Pub Date : 2025-11-09DOI: 10.1016/j.bbamcr.2025.120085
Elena A. Mikhaleva , Natalia V. Akulenko , Oxana M. Olenkina , Yuri A. Abramov , Sergey A. Lavrov , Thoomas A. Leinsoo , Sofia V. Marfina , Georgij Arapidi , Victoria Shender , Sergei S. Ryazansky , Galina L. Kogan , Vladimir A. Gvozdev
The ribosome-associated αβ heterodimeric ubiquitously expressed NAC protein is involved in protein homeostasis in eukaryotes. We previously reported that the germline-specific α and β subunits (gNACαβ) differ from ubiquitously expressed paralogs by the presence of extended intrinsically disordered regions. The embryonic precursors of the germline (pole cells) express both α and β gNAC subunits. CRISPR/Cas9-mediated knockout of the gNAC α- subunit gene resulted in the death of pole cells progenitors, implicating gNAC as an essential component of the germ plasm responsible for the germline development. Immunofluorescence detection was used to track changes in the expression of α- and β-subunits of gNAC during development. The bright fluorescence of the α-subunit in the germarium strongly decreases during oocyte specification and is replaced by the onset of increased β-subunit fluorescence, especially in the posterior part of mature oocytes, in the germ plasm region, where NACα-subunit fluorescent signal is absent. The visualized switches of gNAC subunits presence, as well as detection of separate cytoplasmic fluorescent puncta for α and β gNAC subunits in a germline cells (elongating spermatids in testis or embryonic pole cells), are explained by transient formation the hybrid NAC heterodimers composed of α or β germline subunit and a ubiquitous partner. The formation of hybrids, demonstrated by mass spectrometry analysis in testes, indicates a programed spatiotemporal functioning of both the ubiquitous and germline-specific NAC paralogs to support the germline-specific proteostasis.
{"title":"Developmental spatiotemporal switching of the germline-specific subunits of heterodimeric NAC protein in Drosophila melanogaster","authors":"Elena A. Mikhaleva , Natalia V. Akulenko , Oxana M. Olenkina , Yuri A. Abramov , Sergey A. Lavrov , Thoomas A. Leinsoo , Sofia V. Marfina , Georgij Arapidi , Victoria Shender , Sergei S. Ryazansky , Galina L. Kogan , Vladimir A. Gvozdev","doi":"10.1016/j.bbamcr.2025.120085","DOIUrl":"10.1016/j.bbamcr.2025.120085","url":null,"abstract":"<div><div>The ribosome-associated αβ heterodimeric ubiquitously expressed NAC protein is involved in protein homeostasis in eukaryotes. We previously reported that the germline-specific α and β subunits (gNACαβ) differ from ubiquitously expressed paralogs by the presence of extended intrinsically disordered regions. The embryonic precursors of the germline (pole cells) express both α and β gNAC subunits. CRISPR/Cas9-mediated knockout of the gNAC α- subunit gene resulted in the death of pole cells progenitors, implicating gNAC as an essential component of the germ plasm responsible for the germline development. Immunofluorescence detection was used to track changes in the expression of α- and β-subunits of gNAC during development. The bright fluorescence of the α-subunit in the germarium strongly decreases during oocyte specification and is replaced by the onset of increased β-subunit fluorescence, especially in the posterior part of mature oocytes, in the germ plasm region, where NACα-subunit fluorescent signal is absent. The visualized switches of gNAC subunits presence, as well as detection of separate cytoplasmic fluorescent puncta for α and β gNAC subunits in a germline cells (elongating spermatids in testis or embryonic pole cells), are explained by transient formation the hybrid NAC heterodimers composed of α or β germline subunit and a ubiquitous partner. The formation of hybrids, demonstrated by mass spectrometry analysis in testes, indicates a programed spatiotemporal functioning of both the ubiquitous and germline-specific NAC paralogs to support the germline-specific proteostasis.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120085"},"PeriodicalIF":3.7,"publicationDate":"2025-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145494456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The rising incidence of cancer creates an urgent need to develop faster methods for early detection. Fourier Transform Infrared Spectroscopy (FTIR) spectroscopy is a promising technique to analyse molecular content in biological samples. FTIR has also been used to study structural changes in extracellular vesicles (EVs), revealing alterations in protein secondary structure, protein-to-lipid ratio, and protein phosphorylation levels. We investigated how changes in glycosylation, which naturally occur during tumor progression, affect the previously mentioned parameters and whether these cellular changes are reflected in EV composition. The WM266–4 melanoma cell line, treated with two glycosylation inhibitors, was used as a model. After treatment, EVs were isolated and ATR-FTIR measurements were performed, followed by a comprehensive analysis of the data. Significant differences in protein and lipid content were observed between EV populations treated with glycosylation inhibitors, with greater changes in EVs compared to cells. Glycosylation inhibitors also increased the level of phosphorylated proteins in EVs. Our findings showed that changes in EVs were more dynamic than at the cellular level cells. The presented analysis shows that subtle molecular changes can significantly impact EV cargo and properties. FTIR is a powerful tool for detecting these changes and may aid future cancer diagnostics.
{"title":"ATR-FTIR spectroscopy as a reliable analytical tool in cancer research: Tracking glycosylation-induced protein and lipid alteration in extracellular vesicles","authors":"Magdalena Wilczak , Andrzej Wróbel , Magdalena Surman , Martyna Durak-Kozica , Ewa Ł. Stępień , Małgorzata Przybyło","doi":"10.1016/j.bbamcr.2025.120082","DOIUrl":"10.1016/j.bbamcr.2025.120082","url":null,"abstract":"<div><div>The rising incidence of cancer creates an urgent need to develop faster methods for early detection. Fourier Transform Infrared Spectroscopy (FTIR) spectroscopy is a promising technique to analyse molecular content in biological samples. FTIR has also been used to study structural changes in extracellular vesicles (EVs), revealing alterations in protein secondary structure, protein-to-lipid ratio, and protein phosphorylation levels. We investigated how changes in glycosylation, which naturally occur during tumor progression, affect the previously mentioned parameters and whether these cellular changes are reflected in EV composition. The WM266–4 melanoma cell line, treated with two glycosylation inhibitors, was used as a model. After treatment, EVs were isolated and ATR-FTIR measurements were performed, followed by a comprehensive analysis of the data. Significant differences in protein and lipid content were observed between EV populations treated with glycosylation inhibitors, with greater changes in EVs compared to cells. Glycosylation inhibitors also increased the level of phosphorylated proteins in EVs. Our findings showed that changes in EVs were more dynamic than at the cellular level cells. The presented analysis shows that subtle molecular changes can significantly impact EV cargo and properties. FTIR is a powerful tool for detecting these changes and may aid future cancer diagnostics.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120082"},"PeriodicalIF":3.7,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145450692","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-04DOI: 10.1016/j.bbamcr.2025.120083
Yi-Ni Lu , Wen-Xue Liu , Xiao-Han Wang , Xiong Zhang , Yu-Fen Zheng , Tian-Yin Wang , Feng Chen , Feng Yu
Sepsis-related cardiomyopathy (SCM) is a life-threatening complication of sepsis, characterized by cardiac dysfunction. Although angiopoietin 2 (ANGPT2) is pivotal in the pathological progress of several diseases, its functional involvement in the pathogenesis of SCM remains uncharacterized. In this study, we demonstrated that ANGPT2 is a key regulator of SCM progression, bridging the functional homeostasis between vascular endothelial cells (ECs) and cardiomyocytes. Bioinformatics analysis of a SCM-related gene set derived from public databases revealed that ANGPT2 is a critical regulatory node. Serum ANGPT2 levels correlated with elevated creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI) in vivo. Abnormal cardiac function was observed in cecal ligation and puncture (CLP)-induced sepsis mouse model and adeno-associated virus 9-mediated endothelial cell-specific ANGPT2-overexpressing (AAV9-Angpt2) mice, primarily characterized by reduced left ventricular end-diastolic volume (LVEDV) and left ventricular end-systolic volume (LVESV). Additionally, integrin β1 (ITGB1) and integrin β3 (ITGB3) were upregulated, along with dysregulation of calcium signaling-associated proteins in cardiac tissue. Notably, AAV9-Angpt2 mice exhibited exacerbated CLP-induced cardiac dysfunction. In vitro experiments showed that ANGPT2 significantly reduced the beating frequency of primary neonatal mouse cardiomyocytes (NMCMs), and disrupted intracellular calcium homeostasis in adult mouse cardiomyocytes (AMCMs). Mechanistically, ANGPT2 activates and binds to the extracellular domains of ITGB3, thereby triggering ryanodine receptor 2 (RYR2) phosphorylation and upregulating the reticulum calcium ATPase 2 (ATP2a2) and phospholamban (PLN) expression, while simultaneously suppressing PLN phosphorylation. ITGB3-specific siRNA significantly attenuated the effects of ANGPT2 on calcium signaling proteins and intracellular calcium homeostasis. Collectively, these results suggest that the ANGPT2-ITGB3 signaling axis plays an important role in the pathogenesis of SCM in mice and may support this pathway as a potential therapeutic target during sepsis in humans.
{"title":"Endothelial ANGPT2 impairs cardiomyocyte calcium homeostasis via ITGB3 receptor in murine sepsis-related cardiomyopathy","authors":"Yi-Ni Lu , Wen-Xue Liu , Xiao-Han Wang , Xiong Zhang , Yu-Fen Zheng , Tian-Yin Wang , Feng Chen , Feng Yu","doi":"10.1016/j.bbamcr.2025.120083","DOIUrl":"10.1016/j.bbamcr.2025.120083","url":null,"abstract":"<div><div>Sepsis-related cardiomyopathy (SCM) is a life-threatening complication of sepsis, characterized by cardiac dysfunction. Although angiopoietin 2 (ANGPT2) is pivotal in the pathological progress of several diseases, its functional involvement in the pathogenesis of SCM remains uncharacterized. In this study, we demonstrated that ANGPT2 is a key regulator of SCM progression, bridging the functional homeostasis between vascular endothelial cells (ECs) and cardiomyocytes. Bioinformatics analysis of a SCM-related gene set derived from public databases revealed that ANGPT2 is a critical regulatory node. Serum ANGPT2 levels correlated with elevated creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI) <em>in vivo</em>. Abnormal cardiac function was observed in cecal ligation and puncture (CLP)-induced sepsis mouse model and adeno-associated virus 9-mediated endothelial cell-specific ANGPT2-overexpressing (AAV9-<em>Angpt2</em>) mice, primarily characterized by reduced left ventricular end-diastolic volume (LVEDV) and left ventricular end-systolic volume (LVESV). Additionally, integrin β1 (ITGB1) and integrin β3 (ITGB3) were upregulated, along with dysregulation of calcium signaling-associated proteins in cardiac tissue. Notably, AAV9-<em>Angpt2</em> mice exhibited exacerbated CLP-induced cardiac dysfunction. <em>In vitro</em> experiments showed that ANGPT2 significantly reduced the beating frequency of primary neonatal mouse cardiomyocytes (NMCMs), and disrupted intracellular calcium homeostasis in adult mouse cardiomyocytes (AMCMs). Mechanistically, ANGPT2 activates and binds to the extracellular domains of ITGB3, thereby triggering ryanodine receptor 2 (RYR2) phosphorylation and upregulating the reticulum calcium ATPase 2 (ATP2a2) and phospholamban (PLN) expression, while simultaneously suppressing PLN phosphorylation. ITGB3-specific siRNA significantly attenuated the effects of ANGPT2 on calcium signaling proteins and intracellular calcium homeostasis. Collectively, these results suggest that the ANGPT2-ITGB3 signaling axis plays an important role in the pathogenesis of SCM in mice and may support this pathway as a potential therapeutic target during sepsis in humans.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120083"},"PeriodicalIF":3.7,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145450726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-03DOI: 10.1016/j.bbamcr.2025.120081
Evodie Koutouan , Alejandro Lillo , Ayano Kabashima , Jack W. Sample , Danielle Carlson , Rondell Graham , Rory L. Smoot , Elena B. Pasquale
Although EPHA2 is a receptor tyrosine kinase widely expressed in many cancers, it exhibits a uniquely high frequency of coding sequence mutations in cholangiocarcinoma, a cancer of the biliary tract with dismal prognosis. EPHA2 is extensively studied, but very little is known about the role of EPHA2 cancer mutations. To define the functional significance of EPHA2 mutations in biliary tract cancers, we generated representative EPHA2 mutants and monitored major receptor autophosphorylation sites as indicators of kinase activity-dependent signal transduction (known as forward signaling). We found that missense mutations in the ligand-binding domain abrogate ephrinA ligand binding, while missense mutations in the kinase domain abrogate kinase activity. The effects of missense mutations in other domains were less pronounced and varied depending on the phosphosite. The majority of the EPHA2 mutations are nonsense or frame-shift mutations that introduce early stop codons. They generate EPHA2 truncated forms that lack an intact kinase domain or, in some cases, most of the coding sequence. Several EPHA2 mutants tested inhibited tyrosine phosphorylation of co-expressed EPHA2 wild-type, indicating the ability to exert dominant negative effects. We show that EPHA2 forward signaling in cholangiocytes inhibits the ERK oncogenic pathway and cell proliferation, suggesting that loss-of-function mutations facilitate tumor development in the biliary tract. Indeed, an EPHA2 kinase-inactive mutant, but not EPHA2 wild-type, induced proliferative masses consistent with well differentiated cholangiocarcinoma in a validated mouse model of cholangiocarcinogenesis. Thus, EPHA2 has the attributes of a driver gene with tumor suppressor activity in biliary tract cancers.
{"title":"Frequent EPHA2 receptor mutations in cholangiocarcinoma disrupt receptor forward signaling supporting a tumor suppressor role","authors":"Evodie Koutouan , Alejandro Lillo , Ayano Kabashima , Jack W. Sample , Danielle Carlson , Rondell Graham , Rory L. Smoot , Elena B. Pasquale","doi":"10.1016/j.bbamcr.2025.120081","DOIUrl":"10.1016/j.bbamcr.2025.120081","url":null,"abstract":"<div><div>Although EPHA2 is a receptor tyrosine kinase widely expressed in many cancers, it exhibits a uniquely high frequency of coding sequence mutations in cholangiocarcinoma, a cancer of the biliary tract with dismal prognosis. EPHA2 is extensively studied, but very little is known about the role of EPHA2 cancer mutations. To define the functional significance of EPHA2 mutations in biliary tract cancers, we generated representative EPHA2 mutants and monitored major receptor autophosphorylation sites as indicators of kinase activity-dependent signal transduction (known as forward signaling). We found that missense mutations in the ligand-binding domain abrogate ephrinA ligand binding, while missense mutations in the kinase domain abrogate kinase activity. The effects of missense mutations in other domains were less pronounced and varied depending on the phosphosite. The majority of the EPHA2 mutations are nonsense or frame-shift mutations that introduce early stop codons. They generate EPHA2 truncated forms that lack an intact kinase domain or, in some cases, most of the coding sequence. Several EPHA2 mutants tested inhibited tyrosine phosphorylation of co-expressed EPHA2 wild-type, indicating the ability to exert dominant negative effects. We show that EPHA2 forward signaling in cholangiocytes inhibits the ERK oncogenic pathway and cell proliferation, suggesting that loss-of-function mutations facilitate tumor development in the biliary tract. Indeed, an EPHA2 kinase-inactive mutant, but not EPHA2 wild-type, induced proliferative masses consistent with well differentiated cholangiocarcinoma in a validated mouse model of cholangiocarcinogenesis. Thus, EPHA2 has the attributes of a driver gene with tumor suppressor activity in biliary tract cancers.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120081"},"PeriodicalIF":3.7,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145443855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Enhancer of zeste homolog 2 (EZH2) is often overexpressed in malignant tumors and plays a key role in metastasis by trimethylating lysine 27 on histone H3 (H3K27me3). However, the exact mechanism by which EZH2 facilitates metastasis in the diethylnitrosamine (DEN) and N-nitrosomorpholine (NMOR)-induced hepatocellular carcinoma (HCC) model remains unexplored. In this study, we demonstrated that EZH2 expression is elevated in HCC and is associated with metastasis to lung tissue, with poor overall survival outcomes. EZH2 overexpression in HCC liver tissues inhibited claudin-4 gene expression and promoted the activation of the Wnt/β-catenin signaling pathway. β-catenin signaling activates Lymphoid Enhancer-Binding Factor 1(LEF-1) transcription factors that regulate the expression of genes involved in epithelia-to-mesenchymal transition (EMT) and metastasis. EZH2 overexpression significantly elevates H3K27me3 at the promoter, suppressing claudin-4 expression. This leads to increased levels of Matrix metalloproteinase-9 (MMP-9) and vimentin, enhancing the invasion, migration, and metastasis of hepatocellular carcinoma (HCC) cells. However, inhibiting EZH2 with tazemetostat subsequently enhanced claudin-4 expression by reducing Wnt/β-catenin signaling activity and inhibiting EMT-inducing factors.
{"title":"EZH2-mediated hypermethylation of H3K27me3 downregulates claudin-4 and upregulates the Wnt/β-catenin signaling pathway in hepatocellular carcinoma metastasis","authors":"Smriti Verma , Manisha Yadav , Shobhit Verma , Anurag Kumar Srivastava , Madhav Nilakanth Mugale","doi":"10.1016/j.bbamcr.2025.120076","DOIUrl":"10.1016/j.bbamcr.2025.120076","url":null,"abstract":"<div><div>Enhancer of zeste homolog 2 (EZH2) is often overexpressed in malignant tumors and plays a key role in metastasis by trimethylating lysine 27 on histone H3 (H3K27me3). However, the exact mechanism by which EZH2 facilitates metastasis in the diethylnitrosamine (DEN) and N-nitrosomorpholine (NMOR)-induced hepatocellular carcinoma (HCC) model remains unexplored. In this study, we demonstrated that EZH2 expression is elevated in HCC and is associated with metastasis to lung tissue, with poor overall survival outcomes. EZH2 overexpression in HCC liver tissues inhibited claudin-4 gene expression and promoted the activation of the Wnt/β-catenin signaling pathway. β-catenin signaling activates Lymphoid Enhancer-Binding Factor 1(LEF-1) transcription factors that regulate the expression of genes involved in epithelia-to-mesenchymal transition (EMT) and metastasis. EZH2 overexpression significantly elevates H3K27me3 at the promoter, suppressing claudin-4 expression. This leads to increased levels of Matrix metalloproteinase-9 (MMP-9) and vimentin, enhancing the invasion, migration, and metastasis of hepatocellular carcinoma (HCC) cells. However, inhibiting EZH2 with tazemetostat subsequently enhanced claudin-4 expression by reducing Wnt/β-catenin signaling activity and inhibiting EMT-inducing factors.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120076"},"PeriodicalIF":3.7,"publicationDate":"2025-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145443876","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01DOI: 10.1016/j.bbamcr.2025.120077
Daiming Liu , Ning Liu , Xinxuan Liu , Yi Liu , Hongjia Zhang , Zhuoqian Zhou , Jiawang Zheng , Yi Tian
Wound healing requires the coordinated resolution of inflammation and tissue regeneration, whereby the role played by macrophage polarization is critical. This study investigates whether adipose-derived stem cells (ADSCs) promote wound repair through paracrine dickkopf-related protein 1 (DKK1) and explores their regulatory mechanism on macrophage polarization. In a murine full-thickness skin wound model, ADSCs treatment significantly enhanced re-epithelialization, collagen deposition, and Ki67 expression levels, while reducing local inflammatory cytokines and promoting M2 macrophage polarization. In vitro, ADSCs suppressed proinflammatory cytokines and induced M2 markers in LPS/IFN-γ-stimulated macrophages, an effect that was attenuated upon DKK1 knockdown. Mechanistically, DKK1 enhanced PI3K/AKT and JNK pathway activation both in macrophages and wound tissue. DKK1-deficient ADSCs showed a diminished ability to promote wound healing, reduce inflammation, and modulate macrophage phenotypes. These findings identify DKK1 as a key paracrine mediator of ADSCs-driven immunomodulation and tissue regeneration, thereby providing new insights into optimizing stem cell-based therapies for wound healing.
{"title":"Adipose-derived stem cells (ADSCs)-derived DKK1 promotes cutaneous wound healing by inducing M2 macrophage polarization via PI3K/AKT and JNK activation","authors":"Daiming Liu , Ning Liu , Xinxuan Liu , Yi Liu , Hongjia Zhang , Zhuoqian Zhou , Jiawang Zheng , Yi Tian","doi":"10.1016/j.bbamcr.2025.120077","DOIUrl":"10.1016/j.bbamcr.2025.120077","url":null,"abstract":"<div><div>Wound healing requires the coordinated resolution of inflammation and tissue regeneration, whereby the role played by macrophage polarization is critical. This study investigates whether adipose-derived stem cells (ADSCs) promote wound repair through paracrine dickkopf-related protein 1 (DKK1) and explores their regulatory mechanism on macrophage polarization. In a murine full-thickness skin wound model, ADSCs treatment significantly enhanced re-epithelialization, collagen deposition, and Ki67 expression levels, while reducing local inflammatory cytokines and promoting M2 macrophage polarization. <em>In vitro</em>, ADSCs suppressed proinflammatory cytokines and induced M2 markers in LPS/IFN-γ-stimulated macrophages, an effect that was attenuated upon DKK1 knockdown. Mechanistically, DKK1 enhanced PI3K/AKT and JNK pathway activation both in macrophages and wound tissue. DKK1-deficient ADSCs showed a diminished ability to promote wound healing, reduce inflammation, and modulate macrophage phenotypes. These findings identify DKK1 as a key paracrine mediator of ADSCs-driven immunomodulation and tissue regeneration, thereby providing new insights into optimizing stem cell-based therapies for wound healing.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120077"},"PeriodicalIF":3.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145430363","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The 13th edition of the Tuscany Retreat on Cancer Research and Apoptosis was convened on August 23rd to 30th, 2025, continuing its tradition as a biennial forum for scientific exchange. The Retreat brought together leading investigators focused on cancer biology, programmed cell death, and neurodegenerative disorders. Presentations and discussions encompassed diverse themes, including mechanisms of therapeutic resistance, advances in molecular signaling networks, and the identification of novel cellular targets. All topics were unified by their relevance to the regulation of programmed cell death and the pathological consequences of its disruption. Through this review, we intend to convey the essence of the scientific exchanges and to summarize the major take-home messages derived from the Retreat.
{"title":"13th Tuscany Retreat on Cancer Research and Apoptosis: Genetic profiling, resistance mechanisms and novel treatment concepts in cancer and neurodegeneration","authors":"Femke Speelman-Rooms , Olga Troitskaya , Hannah Coxhead , Claire Naveh , Konstantinos Kelepouras , Marta Manik , Annalena Renner , Dhairya Rajguru , Jelena Budimir , Fatma Isil Yapici , Ophélie Champion","doi":"10.1016/j.bbamcr.2025.120079","DOIUrl":"10.1016/j.bbamcr.2025.120079","url":null,"abstract":"<div><div>The 13th edition of the Tuscany Retreat on Cancer Research and Apoptosis was convened on August 23rd to 30th, 2025, continuing its tradition as a biennial forum for scientific exchange. The Retreat brought together leading investigators focused on cancer biology, programmed cell death, and neurodegenerative disorders. Presentations and discussions encompassed diverse themes, including mechanisms of therapeutic resistance, advances in molecular signaling networks, and the identification of novel cellular targets. All topics were unified by their relevance to the regulation of programmed cell death and the pathological consequences of its disruption. Through this review, we intend to convey the essence of the scientific exchanges and to summarize the major take-home messages derived from the Retreat.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120079"},"PeriodicalIF":3.7,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421290","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-30DOI: 10.1016/j.bbamcr.2025.120080
Qiujie Wang, Yuanyuan Liu, Kai Wang, Ailong Huang, Ni Tang, Pai Peng
O-linked-β-N-acetylglucosamine (O-GlcNAc) modification, also known as O-GlcNAcylation, is a dynamic and reversible protein modification. Aberrant O-GlcNAcylation are associated with the pathogenesis of cancers. DEAD-box helicase 46 (DDX46) is an ATP-dependent RNA helicase associated with cancer development; however, its role and regulation in hepatocellular carcinoma (HCC) remain unclear. In this study, we observed that the level of O-GlcNAcylation of DDX46 was significantly elevated in HCC mouse models and patients. In addition, direct OGT-DDX46 interaction facilitates O-GlcNAcylation at the Ser257 site. Mechanically, we discovered that O-GlcNAcylation enhances the stability of DDX46 by impeding ubiquitin-mediated degradation. Increased expression of DDX46 activates the PI3K/Akt signaling pathway, promoting the proliferation and invasion of HCC. Taken together, our study highlights the critical role of DDX46 O-GlcNAcylation in HCC progression, thus proposing targeted disruption of this cascade as a novel therapeutic strategy for HCC treatment.
O-linked-β- n -乙酰氨基葡萄糖(O-GlcNAc)修饰,又称O-GlcNAc酰化,是一种动态可逆的蛋白质修饰。异常的o - glcn酰化与癌症的发病机制有关。DEAD-box解旋酶46 (DDX46)是一种与癌症发展相关的atp依赖性RNA解旋酶;然而,其在肝细胞癌(HCC)中的作用和调控尚不清楚。在本研究中,我们观察到DDX46的o - glcnac酰化水平在HCC小鼠模型和患者中显著升高。此外,直接OGT-DDX46相互作用促进了Ser257位点的o - glcn酰化。机械地,我们发现o - glcn酰化通过阻碍泛素介导的降解来增强DDX46的稳定性。DDX46表达增加激活PI3K/Akt信号通路,促进HCC的增殖和侵袭。综上所述,我们的研究强调了DDX46 o - glcn酰化在HCC进展中的关键作用,因此提出了靶向破坏该级联作为HCC治疗的新治疗策略。
{"title":"O-GlcNAcylation of DDX46 promotes hepatocellular carcinoma progression by activating the PI3K/Akt signaling pathway","authors":"Qiujie Wang, Yuanyuan Liu, Kai Wang, Ailong Huang, Ni Tang, Pai Peng","doi":"10.1016/j.bbamcr.2025.120080","DOIUrl":"10.1016/j.bbamcr.2025.120080","url":null,"abstract":"<div><div>O-linked-β-<em>N</em>-acetylglucosamine (O-GlcNAc) modification, also known as O-GlcNAcylation, is a dynamic and reversible protein modification. Aberrant O-GlcNAcylation are associated with the pathogenesis of cancers. DEAD-box helicase 46 (DDX46) is an ATP-dependent RNA helicase associated with cancer development; however, its role and regulation in hepatocellular carcinoma (HCC) remain unclear. In this study, we observed that the level of O-GlcNAcylation of DDX46 was significantly elevated in HCC mouse models and patients. In addition, direct OGT-DDX46 interaction facilitates O-GlcNAcylation at the Ser257 site. Mechanically, we discovered that O-GlcNAcylation enhances the stability of DDX46 by impeding ubiquitin-mediated degradation. Increased expression of DDX46 activates the PI3K/Akt signaling pathway, promoting the proliferation and invasion of HCC. Taken together, our study highlights the critical role of DDX46 O-GlcNAcylation in HCC progression, thus proposing targeted disruption of this cascade as a novel therapeutic strategy for HCC treatment.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120080"},"PeriodicalIF":3.7,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145426355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-29DOI: 10.1016/j.bbamcr.2025.120078
Yanhong Liu , Xiaoyu Tong
Chronic migraine (CM) is a widespread neurological disorder with heterogeneous underlying causes. Basic leucine zipper ATF-like transcription factor (Batf) can amplify neuroinflammatory responses, but therapeutic strategies targeting Batf in CM remain unclear. Here, we investigated the role of Batf in a nitroglycerin (NTG)-induced CM mouse model and lipopolysaccharide (LPS)-induced BV-2 cells. Batf and Iba-1 expression were enhanced in the trigeminal nucleus caudalis (TNC) of NTG-induced mice. Batf knockdown mitigated NTG-induced central sensitization, decreased CGRP and c-Fos expression, and inhibited Iba-1, iNOS, IL-18, and IL-1β expression in TNC of NTG-induced mice. Furthermore, the NLRP3 inflammasome in TNC of NTG-induced mice was inhibited by Batf knockdown. Our results suggested that Batf knockdown attenuated central sensitization through mediating microglial activation via the NLRP3 inflammasome in the TNC of CM mice. Batf might be a candidate for migraine treatment.
{"title":"Basic leucine zipper ATF-like transcription factor (Batf) aggravates central sensitization in nitroglycerin-induced chronic migraine mouse model","authors":"Yanhong Liu , Xiaoyu Tong","doi":"10.1016/j.bbamcr.2025.120078","DOIUrl":"10.1016/j.bbamcr.2025.120078","url":null,"abstract":"<div><div>Chronic migraine (CM) is a widespread neurological disorder with heterogeneous underlying causes. Basic leucine zipper ATF-like transcription factor (Batf) can amplify neuroinflammatory responses, but therapeutic strategies targeting Batf in CM remain unclear. Here, we investigated the role of Batf in a nitroglycerin (NTG)-induced CM mouse model and lipopolysaccharide (LPS)-induced BV-2 cells. Batf and Iba-1 expression were enhanced in the trigeminal nucleus caudalis (TNC) of NTG-induced mice. Batf knockdown mitigated NTG-induced central sensitization, decreased CGRP and c-Fos expression, and inhibited Iba-1, iNOS, IL-18, and IL-1β expression in TNC of NTG-induced mice. Furthermore, the NLRP3 inflammasome in TNC of NTG-induced mice was inhibited by Batf knockdown. Our results suggested that Batf knockdown attenuated central sensitization through mediating microglial activation via the NLRP3 inflammasome in the TNC of CM mice. Batf might be a candidate for migraine treatment.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120078"},"PeriodicalIF":3.7,"publicationDate":"2025-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145413205","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Polyomaviruses (PyVs) are small dsDNA viruses replicating in the host cell nucleus, thanks to the viral encoded large tumor antigen (LTA). Aided by recent advances in molecular biology techniques, the list of known PyVs is rapidly growing, revealing unexpected broad sequence, and host heterogenicity. Given their dependence on nuclear localization, LTAs represent an attractive model for studying the nuclear transport process. A comprehensive analysis of the evolution of classical nuclear localization signals (cNLSs) within LTAs encoded by PyVs infecting mammals highlighted strong positional conservation of cNLSs between the LXCXE motif and the origin-binding domain (OBD). Here we extend such analysis to PyVs infecting non-mammalian hosts. We combined biochemical, structural and functional assays to demonstrate that black sea bass (BSB) PyV-LTA is transported into the nucleus by the Importin (IMP)α/β1 heterodimer thanks to the recognition of a bipartite cNLS located downstream of the SF3 helicase domain, rather than between the LXCXE motif and the OBD. Such cNLS binds with high affinity to several IMPα paralogs by simultaneously interacting with the minor and major binding sites. Substitution of NLS key basic residues abrogating binding to IMPα, or co-expression with the well characterized IMPα/β1 inhibitor Bimax2 suppressed nuclear localization. Intriguingly a cNLS could be identified in a similar position in LTAs from other PyVs infecting ray-finned fishes, but not cartilaginous fishes, birds or scorpions, where cNLSs were predicted elsewhere. Our study suggests that LTAs from PyVs infecting different non-mammalian hosts might bear cNLS in distinctive positions, possibly reflecting processes of virus-host adaptation.
{"title":"Importin α/β1 dependent nuclear import of black sea bass polyomavirus large tumor antigen is mediated by a classical NLS located downstream of the SF3 helicase domain","authors":"Mikayla Hoad , Silvia Pavan , Sepehr Nematollahzadeh , Ole Tietz , Jospeh Reeman , Jade K. Forwood , Gualtiero Alvisi","doi":"10.1016/j.bbamcr.2025.120074","DOIUrl":"10.1016/j.bbamcr.2025.120074","url":null,"abstract":"<div><div>Polyomaviruses (PyVs) are small dsDNA viruses replicating in the host cell nucleus, thanks to the viral encoded large tumor antigen (LTA). Aided by recent advances in molecular biology techniques, the list of known PyVs is rapidly growing, revealing unexpected broad sequence, and host heterogenicity. Given their dependence on nuclear localization, LTAs represent an attractive model for studying the nuclear transport process. A comprehensive analysis of the evolution of classical nuclear localization signals (cNLSs) within LTAs encoded by PyVs infecting mammals highlighted strong positional conservation of cNLSs between the LXCXE motif and the origin-binding domain (OBD). Here we extend such analysis to PyVs infecting non-mammalian hosts. We combined biochemical, structural and functional assays to demonstrate that black sea bass (BSB) PyV-LTA is transported into the nucleus by the Importin (IMP)α/β1 heterodimer thanks to the recognition of a bipartite cNLS located downstream of the SF3 helicase domain, rather than between the LXCXE motif and the OBD. Such cNLS binds with high affinity to several IMPα paralogs by simultaneously interacting with the minor and major binding sites. Substitution of NLS key basic residues abrogating binding to IMPα, or co-expression with the well characterized IMPα/β1 inhibitor Bimax2 suppressed nuclear localization. Intriguingly a cNLS could be identified in a similar position in LTAs from other PyVs infecting ray-finned fishes, but not cartilaginous fishes, birds or scorpions, where cNLSs were predicted elsewhere. Our study suggests that LTAs from PyVs infecting different non-mammalian hosts might bear cNLS in distinctive positions, possibly reflecting processes of virus-host adaptation.</div></div>","PeriodicalId":8754,"journal":{"name":"Biochimica et biophysica acta. Molecular cell research","volume":"1873 1","pages":"Article 120074"},"PeriodicalIF":3.7,"publicationDate":"2025-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}