EV分泌和致癌蛋白表达的机制,阐明了它们在EC发病机制中的作用。此外,它还提供了临床前证据,支持启动旨在预防肥胖介导的心肌梗死的 EV 靶向疗法的新型研究。
{"title":"Obesity-induced extracellular vesicles proteins drive the endometrial cancer pathogenesis: therapeutic potential of HO-3867 and Metformin.","authors":"Takahiko Sakaue, Kalpana Deepa Priya Dorayappan, Roman Zingarelli, Wafa Khadraoui, Muralidharan Anbalagan, John Wallbillich, Balazs Bognar, Ross Wanner, Casey Cosgrove, Adrian Suarez, Hironori Koga, G Larry Maxwell, David M O'Malley, David E Cohn, Karuppaiyah Selvendiran","doi":"10.1038/s41388-024-03182-2","DOIUrl":"https://doi.org/10.1038/s41388-024-03182-2","url":null,"abstract":"<p><p>Endometrial cancer (EC) is the leading gynecologic malignancy in the United States with obesity implicated in 57% of cases. This research investigates the molecular complexities of extracellular vesicles (EV) secretion as carriers of oncogenic protein and their involvement in obesity-mediated EC. An understanding of these mechanisms is pivotal for unraveling pathways relevant to obesity-associated EC, thereby guiding the development of innovative prevention and treatment strategies. Our exploration revealed a significant increase in EV secretion carrying oncogenic proteins (TMEM205, STAT5, and FAS) in adipose and uterine tissues/serum samples from obese EC patients compared to control (without cancer). We identified alterations in EV-regulating proteins (Rab7, Rab11, and Rab27a) in obesity-mediated EC patients, adipose/uterine tissues, and serum samples. Through a 24-week analysis of the effects of a 45% kcal high-fat diet (HFD) on mice, we observed increased body weight, increased adipose tissue, enlarged uterine horns, and increased inflammation in the HFD group. This correlated with elevated levels of EV secretion and increased expression of oncogenic proteins TMEM205, FAS, and STAT5 and downregulation of the tumor suppressor gene PIAS3 in adipose and uterine tissues. Furthermore, our study confirmed that adipocyte derived EV increased EC cell proliferation, migration and xenograft tumor growth. Additionally, we identified that the small molecule inhibitors (HO-3867) or Metformin inhibited EV secretion in vitro and in vivo, demonstrating significant inhibition of high glucose or adipocyte-mediated EC cell proliferation and a reduction in body weight and adipose tissue accumulation when administered to HFD mice. Moreover, HO-3867 or Metformin treatment inhibited HFD induced hyperplasia (precursor of EC) by altering the expression of EV-regulated proteins and decreasing oncogenic protein expression levels. This study provides critical insights into the mechanisms underpinning obesity-mediated EV secretion with oncogenic protein expression, shedding light on their role in EC pathogenesis. Additionally, it offers pre-clinical evidence supporting the initiation of novel studies for EV-targeted therapies aimed at preventing obesity-mediated EC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142471607","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Mesenchymal stromal cells as cancer promoters. 间充质基质细胞是癌症的促进因素。
IF 6.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-16 DOI: 10.1038/s41388-024-03183-1
Roula Antoon, Nina Overdevest, Amr H Saleh, Armand Keating

Mesenchymal stromal cells (MSCs) are important cellular constituents of tumor stroma that play an active role in tumor development. Complex interactions between MSCs and cancer promote tumor progression by creating a favorable milieu for tumor cell proliferation, angiogenesis, motility, invasion, and metastasis. The cellular heterogeneity, source of origin, diversity in isolation methods, culture techniques and model systems of MSCs, together with the different tumor subtypes, add to the complexity of MSC-tumor interactions. In this review, we discuss the mechanisms of MSC-mediated tumor promotion and evaluate cell-stromal interactions between cancer cells, MSCs, cells of the tumor microenvironment (TME), and the extracellular matrix (ECM). A more thorough understanding of tumor-MSC interactions is likely to lead to better cancer management.

间充质基质细胞(MSCs)是肿瘤基质的重要细胞成分,在肿瘤发生发展过程中发挥着积极作用。间充质干细胞与癌症之间复杂的相互作用为肿瘤细胞的增殖、血管生成、运动、侵袭和转移创造了有利的环境,从而促进了肿瘤的发展。间充质干细胞的细胞异质性、来源、分离方法、培养技术和模型系统的多样性,以及不同的肿瘤亚型,增加了间充质干细胞与肿瘤相互作用的复杂性。在这篇综述中,我们讨论了间充质干细胞介导的肿瘤促进机制,并评估了癌细胞、间充质干细胞、肿瘤微环境(TME)细胞和细胞外基质(ECM)之间的细胞间质相互作用。更透彻地了解肿瘤与间充质干细胞之间的相互作用可能有助于更好地治疗癌症。
{"title":"Mesenchymal stromal cells as cancer promoters.","authors":"Roula Antoon, Nina Overdevest, Amr H Saleh, Armand Keating","doi":"10.1038/s41388-024-03183-1","DOIUrl":"https://doi.org/10.1038/s41388-024-03183-1","url":null,"abstract":"<p><p>Mesenchymal stromal cells (MSCs) are important cellular constituents of tumor stroma that play an active role in tumor development. Complex interactions between MSCs and cancer promote tumor progression by creating a favorable milieu for tumor cell proliferation, angiogenesis, motility, invasion, and metastasis. The cellular heterogeneity, source of origin, diversity in isolation methods, culture techniques and model systems of MSCs, together with the different tumor subtypes, add to the complexity of MSC-tumor interactions. In this review, we discuss the mechanisms of MSC-mediated tumor promotion and evaluate cell-stromal interactions between cancer cells, MSCs, cells of the tumor microenvironment (TME), and the extracellular matrix (ECM). A more thorough understanding of tumor-MSC interactions is likely to lead to better cancer management.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142471605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Oncogene
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