首页 > 最新文献

Biology Direct最新文献

英文 中文
Identification of metastasis-related genes for predicting prostate cancer diagnosis, metastasis and immunotherapy drug candidates using machine learning approaches. 利用机器学习方法鉴定转移相关基因,以预测前列腺癌诊断、转移和免疫疗法候选药物。
IF 5.7 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-25 DOI: 10.1186/s13062-024-00494-x
YaXuan Wang, Bo Ji, Lu Zhang, Jinfeng Wang, JiaXin He, BeiChen Ding, MingHua Ren

Background: Prostate cancer (PCa) is the second leading cause of tumor-related mortality in men. Metastasis from advanced tumors is the primary cause of death among patients. Identifying novel and effective biomarkers is essential for understanding the mechanisms of metastasis in PCa patients and developing successful interventions.

Methods: Using the GSE8511 and GSE27616 data sets, 21 metastasis-related genes were identified through the weighted gene co-expression network analysis (WGCNA) method. Subsequent functional analysis of these genes was conducted on the gene set cancer analysis (GSCA) website. Cluster analysis was utilized to explore the relationship between these genes, immune infiltration in PCa, and the efficacy of targeted drug IC50 scores. Machine learning algorithms were then employed to construct diagnostic and prognostic models, assessing their predictive accuracy. Additionally, multivariate COX regression analysis highlighted the significant role of POLD1 and examined its association with DNA methylation. Finally, molecular docking and immunohistochemistry experiments were carried out to assess the binding affinity of POLD1 to PCa drugs and its impact on PCa prognosis.

Results: The study identified 21 metastasis-related genes using the WGCNA method, which were found to be associated with DNA damage, hormone AR activation, and inhibition of the RTK pathway. Cluster analysis confirmed a significant correlation between these genes and PCa metastasis, particularly in the context of immunotherapy and targeted therapy drugs. A diagnostic model combining multiple machine learning algorithms showed strong predictive capabilities for PCa diagnosis, while a transfer model using the LASSO algorithm also yielded promising results. POLD1 emerged as a key prognostic gene among the metastatic genes, showing associations with DNA methylation. Molecular docking experiments supported its high affinity with PCa-targeted drugs. Immunohistochemistry experiments further validated that increased POLD1 expression is linked to poor prognosis in PCa patients.

Conclusions: The developed diagnostic and metastasis models provide substantial value for patients with prostate cancer. The discovery of POLD1 as a novel biomarker related to prostate cancer metastasis offers a promising avenue for enhancing treatment of prostate cancer metastasis.

背景:前列腺癌(PCa)是导致男性肿瘤相关死亡的第二大原因。晚期肿瘤的转移是导致患者死亡的主要原因。要想了解 PCa 患者的转移机制并制定成功的干预措施,识别新型有效的生物标记物至关重要:方法:利用 GSE8511 和 GSE27616 数据集,通过加权基因共表达网络分析(WGCNA)方法确定了 21 个转移相关基因。随后在基因组癌症分析(GSCA)网站上对这些基因进行了功能分析。利用聚类分析探讨了这些基因、PCa 中的免疫浸润和靶向药物 IC50 分疗效之间的关系。然后采用机器学习算法构建诊断和预后模型,评估其预测准确性。此外,多变量 COX 回归分析强调了 POLD1 的重要作用,并研究了其与 DNA 甲基化的关联。最后,还进行了分子对接和免疫组化实验,以评估POLD1与PCa药物的结合亲和力及其对PCa预后的影响:研究采用WGCNA方法鉴定了21个转移相关基因,发现这些基因与DNA损伤、激素AR激活和RTK通路抑制有关。聚类分析证实了这些基因与PCa转移之间的显著相关性,尤其是在免疫疗法和靶向治疗药物的背景下。一个结合了多种机器学习算法的诊断模型显示出了很强的PCa诊断预测能力,而一个使用LASSO算法的转移模型也取得了很好的结果。在转移基因中,POLD1 是一个关键的预后基因,显示出与 DNA 甲基化的关联。分子对接实验证明了它与 PCa 靶向药物的高亲和力。免疫组化实验进一步证实,POLD1的表达增加与PCa患者的不良预后有关:结论:所开发的诊断和转移模型对前列腺癌患者具有重要价值。结论:所开发的诊断和转移模型为前列腺癌患者提供了重要价值,POLD1作为与前列腺癌转移相关的新型生物标志物的发现为加强前列腺癌转移治疗提供了一个前景广阔的途径。
{"title":"Identification of metastasis-related genes for predicting prostate cancer diagnosis, metastasis and immunotherapy drug candidates using machine learning approaches.","authors":"YaXuan Wang, Bo Ji, Lu Zhang, Jinfeng Wang, JiaXin He, BeiChen Ding, MingHua Ren","doi":"10.1186/s13062-024-00494-x","DOIUrl":"10.1186/s13062-024-00494-x","url":null,"abstract":"<p><strong>Background: </strong>Prostate cancer (PCa) is the second leading cause of tumor-related mortality in men. Metastasis from advanced tumors is the primary cause of death among patients. Identifying novel and effective biomarkers is essential for understanding the mechanisms of metastasis in PCa patients and developing successful interventions.</p><p><strong>Methods: </strong>Using the GSE8511 and GSE27616 data sets, 21 metastasis-related genes were identified through the weighted gene co-expression network analysis (WGCNA) method. Subsequent functional analysis of these genes was conducted on the gene set cancer analysis (GSCA) website. Cluster analysis was utilized to explore the relationship between these genes, immune infiltration in PCa, and the efficacy of targeted drug IC50 scores. Machine learning algorithms were then employed to construct diagnostic and prognostic models, assessing their predictive accuracy. Additionally, multivariate COX regression analysis highlighted the significant role of POLD1 and examined its association with DNA methylation. Finally, molecular docking and immunohistochemistry experiments were carried out to assess the binding affinity of POLD1 to PCa drugs and its impact on PCa prognosis.</p><p><strong>Results: </strong>The study identified 21 metastasis-related genes using the WGCNA method, which were found to be associated with DNA damage, hormone AR activation, and inhibition of the RTK pathway. Cluster analysis confirmed a significant correlation between these genes and PCa metastasis, particularly in the context of immunotherapy and targeted therapy drugs. A diagnostic model combining multiple machine learning algorithms showed strong predictive capabilities for PCa diagnosis, while a transfer model using the LASSO algorithm also yielded promising results. POLD1 emerged as a key prognostic gene among the metastatic genes, showing associations with DNA methylation. Molecular docking experiments supported its high affinity with PCa-targeted drugs. Immunohistochemistry experiments further validated that increased POLD1 expression is linked to poor prognosis in PCa patients.</p><p><strong>Conclusions: </strong>The developed diagnostic and metastasis models provide substantial value for patients with prostate cancer. The discovery of POLD1 as a novel biomarker related to prostate cancer metastasis offers a promising avenue for enhancing treatment of prostate cancer metastasis.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"50"},"PeriodicalIF":5.7,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11197330/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449705","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Silencing LINC00987 ameliorates adriamycin resistance of acute myeloid leukemia via miR-4458/HMGA2 axis. 沉默LINC00987可通过miR-4458/HMGA2轴改善急性髓性白血病的阿霉素耐药性
IF 5.7 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-24 DOI: 10.1186/s13062-024-00490-1
Yue Liu, Xiao-Ya Zhu, Li-Li Liao, Zhan-Hui Zhang, Tao-Sheng Huang, Ling Zhang, Xi-Wen Jiang, Yi Ma

Background: Most patients with acute myeloid leukemia (AML) eventually develop drug resistance, leading to a poor prognosis. Dysregulated long gene non coding RNAs (lincRNAs) have been implicated in chemoresistance in AML. Unfortunately, the effects of lincRNAs which participate in regulating the Adriamycin (ADR) resistance in AML cells remain unclear. Thus, the purpose of this study is to determine LINC00987 function in ADR-resistant AML.

Methods: In this study, ADR-resistant cells were constructed. LINC00987, miRNAs, and HMGA2 mRNA expression were measured by qRT-PCR. P-GP, BCRP, and HMGA2 protein were measured by Western blot. The proliferation was analyzed by MTS and calculated IC50. Soft agar colony formation assay and TUNEL staining were used to analyze cell colony formation and apoptosis. Xenograft tumor experiment was used to analyze the xenograft tumor growth of ADR-resistant AML.

Results: We found that higher expression of LINC00987 was observed in AML patients and associated with poor overall survival in AML patients. LINC00987 expression was increased in ADR-resistant AML cells, including ADR/MOLM13 and ADR/HL-60 cells. LINC00987 downregulation reduces ADR resistance in ADR/MOLM13 and ADR/HL-60 cells in vitro and in vivo, while LINC00987 overexpression enhanced ADR resistance in MOLM13 and HL-60 cells. Additionally, LINC00987 functions as a competing endogenous RNA for miR-4458 to affect ADR resistance in ADR/MOLM13 and ADR/HL-60 cells. HMGA2 is a target of miR-4458. LINC00987 knockdown and miR-4458 overexpression reduced HMGA2 expression. HMGA2 overexpression enhanced ADR resistance, which reversed the function of LINC00987 silencing in suppressing ADR resistance of ADR/MOLM13 and ADR/HL-60 cells.

Conclusions: Downregulation of LINC00987 weakens ADR resistance by releasing miR-4458 to deplete HMGA2 in ADR/MOLM13 and ADR/HL-60. Therefore, LINC00987 may act as the therapeutic target for treating chemoresistant AML.

背景:大多数急性髓性白血病(AML)患者最终会产生耐药性,导致不良预后。长基因非编码 RNA(lincRNA)的失调与急性髓性白血病的化疗耐药性有关。遗憾的是,参与调节急性髓细胞白血病细胞阿霉素(ADR)耐药性的长基因非编码 RNAs 的作用仍不清楚。因此,本研究旨在确定LINC00987在ADR耐药AML中的功能:方法:本研究构建了 ADR 抗性细胞。通过 qRT-PCR 检测 LINC00987、miRNAs 和 HMGA2 mRNA 的表达。通过 Western 印迹检测 P-GP、BCRP 和 HMGA2 蛋白。用 MTS 分析增殖并计算 IC50。软琼脂集落形成试验和 TUNEL 染色用于分析细胞集落形成和凋亡。异种移植肿瘤实验用于分析ADR耐药AML的异种移植肿瘤生长情况:结果:我们发现 LINC00987 在急性髓细胞性白血病患者中表达较高,并且与急性髓细胞性白血病患者的总生存率较低有关。LINC00987在ADR耐药的AML细胞(包括ADR/MOLM13和ADR/HL-60细胞)中表达增加。LINC00987 下调可降低 ADR/MOLM13 和 ADR/HL-60 细胞在体外和体内对 ADR 的耐药性,而 LINC00987 过表达则会增强 MOLM13 和 HL-60 细胞对 ADR 的耐药性。此外,LINC00987作为miR-4458的竞争内源RNA,影响了ADR/MOLM13和ADR/HL-60细胞的ADR耐药性。HMGA2是miR-4458的靶标。LINC00987 敲除和 miR-4458 过表达会降低 HMGA2 的表达。HMGA2的过表达增强了ADR耐药性,从而逆转了LINC00987沉默抑制ADR/MOLM13和ADR/HL-60细胞ADR耐药性的功能:结论:在ADR/MOLM13和ADR/HL-60细胞中,下调LINC00987可通过释放miR-4458来消耗HMGA2,从而削弱ADR耐药性。因此,LINC00987 可作为治疗化疗耐药 AML 的靶点。
{"title":"Silencing LINC00987 ameliorates adriamycin resistance of acute myeloid leukemia via miR-4458/HMGA2 axis.","authors":"Yue Liu, Xiao-Ya Zhu, Li-Li Liao, Zhan-Hui Zhang, Tao-Sheng Huang, Ling Zhang, Xi-Wen Jiang, Yi Ma","doi":"10.1186/s13062-024-00490-1","DOIUrl":"10.1186/s13062-024-00490-1","url":null,"abstract":"<p><strong>Background: </strong>Most patients with acute myeloid leukemia (AML) eventually develop drug resistance, leading to a poor prognosis. Dysregulated long gene non coding RNAs (lincRNAs) have been implicated in chemoresistance in AML. Unfortunately, the effects of lincRNAs which participate in regulating the Adriamycin (ADR) resistance in AML cells remain unclear. Thus, the purpose of this study is to determine LINC00987 function in ADR-resistant AML.</p><p><strong>Methods: </strong>In this study, ADR-resistant cells were constructed. LINC00987, miRNAs, and HMGA2 mRNA expression were measured by qRT-PCR. P-GP, BCRP, and HMGA2 protein were measured by Western blot. The proliferation was analyzed by MTS and calculated IC50. Soft agar colony formation assay and TUNEL staining were used to analyze cell colony formation and apoptosis. Xenograft tumor experiment was used to analyze the xenograft tumor growth of ADR-resistant AML.</p><p><strong>Results: </strong>We found that higher expression of LINC00987 was observed in AML patients and associated with poor overall survival in AML patients. LINC00987 expression was increased in ADR-resistant AML cells, including ADR/MOLM13 and ADR/HL-60 cells. LINC00987 downregulation reduces ADR resistance in ADR/MOLM13 and ADR/HL-60 cells in vitro and in vivo, while LINC00987 overexpression enhanced ADR resistance in MOLM13 and HL-60 cells. Additionally, LINC00987 functions as a competing endogenous RNA for miR-4458 to affect ADR resistance in ADR/MOLM13 and ADR/HL-60 cells. HMGA2 is a target of miR-4458. LINC00987 knockdown and miR-4458 overexpression reduced HMGA2 expression. HMGA2 overexpression enhanced ADR resistance, which reversed the function of LINC00987 silencing in suppressing ADR resistance of ADR/MOLM13 and ADR/HL-60 cells.</p><p><strong>Conclusions: </strong>Downregulation of LINC00987 weakens ADR resistance by releasing miR-4458 to deplete HMGA2 in ADR/MOLM13 and ADR/HL-60. Therefore, LINC00987 may act as the therapeutic target for treating chemoresistant AML.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"49"},"PeriodicalIF":5.7,"publicationDate":"2024-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11195003/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141442174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PNSC928, a plant-derived compound, specifically disrupts CtBP2-p300 interaction and reduces inflammation in mice with acute respiratory distress syndrome. PNSC928 是一种植物提取的化合物,它能特异性地破坏 CtBP2-p300 的相互作用,减轻急性呼吸窘迫综合征小鼠的炎症反应。
IF 5.7 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-21 DOI: 10.1186/s13062-024-00491-0
Fan Li, Wenqing Yan, Weihua Dong, Zhiping Chen, Zhi Chen

Background: Prior research has highlighted the involvement of a transcriptional complex comprising C-terminal binding protein 2 (CtBP2), histone acetyltransferase p300, and nuclear factor kappa B (NF-κB) in the transactivation of proinflammatory cytokine genes, contributing to inflammation in mice with acute respiratory distress syndrome (ARDS). Nonetheless, it remains uncertain whether the therapeutic targeting of the CtBP2-p300-NF-κB complex holds potential for ARDS suppression.

Methods: An ARDS mouse model was established using lipopolysaccharide (LPS) exposure. RNA-Sequencing (RNA-Seq) was performed on ARDS mice and LPS-treated cells with CtBP2, p300, and p65 knockdown. Small molecules inhibiting the CtBP2-p300 interaction were identified through AlphaScreen. Gene and protein expression levels were quantified using RT-qPCR and immunoblots. Tissue damage was assessed via histological staining.

Key findings: We elucidated the specific role of the CtBP2-p300-NF-κB complex in proinflammatory gene regulation. RNA-seq analysis in LPS-challenged ARDS mice and LPS-treated CtBP2-knockdown (CtBP2KD), p300KD, and p65KD cells revealed its significant impact on proinflammatory genes with minimal effects on other NF-κB targets. Commercial inhibitors for CtBP2, p300, or NF-κB exhibited moderate cytotoxicity in vitro and in vivo, affecting both proinflammatory genes and other targets. We identified a potent inhibitor, PNSC928, for the CtBP2-p300 interaction using AlphaScreen. PNSC928 treatment hindered the assembly of the CtBP2-p300-NF-κB complex, substantially downregulating proinflammatory cytokine gene expression without observable cytotoxicity in normal cells. In vivo administration of PNSC928 significantly reduced CtBP2-driven proinflammatory gene expression in ARDS mice, alleviating inflammation and lung injury, ultimately improving ARDS prognosis.

Conclusion: Our results position PNSC928 as a promising therapeutic candidate to specifically target the CtBP2-p300 interaction and mitigate inflammation in ARDS management.

背景:先前的研究强调,由 C 端结合蛋白 2 (CtBP2)、组蛋白乙酰转移酶 p300 和核因子卡巴 B (NF-κB) 组成的转录复合物参与了促炎细胞因子基因的转录激活,导致了急性呼吸窘迫综合征 (ARDS) 小鼠的炎症。然而,针对 CtBP2-p300-NF-κB 复合物的治疗是否具有抑制 ARDS 的潜力仍不确定:方法:利用脂多糖(LPS)暴露建立 ARDS 小鼠模型。方法:利用脂多糖(LPS)暴露建立了ARDS小鼠模型,对ARDS小鼠和LPS处理过的细胞进行了RNA测序(RNA-Seq),并敲除了CtBP2、p300和p65。通过 AlphaScreen 鉴定了抑制 CtBP2-p300 相互作用的小分子。基因和蛋白质表达水平通过 RT-qPCR 和免疫印迹进行量化。组织损伤通过组织学染色进行评估:我们阐明了 CtBP2-p300-NF-κB 复合物在促炎基因调控中的特殊作用。在LPS挑战的ARDS小鼠和LPS处理的CtBP2-敲除(CtBP2KD)、p300KD和p65KD细胞中进行的RNA-seq分析显示,它对促炎基因有显著影响,而对其他NF-κB靶点的影响则微乎其微。CtBP2、p300 或 NF-κB 的商用抑制剂在体外和体内都表现出中等程度的细胞毒性,对促炎基因和其他靶点都有影响。我们利用 AlphaScreen 发现了一种 CtBP2-p300 相互作用的强效抑制剂 PNSC928。PNSC928 的处理阻碍了 CtBP2-p300-NF-κB 复合物的组装,从而大大降低了促炎细胞因子基因的表达,而且在正常细胞中没有明显的细胞毒性。体内给药 PNSC928 能显著降低 ARDS 小鼠体内 CtBP2 驱动的促炎基因表达,减轻炎症和肺损伤,最终改善 ARDS 的预后:我们的研究结果表明,PNSC928 是一种很有前景的候选疗法,它能特异性地靶向 CtBP2-p300 相互作用,减轻 ARDS 治疗中的炎症反应。
{"title":"PNSC928, a plant-derived compound, specifically disrupts CtBP2-p300 interaction and reduces inflammation in mice with acute respiratory distress syndrome.","authors":"Fan Li, Wenqing Yan, Weihua Dong, Zhiping Chen, Zhi Chen","doi":"10.1186/s13062-024-00491-0","DOIUrl":"10.1186/s13062-024-00491-0","url":null,"abstract":"<p><strong>Background: </strong>Prior research has highlighted the involvement of a transcriptional complex comprising C-terminal binding protein 2 (CtBP2), histone acetyltransferase p300, and nuclear factor kappa B (NF-κB) in the transactivation of proinflammatory cytokine genes, contributing to inflammation in mice with acute respiratory distress syndrome (ARDS). Nonetheless, it remains uncertain whether the therapeutic targeting of the CtBP2-p300-NF-κB complex holds potential for ARDS suppression.</p><p><strong>Methods: </strong>An ARDS mouse model was established using lipopolysaccharide (LPS) exposure. RNA-Sequencing (RNA-Seq) was performed on ARDS mice and LPS-treated cells with CtBP2, p300, and p65 knockdown. Small molecules inhibiting the CtBP2-p300 interaction were identified through AlphaScreen. Gene and protein expression levels were quantified using RT-qPCR and immunoblots. Tissue damage was assessed via histological staining.</p><p><strong>Key findings: </strong>We elucidated the specific role of the CtBP2-p300-NF-κB complex in proinflammatory gene regulation. RNA-seq analysis in LPS-challenged ARDS mice and LPS-treated CtBP2-knockdown (CtBP2<sup>KD</sup>), p300<sup>KD</sup>, and p65<sup>KD</sup> cells revealed its significant impact on proinflammatory genes with minimal effects on other NF-κB targets. Commercial inhibitors for CtBP2, p300, or NF-κB exhibited moderate cytotoxicity in vitro and in vivo, affecting both proinflammatory genes and other targets. We identified a potent inhibitor, PNSC928, for the CtBP2-p300 interaction using AlphaScreen. PNSC928 treatment hindered the assembly of the CtBP2-p300-NF-κB complex, substantially downregulating proinflammatory cytokine gene expression without observable cytotoxicity in normal cells. In vivo administration of PNSC928 significantly reduced CtBP2-driven proinflammatory gene expression in ARDS mice, alleviating inflammation and lung injury, ultimately improving ARDS prognosis.</p><p><strong>Conclusion: </strong>Our results position PNSC928 as a promising therapeutic candidate to specifically target the CtBP2-p300 interaction and mitigate inflammation in ARDS management.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"48"},"PeriodicalIF":5.7,"publicationDate":"2024-06-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11191317/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141431392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The deubiquitinating protein OTUD6B promotes lung adenocarcinoma progression by stabilizing RIPK1. 去泛素化蛋白OTUD6B通过稳定RIPK1促进肺腺癌的进展。
IF 5.7 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-16 DOI: 10.1186/s13062-024-00489-8
Miaomiao Yang, Yujie Wei, Xin He, Changwei Xia

Background: There is growing evidence indicating that deubiquitinating enzymes may contribute to tumor progression and can serve as promising therapeutic targets.

Methods: The overexpression of deubiquitinase OTUD6B in lung adenocarcinoma (LUAD) and its adjacent tissues was analyzed by immunohistochemistry and TCGA/GO database. Survival analysis further supported OTUD6B as a potential target for LUAD treatment. We assessed the effect of OTUD6B on LUAD cell growth using cell viability assays and conducted TUNEL staining, migration, and invasion experiments to investigate the impact of OTUD6B on the apoptosis and metastasis of LUAD cells. Additionally, we established a transplanted tumor model in nude mice to validate our findings in vivo. Finally, using IP mass spectrometry and co-IP experiments, we screened and confirmed the influence of RIPK1 as a substrate of OTUD6B in LUAD.

Results: OTUD6B is highly overexpressed in human LUAD and predicts poor prognosis in LUAD patients. OTUD6B knockdown inhibited the proliferation of LUAD cells and enhanced apoptosis and inhibited metastasis in LUAD cells suppressed. A549 xenografts revealed that OTUD6B deletion can slow down tumour growth. Additionally, OTUD6B can bind to RIPK1, reduce its ubiquitination level and increase its protein stability.

Conclusions: Our results suggest that OTUD6B is a promising clinical target for LUAD treatment and that targeting OTUD6B may constitute an effective anti-LUAD strategy.

背景:越来越多的证据表明,去泛素化酶可能导致肿瘤进展,并可作为有前景的治疗靶点:方法:通过免疫组化和TCGA/GO数据库分析了去泛素化酶OTUD6B在肺腺癌(LUAD)及其邻近组织中的过表达。存活率分析进一步支持 OTUD6B 成为治疗 LUAD 的潜在靶点。我们利用细胞活力测定评估了 OTUD6B 对 LUAD 细胞生长的影响,并进行了 TUNEL 染色、迁移和侵袭实验,以研究 OTUD6B 对 LUAD 细胞凋亡和转移的影响。此外,我们还建立了裸鼠移植肿瘤模型,以在体内验证我们的发现。最后,我们利用 IP 质谱和共 IP 实验筛选并证实了 RIPK1 作为 OTUD6B 底物对 LUAD 的影响:结果:OTUD6B在人类LUAD中高度过表达,并预示着LUAD患者的不良预后。敲除 OTUD6B 可抑制 LUAD 细胞的增殖,增强细胞凋亡,抑制 LUAD 细胞的转移。A549异种移植显示,OTUD6B缺失可减缓肿瘤生长。此外,OTUD6B还能与RIPK1结合,降低其泛素化水平,增加其蛋白稳定性:我们的研究结果表明,OTUD6B是一个很有前景的治疗LUAD的临床靶点,靶向OTUD6B可能是一种有效的抗LUAD策略。
{"title":"The deubiquitinating protein OTUD6B promotes lung adenocarcinoma progression by stabilizing RIPK1.","authors":"Miaomiao Yang, Yujie Wei, Xin He, Changwei Xia","doi":"10.1186/s13062-024-00489-8","DOIUrl":"10.1186/s13062-024-00489-8","url":null,"abstract":"<p><strong>Background: </strong>There is growing evidence indicating that deubiquitinating enzymes may contribute to tumor progression and can serve as promising therapeutic targets.</p><p><strong>Methods: </strong>The overexpression of deubiquitinase OTUD6B in lung adenocarcinoma (LUAD) and its adjacent tissues was analyzed by immunohistochemistry and TCGA/GO database. Survival analysis further supported OTUD6B as a potential target for LUAD treatment. We assessed the effect of OTUD6B on LUAD cell growth using cell viability assays and conducted TUNEL staining, migration, and invasion experiments to investigate the impact of OTUD6B on the apoptosis and metastasis of LUAD cells. Additionally, we established a transplanted tumor model in nude mice to validate our findings in vivo. Finally, using IP mass spectrometry and co-IP experiments, we screened and confirmed the influence of RIPK1 as a substrate of OTUD6B in LUAD.</p><p><strong>Results: </strong>OTUD6B is highly overexpressed in human LUAD and predicts poor prognosis in LUAD patients. OTUD6B knockdown inhibited the proliferation of LUAD cells and enhanced apoptosis and inhibited metastasis in LUAD cells suppressed. A549 xenografts revealed that OTUD6B deletion can slow down tumour growth. Additionally, OTUD6B can bind to RIPK1, reduce its ubiquitination level and increase its protein stability.</p><p><strong>Conclusions: </strong>Our results suggest that OTUD6B is a promising clinical target for LUAD treatment and that targeting OTUD6B may constitute an effective anti-LUAD strategy.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"46"},"PeriodicalIF":5.7,"publicationDate":"2024-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11181667/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141330343","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hypoxia-induced activation of HIF-1alpha/IL-1beta axis in microglia promotes glioma progression via NF-κB-mediated upregulation of heparanase expression. 缺氧诱导的小胶质细胞 HIF-1alpha/IL-1beta 轴激活通过 NF-κB 介导的肝素酶表达上调促进胶质瘤进展。
IF 5.7 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-11 DOI: 10.1186/s13062-024-00487-w
Jinchao Si, Jingya Guo, Xu Zhang, Wei Li, Shen Zhang, Shuyu Shang, Quanwu Zhang

Background: Glioma is a common tumor that occurs in the brain and spinal cord. Hypoxia is a crucial feature of the tumor microenvironment. Tumor-associated macrophages/microglia play a crucial role in the advancement of glioma. This study aims to illuminate the detailed mechanisms by which hypoxia regulates microglia and, consequently, influences the progression of glioma.

Methods: The glioma cell viability and proliferation were analyzed by cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine assay. Wound healing assay and transwell assay were implemented to detect glioma cell migration and invasion, respectively. Enzyme-linked immunosorbent assay was conducted to detect protein levels in cell culture medium. The protein levels in glioma cells and tumor tissues were evaluated using western blot analysis. The histological morphology of tumor tissue was determined by hematoxylin-eosin staining. The protein expression in tumor tissues was determined using immunohistochemistry. Human glioma xenograft in nude mice was employed to test the influence of hypoxic microglia-derived interleukin-1beta (IL-1β) and heparanase (HPSE) on glioma growth in vivo.

Results: Hypoxic HMC3 cells promoted proliferation, migration, and invasion abilities of U251 and U87 cells by secreting IL-1β, which was upregulated by hypoxia-induced activation of hypoxia inducible factor-1alpha (HIF-1α). Besides, IL-1β from HMC3 cells promoted glioma progression and caused activation of nuclear factor-κB (NF-κB) and upregulation of HPSE in vivo. We also confirmed that IL-1β facilitated HPSE expression in U251 and U87 cells by activating NF-κB. Hypoxic HMC3 cells-secreted IL-1β facilitated the proliferation, migration, and invasion of U251 and U87 cells via NF-κB-mediated upregulation of HPSE expression. Finally, we revealed that silencing HPSE curbed the proliferation and metastasis of glioma in mice.

Conclusion: Hypoxia-induced activation of HIF-1α/IL-1β axis in microglia promoted glioma progression via NF-κB-mediated upregulation of HPSE expression.

背景:胶质瘤是发生在大脑和脊髓的一种常见肿瘤。缺氧是肿瘤微环境的一个重要特征。肿瘤相关巨噬细胞/小胶质细胞在胶质瘤的发展过程中起着至关重要的作用。本研究旨在阐明缺氧调节小胶质细胞并进而影响胶质瘤进展的详细机制:方法:通过细胞计数试剂盒-8测定法和5-乙炔基-2'-脱氧尿苷测定法分析胶质瘤细胞的活力和增殖情况。伤口愈合试验和透孔试验分别检测胶质瘤细胞的迁移和侵袭。酶联免疫吸附试验检测细胞培养基中的蛋白质水平。胶质瘤细胞和肿瘤组织中的蛋白质水平则通过 Western 印迹分析进行评估。肿瘤组织的组织学形态由苏木精-伊红染色确定。肿瘤组织中的蛋白质表达采用免疫组化法测定。采用裸鼠进行人胶质瘤异种移植,检测缺氧小胶质细胞衍生的白细胞介素-1β(IL-1β)和肝聚糖酶(HPSE)对胶质瘤体内生长的影响:结果:缺氧的HMC3细胞通过分泌IL-1β促进了U251和U87细胞的增殖、迁移和侵袭能力。此外,来自HMC3细胞的IL-1β促进了胶质瘤的进展,并导致核因子-κB(NF-κB)的活化和体内HPSE的上调。我们还证实,IL-1β通过激活NF-κB促进了HPSE在U251和U87细胞中的表达。缺氧的HMC3细胞分泌的IL-1β通过NF-κB介导的HPSE表达上调促进了U251和U87细胞的增殖、迁移和侵袭。最后,我们发现沉默 HPSE 可抑制小鼠胶质瘤的增殖和转移:结论:缺氧诱导的小胶质细胞HIF-1α/IL-1β轴激活通过NF-κB介导的HPSE表达上调促进了胶质瘤的进展。
{"title":"Hypoxia-induced activation of HIF-1alpha/IL-1beta axis in microglia promotes glioma progression via NF-κB-mediated upregulation of heparanase expression.","authors":"Jinchao Si, Jingya Guo, Xu Zhang, Wei Li, Shen Zhang, Shuyu Shang, Quanwu Zhang","doi":"10.1186/s13062-024-00487-w","DOIUrl":"10.1186/s13062-024-00487-w","url":null,"abstract":"<p><strong>Background: </strong>Glioma is a common tumor that occurs in the brain and spinal cord. Hypoxia is a crucial feature of the tumor microenvironment. Tumor-associated macrophages/microglia play a crucial role in the advancement of glioma. This study aims to illuminate the detailed mechanisms by which hypoxia regulates microglia and, consequently, influences the progression of glioma.</p><p><strong>Methods: </strong>The glioma cell viability and proliferation were analyzed by cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine assay. Wound healing assay and transwell assay were implemented to detect glioma cell migration and invasion, respectively. Enzyme-linked immunosorbent assay was conducted to detect protein levels in cell culture medium. The protein levels in glioma cells and tumor tissues were evaluated using western blot analysis. The histological morphology of tumor tissue was determined by hematoxylin-eosin staining. The protein expression in tumor tissues was determined using immunohistochemistry. Human glioma xenograft in nude mice was employed to test the influence of hypoxic microglia-derived interleukin-1beta (IL-1β) and heparanase (HPSE) on glioma growth in vivo.</p><p><strong>Results: </strong>Hypoxic HMC3 cells promoted proliferation, migration, and invasion abilities of U251 and U87 cells by secreting IL-1β, which was upregulated by hypoxia-induced activation of hypoxia inducible factor-1alpha (HIF-1α). Besides, IL-1β from HMC3 cells promoted glioma progression and caused activation of nuclear factor-κB (NF-κB) and upregulation of HPSE in vivo. We also confirmed that IL-1β facilitated HPSE expression in U251 and U87 cells by activating NF-κB. Hypoxic HMC3 cells-secreted IL-1β facilitated the proliferation, migration, and invasion of U251 and U87 cells via NF-κB-mediated upregulation of HPSE expression. Finally, we revealed that silencing HPSE curbed the proliferation and metastasis of glioma in mice.</p><p><strong>Conclusion: </strong>Hypoxia-induced activation of HIF-1α/IL-1β axis in microglia promoted glioma progression via NF-κB-mediated upregulation of HPSE expression.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"45"},"PeriodicalIF":5.7,"publicationDate":"2024-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11165725/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141305422","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
METTL3 promotes the progression of osteosarcoma through the N6-methyladenosine modification of MCAM via IGF2BP1. METTL3通过IGF2BP1对MCAM进行N6-甲基腺苷修饰,从而促进骨肉瘤的进展。
IF 5.5 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-07 DOI: 10.1186/s13062-024-00486-x
Dongjian Song, Qi Wang, Zechen Yan, Meng Su, Hui Zhang, Longyan Shi, Yingzhong Fan, Qian Zhang, Heying Yang, Da Zhang, Qiuliang Liu

Background: The molecular mechanisms of osteosarcoma (OS) are complex. In this study, we focused on the functions of melanoma cell adhesion molecule (MCAM), methyltransferase 3 (METTL3) and insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) in OS development.

Methods: qRT-PCR assay and western blot assay were performed to determine mRNA and protein expression of MCAM, METTL3, IGF2BP1 and YY1. MTT assay and colony formation assay were conducted to assess cell proliferation. Cell apoptosis, invasion and migration were evaluated by flow cytometry analysis, transwell assay and wound-healing assay, respectively. Methylated RNA Immunoprecipitation (MeRIP), dual-luciferase reporter, Co-IP, RIP and ChIP assays were performed to analyze the relationships of MCAM, METTL3, IGF2BP1 and YY1. The functions of METTL3 and MCAM in tumor growth were explored through in vivo experiments.

Results: MCAM was upregulated in OS, and MCAM overexpression promoted OS cell growth, invasion and migration and inhibited apoptosis. METTL3 and IGF2BP1 were demonstrated to mediate the m6A methylation of MCAM. Functionally, METTL3 or IGF2BP1 silencing inhibited OS cell progression, while MCAM overexpression ameliorated the effects. Transcription factor YY1 promoted the transcription level of METTL3 and regulated METTL3 expression in OS cells. Additionally, METTL3 deficiency suppressed tumor growth in vivo, while MCAM overexpression abated the effect.

Conclusion: YY1/METTL3/IGF2BP1/MCAM axis aggravated OS development, which might provide novel therapy targets for OS.

背景:骨肉瘤(OS)的分子机制十分复杂。方法:通过 qRT-PCR 检测和 Western 印迹检测确定 MCAM、METTL3、IGF2BP1 和 YY1 的 mRNA 和蛋白表达。MTT 试验和集落形成试验用于评估细胞增殖。细胞凋亡、侵袭和迁移分别通过流式细胞仪分析、Transwell 试验和伤口愈合试验进行评估。通过甲基化 RNA 免疫沉淀(MeRIP)、双荧光素酶报告、Co-IP、RIP 和 ChIP 检测分析 MCAM、METTL3、IGF2BP1 和 YY1 的关系。通过体内实验探讨了METTL3和MCAM在肿瘤生长中的功能:结果:MCAM在OS中上调,MCAM过表达促进OS细胞的生长、侵袭和迁移,并抑制细胞凋亡。METTL3和IGF2BP1被证实介导了MCAM的m6A甲基化。在功能上,沉默METTL3或IGF2BP1可抑制OS细胞的进展,而过表达MCAM则可改善其效果。转录因子YY1可促进METTL3的转录水平,并调节METTL3在OS细胞中的表达。此外,METTL3缺乏可抑制肿瘤在体内的生长,而MCAM过表达则可减轻这种影响:结论:YY1/METTL3/IGF2BP1/MCAM轴加重了OS的发展,可能为OS提供了新的治疗靶点。
{"title":"METTL3 promotes the progression of osteosarcoma through the N6-methyladenosine modification of MCAM via IGF2BP1.","authors":"Dongjian Song, Qi Wang, Zechen Yan, Meng Su, Hui Zhang, Longyan Shi, Yingzhong Fan, Qian Zhang, Heying Yang, Da Zhang, Qiuliang Liu","doi":"10.1186/s13062-024-00486-x","DOIUrl":"10.1186/s13062-024-00486-x","url":null,"abstract":"<p><strong>Background: </strong>The molecular mechanisms of osteosarcoma (OS) are complex. In this study, we focused on the functions of melanoma cell adhesion molecule (MCAM), methyltransferase 3 (METTL3) and insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) in OS development.</p><p><strong>Methods: </strong>qRT-PCR assay and western blot assay were performed to determine mRNA and protein expression of MCAM, METTL3, IGF2BP1 and YY1. MTT assay and colony formation assay were conducted to assess cell proliferation. Cell apoptosis, invasion and migration were evaluated by flow cytometry analysis, transwell assay and wound-healing assay, respectively. Methylated RNA Immunoprecipitation (MeRIP), dual-luciferase reporter, Co-IP, RIP and ChIP assays were performed to analyze the relationships of MCAM, METTL3, IGF2BP1 and YY1. The functions of METTL3 and MCAM in tumor growth were explored through in vivo experiments.</p><p><strong>Results: </strong>MCAM was upregulated in OS, and MCAM overexpression promoted OS cell growth, invasion and migration and inhibited apoptosis. METTL3 and IGF2BP1 were demonstrated to mediate the m6A methylation of MCAM. Functionally, METTL3 or IGF2BP1 silencing inhibited OS cell progression, while MCAM overexpression ameliorated the effects. Transcription factor YY1 promoted the transcription level of METTL3 and regulated METTL3 expression in OS cells. Additionally, METTL3 deficiency suppressed tumor growth in vivo, while MCAM overexpression abated the effect.</p><p><strong>Conclusion: </strong>YY1/METTL3/IGF2BP1/MCAM axis aggravated OS development, which might provide novel therapy targets for OS.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"44"},"PeriodicalIF":5.5,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11157866/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141287784","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
M2 macrophage-derived exosomes promote angiogenesis and improve cardiac function after myocardial infarction. M2巨噬细胞衍生的外泌体促进血管生成并改善心肌梗死后的心脏功能
IF 5.7 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-06 DOI: 10.1186/s13062-024-00485-y
Hongzhou Guo, Zeya Li, Bin Xiao, Rongchong Huang

Background: Myocardial infarction (MI) is a major cause of mortality and morbidity worldwide. The intercellular communication in post-infarction angiogenesis remains unclear.

Methods: In this study, we explored the role and mechanism of action of M2 macrophage-derived exosomes (M2-exos) in angiogenesis after MI. M2-exos were harvested and injected intramyocardially at the onset of MI. Two distinct endothelial cells (ECs) were cultured with M2-exos to explore the direct effects on angiogenesis.

Results: We showed that M2-exos improved cardiac function, reduced infarct size, and enhanced angiogenesis after MI. Moreover, M2-exos promoted angiogenesis in vitro; the molecules loaded in the vesicles were responsible for its proangiogenic effects. We further validated that higher abundance of miR-132-3p in M2-exos, which recapitulate their functions, was required for the cardioprotective effects exerted by M2-exos. Mechanistically, miR-132-3p carried by M2-exos down-regulate the expression of THBS1 through direct binding to its 3´UTR and the proangiogenic effects of miR-132-3p were largely reversed by THBS1 overexpression.

Conclusion: Our findings demonstrate that M2-exos promote angiogenesis after MI by transporting miR-132-3p to ECs, and by binding to THBS1 mRNA directly and negatively regulating its expression. These findings highlight the role of M2-exos in cardiac repair and provide novel mechanistic understanding of intercellular communication in post-infarction angiogenesis.

背景:心肌梗死(MI)是全球死亡和发病的主要原因。心肌梗死后血管生成过程中的细胞间通讯仍不清楚:在这项研究中,我们探讨了 M2 巨噬细胞衍生的外泌体(M2-exos)在心肌梗死后血管生成中的作用和作用机制。我们在心肌梗死发生时收获了M2-外泌体并将其注入心肌内。用M2-外泌体培养两种不同的内皮细胞(EC),以探讨其对血管生成的直接影响:结果:我们发现,M2-exos能改善心肌梗死后的心脏功能、缩小梗死面积并促进血管生成。此外,M2-exos 还能促进体外血管生成;囊泡中装载的分子是其促进血管生成作用的原因。我们进一步验证,M2-exos发挥心脏保护作用需要M2-exos中较高丰度的miR-132-3p,它再现了M2-exos的功能。从机制上讲,M2-exos携带的miR-132-3p通过直接与THBS1的3´UTR结合下调THBS1的表达,而THBS1的过表达在很大程度上逆转了miR-132-3p的促血管生成作用:我们的研究结果表明,M2-外显子通过将 miR-132-3p 运送到心肌细胞,并直接与 THBS1 mRNA 结合,负向调节其表达,从而促进心肌梗死后的血管生成。这些发现凸显了M2-exos在心脏修复中的作用,并提供了对心肌梗死后血管生成过程中细胞间通讯的新的机理认识。
{"title":"M2 macrophage-derived exosomes promote angiogenesis and improve cardiac function after myocardial infarction.","authors":"Hongzhou Guo, Zeya Li, Bin Xiao, Rongchong Huang","doi":"10.1186/s13062-024-00485-y","DOIUrl":"10.1186/s13062-024-00485-y","url":null,"abstract":"<p><strong>Background: </strong>Myocardial infarction (MI) is a major cause of mortality and morbidity worldwide. The intercellular communication in post-infarction angiogenesis remains unclear.</p><p><strong>Methods: </strong>In this study, we explored the role and mechanism of action of M2 macrophage-derived exosomes (M2-exos) in angiogenesis after MI. M2-exos were harvested and injected intramyocardially at the onset of MI. Two distinct endothelial cells (ECs) were cultured with M2-exos to explore the direct effects on angiogenesis.</p><p><strong>Results: </strong>We showed that M2-exos improved cardiac function, reduced infarct size, and enhanced angiogenesis after MI. Moreover, M2-exos promoted angiogenesis in vitro; the molecules loaded in the vesicles were responsible for its proangiogenic effects. We further validated that higher abundance of miR-132-3p in M2-exos, which recapitulate their functions, was required for the cardioprotective effects exerted by M2-exos. Mechanistically, miR-132-3p carried by M2-exos down-regulate the expression of THBS1 through direct binding to its 3´UTR and the proangiogenic effects of miR-132-3p were largely reversed by THBS1 overexpression.</p><p><strong>Conclusion: </strong>Our findings demonstrate that M2-exos promote angiogenesis after MI by transporting miR-132-3p to ECs, and by binding to THBS1 mRNA directly and negatively regulating its expression. These findings highlight the role of M2-exos in cardiac repair and provide novel mechanistic understanding of intercellular communication in post-infarction angiogenesis.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"43"},"PeriodicalIF":5.7,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11155164/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141260758","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
YY1 mediated DCUN1D5 transcriptional activation promotes triple-negative breast cancer progression by targeting FN1/PI3K/AKT pathway. YY1 介导的 DCUN1D5 转录激活通过靶向 FN1/PI3K/AKT 通路促进三阴性乳腺癌的进展。
IF 5.5 2区 生物学 Q1 BIOLOGY Pub Date : 2024-06-03 DOI: 10.1186/s13062-024-00481-2
Yuxiang Lin, Yan Li, Xiaobin Chen, Xuan Jin, Meichen Jiang, Han Xiao, Lili Chen, Minyan Chen, Wenzhe Zhang, Hanxi Chen, Qian Nie, Rongrong Guo, Wenhui Guo, Fangmeng Fu, Chuan Wang

Triple-negative breast cancer (TNBC) is more aggressive and has a higher metastasis rate compared with other subtypes of breast cancer. Due to the lack of drug-targetable receptors, chemotherapy is now the only available systemic treatment for TNBC. However, some patients might still develop drug resistance and have poor prognosis. Therefore, novel molecular biomarkers and new treatment targets are urgently needed for patients with TNBC. To provide molecular insights into TNBC progression, we investigated the function and the underlying mechanism of Defective in cullin neddylation 1 domain containing 5 (DCUN1D5) in the regulation of TNBC. By TCGA dataset and surgical specimens with immunohistochemical (IHC) staining method, DCUN1D5 was identified to be significantly upregulated in TNBC tumor tissues and negatively associated with prognosis. A series of in vitro and in vivo experiments were performed to confirm the oncogenic role of DCUN1D5 in TNBC. Overexpression of FN1 or PI3K/AKT activator IGF-1 could restore the proliferative and invasive ability induced by DCUN1D5 knockdown and DCUN1D5 could act as a novel transcriptional target of transcription factor Yin Yang 1 (YY1). In conclusion, YY1-enhanced DCUN1D5 expression could promote TNBC progression by FN1/PI3K/AKT pathway and DCUN1D5 might be a potential prognostic biomarker and therapeutic target for TNBC treatment.

与其他亚型乳腺癌相比,三阴性乳腺癌(TNBC)更具侵袭性,转移率更高。由于缺乏药物靶向受体,化疗是目前治疗 TNBC 的唯一方法。然而,部分患者仍可能产生耐药性,预后较差。因此,TNBC 患者迫切需要新的分子生物标志物和新的治疗靶点。为了提供TNBC进展的分子洞察,我们研究了Defective in cullin neddylation 1 domain containing 5(DCUN1D5)在TNBC调控中的功能及其内在机制。通过TCGA数据集和手术标本的免疫组化(IHC)染色方法,我们发现DCUN1D5在TNBC肿瘤组织中显著上调,并与预后呈负相关。为了证实DCUN1D5在TNBC中的致癌作用,研究人员进行了一系列体外和体内实验。过表达FN1或PI3K/AKT激活剂IGF-1可恢复DCUN1D5敲除诱导的增殖和侵袭能力,DCUN1D5可作为转录因子阴阳1(YY1)的一个新的转录靶点。总之,YY1增强的DCUN1D5表达可通过FN1/PI3K/AKT通路促进TNBC的进展,DCUN1D5可能是TNBC治疗的潜在预后生物标志物和治疗靶点。
{"title":"YY1 mediated DCUN1D5 transcriptional activation promotes triple-negative breast cancer progression by targeting FN1/PI3K/AKT pathway.","authors":"Yuxiang Lin, Yan Li, Xiaobin Chen, Xuan Jin, Meichen Jiang, Han Xiao, Lili Chen, Minyan Chen, Wenzhe Zhang, Hanxi Chen, Qian Nie, Rongrong Guo, Wenhui Guo, Fangmeng Fu, Chuan Wang","doi":"10.1186/s13062-024-00481-2","DOIUrl":"10.1186/s13062-024-00481-2","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is more aggressive and has a higher metastasis rate compared with other subtypes of breast cancer. Due to the lack of drug-targetable receptors, chemotherapy is now the only available systemic treatment for TNBC. However, some patients might still develop drug resistance and have poor prognosis. Therefore, novel molecular biomarkers and new treatment targets are urgently needed for patients with TNBC. To provide molecular insights into TNBC progression, we investigated the function and the underlying mechanism of Defective in cullin neddylation 1 domain containing 5 (DCUN1D5) in the regulation of TNBC. By TCGA dataset and surgical specimens with immunohistochemical (IHC) staining method, DCUN1D5 was identified to be significantly upregulated in TNBC tumor tissues and negatively associated with prognosis. A series of in vitro and in vivo experiments were performed to confirm the oncogenic role of DCUN1D5 in TNBC. Overexpression of FN1 or PI3K/AKT activator IGF-1 could restore the proliferative and invasive ability induced by DCUN1D5 knockdown and DCUN1D5 could act as a novel transcriptional target of transcription factor Yin Yang 1 (YY1). In conclusion, YY1-enhanced DCUN1D5 expression could promote TNBC progression by FN1/PI3K/AKT pathway and DCUN1D5 might be a potential prognostic biomarker and therapeutic target for TNBC treatment.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"42"},"PeriodicalIF":5.5,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11145835/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141236324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lysine-specific methyltransferase Set7/9 in stemness, differentiation, and development. 赖氨酸特异性甲基转移酶Set7/9在干性、分化和发育中的作用
IF 5.5 2区 生物学 Q1 BIOLOGY Pub Date : 2024-05-29 DOI: 10.1186/s13062-024-00484-z
Alexandra Daks, Sergey Parfenyev, Oleg Shuvalov, Olga Fedorova, Alexander Nazarov, Gerry Melino, Nickolai A Barlev

The enzymes performing protein post-translational modifications (PTMs) form a critical post-translational regulatory circuitry that orchestrates literally all cellular processes in the organism. In particular, the balance between cellular stemness and differentiation is crucial for the development of multicellular organisms. Importantly, the fine-tuning of this balance on the genetic level is largely mediated by specific PTMs of histones including lysine methylation. Lysine methylation is carried out by special enzymes (lysine methyltransferases) that transfer the methyl group from S-adenosyl-L-methionine to the lysine residues of protein substrates. Set7/9 is one of the exemplary protein methyltransferases that however, has not been fully studied yet. It was originally discovered as histone H3 lysine 4-specific methyltransferase, which later was shown to methylate a number of non-histone proteins that are crucial regulators of stemness and differentiation, including p53, pRb, YAP, DNMT1, SOX2, FOXO3, and others. In this review we summarize the information available to date on the role of Set7/9 in cellular differentiation and tissue development during embryogenesis and in adult organisms. Finally, we highlight and discuss the role of Set7/9 in pathological processes associated with aberrant cellular differentiation and self-renewal, including the formation of cancer stem cells.

进行蛋白质翻译后修饰(PTM)的酶形成了一个关键的翻译后调控回路,协调着生物体内的所有细胞过程。特别是,细胞干性和分化之间的平衡对多细胞生物体的发育至关重要。重要的是,这种平衡在基因水平上的微调主要是由组蛋白的特定 PTMs(包括赖氨酸甲基化)介导的。赖氨酸甲基化是由特殊的酶(赖氨酸甲基转移酶)完成的,它们将甲基从 S-腺苷-L-蛋氨酸转移到蛋白质底物的赖氨酸残基上。Set7/9 是典型的蛋白质甲基转移酶之一,但尚未得到充分研究。它最初是作为组蛋白 H3 赖氨酸 4 特异性甲基转移酶被发现的,后来被证明能甲基化一些非组蛋白,这些蛋白是干性和分化的关键调节因子,包括 p53、pRb、YAP、DNMT1、SOX2、FOXO3 等。在这篇综述中,我们总结了迄今为止有关Set7/9在胚胎发生和成体生物体内细胞分化和组织发育过程中的作用的信息。最后,我们将重点讨论 Set7/9 在与细胞异常分化和自我更新相关的病理过程(包括癌症干细胞的形成)中的作用。
{"title":"Lysine-specific methyltransferase Set7/9 in stemness, differentiation, and development.","authors":"Alexandra Daks, Sergey Parfenyev, Oleg Shuvalov, Olga Fedorova, Alexander Nazarov, Gerry Melino, Nickolai A Barlev","doi":"10.1186/s13062-024-00484-z","DOIUrl":"10.1186/s13062-024-00484-z","url":null,"abstract":"<p><p>The enzymes performing protein post-translational modifications (PTMs) form a critical post-translational regulatory circuitry that orchestrates literally all cellular processes in the organism. In particular, the balance between cellular stemness and differentiation is crucial for the development of multicellular organisms. Importantly, the fine-tuning of this balance on the genetic level is largely mediated by specific PTMs of histones including lysine methylation. Lysine methylation is carried out by special enzymes (lysine methyltransferases) that transfer the methyl group from S-adenosyl-L-methionine to the lysine residues of protein substrates. Set7/9 is one of the exemplary protein methyltransferases that however, has not been fully studied yet. It was originally discovered as histone H3 lysine 4-specific methyltransferase, which later was shown to methylate a number of non-histone proteins that are crucial regulators of stemness and differentiation, including p53, pRb, YAP, DNMT1, SOX2, FOXO3, and others. In this review we summarize the information available to date on the role of Set7/9 in cellular differentiation and tissue development during embryogenesis and in adult organisms. Finally, we highlight and discuss the role of Set7/9 in pathological processes associated with aberrant cellular differentiation and self-renewal, including the formation of cancer stem cells.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"41"},"PeriodicalIF":5.5,"publicationDate":"2024-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11137904/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141173666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Plant hormones and phenolic acids response to UV-B stress in Rhododendron chrysanthum pall. 杜鹃花对紫外线-B 胁迫的植物激素和酚酸反应
IF 5.5 2区 生物学 Q1 BIOLOGY Pub Date : 2024-05-28 DOI: 10.1186/s13062-024-00483-0
Qi Sun, Xiangqun Li, Li Sun, Mingyi Sun, Hongwei Xu, Xiaofu Zhou

Our study aims to identify the mechanisms involved in regulating the response of Rhodoendron Chrysanthum Pall. (R. chrysanthum) leaves to UV-B exposure; phosphorylated proteomics and metabolomics for phenolic acids and plant hormones were integrated in this study. The results showed that UV-B stress resulted in the accumulation of salicylic acid and the decrease of auxin, jasmonic acid, abscisic acid, cytokinin and gibberellin in R. chrysanthum. The phosphorylated proteins that changed in plant hormone signal transduction pathway and phenolic acid biosynthesis pathway were screened by comprehensive metabonomics and phosphorylated proteomics. In order to construct the regulatory network of R. chrysanthum leaves under UV-B stress, the relationship between plant hormones and phenolic acid compounds was analyzed. It provides a rationale for elucidating the molecular mechanisms of radiation tolerance in plants.

我们的研究旨在确定调控杜鹃花叶片(Rhodoendron Chrysanthum Pall.(本研究整合了磷酸化蛋白质组学、酚酸代谢组学和植物激素代谢组学。结果表明,紫外线-B 胁迫导致 R. Chrysanthum 水杨酸积累,而辅助素、茉莉酸、脱落酸、细胞分裂素和赤霉素减少。通过综合代谢组学和磷酸化蛋白质组学筛选了植物激素信号转导途径和酚酸生物合成途径中发生变化的磷酸化蛋白。通过分析植物激素与酚酸化合物之间的关系,构建了 UV-B 胁迫下菊芋叶片的调控网络。这为阐明植物耐辐射的分子机制提供了理论依据。
{"title":"Plant hormones and phenolic acids response to UV-B stress in Rhododendron chrysanthum pall.","authors":"Qi Sun, Xiangqun Li, Li Sun, Mingyi Sun, Hongwei Xu, Xiaofu Zhou","doi":"10.1186/s13062-024-00483-0","DOIUrl":"10.1186/s13062-024-00483-0","url":null,"abstract":"<p><p>Our study aims to identify the mechanisms involved in regulating the response of Rhodoendron Chrysanthum Pall. (R. chrysanthum) leaves to UV-B exposure; phosphorylated proteomics and metabolomics for phenolic acids and plant hormones were integrated in this study. The results showed that UV-B stress resulted in the accumulation of salicylic acid and the decrease of auxin, jasmonic acid, abscisic acid, cytokinin and gibberellin in R. chrysanthum. The phosphorylated proteins that changed in plant hormone signal transduction pathway and phenolic acid biosynthesis pathway were screened by comprehensive metabonomics and phosphorylated proteomics. In order to construct the regulatory network of R. chrysanthum leaves under UV-B stress, the relationship between plant hormones and phenolic acid compounds was analyzed. It provides a rationale for elucidating the molecular mechanisms of radiation tolerance in plants.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"19 1","pages":"40"},"PeriodicalIF":5.5,"publicationDate":"2024-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11134694/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141161284","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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
Biology Direct
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