Pub Date : 2026-01-09DOI: 10.1016/j.yexcr.2026.114887
Ashesh Sharma , Songon An
ATP-dependent phosphofructokinase (PFK) catalyzes a key committed step in glycolysis. Also called the “gatekeeper” of glycolysis, PFKs catalyze an irreversible phosphorylation of fructose-6-phosphate to fructose-1,6-bisphosphate. While eukaryotic PFKs are more than double the size of prokaryotic PFKs, the overall structure and mechanism of PFKs are largely conserved from prokaryotes to eukaryotes. Usually tetrameric to be active, PFKs are also found to form higher-order structures, such as octamers in yeasts and multimeric filaments in mammals. Due to their central role in glycolysis, the enzymatic activity of PFKs is highly regulated by a myriad of allosteric effectors, post-translational modifications, and various signaling pathways. Such complex regulatory networks of PFKs often result from and/or result in a variety of protein-protein interactions, thus providing the molecular basis of dynamic association and dissociation of PFKs into multienzyme metabolic assemblies (i.e., metabolons) in cells. The concept of metabolon has now become an organizational principle for a cell to regulate the function of PFKs and thus govern glycolysis. Therefore, this article aims to integrate current knowledge in PFK enzymology and cell biology to provide a molecular and cellular framework for repositioning this century-old drug target for innovative therapeutic applications.
{"title":"Phosphofructokinase in glycolysis: Bridging enzymology and cell biology","authors":"Ashesh Sharma , Songon An","doi":"10.1016/j.yexcr.2026.114887","DOIUrl":"10.1016/j.yexcr.2026.114887","url":null,"abstract":"<div><div>ATP-dependent phosphofructokinase (PFK) catalyzes a key committed step in glycolysis. Also called the “gatekeeper” of glycolysis, PFKs catalyze an irreversible phosphorylation of fructose-6-phosphate to fructose-1,6-bisphosphate. While eukaryotic PFKs are more than double the size of prokaryotic PFKs, the overall structure and mechanism of PFKs are largely conserved from prokaryotes to eukaryotes. Usually tetrameric to be active, PFKs are also found to form higher-order structures, such as octamers in yeasts and multimeric filaments in mammals. Due to their central role in glycolysis, the enzymatic activity of PFKs is highly regulated by a myriad of allosteric effectors, post-translational modifications, and various signaling pathways. Such complex regulatory networks of PFKs often result from and/or result in a variety of protein-protein interactions, thus providing the molecular basis of dynamic association and dissociation of PFKs into multienzyme metabolic assemblies (i.e., metabolons) in cells. The concept of metabolon has now become an organizational principle for a cell to regulate the function of PFKs and thus govern glycolysis. Therefore, this article aims to integrate current knowledge in PFK enzymology and cell biology to provide a molecular and cellular framework for repositioning this century-old drug target for innovative therapeutic applications.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"456 1","pages":"Article 114887"},"PeriodicalIF":3.5,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951769","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}
Pub Date : 2026-01-07DOI: 10.1016/j.yexcr.2025.114875
Ruman Chen , Jiqing He , Na An, Mingzhi Xu, Liheng Wang, Yafei Bai
Background
Vascular calcification (VC) is a severe cardiovascular complication of chronic kidney disease (CKD), driven by vascular smooth muscle cell (VSMC) osteogenic trans-differentiation and exacerbated by oxidative stress and cellular dysfunction. Despite its clinical relevance, the molecular mechanisms underlying CKD-associated VC remain incompletely understood. This study investigates the role of Sirtuin 1 (SIRT1) in modulating VC through ferroptosis inhibition and mitophagy activation and examines whether microRNA-181c-5p (miR-181c-5p) contributes to SIRT1 dysregulation in this context.
Methods
A CKD-associated VC model was induced in rats by 5/6 nephrectomy followed by high calcium/phosphate and calcitriol loading, and an in vitro calcification model was established in primary rat VSMCs. SIRT1 was manipulated using AAV9-mediated overexpression in vivo and plasmid overexpression or inhibition in vitro. Upstream regulation of SIRT1 by miR-181c-5p was predicted bioinformatically and validated by RNA pull-down and dual-luciferase assays. Ferroptosis was assessed by redox and Fe2+ indices, and mitophagy by Pink1/Parkin, LC3-II and p62 expression. Rescue experiments employed erastin, Mdivi-1 and Parkin knockdown.
Results
SIRT1 expression was markedly reduced in calcified aortic tissues and VSMCs. SIRT1 overexpression suppressed VC by reducing calcium deposition, downregulating osteogenic markers, and increasing fetuin-A levels. SIRT1 also suppressed ferroptosis by restoring the GSH/GPX4/SLC7A11 axis and limiting ROS and lipid peroxidation, whereas erastin abolished these effects. Mechanistically, miR-181c-5p was found to directly target SIRT1 and promote VSMC calcification by repressing SIRT1. Moreover, SIRT1 promoted mitophagy via the Pink1/Parkin pathway activation. Furthermore, inhibition of mitophagy reversed the anti-ferroptotic effects of SIRT1, confirming their functional interplay.
Conclusions
SIRT1, negatively regulated by miR-181c-5p, mitigates CKD-associated VC by suppressing ferroptosis and activating Pink1/Parkin-dependent mitophagy in VSMCs, highlighting a potential therapeutic axis for vascular protection in CKD.
{"title":"MiR-181c-5p-SIRT1 axis-driven Pink1/Parkin-mediated mitophagy prevents ferroptosis and vascular calcification in chronic kidney disease","authors":"Ruman Chen , Jiqing He , Na An, Mingzhi Xu, Liheng Wang, Yafei Bai","doi":"10.1016/j.yexcr.2025.114875","DOIUrl":"10.1016/j.yexcr.2025.114875","url":null,"abstract":"<div><h3>Background</h3><div>Vascular calcification (VC) is a severe cardiovascular complication of chronic kidney disease (CKD), driven by vascular smooth muscle cell (VSMC) osteogenic trans-differentiation and exacerbated by oxidative stress and cellular dysfunction. Despite its clinical relevance, the molecular mechanisms underlying CKD-associated VC remain incompletely understood. This study investigates the role of Sirtuin 1 (SIRT1) in modulating VC through ferroptosis inhibition and mitophagy activation and examines whether microRNA-181c-5p (miR-181c-5p) contributes to SIRT1 dysregulation in this context.</div></div><div><h3>Methods</h3><div>A CKD-associated VC model was induced in rats by 5/6 nephrectomy followed by high calcium/phosphate and calcitriol loading, and an <em>in vitro</em> calcification model was established in primary rat VSMCs. SIRT1 was manipulated using AAV9-mediated overexpression <em>in vivo</em> and plasmid overexpression or inhibition <em>in vitro</em>. Upstream regulation of SIRT1 by miR-181c-5p was predicted bioinformatically and validated by RNA pull-down and dual-luciferase assays. Ferroptosis was assessed by redox and Fe<sup>2+</sup> indices, and mitophagy by Pink1/Parkin, LC3-II and p62 expression. Rescue experiments employed erastin, Mdivi-1 and Parkin knockdown.</div></div><div><h3>Results</h3><div>SIRT1 expression was markedly reduced in calcified aortic tissues and VSMCs. SIRT1 overexpression suppressed VC by reducing calcium deposition, downregulating osteogenic markers, and increasing fetuin-A levels. SIRT1 also suppressed ferroptosis by restoring the GSH/GPX4/SLC7A11 axis and limiting ROS and lipid peroxidation, whereas erastin abolished these effects. Mechanistically, miR-181c-5p was found to directly target SIRT1 and promote VSMC calcification by repressing SIRT1. Moreover, SIRT1 promoted mitophagy via the Pink1/Parkin pathway activation. Furthermore, inhibition of mitophagy reversed the anti-ferroptotic effects of SIRT1, confirming their functional interplay.</div></div><div><h3>Conclusions</h3><div>SIRT1, negatively regulated by miR-181c-5p, mitigates CKD-associated VC by suppressing ferroptosis and activating Pink1/Parkin-dependent mitophagy in VSMCs, highlighting a potential therapeutic axis for vascular protection in CKD.</div></div><div><h3>Clinical trial registration number</h3><div>Not applicable.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"456 1","pages":"Article 114875"},"PeriodicalIF":3.5,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942971","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}
Pub Date : 2026-01-07DOI: 10.1016/j.yexcr.2026.114888
Reuben J. Hoffmann , AeSoon Bensen , Mark Dane, Jane Arterberry, Rebecca Smith, James Korkola, Pepper Schedin
COX-2, an inducible enzyme key to production of inflammatory prostaglandins, has tumor cell-intrinsic oncogenic activity. Previously, we reported Cys-526-nitrosylated COX-2 (SNO-COX-2) associates with breast cancer progression and poor-prognostic young onset breast cancer. Here, using a 3D culture model of early-stage human breast cancer (MCF10DCIS cells), we report SNO-COX-2, but not non-nitrosylated COX-2, closely associated with mesenchymal cell phenotypes induced by fibrillar Col1. Inhibition of nitric oxide synthase (NOS) activity did not reduce SNO-COX-2 levels, suggesting alternative nitrosylation mechanisms. In 3D MCF10DCIS culture, mesenchymal phenotypes and SNO-COX-2 protein induced by Col1 did not associate with transcription of classic epithelial-to-mesenchymal transition (EMT) markers nor common cancer signaling pathways. Conversely, TGFβ-1 strongly induced EMT- and cancer signaling-related transcripts but was insufficient to increase SNO-COX-2 protein or mesenchymal phenotypes. These data suggest the mesenchymal phenotype and SNO-COX-2 expression in MCF10DCIS are driven by a non-transcriptional mechanism dependent on Col1. We tested 300 additional microenvironmental conditions and find SNO-COX-2 expression is driven by inflammatory, wound-resolving, and cancer-associated TME factors, including TNC, SPP1, decorin, Col1, Col3, INF-γ, and IL-4/13, with specific extracellular matrix-ligand combinations driving both high and low SNO-COX-2 expression. In sum, these observations show that in MCF10DCIS cells, SNO-COX-2 associates with mesenchymal phenotypes more strongly than non-nitrosylated COX-2; expression of classic EMT transcripts is neither sufficient nor necessary for acquisition of mesenchymal phenotypes; and expression of SNO-COX-2 is highly microenvironment-dependent. Future studies evaluating SNO-COX-2 as a biomarker for early-stage breast cancer with increased risk for progression, and its regulation, are warranted.
{"title":"S-nitrosylated COX-2 is a microenvironment-regulated breast cancer cell biomarker of mesenchymal phenotypes","authors":"Reuben J. Hoffmann , AeSoon Bensen , Mark Dane, Jane Arterberry, Rebecca Smith, James Korkola, Pepper Schedin","doi":"10.1016/j.yexcr.2026.114888","DOIUrl":"10.1016/j.yexcr.2026.114888","url":null,"abstract":"<div><div>COX-2, an inducible enzyme key to production of inflammatory prostaglandins, has tumor cell-intrinsic oncogenic activity. Previously, we reported Cys-526-nitrosylated COX-2 (SNO-COX-2) associates with breast cancer progression and poor-prognostic young onset breast cancer. Here, using a 3D culture model of early-stage human breast cancer (MCF10DCIS cells), we report SNO-COX-2, but not non-nitrosylated COX-2, closely associated with mesenchymal cell phenotypes induced by fibrillar Col1. Inhibition of nitric oxide synthase (NOS) activity did not reduce SNO-COX-2 levels, suggesting alternative nitrosylation mechanisms. In 3D MCF10DCIS culture, mesenchymal phenotypes and SNO-COX-2 protein induced by Col1 did not associate with transcription of classic epithelial-to-mesenchymal transition (EMT) markers nor common cancer signaling pathways. Conversely, TGFβ-1 strongly induced EMT- and cancer signaling-related transcripts but was insufficient to increase SNO-COX-2 protein or mesenchymal phenotypes. These data suggest the mesenchymal phenotype and SNO-COX-2 expression in MCF10DCIS are driven by a non-transcriptional mechanism dependent on Col1. We tested 300 additional microenvironmental conditions and find SNO-COX-2 expression is driven by inflammatory, wound-resolving, and cancer-associated TME factors, including TNC, SPP1, decorin, Col1, Col3, INF-γ, and IL-4/13, with specific extracellular matrix-ligand combinations driving both high and low SNO-COX-2 expression. In sum, these observations show that in MCF10DCIS cells, SNO-COX-2 associates with mesenchymal phenotypes more strongly than non-nitrosylated COX-2; expression of classic EMT transcripts is neither sufficient nor necessary for acquisition of mesenchymal phenotypes; and expression of SNO-COX-2 is highly microenvironment-dependent. Future studies evaluating SNO-COX-2 as a biomarker for early-stage breast cancer with increased risk for progression, and its regulation, are warranted.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"456 1","pages":"Article 114888"},"PeriodicalIF":3.5,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942969","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}
Pub Date : 2026-01-06DOI: 10.1016/j.yexcr.2026.114886
Shi Chen , Yue Wang , Jingwen Gao , Jiaxi Lin , Lu Liu , Shiqi Zhu , Lihe Liu , Chunfang Xu , Xiaolin Liu , Manhui Zhu , Jinzhou Zhu
Hepatocyte pyroptosis critically contributes to metabolism-associated fatty liver disease (MAFLD) progression. Fetuin-B (FETUB), a hepatocytokine, promotes pyroptosis by downregulating adiponectin receptor 1 (AdipoR1), thereby activating the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome/GSDMD pathway. This study investigated the role of FETUB in metabolic dysfunction-associated steatohepatitis (MASH) and the therapeutic efficacy of FETUB inhibition. In primary mouse hepatocytes, free fatty acid (FFA) stimulation upregulated FETUB transcription, expression, and secretion, which suppressed membrane AdipoR1 and triggered NLRP3/GSDMD-mediated pyroptosis, exacerbating steatosis. In high-fat diet (HFD)-induced MASH mice, hepatic FETUB expression increased concordantly with AdipoR1 downregulation. FETUB blockade ameliorated hepatic steatosis, inflammation, ballooning, and fibrosis by disrupting this pathway. These findings establish FETUB as a key regulator of NLRP3/GSDMD-driven pyroptosis in MASH and identify it as a promising therapeutic target.
{"title":"Fetuin B drives metabolism-associated steatohepatitis by promoting hepatocyte pyroptosis via NLRP3/GSDMD pathway","authors":"Shi Chen , Yue Wang , Jingwen Gao , Jiaxi Lin , Lu Liu , Shiqi Zhu , Lihe Liu , Chunfang Xu , Xiaolin Liu , Manhui Zhu , Jinzhou Zhu","doi":"10.1016/j.yexcr.2026.114886","DOIUrl":"10.1016/j.yexcr.2026.114886","url":null,"abstract":"<div><div>Hepatocyte pyroptosis critically contributes to metabolism-associated fatty liver disease (MAFLD) progression. Fetuin-B (FETUB), a hepatocytokine, promotes pyroptosis by downregulating adiponectin receptor 1 (AdipoR1), thereby activating the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome/GSDMD pathway. This study investigated the role of FETUB in metabolic dysfunction-associated steatohepatitis (MASH) and the therapeutic efficacy of FETUB inhibition. In primary mouse hepatocytes, free fatty acid (FFA) stimulation upregulated FETUB transcription, expression, and secretion, which suppressed membrane AdipoR1 and triggered NLRP3/GSDMD-mediated pyroptosis, exacerbating steatosis. In high-fat diet (HFD)-induced MASH mice, hepatic FETUB expression increased concordantly with AdipoR1 downregulation. FETUB blockade ameliorated hepatic steatosis, inflammation, ballooning, and fibrosis by disrupting this pathway. These findings establish FETUB as a key regulator of NLRP3/GSDMD-driven pyroptosis in MASH and identify it as a promising therapeutic target.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"456 1","pages":"Article 114886"},"PeriodicalIF":3.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145915416","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}
Pub Date : 2026-01-03DOI: 10.1016/j.yexcr.2026.114885
Kangjie He , Chunxiao Lin , Hengyou Wang , Hongshen Wu , Shanfeng Yin , Chang Tao , Xiang Yan
Bladder cancer is a common malignant tumor of the urinary system, with its malignant progression mechanisms remaining unclear. In this study, a circular RNA (hsa_circ_0040457, hereinafter referred to as "circ-GLG1″), which has been less characterized in bladder cancer, was identified. It is significantly overexpressed in bladder cancer tissues and cells, and closely associated with patients' pTNM stage and poor prognosis. Circ-GLG1 is formed by back-splicing of exons 23–26 of the GLG1 gene, with circular stability and IRES-dependent translational potential. Functional studies confirmed that silencing circ-GLG1 inhibits cellular malignant phenotypes and tumor formation in nude mice, while overexpression promotes progression by activating TGF-β, PI3K-AKT, and MAPK pathways. Mechanistically, transcriptome sequencing identified 555 differentially expressed genes, with GO/KEGG enrichment in cancer-related pathways. Combined with database prediction, KCNJ9 was identified as a key target. Luciferase assays confirmed that circ-GLG1 may sponge miR-346, relieving its inhibitory effect on KCNJ9's 3′UTR. Functional rescue experiments showed that KCNJ9 overexpression reversed the phenotypic and pathway inactivation induced by circ-GLG1 silencing, verifying the "circ-GLG1/miR-346/KCNJ9″ regulatory axis. This study reveals that circ-GLG1 acts as a ceRNA to target KCNJ9, relieving miR-346-mediated post-transcriptional inhibition, with concurrent activation of TGF-β, PI3K-AKT, and MAPK pathways via KCNJ9 that are associated with bladder cancer progression.
{"title":"The circ-GLG1/miR-346/KCNJ9 axis drives malignant progression of bladder cancer by modulating KCNJ9 expression","authors":"Kangjie He , Chunxiao Lin , Hengyou Wang , Hongshen Wu , Shanfeng Yin , Chang Tao , Xiang Yan","doi":"10.1016/j.yexcr.2026.114885","DOIUrl":"10.1016/j.yexcr.2026.114885","url":null,"abstract":"<div><div>Bladder cancer is a common malignant tumor of the urinary system, with its malignant progression mechanisms remaining unclear. In this study, a circular RNA (hsa_circ_0040457, hereinafter referred to as \"circ-GLG1″), which has been less characterized in bladder cancer, was identified. It is significantly overexpressed in bladder cancer tissues and cells, and closely associated with patients' pTNM stage and poor prognosis. Circ-GLG1 is formed by back-splicing of exons 23–26 of the GLG1 gene, with circular stability and IRES-dependent translational potential. Functional studies confirmed that silencing circ-GLG1 inhibits cellular malignant phenotypes and tumor formation in nude mice, while overexpression promotes progression by activating TGF-β, PI3K-AKT, and MAPK pathways. Mechanistically, transcriptome sequencing identified 555 differentially expressed genes, with GO/KEGG enrichment in cancer-related pathways. Combined with database prediction, KCNJ9 was identified as a key target. Luciferase assays confirmed that circ-GLG1 may sponge miR-346, relieving its inhibitory effect on KCNJ9's 3′UTR. Functional rescue experiments showed that KCNJ9 overexpression reversed the phenotypic and pathway inactivation induced by circ-GLG1 silencing, verifying the \"circ-GLG1/miR-346/KCNJ9″ regulatory axis. This study reveals that circ-GLG1 acts as a ceRNA to target KCNJ9, relieving miR-346-mediated post-transcriptional inhibition, with concurrent activation of TGF-β, PI3K-AKT, and MAPK pathways via KCNJ9 that are associated with bladder cancer progression.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"455 2","pages":"Article 114885"},"PeriodicalIF":3.5,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145905790","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}
Pub Date : 2026-01-02DOI: 10.1016/j.yexcr.2025.114884
Yini Wang , Hongquan Liu , Tianqi Wang , Yingying Yang , Xiaohong Ma , Jitao Wu
Objective
Investigate the function of glutathione reductase-associated protein 5 (GLRX5) and its prognostic significance, as well as its association with CAFs and the TME.
Methods
Based on data from TCGA and the GEO databases, this study investigates the expression of GLRX5 in BLCA and its association with clinical outcomes. Using the ssGSEA algorithm, we explored the association between functional features of GLRX5 and BLCA. We validated our findings using in vitro cellular functional assays. We explored the regulatory mechanisms associated with GLRX5 using GO, KEGG, and GSEA. Furthermore, based on single-cell and spatial transcriptomics data from bladder cancer, we analyzed the expression patterns and potential functions of GLRX5 in bladder cancer. The results of the above analyses were experimentally explored and validated.
Results
High expression of GLRX5 in BLCA correlates with malignant biological behavior and poor prognosis. Enrichment analysis indicates that GLRX5 is primarily associated with malignant functional characteristics in bladder cancer, and its expression levels are also linked to EMT, OXPHOS, and FAM. The above analyses have all been validated through in vitro experiments. Single-cell and spatial transcriptomics analyses indicate that GLRX5 is also expressed in CAFs and participates in metabolic pathways. Experiments demonstrated that GLRX5 promotes the activation of CAFs and may enhance tumor cell migration by influencing pathways within the TME.
Conclusion
High expression of GLRX5 in tumor cells promotes malignant biological behavior and predicts poor prognosis. At the same time, high expression of GLRX5 in CAFs promotes tumor cell migration by affecting the TME.
{"title":"GLRX5 is a prognostic marker in bladder cancer and correlates with activation of cancer-associated fibroblasts in the tumor microenvironment","authors":"Yini Wang , Hongquan Liu , Tianqi Wang , Yingying Yang , Xiaohong Ma , Jitao Wu","doi":"10.1016/j.yexcr.2025.114884","DOIUrl":"10.1016/j.yexcr.2025.114884","url":null,"abstract":"<div><h3>Objective</h3><div>Investigate the function of glutathione reductase-associated protein 5 (GLRX5) and its prognostic significance, as well as its association with CAFs and the TME.</div></div><div><h3>Methods</h3><div>Based on data from TCGA and the GEO databases, this study investigates the expression of GLRX5 in BLCA and its association with clinical outcomes. Using the ssGSEA algorithm, we explored the association between functional features of GLRX5 and BLCA. We validated our findings using in vitro cellular functional assays. We explored the regulatory mechanisms associated with GLRX5 using GO, KEGG, and GSEA. Furthermore, based on single-cell and spatial transcriptomics data from bladder cancer, we analyzed the expression patterns and potential functions of GLRX5 in bladder cancer. The results of the above analyses were experimentally explored and validated.</div></div><div><h3>Results</h3><div>High expression of GLRX5 in BLCA correlates with malignant biological behavior and poor prognosis. Enrichment analysis indicates that GLRX5 is primarily associated with malignant functional characteristics in bladder cancer, and its expression levels are also linked to EMT, OXPHOS, and FAM. The above analyses have all been validated through in vitro experiments. Single-cell and spatial transcriptomics analyses indicate that GLRX5 is also expressed in CAFs and participates in metabolic pathways. Experiments demonstrated that GLRX5 promotes the activation of CAFs and may enhance tumor cell migration by influencing pathways within the TME.</div></div><div><h3>Conclusion</h3><div>High expression of GLRX5 in tumor cells promotes malignant biological behavior and predicts poor prognosis. At the same time, high expression of GLRX5 in CAFs promotes tumor cell migration by affecting the TME.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"455 2","pages":"Article 114884"},"PeriodicalIF":3.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145899569","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}
Pub Date : 2025-12-29DOI: 10.1016/j.yexcr.2025.114876
Hatila Tuerxun , JinQiu Li , Qian Liu , Mikrban Tursun , Jin BaoXiao , Chengwei Wang , Ayshamgul Hasim
The immune status is of crucial importance in the development of cervical cancer (CC). MICA/B, as a major histocompatibility complex Class I associated protein, mediates anti-tumor immunity by activating NK cell receptors. However, the precise mechanisms underlying MICA/B-mediated regulation of CC progression remain poorly understood. This study combined spatial transcriptome sequencing and bioinformatics analysis and found that MICA/B was significantly highly expressed in CC tissues and cells, accompanied by more NK cell infiltration. Flow cytometry and Cell Functional assays, Knockdown of MICA/MICB weakens the activation receptor efficacy of NK cells, enhances the inhibitory signal, leads to a decrease in cytotoxicity, and simultaneously upregulates Cyclin expression in CC cells while downregulating BCL-2/BAX. Tumor xenograft models indicated that tumors with MICA knockdown exhibited a growth tendency in the presence of natural killer (NK) cells. Mechanistically, MICA/B regulates inflammatory factors such as IL-6 and CXCL10/11 through the Toll-like signaling pathway, affecting the function of NK cells. Thus, MICA/B expression on cervical cancer cells plays a pivotal role in eliciting NK cell-mediated antitumor immunity. Their downregulation attenuates NK cell function, promoting cervical cancer cell proliferation and survival via the Toll signaling pathway. These findings highlight the potential of targeting MICA/B-NK cell interactions as a therapeutic strategy for cervical cancer.
{"title":"MICA/B-driven NK cell dysfunction promotes cervical cancer via Toll signaling","authors":"Hatila Tuerxun , JinQiu Li , Qian Liu , Mikrban Tursun , Jin BaoXiao , Chengwei Wang , Ayshamgul Hasim","doi":"10.1016/j.yexcr.2025.114876","DOIUrl":"10.1016/j.yexcr.2025.114876","url":null,"abstract":"<div><div>The immune status is of crucial importance in the development of cervical cancer (CC). MICA/B, as a major histocompatibility complex Class I associated protein, mediates anti-tumor immunity by activating NK cell receptors. However, the precise mechanisms underlying MICA/B-mediated regulation of CC progression remain poorly understood. This study combined spatial transcriptome sequencing and bioinformatics analysis and found that MICA/B was significantly highly expressed in CC tissues and cells, accompanied by more NK cell infiltration. Flow cytometry and Cell Functional assays, Knockdown of MICA/MICB weakens the activation receptor efficacy of NK cells, enhances the inhibitory signal, leads to a decrease in cytotoxicity, and simultaneously upregulates Cyclin expression in CC cells while downregulating BCL-2/BAX. Tumor xenograft models indicated that tumors with MICA knockdown exhibited a growth tendency in the presence of natural killer (NK) cells. Mechanistically, MICA/B regulates inflammatory factors such as IL-6 and CXCL10/11 through the Toll-like signaling pathway, affecting the function of NK cells. Thus, MICA/B expression on cervical cancer cells plays a pivotal role in eliciting NK cell-mediated antitumor immunity. Their downregulation attenuates NK cell function, promoting cervical cancer cell proliferation and survival via the Toll signaling pathway. These findings highlight the potential of targeting MICA/B-NK cell interactions as a therapeutic strategy for cervical cancer.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"455 2","pages":"Article 114876"},"PeriodicalIF":3.5,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145877978","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}
Pub Date : 2025-12-27DOI: 10.1016/j.yexcr.2025.114883
Yansong Xu , Yuansong Sun , Chunlin Yin , Fei Xie , He Li
Acute pancreatitis (AP) exhibits marked clinical heterogeneity. To investigate the molecular mechanisms involved in AP, we integrated bioinformatics analysis of public sequencing datasets, which identified differentially expressed genes (DEGs) significantly associated with AP. Subsequently, GO/KEGG enrichment analyses revealed robust involvement of these DEGs in cellular adhesion and MAPK signaling pathways. Protein-protein interaction (PPI) network analysis pinpointed integrin β1 (ITGB1) as the central hub gene, while single-gene gene set enrichment analysis (GSEA) across ontological databases confirmed its significant enrichment in pathways associated with adhesion and inflammation. These findings establish ITGB1 as a pivotal regulator coordinating cell adhesion and inflammatory responses in AP. In murine AP models, ITGB1 protein was significantly upregulated in the pancreas and co-localized specifically with macrophages. In vitro studies using bone marrow-derived macrophages (BMDMs) revealed that ITGB1 upregulation enhanced macrophage-endothelial adhesion and inflammatory cascades through p38 MAPK phosphorylation. Critically, clinical translation studies established the dual diagnostic value of ITGB1. The receiver operating characteristic (ROC) curve exhibited significant discriminatory power for distinguishing patients with AP from healthy controls, along with robust efficacy in stratifying disease severity. In conclusion, ITGB1 orchestrates macrophage-mediated inflammation through p38 MAPK-dependent mechanisms and can function as a biomarker for diagnosis and severity stratification in AP.
{"title":"Integrin β1 contributes to acute pancreatitis by mediating macrophage adhesion and inflammatory cascades","authors":"Yansong Xu , Yuansong Sun , Chunlin Yin , Fei Xie , He Li","doi":"10.1016/j.yexcr.2025.114883","DOIUrl":"10.1016/j.yexcr.2025.114883","url":null,"abstract":"<div><div>Acute pancreatitis (AP) exhibits marked clinical heterogeneity. To investigate the molecular mechanisms involved in AP, we integrated bioinformatics analysis of public sequencing datasets, which identified differentially expressed genes (DEGs) significantly associated with AP. Subsequently, GO/KEGG enrichment analyses revealed robust involvement of these DEGs in cellular adhesion and MAPK signaling pathways. Protein-protein interaction (PPI) network analysis pinpointed integrin β1 (ITGB1) as the central hub gene, while single-gene gene set enrichment analysis (GSEA) across ontological databases confirmed its significant enrichment in pathways associated with adhesion and inflammation. These findings establish ITGB1 as a pivotal regulator coordinating cell adhesion and inflammatory responses in AP. In murine AP models, ITGB1 protein was significantly upregulated in the pancreas and co-localized specifically with macrophages. <em>In vitro</em> studies using bone marrow-derived macrophages (BMDMs) revealed that ITGB1 upregulation enhanced macrophage-endothelial adhesion and inflammatory cascades through p38 MAPK phosphorylation. Critically, clinical translation studies established the dual diagnostic value of ITGB1. The receiver operating characteristic (ROC) curve exhibited significant discriminatory power for distinguishing patients with AP from healthy controls, along with robust efficacy in stratifying disease severity. In conclusion, ITGB1 orchestrates macrophage-mediated inflammation through p38 MAPK-dependent mechanisms and can function as a biomarker for diagnosis and severity stratification in AP.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"455 2","pages":"Article 114883"},"PeriodicalIF":3.5,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145855157","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}
Pub Date : 2025-12-25DOI: 10.1016/j.yexcr.2025.114874
Xiumin Xu , Zeping Zuo , Jinhai Zhu , Jun Wu , Tao Zhang
As the most common and aggressive subtype of renal cancer, clear cell renal cell carcinoma (ccRCC) often shows poor responsiveness to current therapeutic strategies. Although GNA15, a G protein alpha subunit, has been associated with the progression of multiple tumor types, its functional significance in ccRCC remains largely undefined. Public datasets were used to profile GNA15 expression across cancers, and its links to prognosis, genomic diversity, stemness, and immune infiltration were analyzed with multiple computational tools. In ccRCC, transcriptomic and protein expression levels were validated using immunofluorescence and western blotting. Functional assays, including colony formation, transwell migration, tumor spheroid formation, and GSEA, were used to investigate the biological role of GNA15. The effects of GNA15 knockdown were assessed in renal cancer cell lines. GNA15 was aberrantly upregulated in multiple cancers and significantly elevated in ccRCC tissues and cell lines. High GNA15 expression correlated with poor overall survival and advanced clinical stage. It was also positively associated with tumor heterogeneity, stemness, and immunosuppressive microenvironment characteristics, particularly M2 macrophage and neutrophil infiltration. GSEA identified enrichment in oncogenic pathways, including JAK-STAT, Wnt, and Notch signaling. In vitro knockdown of GNA15 reduced tumor cell proliferation, migration, spheroid formation, and expression of stemness markers and PD-L1. Our results highlight GNA15 as a novel oncogenic and immune-related contributor to ccRCC progression, supporting its potential as a prognostic biomarker and therapeutic target.
{"title":"GNA15 as a potential prognostic and immunological biomarker in ccRCC based on bioinformatics analysis and experimental verification","authors":"Xiumin Xu , Zeping Zuo , Jinhai Zhu , Jun Wu , Tao Zhang","doi":"10.1016/j.yexcr.2025.114874","DOIUrl":"10.1016/j.yexcr.2025.114874","url":null,"abstract":"<div><div>As the most common and aggressive subtype of renal cancer, clear cell renal cell carcinoma (ccRCC) often shows poor responsiveness to current therapeutic strategies. Although GNA15, a G protein alpha subunit, has been associated with the progression of multiple tumor types, its functional significance in ccRCC remains largely undefined. Public datasets were used to profile GNA15 expression across cancers, and its links to prognosis, genomic diversity, stemness, and immune infiltration were analyzed with multiple computational tools. In ccRCC, transcriptomic and protein expression levels were validated using immunofluorescence and western blotting. Functional assays, including colony formation, transwell migration, tumor spheroid formation, and GSEA, were used to investigate the biological role of GNA15. The effects of GNA15 knockdown were assessed in renal cancer cell lines. GNA15 was aberrantly upregulated in multiple cancers and significantly elevated in ccRCC tissues and cell lines. High GNA15 expression correlated with poor overall survival and advanced clinical stage. It was also positively associated with tumor heterogeneity, stemness, and immunosuppressive microenvironment characteristics, particularly M2 macrophage and neutrophil infiltration. GSEA identified enrichment in oncogenic pathways, including JAK-STAT, Wnt, and Notch signaling. In vitro knockdown of GNA15 reduced tumor cell proliferation, migration, spheroid formation, and expression of stemness markers and PD-L1. Our results highlight GNA15 as a novel oncogenic and immune-related contributor to ccRCC progression, supporting its potential as a prognostic biomarker and therapeutic target.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"455 2","pages":"Article 114874"},"PeriodicalIF":3.5,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145843526","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}