Pub Date : 2025-11-22DOI: 10.1016/j.yexcr.2025.114838
Yu Zhang , Weiqiang Chen , Kai Yang, Tao Hao, Wenfeng Cao, Shuqiang Dong
In pulmonary hypertension (PH), metabolic enhancement of glycolysis drives a hyperproliferative and apoptosis-resistant phenotype in pulmonary artery smooth muscle cells (PASMCs), which is a key pathological process leading to pulmonary vascular remodeling. We aimed to uncover the role of CTRP1, a gene responsible for regulating glycolysis, in the modulation of PH-related pathogenesis. In the PH mouse and cell models established by hypoxia stimulation, the regulatory mechanism of CTRP1 deletion on the pathological characteristics of PH was explored. CTRP1 levels were significantly upregulated in PH mice, accompanied by an abnormal increase in lactate production and glycolysis-related key protein expressions (HK2 and PDHK1). Inhibition of CTRP1 markedly improved pulmonary artery pressure and right ventricular function in PH mice by reducing glycolysis levels. In vitro experiments further observed that CTRP1 knockdown suppressed the hypoxia-induced hyperproliferation and anti-apoptosis phenotype of PASMCs, with inhibition of glycolysis. Mechanically, downregulated CTRP1 resulted in p-AMPK activation and p-AKT/mTOR inhibition. This beneficial effect was reversed by AMPKα2 deficiency. Overall, CTRP1 deficiency reverses the hypoxia-induced hyperproliferation and antiapoptotic capacity of PASMCs by weakening glycolysis. These results provide evidence for CTRP1 as a potential therapeutic target in PH.
{"title":"Glycolysis inhibition and AMPK activation: a critical role of CTRP1 deficiency in the treatment of hypoxia-induced pulmonary hypertension","authors":"Yu Zhang , Weiqiang Chen , Kai Yang, Tao Hao, Wenfeng Cao, Shuqiang Dong","doi":"10.1016/j.yexcr.2025.114838","DOIUrl":"10.1016/j.yexcr.2025.114838","url":null,"abstract":"<div><div>In pulmonary hypertension (PH), metabolic enhancement of glycolysis drives a hyperproliferative and apoptosis-resistant phenotype in pulmonary artery smooth muscle cells (PASMCs), which is a key pathological process leading to pulmonary vascular remodeling. We aimed to uncover the role of CTRP1, a gene responsible for regulating glycolysis, in the modulation of PH-related pathogenesis. In the PH mouse and cell models established by hypoxia stimulation, the regulatory mechanism of CTRP1 deletion on the pathological characteristics of PH was explored. CTRP1 levels were significantly upregulated in PH mice, accompanied by an abnormal increase in lactate production and glycolysis-related key protein expressions (HK2 and PDHK1). Inhibition of CTRP1 markedly improved pulmonary artery pressure and right ventricular function in PH mice by reducing glycolysis levels. In vitro experiments further observed that CTRP1 knockdown suppressed the hypoxia-induced hyperproliferation and anti-apoptosis phenotype of PASMCs, with inhibition of glycolysis. Mechanically, downregulated CTRP1 resulted in p-AMPK activation and p-AKT/mTOR inhibition. This beneficial effect was reversed by AMPKα2 deficiency. Overall, CTRP1 deficiency reverses the hypoxia-induced hyperproliferation and antiapoptotic capacity of PASMCs by weakening glycolysis. These results provide evidence for CTRP1 as a potential therapeutic target in PH.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 2","pages":"Article 114838"},"PeriodicalIF":3.5,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145596346","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 : 2025-11-22DOI: 10.1016/j.yexcr.2025.114839
Xueshan Pan , Ke Yu , Kai Chen , Jiao Wang , Zheng Huang , Jiewen Wang , Tong Cao , Jia Ma
The mechanisms underlying the development and progression of clear cell renal cell carcinoma (ccRCC) and its sunitinib resistance are elusive. Fbxo45 is a member of the F-box protein family that has been demonstrated to participate in tumorigenesis. However, the role of Fbxo45 in ccRCC progression has not been characterized. This study aims to investigate the biological functions and molecular mechanism of Fbxo45 in ccRCC progression. We found that Fbxo45 knockdown inhibited the viability and motility of ccRCC cells, while Fbxo45 overexpression resulted in the opposite phenotype. Ectopic expression of Fbxo45 promoted tumor growth in mice. Fbxo45 expression was negatively correlated with Erbin expression, which has been reported to mediate anti-tumor activities in ccRCC. Furthermore, Fbxo45 facilitated ccRCC cell viability and motility by inhibiting Erbin. Notably, Fbxo45 upregulation reduced sunitinib sensitivity in ccRCC cells. Our results suggest that Fbxo45 could be a potential target for ccRCC treatment and sunitinib resistance.
{"title":"Fbxo45 promotes cell viability, invasion and sunitinib resistance of clear cell renal cell carcinoma by targeting Erbin","authors":"Xueshan Pan , Ke Yu , Kai Chen , Jiao Wang , Zheng Huang , Jiewen Wang , Tong Cao , Jia Ma","doi":"10.1016/j.yexcr.2025.114839","DOIUrl":"10.1016/j.yexcr.2025.114839","url":null,"abstract":"<div><div>The mechanisms underlying the development and progression of clear cell renal cell carcinoma (ccRCC) and its sunitinib resistance are elusive. Fbxo45 is a member of the F-box protein family that has been demonstrated to participate in tumorigenesis. However, the role of Fbxo45 in ccRCC progression has not been characterized. This study aims to investigate the biological functions and molecular mechanism of Fbxo45 in ccRCC progression. We found that Fbxo45 knockdown inhibited the viability and motility of ccRCC cells, while Fbxo45 overexpression resulted in the opposite phenotype. Ectopic expression of Fbxo45 promoted tumor growth in mice. Fbxo45 expression was negatively correlated with Erbin expression, which has been reported to mediate anti-tumor activities in ccRCC. Furthermore, Fbxo45 facilitated ccRCC cell viability and motility by inhibiting Erbin. Notably, Fbxo45 upregulation reduced sunitinib sensitivity in ccRCC cells. Our results suggest that Fbxo45 could be a potential target for ccRCC treatment and sunitinib resistance.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 2","pages":"Article 114839"},"PeriodicalIF":3.5,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145596335","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 : 2025-11-20DOI: 10.1016/j.yexcr.2025.114829
Ashima Thakur, Shweta Saran
Two-Pore Channel 2 (TPC2) are calcium (Ca2+) release channels regulated by Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP), primarily localized to the endolysosomal system. TPC2 regulates diverse Ca2+-dependent processes such as cell proliferation, development, migration, and autophagy. In the protist Dictyostelium discoideum, intracellular Ca2+ levels and autophagy are key determinants of cell-fate, particularly for stalk cell differentiation. To investigate the involvement of DdTPC2 in NAADP-mediated Ca2+ signalling, we treated wild-type (Ax2) and various tpc2 mutant strains, like overexpressors (tpc2OE), knockout (tpc2-), and rescue (tpc2Res), with both NAADP-AM (NAADP agonist) and trans-Ned-19 (NAADP antagonist). Our findings show that trans-Ned-19 does not specifically inhibit DdTPC2 activity during the proliferative (vegetative) stage, but acts as a specific TPC2 inhibitor in freshly starved cells or during multicellular development, as the phenotypes displayed by trans-Ned-19 treated Ax2 cells were similar to tpc2- cells. Trans-Ned-19 colocalizes with lysosomes, consistent with the subcellular distribution of TPC2. Fluid-phase endocytosis was reduced in tpc2- cells, implicating TPC2 in endolysosomal function. Intracellular calcium measurements demonstrated that NAADP regulates Ca2+ signalling through TPC2, as both NAADP-AM and trans-Ned-19 significantly modulated intracellular free Ca2+ levels in TPC2-expressing strains. Notably, trans-Ned-19 treatment reduced autophagic flux in Ax2 cells to tpc2- levels, demonstrating a positive correlation between TPC2 and autophagic flux, further supported by rapamycin and 3-methyladenine treatment results. This study highlights TPC2 as a pivotal regulator of NAADP-mediated Ca2+ signalling and autophagy in Dictyostelium, with broad implications for understanding these processes in higher eukaryotes.
{"title":"Elucidating the impact of trans-ned-19 on two-Pore channel 2 mutants of Dictyostelium: changes in intracellular calcium levels and subsequent effect on autophagic flux","authors":"Ashima Thakur, Shweta Saran","doi":"10.1016/j.yexcr.2025.114829","DOIUrl":"10.1016/j.yexcr.2025.114829","url":null,"abstract":"<div><div>Two-Pore Channel 2 (TPC2) are calcium (Ca<sup>2+</sup>) release channels regulated by Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP), primarily localized to the endolysosomal system. TPC2 regulates diverse Ca<sup>2+</sup>-dependent processes such as cell proliferation, development, migration, and autophagy. In the protist <em>Dictyostelium discoideum</em>, intracellular Ca<sup>2+</sup> levels and autophagy are key determinants of cell-fate, particularly for stalk cell differentiation. To investigate the involvement of <em>Dd</em>TPC2 in NAADP-mediated Ca<sup>2+</sup> signalling, we treated wild-type (Ax2) and various <em>tpc2</em> mutant strains, like overexpressors (<em>tpc2</em><sup><em>OE</em></sup>), knockout (<em>tpc2</em><sup>-</sup>), and rescue (<em>tpc2</em><sup><em>Res</em></sup>), with both NAADP-AM (NAADP agonist) and trans-Ned-19 (NAADP antagonist). Our findings show that trans-Ned-19 does not specifically inhibit <em>Dd</em>TPC2 activity during the proliferative (vegetative) stage, but acts as a specific TPC2 inhibitor in freshly starved cells or during multicellular development, as the phenotypes displayed by trans-Ned-19 treated Ax2 cells were similar to <em>tpc2</em><sup><em>-</em></sup> cells. Trans-Ned-19 colocalizes with lysosomes, consistent with the subcellular distribution of TPC2. Fluid-phase endocytosis was reduced in <em>tpc2</em><sup>-</sup> cells, implicating TPC2 in endolysosomal function. Intracellular calcium measurements demonstrated that NAADP regulates Ca<sup>2+</sup> signalling through TPC2, as both NAADP-AM and trans-Ned-19 significantly modulated intracellular free Ca<sup>2+</sup> levels in TPC2-expressing strains. Notably, trans-Ned-19 treatment reduced autophagic flux in Ax2 cells to <em>tpc2</em><sup><em>-</em></sup> levels, demonstrating a positive correlation between TPC2 and autophagic flux, further supported by rapamycin and 3-methyladenine treatment results. This study highlights TPC2 as a pivotal regulator of NAADP-mediated Ca<sup>2+</sup> signalling and autophagy in <em>Dictyostelium</em>, with broad implications for understanding these processes in higher eukaryotes.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 2","pages":"Article 114829"},"PeriodicalIF":3.5,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581909","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}
Heterotopic ossification is a pathological process characterized by aberrant bone formation in soft tissues, leading to joint pain, stiffness, functional impairment, and poor quality of life. We used a mouse Achilles tendon heterotopic ossification model and in vitro tendon-derived stem cells (TDSCs) assays to identify the molecular mechanism of metformin in prevention of heterotopic ossification. Metformin significantly attenuated heterotopic ossification, reducing ectopic bone volume and osteogenic gene expression. In vitro, metformin inhibited TDSCs osteogenic differentiation in a dose-dependent manner, decreasing calcium nodule deposition and osteogenic marker expression. Transcriptomic analysis revealed downregulated Nr4a1 expression in metformin-treated heterotopic ossification samples; in vitro experiments confirmed that Nr4a1 activation enhances TDSCs osteogenesis, and Nr4a1 knockdown suppresses osteogenesis. Metformin also reduced Wnt4 and β-catenin expression, suggesting that Nr4a1 promotes heterotopic ossification by positively regulating Wnt/β-catenin signaling. In sum, metformin downregulates expression of Nr4a1 in TDSCs, which suppresses osteogenic differentiation by inhibiting Wnt/β-catenin signaling. As a mediator of TDSC osteogenic differentiation, Nr4a1 may be a therapeutic target for heterotopic ossification.
{"title":"Metformin inhibits heterotopic ossification of mouse Achilles tendon by inhibiting the Nr4a1/Wnt/β-catenin signaling pathway","authors":"Danxia Zheng , Mengyu Jiang , Wenjie Wang , Jianglin Yu , Xiaoyang Qi , Yixin Chen","doi":"10.1016/j.yexcr.2025.114824","DOIUrl":"10.1016/j.yexcr.2025.114824","url":null,"abstract":"<div><div>Heterotopic ossification is a pathological process characterized by aberrant bone formation in soft tissues, leading to joint pain, stiffness, functional impairment, and poor quality of life. We used a mouse Achilles tendon heterotopic ossification model and in vitro tendon-derived stem cells (TDSCs) assays to identify the molecular mechanism of metformin in prevention of heterotopic ossification. Metformin significantly attenuated heterotopic ossification, reducing ectopic bone volume and osteogenic gene expression. In vitro, metformin inhibited TDSCs osteogenic differentiation in a dose-dependent manner, decreasing calcium nodule deposition and osteogenic marker expression. Transcriptomic analysis revealed downregulated Nr4a1 expression in metformin-treated heterotopic ossification samples; in vitro experiments confirmed that Nr4a1 activation enhances TDSCs osteogenesis, and Nr4a1 knockdown suppresses osteogenesis. Metformin also reduced Wnt4 and β-catenin expression, suggesting that Nr4a1 promotes heterotopic ossification by positively regulating Wnt/β-catenin signaling. In sum, metformin downregulates expression of Nr4a1 in TDSCs, which suppresses osteogenic differentiation by inhibiting Wnt/β-catenin signaling. As a mediator of TDSC osteogenic differentiation, Nr4a1 may be a therapeutic target for heterotopic ossification.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 2","pages":"Article 114824"},"PeriodicalIF":3.5,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145534361","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 : 2025-11-13DOI: 10.1016/j.yexcr.2025.114779
Jianbing Hao, Siyu Wang, Lirong Hao
Vascular calcification, a major contributor to cardiovascular morbidity, involves pathological osteogenic transdifferentiation of vascular smooth muscle cell (VSMC) under hyperphosphatemic conditions in chronic kidney disease. This study investigates the role of lincRNA-p21 and small extracellular vesicles in phosphate-induced mouse aortic smooth muscle cell (MASMC) calcification. Exposure to phosphate (2.6 mmol/L) triggered time-dependent calcification, characterized by enhanced calcium deposition, endoplasmic reticulum remodeling, and intracellular calcium accumulation (p < 0.05 vs. 24/48-h controls). Concurrently, the expression of osteogenic markers (BGP, OCN, and OPN) and lincRNA-p21 was significantly upregulated, whereas the expression of contractile phenotype-specific markers (SM22α, SM-MHC, and SM α-actin) was markedly down-regulated. This pattern of gene expression was correlated with MASMC osteogenic transdifferentiation. Small extracellular vesicles isolated from phosphate-treated MASMC exhibited elevated lincRNA-p21 levels (p < 0.05) and induced calcification and apoptosis in recipient cells, suggesting small extracellular vesicles-mediated propagation of calcific signals. Functional studies demonstrated that lincRNA-p21 overexpression exacerbated calcification, apoptosis, and osteogenic marker expression, while its knockdown attenuated these effects (p < 0.05). Time-course analyses revealed lincRNA-p21 dynamically regulates small extracellular vesicles secretion, calcium accumulation, and apoptotic pathways, acting as a molecular switch driving phosphate-induced calcification. These findings establish lincRNA-p21 as a critical mediator of MASMC calcification via small extracellular vesicles-dependent mechanisms, offering insights into therapeutic strategies for vascular calcification.
{"title":"lincRNA-p21 drives apoptosis and calcification of vascular smooth muscle cell via small extracellular vesicles under hyperphosphatemic conditions in chronic kidney disease","authors":"Jianbing Hao, Siyu Wang, Lirong Hao","doi":"10.1016/j.yexcr.2025.114779","DOIUrl":"10.1016/j.yexcr.2025.114779","url":null,"abstract":"<div><div>Vascular calcification, a major contributor to cardiovascular morbidity, involves pathological osteogenic transdifferentiation of vascular smooth muscle cell (VSMC) under hyperphosphatemic conditions in chronic kidney disease. This study investigates the role of lincRNA-p21 and small extracellular vesicles in phosphate-induced mouse aortic smooth muscle cell (MASMC) calcification. Exposure to phosphate (2.6 mmol/L) triggered time-dependent calcification, characterized by enhanced calcium deposition, endoplasmic reticulum remodeling, and intracellular calcium accumulation (p < 0.05 vs. 24/48-h controls). Concurrently, the expression of osteogenic markers (BGP, OCN, and OPN) and lincRNA-p21 was significantly upregulated, whereas the expression of contractile phenotype-specific markers (SM22α, SM-MHC, and SM α-actin) was markedly down-regulated. This pattern of gene expression was correlated with MASMC osteogenic transdifferentiation. Small extracellular vesicles isolated from phosphate-treated MASMC exhibited elevated lincRNA-p21 levels (p < 0.05) and induced calcification and apoptosis in recipient cells, suggesting small extracellular vesicles-mediated propagation of calcific signals. Functional studies demonstrated that lincRNA-p21 overexpression exacerbated calcification, apoptosis, and osteogenic marker expression, while its knockdown attenuated these effects (p < 0.05). Time-course analyses revealed lincRNA-p21 dynamically regulates small extracellular vesicles secretion, calcium accumulation, and apoptotic pathways, acting as a molecular switch driving phosphate-induced calcification. These findings establish lincRNA-p21 as a critical mediator of MASMC calcification via small extracellular vesicles-dependent mechanisms, offering insights into therapeutic strategies for vascular calcification.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 2","pages":"Article 114779"},"PeriodicalIF":3.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145530515","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 : 2025-11-13DOI: 10.1016/j.yexcr.2025.114825
Jingjing Wang , Jinqiu Jia , Zitong Cao , Zeming Cai , Kai Zhang , Jin He , Chunyan Wu , Zuo Wang
Ferroptosis is a phospholipid peroxidation-mediated and iron-dependent cell death form. Inhibiting ferroptosis is a promising strategy for the prevention and treatment of cardiovascular diseases. Annexin A1 (ANXA1) is an endogenous anti-inflammatory mediator that plays an important regulatory role in cardiovascular diseases such as atherosclerosis, especially in inflammation suppression, protecting the heart from injury, and regulating vascular function. Ac2-26 is a synthetic peptide derived from the N-terminal 26 amino acids of ANXA1, which retains its anti-inflammatory properties. However, the regulatory mechanism of ANXA1 in atherosclerosis (AS) is not yet fully understood. This study aims to explore the specific role of ferroptosis in HUVECs and demonstrate that ANXA1 can disrupt ferritinophagy and protect endothelial function. Treatment with Ac2-26 or ANXA1-overexpressing HUVECs alleviated RSL3-induced endothelial cell dysfunction and inhibited lipid peroxidation, as evidenced by a reduction in ferrous ion levels and upregulation of GPX4, FTH1, and SLC7A11 protein expression, along with a downregulation of LC3-II and NCOA4 expression. In contrast, knockdown of ANXA1 in HUVECs failed to suppress the outcome of ferroptosis. Furthermore, co-immunoprecipitation analysis revealed that ANXA1 overexpression prevented ferritin degradation by disrupting the NCOA4-FTH1 protein-protein interaction, reducing the bioavailability of intracellular ferrous ions, thereby blocking ferroptosis. In conclusion, our findings identify a novel mechanism, showing that ANXA1 can inhibit ferroptosis via ferritinophagy, thereby alleviating endothelial dysfunction, which may provide a new therapeutic avenue for AS.
{"title":"ANXA1 inhibits ferroptosis in HUVECs via negative regulation of ferritinophagy","authors":"Jingjing Wang , Jinqiu Jia , Zitong Cao , Zeming Cai , Kai Zhang , Jin He , Chunyan Wu , Zuo Wang","doi":"10.1016/j.yexcr.2025.114825","DOIUrl":"10.1016/j.yexcr.2025.114825","url":null,"abstract":"<div><div>Ferroptosis is a phospholipid peroxidation-mediated and iron-dependent cell death form. Inhibiting ferroptosis is a promising strategy for the prevention and treatment of cardiovascular diseases. Annexin A1 (ANXA1) is an endogenous anti-inflammatory mediator that plays an important regulatory role in cardiovascular diseases such as atherosclerosis, especially in inflammation suppression, protecting the heart from injury, and regulating vascular function. Ac2-26 is a synthetic peptide derived from the N-terminal 26 amino acids of ANXA1, which retains its anti-inflammatory properties. However, the regulatory mechanism of ANXA1 in atherosclerosis (AS) is not yet fully understood. This study aims to explore the specific role of ferroptosis in HUVECs and demonstrate that ANXA1 can disrupt ferritinophagy and protect endothelial function. Treatment with Ac2-26 or ANXA1-overexpressing HUVECs alleviated RSL3-induced endothelial cell dysfunction and inhibited lipid peroxidation, as evidenced by a reduction in ferrous ion levels and upregulation of GPX4, FTH1, and SLC7A11 protein expression, along with a downregulation of LC3-II and NCOA4 expression. In contrast, knockdown of ANXA1 in HUVECs failed to suppress the outcome of ferroptosis. Furthermore, co-immunoprecipitation analysis revealed that ANXA1 overexpression prevented ferritin degradation by disrupting the NCOA4-FTH1 protein-protein interaction, reducing the bioavailability of intracellular ferrous ions, thereby blocking ferroptosis. In conclusion, our findings identify a novel mechanism, showing that ANXA1 can inhibit ferroptosis via ferritinophagy, thereby alleviating endothelial dysfunction, which may provide a new therapeutic avenue for AS.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 1","pages":"Article 114825"},"PeriodicalIF":3.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145530576","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}
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by excessive lipid deposition in hepatocytes. Augmenter of liver regeneration (ALR, encoded by the Gfer gene), a survival factor in liver cells, protects the liver against various injuries, whereas downregulation of ALR expression accelerates MASLD progression in mice. CD36 functions as a long chain free fatty acid transporter, playing a crucial role during pathogenesis of MASLD. Herein, the expression level of CD36 was found to significantly decrease in ALR-transfected (ALR-Tx) HepG2 cells and upregulated in shRNA-ALR (shALR) cells after oleic acid/palmitic acid treatment. Furthermore, CD36 expression was markedly elevated in MASLD model mice with heterozygous Gfer knockdown (Gfer+/−) fed a high-fat diet,as well as model mice with liver-specific Gfer knockout (Gfer-CKO). Lipid uptake increased and lipophagy was suppressed in shALR HepG2 cells, and conversly, lipid uptake reduced and lipophagy was promoted in ALR-Tx HepG2 cells. After downregulation of CD36 expression by siRNA-CD36 in shALR cells, lipid accumulation markedly decreased, and lipophagy was stimulated. Further, the mRNA and protein expression of ATF3 (Activating Transcription Factor 3) were found to substantially elevated and decreased in ALR-Tx cells and shALR cells respectively, and CD36 expression was induced after ATF3 was knock down. Our results thus suggest that lack of ALR aggravates lipid deposition in MASLD partly through the ATF3-CD36 axis.
{"title":"Augmenter of liver regeneration alleviates lipid accumulation in MASLD through ATF3-CD36 axis","authors":"Weichun Xiao , Yuan Wu , Ling Qin , Jing Zhang , Wei An","doi":"10.1016/j.yexcr.2025.114823","DOIUrl":"10.1016/j.yexcr.2025.114823","url":null,"abstract":"<div><div>Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by excessive lipid deposition in hepatocytes. Augmenter of liver regeneration (ALR, encoded by the <em>Gfer</em> gene), a survival factor in liver cells, protects the liver against various injuries, whereas downregulation of ALR expression accelerates MASLD progression in mice. CD36 functions as a long chain free fatty acid transporter, playing a crucial role during pathogenesis of MASLD. Herein, the expression level of CD36 was found to significantly decrease in ALR-transfected (ALR-Tx) HepG2 cells and upregulated in shRNA-ALR (shALR) cells after oleic acid/palmitic acid treatment. Furthermore, CD36 expression was markedly elevated in MASLD model mice with heterozygous <em>Gfer</em> knockdown (Gfer<sup>+/−</sup>) fed a high-fat diet,as well as model mice with liver-specific <em>Gfer</em> knockout (Gfer-CKO). Lipid uptake increased and lipophagy was suppressed in shALR HepG2 cells, and conversly, lipid uptake reduced and lipophagy was promoted in ALR-Tx HepG2 cells. After downregulation of CD36 expression by siRNA-CD36 in shALR cells, lipid accumulation markedly decreased, and lipophagy was stimulated. Further, the mRNA and protein expression of ATF3 (Activating Transcription Factor 3) were found to substantially elevated and decreased in ALR-Tx cells and shALR cells respectively, and CD36 expression was induced after ATF3 was knock down. Our results thus suggest that lack of ALR aggravates lipid deposition in MASLD partly through the ATF3-CD36 axis.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 2","pages":"Article 114823"},"PeriodicalIF":3.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145530556","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 : 2025-11-13DOI: 10.1016/j.yexcr.2025.114828
Chiaki Noguchi , Kalisse I. Horne , Tylor Brewster , Alyssa Duffy , Jeel P. Shah , Amber Theriault , Olivia El Naggar , Soumya Vavilala , Shivani Sheth , Eishi Noguchi
Esophageal squamous cell carcinoma (ESCC) is one of the deadliest forms of squamous cell carcinoma, comprising approximately 90 % of all esophageal cancer cases. We previously demonstrated that the Fanconi anemia DNA repair (FA) pathway mitigates replication stress to preserve self-renewal capacity of esophageal cancer cells, highlighting the critical role of minimizing replication stress in esophageal cancer proliferation. In this study, to further explore the role of replication stress in esophageal cancer growth, we investigated the function of Timeless, a key subunit of the replication fork protection complex essential for preventing replication stress. Our findings reveal that Timeless is upregulated in esophageal cancer cells, and its depletion increases sensitivity to DNA-damaging agents, inducing cellular senescence in esophageal keratinocytes. Timeless depletion also elevates the DNA damage response while reducing the expression of DNA repair proteins associated with the FA pathway and homologous recombination. Furthermore, the loss of Timeless impairs colony-forming ability in soft agar and diminishes the self-renewal capacity required to form 3D organoids. These results suggest that Timeless plays a critical role in facilitating DNA repair and esophageal cancer progression and may represent a promising target for developing effective therapeutic strategies to treat esophageal cancers.
{"title":"Timeless prevents senescence-associated phenotypes and enhances DNA repair to promote esophageal cancer cell growth","authors":"Chiaki Noguchi , Kalisse I. Horne , Tylor Brewster , Alyssa Duffy , Jeel P. Shah , Amber Theriault , Olivia El Naggar , Soumya Vavilala , Shivani Sheth , Eishi Noguchi","doi":"10.1016/j.yexcr.2025.114828","DOIUrl":"10.1016/j.yexcr.2025.114828","url":null,"abstract":"<div><div>Esophageal squamous cell carcinoma (ESCC) is one of the deadliest forms of squamous cell carcinoma, comprising approximately 90 % of all esophageal cancer cases. We previously demonstrated that the Fanconi anemia DNA repair (FA) pathway mitigates replication stress to preserve self-renewal capacity of esophageal cancer cells, highlighting the critical role of minimizing replication stress in esophageal cancer proliferation. In this study, to further explore the role of replication stress in esophageal cancer growth, we investigated the function of Timeless, a key subunit of the replication fork protection complex essential for preventing replication stress. Our findings reveal that Timeless is upregulated in esophageal cancer cells, and its depletion increases sensitivity to DNA-damaging agents, inducing cellular senescence in esophageal keratinocytes. Timeless depletion also elevates the DNA damage response while reducing the expression of DNA repair proteins associated with the FA pathway and homologous recombination. Furthermore, the loss of Timeless impairs colony-forming ability in soft agar and diminishes the self-renewal capacity required to form 3D organoids. These results suggest that Timeless plays a critical role in facilitating DNA repair and esophageal cancer progression and may represent a promising target for developing effective therapeutic strategies to treat esophageal cancers.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"455 1","pages":"Article 114828"},"PeriodicalIF":3.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145530490","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}
Acute kidney injury (AKI) to chronic kidney disease (CKD) transition is a progressive, long-term kidney dysfunction driven by complex pathophysiological processes, including persistent endoplasmic reticulum (ER) stress and impaired autophagy, contributing to fibrosis. Long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3) has been implicated in endoplasmic reticulum (ER) stress and autophagy in several diseases, but its role in kidney injury and fibrosis during AKI-to-CKD transition remains unclear. Our previous transcriptomic analysis revealed that MEG3 is dysregulated during this transition, prompting us to explore its functional role. In this study, we investigated the function of MEG3 in ER stress–autophagy crosstalk during the AKI-to-CKD transition. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9)-mediated MEG3 knockout in NRK-52E cells was confirmed by T7 endonuclease assay, quantitative real-time polymerase chain reaction (qRT-PCR), and fluorescence in-situ hybridization (FISH) assay. Functionally, MEG3 knockout markedly attenuated ER stress and apoptosis, as shown by reduced expression of BiP, CHOP, ATF6, ATF4, p-PERK, p-eIF2α, along with restoration of the Bax/Bcl-2 balance. Autophagy activity was restored, with increased Beclin-1 and LC3B expression and decreased p62 accumulation. Furthermore, fibrotic progression was reduced, as indicated by lower levels of fibronectin and collagen I. Notably, tauroursodeoxycholic acid (TUDCA, 400 μM) acted synergistically with MEG3 knockout, further suppressing ER stress and fibrosis markers compared to either treatment alone. These findings demonstrate that MEG3 promotes maladaptive ER stress and impaired autophagy in tubular epithelial cells, driving AKI-to-CKD transition. Targeting MEG3 through CRISPR-based strategies or in combination with TUDCA may represent a promising therapeutic strategy to mitigate fibrosis and slow disease progression.
{"title":"Targeting LncRNA MEG3 to modulate ER stress and autophagy: A CRISPR/Cas9-based strategy in AKI-to-CKD transition","authors":"Bhupendra Puri , Syamantak Majumder , Anil Bhanudas Gaikwad","doi":"10.1016/j.yexcr.2025.114826","DOIUrl":"10.1016/j.yexcr.2025.114826","url":null,"abstract":"<div><div>Acute kidney injury (AKI) to chronic kidney disease (CKD) transition is a progressive, long-term kidney dysfunction driven by complex pathophysiological processes, including persistent endoplasmic reticulum (ER) stress and impaired autophagy, contributing to fibrosis. Long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3) has been implicated in endoplasmic reticulum (ER) stress and autophagy in several diseases, but its role in kidney injury and fibrosis during AKI-to-CKD transition remains unclear. Our previous transcriptomic analysis revealed that MEG3 is dysregulated during this transition, prompting us to explore its functional role. In this study, we investigated the function of MEG3 in ER stress–autophagy crosstalk during the AKI-to-CKD transition<strong>.</strong> Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9)-mediated MEG3 knockout in NRK-52E cells was confirmed by T7 endonuclease assay, quantitative real-time polymerase chain reaction (qRT-PCR), and fluorescence in-situ hybridization (FISH) assay. Functionally, MEG3 knockout markedly attenuated ER stress and apoptosis, as shown by reduced expression of BiP, CHOP, ATF6, ATF4, p-PERK, p-eIF2α, along with restoration of the Bax/Bcl-2 balance. Autophagy activity was restored, with increased Beclin-1 and LC3B expression and decreased p62 accumulation. Furthermore, fibrotic progression was reduced, as indicated by lower levels of fibronectin and collagen I. Notably, tauroursodeoxycholic acid (TUDCA, 400 μM) acted synergistically with MEG3 knockout, further suppressing ER stress and fibrosis markers compared to either treatment alone. These findings demonstrate that MEG3 promotes maladaptive ER stress and impaired autophagy in tubular epithelial cells, driving AKI-to-CKD transition. Targeting MEG3 through CRISPR-based strategies or in combination with TUDCA may represent a promising therapeutic strategy to mitigate fibrosis and slow disease progression<strong>.</strong></div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 1","pages":"Article 114826"},"PeriodicalIF":3.5,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145512300","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 : 2025-11-10DOI: 10.1016/j.yexcr.2025.114827
Qiang Xu , Ge Xuan , Chongying Zhu , Bingquan Zhu , Zihao An , Meiyuan Jin , Chao Tang
Preeclampsia (PE) is a multisystem disorder with increased maternal and perinatal mortality and morbidity while the treatment of PE remains largely unknown in clinic. Therefore, it is necessary to find more effective therapeutic methods of PE. We report herein that, Tianma Gouteng Decoction (TGD) generates protective effects against PE by activating Wnt 3a/β-catenin signaling and simultaneously suppressing ferroptosis in placental trophoblast cells. Particularly, Gastrodin (Gtd), the active component in TGD, plays a major role in protection against PE. Mechanistically, Gtd up-regulates the Wnt 3a/β-catenin signaling activity by inducing Wnt 3a mRNA expression, resulting in the increased expression of β-catenin-controlled target genes. On the other hand, Gtd-triggered Wnt activation obviously exerts negative effects on ferroptosis by promoting expression levels of the anti-ferroptosis proteins accompanied by the down-regulated reactive oxygen species (ROS) production and total iron content but the up-regulated L-Glutathione (GSH) levels. Consistently, Gtd-administration reveals apparent anti-hypertensive effects in a PE-like mouse model with diminished ferroptosis, whereas deactivation of β-catenin by administration with the specific antagonist ICG001 disrupts the protective effects derived from Gtd. Therefore, our results provide an innovative basis for the role of Gtd as a new therapy for PE.
{"title":"Gastrodin produces therapeutic effects against preeclampsia by activating Wnt 3a signaling and inhibiting ferroptosis","authors":"Qiang Xu , Ge Xuan , Chongying Zhu , Bingquan Zhu , Zihao An , Meiyuan Jin , Chao Tang","doi":"10.1016/j.yexcr.2025.114827","DOIUrl":"10.1016/j.yexcr.2025.114827","url":null,"abstract":"<div><div>Preeclampsia (PE) is a multisystem disorder with increased maternal and perinatal mortality and morbidity while the treatment of PE remains largely unknown in clinic. Therefore, it is necessary to find more effective therapeutic methods of PE. We report herein that, Tianma Gouteng Decoction (TGD) generates protective effects against PE by activating Wnt 3a/β-catenin signaling and simultaneously suppressing ferroptosis in placental trophoblast cells. Particularly, Gastrodin (Gtd), the active component in TGD, plays a major role in protection against PE. Mechanistically, Gtd up-regulates the Wnt 3a/β-catenin signaling activity by inducing <em>Wnt 3a</em> mRNA expression, resulting in the increased expression of β-catenin-controlled target genes. On the other hand, Gtd-triggered Wnt activation obviously exerts negative effects on ferroptosis by promoting expression levels of the anti-ferroptosis proteins accompanied by the down-regulated reactive oxygen species (ROS) production and total iron content but the up-regulated L-Glutathione (GSH) levels. Consistently, Gtd-administration reveals apparent anti-hypertensive effects in a PE-like mouse model with diminished ferroptosis, whereas deactivation of β-catenin by administration with the specific antagonist ICG001 disrupts the protective effects derived from Gtd. Therefore, our results provide an innovative basis for the role of Gtd as a new therapy for PE.</div></div>","PeriodicalId":12227,"journal":{"name":"Experimental cell research","volume":"454 1","pages":"Article 114827"},"PeriodicalIF":3.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145502588","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}