首页 > 最新文献

Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis最新文献

英文 中文
miR-129–2-3p binds SEMA4C to regulate HCC development and inhibit the EMT miR-129-2-3p 与 SEMA4C 结合,调控 HCC 的发展并抑制 EMT
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111872
Siyuan Ma , Chun Pu

Background

Among primary liver cancers, HCC is the most prevalent. Small noncoding RNAs called miRNAs control the expression of downstream target genes to take part in a variety of physiological and pathological processes, including those related to cancer.

Methods

miR-129–2–3p and SEMA4C expression levels were assessed using RT-qPCR. The CCK-8, invasion, and wound healing assays were used to confirm the capacity of HCC cells for proliferation, invasion and migration respectively. Serum SEMA4C levels were detected via ELISA. The RIP and dual-luciferase reporter assays were used to confirm the existence of intergenic binding sites. Cell apoptosis assay and cell cycle assay were performed to detect the apoptosis rate and cycle distribution of cells, and WB was performed to detect the protein expression of SEMA4C, RhoA, ROCK1, E-cadherin, N-cadherin, and vimentin. Furthermore, cancer-inhibiting role of miR-129–2–3p were further confirmed by animal tests.

Results

miR-129–2–3p expression was reduced in HCC tissues and cells. Overexpression of miR-129–2–3p decreased the proliferation, invasion, migration, and EMT in HCC cells, whereas inhibition of miR-129–2–3p had the opposite effects. Our research also showed that SEMA4C was increased in HCC tissues, serum and cells, and that SEMA4C knockdown prevented HCC cell invasion, migration, proliferation, and EMT. Overexpression of SEMA4C reversed the inhibitory effect of miR-129–2–3p on HCC.

Conclusions

Overall, we discovered that through binding to SEMA4C, miR-129–2–3p regulates HCC cell proliferation, invasion, migration, and EMT.

背景在原发性肝癌中,HCC 的发病率最高。方法使用 RT-qPCR 评估 miR-129-2-3p 和 SEMA4C 的表达水平。CCK-8、侵袭和伤口愈合试验分别用于确认 HCC 细胞的增殖、侵袭和迁移能力。通过 ELISA 检测血清 SEMA4C 水平。RIP和双荧光素酶报告实验用于确认基因间结合位点的存在。细胞凋亡检测和细胞周期检测用于检测细胞的凋亡率和周期分布,WB检测SEMA4C、RhoA、ROCK1、E-cadherin、N-cadherin和vimentin的蛋白表达。此外,动物实验进一步证实了 miR-129-2-3p 的抑癌作用。过表达 miR-129-2-3p 会降低 HCC 细胞的增殖、侵袭、迁移和 EMT,而抑制 miR-129-2-3p 则会产生相反的效果。我们的研究还表明,SEMA4C 在 HCC 组织、血清和细胞中均有增高,而 SEMA4C 的敲除可阻止 HCC 细胞的侵袭、迁移、增殖和 EMT。结论总之,我们发现 miR-129-2-3p 通过与 SEMA4C 结合,调控 HCC 细胞的增殖、侵袭、迁移和 EMT。
{"title":"miR-129–2-3p binds SEMA4C to regulate HCC development and inhibit the EMT","authors":"Siyuan Ma ,&nbsp;Chun Pu","doi":"10.1016/j.mrfmmm.2024.111872","DOIUrl":"10.1016/j.mrfmmm.2024.111872","url":null,"abstract":"<div><h3>Background</h3><p>Among primary liver cancers, HCC is the most prevalent. Small noncoding RNAs called miRNAs control the expression of downstream target genes to take part in a variety of physiological and pathological processes, including those related to cancer.</p></div><div><h3>Methods</h3><p>miR-129–2–3p and SEMA4C expression levels were assessed using RT-qPCR. The CCK-8, invasion, and wound healing assays were used to confirm the capacity of HCC cells for proliferation, invasion and migration respectively. Serum SEMA4C levels were detected via ELISA. The RIP and dual-luciferase reporter assays were used to confirm the existence of intergenic binding sites. Cell apoptosis assay and cell cycle assay were performed to detect the apoptosis rate and cycle distribution of cells, and WB was performed to detect the protein expression of SEMA4C, RhoA, ROCK1, E-cadherin, N-cadherin, and vimentin. Furthermore, cancer-inhibiting role of miR-129–2–3p were further confirmed by animal tests.</p></div><div><h3>Results</h3><p>miR-129–2–3p expression was reduced in HCC tissues and cells. Overexpression of miR-129–2–3p decreased the proliferation, invasion, migration, and EMT in HCC cells, whereas inhibition of miR-129–2–3p had the opposite effects. Our research also showed that SEMA4C was increased in HCC tissues, serum and cells, and that SEMA4C knockdown prevented HCC cell invasion, migration, proliferation, and EMT. Overexpression of SEMA4C reversed the inhibitory effect of miR-129–2–3p on HCC.</p></div><div><h3>Conclusions</h3><p>Overall, we discovered that through binding to SEMA4C, miR-129–2–3p regulates HCC cell proliferation, invasion, migration, and EMT.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111872"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141629764","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MicroRNA-138 promotes the progression of multiple myeloma through targeting paired PAX5 MicroRNA-138 通过靶向配对的 PAX5 促进多发性骨髓瘤的进展。
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111869
Xiao Yan , Keting Wang , Cong Shi , Kaihong Xu , Binbin Lai , Shujun Yang , Lixia Sheng , Ping Zhang , Ying Chen , Qitian Mu , Guifang Ouyang

Background

Multiple myeloma cancer stem cells (MMSC) have been considered as the leading cause of multiple myeloma (MM) drug resistance and eventual relapse, microRNAs (miRNAs) collectively participate in the progression of MM. However, the pathogenesis of miR-138 in MMSC is still not fully understood.

Objective

The intention of this study was to investigate the mechanism and role of miR-138 in multiple myeloma.

Method

Bone marrow samples and peripheral blood from patients and normal controls were collected. Use Magnet-based Cancer Stem Cell Isolation Kit to separate and extract MMSC. Real-time quantitative PCR (RT-qPCR) was carried out to determine mRNA level. Western blot was applied to detect protein levels. MTT and flow cytometry were conducted to examine the proliferation and apoptosis of MMSC. Finally, dual-luciferase reporter gene assays were performed to confirm that paired box 5 (PAX5) is a direct target for miR-138.

Results

Compared with normal group, the expression of miR-138 in patients was significantly up-regulated, and the expression of miR-138 was in a negative correlation with PAX5. Additionally, downregulated miR-138 facilitated the apoptosis and inhibited the proliferation of MMSC in vitro and in vivo. Downregulated miR-138 moderated the expression of PAX5, Bcl-2, Bax, and Caspase-3. PAX5 was a direct target of miR-138.

Conclusion

Taken together, miR-138 plays a carcinogenic role in MM, and miR-138 adjusted the proliferation and apoptosis of MMSC by targeting PAX5. miR-138 has the probability of becoming a new medicinal target for the treatment of MM.

背景:多发性骨髓瘤癌干细胞(MMSC)被认为是多发性骨髓瘤(MM)耐药和最终复发的主要原因,微RNA(miRNA)共同参与了MM的进展。然而,miR-138在MMSC中的发病机制仍未完全明了:本研究旨在探讨 miR-138 在多发性骨髓瘤中的作用机制:方法:收集患者和正常对照组的骨髓和外周血样本。使用磁性癌症干细胞分离试剂盒分离并提取 MMSC。采用实时定量 PCR(RT-qPCR)检测 mRNA 水平。采用 Western 印迹检测蛋白质水平。采用 MTT 和流式细胞术检测 MMSC 的增殖和凋亡。最后,通过双荧光素酶报告基因实验证实配对框 5(PAX5)是 miR-138 的直接靶标:结果:与正常组相比,患者体内 miR-138 的表达明显上调,且 miR-138 的表达与 PAX5 呈负相关。此外,下调的 miR-138 可促进 MMSC 的体外和体内凋亡并抑制其增殖。下调的 miR-138 可调节 PAX5、Bcl-2、Bax 和 Caspase-3 的表达。PAX5是miR-138的直接靶标:综上所述,miR-138在MM中起致癌作用,miR-138通过靶向PAX5调节MMSC的增殖和凋亡。
{"title":"MicroRNA-138 promotes the progression of multiple myeloma through targeting paired PAX5","authors":"Xiao Yan ,&nbsp;Keting Wang ,&nbsp;Cong Shi ,&nbsp;Kaihong Xu ,&nbsp;Binbin Lai ,&nbsp;Shujun Yang ,&nbsp;Lixia Sheng ,&nbsp;Ping Zhang ,&nbsp;Ying Chen ,&nbsp;Qitian Mu ,&nbsp;Guifang Ouyang","doi":"10.1016/j.mrfmmm.2024.111869","DOIUrl":"10.1016/j.mrfmmm.2024.111869","url":null,"abstract":"<div><h3>Background</h3><p>Multiple myeloma cancer stem cells (MMSC) have been considered as the leading cause of multiple myeloma (MM) drug resistance and eventual relapse, microRNAs (miRNAs) collectively participate in the progression of MM. However, the pathogenesis of miR-138 in MMSC is still not fully understood.</p></div><div><h3>Objective</h3><p>The intention of this study was to investigate the mechanism and role of miR-138 in multiple myeloma.</p></div><div><h3>Method</h3><p>Bone marrow samples and peripheral blood from patients and normal controls were collected. Use Magnet-based Cancer Stem Cell Isolation Kit to separate and extract MMSC. Real-time quantitative PCR (RT-qPCR) was carried out to determine mRNA level. Western blot was applied to detect protein levels. MTT and flow cytometry were conducted to examine the proliferation and apoptosis of MMSC. Finally, dual-luciferase reporter gene assays were performed to confirm that paired box 5 (PAX5) is a direct target for miR-138.</p></div><div><h3>Results</h3><p>Compared with normal group, the expression of miR-138 in patients was significantly up-regulated, and the expression of miR-138 was in a negative correlation with PAX5. Additionally, downregulated miR-138 facilitated the apoptosis and inhibited the proliferation of MMSC in vitro and in vivo. Downregulated miR-138 moderated the expression of PAX5, Bcl-2, Bax, and Caspase-3. PAX5 was a direct target of miR-138.</p></div><div><h3>Conclusion</h3><p>Taken together, miR-138 plays a carcinogenic role in MM, and miR-138 adjusted the proliferation and apoptosis of MMSC by targeting PAX5. miR-138 has the probability of becoming a new medicinal target for the treatment of MM.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111869"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141500042","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Saikosaponin-d mediates FOXG1 to reverse docetaxel resistance in prostate cancer through oxidative phosphorylation 柴胡皂苷-d 通过氧化磷酸化介导 FOXG1 逆转前列腺癌的多西他赛耐药性
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111875
Jun Meng, Bo Yang, Chang Shu, Shuai Jiang

Background

Prostate cancer (PCa), a prevalent malignancy worldwide, is frequently identified in advanced stages due to the absence of distinctive early symptoms, thereby culminating in the development of chemotherapy-induced drug resistance. Exploring novel resistance mechanisms and identifying new therapeutic agents can facilitate the advancement of more efficacious strategies for PCa treatment.

Methods

Bioinformatics analysis was employed to investigate the expression of FOXG1 in PCa tissues. Subsequently, qRT-PCR was utilized to validate FOXG1 mRNA expression levels in corresponding PCa cell lines. FOXG1 knockdown was performed, and cell proliferation was assessed using CCK-8 assays, while cell migration and invasion capabilities were evaluated through wound healing and Transwell assays. Western blot and Seahorse analyzer were used to measure oxidative phosphorylation (OXPHOS) levels. Additionally, to explore potential approaches to alleviate PCa drug resistance, this study assessed the impact of biologically active saikosaponin-d (SSd) on PCa malignant progression and resistance by regulating FOXG1 expression.

Results

FOXG1 exhibited high expression in PCa tissues and cell lines. Knockdown of FOXG1 inhibited the proliferation, migration, and invasion of PCa cells, while FOXG1 overexpression had the opposite effect and promoted OXPHOS levels. The addition of an OXPHOS inhibitor prevented this outcome. Finally, SSd was shown to suppress FOXG1 expression and reverse docetaxel resistance in PCa cells through the OXPHOS pathway.

Conclusion

This work demonstrated that SSd mediated FOXG1 to reverse malignant progression and docetaxel resistance in PCa through OXPHOS.

背景:前列腺癌(PCa)是一种全球流行的恶性肿瘤,由于没有明显的早期症状,往往在晚期才被发现,从而导致化疗引起的耐药性的产生。探索新的耐药机制和确定新的治疗药物有助于推进更有效的 PCa 治疗策略:方法:采用生物信息学分析研究 PCa 组织中 FOXG1 的表达。随后,利用 qRT-PCR 验证了 FOXG1 mRNA 在相应 PCa 细胞系中的表达水平。进行 FOXG1 基因敲除,用 CCK-8 检测法评估细胞增殖,用伤口愈合和 Transwell 检测法评估细胞迁移和侵袭能力。利用 Western 印迹和海马分析仪测量氧化磷酸化(OXPHOS)水平。此外,为了探索缓解 PCa 耐药性的潜在方法,本研究还评估了生物活性赛可皂甙-d(SSd)通过调节 FOXG1 表达对 PCa 恶性进展和耐药性的影响:结果:FOXG1在PCa组织和细胞系中高表达。结果:FOXG1 在 PCa 组织和细胞系中高表达,敲除 FOXG1 可抑制 PCa 细胞的增殖、迁移和侵袭,而过表达 FOXG1 则会产生相反的效果,促进 OXPHOS 水平。添加 OXPHOS 抑制剂可防止这种结果。最后,研究表明 SSd 可抑制 FOXG1 的表达,并通过 OXPHOS 途径逆转 PCa 细胞对多西他赛的耐药性:这项研究表明,SSd通过OXPHOS介导FOXG1,逆转了PCa的恶性进展和多西他赛耐药性。
{"title":"Saikosaponin-d mediates FOXG1 to reverse docetaxel resistance in prostate cancer through oxidative phosphorylation","authors":"Jun Meng,&nbsp;Bo Yang,&nbsp;Chang Shu,&nbsp;Shuai Jiang","doi":"10.1016/j.mrfmmm.2024.111875","DOIUrl":"10.1016/j.mrfmmm.2024.111875","url":null,"abstract":"<div><h3>Background</h3><p>Prostate cancer (PCa), a prevalent malignancy worldwide, is frequently identified in advanced stages due to the absence of distinctive early symptoms, thereby culminating in the development of chemotherapy-induced drug resistance. Exploring novel resistance mechanisms and identifying new therapeutic agents can facilitate the advancement of more efficacious strategies for PCa treatment.</p></div><div><h3>Methods</h3><p>Bioinformatics analysis was employed to investigate the expression of FOXG1 in PCa tissues. Subsequently, qRT-PCR was utilized to validate FOXG1 mRNA expression levels in corresponding PCa cell lines. FOXG1 knockdown was performed, and cell proliferation was assessed using CCK-8 assays, while cell migration and invasion capabilities were evaluated through wound healing and Transwell assays. Western blot and Seahorse analyzer were used to measure oxidative phosphorylation (OXPHOS) levels. Additionally, to explore potential approaches to alleviate PCa drug resistance, this study assessed the impact of biologically active saikosaponin-d (SSd) on PCa malignant progression and resistance by regulating FOXG1 expression.</p></div><div><h3>Results</h3><p>FOXG1 exhibited high expression in PCa tissues and cell lines. Knockdown of FOXG1 inhibited the proliferation, migration, and invasion of PCa cells, while FOXG1 overexpression had the opposite effect and promoted OXPHOS levels. The addition of an OXPHOS inhibitor prevented this outcome. Finally, SSd was shown to suppress FOXG1 expression and reverse docetaxel resistance in PCa cells through the OXPHOS pathway.</p></div><div><h3>Conclusion</h3><p>This work demonstrated that SSd mediated FOXG1 to reverse malignant progression and docetaxel resistance in PCa through OXPHOS.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111875"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141891408","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NTSR1 promotes epithelial-mesenchymal transition and metastasis in lung adenocarcinoma through the Wnt/β-catenin pathway NTSR1 通过 Wnt/β-catenin 通路促进肺腺癌的上皮-间质转化和转移
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111877
Zhihao Zhang , Dongliang Zhang , Kai Su , Dongqiang Wu , Qiqi Hu , Tianying Jin , Tingting Ye , Rongrong Zhang

Background

Lung adenocarcinoma (LUAD) patients are implicated in poor prognoses and increased mortality rates. Metastasis, as a leading cause of LUAD-related deaths, requires further investigation. Highly metastatic cancer cells often exhibit extensive characteristics of epithelial-mesenchymal transition (EMT). This study attempted to identify novel targets associated with LUAD metastasis and validate their specific molecular mechanisms.

Methods

Bioinformatics was conducted to determine NTSR1 expression in LUAD and the enriched pathways. Immunohistochemical analysis was used to assess NTSR1 expression in LUAD tissue. qRT-PCR examined expressions of NTSR1 and Wnt/β-Catenin pathway-related genes in LUAD cells. Transwell assayed cell migration and invasion. Cell adhesion experiments were conducted to evaluate cell adhesion capacity. Western blot analysis was employed to examine expression of EMT, Wnt/β-Catenin pathway, and cell adhesion markers.

Results

NTSR1 was upregulated in LUAD tissues and cells, and enriched in EMT pathway. Knockdown of NTSR1 reduced migration, invasion, and adhesion abilities in LUAD cells, and inhibited EMT progression and Wnt/β-Catenin pathway. Rescue experiments demonstrated that β-Catenin activator SKL2001 reversed repressive influence of NTSR1 knockdown on LUAD cell malignant phenotypes and EMT progression.

Conclusion

The data obtained in this study suggested that NTSR1 stimulated EMT and metastasis in LUAD via Wnt/β-Catenin pathway. This finding may provide options for overcoming LUAD metastasis.

背景肺腺癌(LUAD)患者预后不良,死亡率升高。转移是导致肺腺癌相关死亡的主要原因,需要进一步研究。高度转移的癌细胞通常表现出上皮-间质转化(EMT)的广泛特征。本研究试图确定与LUAD转移相关的新靶点,并验证其特定的分子机制。方法通过生物信息学方法确定NTSR1在LUAD中的表达以及富集的通路。qRT-PCR检测了LUAD细胞中NTSR1和Wnt/β-Catenin通路相关基因的表达。Transwell检测细胞迁移和侵袭。细胞粘附实验用于评估细胞粘附能力。结果 NTSR1在LUAD组织和细胞中上调,并在EMT通路中富集。敲除 NTSR1 可降低 LUAD 细胞的迁移、侵袭和粘附能力,抑制 EMT 进展和 Wnt/β-Catenin 通路。结论 本研究获得的数据表明,NTSR1 通过 Wnt/β-Catenin 通路刺激 LUAD 细胞的 EMT 和转移。这一发现为克服 LUAD 转移提供了可能。
{"title":"NTSR1 promotes epithelial-mesenchymal transition and metastasis in lung adenocarcinoma through the Wnt/β-catenin pathway","authors":"Zhihao Zhang ,&nbsp;Dongliang Zhang ,&nbsp;Kai Su ,&nbsp;Dongqiang Wu ,&nbsp;Qiqi Hu ,&nbsp;Tianying Jin ,&nbsp;Tingting Ye ,&nbsp;Rongrong Zhang","doi":"10.1016/j.mrfmmm.2024.111877","DOIUrl":"10.1016/j.mrfmmm.2024.111877","url":null,"abstract":"<div><h3>Background</h3><p>Lung adenocarcinoma (LUAD) patients are implicated in poor prognoses and increased mortality rates. Metastasis, as a leading cause of LUAD-related deaths, requires further investigation. Highly metastatic cancer cells often exhibit extensive characteristics of epithelial-mesenchymal transition (EMT). This study attempted to identify novel targets associated with LUAD metastasis and validate their specific molecular mechanisms.</p></div><div><h3>Methods</h3><p>Bioinformatics was conducted to determine NTSR1 expression in LUAD and the enriched pathways. Immunohistochemical analysis was used to assess NTSR1 expression in LUAD tissue. qRT-PCR examined expressions of NTSR1 and Wnt/β-Catenin pathway-related genes in LUAD cells. Transwell assayed cell migration and invasion. Cell adhesion experiments were conducted to evaluate cell adhesion capacity. Western blot analysis was employed to examine expression of EMT, Wnt/β-Catenin pathway, and cell adhesion markers.</p></div><div><h3>Results</h3><p>NTSR1 was upregulated in LUAD tissues and cells, and enriched in EMT pathway. Knockdown of NTSR1 reduced migration, invasion, and adhesion abilities in LUAD cells, and inhibited EMT progression and Wnt/β-Catenin pathway. Rescue experiments demonstrated that β-Catenin activator SKL2001 reversed repressive influence of NTSR1 knockdown on LUAD cell malignant phenotypes and EMT progression.</p></div><div><h3>Conclusion</h3><p>The data obtained in this study suggested that NTSR1 stimulated EMT and metastasis in LUAD via Wnt/β-Catenin pathway. This finding may provide options for overcoming LUAD metastasis.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111877"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142049673","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CircRNA ATF6 suppresses bladder cancer cell proliferation and migration via miR-146a-5p/FLNA axis CircRNA ATF6通过miR-146a-5p/FLNA轴抑制膀胱癌细胞的增殖和迁移
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111876
Bing Lu , Yongqiang Zhou , Zheng Ma , Zhenfan Wang

Background

Bladder cancer (BCa) is the most common malignancy with increasing morbidity and mortality. Circular RNA (circRNA) ATF6 level was downregulated in BCa after GSE92675 CircRNA microarray dataset was analyzed using GEO2R. However, its function and mechanism in BCa remain largely unknown.

Methods

GEO2R and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used to measure levels of circRNA ATF6, microRNA-146a-5p (miR-146a-5p), and filamin A (FLNA). CircRNA ATF6 stability was assessed by actinomycin D and RNase R assays, while circRNA ATF6 cellular localization was examined by FISH experiments in T24 cells. Cell counting kit-8 (CCK-8), colony formation, wound-healing, and transwell assays were used to study circRNA ATF6’s function in growth, motility, and invasion. By examining luciferase, starBase, RNA pull-down, and RNA immunoprecipitation (RIP) experiments, we anticipated and confirmed miR-146a-5p interactions with circRNA ATF6, as well as miR-146a-5p interactions with FLNA. On tumor-bearing mice, in vivo experiments were conducted.

Results

MiR-146a-5p expression in Bca was elevated, while circRNA ATF6 and FLNA were downregulated. CircRNA ATF6 showed better stability in BCa cells, with its expression primarily in the cytoplasm. Upregulating circRNA ATF6 lowered BCa cell viability, colony numbers, and invasion numbers, but broadened their migratory pattern. MiR-146a-5p was directly sponged up by circRNA ATF6, which also detrimentally affected miR-146a-5p levels in BCa. MiR-146a-5p reduced BCa FLNA expression by targeting FLNA. FLNA silencing abolished circRNA ATF6’s mitigating function in BCa cell proliferation, motility, and invasion. In vivo, overexpression of circRNA ATF6 significantly reduced tumor volume and weight.

Conclusion

CircRNA ATF6 suppresses BCa cell growth, migration and invasion through the miR-146a-5p/FLNA axis.

背景膀胱癌(BCa)是最常见的恶性肿瘤,发病率和死亡率不断上升。利用 GEO2R 对 GSE92675 CircRNA 微阵列数据集进行分析后发现,环状 RNA(circRNA)ATF6 在 BCa 中水平下调。方法使用 GEO2R 和反转录定量聚合酶链反应(RT-qPCR)测量 circRNA ATF6、microRNA-146a-5p(miR-146a-5p)和丝胶蛋白 A(FLNA)的水平。循环RNA ATF6的稳定性通过放线菌素D和RNase R检测进行评估,而循环RNA ATF6的细胞定位则通过T24细胞的FISH实验进行检测。细胞计数试剂盒-8(CCK-8)、菌落形成、伤口愈合和透孔试验被用来研究 circRNA ATF6 在生长、运动和侵袭中的功能。通过荧光素酶、starBase、RNA pull-down和RNA免疫沉淀(RIP)实验,我们预测并证实了miR-146a-5p与circRNA ATF6的相互作用,以及miR-146a-5p与FLNA的相互作用。结果miR-146a-5p在Bca中的表达升高,而circRNA ATF6和FLNA则下调。circRNA ATF6 在 BCa 细胞中表现出较好的稳定性,主要在细胞质中表达。上调 circRNA ATF6 会降低 BCa 细胞的活力、集落数和侵袭数,但会扩大其迁移模式。circRNA ATF6直接上调了miR-146a-5p,这也对BCa细胞中的miR-146a-5p水平产生了不利影响。MiR-146a-5p 通过靶向 FLNA 减少了 BCa FLNA 的表达。FLNA沉默会削弱circRNA ATF6在BCa细胞增殖、运动和侵袭中的缓解功能。结论circRNA ATF6通过miR-146a-5p/FLNA轴抑制BCa细胞的生长、迁移和侵袭。
{"title":"CircRNA ATF6 suppresses bladder cancer cell proliferation and migration via miR-146a-5p/FLNA axis","authors":"Bing Lu ,&nbsp;Yongqiang Zhou ,&nbsp;Zheng Ma ,&nbsp;Zhenfan Wang","doi":"10.1016/j.mrfmmm.2024.111876","DOIUrl":"10.1016/j.mrfmmm.2024.111876","url":null,"abstract":"<div><h3>Background</h3><p>Bladder cancer (BCa) is the most common malignancy with increasing morbidity and mortality. Circular RNA (circRNA) ATF6 level was downregulated in BCa after GSE92675 CircRNA microarray dataset was analyzed using GEO2R. However, its function and mechanism in BCa remain largely unknown.</p></div><div><h3>Methods</h3><p>GEO2R and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used to measure levels of circRNA ATF6, microRNA-146a-5p (miR-146a-5p), and filamin A (FLNA). CircRNA ATF6 stability was assessed by actinomycin D and RNase R assays, while circRNA ATF6 cellular localization was examined by FISH experiments in T24 cells. Cell counting kit-8 (CCK-8), colony formation, wound-healing, and transwell assays were used to study circRNA ATF6’s function in growth, motility, and invasion. By examining luciferase, starBase, RNA pull-down, and RNA immunoprecipitation (RIP) experiments, we anticipated and confirmed miR-146a-5p interactions with circRNA ATF6, as well as miR-146a-5p interactions with FLNA. On tumor-bearing mice, <em>in vivo</em> experiments were conducted.</p></div><div><h3>Results</h3><p>MiR-146a-5p expression in Bca was elevated, while circRNA ATF6 and FLNA were downregulated. CircRNA ATF6 showed better stability in BCa cells, with its expression primarily in the cytoplasm. Upregulating circRNA ATF6 lowered BCa cell viability, colony numbers, and invasion numbers, but broadened their migratory pattern. MiR-146a-5p was directly sponged up by circRNA ATF6, which also detrimentally affected miR-146a-5p levels in BCa. MiR-146a-5p reduced BCa FLNA expression by targeting FLNA. FLNA silencing abolished circRNA ATF6’s mitigating function in BCa cell proliferation, motility, and invasion. <em>In vivo</em>, overexpression of circRNA ATF6 significantly reduced tumor volume and weight.</p></div><div><h3>Conclusion</h3><p>CircRNA ATF6 suppresses BCa cell growth, migration and invasion through the miR-146a-5p/FLNA axis.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111876"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142049675","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular dynamics of DNA repair and carcinogen interaction: Implications for cancer initiation, progression, and therapeutic strategies DNA 修复与致癌物质相互作用的分子动力学:癌症发生、发展和治疗策略的意义
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111883
Eman Alyafeai , Eskandar Qaed , Haitham Saad Al-mashriqi , Ahmed Almaamari , Anisa H. Almansory , Fatima Al Futini , Marwa Sultan , Zeyao Tang

The integrity of the genetic material in human cells is continuously challenged by environmental agents and endogenous stresses. Among these, environmental carcinogens are pivotal in initiating complex DNA lesions that can lead to malignant transformations if not properly repaired. This review synthesizes current knowledge on the molecular dynamics of DNA repair mechanisms and their interplay with various environmental carcinogens, providing a comprehensive overview of how these interactions contribute to cancer initiation and progression. We examine key DNA repair pathways including base excision repair, nucleotide excision repair, and double-strand break repair and their regulatory networks, highlighting how defects in these pathways can exacerbate carcinogen-induced damage. Further, we discuss how understanding these molecular interactions offers novel insights into potential therapeutic strategies. This includes leveraging synthetic lethality concepts and designing targeted therapies that exploit specific DNA repair vulnerabilities in cancer cells. By integrating recent advances in molecular biology, genetics, and oncology, this review aims to illuminate the complex landscape of DNA repair and carcinogen-induced carcinogenesis, setting the stage for future research and therapeutic innovations.

人类细胞中遗传物质的完整性不断受到环境因素和内源性压力的挑战。其中,环境致癌物质是引发复杂 DNA 病变的关键因素,如果修复不当,就会导致恶性转化。本综述综合了目前有关 DNA 修复机制的分子动力学及其与各种环境致癌物相互作用的知识,全面概述了这些相互作用如何导致癌症的发生和发展。我们研究了包括碱基切除修复、核苷酸切除修复和双链断裂修复在内的关键 DNA 修复途径及其调控网络,重点介绍了这些途径的缺陷如何加剧致癌物质诱发的损伤。此外,我们还讨论了了解这些分子相互作用如何为潜在的治疗策略提供新的见解。这包括利用合成致死概念和设计靶向疗法,利用癌细胞中特定的 DNA 修复漏洞。通过整合分子生物学、遗传学和肿瘤学的最新进展,本综述旨在阐明 DNA 修复和致癌物诱导的致癌过程的复杂情况,为未来的研究和治疗创新奠定基础。
{"title":"Molecular dynamics of DNA repair and carcinogen interaction: Implications for cancer initiation, progression, and therapeutic strategies","authors":"Eman Alyafeai ,&nbsp;Eskandar Qaed ,&nbsp;Haitham Saad Al-mashriqi ,&nbsp;Ahmed Almaamari ,&nbsp;Anisa H. Almansory ,&nbsp;Fatima Al Futini ,&nbsp;Marwa Sultan ,&nbsp;Zeyao Tang","doi":"10.1016/j.mrfmmm.2024.111883","DOIUrl":"10.1016/j.mrfmmm.2024.111883","url":null,"abstract":"<div><p>The integrity of the genetic material in human cells is continuously challenged by environmental agents and endogenous stresses. Among these, environmental carcinogens are pivotal in initiating complex DNA lesions that can lead to malignant transformations if not properly repaired. This review synthesizes current knowledge on the molecular dynamics of DNA repair mechanisms and their interplay with various environmental carcinogens, providing a comprehensive overview of how these interactions contribute to cancer initiation and progression. We examine key DNA repair pathways including base excision repair, nucleotide excision repair, and double-strand break repair and their regulatory networks, highlighting how defects in these pathways can exacerbate carcinogen-induced damage. Further, we discuss how understanding these molecular interactions offers novel insights into potential therapeutic strategies. This includes leveraging synthetic lethality concepts and designing targeted therapies that exploit specific DNA repair vulnerabilities in cancer cells. By integrating recent advances in molecular biology, genetics, and oncology, this review aims to illuminate the complex landscape of DNA repair and carcinogen-induced carcinogenesis, setting the stage for future research and therapeutic innovations.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111883"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142167503","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Therapeutic potential of curcumin in autophagy modulation: Insights into the role of transcription factor EB 姜黄素在自噬调节中的治疗潜力:对转录因子 EB 作用的见解
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111879
Shabnam Radbakhsh , Prashant Kesharwani , Amirhossein Sahebkar

Transcription factor EB (TFEB) is a basic Helix–Loop–Helix/Leucine Zipper (bHLHZip) class of DNA-binding proteins, which can control the expression of genes included in the autophagy–lysosomal pathway. TFEB regulates the autophagic flux by enhancing lysosome biogenesis, forming autophagosomes, and fusion with lysosomes, thereby facilitating cellular clearance of pathogenic protein structures. Curcumin is a natural polyphenolic molecule with pharmacological properties that make it a potential therapeutic candidate for a wide range of diseases. One of the important curcumin mechanisms of action includes modulation of autophagy through affecting various signaling components such as TFEB. This review discusses in vitro and in vivo evidence on the effects of curcumin on autophagy process via modulating TFEB activity in different disorders.

转录因子 EB(TFEB)是基本螺旋-环-螺旋/亮氨酸拉链(bHLHZip)类 DNA 结合蛋白,可控制自噬-溶酶体途径中基因的表达。TFEB 通过增强溶酶体的生物生成、形成自噬体以及与溶酶体融合来调节自噬通量,从而促进细胞对致病蛋白结构的清除。姜黄素是一种天然多酚分子,其药理特性使其成为治疗多种疾病的潜在候选药物。姜黄素的重要作用机制之一包括通过影响各种信号成分(如 TFEB)来调节自噬。本综述将讨论姜黄素通过调节 TFEB 的活性对不同疾病的自噬过程产生影响的体外和体内证据。
{"title":"Therapeutic potential of curcumin in autophagy modulation: Insights into the role of transcription factor EB","authors":"Shabnam Radbakhsh ,&nbsp;Prashant Kesharwani ,&nbsp;Amirhossein Sahebkar","doi":"10.1016/j.mrfmmm.2024.111879","DOIUrl":"10.1016/j.mrfmmm.2024.111879","url":null,"abstract":"<div><p>Transcription factor EB (TFEB) is a basic Helix–Loop–Helix/Leucine Zipper (bHLHZip) class of DNA-binding proteins, which can control the expression of genes included in the autophagy–lysosomal pathway. TFEB regulates the autophagic flux by enhancing lysosome biogenesis, forming autophagosomes, and fusion with lysosomes, thereby facilitating cellular clearance of pathogenic protein structures. Curcumin is a natural polyphenolic molecule with pharmacological properties that make it a potential therapeutic candidate for a wide range of diseases. One of the important curcumin mechanisms of action includes modulation of autophagy through affecting various signaling components such as TFEB. This review discusses <em>in vitro</em> and <em>in vivo</em> evidence on the effects of curcumin on autophagy process <em>via</em> modulating TFEB activity in different disorders.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111879"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142040854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transcription factor NFYA inhibits ferroptosis in lung adenocarcinoma cells by regulating PEBP1 转录因子 NFYA 通过调控 PEBP1 抑制肺腺癌细胞的铁突变。
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111873
Feng Chen, Tingting Xu, Ni Jin, Digeng Li, Yanfu Ying, Chen Wang

Background

Ferroptosis is an iron-dependent programmed cell death mediated by lipid peroxidation. The purpose was to explore the molecular mechanism by which phosphatidylethanolamine-binding protein 1 (PEBP1) regulates ferroptosis in lung adenocarcinoma (LUAD), hoping to identify novel therapeutic targets for LUAD.

Methods

The expression, enrichment pathways and upstream transcription factors of PEBP1 were analyzed using bioinformatics tools. Dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) experiments were conducted to validate the interaction and binding relationship between PEBP1 and the upstream transcription factor nuclear transcription factor Y subunit α (NFYA). Quantitative reverse transcription PCR (qRT-PCR) was conducted to measure the expression levels of PEBP1 and NFYA mRNA in LUAD cells. Cell viability was detected by cell counting kit-8 assay. In addition, levels of malondialdehyde (MDA), Fe2+, and lipid reactive oxygen species (ROS) were assessed to evaluate ferroptosis levels in LUAD cells.

Results

PEBP1 was downregulated and significantly enriched in the ferroptosis signaling pathway in LUAD. Overexpression of PEBP1 suppressed cell viability remarkably, while levels of MDA, Fe2+, and lipid ROS were increased. Conversely, knockdown of PEBP1 produced the opposite effects. The upstream transcription factor NFYA, predicted to be involved in the regulation of PEBP1, was also upregulated in LUAD. Dual-luciferase reporter assay, ChIP, and molecular experiments revealed that NFYA transcriptionally suppressed the expression of PEBP1, and overexpression of NFYA could reverse the effects caused by PEBP1 overexpression.

Conclusion

PEBP1 regulated ferroptosis in LUAD, and the transcription factor NFYA inhibited ferroptosis in LUAD cells by transcriptionally downregulating PEBP1 expression.

背景:铁变性是一种由脂质过氧化介导的铁依赖性程序性细胞死亡。本研究的目的是探讨磷脂酰乙醇胺结合蛋白1(PEBP1)调控肺腺癌(LUAD)铁凋亡的分子机制,希望能找到治疗LUAD的新靶点:方法:利用生物信息学工具分析了PEBP1的表达、富集途径和上游转录因子。双荧光素酶报告实验和染色质免疫沉淀(ChIP)实验验证了PEBP1与上游转录因子核转录因子Y亚基α(NFYA)之间的相互作用和结合关系。定量反转录 PCR(qRT-PCR)测定了 PEBP1 和 NFYA mRNA 在 LUAD 细胞中的表达水平。细胞活力通过细胞计数试剂盒-8检测。此外,还评估了丙二醛(MDA)、Fe2+和脂质活性氧(ROS)的水平,以评价 LUAD 细胞的铁变态反应水平:结果:在LUAD细胞中,PEBP1被下调,并在铁变态反应信号通路中显著富集。结果:在 LUAD 细胞中,PEBP1 被下调,并在铁氧化信号通路中明显富集。过表达 PEBP1 会明显抑制细胞活力,同时 MDA、Fe2+ 和脂质 ROS 水平升高。相反,敲除 PEBP1 则会产生相反的效果。上游转录因子 NFYA 被认为参与了 PEBP1 的调控,在 LUAD 中也被上调。双荧光素酶报告实验、ChIP和分子实验显示,NFYA转录抑制了PEBP1的表达,而NFYA的过表达可以逆转PEBP1过表达造成的影响:结论:PEBP1调控LUAD细胞的铁突变,转录因子NFYA通过转录下调PEBP1的表达抑制LUAD细胞的铁突变。
{"title":"Transcription factor NFYA inhibits ferroptosis in lung adenocarcinoma cells by regulating PEBP1","authors":"Feng Chen,&nbsp;Tingting Xu,&nbsp;Ni Jin,&nbsp;Digeng Li,&nbsp;Yanfu Ying,&nbsp;Chen Wang","doi":"10.1016/j.mrfmmm.2024.111873","DOIUrl":"10.1016/j.mrfmmm.2024.111873","url":null,"abstract":"<div><h3>Background</h3><p>Ferroptosis is an iron-dependent programmed cell death mediated by lipid peroxidation. The purpose was to explore the molecular mechanism by which phosphatidylethanolamine-binding protein 1 (PEBP1) regulates ferroptosis in lung adenocarcinoma (LUAD), hoping to identify novel therapeutic targets for LUAD.</p></div><div><h3>Methods</h3><p>The expression, enrichment pathways and upstream transcription factors of PEBP1 were analyzed using bioinformatics tools. Dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) experiments were conducted to validate the interaction and binding relationship between PEBP1 and the upstream transcription factor nuclear transcription factor Y subunit α (NFYA). Quantitative reverse transcription PCR (qRT-PCR) was conducted to measure the expression levels of PEBP1 and NFYA mRNA in LUAD cells. Cell viability was detected by cell counting kit-8 assay. In addition, levels of malondialdehyde (MDA), Fe<sup>2+</sup>, and lipid reactive oxygen species (ROS) were assessed to evaluate ferroptosis levels in LUAD cells.</p></div><div><h3>Results</h3><p>PEBP1 was downregulated and significantly enriched in the ferroptosis signaling pathway in LUAD. Overexpression of PEBP1 suppressed cell viability remarkably, while levels of MDA, Fe<sup>2+</sup>, and lipid ROS were increased. Conversely, knockdown of PEBP1 produced the opposite effects. The upstream transcription factor NFYA, predicted to be involved in the regulation of PEBP1, was also upregulated in LUAD. Dual-luciferase reporter assay, ChIP, and molecular experiments revealed that NFYA transcriptionally suppressed the expression of PEBP1, and overexpression of NFYA could reverse the effects caused by PEBP1 overexpression.</p></div><div><h3>Conclusion</h3><p>PEBP1 regulated ferroptosis in LUAD, and the transcription factor NFYA inhibited ferroptosis in LUAD cells by transcriptionally downregulating PEBP1 expression.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111873"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141602398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Knockdown of SDCBP induces autophagy to promote cardiomyocyte growth and angiogenesis in hypoxia/reoxygenation model 在缺氧/复氧模型中敲除 SDCBP 可诱导自噬,促进心肌细胞生长和血管生成
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111885
Ling Gao, Wanqian Liu

Objective

Angina, myocardial infarction, and even mortality can result from myocardial ischemia (MI). Angiogenesis facilitates tissue repair, lessens cell damage, and ensures that ischemic tissues receive blood and oxygen. This study investigated the possible mechanism of syndecan-binding protein (SDCBP) on autophagy and assessed its impact on myocardial ischemia.

Method

A cardiac hypoxia-reoxygenation (H/R) cell model was created for this investigation. Flow cytometry, the cell counting kit-8, and Western blotting were used to measure the damage to cardiomyocytes. Western blotting and immunofluorescence were used to quantify autophagy. Furthermore, assays for tube formation, migration, and Western blotting were used to assess angiogenic capacity. Additionally, the EGFR-PI3K-Akt signaling pathway's activation was found using Western blotting.

Result

In the H/R-induced cardiomyocyte model, there is a rise in the expression of SDCBP. Treatment with H/R markedly boosted apoptosis and considerably decreased cell survival. H/R induction strongly inhibits autophagy, increases P62 expression, and decreases LC3II/I expression. Moreover, H/R induction dramatically reduced the ability to form tubes, migrate, and express VEGF, all of which prevented cell angiogenesis. Furthermore, EGFR-PI3K-Akt signaling pathway expression is strongly inhibited by H/R induction. considerable reduction of H/R-induced cell damage, considerable inhibition of apoptosis, promotion autophagy and angiogenesis, and activation of the EGFR-PI3K-Akt signaling pathway are all possible with SDCBP knockdown.

Conclusion

To summarize, this study demonstrates that via stimulating the EGFR-PI3K-Akt signaling pathway, SDCBP knockdown may mitigate the effects of H/R-induced cardiomyocyte death and encourage autophagy and blood vessel formation. A theoretical foundation for possible myocardial infarction treatment is thus provided.
目的心肌缺血(MI)可导致心绞痛、心肌梗塞,甚至死亡。血管生成有利于组织修复,减轻细胞损伤,确保缺血组织获得血液和氧气。本研究探讨了辛迪卡结合蛋白(SDCBP)对自噬的可能机制,并评估了其对心肌缺血的影响。采用流式细胞术、细胞计数试剂盒-8 和 Western 印迹法测量心肌细胞受损情况。Western 印迹法和免疫荧光法用于量化自噬。此外,还使用管形成、迁移和 Western 印迹法评估血管生成能力。结果 在 H/R 诱导的心肌细胞模型中,SDCBP 的表达上升。结果 在 H/R 诱导的心肌细胞模型中,SDCBP 的表达上升,H/R 处理显著促进细胞凋亡并大大降低细胞存活率。H/R 诱导强烈抑制自噬,增加 P62 表达,减少 LC3II/I 表达。此外,H/R 诱导还显著降低了细胞形成管道、迁移和表达血管内皮生长因子的能力,所有这些都阻碍了细胞的血管生成。此外,H/R 诱导还强烈抑制了表皮生长因子受体-PI3K-Akt 信号通路的表达。敲除 SDCBP 可显著减少 H/R 诱导的细胞损伤,抑制细胞凋亡,促进自噬和血管生成,并激活表皮生长因子受体-PI3K-Akt 信号通路。结论综上所述,本研究表明,通过刺激表皮生长因子受体-PI3K-Akt 信号通路,SDCBP 敲除可减轻 H/R 诱导的心肌细胞死亡的影响,并促进自噬和血管形成。这为可能的心肌梗死治疗提供了理论基础。
{"title":"Knockdown of SDCBP induces autophagy to promote cardiomyocyte growth and angiogenesis in hypoxia/reoxygenation model","authors":"Ling Gao,&nbsp;Wanqian Liu","doi":"10.1016/j.mrfmmm.2024.111885","DOIUrl":"10.1016/j.mrfmmm.2024.111885","url":null,"abstract":"<div><h3>Objective</h3><div>Angina, myocardial infarction, and even mortality can result from myocardial ischemia (MI). Angiogenesis facilitates tissue repair, lessens cell damage, and ensures that ischemic tissues receive blood and oxygen. This study investigated the possible mechanism of syndecan-binding protein (SDCBP) on autophagy and assessed its impact on myocardial ischemia.</div></div><div><h3>Method</h3><div>A cardiac hypoxia-reoxygenation (H/R) cell model was created for this investigation. Flow cytometry, the cell counting kit-8, and Western blotting were used to measure the damage to cardiomyocytes. Western blotting and immunofluorescence were used to quantify autophagy. Furthermore, assays for tube formation, migration, and Western blotting were used to assess angiogenic capacity. Additionally, the EGFR-PI3K-Akt signaling pathway's activation was found using Western blotting.</div></div><div><h3>Result</h3><div>In the H/R-induced cardiomyocyte model, there is a rise in the expression of SDCBP. Treatment with H/R markedly boosted apoptosis and considerably decreased cell survival. H/R induction strongly inhibits autophagy, increases P62 expression, and decreases LC3II/I expression. Moreover, H/R induction dramatically reduced the ability to form tubes, migrate, and express VEGF, all of which prevented cell angiogenesis. Furthermore, EGFR-PI3K-Akt signaling pathway expression is strongly inhibited by H/R induction. considerable reduction of H/R-induced cell damage, considerable inhibition of apoptosis, promotion autophagy and angiogenesis, and activation of the EGFR-PI3K-Akt signaling pathway are all possible with SDCBP knockdown.</div></div><div><h3>Conclusion</h3><div>To summarize, this study demonstrates that via stimulating the EGFR-PI3K-Akt signaling pathway, SDCBP knockdown may mitigate the effects of H/R-induced cardiomyocyte death and encourage autophagy and blood vessel formation. A theoretical foundation for possible myocardial infarction treatment is thus provided.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111885"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142561078","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MicroRNA-532 as a probable diagnostic and therapeutic marker in cancer patients MicroRNA-532 可能是癌症患者的诊断和治疗标志物。
IF 1.5 4区 医学 Q4 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-01 DOI: 10.1016/j.mrfmmm.2024.111874
Malihe Lotfi , Amirhosein Maharati , Amir Abbas Hamidi , Negin Taghehchian , Meysam Moghbeli

The high mortality rate in cancer patients is always one of the main challenges of the health systems globally. Several factors are involved in the high rate of cancer related mortality, including late diagnosis and drug resistance. Cancer is mainly diagnosed in the advanced stages of tumor progression that causes the failure of therapeutic strategies and increases the death rate in these patients. Therefore, assessment of the molecular mechanisms associated with the occurrence of cancer can be effective to introduce early tumor diagnostic markers. MicroRNAs (miRNAs) as the stable non-coding RNAs in the biological body fluids are involved in regulation of cell proliferation, migration, and apoptosis. MiR-532 deregulation has been reported in different tumor types. Therefore, in the present review we discussed the role of miR-532 during tumor growth. It has been shown that miR-532 has mainly a tumor suppressor role through the regulation of transcription factors, chemokines, and signaling pathways such as NF-kB, MAPK, PI3K/AKT, and WNT. In addition to the independent role of miR-532 in regulation of cellular processes, it also functions as a mediator of lncRNAs and circRNAs. Therefore, miR-532 can be considered as a non-invasive diagnostic/prognostic marker as well as a therapeutic target in cancer patients.

癌症患者的高死亡率一直是全球卫生系统面临的主要挑战之一。与癌症相关的高死亡率涉及多个因素,包括诊断过晚和耐药性。癌症主要在肿瘤进展的晚期才被诊断出来,这导致治疗策略失败,增加了这些患者的死亡率。因此,评估与癌症发生相关的分子机制可有效引入早期肿瘤诊断标记物。微RNA(miRNA)作为生物体液中稳定的非编码RNA,参与细胞增殖、迁移和凋亡的调控。不同类型的肿瘤中都有 MiR-532 失调的报道。因此,本综述讨论了 miR-532 在肿瘤生长过程中的作用。研究表明,miR-532 主要通过调节转录因子、趋化因子以及 NF-kB、MAPK、PI3K/AKT 和 WNT 等信号通路发挥抑瘤作用。miR-532 除了在调控细胞过程中发挥独立作用外,还可作为 lncRNA 和 circRNA 的媒介发挥作用。因此,miR-532 可被视为癌症患者的非侵入性诊断/预后标志物和治疗靶点。
{"title":"MicroRNA-532 as a probable diagnostic and therapeutic marker in cancer patients","authors":"Malihe Lotfi ,&nbsp;Amirhosein Maharati ,&nbsp;Amir Abbas Hamidi ,&nbsp;Negin Taghehchian ,&nbsp;Meysam Moghbeli","doi":"10.1016/j.mrfmmm.2024.111874","DOIUrl":"10.1016/j.mrfmmm.2024.111874","url":null,"abstract":"<div><p>The high mortality rate in cancer patients is always one of the main challenges of the health systems globally. Several factors are involved in the high rate of cancer related mortality, including late diagnosis and drug resistance. Cancer is mainly diagnosed in the advanced stages of tumor progression that causes the failure of therapeutic strategies and increases the death rate in these patients. Therefore, assessment of the molecular mechanisms associated with the occurrence of cancer can be effective to introduce early tumor diagnostic markers. MicroRNAs (miRNAs) as the stable non-coding RNAs in the biological body fluids are involved in regulation of cell proliferation, migration, and apoptosis. MiR-532 deregulation has been reported in different tumor types. Therefore, in the present review we discussed the role of miR-532 during tumor growth. It has been shown that miR-532 has mainly a tumor suppressor role through the regulation of transcription factors, chemokines, and signaling pathways such as NF-kB, MAPK, PI3K/AKT, and WNT. In addition to the independent role of miR-532 in regulation of cellular processes, it also functions as a mediator of lncRNAs and circRNAs. Therefore, miR-532 can be considered as a non-invasive diagnostic/prognostic marker as well as a therapeutic target in cancer patients.</p></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"829 ","pages":"Article 111874"},"PeriodicalIF":1.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141581906","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis
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