首页 > 最新文献

bioRxiv - Cancer Biology最新文献

英文 中文
p53 Regulates Nuclear Architecture to Reduce Carcinogen Sensitivity and Mutagenic Potential p53 调节核结构以降低对致癌物质的敏感性和诱变潜力
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.14.613067
Devin A King, Dakota E McCoy, Adrian Perdyan, Jakub Mieczkowski, Thierry Douki, Jennifer A Dionne, Rafael E Herrera, Ashby J Morrison
The p53 tumor suppressor is an indispensable regulator of DNA damage responses that accelerates carcinogenesis when mutated. In this report, we uncover a new mechanism by which p53 maintains genomic integrity in the absence of canonical DNA damage response activation. Specifically, loss of p53 dramatically alters chromatin structure at the nuclear periphery, allowing increased transmission of an environmental carcinogen, ultraviolet (UV) radiation, into the nucleus. Genome-wide mapping of UV-induced DNA lesions in p53-deficient primary cells reveals increased lesion abundance corresponding to regions of high mutation burden in malignant melanomas. These findings uncover a novel role of p53 in the suppression of mutations that contribute to cancer and highlight the critical influence of nuclear architecture in regulating sensitivity to carcinogens.
p53 肿瘤抑制因子是 DNA 损伤反应不可或缺的调节因子,一旦发生突变会加速癌变。在本报告中,我们发现了一种新的机制,通过这种机制,p53 可以在没有典型 DNA 损伤反应激活的情况下维持基因组的完整性。具体来说,p53 的缺失会显著改变核外围的染色质结构,从而增加环境致癌物质紫外线(UV)辐射向细胞核的传递。p53缺陷原代细胞中紫外线诱导的DNA病变的全基因组图谱显示,病变丰度增加与恶性黑色素瘤的高突变负荷区域相对应。这些发现揭示了 p53 在抑制导致癌症的突变方面的新作用,并强调了核结构在调节对致癌物质的敏感性方面的关键影响。
{"title":"p53 Regulates Nuclear Architecture to Reduce Carcinogen Sensitivity and Mutagenic Potential","authors":"Devin A King, Dakota E McCoy, Adrian Perdyan, Jakub Mieczkowski, Thierry Douki, Jennifer A Dionne, Rafael E Herrera, Ashby J Morrison","doi":"10.1101/2024.09.14.613067","DOIUrl":"https://doi.org/10.1101/2024.09.14.613067","url":null,"abstract":"The p53 tumor suppressor is an indispensable regulator of DNA damage responses that accelerates carcinogenesis when mutated. In this report, we uncover a new mechanism by which p53 maintains genomic integrity in the absence of canonical DNA damage response activation. Specifically, loss of p53 dramatically alters chromatin structure at the nuclear periphery, allowing increased transmission of an environmental carcinogen, ultraviolet (UV) radiation, into the nucleus. Genome-wide mapping of UV-induced DNA lesions in p53-deficient primary cells reveals increased lesion abundance corresponding to regions of high mutation burden in malignant melanomas. These findings uncover a novel role of p53 in the suppression of mutations that contribute to cancer and highlight the critical influence of nuclear architecture in regulating sensitivity to carcinogens.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"53 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249757","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting cancer-associated glycosylation for adoptive T cell therapy of gastro-intestinal and gynecological cancers 以癌症相关糖基化为靶点,采用 T 细胞治疗胃肠道和妇科癌症
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.12.612750
Andreas Zingg, Reto Ritschard, Helen Thut, Melanie Buchi, Andreas Holbro, Anton Oseledchyk, Viola Heinzelmann, Andreas Buser, Mascha Binder, Alfred Zippelius, Natalia Rodrigues Mantuano, Heinz Laubli
CAR T cell therapy has provided a significant improvement for patients with chemotherapy-resistant B cell malignancies. However, CAR T cell treatment of patients with solid cancers has been more difficult, in part because of the heterogeneous expression of tumor-specific cell surface antigens. Here, we describe the generation of a fully human CAR targeting altered glycosylation in secretory epithelial cancers. The expression of the target antigen, the truncated, sialylated O-glycan sialyl-Thomsen-Nouveau antigen (STn), was studied with a highly STn-specific antibody across various different tumor tissues. Strong expression was found in a high proportion of gastro-intestinal cancers including pancreatic cancers and in gynecological cancers, in particular ovarian and endometrial tumors. T cells expressing anti-STn CAR were tested in vitro and in vivo. Anti-STn CAR T cells showed activity in mouse models as well as in assays with primary ovarian cancer samples. No significant toxicity was observed in mouse models, although some intraluminal expression of STn was found in gastro-intestinal mouse tissue. Taken together, this fully human anti-STn CAR construct shows promising activity in preclinical tumor models supporting its further evaluation in early clinical trials.
CAR T 细胞疗法大大改善了化疗耐药 B 细胞恶性肿瘤患者的病情。然而,CAR T 细胞治疗实体瘤患者却比较困难,部分原因是肿瘤特异性细胞表面抗原的表达不一致。在这里,我们描述了针对分泌性上皮癌糖基化改变的全人源 CAR 的产生过程。我们用一种高度 STn 特异性的抗体研究了各种不同肿瘤组织中的靶抗原--截短的、糖基化的 O-聚糖 sialyl-Thomsen-Nouveau 抗原(STn)的表达情况。在包括胰腺癌在内的高比例胃肠道癌症和妇科癌症(尤其是卵巢癌和子宫内膜癌)中发现了强表达。对表达抗 STn CAR 的 T 细胞进行了体外和体内测试。抗 STn CAR T 细胞在小鼠模型以及原发性卵巢癌样本的试验中显示出活性。虽然在小鼠胃肠道组织中发现了一些 STn 腔内表达,但在小鼠模型中没有观察到明显的毒性。综上所述,这种全人源抗 STn CAR 构建物在临床前肿瘤模型中显示出良好的活性,支持在早期临床试验中对其进行进一步评估。
{"title":"Targeting cancer-associated glycosylation for adoptive T cell therapy of gastro-intestinal and gynecological cancers","authors":"Andreas Zingg, Reto Ritschard, Helen Thut, Melanie Buchi, Andreas Holbro, Anton Oseledchyk, Viola Heinzelmann, Andreas Buser, Mascha Binder, Alfred Zippelius, Natalia Rodrigues Mantuano, Heinz Laubli","doi":"10.1101/2024.09.12.612750","DOIUrl":"https://doi.org/10.1101/2024.09.12.612750","url":null,"abstract":"CAR T cell therapy has provided a significant improvement for patients with chemotherapy-resistant B cell malignancies. However, CAR T cell treatment of patients with solid cancers has been more difficult, in part because of the heterogeneous expression of tumor-specific cell surface antigens. Here, we describe the generation of a fully human CAR targeting altered glycosylation in secretory epithelial cancers. The expression of the target antigen, the truncated, sialylated O-glycan sialyl-Thomsen-Nouveau antigen (STn), was studied with a highly STn-specific antibody across various different tumor tissues. Strong expression was found in a high proportion of gastro-intestinal cancers including pancreatic cancers and in gynecological cancers, in particular ovarian and endometrial tumors. T cells expressing anti-STn CAR were tested in vitro and in vivo. Anti-STn CAR T cells showed activity in mouse models as well as in assays with primary ovarian cancer samples. No significant toxicity was observed in mouse models, although some intraluminal expression of STn was found in gastro-intestinal mouse tissue. Taken together, this fully human anti-STn CAR construct shows promising activity in preclinical tumor models supporting its further evaluation in early clinical trials.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"53 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Proteogenomic Characterization of Primary Oral Squamous Cell Carcinomas Unveils the Extracellular Matrix Remodeling and Immunosuppressive Microenvironment Linked with Lymph Node Metastasis 原发性口腔鳞状细胞癌的蛋白质基因组特征揭示了与淋巴结转移相关的细胞外基质重塑和免疫抑制微环境
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.12.612653
Yu Liu, Zhenyu Yang, Jingya Jane Pu, Jie Zhong, Ui Soon Khoo, Yuxiong Richard Su, Gao Zhang
Oral squamous cell carcinoma (OSCC) is an increasingly prevalent malignancy worldwide. This study aims to understand molecular alterations associated with lymph node metastasis of OSCC in order to improve treatment strategies. We analyzed a cohort of 46 patients with primary OSCC, including 10 with lymph node metastasis and 36 without. Using a comprehensive multi-omics approach, encompassing genomic, transcriptomic, proteomic, epigenetic, single-cell, and spatial analyses; we integrated data to delineate the molecular landscape of OSCC in the context of lymph node metastasis. Our genomic analysis identified significant mutations in key genes within the MAPK, TGF-b, and WNT signaling pathways, which are essential for tumor development. The proteogenomic analysis highlighted pathways critical for lymph node dissemination and factors contributing to an immunosuppressive tumor microenvironment. Elevated levels of POSTN were found to reorganize the extracellular matrix (ECM), interact with TGF-b disrupt cell cycle regulation, and suppress the immune response by reducing VCAM1 activity. Integrated analyses of single-cell and spatial transcriptome data revealed that cancer-associated fibroblasts (CAFs) secrete TGF-b1/2 promoting cancer cell metastasis through epithelial-mesenchymal transition (EMT). Our integrated multi-omics analysis provides a detailed understanding of molecular mechanisms driving lymph node metastasis of OSCC. These insights could lead to more precise diagnostics and targeted treatments.
口腔鳞状细胞癌(OSCC)是一种在全球日益流行的恶性肿瘤。本研究旨在了解与OSCC淋巴结转移相关的分子改变,从而改进治疗策略。我们分析了46例原发性OSCC患者,其中10例有淋巴结转移,36例无淋巴结转移。我们采用了一种全面的多组学方法,包括基因组、转录组、蛋白质组、表观遗传学、单细胞和空间分析;我们整合了数据,在淋巴结转移的背景下描绘了OSCC的分子图谱。我们的基因组分析发现了MAPK、TGF-b和WNT信号通路中关键基因的重大突变,这些基因对肿瘤的发展至关重要。蛋白质基因组分析突出了对淋巴结扩散至关重要的通路以及导致免疫抑制性肿瘤微环境的因素。研究发现,POSTN水平的升高会重组细胞外基质(ECM),与TGF-b相互作用破坏细胞周期调控,并通过降低VCAM1的活性抑制免疫反应。对单细胞和空间转录组数据的综合分析表明,癌症相关成纤维细胞(CAFs)会分泌TGF-b1/2,通过上皮-间质转化(EMT)促进癌细胞转移。我们的综合多组学分析提供了对驱动OSCC淋巴结转移的分子机制的详细了解。这些见解可能会带来更精确的诊断和靶向治疗。
{"title":"Proteogenomic Characterization of Primary Oral Squamous Cell Carcinomas Unveils the Extracellular Matrix Remodeling and Immunosuppressive Microenvironment Linked with Lymph Node Metastasis","authors":"Yu Liu, Zhenyu Yang, Jingya Jane Pu, Jie Zhong, Ui Soon Khoo, Yuxiong Richard Su, Gao Zhang","doi":"10.1101/2024.09.12.612653","DOIUrl":"https://doi.org/10.1101/2024.09.12.612653","url":null,"abstract":"Oral squamous cell carcinoma (OSCC) is an increasingly prevalent malignancy worldwide. This study aims to understand molecular alterations associated with lymph node metastasis of OSCC in order to improve treatment strategies. We analyzed a cohort of 46 patients with primary OSCC, including 10 with lymph node metastasis and 36 without. Using a comprehensive multi-omics approach, encompassing genomic, transcriptomic, proteomic, epigenetic, single-cell, and spatial analyses; we integrated data to delineate the molecular landscape of OSCC in the context of lymph node metastasis. Our genomic analysis identified significant mutations in key genes within the MAPK, TGF-b, and WNT signaling pathways, which are essential for tumor development. The proteogenomic analysis highlighted pathways critical for lymph node dissemination and factors contributing to an immunosuppressive tumor microenvironment. Elevated levels of POSTN were found to reorganize the extracellular matrix (ECM), interact with TGF-b disrupt cell cycle regulation, and suppress the immune response by reducing VCAM1 activity. Integrated analyses of single-cell and spatial transcriptome data revealed that cancer-associated fibroblasts (CAFs) secrete TGF-b1/2 promoting cancer cell metastasis through epithelial-mesenchymal transition (EMT). Our integrated multi-omics analysis provides a detailed understanding of molecular mechanisms driving lymph node metastasis of OSCC. These insights could lead to more precise diagnostics and targeted treatments.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"199 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249761","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhibition of glutaminase elicits senolysis in therapy-induced senescent melanoma cells. 抑制谷氨酰胺酶可诱导治疗诱导的衰老黑色素瘤细胞发生衰老。
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.12.612728
Justin Kim, Bryce Brunetti, Ayanesh Kumar, Ankit Mangla, Kord Honda, Akihiro Yoshida
The cyclin D1-Cyclin-Dependent Kinases 4 and 6 (CDK4/6) complex is crucial for the development of melanoma. We previously demonstrated that targeting CDK4/6 using small molecule inhibitors (CDK4/6i) suppresses BrafV600E melanoma growth in vitro and in vivo through induction of cellular senescence. Clinical trials investigating CDK4/6i in melanoma have not yielded successful outcomes, underscoring the necessity to enhance the therapeutic efficacy of CDK4/6i. Accumulated research has shown that while senescence initially suppresses cell proliferation, a prolonged state of senescence eventually leads to tumor relapse by altering the tumor microenvironment, suggesting that removal of those senescent cells (in a process referred to as senolysis) is of clinical necessity to facilitate clinical response. We demonstrate that glutaminase 1 (GLS1) expression is specifically upregulated in CDK4/6i-induced senescent BrafV600E melanoma cells. Upregulated GLS1 expression renders BrafV600E melanoma senescent cells vulnerable to GLS1 inhibitor (GLS1i). Furthermore, we demonstrate that this senolytic approach targeting upregulated GLS1 expression is applicable even though those cells developed resistance to the BrafV600E inhibitor vemurafenib, a frequently encountered substantial clinical challenge to treating patients. Thus, this novel senolytic approach may revolutionize current CDK4/6i mediated melanoma treatment if melanoma cells undergo senescence prior to developing resistance to CDK4/6i. Given that we demonstrate that a low dose of vemurafenib induced senescence, which renders BrafV600E melanoma cells susceptible to GLS1i and recent accumulated research shows many cancer cells undergo senescence in response to chemotherapy, radiation, and immunotherapy, this senolytic therapy approach may prove applicable to a wide range of cancer types once senescence and GLS1 expression are induced.
细胞周期蛋白D1-细胞周期蛋白依赖性激酶4和6(CDK4/6)复合物对黑色素瘤的发展至关重要。我们曾证实,使用小分子抑制剂(CDK4/6i)靶向 CDK4/6,可通过诱导细胞衰老抑制 BrafV600E 黑色素瘤在体外和体内的生长。研究CDK4/6i治疗黑色素瘤的临床试验并未取得成功,这凸显了提高CDK4/6i疗效的必要性。累积的研究表明,虽然衰老最初会抑制细胞增殖,但长期的衰老状态最终会通过改变肿瘤微环境而导致肿瘤复发,这表明清除这些衰老细胞(在称为衰老分解的过程中)对促进临床反应具有临床必要性。我们证明,谷氨酰胺酶1(GLS1)的表达在CDK4/6i诱导的衰老BrafV600E黑色素瘤细胞中特异性上调。上调的 GLS1 表达使 BrafV600E 黑色素瘤衰老细胞易受 GLS1 抑制剂(GLS1i)的影响。此外,我们还证明,即使这些细胞对 BrafV600E 抑制剂 vemurafenib 产生耐药性,这种针对 GLS1 表达上调的衰老溶解方法仍然适用,而这正是临床治疗患者时经常遇到的巨大挑战。因此,如果黑色素瘤细胞在对CDK4/6i产生抗药性之前发生衰老,那么这种新型的衰老分解方法可能会彻底改变目前CDK4/6i介导的黑色素瘤治疗方法。鉴于我们证明了低剂量的维莫非尼能诱导衰老,从而使 BrafV600E 黑色素瘤细胞对 GLS1i 易感,而且最近积累的研究表明,许多癌细胞在对化疗、放疗和免疫疗法做出反应时都会发生衰老,因此一旦诱导衰老和 GLS1 表达,这种衰老疗法可能会被证明适用于多种癌症类型。
{"title":"Inhibition of glutaminase elicits senolysis in therapy-induced senescent melanoma cells.","authors":"Justin Kim, Bryce Brunetti, Ayanesh Kumar, Ankit Mangla, Kord Honda, Akihiro Yoshida","doi":"10.1101/2024.09.12.612728","DOIUrl":"https://doi.org/10.1101/2024.09.12.612728","url":null,"abstract":"The cyclin D1-Cyclin-Dependent Kinases 4 and 6 (CDK4/6) complex is crucial for the development of melanoma. We previously demonstrated that targeting CDK4/6 using small molecule inhibitors (CDK4/6i) suppresses BrafV600E melanoma growth in vitro and in vivo through induction of cellular senescence. Clinical trials investigating CDK4/6i in melanoma have not yielded successful outcomes, underscoring the necessity to enhance the therapeutic efficacy of CDK4/6i. Accumulated research has shown that while senescence initially suppresses cell proliferation, a prolonged state of senescence eventually leads to tumor relapse by altering the tumor microenvironment, suggesting that removal of those senescent cells (in a process referred to as senolysis) is of clinical necessity to facilitate clinical response. We demonstrate that glutaminase 1 (GLS1) expression is specifically upregulated in CDK4/6i-induced senescent BrafV600E melanoma cells. Upregulated GLS1 expression renders BrafV600E melanoma senescent cells vulnerable to GLS1 inhibitor (GLS1i). Furthermore, we demonstrate that this senolytic approach targeting upregulated GLS1 expression is applicable even though those cells developed resistance to the BrafV600E inhibitor vemurafenib, a frequently encountered substantial clinical challenge to treating patients. Thus, this novel senolytic approach may revolutionize current CDK4/6i mediated melanoma treatment if melanoma cells undergo senescence prior to developing resistance to CDK4/6i. Given that we demonstrate that a low dose of vemurafenib induced senescence, which renders BrafV600E melanoma cells susceptible to GLS1i and recent accumulated research shows many cancer cells undergo senescence in response to chemotherapy, radiation, and immunotherapy, this senolytic therapy approach may prove applicable to a wide range of cancer types once senescence and GLS1 expression are induced.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"15 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249760","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SOX2 drives fetal reprogramming and reversible dormancy in colorectal cancer SOX2 驱动胎儿重编程和结直肠癌的可逆休眠
Pub Date : 2024-09-16 DOI: 10.1101/2024.09.11.612412
Anna Baulies, Veronica Moncho-Amor, Diana Drago-Garcia, Anna Kucharska, Probir Chakravarty, Manuel Moreno-Valladares, Sara Cruces-Salguero, Florian Hubl, Colin Hutton, Haeun Kim, Ander Matheu, Robin Lovell-Badge, Vivian S.W. Li
Cellular plasticity plays critical roles in tissue regeneration, tumour progression and therapeutic resistance. However, the mechanism underlying this cell state transition remains elusive. Here, we show that the transcription factor SOX2 induces fetal reprogramming and reversible dormancy in colorectal cancer (CRC). SOX2 expression correlates with fetal reprogramming and poor prognosis in human primary and metastatic colorectal adenocarcinomas. In mouse CRC models, rare slow-cycling SOX2+SCA1+ cells are detected in early Apc-deleted mouse tumours that undergo slow clonal expansion over time. In contrast, the SOX2+ clones were found proliferative in advanced Apc-/-;KrasG12D/+;p53-/-;Tgfbr2-/- (AKPT) tumours, accompanied by dynamic cell state reprogramming from LGR5+ to LGR5-SCA1+ cells. Using transgenic mouse models, we demonstrate that ectopic expression of SOX2 inhibits intestinal lineage differentiation and induces fetal reprogramming. SOX2+ cells adopt dynamic cell cycle states depending on its expression level. High SOX2 expression results in hyperproliferation, whereas low SOX2 levels induce senescence-mediated dormancy. We find that loss of p53 can reverse SOX2-induced senescence, in line with the dormant cell state exit of the SOX2+ cells observed in advanced tumours. Finally, SOX2 expression is induced by 5-FU treatment in CRC. SOX2-expressing organoids exhibit increased tolerance to chemotherapy treatment, whilst deletion of SOX2 in AKPT tumour organoids sensitises drug responses. We propose that SOX2-induced plasticity and reversible dormancy promotes tumour progression and drug tolerance in CRC.
细胞可塑性在组织再生、肿瘤进展和抗药性方面发挥着至关重要的作用。然而,这种细胞状态转变的内在机制仍然难以捉摸。在这里,我们发现转录因子 SOX2 可诱导结直肠癌(CRC)的胎儿重编程和可逆休眠。在人类原发性和转移性结直肠腺癌中,SOX2 的表达与胎儿重编程和不良预后相关。在小鼠 CRC 模型中,早期 Apc 缺失的小鼠肿瘤中检测到罕见的慢周期 SOX2+SCA1+ 细胞,随着时间的推移,这些细胞会发生缓慢的克隆扩增。相反,在晚期Apc-/-;KrasG12D/+;p53-/-;Tgfbr2-/-(AKPT)肿瘤中发现SOX2+克隆具有增殖性,并伴随着从LGR5+到LGR5-SCA1+细胞的动态细胞状态重编程。我们利用转基因小鼠模型证明,异位表达 SOX2 会抑制肠系分化并诱导胎儿重编程。SOX2+细胞根据其表达水平的不同而呈现动态的细胞周期状态。SOX2的高表达会导致细胞过度增殖,而低水平的SOX2则会诱导衰老介导的休眠。我们发现,p53 的缺失可以逆转 SOX2 诱导的衰老,这与晚期肿瘤中观察到的 SOX2+ 细胞退出休眠的细胞状态是一致的。最后,5-FU 治疗诱导了 CRC 中 SOX2 的表达。表达 SOX2 的器官组织对化疗的耐受性增强,而在 AKPT 肿瘤器官组织中缺失 SOX2 会使药物反应变得敏感。我们认为,SOX2诱导的可塑性和可逆休眠促进了肿瘤进展和对药物的耐受性。
{"title":"SOX2 drives fetal reprogramming and reversible dormancy in colorectal cancer","authors":"Anna Baulies, Veronica Moncho-Amor, Diana Drago-Garcia, Anna Kucharska, Probir Chakravarty, Manuel Moreno-Valladares, Sara Cruces-Salguero, Florian Hubl, Colin Hutton, Haeun Kim, Ander Matheu, Robin Lovell-Badge, Vivian S.W. Li","doi":"10.1101/2024.09.11.612412","DOIUrl":"https://doi.org/10.1101/2024.09.11.612412","url":null,"abstract":"Cellular plasticity plays critical roles in tissue regeneration, tumour progression and therapeutic resistance. However, the mechanism underlying this cell state transition remains elusive. Here, we show that the transcription factor SOX2 induces fetal reprogramming and reversible dormancy in colorectal cancer (CRC). SOX2 expression correlates with fetal reprogramming and poor prognosis in human primary and metastatic colorectal adenocarcinomas. In mouse CRC models, rare slow-cycling SOX2+SCA1+ cells are detected in early Apc-deleted mouse tumours that undergo slow clonal expansion over time. In contrast, the SOX2+ clones were found proliferative in advanced Apc-/-;KrasG12D/+;p53-/-;Tgfbr2-/- (AKPT) tumours, accompanied by dynamic cell state reprogramming from LGR5+ to LGR5-SCA1+ cells. Using transgenic mouse models, we demonstrate that ectopic expression of SOX2 inhibits intestinal lineage differentiation and induces fetal reprogramming. SOX2+ cells adopt dynamic cell cycle states depending on its expression level. High SOX2 expression results in hyperproliferation, whereas low SOX2 levels induce senescence-mediated dormancy. We find that loss of p53 can reverse SOX2-induced senescence, in line with the dormant cell state exit of the SOX2+ cells observed in advanced tumours. Finally, SOX2 expression is induced by 5-FU treatment in CRC. SOX2-expressing organoids exhibit increased tolerance to chemotherapy treatment, whilst deletion of SOX2 in AKPT tumour organoids sensitises drug responses. We propose that SOX2-induced plasticity and reversible dormancy promotes tumour progression and drug tolerance in CRC.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"8 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Screening activity of brain cancer-derived factors on primary human brain pericytes 筛选脑癌衍生因子对原发性人脑周细胞的活性
Pub Date : 2024-09-16 DOI: 10.1101/2024.09.11.612547
Samuel JC McCullough, Eliene Albers, Akshata Anchan, Jane Yu, Bronwen Connor, Scott Graham
Brain cancers offer poor prognoses to patients accompanied by symptoms that drastically impact the patient and their family. Brain tumours recruit local non-transformed cells to provide trophic support and immunosuppression within the tumour microenvironment, supporting tumour progression. Given the localization and supportive role of pericytes at the brain vasculature, we explored the potential for brain pericytes to contribute to the brain cancer microenvironment. To investigate this, primary brain pericytes were treated with factors commonly upregulated in brain cancers. Changes to brain pericyte cell signalling, inflammatory secretion, and phagocytosis were investigated. The TGFβ superfamily cytokines TGFβ and GDF-15 activated SMAD2/3 and inhibited C/EBP-δ, revealing a potential mechanism behind the pleiotropic action of TGFβ on brain pericytes. IL-17 induced secretion of IL-6 without activating NFκB, STAT1, SMAD2/3, or C/EBP-δ signalling pathways. IL-27 and IFNγ induced STAT1 signalling and significantly reduced pericyte phagocytosis. The remaining brain cancer-derived factors did not induce a measured response, indicating that these factors may act on other cell types or require co-stimulation with other factors to produce significant effects. Together, these findings show potential mechanisms by which brain pericytes contribute to aspects of inflammation and starts to uncover the supportive role brain pericytes may play in brain cancers.
脑癌患者的预后较差,并伴有严重影响患者及其家庭的症状。脑肿瘤会招募局部非转化细胞,在肿瘤微环境中提供营养支持和免疫抑制,从而支持肿瘤的进展。鉴于周细胞在脑血管的定位和支持作用,我们探索了脑周细胞对脑癌微环境做出贡献的可能性。为了研究这一点,我们用脑癌中常见的上调因子处理原代脑周细胞。我们研究了脑周质细胞信号、炎症分泌和吞噬作用的变化。TGFβ超家族细胞因子TGFβ和GDF-15激活了SMAD2/3并抑制了C/EBP-δ,揭示了TGFβ对脑周细胞的多效应背后的潜在机制。IL-17 可诱导 IL-6 的分泌,但不会激活 NFκB、STAT1、SMAD2/3 或 C/EBP-δ 信号通路。IL-27 和 IFNγ 可诱导 STAT1 信号传导,并显著降低周细胞的吞噬能力。其余的脑癌衍生因子没有诱导出可测量的反应,这表明这些因子可能作用于其他类型的细胞,或需要与其他因子共同刺激才能产生显著效果。这些发现共同显示了脑周质对炎症方面起作用的潜在机制,并开始揭示脑周质在脑癌中可能发挥的支持作用。
{"title":"Screening activity of brain cancer-derived factors on primary human brain pericytes","authors":"Samuel JC McCullough, Eliene Albers, Akshata Anchan, Jane Yu, Bronwen Connor, Scott Graham","doi":"10.1101/2024.09.11.612547","DOIUrl":"https://doi.org/10.1101/2024.09.11.612547","url":null,"abstract":"Brain cancers offer poor prognoses to patients accompanied by symptoms that drastically impact the patient and their family. Brain tumours recruit local non-transformed cells to provide trophic support and immunosuppression within the tumour microenvironment, supporting tumour progression. Given the localization and supportive role of pericytes at the brain vasculature, we explored the potential for brain pericytes to contribute to the brain cancer microenvironment. To investigate this, primary brain pericytes were treated with factors commonly upregulated in brain cancers. Changes to brain pericyte cell signalling, inflammatory secretion, and phagocytosis were investigated. The TGFβ superfamily cytokines TGFβ and GDF-15 activated SMAD2/3 and inhibited C/EBP-δ, revealing a potential mechanism behind the pleiotropic action of TGFβ on brain pericytes. IL-17 induced secretion of IL-6 without activating NFκB, STAT1, SMAD2/3, or C/EBP-δ signalling pathways. IL-27 and IFNγ induced STAT1 signalling and significantly reduced pericyte phagocytosis. The remaining brain cancer-derived factors did not induce a measured response, indicating that these factors may act on other cell types or require co-stimulation with other factors to produce significant effects. Together, these findings show potential mechanisms by which brain pericytes contribute to aspects of inflammation and starts to uncover the supportive role brain pericytes may play in brain cancers.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249798","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hypoxic stress dysregulates functions of glioma-associated myeloid cells through epigenomic and transcriptional programs 缺氧应激通过表观基因组和转录程序失调胶质瘤相关髓系细胞的功能
Pub Date : 2024-09-16 DOI: 10.1101/2024.09.12.612769
Monika Dzwigonska, Patrycja Rosa, Beata Kaza, Salwador Cyranowski, Aleksandra Ellert-Miklaszewska, Agata Kominek, Tomasz Obrebski, Anna R Malik, Katarzyna Piwocka, Jakub Mieczkowski, Bozena Kaminska, Katarzyna B Leszczynska
Hypoxia rapidly alters gene expression to allow cellular adaptation to challenging conditions and support tumour growth. Hypoxia also affects the chromatin structure by modifications of histones and DNA methylation. Glioblastoma (GBM) is an aggressive, deadly primary brain tumour for which there is no effective treatment. The tumour microenvironment of GBM is highly heterogeneous, with infiltration of glioma-associated microglia and macrophages (GAMs) and the presence of necrotic, hypoxic regions. The mechanisms through which hypoxia alters the tumour microenvironment and regulates functions of infiltrating immune cells remain poorly understood. Here, we show that hypoxia modulates the expression of myeloid markers in distinct ways: upregulates the monocytic marker Lgals3 expression and downregulates the microglial markers P2ry12 and Tmem119 in microglial and monocytic GAMs in vitro and in vivo, as shown using human and mouse GBM single-cell transcriptomics datasets. The genome-wide hypoxia-dependent transcriptomic changes in microglial cells were determined in microglia-glioma co-cultures. Numerous GAM subtype markers were dysregulated in response to hypoxic stress due to associated changes in chromatin accessibility, as determined using ATACseq. While hypoxia alone drives a decrease of the overall chromatin accessibility at gene promoters, the exposure to glioma cells under hypoxic conditions leads to both increases and decreases of chromatin accessibility at promoter regions in microglial cells. Hypoxia downregulates the chromatin accessibility at the regions enriched in motifs of transcription factors regarded as master regulators of microglial cell identity and function, including SPI1 or IRF8. Overall, our data highlights the importance of hypoxic stress as a strong intratumoral regulator of myeloid cell functions, which adds complexity to the characterisation of particular GAMs subpopulations.
缺氧会迅速改变基因表达,使细胞适应具有挑战性的环境并支持肿瘤生长。缺氧还会通过组蛋白和 DNA 甲基化的修饰影响染色质结构。胶质母细胞瘤(GBM)是一种侵袭性致命原发性脑肿瘤,目前尚无有效的治疗方法。GBM 的肿瘤微环境具有高度异质性,有胶质瘤相关小胶质细胞和巨噬细胞(GAMs)浸润,并存在坏死和缺氧区域。人们对缺氧改变肿瘤微环境和调节浸润免疫细胞功能的机制仍然知之甚少。在这里,我们利用人类和小鼠 GBM 单细胞转录组学数据集显示,缺氧以不同的方式调节髓系标志物的表达:在体外和体内的小胶质细胞和单核细胞 GAMs 中,上调单核细胞标志物 Lgals3 的表达,下调小胶质细胞标志物 P2ry12 和 Tmem119 的表达。在小胶质细胞-胶质瘤共培养物中测定了小胶质细胞中全基因组缺氧依赖性转录组变化。使用 ATACseq 测定发现,由于染色质可及性的相关变化,许多 GAM 亚型标记物在缺氧压力下出现失调。虽然缺氧本身会导致基因启动子的染色质可及性整体下降,但在缺氧条件下暴露于胶质瘤细胞会导致小胶质细胞启动子区域染色质可及性的增加和减少。缺氧会降低富含被认为是小胶质细胞特性和功能主调控因子的转录因子(包括 SPI1 或 IRF8)的区域的染色质可及性。总之,我们的数据凸显了缺氧应激作为瘤内髓系细胞功能强调控因子的重要性,这增加了特定 GAMs 亚群特征描述的复杂性。
{"title":"Hypoxic stress dysregulates functions of glioma-associated myeloid cells through epigenomic and transcriptional programs","authors":"Monika Dzwigonska, Patrycja Rosa, Beata Kaza, Salwador Cyranowski, Aleksandra Ellert-Miklaszewska, Agata Kominek, Tomasz Obrebski, Anna R Malik, Katarzyna Piwocka, Jakub Mieczkowski, Bozena Kaminska, Katarzyna B Leszczynska","doi":"10.1101/2024.09.12.612769","DOIUrl":"https://doi.org/10.1101/2024.09.12.612769","url":null,"abstract":"Hypoxia rapidly alters gene expression to allow cellular adaptation to challenging conditions and support tumour growth. Hypoxia also affects the chromatin structure by modifications of histones and DNA methylation. Glioblastoma (GBM) is an aggressive, deadly primary brain tumour for which there is no effective treatment. The tumour microenvironment of GBM is highly heterogeneous, with infiltration of glioma-associated microglia and macrophages (GAMs) and the presence of necrotic, hypoxic regions. The mechanisms through which hypoxia alters the tumour microenvironment and regulates functions of infiltrating immune cells remain poorly understood. Here, we show that hypoxia modulates the expression of myeloid markers in distinct ways: upregulates the monocytic marker Lgals3 expression and downregulates the microglial markers P2ry12 and Tmem119 in microglial and monocytic GAMs in vitro and in vivo, as shown using human and mouse GBM single-cell transcriptomics datasets. The genome-wide hypoxia-dependent transcriptomic changes in microglial cells were determined in microglia-glioma co-cultures. Numerous GAM subtype markers were dysregulated in response to hypoxic stress due to associated changes in chromatin accessibility, as determined using ATACseq. While hypoxia alone drives a decrease of the overall chromatin accessibility at gene promoters, the exposure to glioma cells under hypoxic conditions leads to both increases and decreases of chromatin accessibility at promoter regions in microglial cells. Hypoxia downregulates the chromatin accessibility at the regions enriched in motifs of transcription factors regarded as master regulators of microglial cell identity and function, including SPI1 or IRF8. Overall, our data highlights the importance of hypoxic stress as a strong intratumoral regulator of myeloid cell functions, which adds complexity to the characterisation of particular GAMs subpopulations.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"59 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MLPA: A Multi-scale Digital Twin Framework for Personalized Cancer Simulation and Treatment Optimization MLPA:用于个性化癌症模拟和治疗优化的多尺度数字双胞胎框架
Pub Date : 2024-09-16 DOI: 10.1101/2024.09.13.612988
Jake Y. Chen, James C Gu
We introduce the Multi-level Parameterized Automata (MLPA), an innovative digital twin model that revolutionizes personalized cancer growth simulation and treatment optimization. MLPA integrates macroscopic electronic health records and microscopic genomic data, employing stochastic cellular automata to model tumor progression and treatment efficacy dynamically. This multi-scale approach enables MLPA to simulate complex cancer behaviors, including metastasis and pharmacological responses, with remarkable precision. Our validation using bioluminescent imaging from mice demonstrates MLPA's exceptional predictive power, achieving an improvement in accuracy over baseline models for tumor growth prediction. The model accurately captures tumors' characteristic S-shaped growth curve and shows high fidelity in simulating various scenarios, from natural progression to aggressive growth and drug treatment responses. MLPA's ability to simulate drug effects through gene pathway perturbation, validated through equivalence testing, underscores its potential as a powerful tool for precision oncology. The framework offers a robust platform for exploring personalized treatment strategies, potentially transforming patient outcomes by optimizing therapy based on individual biological profiles. We present the theoretical foundation, implementation, and validation of MLPA, highlighting its capacity to advance the field of computational oncology and foster more effective, tailored cancer treatment solutions. As we progress towards precision medicine, MLPA stands at the forefront, offering new possibilities in cancer modeling and treatment optimization. The code and imaging dataset used is available at https://github.com/alphamind-club/MLPA.
我们介绍了多级参数化自动机(MLPA),这是一种创新的数字孪生模型,可彻底改变个性化癌症生长模拟和治疗优化。MLPA 整合了宏观电子健康记录和微观基因组数据,采用随机细胞自动机对肿瘤进展和治疗效果进行动态建模。这种多尺度方法使 MLPA 能够非常精确地模拟复杂的癌症行为,包括转移和药理反应。我们利用小鼠的生物发光成像进行了验证,证明了 MLPA 的卓越预测能力,在肿瘤生长预测方面比基线模型的准确性有所提高。该模型准确捕捉了肿瘤特征性的 S 型生长曲线,并在模拟各种情况(从自然进展到侵袭性生长和药物治疗反应)时表现出很高的保真度。通过等效性测试验证,MLPA 能够通过基因通路扰动来模拟药物效应,这突显了它作为精准肿瘤学强大工具的潜力。该框架为探索个性化治疗策略提供了一个强大的平台,通过根据个体生物特征优化治疗,有可能改变患者的预后。我们介绍了 MLPA 的理论基础、实施和验证,强调了它在推动计算肿瘤学领域发展和促进更有效、量身定制的癌症治疗方案方面的能力。随着我们向精准医学迈进,MLPA 站在了最前沿,为癌症建模和治疗优化提供了新的可能性。使用的代码和成像数据集可在 https://github.com/alphamind-club/MLPA 上获取。
{"title":"MLPA: A Multi-scale Digital Twin Framework for Personalized Cancer Simulation and Treatment Optimization","authors":"Jake Y. Chen, James C Gu","doi":"10.1101/2024.09.13.612988","DOIUrl":"https://doi.org/10.1101/2024.09.13.612988","url":null,"abstract":"We introduce the Multi-level Parameterized Automata (MLPA), an innovative digital twin model that revolutionizes personalized cancer growth simulation and treatment optimization. MLPA integrates macroscopic electronic health records and microscopic genomic data, employing stochastic cellular automata to model tumor progression and treatment efficacy dynamically. This multi-scale approach enables MLPA to simulate complex cancer behaviors, including metastasis and pharmacological responses, with remarkable precision. Our validation using bioluminescent imaging from mice demonstrates MLPA's exceptional predictive power, achieving an improvement in accuracy over baseline models for tumor growth prediction. The model accurately captures tumors' characteristic S-shaped growth curve and shows high fidelity in simulating various scenarios, from natural progression to aggressive growth and drug treatment responses. MLPA's ability to simulate drug effects through gene pathway perturbation, validated through equivalence testing, underscores its potential as a powerful tool for precision oncology. The framework offers a robust platform for exploring personalized treatment strategies, potentially transforming patient outcomes by optimizing therapy based on individual biological profiles. We present the theoretical foundation, implementation, and validation of MLPA, highlighting its capacity to advance the field of computational oncology and foster more effective, tailored cancer treatment solutions. As we progress towards precision medicine, MLPA stands at the forefront, offering new possibilities in cancer modeling and treatment optimization. The code and imaging dataset used is available at https://github.com/alphamind-club/MLPA.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"36 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249799","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MITF Targets in Gastrointestinal Stromal Tumors: Implication in Autophagy and Extracellular Vesicle Secretion 胃肠道间质瘤中的 MITF 靶点:自噬和细胞外囊泡分泌的影响
Pub Date : 2024-09-15 DOI: 10.1101/2024.09.10.612253
Elizabeth Proaño-Pérez, Eva Serrano-Candelas, Mario Guerrero, David Gómez-Peregrina, Carlos Llorens, Beatriz Soriano, Ana Gámez-Valero, Marina Herrero-Lorenzo, Eulalia Marti, Cesar Serrano, Margarita Martin
Previous studies have identified Microphthalmia-associated Transcription Factor (MITF) involvement in regulating Gastrointestinal Stromal Tumors (GIST) growth and cell cycle progression. This study uses Chromatin Immunoprecipitation combined with high-throughput sequencing (ChIP-seq) and RNA sequencing to explore MITF-modulated genes in GIST. Our findings reveal that MITF regulates genes involved in lysosome biogenesis, vesicle generation, autophagy, and mTOR signaling pathways. Comparative transcriptome analysis following MITF silencing in GIST cells shows differential enrichment in mTOR signaling, impacting tumor growth and autophagy. In the context of cancer, the interplay between autophagy and extracellular vesicle release can influence tumor progression and metastasis. We examined MITF's role in autophagy and extracellular vesicle (EV) production in GIST, finding that MITF overexpression increases autophagy, as shown by elevated LC3II levels while silencing MITF disrupts autophagosome and autolysosome formation. Despite no significant changes in EV size or number, MITF silencing notably reduces KIT expression in EV content. KIT secretion in EVs has been linked to GIST metastasis, suggesting that MITF is a crucial target for managing tumor growth and metastasis in GIST.
先前的研究发现,微眼球相关转录因子(MITF)参与调控胃肠道间质瘤(GIST)的生长和细胞周期进展。本研究利用染色质免疫共沉淀结合高通量测序(ChIP-seq)和 RNA 测序来探索 GIST 中受 MITF 调控的基因。我们的研究结果表明,MITF调控参与溶酶体生物生成、囊泡生成、自噬和mTOR信号通路的基因。在 GIST 细胞中沉默 MITF 后进行的比较转录组分析表明,mTOR 信号通路在不同程度上富集,影响肿瘤生长和自噬。在癌症中,自噬和细胞外囊泡释放之间的相互作用会影响肿瘤的进展和转移。我们研究了 MITF 在 GIST 中自噬和细胞外囊泡 (EV) 生成中的作用,发现 MITF 过表达会增加自噬,表现为 LC3II 水平升高,而沉默 MITF 则会破坏自噬体和自溶体的形成。尽管EV大小或数量没有明显变化,但沉默MITF会明显减少EV内容物中KIT的表达。EV 中 KIT 的分泌与 GIST 的转移有关,这表明 MITF 是管理 GIST 肿瘤生长和转移的关键靶点。
{"title":"MITF Targets in Gastrointestinal Stromal Tumors: Implication in Autophagy and Extracellular Vesicle Secretion","authors":"Elizabeth Proaño-Pérez, Eva Serrano-Candelas, Mario Guerrero, David Gómez-Peregrina, Carlos Llorens, Beatriz Soriano, Ana Gámez-Valero, Marina Herrero-Lorenzo, Eulalia Marti, Cesar Serrano, Margarita Martin","doi":"10.1101/2024.09.10.612253","DOIUrl":"https://doi.org/10.1101/2024.09.10.612253","url":null,"abstract":"Previous studies have identified Microphthalmia-associated Transcription Factor (MITF) involvement in regulating Gastrointestinal Stromal Tumors (GIST) growth and cell cycle progression. This study uses Chromatin Immunoprecipitation combined with high-throughput sequencing (ChIP-seq) and RNA sequencing to explore MITF-modulated genes in GIST. Our findings reveal that MITF regulates genes involved in lysosome biogenesis, vesicle generation, autophagy, and mTOR signaling pathways. Comparative transcriptome analysis following MITF silencing in GIST cells shows differential enrichment in mTOR signaling, impacting tumor growth and autophagy. In the context of cancer, the interplay between autophagy and extracellular vesicle release can influence tumor progression and metastasis. We examined MITF's role in autophagy and extracellular vesicle (EV) production in GIST, finding that MITF overexpression increases autophagy, as shown by elevated LC3II levels while silencing MITF disrupts autophagosome and autolysosome formation. Despite no significant changes in EV size or number, MITF silencing notably reduces KIT expression in EV content. KIT secretion in EVs has been linked to GIST metastasis, suggesting that MITF is a crucial target for managing tumor growth and metastasis in GIST.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"113 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
U2AF1 S34F enhances tumorigenic potential of lung cells by exhibiting synergy with KRAS mutation and altering response to environmental stress U2AF1 S34F 与 KRAS 突变协同作用并改变对环境压力的反应,从而增强肺细胞的致瘤潜力
Pub Date : 2024-09-15 DOI: 10.1101/2024.09.11.612492
Cindy E Liang, Eva Hrabeta-Robinson, Amit Behera, Carlos Arevalo, Isobel J Fetter, Cameron M Soulette, Alexis M Thornton, Shaheen Sikandar, Angela N Brooks
Although U2AF1S34F is a recurrent splicing factor mutation in lung adenocarcinoma (ADC), U2AF1S34F alone is insufficient for producing tumors in previous models. Because lung ADCs with U2AF1S34F frequently have co-occurring KRAS mutations and smoking histories, we hypothesized that tumor-forming potential arises from U2AF1S34F interacting with oncogenic KRAS and environmental stress. To elucidate the effect of U2AF1S34F co-occurring with a second mutation, we generated human bronchial epithelial cells (HBEC3kt) with co-occurring U2AF1S34F and KRASG12V. Transcriptome analysis revealed that co-occurring U2AF1S34F and KRASG12V differentially impacts inflammatory, cell cycle, and KRAS pathways. Subsequent phenotyping found associated suppressed cytokine production, increased proliferation, anchorage-independent growth, and tumors in mouse xenografts. Interestingly, HBEC3kts harboring only U2AF1S34F display increased splicing in stress granule protein genes and viability in cigarette smoke concentrate. Our results suggest that U2AF1S34F may potentiate transformation by granting precancerous cells survival advantage in environmental stress, permitting accumulation of additional mutations like KRASG12V, which synergize with U2AF1S34F to transform the cell.
虽然 U2AF1S34F 是肺腺癌(ADC)中反复出现的剪接因子突变,但在以前的模型中,U2AF1S34F 本身不足以产生肿瘤。由于带有 U2AF1S34F 的肺腺癌经常同时伴有 KRAS 突变和吸烟史,我们假设肿瘤形成的潜力来自 U2AF1S34F 与致癌 KRAS 和环境压力的相互作用。为了阐明 U2AF1S34F 与第二个突变共存的影响,我们生成了 U2AF1S34F 和 KRASG12V 共存的人支气管上皮细胞(HBEC3kt)。转录组分析表明,U2AF1S34F 和 KRASG12V 共存会对炎症、细胞周期和 KRAS 通路产生不同的影响。随后的表型分析发现,在小鼠异种移植中,相关的细胞因子产生受到抑制、增殖增加、不依赖锚的生长和肿瘤。有趣的是,只携带 U2AF1S34F 的 HBEC3kts 在香烟烟雾浓缩物中显示出应激颗粒蛋白基因剪接的增加和存活率的提高。我们的研究结果表明,U2AF1S34F 可使癌前细胞在环境压力下获得生存优势,从而使 KRASG12V 等其他突变积累,并与 U2AF1S34F 协同使细胞发生转化,从而增强转化的效力。
{"title":"U2AF1 S34F enhances tumorigenic potential of lung cells by exhibiting synergy with KRAS mutation and altering response to environmental stress","authors":"Cindy E Liang, Eva Hrabeta-Robinson, Amit Behera, Carlos Arevalo, Isobel J Fetter, Cameron M Soulette, Alexis M Thornton, Shaheen Sikandar, Angela N Brooks","doi":"10.1101/2024.09.11.612492","DOIUrl":"https://doi.org/10.1101/2024.09.11.612492","url":null,"abstract":"Although U2AF1<sup>S34F</sup> is a recurrent splicing factor mutation in lung adenocarcinoma (ADC), U2AF1<sup>S34F</sup> alone is insufficient for producing tumors in previous models. Because lung ADCs with U2AF1<sup>S34F</sup> frequently have co-occurring KRAS mutations and smoking histories, we hypothesized that tumor-forming potential arises from U2AF1<sup>S34F</sup> interacting with oncogenic KRAS and environmental stress. To elucidate the effect of U2AF1<sup>S34F</sup> co-occurring with a second mutation, we generated human bronchial epithelial cells (HBEC3kt) with co-occurring U2AF1<sup>S34F</sup> and KRAS<sup>G12V</sup>. Transcriptome analysis revealed that co-occurring U2AF1<sup>S34F</sup> and KRAS<sup>G12V</sup> differentially impacts inflammatory, cell cycle, and KRAS pathways. Subsequent phenotyping found associated suppressed cytokine production, increased proliferation, anchorage-independent growth, and tumors in mouse xenografts. Interestingly, HBEC3kts harboring only U2AF1<sup>S34F</sup> display increased splicing in stress granule protein genes and viability in cigarette smoke concentrate. Our results suggest that U2AF1<sup>S34F</sup> may potentiate transformation by granting precancerous cells survival advantage in environmental stress, permitting accumulation of additional mutations like KRAS<sup>G12V</sup>, which synergize with U2AF1<sup>S34F</sup> to transform the cell.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":"40 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249801","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
bioRxiv - Cancer Biology
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