首页 > 最新文献

Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response最新文献

英文 中文
Anti-inflammatory combinatorial therapy to enhance killing efficacy with patient-derived preclinical models 抗炎联合治疗增强患者临床前模型的杀伤效果
Jing Zhang, B. L. Khoo
Many chemotherapeutic drugs induce oxidative stress by accelerating the accumulation of reactive oxygen species (ROS), which triggers the death of cancer cells and then causes severe DNA damage in cancer cells. Here, we proposed using a preclinical microfluidic model to evaluate the combination of doxorubicin and aspirin (DA) for anti-inflammatory therapy using patient-derived circulating tumor cell (CTC) clusters. The preclinical model could perform high-throughput screening of drug combinations and used valves to regulate media inflow for CTC cluster formation. We demonstrated that low-dose aspirin (445–500 mg/ml) and a suboptimal dose of doxorubicin (0.5 D) for seven days could produce higher killing efficacy and significantly reduced the proportion of cancer stem cells and colony-forming ability. Compared with the treatment with doxorubicin alone, the intracellular oxidative activity in the sample under combinatorial DA treatment was reduced, as demonstrated by the intensity of Calcein AM. We demonstrated that the treatment outcomes were mediated by the reduction of COX-2, which was associated with inflammation triggered by ROS. Overall, the preclinical model could be used as a proof of concept to demonstrate the efficacy of anti-inflammatory combinatorial therapies by influencing oxidative stress. Similar research could provide a basis for more DNA-related cancer treatment research in the future.
许多化疗药物通过加速活性氧(ROS)的积累来诱导氧化应激,从而触发癌细胞的死亡,然后在癌细胞中引起严重的DNA损伤。在这里,我们建议使用临床前微流控模型来评估阿霉素和阿司匹林(DA)联合使用患者源性循环肿瘤细胞(CTC)集群进行抗炎治疗的效果。该临床前模型可以进行高通量药物组合筛选,并使用阀门调节CTC簇形成的介质流入。我们证明,低剂量阿司匹林(445-500 mg/ml)和次优剂量阿霉素(0.5 D)持续7天可以产生更高的杀伤效果,并显著降低癌症干细胞的比例和集落形成能力。与阿霉素单独处理相比,复合DA处理的样品细胞内氧化活性降低,这可以从钙黄蛋白AM的强度看出。我们证明了治疗结果是由COX-2的减少介导的,这与ROS引发的炎症有关。总的来说,临床前模型可以作为一种概念证明,通过影响氧化应激来证明抗炎联合疗法的有效性。类似的研究可以为未来更多与dna相关的癌症治疗研究提供基础。
{"title":"Anti-inflammatory combinatorial therapy to enhance killing efficacy with patient-derived preclinical models","authors":"Jing Zhang, B. L. Khoo","doi":"10.3390/iecc2021-09204","DOIUrl":"https://doi.org/10.3390/iecc2021-09204","url":null,"abstract":"Many chemotherapeutic drugs induce oxidative stress by accelerating the accumulation of reactive oxygen species (ROS), which triggers the death of cancer cells and then causes severe DNA damage in cancer cells. Here, we proposed using a preclinical microfluidic model to evaluate the combination of doxorubicin and aspirin (DA) for anti-inflammatory therapy using patient-derived circulating tumor cell (CTC) clusters. The preclinical model could perform high-throughput screening of drug combinations and used valves to regulate media inflow for CTC cluster formation. We demonstrated that low-dose aspirin (445–500 mg/ml) and a suboptimal dose of doxorubicin (0.5 D) for seven days could produce higher killing efficacy and significantly reduced the proportion of cancer stem cells and colony-forming ability. Compared with the treatment with doxorubicin alone, the intracellular oxidative activity in the sample under combinatorial DA treatment was reduced, as demonstrated by the intensity of Calcein AM. We demonstrated that the treatment outcomes were mediated by the reduction of COX-2, which was associated with inflammation triggered by ROS. Overall, the preclinical model could be used as a proof of concept to demonstrate the efficacy of anti-inflammatory combinatorial therapies by influencing oxidative stress. Similar research could provide a basis for more DNA-related cancer treatment research in the future.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89773916","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
MicroRNA Expression Analysis and Biological Pathways in Chemoresistant Non- Small Cell Lung Cancer 化疗耐药非小细胞肺癌的MicroRNA表达分析及生物学途径
S. Agelaki, Krystallia Gourlia, M. Markaki, C. Papadaki, K. Rounis, E. Vorrias, Elena Prokova, E. Lagoudaki, A. Koutsopoulos, I. Tsamardinos, D. Mavroudis
: Platinum-based chemotherapy (CT) is a standard treatment for lung cancer, however a variety of chemoresistance mechanisms can impair its efficacy. MicroRNAs (miRNAs) represent potential biomarkers for the prediction of treatment efficacy in non-small cell lung cancer (NSCLC). We herein used a bioinformatics approach to identify differentially expressed (DE) miRNAs associated with response to platinum-based CT in NSCLC. We identified 6 miRNAs targeting signaling molecules participating in biological pathways involved in cancer and drug resistance. In summary, we developed a 6-miRNA signature that potentially predicts the response to cisplatin in NSCLC and warrants further validation in clinical samples.
铂基化疗(CT)是肺癌的标准治疗方法,但多种化疗耐药机制会影响其疗效。MicroRNAs (miRNAs)是预测非小细胞肺癌(NSCLC)治疗效果的潜在生物标志物。本研究采用生物信息学方法鉴定与非小细胞肺癌铂基CT应答相关的差异表达(DE) mirna。我们发现了6个靶向信号分子的mirna,这些信号分子参与了癌症和耐药的生物学途径。总之,我们开发了一个6-miRNA标记,可以潜在地预测非小细胞肺癌对顺铂的反应,值得在临床样本中进一步验证。
{"title":"MicroRNA Expression Analysis and Biological Pathways in Chemoresistant Non- Small Cell Lung Cancer","authors":"S. Agelaki, Krystallia Gourlia, M. Markaki, C. Papadaki, K. Rounis, E. Vorrias, Elena Prokova, E. Lagoudaki, A. Koutsopoulos, I. Tsamardinos, D. Mavroudis","doi":"10.3390/iecc2021-09206","DOIUrl":"https://doi.org/10.3390/iecc2021-09206","url":null,"abstract":": Platinum-based chemotherapy (CT) is a standard treatment for lung cancer, however a variety of chemoresistance mechanisms can impair its efficacy. MicroRNAs (miRNAs) represent potential biomarkers for the prediction of treatment efficacy in non-small cell lung cancer (NSCLC). We herein used a bioinformatics approach to identify differentially expressed (DE) miRNAs associated with response to platinum-based CT in NSCLC. We identified 6 miRNAs targeting signaling molecules participating in biological pathways involved in cancer and drug resistance. In summary, we developed a 6-miRNA signature that potentially predicts the response to cisplatin in NSCLC and warrants further validation in clinical samples.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90921876","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
ARID1A: The Good, the Bad and the Ugly. ARID1A:好的,坏的和丑陋的。
P. Peinado, Á. Andrades, Juan Sanjuan-Hidalgo, Jeffrey R. Haswell, J. Álvarez-Pérez, F. Slack, Pedro Medina Vico
The chromatin-remodeling complex SWI/SNF is the most mutated remodeler that is currently described in many tumor types. Traditionally, it has been associated with a tumor suppressive role, leading the cellular machinery towards differentiation pathways and DNA repair processes. ARID1A is the most mutated SWI/SNF subunit across all human malignancies. It is also considered as one of the top mutated genes in lung adenocarcinoma (LUAD) and an important driver gene. However, there is a lack of phenotypical studies that confirm the tumor suppressive role of ARID1A in LUAD. We have observed that ARID1A depletion in LUAD cell lines significantly impaired cell viability and promoted apoptosis. At first glance, these results contradicted its initially defined tumor suppressor status and could not be explained by synthetic lethal events involving other SWI/SNF subunits or driver genes. In addition, when we down-regulated ARID1A in a normal lung cell line, we did not see a significant reduction of cell viability, suggesting a tumor context dependency of ARID1A. Moreover, after performing RNA-seq in A549 after ARID1A-knockdown, we observed some up-regulated pathways related with apoptosis and genotoxic stress responses. We found that the depletion of ARID1A enhanced DNA damage in cells and triggered a severe ER stress response that promoted apoptosis. In addition, the protein levels of other subunits of the SWI/SNF complex decreased upon ARID1A, which could explain a decrease of the DNA repair processes. Overall, we conclude that some LUAD cell lines are dependent on ARID1A expression in a tumor-dependent manner. In those contexts, ARID1A loss triggers a DNA damage-induced apoptosis, which could open new therapeutic opportunities.
染色质重塑复合体SWI/SNF是目前在许多肿瘤类型中描述的最易突变的重塑因子。传统上,它与肿瘤抑制作用有关,引导细胞机制走向分化途径和DNA修复过程。ARID1A是所有人类恶性肿瘤中突变最多的SWI/SNF亚基。它也被认为是肺腺癌(LUAD)的顶级突变基因之一和重要的驱动基因。然而,缺乏表型研究证实ARID1A在LUAD中的肿瘤抑制作用。我们观察到在LUAD细胞系中ARID1A缺失会显著损害细胞活力并促进细胞凋亡。乍一看,这些结果与最初定义的肿瘤抑制状态相矛盾,不能用涉及其他SWI/SNF亚基或驱动基因的合成致死事件来解释。此外,当我们下调正常肺细胞系中的ARID1A时,我们没有看到细胞活力的显著降低,这表明ARID1A依赖于肿瘤环境。此外,通过对arid1a敲低后的A549进行rna测序,我们发现了一些与细胞凋亡和基因毒性应激反应相关的上调通路。我们发现ARID1A的缺失会增强细胞中的DNA损伤,并引发严重的内质网应激反应,从而促进细胞凋亡。此外,SWI/SNF复合物的其他亚基的蛋白水平在ARID1A后下降,这可以解释DNA修复过程的减少。总之,我们得出结论,一些LUAD细胞系以肿瘤依赖的方式依赖ARID1A的表达。在这些情况下,ARID1A缺失会引发DNA损伤诱导的细胞凋亡,这可能会开辟新的治疗机会。
{"title":"ARID1A: The Good, the Bad and the Ugly.","authors":"P. Peinado, Á. Andrades, Juan Sanjuan-Hidalgo, Jeffrey R. Haswell, J. Álvarez-Pérez, F. Slack, Pedro Medina Vico","doi":"10.3390/iecc2021-09189","DOIUrl":"https://doi.org/10.3390/iecc2021-09189","url":null,"abstract":"The chromatin-remodeling complex SWI/SNF is the most mutated remodeler that is currently described in many tumor types. Traditionally, it has been associated with a tumor suppressive role, leading the cellular machinery towards differentiation pathways and DNA repair processes. ARID1A is the most mutated SWI/SNF subunit across all human malignancies. It is also considered as one of the top mutated genes in lung adenocarcinoma (LUAD) and an important driver gene. However, there is a lack of phenotypical studies that confirm the tumor suppressive role of ARID1A in LUAD. \u0000We have observed that ARID1A depletion in LUAD cell lines significantly impaired cell viability and promoted apoptosis. At first glance, these results contradicted its initially defined tumor suppressor status and could not be explained by synthetic lethal events involving other SWI/SNF subunits or driver genes. In addition, when we down-regulated ARID1A in a normal lung cell line, we did not see a significant reduction of cell viability, suggesting a tumor context dependency of ARID1A. Moreover, after performing RNA-seq in A549 after ARID1A-knockdown, we observed some up-regulated pathways related with apoptosis and genotoxic stress responses. We found that the depletion of ARID1A enhanced DNA damage in cells and triggered a severe ER stress response that promoted apoptosis. In addition, the protein levels of other subunits of the SWI/SNF complex decreased upon ARID1A, which could explain a decrease of the DNA repair processes. \u0000Overall, we conclude that some LUAD cell lines are dependent on ARID1A expression in a tumor-dependent manner. In those contexts, ARID1A loss triggers a DNA damage-induced apoptosis, which could open new therapeutic opportunities.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73490915","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
Identification of a novel regulator of FANCD2 mediated DNA Repair FANCD2介导的DNA修复新调控因子的鉴定
Wai Yiu Tse, E. Maniati, Jun Wang, M. Lockley, Sarah Martin
{"title":"Identification of a novel regulator of FANCD2 mediated DNA Repair","authors":"Wai Yiu Tse, E. Maniati, Jun Wang, M. Lockley, Sarah Martin","doi":"10.3390/iecc2021-09200","DOIUrl":"https://doi.org/10.3390/iecc2021-09200","url":null,"abstract":"","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74325918","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
5-aminosalicylate–4-thiazolinone hybrid derivatives: A potent modulator of DNA damage response and G2/M cell cycle arrest via ATM/ATR pathway and Cyclin-CDK complex 5-氨基水杨酸- 4-噻唑啉酮杂化衍生物:通过ATM/ATR途径和Cyclin-CDK复合物有效调节DNA损伤反应和G2/M细胞周期阻滞
W. Ramadan, M. Ayad, R. Hamoudi, A. Laham, V. Menon, L. Lozon, H. Abdu-Allah, A. El-Shorbagi, H. Tarazi, R. El-Awady
The last several years have witnessed a tremendous advance in the knowledge of DNA repair and cell cycle mechanisms for the purpose of increasing the treatment efficacy of radiotherapy and DNA damaging agents. Thereby, targeting DNA damage and repair pathways and cell cycle checkpoints become an attractive rationale to optimize treatment strategies through identifying new targets. However, the improved knowledge has increased the complexity of DNA damage response (DDR) and checkpoint pathways which extremely proved challenges in the development of cell cycle and DNA repair targeting drugs. To this end, a novel approach of synthesizing new compounds has been recently introduced which involved accommodating two chemical entities that target several molecules into a single structure. Here we combined 5-aminosalicylic acid and 4-thiazolinone, which both reported to affect DDR and cell cycle progression, in a single structural framework to generate two derivatives named HH32 and HH33. The transcriptomic, in silico, and in vitro analysis has been used to uncover the anti-cancer potential of these two compounds. Both compounds exhibited a high cytotoxic effect against a panel of eight cancer cell lines from different tissue origins and showed a low toxicity profile on normal cells compared to Doxorubicin. The in-silico molecular docking predicts a strong binding of the HH32 and HH33 to cell cycle regulators like CDC2-cyclin B, CDK2-cyclin A complexes, and retinoblastoma. Interestingly, the transcriptomic analysis revealed that DNA double-strand repair and cell cycle are the most affected pathways by HH33 compound. These findings were validated using in vitro models and demonstrated the induction of DNA double-strand breaks and the stimulation of ATM/ATR signaling pathway by HH32 and HH33. In addition to the potent effect of HH compounds on cell cycle progression mediated through upregulation of cyclin-dependent kinase inhibitors and downregulation of G2/M phase cell cycle markers which ultimately arrest the cells at G2/M phase and promote apoptosis. In conclusion, the pleiotropic biological effect of HH32 and HH33 compounds on cancer cells suggests the requirement for assessing their anti-cancer activities in preclinical models which may lead to a new area in the development of potentially therapeutic drugs.
在过去的几年里,为了提高放射治疗和DNA损伤剂的治疗效果,DNA修复和细胞周期机制的知识取得了巨大的进步。因此,靶向DNA损伤和修复途径以及细胞周期检查点成为通过识别新靶点来优化治疗策略的一个有吸引力的理论依据。然而,知识的进步增加了DNA损伤反应(DDR)和检查点途径的复杂性,这给细胞周期和DNA修复靶向药物的开发带来了极大的挑战。为此,最近引入了一种合成新化合物的新方法,该方法涉及容纳两种化学实体,这些化学实体针对几个分子形成一个单一结构。在这里,我们将5-氨基水杨酸和4-噻唑啉酮结合在一个单一的结构框架中,生成了两个名为HH32和HH33的衍生物,这两个衍生物都影响DDR和细胞周期进程。转录组学、硅和体外分析已被用来揭示这两种化合物的抗癌潜力。与阿霉素相比,这两种化合物对来自不同组织来源的八种癌细胞系显示出高细胞毒性作用,对正常细胞显示出低毒性。硅分子对接预测HH32和HH33与细胞周期调节因子如CDC2-cyclin B、CDK2-cyclin a复合物和视网膜母细胞瘤的强结合。有趣的是,转录组学分析显示,DNA双链修复和细胞周期是受HH33化合物影响最大的途径。这些发现在体外模型中得到了验证,并证实了HH32和HH33诱导DNA双链断裂和刺激ATM/ATR信号通路。除了HH化合物通过上调周期蛋白依赖性激酶抑制剂和下调G2/M期细胞周期标记物介导的细胞周期进程的强大作用外,G2/M期细胞周期标记物最终使细胞停滞在G2/M期并促进凋亡。综上所述,HH32和HH33化合物对肿瘤细胞的多效生物学作用提示了在临床前模型中评估其抗癌活性的必要性,这可能为潜在治疗药物的开发开辟一个新的领域。
{"title":"5-aminosalicylate–4-thiazolinone hybrid derivatives: A potent modulator of DNA damage response and G2/M cell cycle arrest via ATM/ATR pathway and Cyclin-CDK complex","authors":"W. Ramadan, M. Ayad, R. Hamoudi, A. Laham, V. Menon, L. Lozon, H. Abdu-Allah, A. El-Shorbagi, H. Tarazi, R. El-Awady","doi":"10.3390/iecc2021-09188","DOIUrl":"https://doi.org/10.3390/iecc2021-09188","url":null,"abstract":"The last several years have witnessed a tremendous advance in the knowledge of DNA repair and cell cycle mechanisms for the purpose of increasing the treatment efficacy of radiotherapy and DNA damaging agents. Thereby, targeting DNA damage and repair pathways and cell cycle checkpoints become an attractive rationale to optimize treatment strategies through identifying new targets. However, the improved knowledge has increased the complexity of DNA damage response (DDR) and checkpoint pathways which extremely proved challenges in the development of cell cycle and DNA repair targeting drugs. To this end, a novel approach of synthesizing new compounds has been recently introduced which involved accommodating two chemical entities that target several molecules into a single structure. Here we combined 5-aminosalicylic acid and 4-thiazolinone, which both reported to affect DDR and cell cycle progression, in a single structural framework to generate two derivatives named HH32 and HH33. The transcriptomic, in silico, and in vitro analysis has been used to uncover the anti-cancer potential of these two compounds. Both compounds exhibited a high cytotoxic effect against a panel of eight cancer cell lines from different tissue origins and showed a low toxicity profile on normal cells compared to Doxorubicin. The in-silico molecular docking predicts a strong binding of the HH32 and HH33 to cell cycle regulators like CDC2-cyclin B, CDK2-cyclin A complexes, and retinoblastoma. Interestingly, the transcriptomic analysis revealed that DNA double-strand repair and cell cycle are the most affected pathways by HH33 compound. These findings were validated using in vitro models and demonstrated the induction of DNA double-strand breaks and the stimulation of ATM/ATR signaling pathway by HH32 and HH33. In addition to the potent effect of HH compounds on cell cycle progression mediated through upregulation of cyclin-dependent kinase inhibitors and downregulation of G2/M phase cell cycle markers which ultimately arrest the cells at G2/M phase and promote apoptosis. In conclusion, the pleiotropic biological effect of HH32 and HH33 compounds on cancer cells suggests the requirement for assessing their anti-cancer activities in preclinical models which may lead to a new area in the development of potentially therapeutic drugs.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"84878403","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
Developing a multidisciplinary strategy to interpret the impact of missense mutations in XPA on NER activity and cisplatin sensitivity 发展多学科策略来解释XPA错义突变对NER活性和顺铂敏感性的影响
Alexandra M. Blee, Bian Li, J. Capra, W. Chazin
Nucleotide excision repair (NER) is an essential DNA damage repair pathway that removes bulky DNA lesions formed by exposure to ultraviolet light, environmental toxins, and platinum (Pt)-based chemotherapeutic drugs that are a standard of care for many cancer types. Mutation or decreased NER gene expression in cancer correlates with improved patient survival after Pt-based chemotherapy. However, the impact of most missense mutations in NER genes is unknown, and few approaches exist to reliably identify nonrecurrent passenger mutations with functional consequences. In this study, a multidisciplinary strategy will be developed to predict, validate, and characterize NER-defective mutations in the essential NER scaffold protein Xeroderma Pigmentosum Complementation Group A (XPA). Computational analyses were used to score NER-deficient versus NER-proficient mutations for further study. Predicted NER-deficient XPA mutants are being expressed in human XPA-deficient cells and screened for both NER activity and cisplatin sensitivity. In-depth biophysical and structural studies are being implemented to elucidate mechanisms of dysfunction. Identifying NER-deficient mutations that may sensitize tumors to Pt-based chemotherapies represents a promising strategy to stratify patients for optimal treatment strategies.
核苷酸切除修复(NER)是一种重要的DNA损伤修复途径,可去除因暴露于紫外线、环境毒素和铂类化疗药物而形成的大块DNA损伤,铂类化疗药物是许多癌症类型的标准治疗方法。肿瘤中NER基因表达的突变或降低与基于pt的化疗后患者生存率的提高相关。然而,大多数误义突变对NER基因的影响是未知的,并且很少有方法可以可靠地识别具有功能后果的非复发性旅客突变。在这项研究中,将开发一种多学科策略来预测、验证和表征必要的NER支架蛋白干皮色素补体组a (XPA)中的NER缺陷突变。计算分析用于对ner缺陷突变和ner精通突变进行评分,以供进一步研究。预测的NER缺陷XPA突变体在人类XPA缺陷细胞中表达,并筛选NER活性和顺铂敏感性。深入的生物物理和结构研究正在实施,以阐明功能障碍的机制。识别可能使肿瘤对基于pt的化疗敏感的ner缺陷突变代表了一种有希望的策略,可以对患者进行分层,以获得最佳治疗策略。
{"title":"Developing a multidisciplinary strategy to interpret the impact of missense mutations in XPA on NER activity and cisplatin sensitivity","authors":"Alexandra M. Blee, Bian Li, J. Capra, W. Chazin","doi":"10.3390/iecc2021-09192","DOIUrl":"https://doi.org/10.3390/iecc2021-09192","url":null,"abstract":"Nucleotide excision repair (NER) is an essential DNA damage repair pathway that removes bulky DNA lesions formed by exposure to ultraviolet light, environmental toxins, and platinum (Pt)-based chemotherapeutic drugs that are a standard of care for many cancer types. Mutation or decreased NER gene expression in cancer correlates with improved patient survival after Pt-based chemotherapy. However, the impact of most missense mutations in NER genes is unknown, and few approaches exist to reliably identify nonrecurrent passenger mutations with functional consequences. In this study, a multidisciplinary strategy will be developed to predict, validate, and characterize NER-defective mutations in the essential NER scaffold protein Xeroderma Pigmentosum Complementation Group A (XPA). Computational analyses were used to score NER-deficient versus NER-proficient mutations for further study. Predicted NER-deficient XPA mutants are being expressed in human XPA-deficient cells and screened for both NER activity and cisplatin sensitivity. In-depth biophysical and structural studies are being implemented to elucidate mechanisms of dysfunction. Identifying NER-deficient mutations that may sensitize tumors to Pt-based chemotherapies represents a promising strategy to stratify patients for optimal treatment strategies.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89512555","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
Excessive new origin firing underlies selective glioma stem cell cytotoxicity induced by replication stress response inhibition 过度的新起源放电是由复制应激反应抑制诱导的选择性胶质瘤干细胞细胞毒性的基础
Emily Clough, K. Strathdee, R. Carruthers
Glioblastoma (GBM) is a treatment refractory cancer of extreme unmet need which exhibits treatment resistance due to a subpopulation of GBM cancer stem cells which have constitutive DNA damage response activation driven by elevated replication stress (RS). RS response inhibition is potently cytotoxic to GSC, however mechanistic understanding will be key to biomarker discovery and successful clinical translation. We investigated response to combined ATR and PARP inhibition (CAiPi) to gain mechanistic insight and inform biomarker development. A panel of patient-derived GBM cell lines were cultured as stem enriched (GSCs) or stem depleted (bulk), to characterise response to combined ATR inhibition (VE821 5μM) and PARP inhibition (Olaparib 1μM), by CellTiter-Glo viability assay. Mechanistic investigations included immunofluorescence of 53BP1 nuclear bodies and DNA fibre analysis. Studies into the importance of PARP trapping included another PARPi Veliparib (1μM), and investigations into inhibition of origin firing used the CDK inhibitor Roscovitine. Responses to CAiPi in a panel of primary paired GBM GSCs vs differentiated progeny were heterogenous. CAiPi is selectively GSC cytotoxic in a subpopulation of tumours. DNA fibre analysis identified increased new origin firing with PARPi, which was correlated with increased PARP trapping. Inhibition of origin firing by exposure to roscovitine rescued the CAiPi cytotoxic phenotype, suggesting origin firing has an important role in selective GSC cytotoxicity. A population of treatment-sensitive GSCs with increased numbers of 53BP1 nuclear bodies in G1 phase with CAiPi were identified, indicative of under-replication of DNA in S phase. Selective GSC cytotoxicity is induced by CAiPi via dysregulation of replication, by both DNA under-replication resulting in DNA lesions, and the novel finding of increased new origin firing in GSC due to PARPi.
胶质母细胞瘤(GBM)是一种治疗难治性癌症,其治疗需求极度未满足,由于GBM癌症干细胞亚群具有由升高的复制应激(RS)驱动的组成性DNA损伤反应激活,因此表现出治疗耐药性。RS反应抑制对GSC具有强大的细胞毒性,但机制的理解将是生物标志物发现和成功临床翻译的关键。我们研究了对ATR和PARP联合抑制(CAiPi)的反应,以获得机制见解并为生物标志物的开发提供信息。通过CellTiter-Glo活性测定,将患者来源的GBM细胞系培养为干细胞富集(GSCs)或干细胞缺失(散装),以表征对ATR (VE821 5μM)和PARP (Olaparib 1μM)联合抑制的反应。机制研究包括53BP1核体的免疫荧光和DNA纤维分析。对PARP捕获重要性的研究包括另一种PARPi Veliparib (1μM),以及使用CDK抑制剂Roscovitine抑制起源激发的研究。在一组配对的原发性GBM GSCs与分化后代中,对CAiPi的反应是异质性的。在肿瘤亚群中,CAiPi具有选择性的GSC细胞毒性。DNA纤维分析发现,PARPi增加了新起源发射,这与PARP捕获增加有关。暴露于罗斯科维汀抑制源放电恢复了CAiPi的细胞毒性表型,表明源放电在选择性GSC细胞毒性中起重要作用。在G1期发现了53BP1核体数量增加的治疗敏感GSCs群体,这表明在S期DNA复制不足。选择性GSC细胞毒性是由CAiPi通过复制失调,DNA复制不足导致DNA损伤,以及新发现的PARPi在GSC中增加的新起源放电引起的。
{"title":"Excessive new origin firing underlies selective glioma stem cell cytotoxicity induced by replication stress response inhibition","authors":"Emily Clough, K. Strathdee, R. Carruthers","doi":"10.3390/iecc2021-09187","DOIUrl":"https://doi.org/10.3390/iecc2021-09187","url":null,"abstract":"\u0000 \u0000 \u0000 Glioblastoma (GBM) is a treatment refractory cancer of extreme unmet need which exhibits treatment resistance due to a subpopulation of GBM cancer stem cells which have constitutive DNA damage response activation driven by elevated replication stress (RS). RS response inhibition is potently cytotoxic to GSC, however mechanistic understanding will be key to biomarker discovery and successful clinical translation.\u0000 We investigated response to combined ATR and PARP inhibition (CAiPi) to gain mechanistic insight and inform biomarker development.\u0000 \u0000 \u0000 \u0000 A panel of patient-derived GBM cell lines were cultured as stem enriched (GSCs) or stem depleted (bulk), to characterise response to combined ATR inhibition (VE821 5μM) and PARP inhibition (Olaparib 1μM), by CellTiter-Glo viability assay.\u0000 Mechanistic investigations included immunofluorescence of 53BP1 nuclear bodies and DNA fibre analysis. Studies into the importance of PARP trapping included another PARPi Veliparib (1μM), and investigations into inhibition of origin firing used the CDK inhibitor Roscovitine.\u0000 \u0000 \u0000 \u0000 Responses to CAiPi in a panel of primary paired GBM GSCs vs differentiated progeny were heterogenous. CAiPi is selectively GSC cytotoxic in a subpopulation of tumours. DNA fibre analysis identified increased new origin firing with PARPi, which was correlated with increased PARP trapping. Inhibition of origin firing by exposure to roscovitine rescued the CAiPi cytotoxic phenotype, suggesting origin firing has an important role in selective GSC cytotoxicity.\u0000 A population of treatment-sensitive GSCs with increased numbers of 53BP1 nuclear bodies in G1 phase with CAiPi were identified, indicative of under-replication of DNA in S phase.\u0000 \u0000 \u0000 \u0000 Selective GSC cytotoxicity is induced by CAiPi via dysregulation of replication, by both DNA under-replication resulting in DNA lesions, and the novel finding of increased new origin firing in GSC due to PARPi.\u0000 \u0000 \u0000 \u0000","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76398579","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
Role of Mitotic slippage in cancer resistance and DNA damage response in the MDA-MB-231-DOX-Treated Cells mda - mb -231- dox处理细胞中有丝分裂滑移在癌症抵抗和DNA损伤反应中的作用
K. Salmina, Felikss Rumnieks, N. Vainshelbaum, D. Pjanova, J. Erenpreisa
{"title":"Role of Mitotic slippage in cancer resistance and DNA damage response in the MDA-MB-231-DOX-Treated Cells","authors":"K. Salmina, Felikss Rumnieks, N. Vainshelbaum, D. Pjanova, J. Erenpreisa","doi":"10.3390/iecc2021-09214","DOIUrl":"https://doi.org/10.3390/iecc2021-09214","url":null,"abstract":"","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86066644","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
Loss of MLH1 regulates a metabolic phenotype in endometrial cancer MLH1的缺失调节子宫内膜癌的代谢表型
Amy Gibson, V. Morales, K. Bianchi, E. Crosbie, G. Ficz, Sarah Martin
{"title":"Loss of MLH1 regulates a metabolic phenotype in endometrial cancer","authors":"Amy Gibson, V. Morales, K. Bianchi, E. Crosbie, G. Ficz, Sarah Martin","doi":"10.3390/iecc2021-09196","DOIUrl":"https://doi.org/10.3390/iecc2021-09196","url":null,"abstract":"","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91137620","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
Development of small molecule NUDT22 inhibitors for uses in cancer. 用于癌症治疗的小分子NUDT22抑制剂的开发。
M. Walter, E. Homan, Tobias Koolmeister, I. Almlöf, T. Helleday, P. Herr
Here we present the characterisation of the so far unstudied NUDIX hydrolase family member NUDT22. We previously identified a unique hydrolase activity of NUDT22 towards UDP-glucose from a family-wide biochemical substrate screen. UDP-glucose hydrolysis results in the production of uridine monophosphate (UMP) and glucose 1-phosphate (G-1-P). We furthermore solved the first crystal structure of NUDT22 in complex with its substrate UDP-glucose [1]. Our mechanistic studies reveal increased replication stress in NUDT22 deficient cells which can be rescued by nucleoside supplementation. We therefore propose the discovery of a novel NUDT22-mediated pyrimidine salvage pathway.Increased replication rates resulting in replication stress is a hallmark of cancer cells and NUDT22 gene expression alterations are present in several cancer tissues, which makes it an interesting new target for the development of small molecule inhibitors for uses in cancer.We employed our NUDT22 crystal structure to perform an in silico docking screen on available small molecule libraries to identify starting points for the development of first-in-class NUDT22 inhibitors. Chemically optimised NUDT22 inhibitors are currently being validated in biochemical assays, cellular target engagement assays, and their cellular activity is being assessed in vitro. [1] M. Carter et al., "Human NUDT22 Is a UDP-Glucose/Galactose Hydrolase Exhibiting a Unique Structural Fold," Structure, vol. 26, no. 2, pp. 295-+, Feb 2018, doi: 10.1016/j.str.2018.01.004.
在这里,我们介绍了迄今为止尚未研究的NUDIX水解酶家族成员NUDT22的特征。我们之前从全家族生化底物筛选中鉴定出NUDT22对udp -葡萄糖的独特水解酶活性。udp -葡萄糖水解产生尿苷单磷酸(UMP)和葡萄糖1-磷酸(G-1-P)。我们进一步解决了NUDT22与底物udp -葡萄糖[1]配合物的第一个晶体结构。我们的机制研究表明,NUDT22缺陷细胞的复制应激增加,可以通过补充核苷来挽救。因此,我们提出发现一种新的nudt22介导的嘧啶回收途径。增加的复制速率导致复制应激是癌细胞的一个标志,NUDT22基因表达改变存在于几种癌症组织中,这使得它成为开发用于癌症的小分子抑制剂的一个有趣的新靶点。我们利用我们的NUDT22晶体结构对可用的小分子文库进行硅对接筛选,以确定开发一流NUDT22抑制剂的起点。化学优化的NUDT22抑制剂目前正在生化分析、细胞靶标接合分析中进行验证,并且正在体外评估其细胞活性。[10] M. Carter等人,“人类NUDT22是一种呈现独特结构折叠的udp -葡萄糖/半乳糖水解酶”,《结构》,第26卷,第2期。2, pp. 295-+, 2018年2月,doi: 10.1016/j.s r.2018.01.004。
{"title":"Development of small molecule NUDT22 inhibitors for uses in cancer.","authors":"M. Walter, E. Homan, Tobias Koolmeister, I. Almlöf, T. Helleday, P. Herr","doi":"10.3390/iecc2021-09197","DOIUrl":"https://doi.org/10.3390/iecc2021-09197","url":null,"abstract":"Here we present the characterisation of the so far unstudied NUDIX hydrolase family member NUDT22. We previously identified a unique hydrolase activity of NUDT22 towards UDP-glucose from a family-wide biochemical substrate screen. UDP-glucose hydrolysis results in the production of uridine monophosphate (UMP) and glucose 1-phosphate (G-1-P). We furthermore solved the first crystal structure of NUDT22 in complex with its substrate UDP-glucose [1]. Our mechanistic studies reveal increased replication stress in NUDT22 deficient cells which can be rescued by nucleoside supplementation. We therefore propose the discovery of a novel NUDT22-mediated pyrimidine salvage pathway.Increased replication rates resulting in replication stress is a hallmark of cancer cells and NUDT22 gene expression alterations are present in several cancer tissues, which makes it an interesting new target for the development of small molecule inhibitors for uses in cancer.We employed our NUDT22 crystal structure to perform an in silico docking screen on available small molecule libraries to identify starting points for the development of first-in-class NUDT22 inhibitors. Chemically optimised NUDT22 inhibitors are currently being validated in biochemical assays, cellular target engagement assays, and their cellular activity is being assessed in vitro. \u0000[1] M. Carter et al., \"Human NUDT22 Is a UDP-Glucose/Galactose Hydrolase Exhibiting a Unique Structural Fold,\" Structure, vol. 26, no. 2, pp. 295-+, Feb 2018, doi: 10.1016/j.str.2018.01.004.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82592440","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
Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response
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