首页 > 最新文献

Oncogenesis最新文献

英文 中文
The branched N-glycan of PD-L1 predicts immunotherapy responses in patients with recurrent/metastatic HNSCC. PD-L1 的支链 N-聚糖可预测复发/转移性 HNSCC 患者的免疫疗法反应。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-02 DOI: 10.1038/s41389-024-00532-3
Huai-Cheng Huang, Yen-Lin Huang, Yi-Ju Chen, Hsin-Yi Wu, Chia-Lang Hsu, Hsiang-Fong Kao, Bin-Chi Liao, Min-Shu Hsieh, Neng-Yu Lin, Yu-Hao Liao, Hsin-Lin Chen, Chun-Nan Chen, Tseng-Cheng Chen, Cheng-Ping Wang, Tsung-Lin Yang, Min-Chuan Huang, Mei-Chun Lin, Pei-Jen Lou

Immunotherapy has revolutionized cancer treatment, but the lack of a reliable predictive biomarker for treatment response remains a challenge. Alpha-1,6-Mannosylglycoprotein 6-β-N-Acetylglucosaminyltransferase 5 (MGAT5) is a key regulator of complex N-glycan synthesis, and its dysregulation is associated with cancer progression. The lectin Phaseolus vulgaris leukoagglutinin (PHA-L) specifically binds to mature MGAT5 products. Previous studies have indicated elevated PHA-L staining in head and neck squamous cell carcinoma (HNSCC), which implies increased activity of MGAT5. However, the specific role of MGAT5 in HNSCC remains unclear. In this study, we found significantly higher PHA-L staining and MGAT5 expression in HNSCC tumors compared to adjacent non-tumor tissues. Using a mass spectrometry (MS)-based glycoproteomic approach, we identified 163 potential protein substrates of MGAT5. Functional analysis revealed that protein substrates of MGAT5 regulated pathways related to T cell proliferation and activation. We further discovered that PD-L1 was among the protein substrates of MGAT5, and the expression of MGAT5 protected tumor cells from cytotoxic T lymphocyte (CTL) killing. Treatment of nivolumab alleviated the protective effects of MGAT5 on CTL activity. Consistently, patients with MGAT5-positive tumors showed improved responses to immunotherapy compared to those with MGAT5-negative tumors. Using purified PD-L1 from HNSCC cells and a glycoproteomic approach, we further deciphered that the N35 and N200 sites carry the majority of complex N-glycans on PD-L1. Our findings highlight the critical role of MGAT5-mediated branched N-glycans on PD-L1 in modulating the interaction with the immune checkpoint receptor PD-1. Consequently, we propose that MGAT5 could serve as a biomarker to predict patients' responses to anti-PD-1 therapy. Furthermore, targeting the branched N-glycans at N35 and N200 of PD-L1 may lead to the development of novel diagnostic and therapeutic approaches.

免疫疗法给癌症治疗带来了革命性的变化,但缺乏可靠的预测治疗反应的生物标志物仍然是一项挑战。α-1,6-甘露糖基糖蛋白 6-β-N-Acetylglucosaminyltransferase 5(MGAT5)是复杂 N-糖合成的关键调控因子,它的失调与癌症进展有关。凝集素 Phaseolus vulgaris leukoagglutinin (PHA-L) 能与成熟的 MGAT5 产物特异性结合。以往的研究表明,头颈部鳞状细胞癌(HNSCC)的 PHA-L 染色升高,这意味着 MGAT5 的活性增加。然而,MGAT5 在 HNSCC 中的具体作用仍不清楚。在这项研究中,我们发现 HNSCC 肿瘤中 PHA-L 染色和 MGAT5 表达明显高于邻近的非肿瘤组织。利用基于质谱(MS)的糖蛋白组学方法,我们确定了 163 种 MGAT5 的潜在蛋白底物。功能分析显示,MGAT5 的蛋白底物调控与 T 细胞增殖和活化相关的通路。我们进一步发现,PD-L1是MGAT5的蛋白底物之一,而MGAT5的表达能保护肿瘤细胞免受细胞毒性T淋巴细胞(CTL)的杀伤。nivolumab的治疗减轻了MGAT5对CTL活性的保护作用。同样,与MGAT5阴性肿瘤患者相比,MGAT5阳性肿瘤患者对免疫疗法的反应更好。利用从 HNSCC 细胞中纯化的 PD-L1 和糖蛋白组学方法,我们进一步破译了 N35 和 N200 位点携带了 PD-L1 上大部分复杂的 N-聚糖。我们的发现凸显了 MGAT5 介导的 PD-L1 上的支化 N-聚糖在调节与免疫检查点受体 PD-1 的相互作用中的关键作用。因此,我们建议将 MGAT5 作为一种生物标记物来预测患者对抗 PD-1 疗法的反应。此外,以 PD-L1 的 N35 和 N200 处的支链 N-糖为靶点可能会开发出新型诊断和治疗方法。
{"title":"The branched N-glycan of PD-L1 predicts immunotherapy responses in patients with recurrent/metastatic HNSCC.","authors":"Huai-Cheng Huang, Yen-Lin Huang, Yi-Ju Chen, Hsin-Yi Wu, Chia-Lang Hsu, Hsiang-Fong Kao, Bin-Chi Liao, Min-Shu Hsieh, Neng-Yu Lin, Yu-Hao Liao, Hsin-Lin Chen, Chun-Nan Chen, Tseng-Cheng Chen, Cheng-Ping Wang, Tsung-Lin Yang, Min-Chuan Huang, Mei-Chun Lin, Pei-Jen Lou","doi":"10.1038/s41389-024-00532-3","DOIUrl":"10.1038/s41389-024-00532-3","url":null,"abstract":"<p><p>Immunotherapy has revolutionized cancer treatment, but the lack of a reliable predictive biomarker for treatment response remains a challenge. Alpha-1,6-Mannosylglycoprotein 6-β-N-Acetylglucosaminyltransferase 5 (MGAT5) is a key regulator of complex N-glycan synthesis, and its dysregulation is associated with cancer progression. The lectin Phaseolus vulgaris leukoagglutinin (PHA-L) specifically binds to mature MGAT5 products. Previous studies have indicated elevated PHA-L staining in head and neck squamous cell carcinoma (HNSCC), which implies increased activity of MGAT5. However, the specific role of MGAT5 in HNSCC remains unclear. In this study, we found significantly higher PHA-L staining and MGAT5 expression in HNSCC tumors compared to adjacent non-tumor tissues. Using a mass spectrometry (MS)-based glycoproteomic approach, we identified 163 potential protein substrates of MGAT5. Functional analysis revealed that protein substrates of MGAT5 regulated pathways related to T cell proliferation and activation. We further discovered that PD-L1 was among the protein substrates of MGAT5, and the expression of MGAT5 protected tumor cells from cytotoxic T lymphocyte (CTL) killing. Treatment of nivolumab alleviated the protective effects of MGAT5 on CTL activity. Consistently, patients with MGAT5-positive tumors showed improved responses to immunotherapy compared to those with MGAT5-negative tumors. Using purified PD-L1 from HNSCC cells and a glycoproteomic approach, we further deciphered that the N35 and N200 sites carry the majority of complex N-glycans on PD-L1. Our findings highlight the critical role of MGAT5-mediated branched N-glycans on PD-L1 in modulating the interaction with the immune checkpoint receptor PD-1. Consequently, we propose that MGAT5 could serve as a biomarker to predict patients' responses to anti-PD-1 therapy. Furthermore, targeting the branched N-glycans at N35 and N200 of PD-L1 may lead to the development of novel diagnostic and therapeutic approaches.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11445275/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142361802","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
NRF2 signaling plays an essential role in cancer progression through the NRF2-GPX2-NOTCH3 axis in head and neck squamous cell carcinoma. 在头颈部鳞状细胞癌中,NRF2 信号通过 NRF2-GPX2-NOTCH3 轴在癌症进展中发挥重要作用。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-27 DOI: 10.1038/s41389-024-00536-z
Xiaoye Jin, Xiayuan Lou, Haoxiang Qi, Chao Zheng, Bo Li, Xuerong Siwu, Ren Liu, Qiaoli Lv, An Zhao, Jian Ruan, Ming Jiang

The activation of nuclear factor erythroid 2-related factor 2 (NRF2) has been observed in various cancers. Yet its exact contribution to the development of head and neck squamous cell carcinoma (HNSCC) remains undetermined. We previously found that NRF2 signaling is critical for the differentiation of squamous basal progenitor cells, while disruption of NRF2 causes basal cell hyperplasia. In this study, we revealed a correlation between elevated NRF2 activity and poor outcomes in HNSCC patients. We demonstrated that NRF2 facilitates tumor proliferation, migration, and invasion, as evidenced by both in vitro and in vivo studies. Significantly, NRF2 augments the expression of the antioxidant enzyme GPX2, thereby enhancing the proliferative, migratory, and invasive properties of HNSCC cells. Activation of GPX2 is critical for sustaining cancer stem cells (CSCs) by up-regulating NOTCH3, a key driver of cancer progression. These results elucidate that NRF2 regulates HNSCC progression through the NRF2-GPX2-NOTCH3 axis. Our findings proposed that pharmacological targeting of the NRF2-GPX2-NOTCH3 axis could be a potential therapeutic approach against HNSCC.

在多种癌症中都观察到了核因子红细胞2相关因子2(NRF2)的活化。然而,它对头颈部鳞状细胞癌(HNSCC)发病的确切作用仍未确定。我们之前发现,NRF2 信号传导对鳞状基底祖细胞的分化至关重要,而 NRF2 信号传导中断会导致基底细胞增生。在本研究中,我们揭示了 NRF2 活性升高与 HNSCC 患者不良预后之间的相关性。我们通过体外和体内研究证明,NRF2 有助于肿瘤的增殖、迁移和侵袭。值得注意的是,NRF2 能增强抗氧化酶 GPX2 的表达,从而增强 HNSCC 细胞的增殖、迁移和侵袭特性。GPX2的激活对于通过上调NOTCH3(癌症进展的关键驱动因素)维持癌症干细胞(CSCs)至关重要。这些结果阐明了NRF2通过NRF2-GPX2-NOTCH3轴调控HNSCC的进展。我们的研究结果表明,以NRF2-GPX2-NOTCH3轴为药理靶点可能是治疗HNSCC的一种潜在方法。
{"title":"NRF2 signaling plays an essential role in cancer progression through the NRF2-GPX2-NOTCH3 axis in head and neck squamous cell carcinoma.","authors":"Xiaoye Jin, Xiayuan Lou, Haoxiang Qi, Chao Zheng, Bo Li, Xuerong Siwu, Ren Liu, Qiaoli Lv, An Zhao, Jian Ruan, Ming Jiang","doi":"10.1038/s41389-024-00536-z","DOIUrl":"https://doi.org/10.1038/s41389-024-00536-z","url":null,"abstract":"<p><p>The activation of nuclear factor erythroid 2-related factor 2 (NRF2) has been observed in various cancers. Yet its exact contribution to the development of head and neck squamous cell carcinoma (HNSCC) remains undetermined. We previously found that NRF2 signaling is critical for the differentiation of squamous basal progenitor cells, while disruption of NRF2 causes basal cell hyperplasia. In this study, we revealed a correlation between elevated NRF2 activity and poor outcomes in HNSCC patients. We demonstrated that NRF2 facilitates tumor proliferation, migration, and invasion, as evidenced by both in vitro and in vivo studies. Significantly, NRF2 augments the expression of the antioxidant enzyme GPX2, thereby enhancing the proliferative, migratory, and invasive properties of HNSCC cells. Activation of GPX2 is critical for sustaining cancer stem cells (CSCs) by up-regulating NOTCH3, a key driver of cancer progression. These results elucidate that NRF2 regulates HNSCC progression through the NRF2-GPX2-NOTCH3 axis. Our findings proposed that pharmacological targeting of the NRF2-GPX2-NOTCH3 axis could be a potential therapeutic approach against HNSCC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11437035/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142351319","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
DKK1 as a chemoresistant protein modulates oxaliplatin responses in colorectal cancer. DKK1 作为一种化疗抗性蛋白可调节奥沙利铂在结直肠癌中的反应。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-27 DOI: 10.1038/s41389-024-00537-y
Chi-Che Hsieh, Ting-Wei Li, Chun-Chun Li, Shang-Hung Chen, You-Lin Wei, Nai-Jung Chiang, Che-Hung Shen

Oxaliplatin is effective against colorectal cancer (CRC), but resistance hampers treatment. We found upregulated Dickkopf-1 (DKK1, a secreted protein) in oxaliplatin-resistant (OR) CRC cell lines and DKK1 levels increased by more than 2-fold in approximately 50% of oxaliplatin-resistant CRC tumors. DKK1 activates AKT via cytoskeleton-associated protein 4 (CKAP4, a DKK1 receptor), modulating oxaliplatin responses in vitro and in vivo. The leucine zipper (LZ) domain of CKAP4 and cysteine-rich domain 1 (CRD1) of secreted DKK1 are crucial for their interaction and AKT signaling. By utilizing the LZ protein, we disrupted DKK1 signaling, enhancing oxaliplatin sensitivity in OR CRC cells and xenograft tumors. This suggests that DKK1 as a chemoresistant factor in CRC via AKT activation. Targeting DKK1 with the LZ protein offers a promising therapeutic strategy for oxaliplatin-resistant CRC with high DKK1 levels. This study sheds light on oxaliplatin resistance mechanisms and proposes an innovative intervention for managing this challenge.

奥沙利铂对结直肠癌(CRC)有效,但耐药性阻碍了治疗。我们发现奥沙利铂耐药(OR)CRC细胞系中的Dickkopf-1(DKK1,一种分泌蛋白)上调,约50%的奥沙利铂耐药CRC肿瘤中DKK1水平增加了2倍以上。DKK1通过细胞骨架相关蛋白4(CKAP4,一种DKK1受体)激活AKT,从而调节体外和体内的奥沙利铂反应。CKAP4 的亮氨酸拉链(LZ)结构域和分泌型 DKK1 的富半胱氨酸结构域 1(CRD1)对于它们之间的相互作用和 AKT 信号转导至关重要。通过利用 LZ 蛋白,我们破坏了 DKK1 信号传导,提高了奥沙利铂在 OR CRC 细胞和异种移植肿瘤中的敏感性。这表明,DKK1是通过AKT激活的CRC化疗耐药因子。用LZ蛋白靶向DKK1为高DKK1水平的奥沙利铂耐药CRC提供了一种有前景的治疗策略。这项研究揭示了奥沙利铂的耐药机制,并提出了应对这一挑战的创新干预措施。
{"title":"DKK1 as a chemoresistant protein modulates oxaliplatin responses in colorectal cancer.","authors":"Chi-Che Hsieh, Ting-Wei Li, Chun-Chun Li, Shang-Hung Chen, You-Lin Wei, Nai-Jung Chiang, Che-Hung Shen","doi":"10.1038/s41389-024-00537-y","DOIUrl":"https://doi.org/10.1038/s41389-024-00537-y","url":null,"abstract":"<p><p>Oxaliplatin is effective against colorectal cancer (CRC), but resistance hampers treatment. We found upregulated Dickkopf-1 (DKK1, a secreted protein) in oxaliplatin-resistant (OR) CRC cell lines and DKK1 levels increased by more than 2-fold in approximately 50% of oxaliplatin-resistant CRC tumors. DKK1 activates AKT via cytoskeleton-associated protein 4 (CKAP4, a DKK1 receptor), modulating oxaliplatin responses in vitro and in vivo. The leucine zipper (LZ) domain of CKAP4 and cysteine-rich domain 1 (CRD1) of secreted DKK1 are crucial for their interaction and AKT signaling. By utilizing the LZ protein, we disrupted DKK1 signaling, enhancing oxaliplatin sensitivity in OR CRC cells and xenograft tumors. This suggests that DKK1 as a chemoresistant factor in CRC via AKT activation. Targeting DKK1 with the LZ protein offers a promising therapeutic strategy for oxaliplatin-resistant CRC with high DKK1 levels. This study sheds light on oxaliplatin resistance mechanisms and proposes an innovative intervention for managing this challenge.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11436992/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142351318","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
TFCP2L1 drives stemness and enhances their resistance to Sorafenib treatment by modulating the NANOG/STAT3 pathway in hepatocellular carcinoma TFCP2L1通过调节肝细胞癌中的NANOG/STAT3通路驱动干性并增强其对索拉非尼治疗的耐受性
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1038/s41389-024-00534-1
Dongbo Qiu, Tiantian Wang, Yi Xiong, Kun Li, Xiusheng Qiu, Yuan Feng, Qinghai Lian, Yunfei Qin, Kunpeng Liu, Qi Zhang, Changchang Jia

Hepatocellular carcinoma (HCC) is a prevalent and aggressive malignancy associated with high risks of recurrence and metastasis. Liver cancer stem cells (CSCs) are increasingly recognized as pivotal drivers of these processes. In our previous research, we demonstrated the involvement of TFCP2L1 in maintaining the pluripotency of embryonic stem cells. However, its relevance to liver CSCs remains unexplored. In this study, we report an inverse correlation between TFCP2L1 protein levels in HCC tissue and patient outcomes. The knockdown of TFCP2L1 significantly reduced HCC cell proliferation, invasion, metastasis, clonal formation, and sphere-forming capacity, while its overexpression enhanced these functions. In addition, experiments using a nude mouse model confirmed TFCP2L1’s essential role in liver CSCs’ function and tumorigenic potential. Mechanistically, we showed that TFCP2L1 promotes the stemness of CSCs by upregulating NANOG, which subsequently activates the JAK/STAT3 pathway, thereby contributing to HCC pathogenesis. Importantly, we identified a specific small molecule targeting TFCP2L1’s active domain, which, in combination with Sorafenib, sensitizes hepatoma cells to treatment. Together, these findings underscore TFCP2L1’s pathological significance in HCC progression, supporting its potential as a prognostic biomarker and therapeutic target in this disease.

肝细胞癌(HCC)是一种常见的侵袭性恶性肿瘤,复发和转移的风险很高。肝癌干细胞(CSCs)越来越被认为是这些过程的关键驱动因素。在我们之前的研究中,我们证实了TFCP2L1参与维持胚胎干细胞的多能性。然而,它与肝脏干细胞的相关性仍未得到探讨。在本研究中,我们报告了HCC组织中TFCP2L1蛋白水平与患者预后之间的反相关性。敲除 TFCP2L1 能显著减少 HCC 细胞的增殖、侵袭、转移、克隆形成和球形成能力,而过表达则能增强这些功能。此外,利用裸鼠模型进行的实验证实了 TFCP2L1 在肝脏 CSCs 功能和致瘤潜能中的重要作用。从机理上讲,我们发现TFCP2L1通过上调NANOG促进CSCs的干性,进而激活JAK/STAT3通路,从而导致HCC发病。重要的是,我们发现了一种靶向TFCP2L1活性结构域的特异性小分子,它与索拉非尼联用可使肝癌细胞对治疗敏感。这些发现共同强调了TFCP2L1在HCC进展中的病理意义,支持其作为该疾病预后生物标志物和治疗靶点的潜力。
{"title":"TFCP2L1 drives stemness and enhances their resistance to Sorafenib treatment by modulating the NANOG/STAT3 pathway in hepatocellular carcinoma","authors":"Dongbo Qiu, Tiantian Wang, Yi Xiong, Kun Li, Xiusheng Qiu, Yuan Feng, Qinghai Lian, Yunfei Qin, Kunpeng Liu, Qi Zhang, Changchang Jia","doi":"10.1038/s41389-024-00534-1","DOIUrl":"https://doi.org/10.1038/s41389-024-00534-1","url":null,"abstract":"<p>Hepatocellular carcinoma (HCC) is a prevalent and aggressive malignancy associated with high risks of recurrence and metastasis. Liver cancer stem cells (CSCs) are increasingly recognized as pivotal drivers of these processes. In our previous research, we demonstrated the involvement of TFCP2L1 in maintaining the pluripotency of embryonic stem cells. However, its relevance to liver CSCs remains unexplored. In this study, we report an inverse correlation between TFCP2L1 protein levels in HCC tissue and patient outcomes. The knockdown of TFCP2L1 significantly reduced HCC cell proliferation, invasion, metastasis, clonal formation, and sphere-forming capacity, while its overexpression enhanced these functions. In addition, experiments using a nude mouse model confirmed TFCP2L1’s essential role in liver CSCs’ function and tumorigenic potential. Mechanistically, we showed that TFCP2L1 promotes the stemness of CSCs by upregulating NANOG, which subsequently activates the JAK/STAT3 pathway, thereby contributing to HCC pathogenesis. Importantly, we identified a specific small molecule targeting TFCP2L1’s active domain, which, in combination with Sorafenib, sensitizes hepatoma cells to treatment. Together, these findings underscore TFCP2L1’s pathological significance in HCC progression, supporting its potential as a prognostic biomarker and therapeutic target in this disease.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142189058","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
Correction: Dihydroartemisinin inhibits prostate cancer via JARID2/miR-7/miR-34a-dependent downregulation of Axl. 更正:双氢青蒿素通过 JARID2/miR-7/miR-34a 依赖性下调 Axl 抑制前列腺癌。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1038/s41389-024-00533-2
Juliano D Paccez, Kristal Duncan, Durairaj Sekar, Ricardo G Correa, Yihong Wang, Xuesong Gu, Manoj Bashin, Kelly Chibale, Towia A Libermann, Luiz F Zerbini
{"title":"Correction: Dihydroartemisinin inhibits prostate cancer via JARID2/miR-7/miR-34a-dependent downregulation of Axl.","authors":"Juliano D Paccez, Kristal Duncan, Durairaj Sekar, Ricardo G Correa, Yihong Wang, Xuesong Gu, Manoj Bashin, Kelly Chibale, Towia A Libermann, Luiz F Zerbini","doi":"10.1038/s41389-024-00533-2","DOIUrl":"https://doi.org/10.1038/s41389-024-00533-2","url":null,"abstract":"","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11393321/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142292657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tumor suppressor BAP1 suppresses disulfidptosis through the regulation of SLC7A11 and NADPH levels 肿瘤抑制因子 BAP1 通过调节 SLC7A11 和 NADPH 水平抑制二硫化硫作用
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1038/s41389-024-00535-0
Jin Wang, Minglin Wang, Shaobo Wu, Yanan Zhu, Kexin Fan, Yuhan Chen, Zhengtao Xiao, Jing Chen, Kangsheng Tu, Dongsheng Huang, Yilei Zhang, Qiuran Xu

BAP1, BRCA1-Associated Protein 1, serves as a novel tumor suppressor through the deubiquitination of monoubiquitination of H2A and subsequent gene transcriptional regulation. Regulated cell death like apoptosis or ferroptosis is considered an essential mechanism mediating tumor suppression. Previous reports, including ours, have demonstrated that BAP1 could promote apoptosis and ferroptosis to inhibit tumor development. Whether BAP1 regulated additional types of cell death remains unclear. Disulfidptosis is a recently identified novel cell death mode characterized by aberrant accumulation of intracellular disulfide (e.g., cystine) and depletion of NADPH. In this study, we first demonstrated that BAP1 could significantly protect disulfidptosis induced by glucose starvation, which is validated by various cell death inhibitors and the accumulation of disulfide bonds in the cytoskeleton proteins. BAP1 is known to inhibit SLC7A11 expression. We found that the protective effect of BAP1 against disulfidptosis was counteracted when overexpressing SLC7A11 or adding additional cystine. Conversely, BAP1-mediated suppression of disulfidptosis was largely abrogated when SLC7A11-mediated cystine uptake was inhibited by the knockout of SLC7A11 or erastin treatment. Besides, high BAP1 expression showed lower NADP+/NADPH levels, which might confer resistance to disulfidptosis. Consistent with these observations, the expression level of BAP1 was also positively correlated with NADPH-related genes in KIRC patients, though the underlying mechanism mediating NADPH regulation remains further investigation. In summary, our results revealed the role of BAP1 in the regulation disulfidptosis and provided new insights into the understanding of disulfidptosis in tumor development.

BAP1,即 BRCA1 相关蛋白 1,通过对 H2A 的单泛素化进行去泛素化以及随后的基因转录调控,成为一种新型肿瘤抑制因子。细胞凋亡或铁凋亡等调节性细胞死亡被认为是介导肿瘤抑制的重要机制。之前的报道(包括我们的研究)表明,BAP1 可促进细胞凋亡和铁凋亡,从而抑制肿瘤的发展。BAP1 是否还能调节其他类型的细胞死亡仍不清楚。二硫化ptosis是最近发现的一种新型细胞死亡模式,其特点是细胞内二硫化物(如胱氨酸)的异常积累和NADPH的耗竭。在这项研究中,我们首次证明了 BAP1 能显著保护葡萄糖饥饿诱导的二硫化ptosis,这一点已被各种细胞死亡抑制剂和细胞骨架蛋白中二硫键的积累所验证。众所周知,BAP1 可抑制 SLC7A11 的表达。我们发现,当过表达 SLC7A11 或添加额外的胱氨酸时,BAP1 对二硫化硫的保护作用会被抵消。相反,当通过敲除 SLC7A11 或厄拉斯汀处理抑制 SLC7A11 介导的胱氨酸摄取时,BAP1 介导的二硫化硫抑制作用在很大程度上被削弱。此外,BAP1的高表达显示了较低的NADP+/NADPH水平,这可能赋予了对二硫化硫的抗性。与这些观察结果一致的是,BAP1的表达水平也与KIRC患者的NADPH相关基因呈正相关,但NADPH调控的潜在机制仍有待进一步研究。总之,我们的研究结果揭示了 BAP1 在调控二硫化硫过程中的作用,为了解二硫化硫在肿瘤发生发展过程中的作用提供了新的视角。
{"title":"Tumor suppressor BAP1 suppresses disulfidptosis through the regulation of SLC7A11 and NADPH levels","authors":"Jin Wang, Minglin Wang, Shaobo Wu, Yanan Zhu, Kexin Fan, Yuhan Chen, Zhengtao Xiao, Jing Chen, Kangsheng Tu, Dongsheng Huang, Yilei Zhang, Qiuran Xu","doi":"10.1038/s41389-024-00535-0","DOIUrl":"https://doi.org/10.1038/s41389-024-00535-0","url":null,"abstract":"<p>BAP1, BRCA1-Associated Protein 1, serves as a novel tumor suppressor through the deubiquitination of monoubiquitination of H2A and subsequent gene transcriptional regulation. Regulated cell death like apoptosis or ferroptosis is considered an essential mechanism mediating tumor suppression. Previous reports, including ours, have demonstrated that BAP1 could promote apoptosis and ferroptosis to inhibit tumor development. Whether BAP1 regulated additional types of cell death remains unclear. Disulfidptosis is a recently identified novel cell death mode characterized by aberrant accumulation of intracellular disulfide (e.g., cystine) and depletion of NADPH. In this study, we first demonstrated that BAP1 could significantly protect disulfidptosis induced by glucose starvation, which is validated by various cell death inhibitors and the accumulation of disulfide bonds in the cytoskeleton proteins. BAP1 is known to inhibit SLC7A11 expression. We found that the protective effect of BAP1 against disulfidptosis was counteracted when overexpressing SLC7A11 or adding additional cystine. Conversely, BAP1-mediated suppression of disulfidptosis was largely abrogated when SLC7A11-mediated cystine uptake was inhibited by the knockout of SLC7A11 or erastin treatment. Besides, high BAP1 expression showed lower NADP<sup>+</sup>/NADPH levels, which might confer resistance to disulfidptosis. Consistent with these observations, the expression level of BAP1 was also positively correlated with NADPH-related genes in KIRC patients, though the underlying mechanism mediating NADPH regulation remains further investigation. In summary, our results revealed the role of BAP1 in the regulation disulfidptosis and provided new insights into the understanding of disulfidptosis in tumor development.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142189059","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
Peptidylarginine deiminase 3 modulates response to neratinib in HER2 positive breast cancer. 肽基精氨酸脱氨酶3调节HER2阳性乳腺癌患者对奈瑞替尼的反应
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-04 DOI: 10.1038/s41389-024-00531-4
Inés Romero-Pérez, Elena Díaz-Rodríguez, Laura Sánchez-Díaz, Juan Carlos Montero, Atanasio Pandiella

Neratinib is a tyrosine kinase inhibitor that is used for the therapy of patients with HER2+ breast tumors. However, despite its clinical benefit, resistance to the drug may arise. Here we have created cellular models of neratinib resistance to investigate the mechanisms underlying such resistance. Chronic neratinib exposure of BT474 human HER2+ breast cancer cells resulted in the selection of several clones resistant to the antiproliferative action of the drug. The clones were characterized biochemically and biologically using a variety of techniques. These clones retained HER2 levels similar to parental cells. Knockdown experiments showed that the neratinib-resistant clones retained oncogenic dependence on HER2. Moreover, the tyrosine phosphorylation status of BT474 and the resistant clones was equally sensitive to neratinib. Transcriptomic and Western analyses showed that peptidylarginine deiminase 3 was overexpressed in the three neratinib-resistant clones studied but was undetectable in BT474 cells. Experiments performed in the neratinib-resistant clones showed that reduction of PADI3 or inhibition of its function restored sensitivity to the antiproliferative action of neratinib. Moreover, overexpression of FLAG-tagged PADI3 in BT474 cells provoked resistance to the antiproliferative action of neratinib. Together, these results uncover a role of PADI3 in the regulation of sensitivity to neratinib in breast cancer cells overexpressing HER2 and open the possibility of using PADI3 inhibitors to fight resistance to neratinib.

奈拉替尼是一种酪氨酸激酶抑制剂,用于治疗HER2+乳腺肿瘤患者。然而,尽管奈拉替尼具有临床疗效,但也可能产生耐药性。在这里,我们创建了奈拉替尼耐药性细胞模型,以研究这种耐药性的机制。BT474人类HER2+乳腺癌细胞长期暴露于奈拉替尼后,筛选出了几个对药物抗增殖作用具有耐药性的克隆。利用多种技术对这些克隆进行了生物化学和生物学鉴定。这些克隆保留了与亲代细胞相似的 HER2 水平。基因敲除实验表明,奈拉替尼耐药克隆保留了对HER2的致癌依赖性。此外,BT474和耐药克隆的酪氨酸磷酸化状态对奈拉替尼同样敏感。转录组和Western分析表明,肽精氨酸脱氨酶3在研究的三个奈拉替尼耐药克隆中过表达,但在BT474细胞中检测不到。在奈拉替尼耐药克隆中进行的实验表明,减少 PADI3 或抑制其功能可恢复对奈拉替尼抗增殖作用的敏感性。此外,在BT474细胞中过表达FLAG标记的PADI3会引起对奈拉替尼抗增殖作用的耐药性。总之,这些结果揭示了PADI3在调控过表达HER2的乳腺癌细胞对奈拉替尼的敏感性中的作用,并为使用PADI3抑制剂对抗奈拉替尼耐药性提供了可能性。
{"title":"Peptidylarginine deiminase 3 modulates response to neratinib in HER2 positive breast cancer.","authors":"Inés Romero-Pérez, Elena Díaz-Rodríguez, Laura Sánchez-Díaz, Juan Carlos Montero, Atanasio Pandiella","doi":"10.1038/s41389-024-00531-4","DOIUrl":"10.1038/s41389-024-00531-4","url":null,"abstract":"<p><p>Neratinib is a tyrosine kinase inhibitor that is used for the therapy of patients with HER2+ breast tumors. However, despite its clinical benefit, resistance to the drug may arise. Here we have created cellular models of neratinib resistance to investigate the mechanisms underlying such resistance. Chronic neratinib exposure of BT474 human HER2+ breast cancer cells resulted in the selection of several clones resistant to the antiproliferative action of the drug. The clones were characterized biochemically and biologically using a variety of techniques. These clones retained HER2 levels similar to parental cells. Knockdown experiments showed that the neratinib-resistant clones retained oncogenic dependence on HER2. Moreover, the tyrosine phosphorylation status of BT474 and the resistant clones was equally sensitive to neratinib. Transcriptomic and Western analyses showed that peptidylarginine deiminase 3 was overexpressed in the three neratinib-resistant clones studied but was undetectable in BT474 cells. Experiments performed in the neratinib-resistant clones showed that reduction of PADI3 or inhibition of its function restored sensitivity to the antiproliferative action of neratinib. Moreover, overexpression of FLAG-tagged PADI3 in BT474 cells provoked resistance to the antiproliferative action of neratinib. Together, these results uncover a role of PADI3 in the regulation of sensitivity to neratinib in breast cancer cells overexpressing HER2 and open the possibility of using PADI3 inhibitors to fight resistance to neratinib.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297914/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141889812","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
Combined Inhibition of PI3K and STAT3 signaling effectively inhibits bladder cancer growth 联合抑制 PI3K 和 STAT3 信号传导可有效抑制膀胱癌生长
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-27 DOI: 10.1038/s41389-024-00529-y
Weidong Peng, Haojie Zhang, Mingwei Yin, Dejie Kong, Liping Kang, Xinkun Teng, Jingjing Wang, Zhimin Chu, Yating Sun, Pengpeng Long, Chengying Cui, Bin Lyu, Jinzhi Zhang, Han Xiao, Mingqing Wu, Yongqiang Wang, Yang Li

Bladder cancer is characterized by aberrant activation of the phosphatidylinositol-3-OH kinase (PI3K) signaling, underscoring the significance of directing therapeutic efforts toward the PI3K pathway as a promising strategy. In this study, we discovered that PI3K serves as a potent therapeutic target for bladder cancer through a high-throughput screening of inhibitory molecules. The PI3K inhibitor demonstrated a robust anti-tumor efficacy, validated both in vitro and in vivo settings. Nevertheless, the feedback activation of JAK1-STAT3 signaling reinstated cell and organoid survival, leading to resistance against the PI3K inhibitor. Mechanistically, the PI3K inhibitor suppresses PTPN11 expression, a negative regulator of the JAK-STAT pathway, thereby activating STAT3. Conversely, restoration of PTPN11 enhances the sensitivity of cancer cells to the PI3K inhibitor. Simultaneous inhibition of both PI3K and STAT3 with small-molecule inhibitors resulted in sustained tumor regression in patient-derived bladder cancer xenografts. These findings advocate for a combinational therapeutic approach targeting both PI3K and STAT3 pathways to achieve enduring cancer eradication in vitro and in vivo, underscoring their promising therapeutic efficacy for treating bladder cancer.

膀胱癌的特点是磷脂酰肌醇-3-OH 激酶(PI3K)信号的异常激活,这突出了将治疗工作引向 PI3K 通路作为一种有前途的策略的重要性。在这项研究中,我们通过高通量筛选抑制分子,发现PI3K是膀胱癌的有效治疗靶点。经体外和体内验证,PI3K 抑制剂具有强大的抗肿瘤功效。然而,JAK1-STAT3 信号的反馈激活恢复了细胞和类器官的存活,导致了对 PI3K 抑制剂的耐药性。从机理上讲,PI3K 抑制剂抑制了 JAK-STAT 通路的负调控因子 PTPN11 的表达,从而激活了 STAT3。相反,恢复 PTPN11 会增强癌细胞对 PI3K 抑制剂的敏感性。用小分子抑制剂同时抑制 PI3K 和 STAT3 可使源自患者的膀胱癌异种移植物的肿瘤持续消退。这些研究结果主张采用针对 PI3K 和 STAT3 通路的联合治疗方法,在体外和体内实现持久的癌症根除,强调了它们对治疗膀胱癌的巨大疗效。
{"title":"Combined Inhibition of PI3K and STAT3 signaling effectively inhibits bladder cancer growth","authors":"Weidong Peng, Haojie Zhang, Mingwei Yin, Dejie Kong, Liping Kang, Xinkun Teng, Jingjing Wang, Zhimin Chu, Yating Sun, Pengpeng Long, Chengying Cui, Bin Lyu, Jinzhi Zhang, Han Xiao, Mingqing Wu, Yongqiang Wang, Yang Li","doi":"10.1038/s41389-024-00529-y","DOIUrl":"https://doi.org/10.1038/s41389-024-00529-y","url":null,"abstract":"<p>Bladder cancer is characterized by aberrant activation of the phosphatidylinositol-3-OH kinase (PI3K) signaling, underscoring the significance of directing therapeutic efforts toward the PI3K pathway as a promising strategy. In this study, we discovered that PI3K serves as a potent therapeutic target for bladder cancer through a high-throughput screening of inhibitory molecules. The PI3K inhibitor demonstrated a robust anti-tumor efficacy, validated both in vitro and in vivo settings. Nevertheless, the feedback activation of JAK1-STAT3 signaling reinstated cell and organoid survival, leading to resistance against the PI3K inhibitor. Mechanistically, the PI3K inhibitor suppresses PTPN11 expression, a negative regulator of the JAK-STAT pathway, thereby activating STAT3. Conversely, restoration of PTPN11 enhances the sensitivity of cancer cells to the PI3K inhibitor. Simultaneous inhibition of both PI3K and STAT3 with small-molecule inhibitors resulted in sustained tumor regression in patient-derived bladder cancer xenografts. These findings advocate for a combinational therapeutic approach targeting both PI3K and STAT3 pathways to achieve enduring cancer eradication in vitro and in vivo, underscoring their promising therapeutic efficacy for treating bladder cancer.</p><figure></figure>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141782188","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
Cell competition in primary and metastatic colorectal cancer. 原发性和转移性结直肠癌中的细胞竞争。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-26 DOI: 10.1038/s41389-024-00530-5
Merel Elise van Luyk, Ana Krotenberg Garcia, Maria Lamprou, Saskia Jacoba Elisabeth Suijkerbuijk

Adult tissues set the scene for a continuous battle between cells, where a comparison of cellular fitness results in the elimination of weaker "loser" cells. This phenomenon, named cell competition, is beneficial for tissue integrity and homeostasis. In fact, cell competition plays a crucial role in tumor suppression, through elimination of early malignant cells, as part of Epithelial Defense Against Cancer. However, it is increasingly apparent that cell competition doubles as a tumor-promoting mechanism. The comparative nature of cell competition means that mutational background, proliferation rate and polarity all factor in to determine the outcome of these processes. In this review, we explore the intricate and context-dependent involvement of cell competition in homeostasis and regeneration, as well as during initiation and progression of primary and metastasized colorectal cancer. We provide a comprehensive overview of molecular and cellular mechanisms governing cell competition and its parallels with regeneration.

成体组织中的细胞之间会进行持续的较量,通过比较细胞的健康状况,淘汰较弱的 "失败者 "细胞。这种现象被称为细胞竞争,有利于组织的完整性和平衡。事实上,作为上皮细胞抗癌防御的一部分,细胞竞争通过消灭早期恶性细胞,在抑制肿瘤方面发挥着至关重要的作用。然而,越来越明显的是,细胞竞争同时也是一种肿瘤促进机制。细胞竞争的比较性质意味着突变背景、增殖率和极性都是决定这些过程结果的因素。在这篇综述中,我们探讨了细胞竞争在平衡和再生过程中,以及在原发性和转移性结直肠癌的发生和发展过程中错综复杂且与环境相关的参与。我们全面概述了细胞竞争的分子和细胞机制及其与再生的相似之处。
{"title":"Cell competition in primary and metastatic colorectal cancer.","authors":"Merel Elise van Luyk, Ana Krotenberg Garcia, Maria Lamprou, Saskia Jacoba Elisabeth Suijkerbuijk","doi":"10.1038/s41389-024-00530-5","DOIUrl":"10.1038/s41389-024-00530-5","url":null,"abstract":"<p><p>Adult tissues set the scene for a continuous battle between cells, where a comparison of cellular fitness results in the elimination of weaker \"loser\" cells. This phenomenon, named cell competition, is beneficial for tissue integrity and homeostasis. In fact, cell competition plays a crucial role in tumor suppression, through elimination of early malignant cells, as part of Epithelial Defense Against Cancer. However, it is increasingly apparent that cell competition doubles as a tumor-promoting mechanism. The comparative nature of cell competition means that mutational background, proliferation rate and polarity all factor in to determine the outcome of these processes. In this review, we explore the intricate and context-dependent involvement of cell competition in homeostasis and regeneration, as well as during initiation and progression of primary and metastasized colorectal cancer. We provide a comprehensive overview of molecular and cellular mechanisms governing cell competition and its parallels with regeneration.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11282291/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141767003","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
USP32 facilitates non-small cell lung cancer progression via deubiquitinating BAG3 and activating RAF-MEK-ERK signaling pathway. USP32 通过去泛素化 BAG3 和激活 RAF-MEK-ERK 信号通路促进非小细胞肺癌的进展。
IF 5.9 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-19 DOI: 10.1038/s41389-024-00528-z
Shuang Li, Lina Yang, Xiaoyan Ding, Hongxiao Sun, Xiaolei Dong, Fanghao Yang, Mengjun Wang, Huhu Zhang, Ya Li, Bing Li, Chunyan Liu

The regulatory significance of ubiquitin-specific peptidase 32 (USP32) in tumor is significant, nevertheless, the biological roles and regulatory mechanisms of USP32 in non-small cell lung cancer (NSCLC) remain unclear. According to our research, USP32 was strongly expressed in NSCLC cell lines and tissues and was linked to a bad prognosis for NSCLC patients. Interference with USP32 resulted in a significant inhibition of NSCLC cell proliferation, migration potential, and EMT development; on the other hand, USP32 overexpression had the opposite effect. To further elucidate the mechanism of action of USP32 in NSCLC, we screened H1299 cells for interacting proteins and found that USP32 interacts with BAG3 (Bcl2-associated athanogene 3) and deubiquitinates and stabilizes BAG3 in a deubiquitinating activity-dependent manner. Functionally, restoration of BAG3 expression abrogated the antitumor effects of USP32 silencing. Furthermore, USP32 increased the phosphorylation level of the RAF/MEK/ERK signaling pathway in NSCLC cells by stabilizing BAG3. In summary, these findings imply that USP32 is critical to the development of NSCLC and could offer a theoretical framework for the clinical diagnosis and management of NSCLC patients in the future.

泛素特异性肽酶32(USP32)在肿瘤中的调控意义重大,然而,USP32在非小细胞肺癌(NSCLC)中的生物学作用和调控机制仍不清楚。根据我们的研究,USP32 在 NSCLC 细胞系和组织中强表达,并且与 NSCLC 患者的不良预后有关。干扰 USP32 会显著抑制 NSCLC 细胞的增殖、迁移潜能和 EMT 的发展;另一方面,USP32 的过表达则会产生相反的效果。为了进一步阐明USP32在NSCLC中的作用机制,我们筛选了H1299细胞中的相互作用蛋白,发现USP32与BAG3(Bcl2-associated athanogene 3)相互作用,并以去泛素活性依赖的方式去泛素化和稳定BAG3。从功能上讲,恢复 BAG3 的表达会减弱 USP32 沉默的抗肿瘤作用。此外,USP32 通过稳定 BAG3 增加了 NSCLC 细胞中 RAF/MEK/ERK 信号通路的磷酸化水平。总之,这些研究结果表明,USP32 对 NSCLC 的发展至关重要,可为今后 NSCLC 患者的临床诊断和管理提供一个理论框架。
{"title":"USP32 facilitates non-small cell lung cancer progression via deubiquitinating BAG3 and activating RAF-MEK-ERK signaling pathway.","authors":"Shuang Li, Lina Yang, Xiaoyan Ding, Hongxiao Sun, Xiaolei Dong, Fanghao Yang, Mengjun Wang, Huhu Zhang, Ya Li, Bing Li, Chunyan Liu","doi":"10.1038/s41389-024-00528-z","DOIUrl":"10.1038/s41389-024-00528-z","url":null,"abstract":"<p><p>The regulatory significance of ubiquitin-specific peptidase 32 (USP32) in tumor is significant, nevertheless, the biological roles and regulatory mechanisms of USP32 in non-small cell lung cancer (NSCLC) remain unclear. According to our research, USP32 was strongly expressed in NSCLC cell lines and tissues and was linked to a bad prognosis for NSCLC patients. Interference with USP32 resulted in a significant inhibition of NSCLC cell proliferation, migration potential, and EMT development; on the other hand, USP32 overexpression had the opposite effect. To further elucidate the mechanism of action of USP32 in NSCLC, we screened H1299 cells for interacting proteins and found that USP32 interacts with BAG3 (Bcl2-associated athanogene 3) and deubiquitinates and stabilizes BAG3 in a deubiquitinating activity-dependent manner. Functionally, restoration of BAG3 expression abrogated the antitumor effects of USP32 silencing. Furthermore, USP32 increased the phosphorylation level of the RAF/MEK/ERK signaling pathway in NSCLC cells by stabilizing BAG3. In summary, these findings imply that USP32 is critical to the development of NSCLC and could offer a theoretical framework for the clinical diagnosis and management of NSCLC patients in the future.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11271578/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141727512","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncogenesis
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号: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