首页 > 最新文献

Oncogenesis最新文献

英文 中文
Spindle function and Wnt pathway inhibition by PBX1 to suppress tumor progression via downregulating DCDC2 in colorectal cancer. 结直肠癌中PBX1抑制纺锤体功能和Wnt通路通过下调DCDC2抑制肿瘤进展。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-02-04 DOI: 10.1038/s41389-023-00448-4
Weigang Dai, Yinan Liu, Tianhao Zhang, Zhixin Huang, Xiang Xu, Zeyu Zhao, Jianqiu Liu, Ertao Zhai, Shirong Cai, Jianhui Chen

PBX1 is a transcription factor that regulates a variety of genes, involved in intracellular lipid metabolism, cell proliferation, and other pathways. The promoting and inhibiting function of PBX1 in various cancer types was extensively discussed, however, there have been no studies on PBX1 proteins in colorectal cancer (CRC). This study aimed to reveal the anti-tumor function of PBX1 in CRC and the underlying molecular mechanism. Bioinformatics analysis revealed that PBX1 is downregulated in CRC, indicating that is a potential antioncogene in CRC. Overexpression of PBX1 suppresses tumor growth and metastasis in vitro and in vivo. Mechanistically, we found that PBX1 acted as a transcription factor that suppressed DCDC2 expression and inhibited spindle function. Moreover, the PBX1-DCDC2 axis controlled the Wnt pathway in CRC cells. Overexpression of DCDC2 restored CRC proliferation, metastasis abilities and Wnt pathway. In conclusion, this study suggests that PBX1 acts as a transcription factor to suppress DCDC2 expression and inhibit cell proliferation and metastasis by disrupting spindle function and the Wnt pathway in CRC.

PBX1是一种调节多种基因的转录因子,参与细胞内脂质代谢、细胞增殖等途径。PBX1在各种癌症类型中的促进和抑制作用已被广泛讨论,但PBX1蛋白在结直肠癌(CRC)中的研究尚未见报道。本研究旨在揭示PBX1在结直肠癌中的抗肿瘤功能及其分子机制。生物信息学分析显示,PBX1在结直肠癌中下调,表明它是结直肠癌中潜在的反基因。在体内和体外,过表达PBX1抑制肿瘤的生长和转移。在机制上,我们发现PBX1作为转录因子抑制DCDC2表达并抑制纺锤体功能。此外,PBX1-DCDC2轴控制CRC细胞中的Wnt通路。过表达DCDC2可恢复结直肠癌的增殖、转移能力和Wnt通路。综上所述,本研究提示PBX1作为一种转录因子,通过破坏结直肠癌的纺锤体功能和Wnt通路,抑制DCDC2的表达,抑制细胞增殖和转移。
{"title":"Spindle function and Wnt pathway inhibition by PBX1 to suppress tumor progression via downregulating DCDC2 in colorectal cancer.","authors":"Weigang Dai,&nbsp;Yinan Liu,&nbsp;Tianhao Zhang,&nbsp;Zhixin Huang,&nbsp;Xiang Xu,&nbsp;Zeyu Zhao,&nbsp;Jianqiu Liu,&nbsp;Ertao Zhai,&nbsp;Shirong Cai,&nbsp;Jianhui Chen","doi":"10.1038/s41389-023-00448-4","DOIUrl":"https://doi.org/10.1038/s41389-023-00448-4","url":null,"abstract":"<p><p>PBX1 is a transcription factor that regulates a variety of genes, involved in intracellular lipid metabolism, cell proliferation, and other pathways. The promoting and inhibiting function of PBX1 in various cancer types was extensively discussed, however, there have been no studies on PBX1 proteins in colorectal cancer (CRC). This study aimed to reveal the anti-tumor function of PBX1 in CRC and the underlying molecular mechanism. Bioinformatics analysis revealed that PBX1 is downregulated in CRC, indicating that is a potential antioncogene in CRC. Overexpression of PBX1 suppresses tumor growth and metastasis in vitro and in vivo. Mechanistically, we found that PBX1 acted as a transcription factor that suppressed DCDC2 expression and inhibited spindle function. Moreover, the PBX1-DCDC2 axis controlled the Wnt pathway in CRC cells. Overexpression of DCDC2 restored CRC proliferation, metastasis abilities and Wnt pathway. In conclusion, this study suggests that PBX1 acts as a transcription factor to suppress DCDC2 expression and inhibit cell proliferation and metastasis by disrupting spindle function and the Wnt pathway in CRC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2023-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9899229/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9228719","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
HLTF promotes hepatocellular carcinoma progression by enhancing SRSF1 stability and activating ERK/MAPK pathway. htf通过增强SRSF1的稳定性和激活ERK/MAPK通路来促进肝细胞癌的进展。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-01-20 DOI: 10.1038/s41389-023-00447-5
Yanan Xu, Shanjia Ke, Shounan Lu, Chaoqun Wang, Zihao Li, Zhigang Feng, Hongjun Yu, Miaoyu Bai, Baolin Qian, Bing Yin, Xinglong Li, Yongliang Hua, Hongchi Jiang, Yong Ma

Helicase-like transcription factor (HLTF) has been found to be involved in the progression of several tumors, but the role of HLTF in hepatocellular carcinoma (HCC) progression has not been studied. Here, our study explored the underlying mechanism of HLTF in HCC progression for the first time. Database analysis and clinical sample examination indicated that HLTF was upregulated in HCC tissues and was related to poor clinicopathological features in patients. Upregulation of HLTF accelerated the growth and metastasis of HCC cells both in vitro and in vivo. Bioinformatics analysis and subsequent experiments revealed that ERK/MAPK signaling pathway activation was vital to HLTF-mediated proliferation and metastasis in HCC cells. Moreover, HLTF was demonstrated to interact with SRSF1 and contribute to its protein stability to activate the ERK/MAPK signaling pathway and enhance HCC growth and metastasis. In addition, miR-511-5p was expressed at a low level in HCC tissues, was negatively correlated HLTF, and regulated HLTF expression. Our study shows that HLTF plays an oncogenic role in HCC progression and provides a novel biomarker and therapeutic target for the diagnosis and treatment of HCC.

解旋酶样转录因子(HLTF)已被发现参与多种肿瘤的进展,但HLTF在肝细胞癌(HCC)进展中的作用尚未被研究。本研究首次探讨了HLTF在HCC进展中的潜在机制。数据库分析和临床样本检查提示HCC组织中HLTF表达上调,与患者临床病理特征较差有关。在体外和体内,HLTF的上调加速了HCC细胞的生长和转移。生物信息学分析和随后的实验表明,ERK/MAPK信号通路的激活对hltf介导的HCC细胞增殖和转移至关重要。此外,HLTF被证明与SRSF1相互作用,并有助于其蛋白稳定性,激活ERK/MAPK信号通路,促进HCC的生长和转移。此外,miR-511-5p在HCC组织中低水平表达,与HLTF呈负相关,并调节HLTF的表达。我们的研究表明,HLTF在HCC的进展中起着致癌作用,为HCC的诊断和治疗提供了新的生物标志物和治疗靶点。
{"title":"HLTF promotes hepatocellular carcinoma progression by enhancing SRSF1 stability and activating ERK/MAPK pathway.","authors":"Yanan Xu,&nbsp;Shanjia Ke,&nbsp;Shounan Lu,&nbsp;Chaoqun Wang,&nbsp;Zihao Li,&nbsp;Zhigang Feng,&nbsp;Hongjun Yu,&nbsp;Miaoyu Bai,&nbsp;Baolin Qian,&nbsp;Bing Yin,&nbsp;Xinglong Li,&nbsp;Yongliang Hua,&nbsp;Hongchi Jiang,&nbsp;Yong Ma","doi":"10.1038/s41389-023-00447-5","DOIUrl":"https://doi.org/10.1038/s41389-023-00447-5","url":null,"abstract":"<p><p>Helicase-like transcription factor (HLTF) has been found to be involved in the progression of several tumors, but the role of HLTF in hepatocellular carcinoma (HCC) progression has not been studied. Here, our study explored the underlying mechanism of HLTF in HCC progression for the first time. Database analysis and clinical sample examination indicated that HLTF was upregulated in HCC tissues and was related to poor clinicopathological features in patients. Upregulation of HLTF accelerated the growth and metastasis of HCC cells both in vitro and in vivo. Bioinformatics analysis and subsequent experiments revealed that ERK/MAPK signaling pathway activation was vital to HLTF-mediated proliferation and metastasis in HCC cells. Moreover, HLTF was demonstrated to interact with SRSF1 and contribute to its protein stability to activate the ERK/MAPK signaling pathway and enhance HCC growth and metastasis. In addition, miR-511-5p was expressed at a low level in HCC tissues, was negatively correlated HLTF, and regulated HLTF expression. Our study shows that HLTF plays an oncogenic role in HCC progression and provides a novel biomarker and therapeutic target for the diagnosis and treatment of HCC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2023-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9859789/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10575771","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: 3
AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer. AKT1调节UHRF1蛋白稳定性,促进前列腺癌对阿比特龙的抵抗。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2023-01-02 DOI: 10.1038/s41389-022-00446-y
Yongming Fu, Tuoyu Cao, Xiaorui Zou, Yubing Ye, Youhong Liu, Yuchong Peng, Tanggang Deng, Linglong Yin, Xiong Li

Oncogenic activation of PI3K/AKT signaling pathway, together with epigenetic aberrations are the characters of castration-resistant prostate cancer (CRPC). UHRF1 as a key epigenetic regulator, plays a critical role in prostate cancer (PCa) development, and its expression is positively correlated with the degree of malignancy. In this present study we investigated the potential regulatory mechanism of AKT1 on UHRF1, and further validated the in vitro and in vivo anticancer efficacy of AKT phosphorylation inhibitor MK2206 in combination with abiraterone. Both UHRF1 and p-AKT aberrantly overexpressed in the abiraterone-resistant PCa cells. Further studies revealed that AKT1 protein interacts with UHRF1, and AKT1 directly phosphorylates UHRF1 via the site Thr-210. MK2206 induced UHRF1 protein degradation by inhibiting AKT1-induced UHRF1 phosphorylation, and then reduced the interaction between UHRF1 and deubiquitinase USP7, while promoted the interaction between UHRF1 and E3 ubiquitin protein ligase BTRC. MK2206 significantly promoted the sensitivity of abiraterone-refractory PCa cells and xenografts to abiraterone by decreasing UHRF1 protein level, and reversed the phenotype of NEPC, evently induced cellular senescence and cell apoptosis. Altogether, our present study for the first time revealed a novel molecular mechanism of abiraterone resistance through PI3K/AKT-UHRF1 pathway, and provided a novel therapeutic modality by targeting PI3K/AKT1 to promote the drug sensitivity of abiraterone in PCa patients.

PI3K/AKT信号通路的致瘤激活以及表观遗传畸变是去势抵抗性前列腺癌(CRPC)的特征。UHRF1作为一种关键的表观遗传调控因子,在前列腺癌(prostate cancer, PCa)的发生发展中起着至关重要的作用,其表达与恶性程度呈正相关。本研究探讨了AKT1对UHRF1的潜在调控机制,进一步验证了AKT磷酸化抑制剂MK2206联合阿比特龙在体外和体内的抗癌效果。UHRF1和p-AKT在阿比特龙耐药PCa细胞中均异常过表达。进一步的研究发现AKT1蛋白与UHRF1相互作用,AKT1通过位点Thr-210直接磷酸化UHRF1。MK2206通过抑制akt1诱导的UHRF1磷酸化诱导UHRF1蛋白降解,进而降低UHRF1与去泛素酶USP7的相互作用,促进UHRF1与E3泛素蛋白连接酶BTRC的相互作用。MK2206通过降低UHRF1蛋白水平,显著提高阿比特龙难愈性PCa细胞和异种移植物对阿比特龙的敏感性,逆转NEPC表型,诱导细胞衰老和细胞凋亡。综上所述,本研究首次通过PI3K/AKT-UHRF1途径揭示了阿比特龙耐药的新分子机制,并提供了一种以PI3K/AKT1为靶点促进阿比特龙在PCa患者药物敏感性的新治疗模式。
{"title":"AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer.","authors":"Yongming Fu,&nbsp;Tuoyu Cao,&nbsp;Xiaorui Zou,&nbsp;Yubing Ye,&nbsp;Youhong Liu,&nbsp;Yuchong Peng,&nbsp;Tanggang Deng,&nbsp;Linglong Yin,&nbsp;Xiong Li","doi":"10.1038/s41389-022-00446-y","DOIUrl":"https://doi.org/10.1038/s41389-022-00446-y","url":null,"abstract":"<p><p>Oncogenic activation of PI3K/AKT signaling pathway, together with epigenetic aberrations are the characters of castration-resistant prostate cancer (CRPC). UHRF1 as a key epigenetic regulator, plays a critical role in prostate cancer (PCa) development, and its expression is positively correlated with the degree of malignancy. In this present study we investigated the potential regulatory mechanism of AKT1 on UHRF1, and further validated the in vitro and in vivo anticancer efficacy of AKT phosphorylation inhibitor MK2206 in combination with abiraterone. Both UHRF1 and p-AKT aberrantly overexpressed in the abiraterone-resistant PCa cells. Further studies revealed that AKT1 protein interacts with UHRF1, and AKT1 directly phosphorylates UHRF1 via the site Thr-210. MK2206 induced UHRF1 protein degradation by inhibiting AKT1-induced UHRF1 phosphorylation, and then reduced the interaction between UHRF1 and deubiquitinase USP7, while promoted the interaction between UHRF1 and E3 ubiquitin protein ligase BTRC. MK2206 significantly promoted the sensitivity of abiraterone-refractory PCa cells and xenografts to abiraterone by decreasing UHRF1 protein level, and reversed the phenotype of NEPC, evently induced cellular senescence and cell apoptosis. Altogether, our present study for the first time revealed a novel molecular mechanism of abiraterone resistance through PI3K/AKT-UHRF1 pathway, and provided a novel therapeutic modality by targeting PI3K/AKT1 to promote the drug sensitivity of abiraterone in PCa patients.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2023-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9807647/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10487103","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: 3
LPA receptor 1 (LPAR1) is a novel interaction partner of Filamin A that promotes Filamin A phosphorylation, MRTF-A transcriptional activity and oncogene-induced senescence. LPA受体1 (LPAR1)是丝蛋白a的新型相互作用伙伴,可促进丝蛋白a磷酸化、MRTF-A转录活性和癌基因诱导的衰老。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-12-28 DOI: 10.1038/s41389-022-00445-z
Andreas Konopa, Melanie A Meier, Miriam J Franz, Emanuele Bernardinelli, Anna-Lena Voegele, Raja Atreya, Silvia Ribback, Stephanie Roessler, Achim Aigner, Kerstin Singer, Stephan Singer, Antonio Sarikas, Susanne Muehlich

Myocardin-related transcription factors A and B (MRTFs) are coactivators of Serum Response Factor (SRF), which controls fundamental biological processes such as cell growth, migration, and differentiation. MRTF and SRF transcriptional activity play an important role in hepatocellular carcinoma (HCC) growth, which represents the second leading cause of cancer-related mortality in humans worldwide. We, therefore, searched for druggable targets in HCC that regulate MRTF/SRF transcriptional activity and can be exploited therapeutically for HCC therapy. We identified the G protein-coupled lysophosphatidic acid receptor 1 (LPAR1) as a novel interaction partner of MRTF-A and Filamin A (FLNA) using fluorescence resonance energy transfer-(FRET) and proximity ligation assay (PLA) in vitro in HCC cells and in vivo in organoids. We found that LPAR1 promotes FLNA phosphorylation at S2152 which enhances the complex formation of FLNA and MRTF-A, actin polymerization, and MRTF transcriptional activity. Pharmacological blockade or depletion of LPAR1 prevents FLNA phosphorylation and complex formation with MRTF-A, resulting in reduced MRTF/SRF target gene expression and oncogene-induced senescence. Thus, inhibition of the LPAR1-FLNA-MRTF-A interaction represents a promising strategy for HCC therapy.

心肌素相关转录因子A和B (mrtf)是血清反应因子(SRF)的共激活因子,SRF控制着基本的生物过程,如细胞生长、迁移和分化。MRTF和SRF转录活性在肝细胞癌(HCC)生长中发挥重要作用,HCC是全球人类癌症相关死亡的第二大原因。因此,我们在HCC中寻找可调节MRTF/SRF转录活性并可用于HCC治疗的药物靶点。我们利用荧光共振能量转移(FRET)和近距离结联实验(PLA)在体外HCC细胞和体内类器官中鉴定了G蛋白偶联溶血磷脂酸受体1 (LPAR1)作为mrtnf - a和丝蛋白a (FLNA)的新型相互作用伙伴。我们发现LPAR1促进FLNA在S2152位点的磷酸化,从而增强FLNA和MRTF- a复合物的形成、肌动蛋白聚合和MRTF转录活性。药物阻断或耗尽LPAR1可阻止FLNA磷酸化并与MRTF- a形成复合物,导致MRTF/SRF靶基因表达降低和癌基因诱导的衰老。因此,抑制LPAR1-FLNA-MRTF-A相互作用是HCC治疗的一种有希望的策略。
{"title":"LPA receptor 1 (LPAR1) is a novel interaction partner of Filamin A that promotes Filamin A phosphorylation, MRTF-A transcriptional activity and oncogene-induced senescence.","authors":"Andreas Konopa,&nbsp;Melanie A Meier,&nbsp;Miriam J Franz,&nbsp;Emanuele Bernardinelli,&nbsp;Anna-Lena Voegele,&nbsp;Raja Atreya,&nbsp;Silvia Ribback,&nbsp;Stephanie Roessler,&nbsp;Achim Aigner,&nbsp;Kerstin Singer,&nbsp;Stephan Singer,&nbsp;Antonio Sarikas,&nbsp;Susanne Muehlich","doi":"10.1038/s41389-022-00445-z","DOIUrl":"https://doi.org/10.1038/s41389-022-00445-z","url":null,"abstract":"<p><p>Myocardin-related transcription factors A and B (MRTFs) are coactivators of Serum Response Factor (SRF), which controls fundamental biological processes such as cell growth, migration, and differentiation. MRTF and SRF transcriptional activity play an important role in hepatocellular carcinoma (HCC) growth, which represents the second leading cause of cancer-related mortality in humans worldwide. We, therefore, searched for druggable targets in HCC that regulate MRTF/SRF transcriptional activity and can be exploited therapeutically for HCC therapy. We identified the G protein-coupled lysophosphatidic acid receptor 1 (LPAR1) as a novel interaction partner of MRTF-A and Filamin A (FLNA) using fluorescence resonance energy transfer-(FRET) and proximity ligation assay (PLA) in vitro in HCC cells and in vivo in organoids. We found that LPAR1 promotes FLNA phosphorylation at S2152 which enhances the complex formation of FLNA and MRTF-A, actin polymerization, and MRTF transcriptional activity. Pharmacological blockade or depletion of LPAR1 prevents FLNA phosphorylation and complex formation with MRTF-A, resulting in reduced MRTF/SRF target gene expression and oncogene-induced senescence. Thus, inhibition of the LPAR1-FLNA-MRTF-A interaction represents a promising strategy for HCC therapy.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9797565/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10512964","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: 2
Correction: Overexpression of SMC4 activates TGFβ/Smad signaling and promotes aggressive phenotype in glioma cells. 更正:SMC4的过表达激活TGFβ/Smad信号传导并促进胶质瘤细胞的侵袭性表型。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-11-18 DOI: 10.1038/s41389-022-00442-2
L Jiang, J Zhou, D Zhong, Y Zhou, W Zhang, W Wu, Z Zhao, W Wang, W Xu, L He, Y Ma, Y Hu, W Zhang, J Li
{"title":"Correction: Overexpression of SMC4 activates TGFβ/Smad signaling and promotes aggressive phenotype in glioma cells.","authors":"L Jiang,&nbsp;J Zhou,&nbsp;D Zhong,&nbsp;Y Zhou,&nbsp;W Zhang,&nbsp;W Wu,&nbsp;Z Zhao,&nbsp;W Wang,&nbsp;W Xu,&nbsp;L He,&nbsp;Y Ma,&nbsp;Y Hu,&nbsp;W Zhang,&nbsp;J Li","doi":"10.1038/s41389-022-00442-2","DOIUrl":"https://doi.org/10.1038/s41389-022-00442-2","url":null,"abstract":"","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9674687/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40695750","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
Ablation of sphingosine kinase 2 suppresses fatty liver-associated hepatocellular carcinoma via downregulation of ceramide transfer protein. 鞘氨醇激酶2的消融通过下调神经酰胺转移蛋白抑制脂肪肝相关肝细胞癌。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-11-04 DOI: 10.1038/s41389-022-00444-0
Xin Tracy Liu, Long Hoa Chung, Da Liu, Jinbiao Chen, Yu Huang, Jonathan D Teo, Xingxing Daisy Han, Yinan Zhao, Fiona H X Guan, Collin Tran, Jun Yup Lee, Timothy A Couttas, Ken Liu, Geoffery W McCaughan, Mark D Gorrell, Anthony S Don, Shubiao Zhang, Yanfei Qi

Hepatocellular carcinoma (HCC) accounts for 90% of primary liver cancer, the third leading cause of cancer-associated death worldwide. With the increasing prevalence of metabolic conditions, non-alcoholic fatty liver disease (NAFLD) is emerging as the fastest-growing HCC risk factor, and it imposes an additional layer of difficulty in HCC management. Dysregulated hepatic lipids are generally believed to constitute a deleterious environment cultivating the development of NAFLD-associated HCC. However, exactly which lipids or lipid regulators drive this process remains elusive. We report herein that sphingosine kinase 2 (SphK2), a key sphingolipid metabolic enzyme, plays a critical role in NAFLD-associated HCC. Ablation of Sphk2 suppressed HCC development in NAFLD livers via inhibition of hepatocyte proliferation both in vivo and in vitro. Mechanistically, SphK2 deficiency led to downregulation of ceramide transfer protein (CERT) that, in turn, decreased the ratio of pro-cancer sphingomyelin (SM) to anti-cancer ceramide. Overexpression of CERT restored hepatocyte proliferation, colony growth and cell cycle progression. In conclusion, the current study demonstrates that SphK2 is an essential lipid regulator in NAFLD-associated HCC, providing experimental evidence to support clinical trials of SphK2 inhibitors as systemic therapies against HCC.

肝细胞癌(HCC)占原发性肝癌的90%,是全球癌症相关死亡的第三大原因。随着代谢疾病的日益流行,非酒精性脂肪性肝病(NAFLD)正成为增长最快的HCC危险因素,并给HCC治疗带来了额外的困难。一般认为,肝脏脂质失调构成了一个有害的环境,促进了nafld相关HCC的发展。然而,究竟是哪种脂质或脂质调节剂驱动了这一过程仍然难以捉摸。我们在此报道鞘磷脂激酶2 (SphK2)是一种关键的鞘脂代谢酶,在nafld相关的HCC中起关键作用。在体内和体外,消融Sphk2通过抑制肝细胞增殖来抑制NAFLD肝脏中HCC的发展。机制上,SphK2缺乏导致神经酰胺传递蛋白(CERT)下调,进而降低促癌鞘磷脂(SM)与抗癌神经酰胺的比例。过表达CERT可恢复肝细胞增殖、集落生长和细胞周期进程。总之,目前的研究表明SphK2是nafld相关HCC中必需的脂质调节剂,为SphK2抑制剂作为HCC全身治疗的临床试验提供了实验证据。
{"title":"Ablation of sphingosine kinase 2 suppresses fatty liver-associated hepatocellular carcinoma via downregulation of ceramide transfer protein.","authors":"Xin Tracy Liu,&nbsp;Long Hoa Chung,&nbsp;Da Liu,&nbsp;Jinbiao Chen,&nbsp;Yu Huang,&nbsp;Jonathan D Teo,&nbsp;Xingxing Daisy Han,&nbsp;Yinan Zhao,&nbsp;Fiona H X Guan,&nbsp;Collin Tran,&nbsp;Jun Yup Lee,&nbsp;Timothy A Couttas,&nbsp;Ken Liu,&nbsp;Geoffery W McCaughan,&nbsp;Mark D Gorrell,&nbsp;Anthony S Don,&nbsp;Shubiao Zhang,&nbsp;Yanfei Qi","doi":"10.1038/s41389-022-00444-0","DOIUrl":"https://doi.org/10.1038/s41389-022-00444-0","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) accounts for 90% of primary liver cancer, the third leading cause of cancer-associated death worldwide. With the increasing prevalence of metabolic conditions, non-alcoholic fatty liver disease (NAFLD) is emerging as the fastest-growing HCC risk factor, and it imposes an additional layer of difficulty in HCC management. Dysregulated hepatic lipids are generally believed to constitute a deleterious environment cultivating the development of NAFLD-associated HCC. However, exactly which lipids or lipid regulators drive this process remains elusive. We report herein that sphingosine kinase 2 (SphK2), a key sphingolipid metabolic enzyme, plays a critical role in NAFLD-associated HCC. Ablation of Sphk2 suppressed HCC development in NAFLD livers via inhibition of hepatocyte proliferation both in vivo and in vitro. Mechanistically, SphK2 deficiency led to downregulation of ceramide transfer protein (CERT) that, in turn, decreased the ratio of pro-cancer sphingomyelin (SM) to anti-cancer ceramide. Overexpression of CERT restored hepatocyte proliferation, colony growth and cell cycle progression. In conclusion, the current study demonstrates that SphK2 is an essential lipid regulator in NAFLD-associated HCC, providing experimental evidence to support clinical trials of SphK2 inhibitors as systemic therapies against HCC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9636415/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666400","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: 5
DUSP5P1 promotes gastric cancer metastasis and platinum drug resistance. DUSP5P1促进胃癌转移和铂类药物耐药。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-10-28 DOI: 10.1038/s41389-022-00441-3
Xiaohong Wang, Lianhai Zhang, Qiaoyi Liang, Chi Chun Wong, Huarong Chen, Hongyan Gou, Yujuan Dong, Weixin Liu, Ziyu Li, Jiafu Ji, Jun Yu

We elucidated the functional significance and molecular mechanisms of DUSP5P1 lncRNA (dual specificity phosphatase 5 pseudogene 1) in gastric carcinogenesis. We demonstrated that gastric cancer (GC) patients with high DUSP5P1 expression had shortened survival in two independent cohorts. DUSP5P1 promoted GC cell migration and invasion in vitro and metastasis in vivo. Mechanistically, DUSP5P1 activated ARHGAP5 transcription by directly binding to the promoter of ARHGAP5 with a binding motif of TATGTG. RNA-seq revealed that ARHGAP5 activated focal adhesion and MAPK signaling pathways to promote GC metastasis. DUSP5P1 also dysregulated platinum drug resistance pathway. Consistently, DUSP5P1 overexpression in GC cells antagonized cytotoxic effect of Oxaliplatin, and shDUSP5P1 plus Oxaliplatin exerted synergistic effect on inhibiting GC metastasis in vitro and in vivo. DUSP5P1 depletion also suppressed the growth of platinum drug-resistant PDO models. In conclusion, DUSP5P1 promoted GC metastasis by directly modulating ARHGAP5 expression to activate focal adhesion and MAPK pathways, serves as therapeutic target for platinum drug resistant GC, and is an independent prognostic factor in GC.

我们阐明了DUSP5P1 lncRNA(双特异性磷酸酶5假基因1)在胃癌发生中的功能意义和分子机制。我们在两个独立的队列中证明了高DUSP5P1表达的胃癌(GC)患者的生存期缩短。DUSP5P1促进GC细胞在体外的迁移侵袭和体内的转移。机制上,DUSP5P1通过TATGTG结合基序直接结合ARHGAP5的启动子,激活ARHGAP5的转录。RNA-seq结果显示,ARHGAP5激活局灶黏附和MAPK信号通路,促进胃癌转移。DUSP5P1也会调控铂类药物耐药通路。在GC细胞中,DUSP5P1的过表达一致地拮抗奥沙利铂的细胞毒作用,shDUSP5P1与奥沙利铂在体外和体内均具有协同抑制GC转移的作用。DUSP5P1缺失也抑制了铂耐药PDO模型的生长。综上所述,DUSP5P1通过直接调节ARHGAP5表达激活局灶黏附和MAPK通路促进胃癌转移,是铂类耐药胃癌的治疗靶点,是胃癌的独立预后因素。
{"title":"DUSP5P1 promotes gastric cancer metastasis and platinum drug resistance.","authors":"Xiaohong Wang,&nbsp;Lianhai Zhang,&nbsp;Qiaoyi Liang,&nbsp;Chi Chun Wong,&nbsp;Huarong Chen,&nbsp;Hongyan Gou,&nbsp;Yujuan Dong,&nbsp;Weixin Liu,&nbsp;Ziyu Li,&nbsp;Jiafu Ji,&nbsp;Jun Yu","doi":"10.1038/s41389-022-00441-3","DOIUrl":"https://doi.org/10.1038/s41389-022-00441-3","url":null,"abstract":"<p><p>We elucidated the functional significance and molecular mechanisms of DUSP5P1 lncRNA (dual specificity phosphatase 5 pseudogene 1) in gastric carcinogenesis. We demonstrated that gastric cancer (GC) patients with high DUSP5P1 expression had shortened survival in two independent cohorts. DUSP5P1 promoted GC cell migration and invasion in vitro and metastasis in vivo. Mechanistically, DUSP5P1 activated ARHGAP5 transcription by directly binding to the promoter of ARHGAP5 with a binding motif of TATGTG. RNA-seq revealed that ARHGAP5 activated focal adhesion and MAPK signaling pathways to promote GC metastasis. DUSP5P1 also dysregulated platinum drug resistance pathway. Consistently, DUSP5P1 overexpression in GC cells antagonized cytotoxic effect of Oxaliplatin, and shDUSP5P1 plus Oxaliplatin exerted synergistic effect on inhibiting GC metastasis in vitro and in vivo. DUSP5P1 depletion also suppressed the growth of platinum drug-resistant PDO models. In conclusion, DUSP5P1 promoted GC metastasis by directly modulating ARHGAP5 expression to activate focal adhesion and MAPK pathways, serves as therapeutic target for platinum drug resistant GC, and is an independent prognostic factor in GC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9616843/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40654767","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: 3
The SUMO protease SENP1 promotes aggressive behaviors of high HIF2α expressing renal cell carcinoma cells SUMO蛋白酶SENP1促进高表达HIF2α的肾细胞癌细胞的侵袭行为
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-10-25 DOI: 10.1038/s41389-022-00440-4
M. Lee, Kyung E Sung, David Beebe, Wei-Ren Huang, Danielle Shapiro, S. Miyamoto, E. Abel
{"title":"The SUMO protease SENP1 promotes aggressive behaviors of high HIF2α expressing renal cell carcinoma cells","authors":"M. Lee, Kyung E Sung, David Beebe, Wei-Ren Huang, Danielle Shapiro, S. Miyamoto, E. Abel","doi":"10.1038/s41389-022-00440-4","DOIUrl":"https://doi.org/10.1038/s41389-022-00440-4","url":null,"abstract":"","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44939508","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: 3
Correction: Mitochondrial dysfunction and impaired growth of glioblastoma cell lines caused by antimicrobial agents inducing ferroptosis under glucose starvation. 更正:葡萄糖饥饿下抗微生物药物诱导铁下垂引起的胶质母细胞瘤细胞系线粒体功能障碍和生长受损。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-10-19 DOI: 10.1038/s41389-022-00443-1
Kenji Miki, Mikako Yagi, Koji Yoshimoto, Dongchon Kang, Takeshi Uchiumi
{"title":"Correction: Mitochondrial dysfunction and impaired growth of glioblastoma cell lines caused by antimicrobial agents inducing ferroptosis under glucose starvation.","authors":"Kenji Miki,&nbsp;Mikako Yagi,&nbsp;Koji Yoshimoto,&nbsp;Dongchon Kang,&nbsp;Takeshi Uchiumi","doi":"10.1038/s41389-022-00443-1","DOIUrl":"https://doi.org/10.1038/s41389-022-00443-1","url":null,"abstract":"","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9581926/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40655140","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
Retraction Note: Cadherin 6 is activated by Epstein-Barr virus LMP1 to mediate EMT and metastasis as an interplay node of multiple pathways in nasopharyngeal carcinoma. 注:在鼻咽癌中,钙粘蛋白6被Epstein-Barr病毒LMP1激活介导EMT和转移,是多途径相互作用的节点。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2022-10-17 DOI: 10.1038/s41389-022-00439-x
L-L Zuo, J Zhang, L- Z Liu, Q Zhou, S- J Du, S- Y Xin, Z- P Ning, J Yang, H- B Yu, W- X Yue, J Wang, F- X Zhu, G- Y Li, J- H Lu
{"title":"Retraction Note: Cadherin 6 is activated by Epstein-Barr virus LMP1 to mediate EMT and metastasis as an interplay node of multiple pathways in nasopharyngeal carcinoma.","authors":"L-L Zuo,&nbsp;J Zhang,&nbsp;L- Z Liu,&nbsp;Q Zhou,&nbsp;S- J Du,&nbsp;S- Y Xin,&nbsp;Z- P Ning,&nbsp;J Yang,&nbsp;H- B Yu,&nbsp;W- X Yue,&nbsp;J Wang,&nbsp;F- X Zhu,&nbsp;G- Y Li,&nbsp;J- H Lu","doi":"10.1038/s41389-022-00439-x","DOIUrl":"https://doi.org/10.1038/s41389-022-00439-x","url":null,"abstract":"","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2022-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9576719/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40395851","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: 2
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