首页 > 最新文献

Experimental cell research最新文献

英文 中文
EBP50 regulates senescence and focal adhesion in endometrial carcinoma
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1016/j.yexcr.2025.114465
Ako Yokoi , Ryoya Ogomori , Yasuko Oguri, Miki Hashimura, Makoto Saegusa
Ezrin-radixin-moesin (ERM)-binding phosphoprotein 50 (EBP50) is a multifunctional scaffold protein that is highly expressed in polarized epithelial cells. Here, we focused on the functional roles of EBP50 in endometrial carcinoma (Em Ca). We analyzed immunohistochemical sections from 121 Em Ca and 30 normal samples. We also characterized EBP50 overexpression or knockout (KO) Em Ca cell lines. High levels of membranous (Me) EBP50 expression were observed in endometrial tissues from normal menstrual cycles, in contrast to the transient upregulation of cytoplasmic (Cyt) EBP50 in tissues in the proliferative phase; this was probably in response to estrogenic effects. There was a significant stepwise reduction of Me-EBP50 expression from grade (G) 1 to G3 Em Cas, which was consistent with the loss of glandular structures. Conversely, Cyt-EBP50 levels increased with in the higher tumor grades. Low Me-EBP50 expression was significantly associated with tumor lymphovascular invasion and short overall survival. Whereas EBP50 KO led to senescence and reduced proliferation and motility, overexpression elicited the opposite phenotypes. Moreover, the number of focal adhesions (FAs), which mediate cell migration, was significantly increased in EBP50 overexpressing cells but decreased in the KO cells. In conclusion, Me- and/or Cyt-EBP50 expression contributes to acceleration of cell motility through enhancement of FA formation, and inhibits senescence to promote cytokinesis. Together, these effects contribute to Em Ca aggressiveness.
{"title":"EBP50 regulates senescence and focal adhesion in endometrial carcinoma","authors":"Ako Yokoi ,&nbsp;Ryoya Ogomori ,&nbsp;Yasuko Oguri,&nbsp;Miki Hashimura,&nbsp;Makoto Saegusa","doi":"10.1016/j.yexcr.2025.114465","DOIUrl":"10.1016/j.yexcr.2025.114465","url":null,"abstract":"<div><div>Ezrin-radixin-moesin (ERM)-binding phosphoprotein 50 (EBP50) is a multifunctional scaffold protein that is highly expressed in polarized epithelial cells. Here, we focused on the functional roles of EBP50 in endometrial carcinoma (Em Ca). We analyzed immunohistochemical sections from 121 Em Ca and 30 normal samples. We also characterized EBP50 overexpression or knockout (KO) Em Ca cell lines. High levels of membranous (Me) EBP50 expression were observed in endometrial tissues from normal menstrual cycles, in contrast to the transient upregulation of cytoplasmic (Cyt) EBP50 in tissues in the proliferative phase; this was probably in response to estrogenic effects. There was a significant stepwise reduction of Me-EBP50 expression from grade (G) 1 to G3 Em Cas, which was consistent with the loss of glandular structures. Conversely, Cyt-EBP50 levels increased with in the higher tumor grades. Low Me-EBP50 expression was significantly associated with tumor lymphovascular invasion and short overall survival. Whereas EBP50 KO led to senescence and reduced proliferation and motility, overexpression elicited the opposite phenotypes. Moreover, the number of focal adhesions (FAs), which mediate cell migration, was significantly increased in EBP50 overexpressing cells but decreased in the KO cells. In conclusion, Me- and/or Cyt-EBP50 expression contributes to acceleration of cell motility through enhancement of FA formation, and inhibits senescence to promote cytokinesis. Together, these effects contribute to Em Ca aggressiveness.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"446 1","pages":"Article 114465"},"PeriodicalIF":3.3,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143437187","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
USP48 inhibits colorectal cancer progression and promotes M1-like macrophage polarization by stabilizing TAK1
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1016/j.yexcr.2025.114469
Xinwen Zhang , Jiawei Zhao , Guangying Qi , Yujing Chen , Xiaotong Guo , Juzheng Zhang , Siqi Chen , Xiaochen Xu , Jiayuan Feng , Qinyu Zhang , Bin Gao , Zhenran Wang , Jiamin Jin
Ubiquitination and deubiquitination have emerged as pivotal regulators of the development of colorectal cancer (CRC). However, the precise role of USP48 in CRC tumorigenesis is poorly understood. In this study, immunohistochemistry, protein blotting, MTT assays, plate cloning, scratch assays, transwell assays, and Hoechst 33258 staining were utilized to assess the expression level of USP48 and its involvement in CRC. The interaction between USP48 and Transforming growth factor-β activated kinase-1(TAK1) was confirmed using co-IP. Additionally, the impact of deubiquitination on downstream signaling was determined through qRT-PCR. Furthermore, the associations between USP48 and tumor-associated macrophages within the tumor microenvironment were investigated using flow cytometry. The findings of our study demonstrated that USP48 expression is downregulated in CRC patients. Through deubiquitination, USP48 interacts with and stabilizes TAK1, leading to the inhibition of TAK1-triggered NF-κB activation and effectively suppresses CRC tumorigenesis. Moreover, this study showed a positive correlation between USP48 expression and M1-type TAM polarization, revealed the potential of USP48 as a molecular target for the effective treatment of CRC in the future.
泛素化和去泛素化已成为结直肠癌(CRC)发病的关键调节因子。然而,USP48 在 CRC 肿瘤发生中的确切作用却鲜为人知。本研究利用免疫组化、蛋白印迹、MTT 试验、平板克隆、划痕试验、transwell 试验和 Hoechst 33258 染色来评估 USP48 的表达水平及其在 CRC 中的参与情况。利用共印迹技术证实了 USP48 与转化生长因子-β 激活激酶-1(TAK1)之间的相互作用。此外,还通过 qRT-PCR 确定了去泛素化对下游信号转导的影响。此外,还使用流式细胞术研究了 USP48 与肿瘤微环境中的肿瘤相关巨噬细胞之间的关联。我们的研究结果表明,USP48 在 CRC 患者中表达下调。通过去泛素化,USP48 与 TAK1 相互作用并稳定 TAK1,从而抑制 TAK1 触发的 NF-κB 激活,有效抑制 CRC 肿瘤发生。此外,该研究还显示 USP48 的表达与 M1 型 TAM 极化之间存在正相关,揭示了 USP48 作为分子靶点在未来有效治疗 CRC 的潜力。
{"title":"USP48 inhibits colorectal cancer progression and promotes M1-like macrophage polarization by stabilizing TAK1","authors":"Xinwen Zhang ,&nbsp;Jiawei Zhao ,&nbsp;Guangying Qi ,&nbsp;Yujing Chen ,&nbsp;Xiaotong Guo ,&nbsp;Juzheng Zhang ,&nbsp;Siqi Chen ,&nbsp;Xiaochen Xu ,&nbsp;Jiayuan Feng ,&nbsp;Qinyu Zhang ,&nbsp;Bin Gao ,&nbsp;Zhenran Wang ,&nbsp;Jiamin Jin","doi":"10.1016/j.yexcr.2025.114469","DOIUrl":"10.1016/j.yexcr.2025.114469","url":null,"abstract":"<div><div>Ubiquitination and deubiquitination have emerged as pivotal regulators of the development of colorectal cancer (CRC). However, the precise role of USP48 in CRC tumorigenesis is poorly understood. In this study, immunohistochemistry, protein blotting, MTT assays, plate cloning, scratch assays, transwell assays, and Hoechst 33258 staining were utilized to assess the expression level of USP48 and its involvement in CRC. The interaction between USP48 and Transforming growth factor-β activated kinase-1(TAK1) was confirmed using co-IP. Additionally, the impact of deubiquitination on downstream signaling was determined through qRT-PCR. Furthermore, the associations between USP48 and tumor-associated macrophages within the tumor microenvironment were investigated using flow cytometry. The findings of our study demonstrated that USP48 expression is downregulated in CRC patients. Through deubiquitination, USP48 interacts with and stabilizes TAK1, leading to the inhibition of TAK1-triggered NF-κB activation and effectively suppresses CRC tumorigenesis. Moreover, this study showed a positive correlation between USP48 expression and M1-type TAM polarization, revealed the potential of USP48 as a molecular target for the effective treatment of CRC in the future.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"446 1","pages":"Article 114469"},"PeriodicalIF":3.3,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143437792","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Threonine and tyrosine kinase promotes multiple myeloma progression by regulating regucalcin expression.
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-15 DOI: 10.1016/j.yexcr.2025.114454
Xiaofeng Zhu, Zuxi Feng, Xiaohuan Peng, Tianning Di, YanHong Li, Jun Bai, Tao Ma, Lijuan Li, Liansheng Zhang

Multiple myeloma (MM) is a malignant proliferative disorder of plasma cells and remains an incurable disease. Threonine and tyrosine kinase (TTK) is a dual-specific protein kinase that targets serine/threonine and tyrosine residues for phosphorylation. Its elevated expression has been linked to unfavorable outcomes in several types of cancer. Although the role of TTK in MM are still incompletely understood. In this research, we assessed TTK mRNA and protein expression levels in 51 MM patients and 30 healthy donors using qRT-PCR and western blotting. The impact of TTK expression on MM cell apoptosis, proliferation, and the cell cycle were assessed through CCK-8 assay, flow cytometry, and western blotting. Our findings revealed a significant overexpression of TTK in multiple myeloma patients and cell lines. TTK knockdown promoted apoptosis and G0/G1 phase arrest while inhibiting proliferation in MM cells, whereas TTK overexpression reduced apoptosis and G0/G1 phase arrest, enhancing proliferation in MM cells. Next, we identified regucalcin (RGN) as a downstream target of TTK through proteomic analysis. In NDMM, the expression of RGN was decreased. Cell function experiments showed that RGN knockdown significantly promoted MM cell proliferation, inhibited apoptosis and reduced cell cycle arrest, and reversed the increased apoptosis, weakened proliferation, and enhanced cell cycle arrest caused by TTK knockdown. Finally, a xenograft mouse model showed that TTK significantly promotes MM development. In summary, we demonstrated that the TTK-RGN axis regulates cell apoptosis, G0/G1 phase arrest, and proliferation in MM, highlighting TTK as a potential target for therapeutic intervention in this cancer.

{"title":"Threonine and tyrosine kinase promotes multiple myeloma progression by regulating regucalcin expression.","authors":"Xiaofeng Zhu, Zuxi Feng, Xiaohuan Peng, Tianning Di, YanHong Li, Jun Bai, Tao Ma, Lijuan Li, Liansheng Zhang","doi":"10.1016/j.yexcr.2025.114454","DOIUrl":"https://doi.org/10.1016/j.yexcr.2025.114454","url":null,"abstract":"<p><p>Multiple myeloma (MM) is a malignant proliferative disorder of plasma cells and remains an incurable disease. Threonine and tyrosine kinase (TTK) is a dual-specific protein kinase that targets serine/threonine and tyrosine residues for phosphorylation. Its elevated expression has been linked to unfavorable outcomes in several types of cancer. Although the role of TTK in MM are still incompletely understood. In this research, we assessed TTK mRNA and protein expression levels in 51 MM patients and 30 healthy donors using qRT-PCR and western blotting. The impact of TTK expression on MM cell apoptosis, proliferation, and the cell cycle were assessed through CCK-8 assay, flow cytometry, and western blotting. Our findings revealed a significant overexpression of TTK in multiple myeloma patients and cell lines. TTK knockdown promoted apoptosis and G0/G1 phase arrest while inhibiting proliferation in MM cells, whereas TTK overexpression reduced apoptosis and G0/G1 phase arrest, enhancing proliferation in MM cells. Next, we identified regucalcin (RGN) as a downstream target of TTK through proteomic analysis. In NDMM, the expression of RGN was decreased. Cell function experiments showed that RGN knockdown significantly promoted MM cell proliferation, inhibited apoptosis and reduced cell cycle arrest, and reversed the increased apoptosis, weakened proliferation, and enhanced cell cycle arrest caused by TTK knockdown. Finally, a xenograft mouse model showed that TTK significantly promotes MM development. In summary, we demonstrated that the TTK-RGN axis regulates cell apoptosis, G0/G1 phase arrest, and proliferation in MM, highlighting TTK as a potential target for therapeutic intervention in this cancer.</p>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":" ","pages":"114454"},"PeriodicalIF":3.3,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ACSS3 promotes the tumorigenesis of non-small cell lung cancer via suppressing p53-mediated ferroptosis.
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-15 DOI: 10.1016/j.yexcr.2025.114438
Jing Zhang, Xiuhong Wang, Jingyi Wang, Xiao Wen, Siyuan Chen, Tao Wang, Bei Wang, Wenquan Hu

Non-small cell lung cancer (NSCLC) is a subtype of the most frequently diagnosed cancer, causing a considerable number of deaths globally. Mitochondrial dysfunction was found to promote malignant progression. However, the underlying mechanism remains unclear. Acyl-CoA synthetase short chain family member 3 (ACSS3) is mainly located in mitochondria, which abnormal regulation is usually accompanied by the occurrence and development of tumors. In this study, we found that the expression level of ACSS3 was correlated with poor prognosis in patients with NSCLC. Moreover, we demonstrated that ACSS3 knockdown led to mitochondrial contraction, increased reactive oxygen species levels, decreased mitochondrial membrane potential, and subsequently inhibited tumor growth of NSCLC cells in vitro and in vivo, whereas its overexpression promoted these processes. Mechanistically, ACSS3 knockdown promoted ferroptosis through transcriptional control of SLC7A11 and GPX4. Further investigations indicated that ACSS3 loss inhibited the SLC7A11/GPX4 axis by enhancing p53 stability. Taken together, our data confirmed that ACSS3 promotes NSCLC tumorigenesis through inhibiting the p53-mediated ferroptosis. Hence, ACSS3 emerges as a promising therapeutic target for NSCLC treatment.

{"title":"ACSS3 promotes the tumorigenesis of non-small cell lung cancer via suppressing p53-mediated ferroptosis.","authors":"Jing Zhang, Xiuhong Wang, Jingyi Wang, Xiao Wen, Siyuan Chen, Tao Wang, Bei Wang, Wenquan Hu","doi":"10.1016/j.yexcr.2025.114438","DOIUrl":"https://doi.org/10.1016/j.yexcr.2025.114438","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) is a subtype of the most frequently diagnosed cancer, causing a considerable number of deaths globally. Mitochondrial dysfunction was found to promote malignant progression. However, the underlying mechanism remains unclear. Acyl-CoA synthetase short chain family member 3 (ACSS3) is mainly located in mitochondria, which abnormal regulation is usually accompanied by the occurrence and development of tumors. In this study, we found that the expression level of ACSS3 was correlated with poor prognosis in patients with NSCLC. Moreover, we demonstrated that ACSS3 knockdown led to mitochondrial contraction, increased reactive oxygen species levels, decreased mitochondrial membrane potential, and subsequently inhibited tumor growth of NSCLC cells in vitro and in vivo, whereas its overexpression promoted these processes. Mechanistically, ACSS3 knockdown promoted ferroptosis through transcriptional control of SLC7A11 and GPX4. Further investigations indicated that ACSS3 loss inhibited the SLC7A11/GPX4 axis by enhancing p53 stability. Taken together, our data confirmed that ACSS3 promotes NSCLC tumorigenesis through inhibiting the p53-mediated ferroptosis. Hence, ACSS3 emerges as a promising therapeutic target for NSCLC treatment.</p>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":" ","pages":"114438"},"PeriodicalIF":3.3,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440250","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
BRAF V600E in Cancer: Exploring Structural Complexities, Mutation Profiles, and Pathway Dysregulation.
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-15 DOI: 10.1016/j.yexcr.2025.114440
Jayhind Bharti, Priyadharshini Gogu, Sarvesh Kumar Pandey, Amita Verma, Jagat Pal Yadav, Ankit Kumar Singh, Pradeep Kumar, Ashish Ranjan Dwivedi, Prateek Pathak

BRAF, a fundamental component of cellular signaling pathways regulating growth and survival, is frequently mutated in cancer development. Among entire BRAF mutations, the V600E substitution stands out as a dominant alteration in various malignancies, including melanoma, colorectal cancer, and thyroid cancer. Understanding the structural differences between wild-type BRAF and BRAFV600E is crucial for elucidating the molecular mechanisms underpinnings tumorigenesis and identifying dysregulation associated with the same. V600E mutation results in a constitutively active kinase domain, leading to dysregulated downstream signaling independent of extracellular stimuli. This sustained activation promotes cell proliferation, survival, angiogenesis, and hallmark features of the cancer cells. The study describes three distinct classes of BRAF mutations where Class 1 mutations predominantly involve point mutations within the BRAF gene, while Class 2 encompasses in-frame insertions and deletions, and Class 3 comprises gene fusions with large-scale chromosomal rearrangements. Further, we have discussed dysregulated pathways associated with mutation of BRAFV600E, which includes MAPK/ERK, PI3K/AKT/mTOR, TP53, DNA damage response, and WNT/β-Catenin from schematic representation. In the current review, we have shown how these dysregulated pathways play pivotal roles in tumorigenesis, tumor progression in BRAF-mutant cancers and highlighted the critical role of BRAF dysregulation in cancer development followed by its therapeutic implications of targeting dysregulated pathways in BRAF-driven malignancies.

{"title":"BRAF V600E in Cancer: Exploring Structural Complexities, Mutation Profiles, and Pathway Dysregulation.","authors":"Jayhind Bharti, Priyadharshini Gogu, Sarvesh Kumar Pandey, Amita Verma, Jagat Pal Yadav, Ankit Kumar Singh, Pradeep Kumar, Ashish Ranjan Dwivedi, Prateek Pathak","doi":"10.1016/j.yexcr.2025.114440","DOIUrl":"https://doi.org/10.1016/j.yexcr.2025.114440","url":null,"abstract":"<p><p>BRAF, a fundamental component of cellular signaling pathways regulating growth and survival, is frequently mutated in cancer development. Among entire BRAF mutations, the V600E substitution stands out as a dominant alteration in various malignancies, including melanoma, colorectal cancer, and thyroid cancer. Understanding the structural differences between wild-type BRAF and BRAFV600E is crucial for elucidating the molecular mechanisms underpinnings tumorigenesis and identifying dysregulation associated with the same. V600E mutation results in a constitutively active kinase domain, leading to dysregulated downstream signaling independent of extracellular stimuli. This sustained activation promotes cell proliferation, survival, angiogenesis, and hallmark features of the cancer cells. The study describes three distinct classes of BRAF mutations where Class 1 mutations predominantly involve point mutations within the BRAF gene, while Class 2 encompasses in-frame insertions and deletions, and Class 3 comprises gene fusions with large-scale chromosomal rearrangements. Further, we have discussed dysregulated pathways associated with mutation of BRAFV600E, which includes MAPK/ERK, PI3K/AKT/mTOR, TP53, DNA damage response, and WNT/β-Catenin from schematic representation. In the current review, we have shown how these dysregulated pathways play pivotal roles in tumorigenesis, tumor progression in BRAF-mutant cancers and highlighted the critical role of BRAF dysregulation in cancer development followed by its therapeutic implications of targeting dysregulated pathways in BRAF-driven malignancies.</p>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":" ","pages":"114440"},"PeriodicalIF":3.3,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440320","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Modeling amyotrophic lateral sclerosis with amniotic membrane-derived mesenchymal stem cells: A novel approach for disease modeling.
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-15 DOI: 10.1016/j.yexcr.2025.114449
B S Soumya, Naisarg Gamit, Manasi Patil, V P Shreenidhi, Arun Dharmarajan, Sudha Warrier

Advancement of therapeutics for neurodegenerative diseases like amyotrophic lateral sclerosis (ALS) has been predominantly hampered by the dearth of relevant disease models. Despite numerous animal models, significant challenges remain in correlating these with human disease complexities. In this study, the ALS model was created using amniotic membrane-derived mesenchymal stem cells (AM-MSCs) which were differentiated into motor neurons (MN) with specific MN induction media and transiently transfected with mutated human SOD1 G93A plasmid to induce ALS-like condition. Characterization included gene expression analysis, immunocytochemistry, flow cytometry, and Western blot. Functional assays assessed the extent of degeneration and model efficiency. AM-MSCs demonstrated multipotency and were positive for MSC markers. Upon differentiation, the expression of MN markers like MNX1, Olig2, and ChAT were found to be elevated. SOD1 G93A overexpression, downregulated MN markers, upregulated NURR1 gene, reduced acetylcholine (ACh), reduced glutathione, and elevated oxidative stress markers. This robust in-vitro ALS model derived from AM-MSCs offers an alternative to animal models to provide an efficient and cost-effective platform to conduct rapid drug screening.

{"title":"Modeling amyotrophic lateral sclerosis with amniotic membrane-derived mesenchymal stem cells: A novel approach for disease modeling.","authors":"B S Soumya, Naisarg Gamit, Manasi Patil, V P Shreenidhi, Arun Dharmarajan, Sudha Warrier","doi":"10.1016/j.yexcr.2025.114449","DOIUrl":"https://doi.org/10.1016/j.yexcr.2025.114449","url":null,"abstract":"<p><p>Advancement of therapeutics for neurodegenerative diseases like amyotrophic lateral sclerosis (ALS) has been predominantly hampered by the dearth of relevant disease models. Despite numerous animal models, significant challenges remain in correlating these with human disease complexities. In this study, the ALS model was created using amniotic membrane-derived mesenchymal stem cells (AM-MSCs) which were differentiated into motor neurons (MN) with specific MN induction media and transiently transfected with mutated human SOD1 G93A plasmid to induce ALS-like condition. Characterization included gene expression analysis, immunocytochemistry, flow cytometry, and Western blot. Functional assays assessed the extent of degeneration and model efficiency. AM-MSCs demonstrated multipotency and were positive for MSC markers. Upon differentiation, the expression of MN markers like MNX1, Olig2, and ChAT were found to be elevated. SOD1 G93A overexpression, downregulated MN markers, upregulated NURR1 gene, reduced acetylcholine (ACh), reduced glutathione, and elevated oxidative stress markers. This robust in-vitro ALS model derived from AM-MSCs offers an alternative to animal models to provide an efficient and cost-effective platform to conduct rapid drug screening.</p>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":" ","pages":"114449"},"PeriodicalIF":3.3,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440273","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","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 PLOD2 inhibits pulmonary artery smooth muscle cell glycolysis under chronic intermittent hypoxia via PI3K/AKT signal
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-15 DOI: 10.1016/j.yexcr.2025.114453
Shenwen Yi , Tiange Qu , Heling Wu, Chenyu Xu, Jun Xu, Fei Yu, Liang Ye

Objective

This study aimed to investigate the role and potential mechanism of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) in chronic intermittent hypoxia (CIH)-induced mice and pulmonary arterial smooth muscle cells (PASMCs).

Methods

CIH mouse model was pre-injected with AAV-shPLOD2 by tail vein, and the pathological changes of lung was evaluated using hematoxylin-eosin (H&E) and α-SMA immunostaining. Enriched KEGG pathway analyses of PLOD2 targeted genes were performed using GSE11341 and GSE131425 datasets. Next, primary PASMCs were exposed to CIH environment, and then measured its proliferation, migration and glycolysis by CCK8, EdU assay, wound healing assay, Transwell and western blotting.

Results

PLOD2 expression was increased in the lungs of CIH-induced mice and in PASMCs under CIH conditions. Moreover, glycolysis and PI3K/AKT pathway were regulated by PLOD2. Silencing of PLOD2 significantly inhibited the increase of RV/(LV + S) and RVSP, alleviated pathological changes of lung in CIH-induced mice and restrained the proliferation, migration, glycolysis and activation of PI3K/AKT in CIH-induced PASMCs. The inhibitory effects of PLOD2 silencing on PASMC proliferation and migration were accelerated by 2-DG (an inhibitor of glycolysis) and were reversed by lactate (the end product of glycolysis). In addition, the inhibitory effects of PLOD2 silencing on PASMC proliferation, migration and glycolysis were accelerated by PI3K/AKT inhibitor LY294002 and were reversed by the agonist 740Y-P.

Conclusions

Silencing of PLOD2 inhibits PI3K/AKT signaling to limit PASMC glycolysis which allows PASMC proliferation and migration in CIH-induced pulmonary arterial hypertension (PAH).
{"title":"Knockdown of PLOD2 inhibits pulmonary artery smooth muscle cell glycolysis under chronic intermittent hypoxia via PI3K/AKT signal","authors":"Shenwen Yi ,&nbsp;Tiange Qu ,&nbsp;Heling Wu,&nbsp;Chenyu Xu,&nbsp;Jun Xu,&nbsp;Fei Yu,&nbsp;Liang Ye","doi":"10.1016/j.yexcr.2025.114453","DOIUrl":"10.1016/j.yexcr.2025.114453","url":null,"abstract":"<div><h3>Objective</h3><div>This study aimed to investigate the role and potential mechanism of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) in chronic intermittent hypoxia (CIH)-induced mice and pulmonary arterial smooth muscle cells (PASMCs).</div></div><div><h3>Methods</h3><div>CIH mouse model was pre-injected with AAV-shPLOD2 by tail vein, and the pathological changes of lung was evaluated using hematoxylin-eosin (H&amp;E) and α-SMA immunostaining. Enriched KEGG pathway analyses of PLOD2 targeted genes were performed using GSE11341 and GSE131425 datasets. Next, primary PASMCs were exposed to CIH environment, and then measured its proliferation, migration and glycolysis by CCK8, EdU assay, wound healing assay, Transwell and western blotting.</div></div><div><h3>Results</h3><div>PLOD2 expression was increased in the lungs of CIH-induced mice and in PASMCs under CIH conditions. Moreover, glycolysis and PI3K/AKT pathway were regulated by PLOD2. Silencing of PLOD2 significantly inhibited the increase of RV/(LV + S) and RVSP, alleviated pathological changes of lung in CIH-induced mice and restrained the proliferation, migration, glycolysis and activation of PI3K/AKT in CIH-induced PASMCs. The inhibitory effects of PLOD2 silencing on PASMC proliferation and migration were accelerated by 2-DG (an inhibitor of glycolysis) and were reversed by lactate (the end product of glycolysis). In addition, the inhibitory effects of PLOD2 silencing on PASMC proliferation, migration and glycolysis were accelerated by PI3K/AKT inhibitor LY294002 and were reversed by the agonist 740Y-P.</div></div><div><h3>Conclusions</h3><div>Silencing of PLOD2 inhibits PI3K/AKT signaling to limit PASMC glycolysis which allows PASMC proliferation and migration in CIH-induced pulmonary arterial hypertension (PAH).</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"446 1","pages":"Article 114453"},"PeriodicalIF":3.3,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143437791","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MYG1 interacts with HSP90 to promote breast cancer progression through Wnt/β-catenin and Notch signaling pathways.
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-14 DOI: 10.1016/j.yexcr.2025.114448
Xuming Liu, Yurong Zhu, Wenqing Huang, Jianxiong Chen, Jiawen Lan, Xiaoli Long, Jun Zhou

Background: As an evolutionarily conserved gene involved in embryonic development, cell differentiation, and immune metabolism, MYG1 exhibits a dynamic expression pattern related to development in human and mouse embryonic tissues, especially upregulates in undifferentiated or pluripotent stem cells. However, MYG1 has been poorly studied in breast cancer and its functional mechanism still remains unclear.

Method: Immunohistochemistry and immunofluorescence were used to study MYG1 expression and localization in breast cancer. Lentivirus transfection combined with CCK8, colony formation, matrix gel experiment and breast fat pad tumor formation in nude mice were used for in vivo and in vitro functional assessment. GSEA enrichment analysis, immunofluorescence and western blot were conducted to explore functional mechanism.

Result: MYG1 expression was upregulated in breast cancer and its higher expression correlated with a variety of clinicopathological characteristics indicating poor prognosis. In vitro and in vivo experiments showed that overexpression of MYG1 promoted breast cancer cells proliferation, migration, invasion and tumorigenesis, while downregulation of MYG1 had an opposite effect. Mechanistically, MYG1 interacted with HSP90 to significantly activate Wnt/β-catenin and Notch signaling pathways in breast cancer cells, thus promoting EMT, cell cycle process and breast cancer progression.

Conclusion: MYG1 is highly expressed in breast cancer and functions as an oncogene. Mechanistically, MYG1 interacts with HSP90 to accelerate EMT and cell cycle process by activating both Wnt/β-catenin and Notch signaling pathways.

{"title":"MYG1 interacts with HSP90 to promote breast cancer progression through Wnt/β-catenin and Notch signaling pathways.","authors":"Xuming Liu, Yurong Zhu, Wenqing Huang, Jianxiong Chen, Jiawen Lan, Xiaoli Long, Jun Zhou","doi":"10.1016/j.yexcr.2025.114448","DOIUrl":"https://doi.org/10.1016/j.yexcr.2025.114448","url":null,"abstract":"<p><strong>Background: </strong>As an evolutionarily conserved gene involved in embryonic development, cell differentiation, and immune metabolism, MYG1 exhibits a dynamic expression pattern related to development in human and mouse embryonic tissues, especially upregulates in undifferentiated or pluripotent stem cells. However, MYG1 has been poorly studied in breast cancer and its functional mechanism still remains unclear.</p><p><strong>Method: </strong>Immunohistochemistry and immunofluorescence were used to study MYG1 expression and localization in breast cancer. Lentivirus transfection combined with CCK8, colony formation, matrix gel experiment and breast fat pad tumor formation in nude mice were used for in vivo and in vitro functional assessment. GSEA enrichment analysis, immunofluorescence and western blot were conducted to explore functional mechanism.</p><p><strong>Result: </strong>MYG1 expression was upregulated in breast cancer and its higher expression correlated with a variety of clinicopathological characteristics indicating poor prognosis. In vitro and in vivo experiments showed that overexpression of MYG1 promoted breast cancer cells proliferation, migration, invasion and tumorigenesis, while downregulation of MYG1 had an opposite effect. Mechanistically, MYG1 interacted with HSP90 to significantly activate Wnt/β-catenin and Notch signaling pathways in breast cancer cells, thus promoting EMT, cell cycle process and breast cancer progression.</p><p><strong>Conclusion: </strong>MYG1 is highly expressed in breast cancer and functions as an oncogene. Mechanistically, MYG1 interacts with HSP90 to accelerate EMT and cell cycle process by activating both Wnt/β-catenin and Notch signaling pathways.</p>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":" ","pages":"114448"},"PeriodicalIF":3.3,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143432702","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell death-associated lncRNAs in cancer immunopathogenesis: An exploration of molecular mechanisms and signaling pathways
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-11 DOI: 10.1016/j.yexcr.2025.114439
Safia Obaidur Rab , Ahmed Hussein Zwamel , Enwa Felix Oghenemaro , Muktesh Chandra , Irwanjot Kaur , Bindu Rani , Vikrant Abbot , M. Ravi Kumar , Muhammad Ikram Ullah , Abhinav Kumar
Cancer remains one of the foremost causes of mortality worldwide, highlighting the urgent need for novel therapeutic targets due to the insufficient efficacy and adverse side effects associated with existing cancer treatments. Long non-coding RNAs (lncRNAs), defined as RNA transcripts longer than 200 nucleotides, have emerged as pivotal regulators in the initiation and progression of various malignancies. In oncology, programmed cell death (PCD) serves as the primary mechanism for tumor cell elimination, comprising processes such as apoptosis, pyroptosis, autophagy, and ferroptosis. Recent studies have elucidated a substantial relationship between lncRNAs and these PCD pathways, indicating that lncRNAs can modulate the apoptotic and non-apoptotic death mechanisms. This regulation may influence not only the dynamics of cancer progression but also the therapeutic response to clinical interventions. This review delves into the intricate role of lncRNAs within the context of PCD in cancer, unveiling the underlying pathogenic mechanisms while proposing innovative strategies for cancer therapy. Additionally, it discusses the potential therapeutic implications of targeting lncRNAs in PCD and related signaling pathways, aiming to enhance treatment outcomes for patients facing cancer.
{"title":"Cell death-associated lncRNAs in cancer immunopathogenesis: An exploration of molecular mechanisms and signaling pathways","authors":"Safia Obaidur Rab ,&nbsp;Ahmed Hussein Zwamel ,&nbsp;Enwa Felix Oghenemaro ,&nbsp;Muktesh Chandra ,&nbsp;Irwanjot Kaur ,&nbsp;Bindu Rani ,&nbsp;Vikrant Abbot ,&nbsp;M. Ravi Kumar ,&nbsp;Muhammad Ikram Ullah ,&nbsp;Abhinav Kumar","doi":"10.1016/j.yexcr.2025.114439","DOIUrl":"10.1016/j.yexcr.2025.114439","url":null,"abstract":"<div><div>Cancer remains one of the foremost causes of mortality worldwide, highlighting the urgent need for novel therapeutic targets due to the insufficient efficacy and adverse side effects associated with existing cancer treatments. Long non-coding RNAs (lncRNAs), defined as RNA transcripts longer than 200 nucleotides, have emerged as pivotal regulators in the initiation and progression of various malignancies. In oncology, programmed cell death (PCD) serves as the primary mechanism for tumor cell elimination, comprising processes such as apoptosis, pyroptosis, autophagy, and ferroptosis. Recent studies have elucidated a substantial relationship between lncRNAs and these PCD pathways, indicating that lncRNAs can modulate the apoptotic and non-apoptotic death mechanisms. This regulation may influence not only the dynamics of cancer progression but also the therapeutic response to clinical interventions. This review delves into the intricate role of lncRNAs within the context of PCD in cancer, unveiling the underlying pathogenic mechanisms while proposing innovative strategies for cancer therapy. Additionally, it discusses the potential therapeutic implications of targeting lncRNAs in PCD and related signaling pathways, aiming to enhance treatment outcomes for patients facing cancer.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"446 1","pages":"Article 114439"},"PeriodicalIF":3.3,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413803","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bee venom: Yesterday's enemy becomes modern medicine for skin cancer
IF 3.3 3区 生物学 Q3 CELL BIOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.yexcr.2025.114435
Amr Ahmed El-Arabey , Hamed A. Ghramh
Malignant melanoma is one of the most lethal human malignancies, particularly when it spreads from its initial site in the skin to distant locations with few therapeutic options. While a range of treatment approaches exist, such as chemotherapy, radiation, immunotherapy, and targeted therapy, they typically fail to treat skin cancer, particularly in its late stages. The complex cellular and molecular mechanisms that drive melanoma growth and metastatic dissemination are both varied and complicated, posing significant challenges to the development of effective treatment approaches. As the incidence and burden of this malignancy increase, there is an urgent need for innovative therapeutic techniques. Therefore, it is vital to research alternate therapy options. Several research undertaken in recent years have found that bee venom influences a variety of cancers. The more research into using bee venom to cure skin cancer, the less attention it receives. In this context, the purpose of this proposal is to review a comprehensive assessment of the clinical impact of bee venom against skin cancer, as well as to highlight challenges and excitement down the road.
{"title":"Bee venom: Yesterday's enemy becomes modern medicine for skin cancer","authors":"Amr Ahmed El-Arabey ,&nbsp;Hamed A. Ghramh","doi":"10.1016/j.yexcr.2025.114435","DOIUrl":"10.1016/j.yexcr.2025.114435","url":null,"abstract":"<div><div>Malignant melanoma is one of the most lethal human malignancies, particularly when it spreads from its initial site in the skin to distant locations with few therapeutic options. While a range of treatment approaches exist, such as chemotherapy, radiation, immunotherapy, and targeted therapy, they typically fail to treat skin cancer, particularly in its late stages. The complex cellular and molecular mechanisms that drive melanoma growth and metastatic dissemination are both varied and complicated, posing significant challenges to the development of effective treatment approaches. As the incidence and burden of this malignancy increase, there is an urgent need for innovative therapeutic techniques. Therefore, it is vital to research alternate therapy options. Several research undertaken in recent years have found that bee venom influences a variety of cancers. The more research into using bee venom to cure skin cancer, the less attention it receives. In this context, the purpose of this proposal is to review a comprehensive assessment of the clinical impact of bee venom against skin cancer, as well as to highlight challenges and excitement down the road.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"445 2","pages":"Article 114435"},"PeriodicalIF":3.3,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143372444","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","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
Experimental cell research
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