首页 > 最新文献

Oncoscience最新文献

英文 中文
Correction: Lactate dehydrogenase A inhibition by small molecular entities: steps in the right direction. 更正:乳酸脱氢酶A的小分子实体抑制:在正确的方向上的步骤。
Pub Date : 2021-03-30 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.529
Btissame El Hassouni, Marika Franczak, Mjriam Capula, Christian M Vonk, Valentina M Gomez, Ryszard T Smolenski, Carlotta Granchi, Godefridus J Peters, Filippo Minutolo, Elisa Giovannetti

[This corrects the article DOI: 10.18632/oncoscience.519.].

[这更正了文章DOI: 10.18632/ onscience .519]。
{"title":"Correction: Lactate dehydrogenase A inhibition by small molecular entities: steps in the right direction.","authors":"Btissame El Hassouni,&nbsp;Marika Franczak,&nbsp;Mjriam Capula,&nbsp;Christian M Vonk,&nbsp;Valentina M Gomez,&nbsp;Ryszard T Smolenski,&nbsp;Carlotta Granchi,&nbsp;Godefridus J Peters,&nbsp;Filippo Minutolo,&nbsp;Elisa Giovannetti","doi":"10.18632/oncoscience.529","DOIUrl":"https://doi.org/10.18632/oncoscience.529","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.18632/oncoscience.519.].</p>","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8051325/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38897114","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
The helix-loop-helix transcriptional regulator Id4 is required for terminal differentiation of luminal epithelial cells in the prostate. 前列腺管腔上皮细胞的终极分化需要螺旋环螺旋转录调节因子Id4。
Pub Date : 2021-03-24 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.524
Dhanushka Hewa Bostanthirige, Shravan K Komaragiri, Jugal B Joshi, Majid Alzahrani, Isha Saini, Sanjay Jain, Nathan J Bowen, Matthew C Havrda, Jaideep Chaudhary

Inhibitor of differentiation 4 (Id4), a member of the helix-loop-helix family of transcriptional regulators has emerged as a tumor suppressor in prostate cancer. In this study we investigated the effect of loss of Id4 (Id4-/-) on mouse prostate development. Histological analysis was performed on prostates from 25 days, 3 months and 6 months old Id4-/- mice. Expression of Amacr, Ck8, Ck18, Fkbp51, Fkbp52, androgen receptor, Pten, sca-1 and Nkx3.1 was investigated by immunohistochemistry. Results were compared to the prostates from Nkx3.1-/- mice. Id4-/- mice had smaller prostates with fewer and smaller tubules. Subtle PIN like lesions were observed at 6mo. Decreased Nkx3.1 and Pten and increased stem cell marker sca-1, PIN marker Amacr and basal cell marker p63 was observed at all ages. Persistent Ck8 and Ck18 expression suggested that loss of Id4 results in epithelial commitment but not terminal differentiation in spite of active Ar. Loss of Id4 attenuates normal prostate development and promotes hyperplasia/ dysplasia with PIN like lesions. The results suggest that loss of Id4 maintains stem cell phenotype of "luminal committed basal cells", identifying a unique prostate developmental pathway regulated by Id4.

分化抑制因子 4(Id4)是转录调控因子螺旋环螺旋家族的成员,已成为前列腺癌的肿瘤抑制因子。在这项研究中,我们研究了Id4(Id4-/-)缺失对小鼠前列腺发育的影响。我们对 25 天、3 个月和 6 个月大的 Id4-/- 小鼠的前列腺进行了组织学分析。免疫组化法检测了Amacr、Ck8、Ck18、Fkbp51、Fkbp52、雄激素受体、Pten、sca-1和Nkx3.1的表达。结果与 Nkx3.1-/- 小鼠的前列腺进行了比较。Id4-/-小鼠的前列腺较小,肾小管较少且较小。6个月时观察到类似PIN的细微病变。在所有年龄段都观察到 Nkx3.1 和 Pten 减少,干细胞标记物 sca-1、PIN 标记物 Amacr 和基底细胞标记物 p63 增加。Ck8 和 Ck18 的持续表达表明,尽管 Ar 活跃,但 Id4 的缺失会导致上皮承诺,而不是终极分化。Id4的缺失会削弱前列腺的正常发育,促进前列腺增生/发育不良,出现类似PIN的病变。结果表明,Id4缺失可维持 "管腔承诺基底细胞 "的干细胞表型,从而确定了由Id4调控的独特前列腺发育途径。
{"title":"The helix-loop-helix transcriptional regulator Id4 is required for terminal differentiation of luminal epithelial cells in the prostate.","authors":"Dhanushka Hewa Bostanthirige, Shravan K Komaragiri, Jugal B Joshi, Majid Alzahrani, Isha Saini, Sanjay Jain, Nathan J Bowen, Matthew C Havrda, Jaideep Chaudhary","doi":"10.18632/oncoscience.524","DOIUrl":"10.18632/oncoscience.524","url":null,"abstract":"<p><p>Inhibitor of differentiation 4 (Id4), a member of the helix-loop-helix family of transcriptional regulators has emerged as a tumor suppressor in prostate cancer. In this study we investigated the effect of loss of Id4 (<i>Id4-/-</i>) on mouse prostate development. Histological analysis was performed on prostates from 25 days, 3 months and 6 months old <i>Id4-/-</i> mice. Expression of Amacr, Ck8, Ck18, Fkbp51, Fkbp52, androgen receptor, Pten, sca-1 and Nkx3.1 was investigated by immunohistochemistry. Results were compared to the prostates from <i>Nkx3.1-/-</i> mice. <i>Id4-/-</i> mice had smaller prostates with fewer and smaller tubules. Subtle PIN like lesions were observed at 6mo. Decreased Nkx3.1 and Pten and increased stem cell marker sca-1, PIN marker Amacr and basal cell marker p63 was observed at all ages. Persistent Ck8 and Ck18 expression suggested that loss of Id4 results in epithelial commitment but not terminal differentiation in spite of active Ar. Loss of Id4 attenuates normal prostate development and promotes hyperplasia/ dysplasia with PIN like lesions. The results suggest that loss of Id4 maintains stem cell phenotype of \"luminal committed basal cells\", identifying a unique prostate developmental pathway regulated by Id4.</p>","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8045964/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38897109","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Balancing efficacy and quality of life measurements among metastatic renal cell carcinoma (RCC) studies. 转移性肾细胞癌(RCC)研究中平衡疗效和生活质量测量。
Pub Date : 2021-03-21 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.528
Jeanny B Aragon-Ching

Metastatic renal cell carcinoma (mRCC) treatments have rapidly evolved in the last few years. While vascular endothelial growth factor (VEGF) inhibition had previously been the mainstay of treatment for first-line advanced RCC therapy in the past decade, it has now rapidly changed into combination checkpoint inhibitors with or without VEGF TKIs, although there remains a role for VEGF tyrosine kinase inhibitor monotherapy for patients with favorable-risk disease and for those with intermediate and poor-risk disease with the use of cabozantinib. Perspectives on the Quality-adjusted survival Time without Symptoms of disease or Toxicity (Q-TWiST) analysis for the CABOSUN trial, as well as different aspects of efficacy regarding different first-line therapy for advanced or metastatic RCC are discussed herein.

转移性肾细胞癌(mRCC)的治疗方法在过去几年中迅速发展。虽然血管内皮生长因子(VEGF)抑制在过去十年中一直是一线晚期RCC治疗的主要治疗方法,但现在它已迅速转变为联合检查点抑制剂与VEGF TKIs,尽管VEGF酪氨酸激酶抑制剂单药治疗对有利风险疾病患者和使用cabozantinib的中、低风险疾病患者仍有作用。本文讨论了CABOSUN试验的无疾病症状或毒性的质量调整生存时间(Q-TWiST)分析的观点,以及不同一线治疗对晚期或转移性RCC疗效的不同方面。
{"title":"Balancing efficacy and quality of life measurements among metastatic renal cell carcinoma (RCC) studies.","authors":"Jeanny B Aragon-Ching","doi":"10.18632/oncoscience.528","DOIUrl":"https://doi.org/10.18632/oncoscience.528","url":null,"abstract":"<p><p>Metastatic renal cell carcinoma (mRCC) treatments have rapidly evolved in the last few years. While vascular endothelial growth factor (VEGF) inhibition had previously been the mainstay of treatment for first-line advanced RCC therapy in the past decade, it has now rapidly changed into combination checkpoint inhibitors with or without VEGF TKIs, although there remains a role for VEGF tyrosine kinase inhibitor monotherapy for patients with favorable-risk disease and for those with intermediate and poor-risk disease with the use of cabozantinib. Perspectives on the Quality-adjusted survival Time without Symptoms of disease or Toxicity (Q-TWiST) analysis for the CABOSUN trial, as well as different aspects of efficacy regarding different first-line therapy for advanced or metastatic RCC are discussed herein.</p>","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8045961/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38897113","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Upper gastrointestinal cancers in Lynch syndrome: the time for surveillance is now. Lynch综合征的上消化道癌症:现在是监测的时候了。
Pub Date : 2021-03-21 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.525
Shria Kumar, Bryson W Katona
{"title":"Upper gastrointestinal cancers in Lynch syndrome: the time for surveillance is now.","authors":"Shria Kumar,&nbsp;Bryson W Katona","doi":"10.18632/oncoscience.525","DOIUrl":"https://doi.org/10.18632/oncoscience.525","url":null,"abstract":"","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8045978/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38897110","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Causation of increased prostate cancer in young men. 年轻男性前列腺癌发病率增加的原因。
Pub Date : 2021-03-20 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.527
Archie Bleyer, Filippo Spreafico, Ronald Barr
{"title":"Causation of increased prostate cancer in young men.","authors":"Archie Bleyer, Filippo Spreafico, Ronald Barr","doi":"10.18632/oncoscience.527","DOIUrl":"10.18632/oncoscience.527","url":null,"abstract":"","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8045966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38897112","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AKT as a therapeutic target for autophagy induction and cancer therapy. AKT作为自噬诱导和癌症治疗的治疗靶点。
Pub Date : 2021-03-19 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.526
Qi Wu, Guido Kroemer, Oliver Kepp
Qi Wu1,2,3, Yi Tu1, Guido Kroemer2,3,4,5 and Oliver Kepp2,3 1 Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China 2 Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France 3 Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France 4 Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China 5 Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France 6 Karolinska Institutet, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
{"title":"AKT as a therapeutic target for autophagy induction and cancer therapy.","authors":"Qi Wu,&nbsp;Guido Kroemer,&nbsp;Oliver Kepp","doi":"10.18632/oncoscience.526","DOIUrl":"https://doi.org/10.18632/oncoscience.526","url":null,"abstract":"Qi Wu1,2,3, Yi Tu1, Guido Kroemer2,3,4,5 and Oliver Kepp2,3 1 Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China 2 Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France 3 Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France 4 Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China 5 Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France 6 Karolinska Institutet, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8045973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38897111","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Monitoring vascular normalization: new opportunities for mitochondrial inhibitors in breast cancer. 监测血管正常化:线粒体抑制剂在乳腺癌中的新机会。
Pub Date : 2021-02-25 eCollection Date: 2021-01-01 DOI: 10.18632/oncoscience.523
Silvana Mouron, Maria J Bueno, Manuel Muñoz, Miguel Quintela-Fandino

Preclinical evidence indicates the potential of targeting mitochondrial respiration as a therapeutic strategy. We previously demonstrated that mitochondrial inhibitors' efficacy was restricted to a metabolic context in which mitochondrial respiration was the predominant energy source, a situation achievable by inducing vascular normalization/hypoxia correction with antiangiogenics. Using molecular imaging, we showed how the same antiangiogenic agent may display different normalizing properties in patients with the same tumor type. This is of key importance, since patients experiencing normalization seem to get more benefit from standard chemotherapy combinations, and also could be eligible for combination with antimitochondrial agents. This scenario emphasizes the need for monitoring vascular normalization in order to optimize the use of antiangiogenics. We have also proposed a method to evaluate anti-mitochondrial agents' pharmacodynamics; despite promising accuracy in animal studies the clinical results were inconclusive, highlighting the need for research in this field. Regarding patients that respond to antiangiogenics increasing vessel abnormality, in this case an immunosuppressive tumor microenvironment is generated. Whether anti-mitochondrial agents can positively modulate the activity of T effector cell subpopulations remains an area of active research. Our research sheds light on the importance of refining the use of antiangiogenics, highlighting the relevance of tracing vascular normalization as a potential biomarker for antiangiogenics to assist patient-tailored medicine and exploring the role of mitochondrial inhibitors in the context of vascular normalization and correction of hypoxia.

临床前证据表明靶向线粒体呼吸作为一种治疗策略的潜力。我们之前已经证明,线粒体抑制剂的功效仅限于线粒体呼吸是主要能量来源的代谢环境,这种情况可以通过诱导血管正常化/抗血管生成的缺氧纠正来实现。利用分子成像,我们展示了相同的抗血管生成药物如何在相同肿瘤类型的患者中显示不同的正常化特性。这是至关重要的,因为经历正常化的患者似乎从标准化疗组合中获益更多,并且也有资格与抗线粒体药物联合使用。这种情况强调了监测血管正常化的必要性,以便优化抗血管生成药物的使用。我们还提出了一种评估抗线粒体药物药效学的方法;尽管在动物研究中有希望的准确性,但临床结果是不确定的,突出了该领域研究的必要性。对于抗血管生成药物反应增加血管异常的患者,在这种情况下会产生免疫抑制的肿瘤微环境。抗线粒体药物是否能积极调节T效应细胞亚群的活性仍然是一个活跃的研究领域。我们的研究揭示了细化抗血管生成使用的重要性,强调了追踪血管正常化作为抗血管生成的潜在生物标志物的相关性,以协助患者量身定制药物,并探索线粒体抑制剂在血管正常化和缺氧纠正中的作用。
{"title":"Monitoring vascular normalization: new opportunities for mitochondrial inhibitors in breast cancer.","authors":"Silvana Mouron,&nbsp;Maria J Bueno,&nbsp;Manuel Muñoz,&nbsp;Miguel Quintela-Fandino","doi":"10.18632/oncoscience.523","DOIUrl":"https://doi.org/10.18632/oncoscience.523","url":null,"abstract":"<p><p>Preclinical evidence indicates the potential of targeting mitochondrial respiration as a therapeutic strategy. We previously demonstrated that mitochondrial inhibitors' efficacy was restricted to a metabolic context in which mitochondrial respiration was the predominant energy source, a situation achievable by inducing vascular normalization/hypoxia correction with antiangiogenics. Using molecular imaging, we showed how the same antiangiogenic agent may display different normalizing properties in patients with the same tumor type. This is of key importance, since patients experiencing normalization seem to get more benefit from standard chemotherapy combinations, and also could be eligible for combination with antimitochondrial agents. This scenario emphasizes the need for monitoring vascular normalization in order to optimize the use of antiangiogenics. We have also proposed a method to evaluate anti-mitochondrial agents' pharmacodynamics; despite promising accuracy in animal studies the clinical results were inconclusive, highlighting the need for research in this field. Regarding patients that respond to antiangiogenics increasing vessel abnormality, in this case an immunosuppressive tumor microenvironment is generated. Whether anti-mitochondrial agents can positively modulate the activity of T effector cell subpopulations remains an area of active research. Our research sheds light on the importance of refining the use of antiangiogenics, highlighting the relevance of tracing vascular normalization as a potential biomarker for antiangiogenics to assist patient-tailored medicine and exploring the role of mitochondrial inhibitors in the context of vascular normalization and correction of hypoxia.</p>","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8018703/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38885688","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Uncovering the deubiquitinase activity landscape of breast cancer. 揭示乳腺癌去泛素酶活性格局。
Pub Date : 2020-11-09 eCollection Date: 2020-11-01 DOI: 10.18632/oncoscience.518
Sijia Liu, Peter Ten Dijke

Breast cancer is a highly heterogeneous disease with dynamic changes in the tumor microenvironment. Precision medicine will in the future provide the possibility to treat each individual cancer patient with the right (combination) therapy specifically tailored to personal needs. However, in order to accomplish this, more accurate biomarkers for precise diagnosis, prognosis, therapy response, and target-specific drugs are required. Although an increasing number of (epi)genetic driving alterations have been reported in breast cancer, the major stumbling block for clinical application of many of them is that they are difficult to therapeutically target. Deubiquitinases (DUBs) are emerging drug targets that play important roles in cancer progression. Hence, we devoted our efforts to uncover the global DUB activity landscape of breast cancer in order to discover potential novel biomarkers or therapeutic targets. We developed a specific DUB activity-based inhibitor and probe and applied it to obtain new insights into breast cancer.

乳腺癌是一种高度异质性的疾病,肿瘤微环境发生动态变化。精准医疗将在未来为每个癌症患者提供针对个人需求量身定制的正确(联合)疗法。然而,为了实现这一目标,需要更准确的生物标志物来精确诊断、预后、治疗反应和靶向药物。尽管越来越多的(epi)基因驱动改变在乳腺癌中被报道,但其中许多基因驱动改变的临床应用的主要障碍是它们难以靶向治疗。去泛素酶(DUBs)是新兴的药物靶点,在癌症进展中起重要作用。因此,我们致力于揭示乳腺癌的全球DUB活性格局,以发现潜在的新型生物标志物或治疗靶点。我们开发了一种特定的基于DUB活性的抑制剂和探针,并将其应用于乳腺癌的研究中。
{"title":"Uncovering the deubiquitinase activity landscape of breast cancer.","authors":"Sijia Liu,&nbsp;Peter Ten Dijke","doi":"10.18632/oncoscience.518","DOIUrl":"https://doi.org/10.18632/oncoscience.518","url":null,"abstract":"<p><p>Breast cancer is a highly heterogeneous disease with dynamic changes in the tumor microenvironment. Precision medicine will in the future provide the possibility to treat each individual cancer patient with the right (combination) therapy specifically tailored to personal needs. However, in order to accomplish this, more accurate biomarkers for precise diagnosis, prognosis, therapy response, and target-specific drugs are required. Although an increasing number of (epi)genetic driving alterations have been reported in breast cancer, the major stumbling block for clinical application of many of them is that they are difficult to therapeutically target. Deubiquitinases (DUBs) are emerging drug targets that play important roles in cancer progression. Hence, we devoted our efforts to uncover the global DUB activity landscape of breast cancer in order to discover potential novel biomarkers or therapeutic targets. We developed a specific DUB activity-based inhibitor and probe and applied it to obtain new insights into breast cancer.</p>","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7781487/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38829905","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dysregulated PJA1-TGF-β signaling in cancer stem cell-associated liver cancers. PJA1-TGF-β信号在癌症干细胞相关性肝癌中的失调
Pub Date : 2020-11-01 DOI: 10.18632/oncoscience.522
Jian Chen, Julian A Gingold

The transforming growth factor beta (TGF-β) signaling pathway plays important roles in cell differentiation, stem cell modulation, organ lineage, and immune suppression. TGF-β signaling is negatively regulated by the ubiquitin-proteasome pathway. Although mouse models of cancer arising from a defective TGF-β pathway clearly demonstrate the tumor-suppressive role of TGF-β, the underlying mechanism by which a defective TGF-β pathway triggers liver cancer development is poorly understood. This review summarizes key findings from our recent studies connecting TGF-β to hepatic oncogenesis and highlights the vulnerability of TGF-β signaling to PJA1-mediated ubiquitination. TGF-β, together with the chromatin insulator CCCTC-binding factor (CTCF), epigenetically and transcriptionally regulate tumor promoter genes, including IGF2 and TERT, in TGF-β-defective mice and in human liver cancers. Dysfunction of the TGF-β-regulated SPTBN1/SMAD3/CTCF complex increases stem cell-like properties in hepatocellular carcinoma (HCC) cells and enhances tumorigenesis in tumor-initiating cells in a mouse model. PJA1, a novel E3 ubiquitin ligase, is a key negative regulator of TGF-β signaling. PJA1 overexpression is detected in HCCs and is sufficient to suppress SMAD3- and SPTBN1-mediated TGF-β tumor suppressor signaling, promoting HCC proliferation. Dysregulated PJA1-TGF-β signaling activates oncogenic genes and promotes tumorigenesis in human liver cancers. In addition, inhibition of PJA1 by treatment with E3 ligase inhibitors restores TGF-β tumor-suppressor function and suppresses liver cancer progression. These new findings suggest potential therapeutic avenues for targeting dysregulated PJA1-TGF-β signaling via cancer stem cells in liver cancers.

转化生长因子β (TGF-β)信号通路在细胞分化、干细胞调控、器官谱系和免疫抑制等方面发挥着重要作用。TGF-β信号被泛素-蛋白酶体通路负调控。虽然由缺陷的TGF-β通路引起的癌症小鼠模型清楚地证明了TGF-β的肿瘤抑制作用,但缺陷的TGF-β通路引发肝癌发展的潜在机制尚不清楚。这篇综述总结了我们最近关于TGF-β与肝癌发生的研究的主要发现,并强调了TGF-β信号传导对pja1介导的泛素化的脆弱性。TGF-β与染色质绝缘子cctc结合因子(CTCF)在TGF-β缺陷小鼠和人肝癌中通过表观遗传和转录调控肿瘤启动子基因,包括IGF2和TERT。在小鼠模型中,TGF-β调控的SPTBN1/SMAD3/CTCF复合物功能障碍可增加肝细胞癌(HCC)细胞的干细胞样特性,并促进肿瘤起始细胞的肿瘤发生。PJA1是一种新的E3泛素连接酶,是TGF-β信号传导的关键负调控因子。PJA1在HCC中过表达,足以抑制SMAD3-和sptbn1介导的TGF-β肿瘤抑制信号,促进HCC增殖。PJA1-TGF-β信号通路失调激活致癌基因,促进人类肝癌的发生。此外,E3连接酶抑制剂对PJA1的抑制可以恢复TGF-β肿瘤抑制功能并抑制肝癌的进展。这些新发现提示了通过肝癌干细胞靶向PJA1-TGF-β信号失调的潜在治疗途径。
{"title":"Dysregulated PJA1-TGF-β signaling in cancer stem cell-associated liver cancers.","authors":"Jian Chen,&nbsp;Julian A Gingold","doi":"10.18632/oncoscience.522","DOIUrl":"https://doi.org/10.18632/oncoscience.522","url":null,"abstract":"<p><p>The transforming growth factor beta (TGF-β) signaling pathway plays important roles in cell differentiation, stem cell modulation, organ lineage, and immune suppression. TGF-β signaling is negatively regulated by the ubiquitin-proteasome pathway. Although mouse models of cancer arising from a defective TGF-β pathway clearly demonstrate the tumor-suppressive role of TGF-β, the underlying mechanism by which a defective TGF-β pathway triggers liver cancer development is poorly understood. This review summarizes key findings from our recent studies connecting TGF-β to hepatic oncogenesis and highlights the vulnerability of TGF-β signaling to PJA1-mediated ubiquitination. TGF-β, together with the chromatin insulator CCCTC-binding factor (CTCF), epigenetically and transcriptionally regulate tumor promoter genes, including IGF2 and TERT, in TGF-β-defective mice and in human liver cancers. Dysfunction of the TGF-β-regulated SPTBN1/SMAD3/CTCF complex increases stem cell-like properties in hepatocellular carcinoma (HCC) cells and enhances tumorigenesis in tumor-initiating cells in a mouse model. PJA1, a novel E3 ubiquitin ligase, is a key negative regulator of TGF-β signaling. PJA1 overexpression is detected in HCCs and is sufficient to suppress SMAD3- and SPTBN1-mediated TGF-β tumor suppressor signaling, promoting HCC proliferation. Dysregulated PJA1-TGF-β signaling activates oncogenic genes and promotes tumorigenesis in human liver cancers. In addition, inhibition of PJA1 by treatment with E3 ligase inhibitors restores TGF-β tumor-suppressor function and suppresses liver cancer progression. These new findings suggest potential therapeutic avenues for targeting dysregulated PJA1-TGF-β signaling via cancer stem cells in liver cancers.</p>","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7781490/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10343114","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 9
Getting quantitative on the effects of somatic mutation on cancer. 定量研究体细胞突变对癌症的影响。
Pub Date : 2020-10-27 eCollection Date: 2020-11-01 DOI: 10.18632/oncoscience.521
Jeffrey P Townsend
Numerous powerful bioinformatic analyses of cancer tumor sequencing have applied sophisticated mutation calling, determining the key cancer-causing variants and quantifying their prevalence. The calculations of prevalence of a mutation across tumors and the determination of the statistical significance of whether it is a driver are the “shoulders” that have enabled the build-up to the most useful metrics about cancer variants—metrics which quantify the effect of the variant on replication and survival of the cancer lineage. Ostrow et al [1] effectively and comprehensively applied ratios of non-synonymous change to synonymous change to quantify natural selection in the somatic evolution of cancer, an approach that has been followed by others in different ways and contexts since then [2–4]. Martincorena et al [2] performed a cogent gene-wide analysis using mutation signatures c.f. [5] on the larger data sets available three years later. More recently, it was revealed that previous studies have reported variant prevalence and P value, but have not reported cancer effect sizes, which quantify the effect of natural selection on somatic mutations within cancer cell lineages. Deconvolving the baseline variant mutation rates enables estimation of the selection intensity of individual mutations [6, 7]—a quantification that should be directly interpreted as a cancer lineage effect size that should be used in decision-making. This measure of the effect of specific somatic mutations on cancer cell proliferation and survival should be widely appreciated as a primary consideration of precisionmedicine tumor boards, which are in operation at hospitals around the world. Effect sizes of somatic mutations should also be a key consideration in the initiation and design of precision medicine clinical trials: the number of trials has been increasing so rapidly that some have argued that demand is vastly outpacing the supply of enrollable patients [8]. Effect sizes should guide target selection in pharmacological development, an approximately three billion dollar industry [9]. And gene-specific site-specific effect sizes should guide basic research prioritization toward those important components of molecular and cell biology that have long-term potential to lead to therapies and cures for cancer. There appear to be increasing numbers of drivers in each cancer as we examine larger and larger datasets [10], and each driver has its own quantitative effect on cancer [11]. Decoupling the contributions of mutation and cancer lineage selection to the frequency of somatic variants among tumors is critical to understanding— and predicting—the therapeutic potential of different interventions [12]. Importantly, antagonistic and synergistic epistasis among mutations also impacts the potential therapeutic benefit of targeted drug development [13]. Active use of these quantitative approaches are essential to furthering basic research on cancer, informing clinical practice, and pro
{"title":"Getting quantitative on the effects of somatic mutation on cancer.","authors":"Jeffrey P Townsend","doi":"10.18632/oncoscience.521","DOIUrl":"https://doi.org/10.18632/oncoscience.521","url":null,"abstract":"Numerous powerful bioinformatic analyses of cancer tumor sequencing have applied sophisticated mutation calling, determining the key cancer-causing variants and quantifying their prevalence. The calculations of prevalence of a mutation across tumors and the determination of the statistical significance of whether it is a driver are the “shoulders” that have enabled the build-up to the most useful metrics about cancer variants—metrics which quantify the effect of the variant on replication and survival of the cancer lineage. Ostrow et al [1] effectively and comprehensively applied ratios of non-synonymous change to synonymous change to quantify natural selection in the somatic evolution of cancer, an approach that has been followed by others in different ways and contexts since then [2–4]. Martincorena et al [2] performed a cogent gene-wide analysis using mutation signatures c.f. [5] on the larger data sets available three years later. More recently, it was revealed that previous studies have reported variant prevalence and P value, but have not reported cancer effect sizes, which quantify the effect of natural selection on somatic mutations within cancer cell lineages. Deconvolving the baseline variant mutation rates enables estimation of the selection intensity of individual mutations [6, 7]—a quantification that should be directly interpreted as a cancer lineage effect size that should be used in decision-making. This measure of the effect of specific somatic mutations on cancer cell proliferation and survival should be widely appreciated as a primary consideration of precisionmedicine tumor boards, which are in operation at hospitals around the world. Effect sizes of somatic mutations should also be a key consideration in the initiation and design of precision medicine clinical trials: the number of trials has been increasing so rapidly that some have argued that demand is vastly outpacing the supply of enrollable patients [8]. Effect sizes should guide target selection in pharmacological development, an approximately three billion dollar industry [9]. And gene-specific site-specific effect sizes should guide basic research prioritization toward those important components of molecular and cell biology that have long-term potential to lead to therapies and cures for cancer. There appear to be increasing numbers of drivers in each cancer as we examine larger and larger datasets [10], and each driver has its own quantitative effect on cancer [11]. Decoupling the contributions of mutation and cancer lineage selection to the frequency of somatic variants among tumors is critical to understanding— and predicting—the therapeutic potential of different interventions [12]. Importantly, antagonistic and synergistic epistasis among mutations also impacts the potential therapeutic benefit of targeted drug development [13]. Active use of these quantitative approaches are essential to furthering basic research on cancer, informing clinical practice, and pro","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2020-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7781488/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38829904","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncoscience
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