首页 > 最新文献

Oncoscience最新文献

英文 中文
STAT3 and mTOR: co-operating to drive HIF and angiogenesis STAT3和mTOR:协同驱动HIF和血管生成
Pub Date : 2015-11-23 DOI: 10.18632/ONCOSCIENCE.272
K. Dodd, A. Tee
Our understanding of angiogenic signalling has been significantly enhanced through studies of a rare genetic disorder called Tuberous Sclerosis Complex (TSC). TSC patients are predisposed to highly vascularised tumours, where renal angiomyolipomas produce high levels of vascular endothelial growth factor (VEGF) that can be readily detected. It is well established that VEGF is driven through hypoxic signalling, with the transcription factor hypoxia inducible factor-1α (HIF-1α) playing a crucial role in its expression. Early studies using cell line models of TSC uncovered that the mammalian target of rapamycin complex 1 (mTORC1) is a key mediator of HIF-1α synthesis, and highlighted the anti-angiogenic properties of mTORC1 inhibitors [1]. Herein we review our recent findings characterising mTORC1 mediated regulation of HIF-1α and discuss the clinical implications of our work. We demonstrated that mTORC1 drives HIF-1α expression via three mechanisms, promoting not only the transcription of HIF-1α mRNA via signal transducer and activator of transcription 3 (STAT3), but also its translation via both eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1 (S6K1). This drive in HIF-1α activity downstream of mTORC1 explains why the tumours which present in TSC are so heavily vascularised, and accounts for the anti-tumorigenic properties of mTOR inhibitors used in this setting. In concordance with this, we observe a 10 fold-increase in HIF-1α transcriptional activity under hypoxia with TSC2 loss, highlighting the significant impact mTORC1 activation can have on HIF-1α. Whilst mTORC1 can promote the transcriptional activity of STAT3 through direct phosphorylation of Ser727, STAT3 is also subject to regulation from a number of different cytokines and growth factors which signal through the receptor tyrosine kinase JAK2 [2]. Both JAK2/STAT3 and mTORC1 signalling pathways are frequently activated in a wide range of malignancies and converge at the level of HIF-1α (see Figure ​Figure1).1). Whilst mTOR inhibitors are effective at blocking Ser727 phosphorylation of STAT3, we were able to completely abolish HIF-1α expression by targeting both the JAK2-mediated Tyr705 phosphorylation site and the mTORC1-mediated Ser727 site. Our work indicates that targeting STAT3 in parallel to mTORC1 could enhance the anti-angiogenic and anti-tumorigenic properties of mTOR inhibitors that are currently in clinical use [3]. Figure 1 Multifaceted regulation of HIF-1α/VEGF-A via mTORC1 and STAT3 Growth of tumours in renal cell carcinoma (RCC) is highly dependent on mTORC1, HIF and VEGF which drive a pro-angiogenic response. In the microenvironment of the kidney, angiogenic signalling is crucial for metabolic transformation and malignancy. Although there has been much investment into drug discovery and the development of inhibitors that directly inhibit HIF, none of these compounds are currently suitable for clinical use. C
STAT3在正常细胞功能中也是必不可少的,这表明STAT3抑制剂的副作用应该很低。总的来说,我们的工作强调了开发临床可用的STAT3抑制剂用于癌症治疗的必要性。
{"title":"STAT3 and mTOR: co-operating to drive HIF and angiogenesis","authors":"K. Dodd, A. Tee","doi":"10.18632/ONCOSCIENCE.272","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.272","url":null,"abstract":"Our understanding of angiogenic signalling has been significantly enhanced through studies of a rare genetic disorder called Tuberous Sclerosis Complex (TSC). TSC patients are predisposed to highly vascularised tumours, where renal angiomyolipomas produce high levels of vascular endothelial growth factor (VEGF) that can be readily detected. It is well established that VEGF is driven through hypoxic signalling, with the transcription factor hypoxia inducible factor-1α (HIF-1α) playing a crucial role in its expression. Early studies using cell line models of TSC uncovered that the mammalian target of rapamycin complex 1 (mTORC1) is a key mediator of HIF-1α synthesis, and highlighted the anti-angiogenic properties of mTORC1 inhibitors [1]. Herein we review our recent findings characterising mTORC1 mediated regulation of HIF-1α and discuss the clinical implications of our work. \u0000 \u0000We demonstrated that mTORC1 drives HIF-1α expression via three mechanisms, promoting not only the transcription of HIF-1α mRNA via signal transducer and activator of transcription 3 (STAT3), but also its translation via both eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1 (S6K1). This drive in HIF-1α activity downstream of mTORC1 explains why the tumours which present in TSC are so heavily vascularised, and accounts for the anti-tumorigenic properties of mTOR inhibitors used in this setting. In concordance with this, we observe a 10 fold-increase in HIF-1α transcriptional activity under hypoxia with TSC2 loss, highlighting the significant impact mTORC1 activation can have on HIF-1α. \u0000 \u0000Whilst mTORC1 can promote the transcriptional activity of STAT3 through direct phosphorylation of Ser727, STAT3 is also subject to regulation from a number of different cytokines and growth factors which signal through the receptor tyrosine kinase JAK2 [2]. Both JAK2/STAT3 and mTORC1 signalling pathways are frequently activated in a wide range of malignancies and converge at the level of HIF-1α (see Figure ​Figure1).1). Whilst mTOR inhibitors are effective at blocking Ser727 phosphorylation of STAT3, we were able to completely abolish HIF-1α expression by targeting both the JAK2-mediated Tyr705 phosphorylation site and the mTORC1-mediated Ser727 site. Our work indicates that targeting STAT3 in parallel to mTORC1 could enhance the anti-angiogenic and anti-tumorigenic properties of mTOR inhibitors that are currently in clinical use [3]. \u0000 \u0000 \u0000 \u0000Figure 1 \u0000 \u0000Multifaceted regulation of HIF-1α/VEGF-A via mTORC1 and STAT3 \u0000 \u0000 \u0000 \u0000Growth of tumours in renal cell carcinoma (RCC) is highly dependent on mTORC1, HIF and VEGF which drive a pro-angiogenic response. In the microenvironment of the kidney, angiogenic signalling is crucial for metabolic transformation and malignancy. Although there has been much investment into drug discovery and the development of inhibitors that directly inhibit HIF, none of these compounds are currently suitable for clinical use. C","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"44 1","pages":"913 - 914"},"PeriodicalIF":0.0,"publicationDate":"2015-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90582742","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: 13
The role of ICAM-2 in neuroblastoma ICAM-2在神经母细胞瘤中的作用
Pub Date : 2015-11-23 DOI: 10.18632/ONCOSCIENCE.273
K. Yoon, A. Miller, K. Kreitzburg
The role of immunoglobulin superfamily cell adhesion molecules (CAMs) in facilitating immune responses in normal and tumor cells is well established. Cell adhesion molecule-1 (CADM1), for example, suppresses development of mouse mammary tumor cell metastasis by interacting with CD8+ T cells in immune-competent hosts [1]. Similarly, co-expression of Intercellular Adhesion Molecule-2 (ICAM-2) and chemokine C-X-C motif ligand 17 (CXCL17) elicits anti-tumor immune responses and suppresses tumor growth [2]. Although controversial, current literature suggests that proteins of the immunoglobulin superfamily CAMs also have functions that may well be distinct from their roles in mediating immune responses. Junctional adhesion molecule-A (JAM-A), for example, negatively regulates breast cancer cell invasiveness by disrupting tight junctions [3]; and a member of the B7 family of the immunoglobulin superfamily proteins, B7-H3, impairs osteogenic differentiation in vitro and in vivo [4]. Our lab demonstrated that ICAM-2 inhibited the development of disseminated neuroblastoma tumors in a preclinical model of metastatic neuroblastoma [5-7]. This inhibition depended on the interaction of ICAM-2 with the actin cytoskeletal linker protein α-actinin, an interaction that inhibited cell motility [7]. Ectopic expression of ICAM-2 did not affect the tumorigenic potential of neuroblastoma cells [7]. Importantly, immunohistochemical analyses of primary neuroblastoma tumor specimens demonstrated that neuroblastoma cells expressing ICAM-2 are phenotypically and histologically those recognized clinically to have limited metastatic potential [5]. Since metastatic disease is responsible for >90% of cancer-related deaths for multiple types of solid tumors, we suggest that elucidation of the molecular mechanism by which ICAM-2 suppresses the metastatic potential of neuroblastoma cells would identify proteins or pathways that might be exploited therapeutically to prevent metastatic disease progression. In normal tissues ICAM-2 is expressed predominantly by neovascular endothelial cells, and at lower levels by established vasculature and some leukocytes. The 202 amino acids comprising its extracellular domain mediate binding of ICAM-2 on endothelial cells to β2-integrins on the surface of leukocytes, to facilitate migration of neutrophils through the vascular endothelium as a component of immune reactions [8]. In neuroblastoma cells ICAM-2 inhibits cell motility independent of immune response, as we observed this inhibition in wound healing and modified Boyden chamber assays in vitro, assays that clearly lack an immune component [5,7]. In silico modeling indicated that ICAM-2 with mutations in the proposed α-actinin binding domain had a more ‘closed’ configuration than the wild type protein, and predicted that these ICAM-2 mutants would not interact with α-actinin [7]. Co-immunoprecipitation experiments confirmed in silico predictions [7]. In support of these findings, t
免疫球蛋白超家族细胞粘附分子(CAMs)在促进正常细胞和肿瘤细胞免疫应答中的作用已经得到了很好的证实。例如,细胞粘附分子-1 (CADM1)通过与免疫活性宿主中的CD8+ T细胞相互作用,抑制小鼠乳腺肿瘤细胞转移的发展[1]。同样,细胞间粘附分子-2 (ICAM-2)和趋化因子C-X-C基序配体17 (CXCL17)的共表达引发抗肿瘤免疫反应并抑制肿瘤生长[2]。尽管存在争议,但目前的文献表明,免疫球蛋白超家族CAMs的蛋白质也具有与其介导免疫反应的作用截然不同的功能。例如,连接粘附分子- a (JAM-A)通过破坏紧密连接来负向调节乳腺癌细胞的侵袭性[3];以及免疫球蛋白超家族蛋白B7家族的成员B7- h3,在体外和体内都能阻碍成骨分化[4]。我们的实验室在转移性神经母细胞瘤临床前模型中证实,ICAM-2抑制播散性神经母细胞瘤肿瘤的发展[5-7]。这种抑制依赖于ICAM-2与肌动蛋白细胞骨架连接蛋白α-肌动蛋白的相互作用,这种相互作用抑制细胞运动[7]。异位表达ICAM-2不影响神经母细胞瘤细胞的致瘤潜能[7]。重要的是,原发性神经母细胞瘤肿瘤标本的免疫组织化学分析表明,在表型和组织学上表达ICAM-2的神经母细胞瘤细胞在临床上被认为具有有限的转移潜力[5]。由于转移性疾病是多种类型实体肿瘤中90%以上的癌症相关死亡的原因,我们建议阐明ICAM-2抑制神经母细胞瘤细胞转移潜能的分子机制,从而确定可能用于治疗预防转移性疾病进展的蛋白质或途径。在正常组织中,ICAM-2主要由新生血管内皮细胞表达,在已建立的血管和一些白细胞中表达的水平较低。其胞外结构域由202个氨基酸组成,介导内皮细胞上的ICAM-2与白细胞表面的β2整合素结合,促进中性粒细胞通过血管内皮迁移,作为免疫反应的一个组成部分[8]。在神经母细胞瘤细胞中,ICAM-2抑制独立于免疫反应的细胞运动,正如我们在体外伤口愈合和改良Boyden室实验中观察到的那样,这种抑制作用显然缺乏免疫成分[5,7]。计算机模拟表明,在α-肌动蛋白结合区域发生突变的ICAM-2比野生型蛋白具有更“封闭”的结构,并预测这些ICAM-2突变体不会与α-肌动蛋白相互作用[7]。共免疫沉淀实验证实了计算机预测[7]。为了支持这些发现,ICAM-2与α-肌动蛋白的相互作用对于抑制体内播散性肿瘤的发展至关重要[7]。如前所述,ICAM-2不影响致瘤潜力,未发表的数据显示ICAM-2对上皮-间质转化(EMT)或干性标志物的表达没有影响。尽管我们认为ICAM-2在体外抑制肿瘤细胞运动是ICAM-2参与不同于免疫反应的细胞功能的有力证据,但我们承认转移性肿瘤进展的复杂性,并提出我们的数据提出了关于ICAM-2抑制神经母细胞瘤细胞运动的机制的多种假设。第一种假设是,ICAM-2与α-肌动蛋白的细胞内相互作用启动了“由内到外”的信号传导,并导致ICAM-2的细胞外结构域的构象变化,从而促进(或抑制)与特定的细胞外基质蛋白的相互作用,这些蛋白在细胞运动中起着不可或缺的作用。其次,ICAM-2与α-肌动蛋白的相互作用改变了α-肌动蛋白的构象,促进(或抑制)这种肌动蛋白细胞骨架蛋白与其他结合伙伴的结合,ICAM-2充当α-肌动蛋白的“激活剂”(或抑制剂),而不是作为膜锚蛋白。基于未发表的微阵列数据的第三种假设是,ICAM-2表达间接上调参与局灶黏附形成和维持的蛋白酪氨酸磷酸酶(PTPs),如局灶黏附激酶(FAK)、Src或Rac和Rho GTPases,它们在肿瘤细胞运动中都起着至关重要的作用。我们的数据显示,ICAM-2抑制肿瘤细胞的运动和抑制神经母细胞瘤细胞的转移潜能。我们建议阐明与ICAM-2表达相关的分子事件将确定调节这种细胞类型转移过程的关键蛋白相互作用。
{"title":"The role of ICAM-2 in neuroblastoma","authors":"K. Yoon, A. Miller, K. Kreitzburg","doi":"10.18632/ONCOSCIENCE.273","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.273","url":null,"abstract":"The role of immunoglobulin superfamily cell adhesion molecules (CAMs) in facilitating immune responses in normal and tumor cells is well established. Cell adhesion molecule-1 (CADM1), for example, suppresses development of mouse mammary tumor cell metastasis by interacting with CD8+ T cells in immune-competent hosts [1]. Similarly, co-expression of Intercellular Adhesion Molecule-2 (ICAM-2) and chemokine C-X-C motif ligand 17 (CXCL17) elicits anti-tumor immune responses and suppresses tumor growth [2]. Although controversial, current literature suggests that proteins of the immunoglobulin superfamily CAMs also have functions that may well be distinct from their roles in mediating immune responses. Junctional adhesion molecule-A (JAM-A), for example, negatively regulates breast cancer cell invasiveness by disrupting tight junctions [3]; and a member of the B7 family of the immunoglobulin superfamily proteins, B7-H3, impairs osteogenic differentiation in vitro and in vivo [4]. \u0000 \u0000Our lab demonstrated that ICAM-2 inhibited the development of disseminated neuroblastoma tumors in a preclinical model of metastatic neuroblastoma [5-7]. This inhibition depended on the interaction of ICAM-2 with the actin cytoskeletal linker protein α-actinin, an interaction that inhibited cell motility [7]. Ectopic expression of ICAM-2 did not affect the tumorigenic potential of neuroblastoma cells [7]. Importantly, immunohistochemical analyses of primary neuroblastoma tumor specimens demonstrated that neuroblastoma cells expressing ICAM-2 are phenotypically and histologically those recognized clinically to have limited metastatic potential [5]. Since metastatic disease is responsible for >90% of cancer-related deaths for multiple types of solid tumors, we suggest that elucidation of the molecular mechanism by which ICAM-2 suppresses the metastatic potential of neuroblastoma cells would identify proteins or pathways that might be exploited therapeutically to prevent metastatic disease progression. \u0000 \u0000In normal tissues ICAM-2 is expressed predominantly by neovascular endothelial cells, and at lower levels by established vasculature and some leukocytes. The 202 amino acids comprising its extracellular domain mediate binding of ICAM-2 on endothelial cells to β2-integrins on the surface of leukocytes, to facilitate migration of neutrophils through the vascular endothelium as a component of immune reactions [8]. In neuroblastoma cells ICAM-2 inhibits cell motility independent of immune response, as we observed this inhibition in wound healing and modified Boyden chamber assays in vitro, assays that clearly lack an immune component [5,7]. \u0000 \u0000In silico modeling indicated that ICAM-2 with mutations in the proposed α-actinin binding domain had a more ‘closed’ configuration than the wild type protein, and predicted that these ICAM-2 mutants would not interact with α-actinin [7]. Co-immunoprecipitation experiments confirmed in silico predictions [7]. In support of these findings, t","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"17 1","pages":"915 - 916"},"PeriodicalIF":0.0,"publicationDate":"2015-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87665668","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: 3
Targeting BCL6 and STAT3 in triple negative breast cancer: the one-two punch? 靶向BCL6和STAT3治疗三阴性乳腺癌:组合拳?
Pub Date : 2015-11-21 DOI: 10.18632/ONCOSCIENCE.270
Sarah R Walker, D. Frank
Breast cancer remains the second leading cause of cancer deaths in women in the United States. Triple negative breast cancer, tumors lacking estrogen receptor, progesterone receptor, and Her2, only comprise about 20–30% of breast tumors diagnosed. However, due to the lack of specific targeted therapy as exists for ER+ or Her2+ tumors, they account for many of the breast cancer deaths. To develop targeted therapy for triple negative breast cancer, inhibiting two targets may be necessary. One new target of interest is the transcriptional modulator BCL6 which has been recently identified as playing an important role in breast cancer [1–3]. BCL6 has been characterized as a transcriptional repressor that recruits various corepressor complexes to repress its target genes; however, genes have also been identified that are upregulated by BCL6 [1, 2], including Zeb1 which is involved in promoting EMT [3]. While the roles of BCL6 in preventing terminal differentiation of B cells in the germinal center and promoting diffuse large B cell lymphoma are well known, little is currently known about the roles of BCL6 in solid tissue. BCL6 is amplified in ∼50% of breast tumors and is expressed in most breast cancer cell lines, including triple negative breast cancer cell lines [1]. Furthermore, BCL6 expression is correlated with disease progression and poor overall survival [2], and targeting BCL6 results in reduced growth and loss of breast cancer cell viability [1]. Importantly, triple negative breast cancer cell lines were among the most sensitive to BCL6 inhibition. In the normal mammary gland, BCL6 has been shown to prevent terminal differentiation and milk production, in part due to competition with signal transducer and activator of transcription 5 (STAT5) for regulation of target genes [4]. STATs are latent transcription factors that remain in the cytoplasm until activated by tyrosine phosphorylation often via Jak kinases. They then translocate to the nucleus as an active dimer and modulate transcription of target genes. There are seven members of the STAT family and four members, STAT1, STAT3, STAT5a, and STAT5b regulate the expression of BCL6. While STAT1 and STAT3 upregulate BCL6 expression, STAT5 (5a and 5b) downregulates BCL6 expression [5]. All four STATs have been shown to play roles in breast cancer; however, STAT3 activation has been linked to more aggressive types. While STAT3 activation can occur in any subtype of breast cancer [5], STAT3 activation is restricted largely to triple negative breast cancer cell lines, and STAT3 signaling has been shown to be important for the survival of triple negative breast tumors [6]. Both BCL6 and STAT3 play critical roles in triple negative breast cancer, including promoting survival and EMT, through modulating largely distinct target genes. Thus, could targeting these two factors together be a useful strategy for specifically treating triple negative breast cancer? Inhibition of BCL6 by siRNA or the
乳腺癌仍然是美国妇女癌症死亡的第二大原因。三阴性乳腺癌,即缺乏雌激素受体、孕激素受体和Her2的肿瘤,仅占确诊乳腺肿瘤的20-30%。然而,由于缺乏针对ER+或Her2+肿瘤的特异性靶向治疗,它们导致了许多乳腺癌死亡。为了开发针对三阴性乳腺癌的靶向治疗,可能需要抑制两个靶点。一个令人感兴趣的新靶点是转录调节剂BCL6,它最近被发现在乳腺癌中起重要作用[1-3]。BCL6被认为是一种转录抑制因子,它可以招募各种辅助抑制因子复合物来抑制其靶基因;然而,也发现了BCL6上调的基因[1,2],包括参与促进EMT[3]的Zeb1。虽然BCL6在阻止生发中心B细胞终末分化和促进弥漫性大B细胞淋巴瘤中的作用是众所周知的,但目前对BCL6在实体组织中的作用知之甚少。BCL6在约50%的乳腺肿瘤中扩增,并在大多数乳腺癌细胞系中表达,包括三阴性乳腺癌细胞系[1]。此外,BCL6表达与疾病进展和较差的总生存期[2]相关,靶向BCL6可导致乳腺癌细胞生长减少和活力丧失[1]。重要的是,三阴性乳腺癌细胞系对BCL6的抑制最为敏感。在正常乳腺中,BCL6已被证明可以阻止终末分化和泌乳,部分原因是BCL6与信号换能器和转录激活因子5 (STAT5)竞争调控靶基因[4]。STATs是潜伏在细胞质中的转录因子,通常通过Jak激酶被酪氨酸磷酸化激活。然后它们作为活性二聚体转移到细胞核并调节目标基因的转录。STAT家族共有7个成员,其中STAT1、STAT3、STAT5a、STAT5b 4个成员调控BCL6的表达。STAT1和STAT3上调BCL6表达,而STAT5 (5a和5b)下调BCL6表达[5]。所有四种stat都被证明在乳腺癌中起作用;然而,STAT3的激活与更具侵略性的类型有关。虽然STAT3激活可以发生在任何亚型乳腺癌[5]中,但STAT3激活主要局限于三阴性乳腺癌细胞系,STAT3信号传导已被证明对三阴性乳腺癌[6]的存活很重要。BCL6和STAT3都在三阴性乳腺癌中发挥关键作用,包括通过调节不同的靶基因促进生存和EMT。因此,针对这两个因素是否可以作为治疗三阴性乳腺癌的有效策略呢?用siRNA或拟肽i - bpi[7]抑制BCL6,同时用Jak激酶抑制剂TG101348或nifuroxazide抑制STAT3,可增强对三阴性乳腺癌细胞系[1]的杀伤。这些发现表明,同时针对两种致癌转录调节剂的靶向治疗可能是治疗三阴性乳腺癌的新途径。
{"title":"Targeting BCL6 and STAT3 in triple negative breast cancer: the one-two punch?","authors":"Sarah R Walker, D. Frank","doi":"10.18632/ONCOSCIENCE.270","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.270","url":null,"abstract":"Breast cancer remains the second leading cause of cancer deaths in women in the United States. Triple negative breast cancer, tumors lacking estrogen receptor, progesterone receptor, and Her2, only comprise about 20–30% of breast tumors diagnosed. However, due to the lack of specific targeted therapy as exists for ER+ or Her2+ tumors, they account for many of the breast cancer deaths. To develop targeted therapy for triple negative breast cancer, inhibiting two targets may be necessary. \u0000 \u0000One new target of interest is the transcriptional modulator BCL6 which has been recently identified as playing an important role in breast cancer [1–3]. BCL6 has been characterized as a transcriptional repressor that recruits various corepressor complexes to repress its target genes; however, genes have also been identified that are upregulated by BCL6 [1, 2], including Zeb1 which is involved in promoting EMT [3]. While the roles of BCL6 in preventing terminal differentiation of B cells in the germinal center and promoting diffuse large B cell lymphoma are well known, little is currently known about the roles of BCL6 in solid tissue. BCL6 is amplified in ∼50% of breast tumors and is expressed in most breast cancer cell lines, including triple negative breast cancer cell lines [1]. Furthermore, BCL6 expression is correlated with disease progression and poor overall survival [2], and targeting BCL6 results in reduced growth and loss of breast cancer cell viability [1]. Importantly, triple negative breast cancer cell lines were among the most sensitive to BCL6 inhibition. In the normal mammary gland, BCL6 has been shown to prevent terminal differentiation and milk production, in part due to competition with signal transducer and activator of transcription 5 (STAT5) for regulation of target genes [4]. \u0000 \u0000STATs are latent transcription factors that remain in the cytoplasm until activated by tyrosine phosphorylation often via Jak kinases. They then translocate to the nucleus as an active dimer and modulate transcription of target genes. There are seven members of the STAT family and four members, STAT1, STAT3, STAT5a, and STAT5b regulate the expression of BCL6. While STAT1 and STAT3 upregulate BCL6 expression, STAT5 (5a and 5b) downregulates BCL6 expression [5]. All four STATs have been shown to play roles in breast cancer; however, STAT3 activation has been linked to more aggressive types. While STAT3 activation can occur in any subtype of breast cancer [5], STAT3 activation is restricted largely to triple negative breast cancer cell lines, and STAT3 signaling has been shown to be important for the survival of triple negative breast tumors [6]. \u0000 \u0000Both BCL6 and STAT3 play critical roles in triple negative breast cancer, including promoting survival and EMT, through modulating largely distinct target genes. Thus, could targeting these two factors together be a useful strategy for specifically treating triple negative breast cancer? Inhibition of BCL6 by siRNA or the ","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"79 1","pages":"912 - 912"},"PeriodicalIF":0.0,"publicationDate":"2015-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82403705","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: 8
A predictive signature for therapy assignment and risk assessment in prostate cancer 前列腺癌治疗分配和风险评估的预测性特征
Pub Date : 2015-11-20 DOI: 10.18632/ONCOSCIENCE.271
D. Bonci, R. De Maria
Prostate cancer remains the second leading cause of death in men. It is imperative to improve patient management in identifying bio-markers for personalized treatment. We demonstrated miR-15/miR-16 loss and miR-21 up-regulation and deregulation of their target genes, which represent a promising signature for ameliorating therapy assignment and risk assessment in prostate cancer.
前列腺癌仍然是男性死亡的第二大原因。提高患者管理水平,识别生物标志物,实现个性化治疗势在必行。我们证明了miR-15/miR-16缺失和miR-21靶基因的上调和解除调控,这代表了改善前列腺癌治疗分配和风险评估的有希望的标志。
{"title":"A predictive signature for therapy assignment and risk assessment in prostate cancer","authors":"D. Bonci, R. De Maria","doi":"10.18632/ONCOSCIENCE.271","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.271","url":null,"abstract":"Prostate cancer remains the second leading cause of death in men. It is imperative to improve patient management in identifying bio-markers for personalized treatment. We demonstrated miR-15/miR-16 loss and miR-21 up-regulation and deregulation of their target genes, which represent a promising signature for ameliorating therapy assignment and risk assessment in prostate cancer.","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"8 1","pages":"920 - 923"},"PeriodicalIF":0.0,"publicationDate":"2015-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86955882","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: 5
MicroRNA-155 regulates tumor myeloid-derived suppressive cells MicroRNA-155调控肿瘤髓源性抑制细胞
Pub Date : 2015-11-19 DOI: 10.18632/ONCOSCIENCE.269
Siqi Chen, Yi Zhang, Bin Zhang
MicroRNA is small non-coding RNA and can lead to translational repression or target degradation by base-pairing with complementary sequences of mRNA molecules. MicroRNA-155 (miR-155), one of the most studied microRNA, is the first one to be reported as oncogenic [1]. miR-155 is over expressed in a long list of both hematological and solid tumors and is of paramount importance in cancer diagnosis and prognosis. However, how miR-155 particularly in host immune system regulates the tumor progression remains poorly understood. Our study underscores a contextual role of miR-155 in regulating tumor growth and tumor immunity via distinct immune subsets within tumors [2]. We conclude that the balance of different effects between those immune cell populations, which are regulated by miR-155, appears to determine whether miR-155 promotes or inhibits tumor growth [2]. We demonstrated that host miR-155 deficiency promoted antitumor T cell immunity in multiple transplanted tumor models. Further analysis of immune cell compartments revealed that miR-155 was required for the accumulation and suppressive function of myeloid-derived suppressive cells (MDSC) in the tumor microenvironment. Apart from the direct modulation on MDSC, miR-155 was also required for the MDSC-mediated CD4+Foxp3+ regulatory T cells (Treg) induction. On the other hand, miR-155 deficiency hampered the antitumor responses of both dendritic cells and T cells. Therefore, it appears that in our tumor models, miR-155 mediated a dominant immunosuppressive effect by MDSC, leading to the enhanced overall antitumor immunity in miR-155 deficient hosts. Reduced colon inflammation and decreased colorectal carcinogenesis were also found in miR-155 deficient mice when azoxymethane (AOM) and dextran sodium sulphate (DSS) were combined to induce colon lesions. Furthermore, miR-155 was upregulated in MDSC either from tumor-bearing hosts or generated from bone marrow progenitors by GM-CSF and IL-6. These results support the notion that miR-155 is a prototypical microRNA bridging inflammation and cancer development [3]. Although miR-155 may regulate tumor growth in an intrinsic manner, it is likely that inflammation promotes the accumulation of functional MDSC by increased miR-155 that dampens the immune surveillance and antitumor immunity, thereby facilitating tumor growth. To identify the molecular mechanisms by which miR-155 regulates MDSC (Figure ​(Figure1),1), we found that miR155 retained the suppressive activity of MDSCs through inhibiting SOCS1. Moreover, inverse correlations between miR-155 expression and SHIP-1/SOCS1 expression were established in MDSC. As SHIP-1 was recently reported as a target of miR155 specifically in MDSC expansion [4], these results suggest both SHIP-1 and SOCS1 as target genes of miR-155 during functional MDSC generation. SOCS1 also restricted arginase I activity [5], which otherwise would limit the efficiency of MDSC proinflammatory responses. Indeed, we showed that
MicroRNA是一种小的非编码RNA,可以通过与mRNA分子互补序列的碱基配对导致翻译抑制或靶标降解。microRNA -155 (miR-155)是研究最多的microRNA之一,也是最早被报道致癌的microRNA[1]。miR-155在一长串血液学和实体肿瘤中都过表达,在癌症诊断和预后中至关重要。然而,特别是在宿主免疫系统中miR-155如何调节肿瘤进展仍然知之甚少。我们的研究强调了miR-155通过肿瘤内不同的免疫亚群调节肿瘤生长和肿瘤免疫的背景作用[2]。我们得出结论,受miR-155调节的免疫细胞群之间的不同作用的平衡似乎决定了miR-155是促进还是抑制肿瘤生长[2]。我们证明了宿主miR-155缺乏在多种移植肿瘤模型中促进抗肿瘤T细胞免疫。对免疫细胞区室的进一步分析表明,miR-155是肿瘤微环境中髓源性抑制细胞(MDSC)积累和抑制功能所必需的。除了直接调节MDSC外,miR-155还需要用于MDSC介导的CD4+Foxp3+调节性T细胞(Treg)诱导。另一方面,miR-155缺失阻碍了树突状细胞和T细胞的抗肿瘤反应。因此,在我们的肿瘤模型中,miR-155介导了MDSC的显性免疫抑制作用,导致miR-155缺陷宿主的整体抗肿瘤免疫增强。当偶氮甲烷(AOM)和葡聚糖硫酸钠(DSS)联合诱导结肠病变时,miR-155缺陷小鼠的结肠炎症减少,结直肠癌发生减少。此外,miR-155在来自肿瘤宿主的MDSC或由GM-CSF和IL-6从骨髓祖细胞产生的MDSC中上调。这些结果支持了miR-155是连接炎症和癌症发展的典型microRNA的观点[3]。尽管miR-155可能以内在方式调节肿瘤生长,但很可能炎症通过miR-155的增加促进功能性MDSC的积累,从而抑制免疫监视和抗肿瘤免疫,从而促进肿瘤生长。为了确定miR-155调节MDSC的分子机制(图(Figure1),1),我们发现miR155通过抑制SOCS1保留了MDSCs的抑制活性。此外,在MDSC中miR-155表达与SHIP-1/SOCS1表达之间建立了负相关。由于SHIP-1最近被报道为miR155在MDSC扩增过程中的特异性靶标[4],这些结果表明,在功能性MDSC生成过程中,SHIP-1和SOCS1都是miR-155的靶基因。SOCS1还限制精氨酸酶I的活性[5],否则会限制MDSC促炎反应的效率。事实上,我们发现miR-155−/−MDSC的精氨酸酶活性水平低于WT对应物,并且用特异性抑制剂抑制精氨酸酶-1完全消除了WT MDSC的抑制活性,并且不影响miR-155−/−MDSC。我们的数据表明,miR-155可能通过靶向SOCS1来调节精氨酸酶依赖的MDSC抑制功能。更有趣的是,我们观察到miR-155−/−MDSC中MMP-9和VEGF的产生减少,这可能会限制肿瘤血管生成。考虑到癌细胞表达miR-155对肿瘤血管生成的贡献[6],进一步的研究将确定miR-155是否通过癌细胞和肿瘤内的MDSC调节肿瘤血管生成。值得注意的是,我们关于宿主miR155缺失和肿瘤生长的研究结果与近期其他研究不同[7,8]。所使用的肿瘤细胞系的差异可能会改变肿瘤微环境中个体免疫细胞亚群的积累,这可能解释了这种差异。主要免疫群体的程度和调节在不同的肿瘤类型和/或肿瘤分期可能有所不同。因此,miR-155的增加可能是平衡肿瘤内抗和促肿瘤免疫成分的关键参与者。在我们给定的肿瘤模型系统中,我们提供了明确的证据表明miR-155以mdsc依赖的方式促进肿瘤生长,这通过体内的“耗竭”和“转移”策略得到了体现。综上所述,我们的研究强调了仔细评估miR-155在不同免疫细胞亚群中的内在作用的本质,其中miR-155可能对抗肿瘤免疫具有保护作用或有害作用。因此,开发细胞特异性的抗mir -155癌症治疗方法是安全且重要的。越来越明显的是,miR-155作为“OncomiR”与“ImmunomiR”协同作用,协调癌症的生长和进展。
{"title":"MicroRNA-155 regulates tumor myeloid-derived suppressive cells","authors":"Siqi Chen, Yi Zhang, Bin Zhang","doi":"10.18632/ONCOSCIENCE.269","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.269","url":null,"abstract":"MicroRNA is small non-coding RNA and can lead to translational repression or target degradation by base-pairing with complementary sequences of mRNA molecules. MicroRNA-155 (miR-155), one of the most studied microRNA, is the first one to be reported as oncogenic [1]. miR-155 is over expressed in a long list of both hematological and solid tumors and is of paramount importance in cancer diagnosis and prognosis. However, how miR-155 particularly in host immune system regulates the tumor progression remains poorly understood. Our study underscores a contextual role of miR-155 in regulating tumor growth and tumor immunity via distinct immune subsets within tumors [2]. We conclude that the balance of different effects between those immune cell populations, which are regulated by miR-155, appears to determine whether miR-155 promotes or inhibits tumor growth [2]. \u0000 \u0000We demonstrated that host miR-155 deficiency promoted antitumor T cell immunity in multiple transplanted tumor models. Further analysis of immune cell compartments revealed that miR-155 was required for the accumulation and suppressive function of myeloid-derived suppressive cells (MDSC) in the tumor microenvironment. Apart from the direct modulation on MDSC, miR-155 was also required for the MDSC-mediated CD4+Foxp3+ regulatory T cells (Treg) induction. On the other hand, miR-155 deficiency hampered the antitumor responses of both dendritic cells and T cells. Therefore, it appears that in our tumor models, miR-155 mediated a dominant immunosuppressive effect by MDSC, leading to the enhanced overall antitumor immunity in miR-155 deficient hosts. \u0000 \u0000Reduced colon inflammation and decreased colorectal carcinogenesis were also found in miR-155 deficient mice when azoxymethane (AOM) and dextran sodium sulphate (DSS) were combined to induce colon lesions. Furthermore, miR-155 was upregulated in MDSC either from tumor-bearing hosts or generated from bone marrow progenitors by GM-CSF and IL-6. These results support the notion that miR-155 is a prototypical microRNA bridging inflammation and cancer development [3]. Although miR-155 may regulate tumor growth in an intrinsic manner, it is likely that inflammation promotes the accumulation of functional MDSC by increased miR-155 that dampens the immune surveillance and antitumor immunity, thereby facilitating tumor growth. \u0000 \u0000To identify the molecular mechanisms by which miR-155 regulates MDSC (Figure ​(Figure1),1), we found that miR155 retained the suppressive activity of MDSCs through inhibiting SOCS1. Moreover, inverse correlations between miR-155 expression and SHIP-1/SOCS1 expression were established in MDSC. As SHIP-1 was recently reported as a target of miR155 specifically in MDSC expansion [4], these results suggest both SHIP-1 and SOCS1 as target genes of miR-155 during functional MDSC generation. SOCS1 also restricted arginase I activity [5], which otherwise would limit the efficiency of MDSC proinflammatory responses. Indeed, we showed that ","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"21 1","pages":"910 - 911"},"PeriodicalIF":0.0,"publicationDate":"2015-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89357128","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: 1
EGFR mutations in sinonasal squamous tumors: oncogenic and therapeutic implications 鼻窦鳞状肿瘤中的EGFR突变:致癌和治疗意义
Pub Date : 2015-11-19 DOI: 10.18632/ONCOSCIENCE.268
A. Udager, J. Mchugh, K. Elenitoba-Johnson, N. Brown
Sinonasal squamous cell carcinoma (SNSCC) and sinonasal papillomas constitute a diverse group of epithelial tumors arising in the sinonasal tract [1]. Sinonasal papillomas are benign tumors classified into three distinct histologic types: exophytic (fungiform), inverted, and oncocytic. While exophytic sinonasal papillomas (ESP) arise from the nasal septum and are only rarely associated with SNSCC, inverted sinonasal papillomas (ISP) and oncocytic sinonasal papillomas (OSP) typically arise from the lateral portion of the nasal cavity and are more frequently associated with synchronous or metachronous SNSCC – up to 25%, depending on the study [2]. The etiology of sinonasal tumors is a topic of current debate [1]. While ESP is associated with infection by low-risk human papillomavirus (HPV) in 55% - 65% [1, 3], most studies have demonstrated significantly lower HPV detections rates for ISP [3, 4]. Similarly, less than half of SNSCC are associated with HPV infection [3, 4] and the incidence in SNSCC associated with ISP may be even lower [5]. These data suggest that while HPV infection may play a role in the pathogenesis of a subset of these tumors, it is not the only factor involved in SNP and SNSCC oncogenesis. In a recent study, our group identified activating somatic EGFR mutations in 88% of ISP and 77% of SNSCC associated with ISP [6]. Importantly, while a variety of different EGFR mutations were found in these tumors, concordant EGFR genotypes were identified for all matched pairs of ISP and synchronous or metachronous SNSCC. Therefore, this study provided the first molecular evidence to support the role of ISP as a precursor lesion for SNSCC. In addition, EGFR mutation status was a significant prognostic factor for ISP, with EGFR wild-type tumors showing earlier progression to SNSCC. No EGFR mutations were identified in ESP, OSP, or SNSCC not associated with ISP, suggesting that the ISP/SNSCC disease spectrum is biologically distinct from these other sinonasal squamous tumors. The oncogenic role of EGFR mutations was supported in this study by functional experiments. In cell lines derived from ISP-associated SNSCC, EGFR mutations were shown to result in activation of EGFR as well as downstream constituents of the MAPK and PI3K/AKT/mTOR signaling pathways. Taken together, these functional studies and the high frequency of EGFR mutations suggest that dysregulated EGFR signaling plays a central role in the oncogenesis of ISP and associated SNSCC – a finding that is an apparent departure from prior paradigms involving HPV infection [1]. These findings, however, do not strictly preclude a role for HPV in these tumors. The HPV-associated E5 oncoprotein has been shown to inhibit EGFR degradation, alter endosomal trafficking of EGFR and activate proteins downstream of EGFR in a ligand-independent manner [7] (Figure ​(Figure1).1). Thus, it is plausible that altered EGFR signaling itself – either as a result of somatic activating EGFR mutations
鼻窦鳞状细胞癌(SNSCC)和鼻窦乳头状瘤构成了一组起源于鼻窦的上皮性肿瘤。鼻乳头状瘤是良性肿瘤,分为三种不同的组织学类型:外生性(真菌状)、倒置型和嗜瘤细胞型。外生性鼻窦乳头状瘤(ESP)起源于鼻中隔,很少与SNSCC相关,而内翻性鼻窦乳头状瘤(ISP)和嗜瘤性鼻窦乳头状瘤(OSP)通常起源于鼻腔外侧,更常与同步或异时性SNSCC相关——根据研究范围的不同,这一比例高达25%。鼻窦肿瘤的病因是目前争论的一个话题。虽然ESP与低风险人乳头瘤病毒(HPV)感染的相关性为55% - 65%[1,3],但大多数研究表明,ISP的HPV检出率明显较低[3,4]。同样,不到一半的SNSCC与HPV感染相关[3,4],而与ISP相关的SNSCC的发病率可能更低。这些数据表明,虽然HPV感染可能在这些肿瘤的一个亚群的发病机制中发挥作用,但它并不是SNP和SNSCC肿瘤发生的唯一因素。在最近的一项研究中,我们的研究小组在88%的ISP和77%的SNSCC中发现了与ISP[6]相关的激活体细胞EGFR突变。重要的是,虽然在这些肿瘤中发现了各种不同的EGFR突变,但在所有匹配的ISP和同步或异时SNSCC中发现了一致的EGFR基因型。因此,本研究为支持ISP作为SNSCC前体病变的作用提供了第一个分子证据。此外,EGFR突变状态是ISP的一个重要预后因素,EGFR野生型肿瘤更早发展为SNSCC。在与ISP无关的ESP、OSP或SNSCC中未发现EGFR突变,这表明ISP/SNSCC疾病谱系在生物学上与这些其他鼻窦鳞状肿瘤不同。本研究通过功能实验证实了EGFR突变的致癌作用。在来自isp相关SNSCC的细胞系中,EGFR突变被证明可以激活EGFR以及MAPK和PI3K/AKT/mTOR信号通路的下游成分。综上所述,这些功能性研究和EGFR突变的高频率表明,EGFR信号失调在ISP和相关SNSCC的肿瘤发生中起着核心作用——这一发现明显偏离了先前涉及HPV感染的范式。然而,这些发现并不能完全排除HPV在这些肿瘤中的作用。hpv相关的E5癌蛋白已被证明可以抑制EGFR降解,改变EGFR的内体运输,并以不依赖配体的方式激活EGFR下游的蛋白质[7](图(图1))。因此,似乎改变的EGFR信号本身——无论是体细胞激活的EGFR突变还是hpv相关的E5癌蛋白——在ISP的发展和进化中很重要。虽然现在有明确的证据支持ISP作为SNSCC的前体的作用,但其进展机制仍不明确。良性肿瘤的进展往往受到癌基因诱导的衰老的限制,而逃避衰老和恶性转化往往与肿瘤抑制蛋白(如p53或Rb)的功能丧失有关。这些肿瘤抑制因子在癌症中的失活通常是体细胞突变的结果;然而,hpv相关的E6和E7蛋白破坏p53和Rb功能是[1]的另一个潜在机制(图(图1))。未来的研究需要将EGFR突变与HPV感染状态联系起来,并阐明从ISP到SNSCC的进展机制。手术切除和放疗是目前SNSCC的治疗选择,化疗通常用于局部晚期或转移性SNSCC。然而,在这些治疗方案中,SNSCC与40%的5年死亡率相关。在ISP和相关SNSCC中发现EGFR突变可能为这些肿瘤的首次靶向治疗提供了机会。目前,EGFR抑制剂治疗是EGFR突变晚期肺癌的标准治疗方法。与EGFR外显子19缺失的肺癌相比,外显子20插入的肺癌通常对目前可用的可逆EGFR抑制剂吉非替尼和厄洛替尼具有耐药性。然而,不可逆的EGFR抑制剂已被证明对外显子20突变的肺癌具有更强的体外作用,探索其治疗潜力的临床试验正在进行中。
{"title":"EGFR mutations in sinonasal squamous tumors: oncogenic and therapeutic implications","authors":"A. Udager, J. Mchugh, K. Elenitoba-Johnson, N. Brown","doi":"10.18632/ONCOSCIENCE.268","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.268","url":null,"abstract":"Sinonasal squamous cell carcinoma (SNSCC) and sinonasal papillomas constitute a diverse group of epithelial tumors arising in the sinonasal tract [1]. Sinonasal papillomas are benign tumors classified into three distinct histologic types: exophytic (fungiform), inverted, and oncocytic. While exophytic sinonasal papillomas (ESP) arise from the nasal septum and are only rarely associated with SNSCC, inverted sinonasal papillomas (ISP) and oncocytic sinonasal papillomas (OSP) typically arise from the lateral portion of the nasal cavity and are more frequently associated with synchronous or metachronous SNSCC – up to 25%, depending on the study [2]. \u0000 \u0000The etiology of sinonasal tumors is a topic of current debate [1]. While ESP is associated with infection by low-risk human papillomavirus (HPV) in 55% - 65% [1, 3], most studies have demonstrated significantly lower HPV detections rates for ISP [3, 4]. Similarly, less than half of SNSCC are associated with HPV infection [3, 4] and the incidence in SNSCC associated with ISP may be even lower [5]. These data suggest that while HPV infection may play a role in the pathogenesis of a subset of these tumors, it is not the only factor involved in SNP and SNSCC oncogenesis. \u0000 \u0000In a recent study, our group identified activating somatic EGFR mutations in 88% of ISP and 77% of SNSCC associated with ISP [6]. Importantly, while a variety of different EGFR mutations were found in these tumors, concordant EGFR genotypes were identified for all matched pairs of ISP and synchronous or metachronous SNSCC. Therefore, this study provided the first molecular evidence to support the role of ISP as a precursor lesion for SNSCC. In addition, EGFR mutation status was a significant prognostic factor for ISP, with EGFR wild-type tumors showing earlier progression to SNSCC. No EGFR mutations were identified in ESP, OSP, or SNSCC not associated with ISP, suggesting that the ISP/SNSCC disease spectrum is biologically distinct from these other sinonasal squamous tumors. \u0000 \u0000The oncogenic role of EGFR mutations was supported in this study by functional experiments. In cell lines derived from ISP-associated SNSCC, EGFR mutations were shown to result in activation of EGFR as well as downstream constituents of the MAPK and PI3K/AKT/mTOR signaling pathways. Taken together, these functional studies and the high frequency of EGFR mutations suggest that dysregulated EGFR signaling plays a central role in the oncogenesis of ISP and associated SNSCC – a finding that is an apparent departure from prior paradigms involving HPV infection [1]. These findings, however, do not strictly preclude a role for HPV in these tumors. The HPV-associated E5 oncoprotein has been shown to inhibit EGFR degradation, alter endosomal trafficking of EGFR and activate proteins downstream of EGFR in a ligand-independent manner [7] (Figure ​(Figure1).1). Thus, it is plausible that altered EGFR signaling itself – either as a result of somatic activating EGFR mutations ","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"7 1","pages":"908 - 909"},"PeriodicalIF":0.0,"publicationDate":"2015-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78435473","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: 8
An added dimension to tumour TRAIL sensitivity 增加了肿瘤TRAIL敏感性的维度
Pub Date : 2015-11-18 DOI: 10.18632/ONCOSCIENCE.267
Sandra Healy, L. O’Leary, E. Szegezdi
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF cytokine family and a selective inducer of apoptosis in a range of tumour cells, but not in healthy normal, untransformed cells. It is expressed by natural killer cells and natural killer-T cells when they encounter malignantly transformed cells and it is a key effector molecule in tumour immune surveillance. TRAIL has 5 receptors, which is the highest receptor promiscuity in the TNF ligand family. It binds to death receptor 4 (DR4) or DR5 on the surface of target cells [1] and initiates a conformational change which promotes association of the receptors with FADD facilitating pro-caspase-8 and/or pro-caspase-10 recruitment which then activates effector caspases to execute cell death [2]. Signalling through DR4 and DR5 can also activate pro-inflammatory intracellular molecules such as MAPK, PKB and NF-κB and overexpression of DR4 or DR5 has been shown to stimulate the release of inflammatory cytokines [3]. However, TRAIL also has three regulatory receptors. Two of these, decoy receptor 1(DcR1) and DcR2 are membrane bound and the third regulatory receptor, osteoprotegerin is a secreted protein. DcRs regulate TRAIL-induced apoptosis by either sequestering TRAIL from the death receptors or by forming inactive, heteromeric DcR1/2–DR4/5 complexes [1]. Indeed, DcRs have been shown to be highly expressed in a number of tumour tissues such as acute myeloid leukaemia, prostate cancer and breast cancer and their expression is linked with poor prognosis [4]. However DcR expression in tumour cells does not correlate with TRAIL sensitivity and non-transformed cells do not require DcRs to be protected from TRAIL-induced apoptosis, suggesting that the in vivo role of the DcRs may be more complex than originally thought [5] The tumor-specific cytotoxicity of TRAIL has been exploited as a therapeutic strategy by utilizing recombinant versions of TRAIL and agonistic antibodies against DR4 and DR5 [6]. While recombinant soluble human TRAIL was highly potent against a broad range of tumours in vitro and in pre-clinical studies, in clinical trials TRAIL has failed to exhibit the same potency [6]. One of the major shortcomings of the preclinical models was the lack of assessment of the contribution of the tumour microenvironment (TME). The TME consists of various cell types, soluble factors and signals from the extracellular matrix, and is in a reciprocal interaction with the tumour cells. It is thus important to understand the interplay of different cell types in the tumour microenvironment and the effect of the factors they express and secrete on tumour growth, development and resistance to therapy. Figure 1 Cell autonomous and supracellular regulation of TRAIL-sensitivity by decoy receptor 1 (DcR1) and -2 The study by O'Leary and colleagues explored the hypothesis that DcRs exerted a ‘supracellular level control’ of TRAIL-sensitivity rather than simply regula
肿瘤坏死因子相关凋亡诱导配体(TRAIL)是TNF细胞因子家族的一员,是多种肿瘤细胞的选择性凋亡诱导剂,但在健康的正常未转化细胞中不存在。它由自然杀伤细胞和自然杀伤t细胞在遇到恶性转化细胞时表达,是肿瘤免疫监视的关键效应分子。TRAIL有5个受体,是TNF配体家族中受体混杂性最高的。它与靶细胞表面的死亡受体4 (DR4)或DR5结合[1],并引发构象变化,促进受体与FADD的结合,促进前caspase-8和/或前caspase-10的募集,然后激活效应caspase执行细胞死亡[2]。通过DR4和DR5的信号传导还可以激活细胞内促炎分子如MAPK、PKB和NF-κB,并且DR4或DR5的过表达已被证明可以刺激炎症细胞因子的释放[3]。然而,TRAIL也有三种调节受体。其中两个,诱饵受体1(DcR1)和DcR2是膜结合的,第三个调节受体,骨保护素是一种分泌蛋白。DcRs通过从死亡受体中分离TRAIL或形成非活性的异聚DcR1/2-DR4/5复合物来调节TRAIL诱导的细胞凋亡[1]。事实上,DcRs已被证明在许多肿瘤组织中高表达,如急性髓性白血病、前列腺癌和乳腺癌,其表达与预后不良有关[4]。然而,肿瘤细胞中的DcR表达与TRAIL敏感性无关,非转化细胞不需要保护DcR免受TRAIL诱导的细胞凋亡,这表明DcR在体内的作用可能比最初认为的要复杂[5]。TRAIL的肿瘤特异性细胞毒性已被利用为一种治疗策略,利用TRAIL的重组版本和针对DR4和DR5的激动抗体[6]。虽然在体外和临床前研究中,重组可溶性人TRAIL对多种肿瘤具有很强的效力,但在临床试验中,TRAIL未能表现出相同的效力[6]。临床前模型的主要缺点之一是缺乏对肿瘤微环境(TME)的贡献的评估。TME由各种细胞类型、可溶性因子和来自细胞外基质的信号组成,并与肿瘤细胞相互作用。因此,了解肿瘤微环境中不同细胞类型的相互作用以及它们表达和分泌的因子对肿瘤生长、发展和治疗耐药性的影响是很重要的。诱骗受体1 (DcR1)和-2对TRAIL敏感性的细胞自主和超细胞调控O' leary及其同事的研究探索了DcRs对TRAIL敏感性的“超细胞水平调控”,而不是简单地在细胞自主水平上调控TRAIL抗性的假设[7]。他们首先检测了诱饵受体在肿瘤细胞、肿瘤基质和非恶性肿瘤邻近组织中的表达。有趣的是,他们发现DcR1和DcR2在组织中普遍表达,但组织基质只表达DcR1。为了确定表达dcr的基质细胞是否影响共享相同微环境的肿瘤细胞的TRAIL敏感性,他们产生并表征了dcr不敏感的TRAIL突变体,并使用数学建模、基于细胞的测定和基质/肿瘤共培养系统的组合,使他们能够模拟相邻基质细胞表达的dcr对肿瘤细胞中DR4/5激活的影响。他们发现,基质dcr显著降低TRAIL诱导的DR4/DR5激活,并保护肿瘤细胞免受TRAIL的侵害。作者在三维混合细胞型(间质瘤)球形肿瘤模型中证实了这一发现。有趣的是,他们还观察到TNF增加了成纤维细胞中DcR的表面表达,这增加了炎症环境诱导DcR表达并促进TRAIL抵抗的可能性。这项研究清楚地表明,肿瘤微环境中的基质dcr可以发挥影响肿瘤细胞的跨细胞调节作用,并强调了开发治疗性TRAIL变体的重要性,这种变体可以选择性地激活两种诱导死亡的TRAIL受体,但不会被基质组织上存在的诱饵受体清除。未来的研究将有望建立一个前馈信号回路,驱动整体组织敏感性,以实现肿瘤根除。
{"title":"An added dimension to tumour TRAIL sensitivity","authors":"Sandra Healy, L. O’Leary, E. Szegezdi","doi":"10.18632/ONCOSCIENCE.267","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.267","url":null,"abstract":"Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF cytokine family and a selective inducer of apoptosis in a range of tumour cells, but not in healthy normal, untransformed cells. It is expressed by natural killer cells and natural killer-T cells when they encounter malignantly transformed cells and it is a key effector molecule in tumour immune surveillance. TRAIL has 5 receptors, which is the highest receptor promiscuity in the TNF ligand family. It binds to death receptor 4 (DR4) or DR5 on the surface of target cells [1] and initiates a conformational change which promotes association of the receptors with FADD facilitating pro-caspase-8 and/or pro-caspase-10 recruitment which then activates effector caspases to execute cell death [2]. Signalling through DR4 and DR5 can also activate pro-inflammatory intracellular molecules such as MAPK, PKB and NF-κB and overexpression of DR4 or DR5 has been shown to stimulate the release of inflammatory cytokines [3]. However, TRAIL also has three regulatory receptors. Two of these, decoy receptor 1(DcR1) and DcR2 are membrane bound and the third regulatory receptor, osteoprotegerin is a secreted protein. DcRs regulate TRAIL-induced apoptosis by either sequestering TRAIL from the death receptors or by forming inactive, heteromeric DcR1/2–DR4/5 complexes [1]. Indeed, DcRs have been shown to be highly expressed in a number of tumour tissues such as acute myeloid leukaemia, prostate cancer and breast cancer and their expression is linked with poor prognosis [4]. However DcR expression in tumour cells does not correlate with TRAIL sensitivity and non-transformed cells do not require DcRs to be protected from TRAIL-induced apoptosis, suggesting that the in vivo role of the DcRs may be more complex than originally thought [5] \u0000 \u0000The tumor-specific cytotoxicity of TRAIL has been exploited as a therapeutic strategy by utilizing recombinant versions of TRAIL and agonistic antibodies against DR4 and DR5 [6]. While recombinant soluble human TRAIL was highly potent against a broad range of tumours in vitro and in pre-clinical studies, in clinical trials TRAIL has failed to exhibit the same potency [6]. One of the major shortcomings of the preclinical models was the lack of assessment of the contribution of the tumour microenvironment (TME). The TME consists of various cell types, soluble factors and signals from the extracellular matrix, and is in a reciprocal interaction with the tumour cells. It is thus important to understand the interplay of different cell types in the tumour microenvironment and the effect of the factors they express and secrete on tumour growth, development and resistance to therapy. \u0000 \u0000 \u0000 \u0000Figure 1 \u0000 \u0000Cell autonomous and supracellular regulation of TRAIL-sensitivity by decoy receptor 1 (DcR1) and -2 \u0000 \u0000 \u0000 \u0000The study by O'Leary and colleagues explored the hypothesis that DcRs exerted a ‘supracellular level control’ of TRAIL-sensitivity rather than simply regula","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"9 1","pages":"906 - 907"},"PeriodicalIF":0.0,"publicationDate":"2015-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88893847","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: 4
Runx1: a new driver in neurofibromagenesis Runx1:神经纤维生成的新驱动因子
Pub Date : 2015-11-17 DOI: 10.18632/ONCOSCIENCE.266
Jianqiang Wu, Gang Huang, N. Ratner
The Runt-related transcription factor-1 (RUNX1 or AML1) encodes a transcription factor that serves as a master developmental regulator. It is important for hematopoiesis, angiogenesis, maturation of megakaryocytes, and differentiation of T and B cells [1]. Runx1 is also important for neuronal development and glial cell differentiation [2]. Runx1 is integrated into a complex regulatory network which acts both at the transcriptional and post-transcriptional levels. Runx1 activity can be regulated by several posttranslational modifications, including phosphorylation, de-phosphorylation, SUMOylation, acetylation, methylation and ubiquitination. These modifications control various aspects of transcriptional factors' activities such as auto inhibition, dimerization and ubiquitin-mediated degradation [3]. Besides its developmental determination role, RUNX1 is involved in malignant tumor formation. Reports have shown that RUNX1 is frequently de-regulated and has paradoxical effects in human cancers, in which it can function either as a tumor suppressor or oncogene [3, 4]. RUNX1 has been implicated as a tumor suppressor in several solid tumors including breast cancer, esophageal adenocarcinoma, colon cancer and possibly prostate cancer but acts as an oncogene in head/neck squamous cell carcinomas, endometrial cancer, and epithelial cancer [3, 4]. Because Runx1 is a sequence specific DNA-binding transcription factor, whether it functions as oncogene or tumor suppressor is dependent on its interaction with specific co-regulatory proteins. We recently showed that RUNX1 acts as an oncogene in the context of loss of neurofibromatosis type 1 (Nf1). Instead of chromosomal translocation and mutation frequently detected in other cancers, Runx1 is overexpressed in human and mouse neurofibroma-initiating cells, both at the messenger RNA and protein levels. Specifically, loss of Nf1 increases number of embryonic day 12.5 Runx1+/Blbp+ Schwann cell progenitors that enable neurofibroma formation in a mouse model (Figure ​(Figure1).1). Targeted genetic deletion of RUNX1 in Schwann cells and Schwann cell progenitors delays mouse neurofibroma formation in vivo (5). Figure 1 Model of neurofibromagenesis It is not clear how loss of Nf1 induces Runx1 overexpression and serves as an oncogene. There are several potential possibilities: 1) NF1 is known to encode a Ras-GTPase activating protein (Ras-GAP) and the Ras-MEK-ERK pathway is important for Nf1 neurofibroma formation [6]. Runx1 may be phosphorylated by the elevated MEK signaling to initiate the tumor formation process. 2) Elevated Wnt or Notch signaling can directly or indirectly activate Runx1, which can accelerate G1-S transition and stimulates cell proliferation. Consistently, our results show that loss of Runx1 in Schwann cells decreased cell proliferation by activating Trp53-p21 or increased cell apoptosis by inhibiting anti-apoptotic gene Bcl-2 in the context of Nf1−/− Schwann cell environment
runt相关转录因子-1 (RUNX1或AML1)编码一种转录因子,作为主要的发育调节因子。它对造血、血管生成、巨核细胞成熟以及T细胞和B细胞分化具有重要作用[1]。Runx1在神经元发育和胶质细胞分化中也很重要[2]。Runx1被整合到一个复杂的调控网络中,该网络在转录和转录后水平上都起作用。Runx1的活性可以通过多种翻译后修饰来调节,包括磷酸化、去磷酸化、SUMOylation、乙酰化、甲基化和泛素化。这些修饰控制转录因子活性的各个方面,如自抑制、二聚化和泛素介导的降解[3]。RUNX1除了具有发育决定作用外,还参与恶性肿瘤的形成。有报道表明,RUNX1经常被去调控,并在人类癌症中具有矛盾的作用,在这种作用中,它既可以作为肿瘤抑制因子,也可以作为致癌基因[3,4]。RUNX1在包括乳腺癌、食管癌、结肠癌和可能的前列腺癌在内的几种实体肿瘤中被认为是肿瘤抑制因子,但在头颈部鳞状细胞癌、子宫内膜癌和上皮癌中作为癌基因[3,4]。由于Runx1是一种序列特异性的dna结合转录因子,所以它是作为致癌基因还是肿瘤抑制因子,取决于它与特异性共调控蛋白的相互作用。我们最近发现RUNX1在1型神经纤维瘤病(Nf1)缺失的情况下作为致癌基因。与在其他癌症中经常检测到的染色体易位和突变不同,Runx1在人和小鼠神经纤维瘤起始细胞中在信使RNA和蛋白质水平上都过表达。具体来说,Nf1的缺失增加了小鼠模型中能够形成神经纤维瘤的Runx1+/Blbp+雪旺细胞祖细胞的胚胎天数12.5天(图1)。雪旺细胞和雪旺细胞祖细胞中RUNX1基因的靶向缺失延缓了小鼠体内神经纤维瘤的形成(5)。图1神经纤维增生模型。目前尚不清楚Nf1缺失如何诱导RUNX1过表达并作为致癌基因。有几种潜在的可能性:1)已知NF1编码Ras-GTPase激活蛋白(Ras-GAP), Ras-MEK-ERK通路对NF1神经纤维瘤的形成很重要[6]。Runx1可能通过MEK信号的升高而磷酸化,从而启动肿瘤的形成过程。2) Wnt或Notch信号升高可直接或间接激活Runx1,加速G1-S转变,刺激细胞增殖。同样,我们的研究结果表明,在Nf1−/−雪旺细胞环境下,Runx1在雪旺细胞中的缺失通过激活Trp53-p21来降低细胞增殖,或者通过抑制抗凋亡基因Bcl-2来增加细胞凋亡。神经纤维瘤细胞内Runx1的升高可能通过Trp53-p21改变细胞命运(即增殖或分化)。需要进一步的实验来确定Runx1fl/fl、Nf1fl/fl、Bcl-2如何被激活或失活。Dhhre肿瘤。3) Runx1可能与染色质重塑复合体、SWI/SNF或多梳抑制复合体等表观遗传调控因子相互作用,通过翻译后修饰影响Runx1活性[7]。总的来说,我们的研究支持Runx1在神经纤维瘤发生和/或维持中的致癌作用,但其潜在机制尚不清楚。随着对RUNX1生物学认识的不断增加,靶向转录因子RUNX1或RUNX1通路可能为神经纤维瘤及其他RUNX1过表达肿瘤提供新的治疗方法。
{"title":"Runx1: a new driver in neurofibromagenesis","authors":"Jianqiang Wu, Gang Huang, N. Ratner","doi":"10.18632/ONCOSCIENCE.266","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.266","url":null,"abstract":"The Runt-related transcription factor-1 (RUNX1 or AML1) encodes a transcription factor that serves as a master developmental regulator. It is important for hematopoiesis, angiogenesis, maturation of megakaryocytes, and differentiation of T and B cells [1]. Runx1 is also important for neuronal development and glial cell differentiation [2]. Runx1 is integrated into a complex regulatory network which acts both at the transcriptional and post-transcriptional levels. Runx1 activity can be regulated by several posttranslational modifications, including phosphorylation, de-phosphorylation, SUMOylation, acetylation, methylation and ubiquitination. These modifications control various aspects of transcriptional factors' activities such as auto inhibition, dimerization and ubiquitin-mediated degradation [3]. \u0000 \u0000Besides its developmental determination role, RUNX1 is involved in malignant tumor formation. Reports have shown that RUNX1 is frequently de-regulated and has paradoxical effects in human cancers, in which it can function either as a tumor suppressor or oncogene [3, 4]. RUNX1 has been implicated as a tumor suppressor in several solid tumors including breast cancer, esophageal adenocarcinoma, colon cancer and possibly prostate cancer but acts as an oncogene in head/neck squamous cell carcinomas, endometrial cancer, and epithelial cancer [3, 4]. Because Runx1 is a sequence specific DNA-binding transcription factor, whether it functions as oncogene or tumor suppressor is dependent on its interaction with specific co-regulatory proteins. \u0000 \u0000We recently showed that RUNX1 acts as an oncogene in the context of loss of neurofibromatosis type 1 (Nf1). Instead of chromosomal translocation and mutation frequently detected in other cancers, Runx1 is overexpressed in human and mouse neurofibroma-initiating cells, both at the messenger RNA and protein levels. Specifically, loss of Nf1 increases number of embryonic day 12.5 Runx1+/Blbp+ Schwann cell progenitors that enable neurofibroma formation in a mouse model (Figure ​(Figure1).1). Targeted genetic deletion of RUNX1 in Schwann cells and Schwann cell progenitors delays mouse neurofibroma formation in vivo (5). \u0000 \u0000 \u0000 \u0000Figure 1 \u0000 \u0000Model of neurofibromagenesis \u0000 \u0000 \u0000 \u0000It is not clear how loss of Nf1 induces Runx1 overexpression and serves as an oncogene. There are several potential possibilities: 1) NF1 is known to encode a Ras-GTPase activating protein (Ras-GAP) and the Ras-MEK-ERK pathway is important for Nf1 neurofibroma formation [6]. Runx1 may be phosphorylated by the elevated MEK signaling to initiate the tumor formation process. 2) Elevated Wnt or Notch signaling can directly or indirectly activate Runx1, which can accelerate G1-S transition and stimulates cell proliferation. Consistently, our results show that loss of Runx1 in Schwann cells decreased cell proliferation by activating Trp53-p21 or increased cell apoptosis by inhibiting anti-apoptotic gene Bcl-2 in the context of Nf1−/− Schwann cell environment","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"379 1","pages":"904 - 905"},"PeriodicalIF":0.0,"publicationDate":"2015-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76614638","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: 3
Kinetochore-microtube attachments in cancer therapy 着丝细胞-微管附件在癌症治疗中的应用
Pub Date : 2015-11-16 DOI: 10.18632/ONCOSCIENCE.265
D. Del Bufalo, F. Degrassi
The process of cell division represents an extraordinary target to develop antitumor therapies. Indeed, a large number of clinically relevant anti-cancer drugs, such as taxanes and vinca alkaloids, target mitosis by stimulating or inhibiting microtubule (MT) polymerization. During the past decades anti-tubulin drugs have proven very effective against a wide range of tumors. However, collateral effects, such as myelosuppression and MT disruption in non-dividing tissues, including brain, are common. Recently, the increased understanding of the cell division process and the identification of several signaling pathways controlling mitosis have provided novel opportunities for cancer drug discovery. Consequently, mitotic proteins have become attractive targets to develop molecular cancer therapeutics. In this scenario, kinetochores (KTs) represent an attractive therapeutic target in light of their fundamental role in driving chromosome segregation and controlling chromosome segregation errors. Indeed, cells require a fine regulation of the kinetochore-microtubule (KT-MT) attachment stability to prevent chromosome instability, and KT-MT attachment dynamics is often deregulated in tumour cells [2]. Chromosome instability is commonly accepted as a driving force in the development of cancer, but more recent work has demonstrated that extensive chromosome missegregation may be detrimental to cancer cells and act as a tumor suppression mechanism [3]. In light of this double role of chromosome instability in cancer, we have explored the hypothesis that interfering with KT-MT attachment dynamics could drive massive chromosome missegregation and kill tumor cells. Highly Expressed in Cancer protein 1 (Hec1) is a constituent of the evolutionary conserved Ndc80 complex, the molecular connector between KTs and MTs. Among the subunits of the Ndc80 complex, Hec1 directly interacts with MTs and regulates KT-MT dynamics and attachment stability [3]. Importantly, Hec1 is frequently overexpressed in cancer. We previously demonstrated that expression of Hec1 fused with the enhanced green fluorescent protein (EGFP) tag at its N-terminus (EGFP-Hec1), the protein domain that regulates MT attachment dynamics, led to a strong accumulation of this modified protein, which acted as a dominant negative mutant over the endogenous Hec1. Mitotic cells expressing a N-terminus tagged Hec1 accumulated lateral KT-MT attachments and underwent a spindle assembly checkpoint (SAC) dependent mitotic arrest associated with the formation of multipolar spindles [4]. We further showed that expression of an inducible N-terminus modified Hec1 completely abolished in vitro growth of EGFP-Hec1 expressing HeLa cells but had no effects on untransformed human fibroblasts or epithelial cells [5]. These in vitro cell-based data were validated in vivo by showing that inducible EGFP-Hec1 expression strongly inhibited tumor growth in a HeLa xenograft mouse model [5]. Strikingly, in both in vitro and in vivo
细胞分裂过程是开发抗肿瘤疗法的重要靶点。事实上,大量临床相关的抗癌药物,如紫杉烷和长春花生物碱,都是通过刺激或抑制微管(MT)聚合来靶向有丝分裂的。在过去的几十年里,抗微管蛋白药物已被证明对多种肿瘤非常有效。然而,附带效应,如骨髓抑制和MT破坏非分裂组织,包括脑,是常见的。最近,对细胞分裂过程的进一步了解和控制有丝分裂的几个信号通路的鉴定为癌症药物的发现提供了新的机会。因此,有丝分裂蛋白已成为开发分子癌症治疗的有吸引力的靶点。在这种情况下,着丝点(KTs)代表了一个有吸引力的治疗靶点,因为它们在驱动染色体分离和控制染色体分离错误方面起着基本作用。事实上,细胞需要对着丝点-微管(KT-MT)的附着稳定性进行精细调节,以防止染色体不稳定,而在肿瘤细胞中,KT-MT的附着动力学通常是不受调节的[2]。染色体不稳定被普遍认为是癌症发展的驱动力,但最近的研究表明,广泛的染色体错分离可能对癌细胞有害,并起到肿瘤抑制机制的作用[3]。鉴于染色体不稳定性在癌症中的双重作用,我们探索了干扰KT-MT附着动力学可能导致大量染色体错分离并杀死肿瘤细胞的假设。在Cancer protein 1 (Hec1)中高表达,是进化保守的Ndc80复合物的一个组成部分,Ndc80复合物是kt和mt之间的分子连接物,在Ndc80复合物的亚基中,Hec1直接与mt相互作用,调节KT-MT动力学和附着稳定性[3]。重要的是,Hec1在癌症中经常过表达。我们之前已经证明,Hec1在其n端与增强型绿色荧光蛋白(EGFP)标签(EGFP-Hec1)(调节MT附着动力学的蛋白质结构域)融合的表达导致这种修饰蛋白的强烈积累,作为内源性Hec1的显性负突变体。表达n端标记Hec1的有丝分裂细胞积累了侧向KT-MT附着,并经历了与多极纺锤体形成相关的纺锤体组装检查点(SAC)依赖的有丝分裂停滞[4]。我们进一步发现,诱导型n端修饰的Hec1的表达完全抑制了表达HeLa细胞的EGFP-Hec1的体外生长,但对未转化的人成纤维细胞或上皮细胞没有影响[5]。这些基于体外细胞的数据在体内得到验证,表明可诱导的EGFP-Hec1表达强烈抑制HeLa异种移植小鼠模型中的肿瘤生长[5]。引人注目的是,在体外和体内模型中,表达EGFP-Hec1的细胞在有丝分裂中永久停止并产生多极纺锤体。表达EGFP-Hec1的细胞的实时成像显示,多极纺锤体内染色体分离受损诱导有丝分裂灾难,通过诱导有丝分裂引起的凋亡死亡,或细胞分裂失败和多核来鉴定。最后,对MT通量率和KT周转率的测量表明,EGFP-Hec1增加了KT-MT附着的稳定性,这表明稳定KT-MT附着动力学是一种很有前途的治疗方法[5]。与KT-MT附着动力学是抗癌作用的分子靶点一致,在c端表达he1与EGFP融合,不影响KT-MT附着动力学,对癌细胞增殖没有显著影响[5]。总的来说,我们的研究结果表明,多极纺锤体内大量的染色体错分离可以通过激活有丝分裂突变过程来杀死肿瘤细胞。在我们的实验模型中,多极性的诱导是由细胞在前期的时间延长引起的,这促进了内聚疲劳(不协调的着丝粒内聚释放),并通过泄漏分离酶激活导致中心粒脱离,如图11所示[6,7]。在控制SAC沉默的蛋白质耗尽或抑制后期促进复合物/cdc20 (APC/C)活性后,内聚疲劳依赖性多极性诱导的癌细胞死亡已被证明(图(图1)),其中一些治疗方法被发现在避免有丝分裂滑移和产生癌细胞死亡方面比MT抑制剂更有效(7,8)。这些研究和我们的工作表明,刺激纺锤体多极可以作为一种抗癌策略,通过激活多极有丝分裂后的有丝分裂灾难。 此外,他们指出,靶向参与调节KT-MT附着动力学,纠正KT-MT错误附着或沉默纺锤体组装检查点的机制可能是抗癌策略发展的新前沿。
{"title":"Kinetochore-microtube attachments in cancer therapy","authors":"D. Del Bufalo, F. Degrassi","doi":"10.18632/ONCOSCIENCE.265","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.265","url":null,"abstract":"The process of cell division represents an extraordinary target to develop antitumor therapies. Indeed, a large number of clinically relevant anti-cancer drugs, such as taxanes and vinca alkaloids, target mitosis by stimulating or inhibiting microtubule (MT) polymerization. During the past decades anti-tubulin drugs have proven very effective against a wide range of tumors. However, collateral effects, such as myelosuppression and MT disruption in non-dividing tissues, including brain, are common. Recently, the increased understanding of the cell division process and the identification of several signaling pathways controlling mitosis have provided novel opportunities for cancer drug discovery. Consequently, mitotic proteins have become attractive targets to develop molecular cancer therapeutics. In this scenario, kinetochores (KTs) represent an attractive therapeutic target in light of their fundamental role in driving chromosome segregation and controlling chromosome segregation errors. Indeed, cells require a fine regulation of the kinetochore-microtubule (KT-MT) attachment stability to prevent chromosome instability, and KT-MT attachment dynamics is often deregulated in tumour cells [2]. Chromosome instability is commonly accepted as a driving force in the development of cancer, but more recent work has demonstrated that extensive chromosome missegregation may be detrimental to cancer cells and act as a tumor suppression mechanism [3]. In light of this double role of chromosome instability in cancer, we have explored the hypothesis that interfering with KT-MT attachment dynamics could drive massive chromosome missegregation and kill tumor cells. Highly Expressed in Cancer protein 1 (Hec1) is a constituent of the evolutionary conserved Ndc80 complex, the molecular connector between KTs and MTs. Among the subunits of the Ndc80 complex, Hec1 directly interacts with MTs and regulates KT-MT dynamics and attachment stability [3]. Importantly, Hec1 is frequently overexpressed in cancer. We previously demonstrated that expression of Hec1 fused with the enhanced green fluorescent protein (EGFP) tag at its N-terminus (EGFP-Hec1), the protein domain that regulates MT attachment dynamics, led to a strong accumulation of this modified protein, which acted as a dominant negative mutant over the endogenous Hec1. Mitotic cells expressing a N-terminus tagged Hec1 accumulated lateral KT-MT attachments and underwent a spindle assembly checkpoint (SAC) dependent mitotic arrest associated with the formation of multipolar spindles [4]. We further showed that expression of an inducible N-terminus modified Hec1 completely abolished in vitro growth of EGFP-Hec1 expressing HeLa cells but had no effects on untransformed human fibroblasts or epithelial cells [5]. These in vitro cell-based data were validated in vivo by showing that inducible EGFP-Hec1 expression strongly inhibited tumor growth in a HeLa xenograft mouse model [5]. Strikingly, in both in vitro and in vivo","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"43 1","pages":"902 - 903"},"PeriodicalIF":0.0,"publicationDate":"2015-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82653381","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
Snail1 controls cooperative cell plasticity during metastasis Snail1控制转移过程中的协同细胞可塑性
Pub Date : 2015-11-16 DOI: 10.18632/ONCOSCIENCE.262
J. Baulida
Mortality in cancer is strongly associated with the capacity of tumor cells to spread and critically affect other tissues and organs. Genetic mutations accumulated by tumor cells and cross-signaling between tumor and host cells underlie the formation of metastasis. Cancer-activated fibroblasts (CAFs), which are host fibroblasts activated by tumor signaling, can alter tumor cell behavior by both paracrine signaling (secreting diffusible molecules) and mechanical signaling (modifying the composition and organization of the stroma). These fibroblasts resemble myofibroblasts (MFs) of the granulation tissue generated during wound healing, which produce a rigid desmoplastic stroma rich in signaling molecules and cross-linked extracellular fibers. Desmoplasia favors malignant tumor cell properties such as mobility, stemness, and even resistance to pharmacological insults [1].
癌症的死亡率与肿瘤细胞扩散并严重影响其他组织和器官的能力密切相关。肿瘤细胞积累的基因突变和肿瘤与宿主细胞之间的交叉信号传导是转移形成的基础。癌症激活的成纤维细胞(CAFs)是一种被肿瘤信号激活的宿主成纤维细胞,它可以通过旁分泌信号(分泌扩散分子)和机械信号(改变基质的组成和组织)改变肿瘤细胞的行为。这些成纤维细胞类似于伤口愈合过程中产生的肉芽组织中的肌成纤维细胞(MFs),后者产生一种富含信号分子和交联细胞外纤维的刚性结缔组织基质。结缔组织增生有利于恶性肿瘤细胞的特性,如移动性、干性,甚至抵抗药物损伤[1]。
{"title":"Snail1 controls cooperative cell plasticity during metastasis","authors":"J. Baulida","doi":"10.18632/ONCOSCIENCE.262","DOIUrl":"https://doi.org/10.18632/ONCOSCIENCE.262","url":null,"abstract":"Mortality in cancer is strongly associated with the capacity of tumor cells to spread and critically affect other tissues and organs. Genetic mutations accumulated by tumor cells and cross-signaling between tumor and host cells underlie the formation of metastasis. Cancer-activated fibroblasts (CAFs), which are host fibroblasts activated by tumor signaling, can alter tumor cell behavior by both paracrine signaling (secreting diffusible molecules) and mechanical signaling (modifying the composition and organization of the stroma). These fibroblasts resemble myofibroblasts (MFs) of the granulation tissue generated during wound healing, which produce a rigid desmoplastic stroma rich in signaling molecules and cross-linked extracellular fibers. Desmoplasia favors malignant tumor cell properties such as mobility, stemness, and even resistance to pharmacological insults [1].","PeriodicalId":94164,"journal":{"name":"Oncoscience","volume":"33 1","pages":"898 - 899"},"PeriodicalIF":0.0,"publicationDate":"2015-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73297482","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: 2
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