Molecular Cancer Research最新文献

筛选
英文 中文
Editor's Note: Targeting Activating Transcription Factor 3 by Galectin-9 Induces Apoptosis and Overcomes Various Types of Treatment Resistance in Chronic Myelogenous Leukemia. 编者按:Galectin-9靶向激活转录因子3可诱导凋亡并克服慢性骨髓性白血病的各种治疗耐药性。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-08-02 DOI: 10.1158/1541-7786.MCR-24-0507
Junya Kuroda, Mio Yamamoto, Hisao Nagoshi, Tsutomu Kobayashi, Nana Sasaki, Yuji Shimura, Shigeo Horiike, Shinya Kimura, Akira Yamauchi, Mitsuomi Hirashima, Masafumi Taniwaki
{"title":"Editor's Note: Targeting Activating Transcription Factor 3 by Galectin-9 Induces Apoptosis and Overcomes Various Types of Treatment Resistance in Chronic Myelogenous Leukemia.","authors":"Junya Kuroda, Mio Yamamoto, Hisao Nagoshi, Tsutomu Kobayashi, Nana Sasaki, Yuji Shimura, Shigeo Horiike, Shinya Kimura, Akira Yamauchi, Mitsuomi Hirashima, Masafumi Taniwaki","doi":"10.1158/1541-7786.MCR-24-0507","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-24-0507","url":null,"abstract":"","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141875453","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
MYC Family Amplification Dictates Sensitivity to BET Bromodomain Protein Inhibitor Mivebresib (ABBV075) in Small-Cell Lung Cancer. MYC家族扩增决定了小细胞肺癌患者对BET溴域蛋白抑制剂Mivebresib(ABBV-075)的敏感性。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-08-02 DOI: 10.1158/1541-7786.MCR-23-0599
Joshua P Plotnik, Zheng Zha, Weiguo Feng, Irene Lee, Jacob Riehm, Ryan A McClure, Stephanie Sandoval, Tamar Uziel, Erin Murphy, Xin Lu, Lloyd T Lam
{"title":"MYC Family Amplification Dictates Sensitivity to BET Bromodomain Protein Inhibitor Mivebresib (ABBV075) in Small-Cell Lung Cancer.","authors":"Joshua P Plotnik, Zheng Zha, Weiguo Feng, Irene Lee, Jacob Riehm, Ryan A McClure, Stephanie Sandoval, Tamar Uziel, Erin Murphy, Xin Lu, Lloyd T Lam","doi":"10.1158/1541-7786.MCR-23-0599","DOIUrl":"10.1158/1541-7786.MCR-23-0599","url":null,"abstract":"<p><p>Small-cell lung cancer (SCLC) accounts for nearly 15% of all lung cancers. Although patients respond to first-line therapy readily, rapid relapse is inevitable, with few treatment options in the second-line setting. Here, we describe SCLC cell lines harboring amplification of MYC and MYCN but not MYCL1 or non-amplified MYC cell lines exhibit superior sensitivity to treatment with the pan-BET bromodomain protein inhibitor mivebresib (ABBV075). Silencing MYC and MYCN partially rescued SCLC cell lines harboring these respective amplifications from the antiproliferative effects of mivebresib. Further characterization of genome-wide binding of MYC, MYCN, and MYCL1 uncovered unique enhancer and epigenetic preferences. Implications: Our study suggests that chromatin landscapes can establish cell states with unique gene expression programs, conveying sensitivity to epigenetic inhibitors such as mivebresib.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294817/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140921218","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
Neem Leaf Glycoprotein Disrupts Exhausted CD8+ T-Cell-Mediated Cancer Stem Cell Aggression. 印楝叶糖蛋白能破坏 CD8+ T 细胞介导的癌症干细胞侵袭。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-08-02 DOI: 10.1158/1541-7786.MCR-23-0993
Mohona Chakravarti, Saurav Bera, Sukanya Dhar, Anirban Sarkar, Pritha Roy Choudhury, Nilanjan Ganguly, Juhina Das, Jasmine Sultana, Aishwarya Guha, Souradeep Biswas, Tapasi Das, Subhadip Hajra, Saptak Banerjee, Rathindranath Baral, Anamika Bose
{"title":"Neem Leaf Glycoprotein Disrupts Exhausted CD8+ T-Cell-Mediated Cancer Stem Cell Aggression.","authors":"Mohona Chakravarti, Saurav Bera, Sukanya Dhar, Anirban Sarkar, Pritha Roy Choudhury, Nilanjan Ganguly, Juhina Das, Jasmine Sultana, Aishwarya Guha, Souradeep Biswas, Tapasi Das, Subhadip Hajra, Saptak Banerjee, Rathindranath Baral, Anamika Bose","doi":"10.1158/1541-7786.MCR-23-0993","DOIUrl":"10.1158/1541-7786.MCR-23-0993","url":null,"abstract":"<p><p>Targeting exhausted CD8+ T-cell (TEX)-induced aggravated cancer stem cells (CSC) holds immense therapeutic potential. In this regard, immunomodulation via Neem Leaf Glycoprotein (NLGP), a plant-derived glycoprotein immunomodulator is explored. Since former reports have proven immune dependent-tumor restriction of NLGP across multiple tumor models, we hypothesized that NLGP might reprogram and rectify TEX to target CSCs successfully. In this study, we report that NLGP's therapeutic administration significantly reduced TEX-associated CSC virulence in in vivo B16-F10 melanoma tumor model. A similar trend was observed in in vitro generated TEX and B16-F10/MCF7 coculture setups. NLGP rewired CSCs by downregulating clonogenicity, multidrug resistance phenotypes and PDL1, OCT4, and SOX2 expression. Cell cycle analysis revealed that NLGP educated-TEX efficiently pushed CSCs out of quiescent phase (G0G1) into synthesis phase (S), supported by hyper-phosphorylation of G0G1-S transitory cyclins and Rb proteins. This rendered quiescent CSCs susceptible to S-phase-targeting chemotherapeutic drugs like 5-fluorouracil (5FU). Consequently, combinatorial treatment of NLGP and 5FU brought optimal CSC-targeting efficiency with an increase in apoptotic bodies and proapoptotic BID expression. Notably a strong nephron-protective effect of NLGP was also observed, which prevented 5FU-associated toxicity. Furthermore, Dectin-1-mediated NLGP uptake and subsequent alteration of Notch1 and mTOR axis were deciphered as the involved signaling network. This observation unveiled Dectin-1 as a potent immunotherapeutic drug target to counter T-cell exhaustion. Cumulatively, NLGP immunotherapy alleviated exhausted CD8+ T-cell-induced CSC aggravation. Implications: Our study recommends that NLGP immunotherapy can be utilized to counter ramifications of T-cell exhaustion and to target therapy elusive aggressive CSCs without evoking toxicity.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140921288","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
Long Noncoding RNA MSL3P1 Regulates CUL3 mRNA Cytoplasmic Transport and Stability and Promotes Lung Adenocarcinoma Metastasis. 长非编码RNA MSL3P1调控CUL3 mRNA的胞质转运和稳定性并促进肺腺癌转移。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-08-02 DOI: 10.1158/1541-7786.MCR-23-0977
Ming-Ming Shao, Xin Li, Rui-Qi Wei, Qing-Yu Chen, Xin Zhang, Xin Qiao, Hui Li
{"title":"Long Noncoding RNA MSL3P1 Regulates CUL3 mRNA Cytoplasmic Transport and Stability and Promotes Lung Adenocarcinoma Metastasis.","authors":"Ming-Ming Shao, Xin Li, Rui-Qi Wei, Qing-Yu Chen, Xin Zhang, Xin Qiao, Hui Li","doi":"10.1158/1541-7786.MCR-23-0977","DOIUrl":"10.1158/1541-7786.MCR-23-0977","url":null,"abstract":"<p><p>Lung adenocarcinoma (LUAD) is the most prevalent histological type of lung cancer. Previous studies have reported that specific long noncoding RNAs (lncRNA) are involved in cancer development and progression. The phenotype and mechanism of ENST00000440028, named MSL3P1, an lncRNA referred to as a cancer-testis gene with potential roles in tumorigenesis and progression, have not been reported. MSL3P1 is overexpressed in LUAD tumor tissues, which is significantly associated with clinical characteristics, metastasis, and poor clinical prognosis. MSL3P1 promotes the metastasis of LUAD in vitro and in vivo. The enhancer reprogramming in LUAD tumor tissue is the major driver of the aberrant expression of MSL3P1. Mechanistically, owing to the competitive binding to CUL3 mRNA with ZFC3H1 protein (a protein involved in targeting polyadenylated RNA to exosomes and promoting the degradation of target mRNA), MSL3P1 can prevent the ZFC3H1-mediated RNA degradation of CUL3 mRNA and transport it to the cytoplasm. This activates the downstream epithelial-to-mesenchymal transition signaling pathway and promotes tumor invasion and metastasis. Implications: This study indicates that lncRNA MSL3P1 regulates CUL3 mRNA stability and promotes metastasis and holds potential as a prognostic biomarker and therapeutic target in LUAD.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140892229","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
Ruthenium drug BOLD-100 regulates BRAFMT colorectal cancer cell apoptosis through AhR/ROS/ATR signaling axis modulation. 钌药物 BOLD-100 通过 AhR/ROS/ATR 信号轴调节 BRAFMT 大肠癌细胞凋亡。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-07-31 DOI: 10.1158/1541-7786.MCR-24-0151
Daryl Griffin, Robbie Carson, Debbie Moss, Tamas Sessler, Deborah Lavin, Vijay K Tiwari, Shivaali Karelia, Richard Kennedy, Kienan I Savage, Simon McDade, Adam Carie, Jim Pankovich, Mark Bazett, Sandra Van Schaeybroeck
{"title":"Ruthenium drug BOLD-100 regulates BRAFMT colorectal cancer cell apoptosis through AhR/ROS/ATR signaling axis modulation.","authors":"Daryl Griffin, Robbie Carson, Debbie Moss, Tamas Sessler, Deborah Lavin, Vijay K Tiwari, Shivaali Karelia, Richard Kennedy, Kienan I Savage, Simon McDade, Adam Carie, Jim Pankovich, Mark Bazett, Sandra Van Schaeybroeck","doi":"10.1158/1541-7786.MCR-24-0151","DOIUrl":"10.1158/1541-7786.MCR-24-0151","url":null,"abstract":"<p><p>Patients with class I V600EBRAF-mutant (MT) colorectal cancer (CRC) have a poor prognosis and their response to combined anti-BRAF/EGFR inhibition remains limited. There is clearly an unmet need in further understanding the biology of V600EBRAFMT CRC. We have used differential gene expression of BRAFWT and MT CRC cells to identify pathways underpinning BRAFMT CRC. We tested a panel of molecularly/genetically subtyped CRC cells for their sensitivity to the Unfolded Protein Response (UPR) activator BOLD-100. To identify novel combination strategies for BOLD-100, we performed RNA sequencing and high-throughput drug screening. Pathway enrichment analysis identified that the UPR and DNA repair pathways were significantly enriched in BRAFMT CRC. We found that oncogenic BRAF plays a crucial role in mediating response to BOLD-100. Using a systems biology approach, we identified V600EBRAFMT-dependent activation of the replication stress response kinase ATR as a key mediator of resistance to BOLD-100. Further analysis identified acute increases in BRAFMT-dependent-reactive oxygen species (ROS) levels following treatment with BOLD-100 that was demonstrated to promote ATR/CHK1 activation and apoptosis. Furthermore, activation of ROS/ATR/CHK1 following BOLD-100 was found to be mediated through the AHR transcription factor and CYP1A1. Importantly, pharmacological blockade of this resistance pathway with ATR inhibitors synergistically increased BOLD-100-induced apoptosis and growth inhibition in BRAFMT models. These results unveil possible novel therapeutic opportunity for BRAFMT CRC. Implications: BOLD-100 induces BRAFMT-dependent replication stress, and targeted strategies against replication stress (eg. by using ATR inhibitors) in combination with BOLD-100 may serve as a potential novel therapeutic strategy for clinically aggressive BRAFMT CRC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7616621/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141856110","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
GRAIL1 stabilizes misfolded mutant p53 through a ubiquitin ligase-independent, chaperone regulatory function. GRAIL1 通过独立于泛素连接酶的伴侣调节功能稳定折叠错误的突变 p53。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-07-17 DOI: 10.1158/1541-7786.MCR-24-0361
Paramita Ray, Sangeeta Jaiswal, Daysha Ferrer-Torres, Zhuwen Wang, Derek Nancarrow, Meghan Curtin, May San Martinho, Shannon M Lacy, Srimathi Kasturirangan, Dafydd Thomas, Jason R Spence, Matthias C Truttmann, Kiran H Lagisetty, Theodore S Lawrence, Thomas D Wang, David G Beer, Dipankar Ray
{"title":"GRAIL1 stabilizes misfolded mutant p53 through a ubiquitin ligase-independent, chaperone regulatory function.","authors":"Paramita Ray, Sangeeta Jaiswal, Daysha Ferrer-Torres, Zhuwen Wang, Derek Nancarrow, Meghan Curtin, May San Martinho, Shannon M Lacy, Srimathi Kasturirangan, Dafydd Thomas, Jason R Spence, Matthias C Truttmann, Kiran H Lagisetty, Theodore S Lawrence, Thomas D Wang, David G Beer, Dipankar Ray","doi":"10.1158/1541-7786.MCR-24-0361","DOIUrl":"10.1158/1541-7786.MCR-24-0361","url":null,"abstract":"<p><p>Frequent (>70%) TP53 mutations often promote its protein stabilization, driving esophageal adenocarcinoma (EAC) development linked to poor survival and therapy resistance. We previously reported that during Barrett's (BE) progression to EAC, an isoform switch occurs in the E3 ubiquitin ligase RNF128 (aka GRAIL - gene related to anergy in lymphocytes), enriching isoform 1 (hereby GRAIL1) and, stabilizing the mutant p53 protein. Consequently, GRAIL1 knockdown degrades mutant p53. But how GRAIL1 stabilizes the mutant p53 protein remains unclear. In search for a mechanism, here we performed biochemical and cell biology studies to identify that GRAIL has a binding domain (315-PMCKCDILKA-325) for Hsp40/DNAJ. This interaction can influence DNAJ chaperone activity to modulate misfolded mutant p53 stability. As predicted, either the overexpression of a GRAIL fragment (Frag-J) encompassing the DNAJ binding domain, or a cell permeable peptide (Pep-J) encoding the above 10 amino acids, can bind and inhibit DNAJ-Hsp70 co-chaperone activity thus degrading misfolded mutant p53. Consequently, either Frag-J or Pep-J can reduce the survival of mutant p53 containing dysplastic BE and EAC cells and inhibit growth of patient-derived dysplastic BE organoids (PDOs) in 3D cultures. The misfolded mutant p53 targeting and growth inhibitory effects of Pep-J is comparable to simvastatin, a cholesterol lowering drug, that can degrade misfolded mutant p53 also via inhibiting DNAJA1, although by a distinct mechanism. Implications: We identified a novel ubiquitin ligase independent, chaperone regulating domain in GRAIL and further synthesized a first-in-class novel misfolded mutant p53 degrading peptide having future translational potential.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141634059","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
HnRNPAB promotes pancreatic ductal adenocarcinoma extravasation and liver metastasis by stablizing MYC mRNA. HnRNPAB 通过稳定 MYC mRNA 促进胰腺导管腺癌的外渗和肝转移。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-07-05 DOI: 10.1158/1541-7786.MCR-24-0110
Ke Lei, Mingyue Sun, Xianghan Chen, Jia Wang, Xiaolan Liu, Ying Ning, Shuai Ping, Ruining Gong, Yu Zhang, Gong Qing, Chenyang Zhao, He Ren
{"title":"HnRNPAB promotes pancreatic ductal adenocarcinoma extravasation and liver metastasis by stablizing MYC mRNA.","authors":"Ke Lei, Mingyue Sun, Xianghan Chen, Jia Wang, Xiaolan Liu, Ying Ning, Shuai Ping, Ruining Gong, Yu Zhang, Gong Qing, Chenyang Zhao, He Ren","doi":"10.1158/1541-7786.MCR-24-0110","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-24-0110","url":null,"abstract":"<p><p>Heterogeneous nuclear ribonucleoprotein AB (hnRNPAB) is considered a cancer-promoting heterogeneous nuclear ribonucleoprotein in many cancers, but its function in pancreatic ductal adenocarcinoma (PDAC) is poorly understood. HnRNPAB was highly expressed in PDAC tissues compared to normal pancreatic tissues, and high expression of hnRNPAB was associated with poor overall survival and recurrence-free survival in PDAC patients. HnRNPAB promotes migration and invasion of PDAC cells in vitro. In xenograft tumor mouse models, hnRNPAB deprivation significantly attenuated liver metastasis. HnRNPAB mRNA and protein levels are positively associated with MYC in PDAC cells. Mechanistically, hnRNPAB bound to MYC mRNA and prolonged its half-life of MYC mRNA. HnRNPAB induced PDAC cells to secret CXCL8 via MYC, which promoted neutrophils recruitment and facilitated tumor cells entrancing into the hepatic parenchyma. These findings point to a novel regulatory mechanism via which hnRNPAB promotes PDAC metastasis. Implications: Hnrnpab participates in the post-transcriptional regulation of the oncogene MYC by binding and stabilizing MYC mRNA, thereby promoting liver metastasis in PDAC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141534886","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
Distinct Regulation of ASCL1 by the Cell Cycle and Chemotherapy in Small Cell Lung Cancer. 细胞周期和化疗对小细胞肺癌中 ASCL1 的不同调控。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-07-02 DOI: 10.1158/1541-7786.MCR-23-0405
Yuning Liu, Qingzhe Wu, Bin Jiang, Tingting Hou, Chuanqiang Wu, Ming Wu, Hai Song
{"title":"Distinct Regulation of ASCL1 by the Cell Cycle and Chemotherapy in Small Cell Lung Cancer.","authors":"Yuning Liu, Qingzhe Wu, Bin Jiang, Tingting Hou, Chuanqiang Wu, Ming Wu, Hai Song","doi":"10.1158/1541-7786.MCR-23-0405","DOIUrl":"10.1158/1541-7786.MCR-23-0405","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) is an aggressive and lethal malignancy. Achaete-scute homolog 1 (ASCL1) is essential for the initiation of SCLC in mice and the development of pulmonary neuroendocrine cells (PNEC), which are the major cells of origin for SCLC. However, the regulatory mechanism of ASCL1 in SCLC remains elusive. Here, we found that ASCL1 expression gradually increases as the tumors grow in a mouse SCLC model, and is regulated by the cell cycle. Mechanistically, CDK2-CyclinA2 complex phosphorylates ASCL1, which results in increased proteasome-mediated ASCL1 protein degradation by E3 ubiquitin ligase HUWE1 during mitosis. TCF3 promotes the multisite phosphorylation of ASCL1 through the CDK2-CyclinA2 complex and the interaction between ASCL1 and TCF3 protects ASCL1 from degradation. The dissociation of TCF3 from ASCL1 during mitosis accelerates the degradation of ASCL1. In addition, chemotherapy drugs greatly reduce the transcription of ASCL1 in SCLC cells. Depletion of ASCL1 sensitizes SCLC cells to chemotherapy drugs. Together, our study demonstrates that ASCL1 is a cell-cycle-regulated protein and provides a theoretical basis for applying cell-cycle-related antitumor drugs in SCLC treatment. Implications:Our study revealed a novel regulatory mechanism of ASCL1 by cell cycle and chemotherapy drugs in SCLC. Treating patients with SCLC with a combination of ASCL1-targeting therapy and chemotherapy drugs could potentially be beneficial.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11217739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140175770","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
Low- and High-Grade Glioma-Associated Vascular Cells Differentially Regulate Tumor Growth. 低级别和高级别胶质瘤相关血管细胞对肿瘤生长有不同的调节作用。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-07-02 DOI: 10.1158/1541-7786.MCR-23-1069
Sree Deepthi Muthukrishnan, Haocheng Qi, David Wang, Lubayna Elahi, Amy Pham, Alvaro G Alvarado, Tie Li, Fuying Gao, Riki Kawaguchi, Albert Lai, Harley I Kornblum
{"title":"Low- and High-Grade Glioma-Associated Vascular Cells Differentially Regulate Tumor Growth.","authors":"Sree Deepthi Muthukrishnan, Haocheng Qi, David Wang, Lubayna Elahi, Amy Pham, Alvaro G Alvarado, Tie Li, Fuying Gao, Riki Kawaguchi, Albert Lai, Harley I Kornblum","doi":"10.1158/1541-7786.MCR-23-1069","DOIUrl":"10.1158/1541-7786.MCR-23-1069","url":null,"abstract":"<p><p>A key feature distinguishing high-grade glioma (HG) from low-grade glioma (LG) is the extensive neovascularization and endothelial hyperproliferation. Prior work has shown that tumor-associated vasculature from HG is molecularly and functionally distinct from normal brain vasculature and expresses higher levels of protumorigenic factors that promote glioma growth and progression. However, it remains unclear whether vessels from LG also express protumorigenic factors, and to what extent they functionally contribute to glioma growth. Here, we profile the transcriptomes of glioma-associated vascular cells (GVC) from IDH-mutant (mIDH) LG and IDH-wild-type (wIDH) HG and show that they exhibit significant molecular and functional differences. LG-GVC show enrichment of extracellular matrix-related gene sets and sensitivity to antiangiogenic drugs, whereas HG-GVC display an increase in immune response-related gene sets and antiangiogenic resistance. Strikingly, conditioned media from LG-GVC inhibits the growth of wIDH glioblastoma cells, whereas HG-GVC promotes growth. In vivo cotransplantation of LG-GVC with tumor cells reduces growth, whereas HG-GVC enhances tumor growth in orthotopic xenografts. We identify ASPORIN (ASPN), a small leucine-rich repeat proteoglycan, highly enriched in LG-GVC as a growth suppressor of wIDH glioblastoma cells in vitro and in vivo. Together, these findings indicate that GVC from LG and HG are molecularly and functionally distinct and differentially regulate tumor growth. Implications: This study demonstrated that vascular cells from IDH-mutant LG and IDH-wild-type HG exhibit distinct molecular signatures and have differential effects on tumor growth via regulation of ASPN-TGFβ1-GPM6A signaling.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11217726/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140028464","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
Hepatitis B Virus-Mediated m6A Demethylation Increases Hepatocellular Carcinoma Stemness and Immune Escape. 乙型肝炎病毒介导的 m6A 去甲基化增加了肝细胞癌的干性和免疫逃逸。
IF 4.1 2区 医学
Molecular Cancer Research Pub Date : 2024-07-02 DOI: 10.1158/1541-7786.MCR-23-0720
Yuting Meng, Zheyue Shu, Xueyao Wang, Liang Hong, Baohua Wang, Jingjing Jiang, Kangxin He, Qingyi Cao, Fan Shi, Hai Wang, Lan Gong, Hongyan Diao
{"title":"Hepatitis B Virus-Mediated m6A Demethylation Increases Hepatocellular Carcinoma Stemness and Immune Escape.","authors":"Yuting Meng, Zheyue Shu, Xueyao Wang, Liang Hong, Baohua Wang, Jingjing Jiang, Kangxin He, Qingyi Cao, Fan Shi, Hai Wang, Lan Gong, Hongyan Diao","doi":"10.1158/1541-7786.MCR-23-0720","DOIUrl":"10.1158/1541-7786.MCR-23-0720","url":null,"abstract":"<p><p>Hepatitis B viral (HBV) persistent infection plays a significant role in hepatocellular carcinoma (HCC) tumorigenesis. Many studies have revealed the pivotal roles of N6-methyladenosine (m6A) in multiple cancers, while the regulatory mechanism in stemness maintenance of HBV persistent infection-related HCC remains elusive. Here, we demonstrated that the level of m6A modification was downregulated by HBV in HBV-positive HCC, through enhanced stability of ALKBH5 mRNA. More specifically, we also identified that ALKBH5 mRNA was functionally required for the stemness maintenance and self-renewal in the HBV-positive HCC, but dispensable in HBV-negative HCC. Mechanistically, ALKBH5 demethylated the m6A modification in the 3' untranslated region of the oncogenic gene SNAI2 to prevent the recognition of YTHDF2 therewith stabilize SNAI2 transcripts, contributing to cancer stem cell traits in HBV-positive HCC. Moreover, the expression of SNAI2 reversed the suppression of stemness properties by knocking down ALKBH5. In addition, ALKBH5/SNAI2 axis accelerates tumor immune evasion through activated ligand of immune checkpoint CD155. Our study unveiled that the ALKBH5 induces m6A demethylation of the SNAI2 as a key regulator in HBV-related HCC, and identifies the function of ALKBH5/SNAI2/YTHDF2 axis in promoting the stem-like cells phenotype and immune escape during HBV infection.</p><p><strong>Implications: </strong>HBV promotes HCC stemness maintenance through elevate m6A modification of SNAI2 in an ALKBH5-YTHDF2-dependent manner and increases the expression of the ligand of immune checkpoint CD155.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11217737/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140306245","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信