首页 > 最新文献

Cancer Genomics & Proteomics最新文献

英文 中文
Gene Expression Profiling Regulated by lncRNA H19 Using Bioinformatic Analyses in Glioma Cell Lines. 利用生物信息学分析脑胶质瘤细胞系中受 lncRNA H19 调控的基因表达谱。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20477
Yeonsoo Chae, Jungwook Roh, Mijung Im, Wonyi Jang, Boseong Kim, Jihoon Kang, Buhyun Youn, Wanyeon Kim

Background/aim: Glioma, the most common type of primary brain tumor, is characterized by high malignancy, recurrence, and mortality. Long non-coding RNA (lncRNA) H19 is a potential biomarker for glioma diagnosis and treatment due to its overexpression in human glioma tissues and its involvement in cell division and metastasis regulation. This study aimed to identify potential therapeutic targets involved in glioma development by analyzing gene expression profiles regulated by H19.

Materials and methods: To elucidate the role of H19 in A172 and U87MG glioma cell lines, cell counting, colony formation, and wound healing assays were conducted. RNA-seq data analysis and bioinformatics analyses were performed to reveal the molecular interactions of H19.

Results: Cell-based experiments showed that elevated H19 levels were related to cancer cell survival, proliferation, and migration. Bioinformatics analyses identified 2,084 differentially expressed genes (DEGs) influenced by H19 which are involved in cancer progression. Specifically, ANXA5, CLEC18B, RAB42, CXCL8, OASL, USP18, and CDCP1 were positively correlated with H19 expression, while CSDC2 and FOXO4 were negatively correlated. These DEGs were predicted to function as oncogenes or tumor suppressors in gliomas, in association with H19.

Conclusion: These findings highlight H19 and its associated genes as potential diagnostic and therapeutic targets for gliomas, emphasizing their clinical significance in patient survival.

背景/目的:胶质瘤是最常见的原发性脑肿瘤,具有恶性程度高、复发率高和死亡率高的特点。长非编码 RNA(lncRNA)H19 是胶质瘤诊断和治疗的潜在生物标志物,因为它在人类胶质瘤组织中过度表达,并参与细胞分裂和转移调控。本研究旨在通过分析 H19 调控的基因表达谱,确定参与胶质瘤发展的潜在治疗靶点:为了阐明H19在A172和U87MG胶质瘤细胞系中的作用,研究人员进行了细胞计数、集落形成和伤口愈合试验。进行了 RNA-seq 数据分析和生物信息学分析,以揭示 H19 的分子相互作用:基于细胞的实验表明,H19水平的升高与癌细胞的存活、增殖和迁移有关。生物信息学分析发现了 2,084 个受 H19 影响的差异表达基因(DEGs),这些基因参与了癌症进展。具体来说,ANXA5、CLEC18B、RAB42、CXCL8、OASL、USP18和CDCP1与H19的表达呈正相关,而CSDC2和FOXO4呈负相关。预测这些 DEGs 与 H19 相关,可在胶质瘤中发挥致癌基因或肿瘤抑制因子的作用:这些发现强调了H19及其相关基因是胶质瘤潜在的诊断和治疗靶点,并强调了它们对患者生存的临床意义。
{"title":"Gene Expression Profiling Regulated by lncRNA H19 Using Bioinformatic Analyses in Glioma Cell Lines.","authors":"Yeonsoo Chae, Jungwook Roh, Mijung Im, Wonyi Jang, Boseong Kim, Jihoon Kang, Buhyun Youn, Wanyeon Kim","doi":"10.21873/cgp.20477","DOIUrl":"10.21873/cgp.20477","url":null,"abstract":"<p><strong>Background/aim: </strong>Glioma, the most common type of primary brain tumor, is characterized by high malignancy, recurrence, and mortality. Long non-coding RNA (lncRNA) H19 is a potential biomarker for glioma diagnosis and treatment due to its overexpression in human glioma tissues and its involvement in cell division and metastasis regulation. This study aimed to identify potential therapeutic targets involved in glioma development by analyzing gene expression profiles regulated by H19.</p><p><strong>Materials and methods: </strong>To elucidate the role of H19 in A172 and U87MG glioma cell lines, cell counting, colony formation, and wound healing assays were conducted. RNA-seq data analysis and bioinformatics analyses were performed to reveal the molecular interactions of H19.</p><p><strong>Results: </strong>Cell-based experiments showed that elevated H19 levels were related to cancer cell survival, proliferation, and migration. Bioinformatics analyses identified 2,084 differentially expressed genes (DEGs) influenced by H19 which are involved in cancer progression. Specifically, ANXA5, CLEC18B, RAB42, CXCL8, OASL, USP18, and CDCP1 were positively correlated with H19 expression, while CSDC2 and FOXO4 were negatively correlated. These DEGs were predicted to function as oncogenes or tumor suppressors in gliomas, in association with H19.</p><p><strong>Conclusion: </strong>These findings highlight H19 and its associated genes as potential diagnostic and therapeutic targets for gliomas, emphasizing their clinical significance in patient survival.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"608-621"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534032/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521084","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Extensive DNA Damage and Loss of Cell Viability Occur Synergistically With the Combination of Recombinant Methioninase and Paclitaxel on Pancreatic Cancer Cells which Report DNA-Damage Response in Real Time. 重组甲硫氨酸酶和紫杉醇对胰腺癌细胞的协同作用会导致广泛的 DNA 损伤和细胞活力丧失,而紫杉醇会实时报告 DNA 损伤反应。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20475
Sei Morinaga, Qinghong Han, Kohei Mizuta, Byung Mo Kang, Motokazu Sato, Michael Bouvet, Norio Yamamoto, Katsuhiro Hayashi, Hiroaki Kimura, Shinji Miwa, Kentaro Igarashi, Takashi Higuchi, Hiroyuki Tsuchiya, Satoru Demura, Robert M Hoffman

Background/aim: Methionine restriction selectively arrests cancer cells during the S-phase of the cell cycle. We hypothesized that DNA damage may occur in S-phase in cancer cells during methionine restriction. To determine if this occurs, we used MiaPaCa-2Tet-On 53BP1-green fluorescent protein (GFP) pancreatic cancer cells, which report GFP fluorescence in real time after DNA-damage response (DDR) in these cells. We also determined whether a chemotherapy drug in combination with methionine restriction increases the rate of DNA damage.

Materials and methods: MiaPaCa-2Tet-On 53BP1-GFP cells were used for in vitro experiments. The 25% and 50% inhibitory concentrations (IC25 and IC50, respectively) of recombinant methioninase (rMETase) and paclitaxel on MiaPaCa-2Tet-On 53BP1-GFP pancreatic cancer cells were determined. Cell viability and DDR with rMETase alone, paclitaxel alone, and their combination were measured in MiaPaCa-2Tet-On 53BP1-GFP cells.

Results: The IC25 of rMETase on MiaPaCa-2Tet-On 53BP1-GFP cells was 1.66 U/ml. The IC25 for paclitaxel on MiaPaCa-2Tet-On 53BP1-GFP cells was 3.31 nM. The combination of rMETase and paclitaxel synergistically reduced the viability of MiaPaCa-2Tet-On 53BP1-GFP cells. The IC50 of paclitacel on MiaPaCa-2Tet-On 53BP1-GFP cells was 5.1 nM. The IC50 of rMETase on MiaPaCa-2Tet-On 53BP1-GFP cells was 2.3 U/ml. The combination of rMETase (IC50) plus paclitaxel (IC50) on MiaPaCa-2Tet-On 53BP1-GFP cells also caused more DNA damage than either agent alone.

Conclusion: The present study suggests the synergy of methionine restriction and chemotherapy is due, at least in part, to DNA damage of cancer cells.

背景/目的:蛋氨酸限制可选择性地使癌细胞停滞在细胞周期的 S 期。我们推测,在蛋氨酸限制过程中,癌细胞的 S 期可能会发生 DNA 损伤。为了确定是否会发生这种情况,我们使用了 MiaPaCa-2Tet-On 53BP1 绿色荧光蛋白(GFP)胰腺癌细胞,这些细胞在发生 DNA 损伤反应(DDR)后会实时发出 GFP 荧光。我们还测定了化疗药物与蛋氨酸限制结合是否会增加DNA损伤率:体外实验使用 MiaPaCa-2Tet-On 53BP1-GFP 细胞。测定了重组蛋氨酸酶(rMETase)和紫杉醇对 MiaPaCa-2Tet-On 53BP1-GFP 胰腺癌细胞的 25% 和 50% 抑制浓度(IC25 和 IC50)。在 MiaPaCa-2Tet-On 53BP1-GFP 细胞中测定了单独使用 rMETase、单独使用紫杉醇以及它们联合使用时的细胞活力和 DDR:结果:rMETase 对 MiaPaCa-2Tet-On 53BP1-GFP 细胞的 IC25 为 1.66 U/ml 。紫杉醇对 MiaPaCa-2Tet-On 53BP1-GFP 细胞的 IC25 为 3.31 nM。rMETase 和紫杉醇联合使用可协同降低 MiaPaCa-2Tet-On 53BP1-GFP 细胞的活力。紫杉醇对 MiaPaCa-2Tet-On 53BP1-GFP 细胞的 IC50 为 5.1 nM。rMETase 对 MiaPaCa-2Tet-On 53BP1-GFP 细胞的 IC50 为 2.3 U/ml。在 MiaPaCa-2Tet-On 53BP1-GFP 细胞上,rMETase(IC50)和紫杉醇(IC50)的组合也比单独使用其中一种药物造成更多的 DNA 损伤:本研究表明,蛋氨酸限制与化疗的协同作用至少部分是由于癌细胞的 DNA 损伤所致。
{"title":"Extensive DNA Damage and Loss of Cell Viability Occur Synergistically With the Combination of Recombinant Methioninase and Paclitaxel on Pancreatic Cancer Cells which Report DNA-Damage Response in Real Time.","authors":"Sei Morinaga, Qinghong Han, Kohei Mizuta, Byung Mo Kang, Motokazu Sato, Michael Bouvet, Norio Yamamoto, Katsuhiro Hayashi, Hiroaki Kimura, Shinji Miwa, Kentaro Igarashi, Takashi Higuchi, Hiroyuki Tsuchiya, Satoru Demura, Robert M Hoffman","doi":"10.21873/cgp.20475","DOIUrl":"10.21873/cgp.20475","url":null,"abstract":"<p><strong>Background/aim: </strong>Methionine restriction selectively arrests cancer cells during the S-phase of the cell cycle. We hypothesized that DNA damage may occur in S-phase in cancer cells during methionine restriction. To determine if this occurs, we used MiaPaCa-2<sup>Tet-On</sup> 53BP1-green fluorescent protein (GFP) pancreatic cancer cells, which report GFP fluorescence in real time after DNA-damage response (DDR) in these cells. We also determined whether a chemotherapy drug in combination with methionine restriction increases the rate of DNA damage.</p><p><strong>Materials and methods: </strong>MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells were used for in vitro experiments. The 25% and 50% inhibitory concentrations (IC<sub>25</sub> and IC<sub>50</sub>, respectively) of recombinant methioninase (rMETase) and paclitaxel on MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP pancreatic cancer cells were determined. Cell viability and DDR with rMETase alone, paclitaxel alone, and their combination were measured in MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells.</p><p><strong>Results: </strong>The IC<sub>25</sub> of rMETase on MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells was 1.66 U/ml. The IC<sub>25</sub> for paclitaxel on MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells was 3.31 nM. The combination of rMETase and paclitaxel synergistically reduced the viability of MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells. The IC<sub>50</sub> of paclitacel on MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells was 5.1 nM. The IC<sub>50</sub> of rMETase on MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells was 2.3 U/ml. The combination of rMETase (IC<sub>50</sub>) plus paclitaxel (IC<sub>50</sub>) on MiaPaCa-2<sup>Tet-On</sup> 53BP1-GFP cells also caused more DNA damage than either agent alone.</p><p><strong>Conclusion: </strong>The present study suggests the synergy of methionine restriction and chemotherapy is due, at least in part, to DNA damage of cancer cells.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"585-590"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521082","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GD2 in Breast Cancer: A Potential Biomarker and Therapeutic Target. 乳腺癌中的 GD2:潜在的生物标记物和治疗靶点
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20471
Kefah Mokbel

Expression of disialoganglioside GD2 in normal tissues is primarily limited to the central nervous system, peripheral sensory nerve fibers, dermal melanocytes, lymphocytes, and mesenchymal stem cells. Its widespread overexpression in various cancer types allows it to be classified as a tumor-associated antigen with potential diagnostic and therapeutic implications. This article reviews the synthesis pathways of GD2 and its role in cancer cell adhesion, proliferation, and metastasis with a focus on breast cancer. GD2 appears to be overexpressed on the outer membrane of most breast cancer cells and breast cancer stem cells (BCSCs) and is closely linked to epithelial-mesenchymal transition (EMT). GD3 synthase (GD3S) is considered to be the rate-determining step in GD2 synthesis. Clinical studies indicate that GD2 expression is increased in 35-70% of breast cancer samples, with higher levels in triple-negative breast cancer (TNBC). This overexpression correlates with more aggressive tumor features and worse prognosis. Therapeutic targeting of GD2 with monoclonal antibodies (moABs) like dinutuximab and naxitamab has demonstrated anti-cancer activity in preclinical cancer models and human clinical trials against high-risk neuroblastoma reducing tumor growth and enhancing survival. GD2-specific chimeric antigen receptor (CAR) T-cell therapy and GD3S inhibition present other promising therapeutic strategies to improve clinical outcomes. Furthermore, GD2-targeted vaccines are currently being investigated in cancer therapy. This narrative review article underscores the critical role of GD2 in breast cancer pathogenesis and highlights the promising therapeutic opportunities it offers. It advocates for the initiation of clinical trials to further explore the potential of GD2-targeted treatment in combination with standard breast cancer therapies.

二异麦角酰肌苷 GD2 在正常组织中的表达主要局限于中枢神经系统、外周感觉神经纤维、真皮黑色素细胞、淋巴细胞和间充质干细胞。它在各种癌症类型中的广泛过表达使其被归类为一种具有潜在诊断和治疗意义的肿瘤相关抗原。本文以乳腺癌为重点,回顾了 GD2 的合成途径及其在癌细胞粘附、增殖和转移中的作用。GD2 似乎在大多数乳腺癌细胞和乳腺癌干细胞(BCSCs)的外膜上过度表达,并与上皮-间质转化(EMT)密切相关。GD3 合成酶(GD3S)被认为是 GD2 合成的决定性步骤。临床研究表明,35%-70% 的乳腺癌样本中 GD2 表达增加,三阴性乳腺癌(TNBC)中的表达水平更高。这种过表达与更具侵袭性的肿瘤特征和更差的预后相关。利用地纽昔单抗和纳昔单抗等单克隆抗体(moABs)对 GD2 进行靶向治疗,已在临床前癌症模型和针对高危神经母细胞瘤的人体临床试验中显示出抗癌活性,可减少肿瘤生长并提高存活率。GD2特异性嵌合抗原受体(CAR)T细胞疗法和GD3S抑制剂也是改善临床疗效的治疗策略。此外,GD2靶向疫苗目前也正在癌症治疗中进行研究。这篇叙述性综述文章强调了 GD2 在乳腺癌发病机制中的关键作用,并着重指出了它所提供的大有可为的治疗机会。文章主张启动临床试验,进一步探索 GD2 靶向治疗与标准乳腺癌疗法相结合的潜力。
{"title":"GD2 in Breast Cancer: A Potential Biomarker and Therapeutic Target.","authors":"Kefah Mokbel","doi":"10.21873/cgp.20471","DOIUrl":"10.21873/cgp.20471","url":null,"abstract":"<p><p>Expression of disialoganglioside GD2 in normal tissues is primarily limited to the central nervous system, peripheral sensory nerve fibers, dermal melanocytes, lymphocytes, and mesenchymal stem cells. Its widespread overexpression in various cancer types allows it to be classified as a tumor-associated antigen with potential diagnostic and therapeutic implications. This article reviews the synthesis pathways of GD2 and its role in cancer cell adhesion, proliferation, and metastasis with a focus on breast cancer. GD2 appears to be overexpressed on the outer membrane of most breast cancer cells and breast cancer stem cells (BCSCs) and is closely linked to epithelial-mesenchymal transition (EMT). GD3 synthase (GD3S) is considered to be the rate-determining step in GD2 synthesis. Clinical studies indicate that GD2 expression is increased in 35-70% of breast cancer samples, with higher levels in triple-negative breast cancer (TNBC). This overexpression correlates with more aggressive tumor features and worse prognosis. Therapeutic targeting of GD2 with monoclonal antibodies (moABs) like dinutuximab and naxitamab has demonstrated anti-cancer activity in preclinical cancer models and human clinical trials against high-risk neuroblastoma reducing tumor growth and enhancing survival. GD2-specific chimeric antigen receptor (CAR) T-cell therapy and GD3S inhibition present other promising therapeutic strategies to improve clinical outcomes. Furthermore, GD2-targeted vaccines are currently being investigated in cancer therapy. This narrative review article underscores the critical role of GD2 in breast cancer pathogenesis and highlights the promising therapeutic opportunities it offers. It advocates for the initiation of clinical trials to further explore the potential of GD2-targeted treatment in combination with standard breast cancer therapies.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"549-556"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534030/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Impact of Hypoxia and the Levels of Transcription Factor HIF-1α and JMJD1A on Epithelial-Mesenchymal Transition in Head and Neck Squamous Cell Carcinoma Cell Lines. 缺氧及转录因子 HIF-1α 和 JMJD1A 水平对头颈部鳞状细胞癌细胞系上皮-间质转化的影响
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20476
Armin VON Fournier, Christian Wilhelm, Clara Tirtey, Manuel Stöth, Totta Ehret Kasemo, Stephan Hackenberg, Agmal Scherzad

Background/aim: This study aimed to assess the impact of hypoxia on epithelial-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC), focusing on the involvement of transcription factors hypoxia inducible factor 1 (HIF-1α) and Jumonji Domain-Containing Protein 1A (JMJD1A).

Materials and methods: FaDu and Cal33 cell lines were subjected to hypoxic and normoxic conditions. Cell proliferation was quantified electronically, while PCR and western blot analyses were used to measure mRNA and protein levels of HIF-1α, JMJD1A, and EMT markers. EMT was further characterized through immunofluorescence, migration, and invasion assays.

Results: Hypoxic conditions significantly reduced cell proliferation after 48 hours in both cell lines. HIF-1α mRNA levels increased initially during short-term hypoxia but declined thereafter, while JMJD1A mRNA levels showed a sustained increase with prolonged hypoxia. Western blot analysis revealed contrasting trends in protein levels. EMT marker expression varied markedly over time at both the mRNA and protein levels, suggesting EMT induction in hypoxia within 24 hours. Immunofluorescence, migration, and invasion assays supported these findings.

Conclusion: The study provides evidence of hypoxia-induced EMT in HNSCC, although conflicting results suggest a complex interplay among molecular regulators involved in this process.

背景/目的:本研究旨在评估缺氧对头颈部鳞状细胞癌(HNSCC)上皮-间质转化(EMT)的影响,重点关注转录因子缺氧诱导因子1(HIF-1α)和含Jumonji域蛋白1A(JMJD1A)的参与:将FaDu和Cal33细胞系置于缺氧和常氧条件下。细胞增殖以电子方式量化,PCR和Western印迹分析用于测量HIF-1α、JMJD1A和EMT标记物的mRNA和蛋白水平。通过免疫荧光、迁移和侵袭实验进一步确定了 EMT 的特征:结果:48 小时后,缺氧条件明显降低了两种细胞系的细胞增殖。HIF-1α mRNA水平最初在短期缺氧时升高,但随后下降,而JMJD1A mRNA水平则随着缺氧时间的延长而持续升高。Western 印迹分析显示了蛋白质水平的对比趋势。EMT标记物在mRNA和蛋白质水平上的表达随时间变化明显,表明缺氧在24小时内诱导了EMT。免疫荧光、迁移和侵袭试验证实了这些发现:结论:本研究提供了缺氧诱导 HNSCC EMT 的证据,尽管相互矛盾的结果表明参与这一过程的分子调控因子之间存在复杂的相互作用。
{"title":"Impact of Hypoxia and the Levels of Transcription Factor HIF-1α and JMJD1A on Epithelial-Mesenchymal Transition in Head and Neck Squamous Cell Carcinoma Cell Lines.","authors":"Armin VON Fournier, Christian Wilhelm, Clara Tirtey, Manuel Stöth, Totta Ehret Kasemo, Stephan Hackenberg, Agmal Scherzad","doi":"10.21873/cgp.20476","DOIUrl":"10.21873/cgp.20476","url":null,"abstract":"<p><strong>Background/aim: </strong>This study aimed to assess the impact of hypoxia on epithelial-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC), focusing on the involvement of transcription factors hypoxia inducible factor 1 (HIF-1α) and Jumonji Domain-Containing Protein 1A (JMJD1A).</p><p><strong>Materials and methods: </strong>FaDu and Cal33 cell lines were subjected to hypoxic and normoxic conditions. Cell proliferation was quantified electronically, while PCR and western blot analyses were used to measure mRNA and protein levels of HIF-1α, JMJD1A, and EMT markers. EMT was further characterized through immunofluorescence, migration, and invasion assays.</p><p><strong>Results: </strong>Hypoxic conditions significantly reduced cell proliferation after 48 hours in both cell lines. HIF-1α mRNA levels increased initially during short-term hypoxia but declined thereafter, while JMJD1A mRNA levels showed a sustained increase with prolonged hypoxia. Western blot analysis revealed contrasting trends in protein levels. EMT marker expression varied markedly over time at both the mRNA and protein levels, suggesting EMT induction in hypoxia within 24 hours. Immunofluorescence, migration, and invasion assays supported these findings.</p><p><strong>Conclusion: </strong>The study provides evidence of hypoxia-induced EMT in HNSCC, although conflicting results suggest a complex interplay among molecular regulators involved in this process.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"591-607"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cordycepin Activates Autophagy to Suppress FGF9-induced TM3 Mouse Leydig Progenitor Cell Proliferation. 虫草素能激活自噬作用,抑制 FGF9 诱导的 TM3 小鼠雷迪格祖细胞增殖
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20479
Su-Zhen Wu, Yu-Yan Lan, Chin-Ying Chen, Li-Ching Chen, Bu-Miin Huang

Background/aim: Fibroblast growth factor 9 (FGF9) is a member of the human FGF family known for its pivotal roles in various biological processes, such as cell proliferation, tissue repair, and male sex determination including testis formation. Cordycepin, a bioactive compound found in Cordyceps sinensis, exhibits potent antitumor effects by triggering apoptosis and/or autophagy pathways. Our research has unveiled that FGF9 promotes proliferation and tumorigenesis in MA-10 mouse Leydig tumor cells, as the phenomena are effectively countered by cordycepin through apoptosis induction. Moreover, we have observed FGF9-mediated stimulation of proliferation and tumorigenesis in TM3 mouse Leydig progenitor cells, prompting an investigation into the potential inhibitory effect of cordycepin on TM3 cell proliferation under FGF9 treatment. Hence, we hypothesized that cordycepin induces cell death via apoptosis and/or autophagy in FGF9-treated TM3 cells.

Materials and methods: TM3 cells were treated with cordycepin and/or FGF9, and the flow cytometry, immunofluorescent plus western blotting assays were used to determine how cordycepin regulated Leydig cell death under FGF9 treatment.

Results: Our findings reveal that cordycepin restricts cell viability and colony formation while inducing morphological alterations associated with cell death in FGF9-treated TM3 cells. Surprisingly, cordycepin fails to elicit the expression of key apoptotic markers, suggesting an alternate mechanism of action. Although the expression of certain autophagy-related proteins remains unaltered, a significant up-regulation of LC3-II, indicative of autophagy, is observed in cordycepin-treated TM3 cells under FGF9 influence. Moreover, the inhibition of autophagy by chloroquine reverses cordycepin-induced TM3 cell death, highlighting the crucial role of autophagy in this process.

Conclusion: Our study demonstrates that cordycepin activates autophagy to induce cell death in TM3 cells under FGF9 treatment conditions.

背景/目的:成纤维细胞生长因子 9(FGF9)是人类成纤维细胞生长因子家族的成员,在细胞增殖、组织修复和男性性别决定(包括睾丸形成)等各种生物过程中发挥着关键作用。冬虫夏草素是冬虫夏草中发现的一种生物活性化合物,它能通过触发细胞凋亡和/或自噬途径发挥强大的抗肿瘤作用。我们的研究揭示了 FGF9 会促进 MA-10 小鼠 Leydig 肿瘤细胞的增殖和肿瘤发生,而虫草素通过诱导细胞凋亡有效地对抗了这一现象。此外,我们还观察到 FGF9 介导的刺激 TM3 小鼠 Leydig 祖细胞增殖和肿瘤发生的现象,这促使我们研究虫草素在 FGF9 处理下对 TM3 细胞增殖的潜在抑制作用。因此,我们假设虫草素通过凋亡和/或自噬诱导 FGF9 处理的 TM3 细胞死亡:用虫草素和/或FGF9处理TM3细胞,采用流式细胞术、免疫荧光和Western印迹法检测虫草素如何调控FGF9处理下的Leydig细胞死亡:结果:我们的研究结果表明,虫草素限制了 FGF9 处理的 TM3 细胞的细胞活力和集落形成,同时诱导了与细胞死亡相关的形态学改变。令人惊讶的是,虫草素未能诱导关键凋亡标志物的表达,这表明虫草素有另一种作用机制。虽然某些自噬相关蛋白的表达未发生改变,但在 FGF9 的影响下,在虫草素处理的 TM3 细胞中观察到 LC3-II 的显著上调,这表明了自噬。此外,氯喹对自噬的抑制逆转了虫草素诱导的 TM3 细胞死亡,突出了自噬在这一过程中的关键作用:我们的研究表明,在FGF9处理条件下,虫草素能激活自噬,诱导TM3细胞死亡。
{"title":"Cordycepin Activates Autophagy to Suppress FGF9-induced TM3 Mouse Leydig Progenitor Cell Proliferation.","authors":"Su-Zhen Wu, Yu-Yan Lan, Chin-Ying Chen, Li-Ching Chen, Bu-Miin Huang","doi":"10.21873/cgp.20479","DOIUrl":"10.21873/cgp.20479","url":null,"abstract":"<p><strong>Background/aim: </strong>Fibroblast growth factor 9 (FGF9) is a member of the human FGF family known for its pivotal roles in various biological processes, such as cell proliferation, tissue repair, and male sex determination including testis formation. Cordycepin, a bioactive compound found in Cordyceps sinensis, exhibits potent antitumor effects by triggering apoptosis and/or autophagy pathways. Our research has unveiled that FGF9 promotes proliferation and tumorigenesis in MA-10 mouse Leydig tumor cells, as the phenomena are effectively countered by cordycepin through apoptosis induction. Moreover, we have observed FGF9-mediated stimulation of proliferation and tumorigenesis in TM3 mouse Leydig progenitor cells, prompting an investigation into the potential inhibitory effect of cordycepin on TM3 cell proliferation under FGF9 treatment. Hence, we hypothesized that cordycepin induces cell death via apoptosis and/or autophagy in FGF9-treated TM3 cells.</p><p><strong>Materials and methods: </strong>TM3 cells were treated with cordycepin and/or FGF9, and the flow cytometry, immunofluorescent plus western blotting assays were used to determine how cordycepin regulated Leydig cell death under FGF9 treatment.</p><p><strong>Results: </strong>Our findings reveal that cordycepin restricts cell viability and colony formation while inducing morphological alterations associated with cell death in FGF9-treated TM3 cells. Surprisingly, cordycepin fails to elicit the expression of key apoptotic markers, suggesting an alternate mechanism of action. Although the expression of certain autophagy-related proteins remains unaltered, a significant up-regulation of LC3-II, indicative of autophagy, is observed in cordycepin-treated TM3 cells under FGF9 influence. Moreover, the inhibition of autophagy by chloroquine reverses cordycepin-induced TM3 cell death, highlighting the crucial role of autophagy in this process.</p><p><strong>Conclusion: </strong>Our study demonstrates that cordycepin activates autophagy to induce cell death in TM3 cells under FGF9 treatment conditions.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"630-644"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521081","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GULP1 as a Downstream Effector of the Estrogen Receptor-β Modulates Cisplatin Sensitivity in Bladder Cancer. GULP1 作为雌激素受体-β的下游效应因子调节膀胱癌患者对顺铂的敏感性
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20472
Tomoyuki Tatenuma, Takuo Matsukawa, Takuro Goto, Guiyang Jiang, Adhya Sharma, Mohammad Amin Elahi Najafi, Yuki Teramoto, Hiroshi Miyamoto

Background/aim: Precise molecular mechanisms underlying resistance to cisplatin-based chemotherapy remain unclear, while the activity of estrogen receptor-β (ERβ) has been suggested to be associated with chemosensitivity in urothelial cancer. We aimed to determine if GULP1, an adapter protein known to facilitate phagocytosis, could represent a downstream effector of ERβ and thereby modulate cisplatin sensitivity in bladder cancer.

Materials and methods: GULP1 expression and cisplatin cytotoxicity were compared in bladder cancer lines. Immunohistochemistry was used to determine the expression of GULP1 and ERβ in two sets of tissue microarray (TMA) consisting of transurethral resection specimens.

Results: The levels of GULP1 expression were considerably higher in ERβ-knockdown sublines than in the respective control ERβ-positive sublines. Estradiol treatment reduced GULP1 expression in ERα-negative/ERβ-positive lines, which was restored by the anti-estrogen tamoxifen. Chromatin immunoprecipitation assay revealed the binding of ERβ to the GULP1 promoter in bladder cancer cells. Moreover, GULP1 knockdown sublines were significantly more resistant to cisplatin treatment, but not to other chemotherapeutic agents, including gemcitabine, methotrexate, vinblastine, and doxorubicin. In the first set of TMA (n=129), the expression of ERβ and GULP1 was inversely correlated (p=0.023), and ERβ(-)/GULP1(+) in 51 muscle-invasive tumors was associated with significantly lower risk of disease progression and cancer-specific mortality. Similarly, in the second set (n=43), patients with ERβ(-)/GULP1(+) muscle-invasive disease were significantly (p=0.021) more likely to be responders to cisplatin-based neoadjuvant chemotherapy before radical cystectomy.

Conclusion: ERβ activation was found to reduce the expression of GULP1 as a direct downstream target in bladder cancer cells, resulting in the induction of cisplatin resistance.

背景/目的:顺铂类化疗耐药性的确切分子机制仍不清楚,而雌激素受体-β(ERβ)的活性被认为与尿路上皮癌的化疗敏感性有关。我们的目的是确定GULP1(一种已知促进吞噬作用的适配蛋白)是否代表ERβ的下游效应物,从而调节膀胱癌的顺铂敏感性:比较膀胱癌细胞系中 GULP1 的表达和顺铂的细胞毒性。免疫组化法测定了两组经尿道切除标本组织芯片(TMA)中 GULP1 和 ERβ 的表达:结果:在ERβ敲除亚系中,GULP1的表达水平明显高于ERβ阳性对照亚系。雌二醇处理降低了ERα阴性/ERβ阳性株系中GULP1的表达,而抗雌激素他莫昔芬可恢复这种表达。染色质免疫共沉淀分析显示,在膀胱癌细胞中,ERβ与GULP1启动子结合。此外,GULP1基因敲除亚系对顺铂治疗的耐药性明显增强,但对其他化疗药物(包括吉西他滨、甲氨蝶呤、长春新碱和多柔比星)的耐药性则没有增强。在第一组TMA(n=129)中,ERβ和GULP1的表达呈反向相关(p=0.023),51例肌层浸润性肿瘤中的ERβ(-)/GULP1(+)与疾病进展风险和癌症特异性死亡率显著降低相关。同样,在第二组(n=43)中,ERβ(-)/GULP1(+)肌浸润性疾病患者在根治性膀胱切除术前接受顺铂新辅助化疗的几率明显增加(p=0.021):结论:研究发现,ERβ活化会降低作为膀胱癌细胞直接下游靶点的GULP1的表达,从而诱导顺铂耐药。
{"title":"GULP1 as a Downstream Effector of the Estrogen Receptor-β Modulates Cisplatin Sensitivity in Bladder Cancer.","authors":"Tomoyuki Tatenuma, Takuo Matsukawa, Takuro Goto, Guiyang Jiang, Adhya Sharma, Mohammad Amin Elahi Najafi, Yuki Teramoto, Hiroshi Miyamoto","doi":"10.21873/cgp.20472","DOIUrl":"10.21873/cgp.20472","url":null,"abstract":"<p><strong>Background/aim: </strong>Precise molecular mechanisms underlying resistance to cisplatin-based chemotherapy remain unclear, while the activity of estrogen receptor-β (ERβ) has been suggested to be associated with chemosensitivity in urothelial cancer. We aimed to determine if GULP1, an adapter protein known to facilitate phagocytosis, could represent a downstream effector of ERβ and thereby modulate cisplatin sensitivity in bladder cancer.</p><p><strong>Materials and methods: </strong>GULP1 expression and cisplatin cytotoxicity were compared in bladder cancer lines. Immunohistochemistry was used to determine the expression of GULP1 and ERβ in two sets of tissue microarray (TMA) consisting of transurethral resection specimens.</p><p><strong>Results: </strong>The levels of GULP1 expression were considerably higher in ERβ-knockdown sublines than in the respective control ERβ-positive sublines. Estradiol treatment reduced GULP1 expression in ERα-negative/ERβ-positive lines, which was restored by the anti-estrogen tamoxifen. Chromatin immunoprecipitation assay revealed the binding of ERβ to the GULP1 promoter in bladder cancer cells. Moreover, GULP1 knockdown sublines were significantly more resistant to cisplatin treatment, but not to other chemotherapeutic agents, including gemcitabine, methotrexate, vinblastine, and doxorubicin. In the first set of TMA (n=129), the expression of ERβ and GULP1 was inversely correlated (p=0.023), and ERβ(-)/GULP1(+) in 51 muscle-invasive tumors was associated with significantly lower risk of disease progression and cancer-specific mortality. Similarly, in the second set (n=43), patients with ERβ(-)/GULP1(+) muscle-invasive disease were significantly (p=0.021) more likely to be responders to cisplatin-based neoadjuvant chemotherapy before radical cystectomy.</p><p><strong>Conclusion: </strong>ERβ activation was found to reduce the expression of GULP1 as a direct downstream target in bladder cancer cells, resulting in the induction of cisplatin resistance.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"557-565"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534028/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521085","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Serum Exosomes Expressing CD9, CD63 and HER2 From Breast-Cancer Patients Decreased After Surgery of the Primary Tumor: A Potential Biomarker of Tumor Burden. 乳腺癌患者血清中表达 CD9、CD63 和 HER2 的外泌体在原发肿瘤手术后减少:肿瘤负担的潜在生物标志物。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20474
Sachiko Inubushi, Tomonari Kunihisa, Marina Kuniyasu, Shotaro Inoue, Mayuko Yamamoto, Yuji Yamashita, Mayuko Miki, Sachiko Mizumoto, Motoi Baba, Robert M Hoffman, Hirokazu Tanino

Background/aim: Exosomes are extracellular vesicles produced by both normal and cancer cells. Previous research has demonstrated that circulating exosomes derived from cancer cells may create a niche for future metastasis, distant from the primary tumor. In the present report, circulating exosomes were captured and quantified based on exosome-surface proteins in pre- and post-operative serum of breast cancer patients, focusing on the exosome markers CD9 and CD63, as well as HER2, a therapeutic target for breast cancer.

Materials and methods: Eight breast cancer patients were recruited, and their pre- and post-operative serum samples were analyzed for CD63 and CD9; or CD9 and human epidermal growth factor receptor-2 (HER2), double-positive exosomes. An ExoCounter with antibody-conjugated beads was used to capture serum-derived exosomes. Sera from patients with tumors larger than 10 mm were used for analysis. The resected breast cancer was also histopathologically analyzed for the presence of HER2.

Results: CD63 and CD9 double-positive serum exosomes and CD9 and HER2 double-positive serum exosomes decreased after surgery in breast-cancer patients whose tumors expressed HER2, as determined by histopathological analysis.

Conclusion: Serum exosomes expressing CD9, CD63 and HER2 are candidate biomarkers of tumor burden in HER2-positive breast-cancer patients.

背景/目的:外泌体是由正常细胞和癌细胞产生的细胞外囊泡。先前的研究表明,从癌细胞中提取的循环外泌体可能会为未来远离原发肿瘤的转移创造一个环境。本报告根据乳腺癌患者术前和术后血清中的外泌体表面蛋白对循环外泌体进行了捕获和量化,重点研究了外泌体标记物CD9和CD63以及乳腺癌的治疗靶点HER2:招募了八名乳腺癌患者,对他们术前和术后血清样本中的 CD63 和 CD9,或 CD9 和人表皮生长因子受体-2(HER2)双阳性外泌体进行分析。使用带有抗体结合珠的 ExoCounter 捕捉血清中的外泌体。分析所用的血清来自肿瘤大于 10 毫米的患者。对切除的乳腺癌也进行了组织病理学分析,以确定是否存在 HER2:结果:经组织病理学分析确定,肿瘤表达HER2的乳腺癌患者术后血清外泌体CD63和CD9双阳性减少,血清外泌体CD9和HER2双阳性减少:表达 CD9、CD63 和 HER2 的血清外泌体是 HER2 阳性乳腺癌患者肿瘤负荷的候选生物标志物。
{"title":"Serum Exosomes Expressing CD9, CD63 and HER2 From Breast-Cancer Patients Decreased After Surgery of the Primary Tumor: A Potential Biomarker of Tumor Burden.","authors":"Sachiko Inubushi, Tomonari Kunihisa, Marina Kuniyasu, Shotaro Inoue, Mayuko Yamamoto, Yuji Yamashita, Mayuko Miki, Sachiko Mizumoto, Motoi Baba, Robert M Hoffman, Hirokazu Tanino","doi":"10.21873/cgp.20474","DOIUrl":"10.21873/cgp.20474","url":null,"abstract":"<p><strong>Background/aim: </strong>Exosomes are extracellular vesicles produced by both normal and cancer cells. Previous research has demonstrated that circulating exosomes derived from cancer cells may create a niche for future metastasis, distant from the primary tumor. In the present report, circulating exosomes were captured and quantified based on exosome-surface proteins in pre- and post-operative serum of breast cancer patients, focusing on the exosome markers CD9 and CD63, as well as HER2, a therapeutic target for breast cancer.</p><p><strong>Materials and methods: </strong>Eight breast cancer patients were recruited, and their pre- and post-operative serum samples were analyzed for CD63 and CD9; or CD9 and human epidermal growth factor receptor-2 (HER2), double-positive exosomes. An ExoCounter with antibody-conjugated beads was used to capture serum-derived exosomes. Sera from patients with tumors larger than 10 mm were used for analysis. The resected breast cancer was also histopathologically analyzed for the presence of HER2.</p><p><strong>Results: </strong>CD63 and CD9 double-positive serum exosomes and CD9 and HER2 double-positive serum exosomes decreased after surgery in breast-cancer patients whose tumors expressed HER2, as determined by histopathological analysis.</p><p><strong>Conclusion: </strong>Serum exosomes expressing CD9, CD63 and HER2 are candidate biomarkers of tumor burden in HER2-positive breast-cancer patients.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"580-584"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534029/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521087","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparative Proteomics of ccRCC Cell Lines to Identify Kidney Cancer Progression Factors. ccRCC细胞系的比较蛋白质组学鉴定肾癌进展因素
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20480
Juhee Park, Hyunchae Sim, Eun Hye Lee, Bum Soo Kim, Jae-Wook Chung, Yun-Sok Ha, Tae Gyun Kwon, Sangkyu Lee, Jun Nyung Lee

Background/aim: Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer, accounting for approximately 75% of kidney cancers. The objective of this study was to identify novel progression markers for ccRCC based on proteomics, with the goal of stage determination and early diagnosis of kidney cancer patients.

Materials and methods: We performed quantitative global proteomics coupled with Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) and high-resolution tandem mass spectrometry on kidney-derived cells, including HEK-293, 786-O (primary ccRCC), and Caki-1 (metastatic ccRCC) cells, to investigate the novel progression factors of ccRCC.

Results: In this study, a total of 1,106 proteins were quantified. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted for differentially expressed proteins (DEPs) that were increased in ccRCC cells compared to HEK-293 cells. Ultimately, 99 DEPs including 75 up-regulated and 24 down-regulated proteins, that were significantly altered in both ccRCC cells, were identified. Among DEPs, vimentin was identified as the most significantly changed protein. Its increased expression in ccRCC was verified through immunoblotting in ccRCC cell lines and immunohistochemistry in kidney tumors.

Conclusion: From the global proteomics data detected in ccRCC, we propose 99 DEPs including vimentin as progression factors.

背景/目的:透明细胞肾细胞癌(ccRCC)是最常见的肾癌类型,约占肾癌的75%。本研究的目的是基于蛋白质组学鉴定ccRCC的新型进展标志物,从而对肾癌患者进行分期判断和早期诊断:我们对HEK-293、786-O(原发性ccRCC)和Caki-1(转移性ccRCC)细胞等肾源细胞进行了定量全局蛋白质组学研究,并结合细胞培养中氨基酸稳定同位素标记(SILAC)和高分辨率串联质谱分析,以研究ccRCC的新型进展因子:本研究共定量分析了 1,106 种蛋白质。与 HEK-293 细胞相比,ccRCC 细胞中增加的差异表达蛋白(DEPs)通过基因本体和京都基因组百科全书(KEGG)进行了分析。最终确定了 99 个 DEPs,包括 75 个上调蛋白和 24 个下调蛋白,它们在两种 ccRCC 细胞中都发生了显著变化。在 DEPs 中,波形蛋白被确定为变化最明显的蛋白质。通过免疫印迹法检测ccRCC细胞系和免疫组化法检测肾脏肿瘤,证实了波形蛋白在ccRCC中的表达增加:结论:从ccRCC中检测到的全局蛋白质组学数据中,我们发现包括波形蛋白在内的99种DEPs是导致ccRCC进展的因素。
{"title":"Comparative Proteomics of ccRCC Cell Lines to Identify Kidney Cancer Progression Factors.","authors":"Juhee Park, Hyunchae Sim, Eun Hye Lee, Bum Soo Kim, Jae-Wook Chung, Yun-Sok Ha, Tae Gyun Kwon, Sangkyu Lee, Jun Nyung Lee","doi":"10.21873/cgp.20480","DOIUrl":"10.21873/cgp.20480","url":null,"abstract":"<p><strong>Background/aim: </strong>Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer, accounting for approximately 75% of kidney cancers. The objective of this study was to identify novel progression markers for ccRCC based on proteomics, with the goal of stage determination and early diagnosis of kidney cancer patients.</p><p><strong>Materials and methods: </strong>We performed quantitative global proteomics coupled with Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) and high-resolution tandem mass spectrometry on kidney-derived cells, including HEK-293, 786-O (primary ccRCC), and Caki-1 (metastatic ccRCC) cells, to investigate the novel progression factors of ccRCC.</p><p><strong>Results: </strong>In this study, a total of 1,106 proteins were quantified. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted for differentially expressed proteins (DEPs) that were increased in ccRCC cells compared to HEK-293 cells. Ultimately, 99 DEPs including 75 up-regulated and 24 down-regulated proteins, that were significantly altered in both ccRCC cells, were identified. Among DEPs, vimentin was identified as the most significantly changed protein. Its increased expression in ccRCC was verified through immunoblotting in ccRCC cell lines and immunohistochemistry in kidney tumors.</p><p><strong>Conclusion: </strong>From the global proteomics data detected in ccRCC, we propose 99 DEPs including vimentin as progression factors.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"645-652"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534031/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SF3B4 Regulates Cellular Senescence and Suppresses Therapy-induced Senescence of Cancer Cells. SF3B4 调节细胞衰老并抑制治疗诱导的癌细胞衰老
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20478
Seungyeon Yang, Minbeom Ko, Soojung Claire Hur, Eun Kyung Lee, Seung Min Jeong

Background/aim: Cellular senescence is a state in which cells permanently exit the cell cycle, preventing tumor growth, but it can also contribute to aging and chronic inflammation. Senescence induced by cancer therapies, known as therapy-induced senescence (TIS), halts cancer cell proliferation and prevents metastasis. TIS has been investigated as an important therapeutic approach that could minimize cytotoxicity effects. This study aimed to elucidate the role of splicing factor 3B subunit 4 (SF3B4) in cellular senescence and TIS in cancer cells.

Materials and methods: β-galactosidase staining was used to examine senescence induction. SF3B4 and p21 expression were determined by RT-qPCR and western blot. Cell proliferation and cell death were evaluated.

Results: SF3B4 expression decreases in replicative senescent human fibroblasts and its knockdown induces senescence via a p21-dependent pathway. In A549 non-small cell lung cancer (NSCLC) cells, SF3B4 knockdown also increased senescence markers. Notably, SF3B4 overexpression mitigated doxorubicin-induced senescence in A549 cells.

Conclusion: SF3B4 regulates senescence, and this study highlights its potential as a therapeutic target for developing better cancer treatment strategies by leveraging TIS to suppress tumor growth and enhance treatment efficacy.

背景/目的:细胞衰老是细胞永久退出细胞周期的一种状态,可防止肿瘤生长,但也可导致衰老和慢性炎症。由癌症疗法诱导的衰老,即治疗诱导衰老(TIS),可阻止癌细胞增殖并防止转移。治疗诱导衰老是一种重要的治疗方法,可将细胞毒性效应降至最低。本研究旨在阐明剪接因子 3B 亚基 4(SF3B4)在癌细胞衰老和 TIS 中的作用。通过 RT-qPCR 和 Western 印迹检测 SF3B4 和 p21 的表达。对细胞增殖和细胞死亡进行了评估:结果:在复制衰老的人成纤维细胞中,SF3B4 的表达下降,其敲除通过 p21 依赖性途径诱导衰老。在 A549 非小细胞肺癌(NSCLC)细胞中,SF3B4 基因敲除也会增加衰老标志物。值得注意的是,SF3B4的过表达减轻了多柔比星诱导的A549细胞衰老:SF3B4调控衰老,本研究强调了其作为治疗靶点的潜力,通过利用TIS抑制肿瘤生长并提高治疗效果,从而开发出更好的癌症治疗策略。
{"title":"SF3B4 Regulates Cellular Senescence and Suppresses Therapy-induced Senescence of Cancer Cells.","authors":"Seungyeon Yang, Minbeom Ko, Soojung Claire Hur, Eun Kyung Lee, Seung Min Jeong","doi":"10.21873/cgp.20478","DOIUrl":"10.21873/cgp.20478","url":null,"abstract":"<p><strong>Background/aim: </strong>Cellular senescence is a state in which cells permanently exit the cell cycle, preventing tumor growth, but it can also contribute to aging and chronic inflammation. Senescence induced by cancer therapies, known as therapy-induced senescence (TIS), halts cancer cell proliferation and prevents metastasis. TIS has been investigated as an important therapeutic approach that could minimize cytotoxicity effects. This study aimed to elucidate the role of splicing factor 3B subunit 4 (SF3B4) in cellular senescence and TIS in cancer cells.</p><p><strong>Materials and methods: </strong>β-galactosidase staining was used to examine senescence induction. SF3B4 and p21 expression were determined by RT-qPCR and western blot. Cell proliferation and cell death were evaluated.</p><p><strong>Results: </strong>SF3B4 expression decreases in replicative senescent human fibroblasts and its knockdown induces senescence via a p21-dependent pathway. In A549 non-small cell lung cancer (NSCLC) cells, SF3B4 knockdown also increased senescence markers. Notably, SF3B4 overexpression mitigated doxorubicin-induced senescence in A549 cells.</p><p><strong>Conclusion: </strong>SF3B4 regulates senescence, and this study highlights its potential as a therapeutic target for developing better cancer treatment strategies by leveraging TIS to suppress tumor growth and enhance treatment efficacy.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"622-629"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534033/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521088","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Suppressing Expression of SERPINE1/PAI1 Through Activation of GPER1 Reduces Progression of Vulvar Carcinoma. 通过激活 GPER1 抑制 SERPINE1/PAI1 的表达可减轻外阴癌的恶化。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2024-11-01 DOI: 10.21873/cgp.20473
Tammy Doelker, Julia Gallwas, Carsten Gründker

Background/aim: The serine proteinase inhibitor 1 (SERPINE1) gene codes for the plasminogen activator inhibitor 1 (PAI1) protein and is thought to play a tumor supportive role in various cancers. In this work we aimed to uncover the role PAI1 plays in the proliferation, migration, and invasion of vulvar cancer (VC), and define the protein's function as an oncogene or tumor suppressor.

Materials and methods: Through treatment with an agonist (G1) and antagonist (G36) of G-coupled estrogen receptor 1 (GPER1), an upstream regulator of SERPINE1 expression, and a forward transfection knockdown protocol, the expression of SERPINE1/PAI1 in VC cells was altered. The effects these altered SERPINE1/PAI1 levels had on tumor cell functions were then examined. Proliferation was analyzed using the resazurin assay, while migration was studied via the gap closure assay. Through colony- and tumor sphere- formation assays clonogenicity was tested, and western blots showed protein expression.

Results: In A431 VC cells, when the levels of PAI1 were reduced via knockdown or treatment with G1, migration, proliferation, and colony growth was reduced. Treatment with G36 increased expression of PAI1 and increased migration and colony size in CAL39 cells.

Conclusion: Based on the findings in this study, suppressing PAI1 expression in VC cells appears to reduce their progression and tumorigenic potential. Therefore, PAI1 could possibly function as an oncogene in VC. GPER1 appears to be a suitable target for suppressing PAI1 in VC.

背景/目的:丝氨酸蛋白酶抑制剂 1(SERPINE1)基因编码纤溶酶原激活物抑制剂 1(PAI1)蛋白,被认为在多种癌症中发挥肿瘤支持作用。在这项工作中,我们旨在揭示 PAI1 在外阴癌(VC)的增殖、迁移和侵袭中所起的作用,并确定该蛋白作为癌基因或肿瘤抑制因子的功能:通过使用G-偶联雌激素受体1(GPER1)的激动剂(G1)和拮抗剂(G36)(GPER1是SERPINE1表达的上游调控因子)以及正向转染敲除方案,VC细胞中SERPINE1/PAI1的表达发生了改变。然后研究了这些改变的 SERPINE1/PAI1 水平对肿瘤细胞功能的影响。增殖是通过resazurin检测法进行分析的,而迁移则是通过缝隙闭合检测法进行研究的。通过集落和瘤球形成试验检测了克隆生成性,并通过 Western 印迹显示了蛋白质的表达:结果:在 A431 VC 细胞中,当 PAI1 的水平通过敲除或用 G1 处理而降低时,细胞的迁移、增殖和集落生长都会降低。结果:在 A431 VC 细胞中,当 PAI1 的水平通过敲除或 G1 处理而降低时,细胞的迁移、增殖和集落生长都会减少:结论:根据本研究的发现,抑制 PAI1 在 VC 细胞中的表达似乎能降低其进展和致瘤潜能。因此,PAI1 有可能是 VC 中的致癌基因。GPER1似乎是抑制PAI1在VC中表达的合适靶点。
{"title":"Suppressing Expression of SERPINE1/PAI1 Through Activation of GPER1 Reduces Progression of Vulvar Carcinoma.","authors":"Tammy Doelker, Julia Gallwas, Carsten Gründker","doi":"10.21873/cgp.20473","DOIUrl":"10.21873/cgp.20473","url":null,"abstract":"<p><strong>Background/aim: </strong>The serine proteinase inhibitor 1 (SERPINE1) gene codes for the plasminogen activator inhibitor 1 (PAI1) protein and is thought to play a tumor supportive role in various cancers. In this work we aimed to uncover the role PAI1 plays in the proliferation, migration, and invasion of vulvar cancer (VC), and define the protein's function as an oncogene or tumor suppressor.</p><p><strong>Materials and methods: </strong>Through treatment with an agonist (G1) and antagonist (G36) of G-coupled estrogen receptor 1 (GPER1), an upstream regulator of SERPINE1 expression, and a forward transfection knockdown protocol, the expression of SERPINE1/PAI1 in VC cells was altered. The effects these altered SERPINE1/PAI1 levels had on tumor cell functions were then examined. Proliferation was analyzed using the resazurin assay, while migration was studied via the gap closure assay. Through colony- and tumor sphere- formation assays clonogenicity was tested, and western blots showed protein expression.</p><p><strong>Results: </strong>In A431 VC cells, when the levels of PAI1 were reduced via knockdown or treatment with G1, migration, proliferation, and colony growth was reduced. Treatment with G36 increased expression of PAI1 and increased migration and colony size in CAL39 cells.</p><p><strong>Conclusion: </strong>Based on the findings in this study, suppressing PAI1 expression in VC cells appears to reduce their progression and tumorigenic potential. Therefore, PAI1 could possibly function as an oncogene in VC. GPER1 appears to be a suitable target for suppressing PAI1 in VC.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"21 6","pages":"566-579"},"PeriodicalIF":2.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11534035/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer Genomics & Proteomics
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