Pub Date : 2025-01-20DOI: 10.1016/j.cellsig.2025.111620
Shousen Hu , Zian Wang , Xu Ding , Daoke Yao , Yue Du , Xiangzhen Kong
Cetuximab resistance is a significant challenge in the treatment of head and neck squamous cell carcinoma (HNSCC). In this study, cetuximab-resistant HNSCC cell lines were established, and untargeted metabolomics was used to detect differences in metabolite profiles between sensitive and resistant cell lines. It was found that glutathione metabolism significantly differed between the sensitive and resistant lines. Combining these findings with transcriptome data, correlation analysis of metabolites revealed that IDH2 regulated glutathione metabolism and contributed to cetuximab resistance in FaDu cells. In vitro experiments showed that IDH2 was highly expressed in FaDu-CR cells, and IDH2 knockdown significantly enhanced the sensitivity of FaDu and FaDu-CR cells to cetuximab. IDH2 knockdown reduced GSH levels and GPX4 expression in FaDu and FaDu-CR cells under cetuximab treatment, while increasing lipid ROS levels. In vivo experiments demonstrated that IDH2 knockdown decreased the tumorigenic ability of FaDu-CR cells in nude mice treated with cetuximab, as well as reduced GPX4 and Ki67 levels in tumor tissues. In conclusion, IDH2 regulated glutathione metabolism and contributed to cetuximab resistance in HNSCC. This study explores strategies to ameliorate cetuximab resistance in HNSCC preclinical models, providing new insights for reversing cetuximab resistance in HNSCC.
{"title":"The roles of IDH2 and glutathione metabolism in cetuximab resistance in head and neck squamous cell carcinoma investigated by metabolomics and transcriptomics","authors":"Shousen Hu , Zian Wang , Xu Ding , Daoke Yao , Yue Du , Xiangzhen Kong","doi":"10.1016/j.cellsig.2025.111620","DOIUrl":"10.1016/j.cellsig.2025.111620","url":null,"abstract":"<div><div>Cetuximab resistance is a significant challenge in the treatment of head and neck squamous cell carcinoma (HNSCC). In this study, cetuximab-resistant HNSCC cell lines were established, and untargeted metabolomics was used to detect differences in metabolite profiles between sensitive and resistant cell lines. It was found that glutathione metabolism significantly differed between the sensitive and resistant lines. Combining these findings with transcriptome data, correlation analysis of metabolites revealed that IDH2 regulated glutathione metabolism and contributed to cetuximab resistance in FaDu cells. <em>In vitro</em> experiments showed that IDH2 was highly expressed in FaDu-CR cells, and IDH2 knockdown significantly enhanced the sensitivity of FaDu and FaDu-CR cells to cetuximab. IDH2 knockdown reduced GSH levels and GPX4 expression in FaDu and FaDu-CR cells under cetuximab treatment, while increasing lipid ROS levels. <em>In vivo</em> experiments demonstrated that IDH2 knockdown decreased the tumorigenic ability of FaDu-CR cells in nude mice treated with cetuximab, as well as reduced GPX4 and Ki67 levels in tumor tissues. In conclusion, IDH2 regulated glutathione metabolism and contributed to cetuximab resistance in HNSCC. This study explores strategies to ameliorate cetuximab resistance in HNSCC preclinical models, providing new insights for reversing cetuximab resistance in HNSCC.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111620"},"PeriodicalIF":4.4,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022398","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 : 2025-01-16DOI: 10.1016/j.cellsig.2025.111610
Guangfa Xia , Jing Qian , Yu Wang , Fei Xiao
Background
While TRPA1 serves as a therapeutic target for nociceptive pain, its role in acute visceral pain induced by uterine cervical dilation (UCD) remains an enigma. This study aims to elucidate the upstream and downstream mechanisms of TRPA1 in the context of UCD-induced acute visceral pain.
Methods
The UCD rats were administered with SAH (inhibitor of the METTL3-METTL14 complex) via intrathecal tubing. Validate UCD model by measuring spinal c-Fos expression and EMG. The levels of TRPA1 and p-NR2B were evaluated by qRT-PCR and western blot,and m6A level was detected by the kit. RNA Immunoprecipitation was adopted to determine the binding between TRPA1 and METTL14. Neurons were isolated from rat dorsal root ganglia (DRG), exposed to SAH treatment, and subsequently subjected to actinomycin D experiments.
Results
In the UCD model, cervical dilation causes an increase in EMG signal and spinal cord c-Fos expression. At the same time, the levels of TRPA1, p-NR2B, METTL14, and m6A increased in a stimulus intensity-dependent manner. Intrathecal SAH, a METTL3-METTL14 inhibitor, alleviated UCD-induced pain and reversed above indicators. Further investigation revealed that METTL14 binds to TRPA1, increasing TRPA1 mRNA stability via m6A modification.
Conclusion
METTL14 stabilizes TRPA1 through m6A modification, thereby promoting NR2B phosphorylation, culminating in acute visceral pain induced by UCD.
{"title":"METTL14-mediated m6A modification of TRPA1 promotes acute visceral pain induced by uterine cervical dilation by promoting NR2B phosphorylation","authors":"Guangfa Xia , Jing Qian , Yu Wang , Fei Xiao","doi":"10.1016/j.cellsig.2025.111610","DOIUrl":"10.1016/j.cellsig.2025.111610","url":null,"abstract":"<div><h3>Background</h3><div>While TRPA1 serves as a therapeutic target for nociceptive pain, its role in acute visceral pain induced by uterine cervical dilation (UCD) remains an enigma. This study aims to elucidate the upstream and downstream mechanisms of TRPA1 in the context of UCD-induced acute visceral pain.</div></div><div><h3>Methods</h3><div>The UCD rats were administered with SAH (inhibitor of the METTL3-METTL14 complex) via intrathecal tubing. Validate UCD model by measuring spinal c-Fos expression and EMG. The levels of TRPA1 and p-NR2B were evaluated by qRT-PCR and western blot,and m6A level was detected by the kit. RNA Immunoprecipitation was adopted to determine the binding between TRPA1 and METTL14. Neurons were isolated from rat dorsal root ganglia (DRG), exposed to SAH treatment, and subsequently subjected to actinomycin D experiments.</div></div><div><h3>Results</h3><div>In the UCD model, cervical dilation causes an increase in EMG signal and spinal cord c-Fos expression. At the same time, the levels of TRPA1, p-NR2B, METTL14, and m6A increased in a stimulus intensity-dependent manner. Intrathecal SAH, a METTL3-METTL14 inhibitor, alleviated UCD-induced pain and reversed above indicators. Further investigation revealed that METTL14 binds to TRPA1, increasing TRPA1 mRNA stability via m6A modification.</div></div><div><h3>Conclusion</h3><div>METTL14 stabilizes TRPA1 through m6A modification, thereby promoting NR2B phosphorylation, culminating in acute visceral pain induced by UCD.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111610"},"PeriodicalIF":4.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000995","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}
Allergic asthma is a chronic airway inflammatory reaction that seriously affects people's quality of life and even endangers their lives. The aim of this study was to explore the role of epicatechin (EC) on asthma and its potential mechanism. A mice model of allergic asthma was established by intraperitoneal injection of ovalbumin (OVA) with aluminum hydrogen solution, and nebulized inhalation of OVA to stimulate. EC (10, 20, 40 mg/kg) was administered 30 min before nebulization for three consecutive days. The results showed that EC attenuated OVA-induced lung injury, inflammatory cell infiltration, IgE, and inflammatory cytokine production. EC also inhibited OVA-induced NF-κB activation and increased Nrf2 and HO-1 expression. 16S rRNA sequencing analysis demonstrated that at genus level, EC significantly increased the abundance of Lachnospiraceae_NK4A136_group, Ligilactobacillus, Alloprevotella. Meanwhile, EC inhibited the abundance of Clostridia UCG-014, Helicobacter, Paramuribaculum, and Escherichia-Shigella. In conclusion, EC can effectively alleviate the symptoms of asthma in mice, which may through regulating the composition of gut microbiota and inhibiting inflammatory response.
{"title":"Epicatechin protects mice against OVA-induced asthma through inhibiting airway inflammation and modulating gut microbiota","authors":"Yang Zheng , Dengyu Gao , Hongyang Xie , Huafeng Geng","doi":"10.1016/j.cellsig.2025.111609","DOIUrl":"10.1016/j.cellsig.2025.111609","url":null,"abstract":"<div><div>Allergic asthma is a chronic airway inflammatory reaction that seriously affects people's quality of life and even endangers their lives. The aim of this study was to explore the role of epicatechin (EC) on asthma and its potential mechanism. A mice model of allergic asthma was established by intraperitoneal injection of ovalbumin (OVA) with aluminum hydrogen solution, and nebulized inhalation of OVA to stimulate. EC (10, 20, 40 mg/kg) was administered 30 min before nebulization for three consecutive days. The results showed that EC attenuated OVA-induced lung injury, inflammatory cell infiltration, IgE, and inflammatory cytokine production. EC also inhibited OVA-induced NF-κB activation and increased Nrf2 and HO-1 expression. 16S rRNA sequencing analysis demonstrated that at genus level, EC significantly increased the abundance of Lachnospiraceae_NK4A136_group, Ligilactobacillus, Alloprevotella. Meanwhile, EC inhibited the abundance of Clostridia UCG-014, Helicobacter, Paramuribaculum, and Escherichia-Shigella. In conclusion, EC can effectively alleviate the symptoms of asthma in mice, which may through regulating the composition of gut microbiota and inhibiting inflammatory response.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111609"},"PeriodicalIF":4.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000996","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 : 2025-01-16DOI: 10.1016/j.cellsig.2025.111604
Yining Liu , Zelei Yang , Nan Lin , Yanxin Liu , Huaxia Chen
Purpose
This study investigated the effects and related mechanisms of Vestigial-like family member 3 (VGLL3) on keloid fibroblast (KF) proliferation, apoptosis, collagen production, and glycolysis.
Methods
Western blot, qRT-PCR, and immunohistochemistry were used for determining VGLL3 expression. KF viability, proliferation, and apoptosis were assessed using CCK-8 assay, EdU assay, and flow cytometry. Changes in the protein expression levels of α-SMA, fibronectin, collagen I, and collagen III were examined utilizing western blotting. The pathways related to VGLL3 were analyzed using Gene Set Enrichment Analysis. Changes in glycolysis were assessed by measuring oxygen consumption rate (OCR), extracellular acidification rate (ECAR), glucose uptake, and lactate production. WNT2 and β-catenin protein levels were measured using western blotting.
Results
VGLL3 was upregulated in human keloid tissues. In KFs, overexpression of VGLL3 inhibited cell apoptosis, promoted cell proliferation and protein expression of α-SMA, fibronectin, collagen I, and collagen III. Moreover, it reduced OCR level, and increased the levels of ECAR, glucose uptake, and lactate production. On the other hand, the knockdown of VGLL3 had the opposite effect. WNT2 and β-catenin protein levels were enhanced by overexpression of VGLL3 and reduced by VGLL3 knockdown. Silencing of WNT2 reversed the effects of VGLL3 on apoptosis, proliferation, collagen production, and glycolysis in KFs.
Conclusions
VGLL3 promoted glycolysis in KFs and keloid progression, which was achieved through the activation of Wnt signaling pathway. Therefore, targeting VGLL3 may be a promising therapeutic strategy for the treatment of keloids.
{"title":"Highly expressed VGLL3 in keloid fibroblasts promotes glycolysis and collagen production via the activation of Wnt/β-catenin signaling","authors":"Yining Liu , Zelei Yang , Nan Lin , Yanxin Liu , Huaxia Chen","doi":"10.1016/j.cellsig.2025.111604","DOIUrl":"10.1016/j.cellsig.2025.111604","url":null,"abstract":"<div><h3>Purpose</h3><div>This study investigated the effects and related mechanisms of Vestigial-like family member 3 (VGLL3) on keloid fibroblast (KF) proliferation, apoptosis, collagen production, and glycolysis.</div></div><div><h3>Methods</h3><div>Western blot, qRT-PCR, and immunohistochemistry were used for determining VGLL3 expression. KF viability, proliferation, and apoptosis were assessed using CCK-8 assay, EdU assay, and flow cytometry. Changes in the protein expression levels of α-SMA, fibronectin, collagen I, and collagen III were examined utilizing western blotting. The pathways related to VGLL3 were analyzed using Gene Set Enrichment Analysis. Changes in glycolysis were assessed by measuring oxygen consumption rate (OCR), extracellular acidification rate (ECAR), glucose uptake, and lactate production. WNT2 and β-catenin protein levels were measured using western blotting.</div></div><div><h3>Results</h3><div>VGLL3 was upregulated in human keloid tissues. In KFs, overexpression of <em>VGLL3</em> inhibited cell apoptosis, promoted cell proliferation and protein expression of α-SMA, fibronectin, collagen I, and collagen III. Moreover, it reduced OCR level, and increased the levels of ECAR, glucose uptake, and lactate production. On the other hand, the knockdown of <em>VGLL3</em> had the opposite effect. WNT2 and β-catenin protein levels were enhanced by overexpression of <em>VGLL3</em> and reduced by <em>VGLL3</em> knockdown. Silencing of <em>WNT2</em> reversed the effects of VGLL3 on apoptosis, proliferation, collagen production, and glycolysis in KFs.</div></div><div><h3>Conclusions</h3><div>VGLL3 promoted glycolysis in KFs and keloid progression, which was achieved through the activation of Wnt signaling pathway. Therefore, targeting VGLL3 may be a promising therapeutic strategy for the treatment of keloids.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111604"},"PeriodicalIF":4.4,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000998","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 : 2025-01-14DOI: 10.1016/j.cellsig.2025.111608
Jian Lu , Feng Ding , Yongjie Sun , Yu Zhao , Wenbiao Ma , Huan Zhang , Bo Shi
This study utilizes single-cell RNA sequencing data to reveal the transcriptomic characteristics of breast cancer and normal epithelial cells. Nine significant cell populations were identified through stringent quality control and batch effect correction. Further classification of breast cancer epithelial cells based on the PAM50 method and clinical subtypes highlighted significant heterogeneity between triple-negative breast cancer (TNBC) and non-triple-negative breast cancer (NTNBC). The study also analyzed myeloid cells and tumor-infiltrating lymphocytes (TILs) within the breast cancer immune microenvironment, identifying 14 TIL subpopulations and assessing their proportion variations across different patients. The CellChat tool revealed a complex cellular communication network within the tumor microenvironment, showing notable differences in communication intensity and patterns between TNBC and NTNBC patients. Additionally, the key regulatory role of the senescence-associated gene MDH1 in breast cancer was confirmed, and its impact on drug sensitivity was explored. Finally, it was discovered that the phytosterol Schottenol inhibits breast cancer cell proliferation by downregulating MDH1 expression and enhances sensitivity to paclitaxel. These findings provide new insights into MDH1 as a therapeutic target and suggest Schottenol as a potential strategy to overcome breast cancer drug resistance.
{"title":"Unveiling the role of MDH1 in breast cancer drug resistance through single-cell sequencing and schottenol intervention","authors":"Jian Lu , Feng Ding , Yongjie Sun , Yu Zhao , Wenbiao Ma , Huan Zhang , Bo Shi","doi":"10.1016/j.cellsig.2025.111608","DOIUrl":"10.1016/j.cellsig.2025.111608","url":null,"abstract":"<div><div>This study utilizes single-cell RNA sequencing data to reveal the transcriptomic characteristics of breast cancer and normal epithelial cells. Nine significant cell populations were identified through stringent quality control and batch effect correction. Further classification of breast cancer epithelial cells based on the PAM50 method and clinical subtypes highlighted significant heterogeneity between triple-negative breast cancer (TNBC) and non-triple-negative breast cancer (NTNBC). The study also analyzed myeloid cells and tumor-infiltrating lymphocytes (TILs) within the breast cancer immune microenvironment, identifying 14 TIL subpopulations and assessing their proportion variations across different patients. The CellChat tool revealed a complex cellular communication network within the tumor microenvironment, showing notable differences in communication intensity and patterns between TNBC and NTNBC patients. Additionally, the key regulatory role of the senescence-associated gene MDH1 in breast cancer was confirmed, and its impact on drug sensitivity was explored. Finally, it was discovered that the phytosterol Schottenol inhibits breast cancer cell proliferation by downregulating MDH1 expression and enhances sensitivity to paclitaxel. These findings provide new insights into MDH1 as a therapeutic target and suggest Schottenol as a potential strategy to overcome breast cancer drug resistance.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111608"},"PeriodicalIF":4.4,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143001000","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 : 2025-01-13DOI: 10.1016/j.cellsig.2025.111606
Ying Kong , Zongming Jia , Yizhang Sun , Lichen Jin , Tong Zhang , Qiya Xu , Yuhua Huang
Clear cell renal cell carcinoma (ccRCC) is a common clinical tumor of the urinary system. The lack of effective diagnostic and treatment options poses a serious challenge to clinical treatment. Therefore, identifying effective molecular targets has become one of the potential means to treat this disease. Firstly, the analysis of the TCGA database found that PLAC1 was abnormally highly expressed in ccRCC and was negatively correlated with patient prognosis. Western blotting and immunofluorescence experiments further verified that PLAC1 was highly expressed in ccRCC patients, and knockdown of PLAC1 inhibited the development of ccRCC in vitro. Last, high-throughput virtual screening technology (HTVS) was performed to identify two molecular inhibitors ,AmB and Cana, which were able to reduce the expression of PLAC1 and inhibited the progression of ccRCC. In conclusion, the current investigation indicated that the PLAC1 could serve as a prognostic biomarker, and AmB and Cana inhibit the progression of ccRCC by reducing PLAC1, making it a potential therapeutic option for ccRCC.
{"title":"Identification of PLAC1 as a prognostic biomarker and molecular target in clear cell renal cell carcinoma","authors":"Ying Kong , Zongming Jia , Yizhang Sun , Lichen Jin , Tong Zhang , Qiya Xu , Yuhua Huang","doi":"10.1016/j.cellsig.2025.111606","DOIUrl":"10.1016/j.cellsig.2025.111606","url":null,"abstract":"<div><div>Clear cell renal cell carcinoma (ccRCC) is a common clinical tumor of the urinary system. The lack of effective diagnostic and treatment options poses a serious challenge to clinical treatment. Therefore, identifying effective molecular targets has become one of the potential means to treat this disease. Firstly, the analysis of the TCGA database found that PLAC1 was abnormally highly expressed in ccRCC and was negatively correlated with patient prognosis. Western blotting and immunofluorescence experiments further verified that PLAC1 was highly expressed in ccRCC patients, and knockdown of PLAC1 inhibited the development of ccRCC <em>in vitro</em>. Last, high-throughput virtual screening technology (HTVS) was performed to identify two molecular inhibitors ,AmB and Cana, which were able to reduce the expression of PLAC1 and inhibited the progression of ccRCC. In conclusion, the current investigation indicated that the PLAC1 could serve as a prognostic biomarker, and AmB and Cana inhibit the progression of ccRCC by reducing PLAC1, making it a potential therapeutic option for ccRCC.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111606"},"PeriodicalIF":4.4,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-13DOI: 10.1016/j.cellsig.2025.111602
Bin Li , Guohu Di , Huanhuan Ge , Peirong Song , Wenshuo Han , Hetong Sun , Dianqiang Wang , Peng Chen , Ye Wang
During the proliferative phase of liver regeneration, insufficient regulation of hepatocyte hydrogen peroxide (H2O2) overproduction can result in oxidative stress and hepatocyte death. This study aims to investigate the influence of Aquaporin 5 (Aqp5) on liver regeneration by evaluating its role in reactive oxygen species (ROS) generation and NLRP3-GSDMD-mediated pyroptosis. A 70 % partial hepatectomy (PHx) model was established in Aqp5−/− mice to evaluate the pathological changes in the liver. Reactive oxygen species (ROS) production was assessed using a dichlorodihydrofluorescein diacetate (DCFH-DA) assay. Aqp5 deficiency significantly increased ROS production, the number of TUNEL-positive cells, and disrupted mitochondrial membrane potential in the liver of Aqp5-deficient mice. The impact of Aqp5 on ROS/NLRP3/Gasdermin-D (GSDMD)-mediated pyroptosis was examined through the administration of N-acetyl-L-cysteine (NAC, an ROS scavenger) or disulfiram (DSF, a GSDMD inhibitor). In Aqp5-deficient mice, the regenerative liver exhibited increased expression of NLRP3, enhanced activation of caspase-1 and GSDMD, as well as elevated secretion of IL-1β. Treatment with DSF significantly attenuated GSDMD-mediated pyroptosis triggered by Aqp5 deficiency in the regenerating liver. Furthermore, the administration of NAC to Aqp5-deficient mice resulted in a reduction in the expression levels of NLRP3, the activity levels of caspase-1 and GSDMD, as well as the release of IL-1β. Our findings indicate that the deficiency of Aqp5 facilitates GSDMD activation through the production of ROS. The suppression of ROS or inhibition of GSDMD significantly alleviates the damage and pyroptosis observed in Aqp5-deficient regenerative liver.
{"title":"Aquaporin-5 facilitates liver regeneration following hepatectomy via ROS/GSDMD pathway","authors":"Bin Li , Guohu Di , Huanhuan Ge , Peirong Song , Wenshuo Han , Hetong Sun , Dianqiang Wang , Peng Chen , Ye Wang","doi":"10.1016/j.cellsig.2025.111602","DOIUrl":"10.1016/j.cellsig.2025.111602","url":null,"abstract":"<div><div>During the proliferative phase of liver regeneration, insufficient regulation of hepatocyte hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) overproduction can result in oxidative stress and hepatocyte death. This study aims to investigate the influence of Aquaporin 5 (Aqp5) on liver regeneration by evaluating its role in reactive oxygen species (ROS) generation and NLRP3-GSDMD-mediated pyroptosis. A 70 % partial hepatectomy (PHx) model was established in Aqp5<sup>−/−</sup> mice to evaluate the pathological changes in the liver. Reactive oxygen species (ROS) production was assessed using a dichlorodihydrofluorescein diacetate (DCFH-DA) assay. Aqp5 deficiency significantly increased ROS production, the number of TUNEL-positive cells, and disrupted mitochondrial membrane potential in the liver of Aqp5-deficient mice. The impact of Aqp5 on ROS/NLRP3/Gasdermin-D (GSDMD)-mediated pyroptosis was examined through the administration of <em>N</em>-acetyl-L-cysteine (NAC, an ROS scavenger) or disulfiram (DSF, a GSDMD inhibitor). In Aqp5-deficient mice, the regenerative liver exhibited increased expression of NLRP3, enhanced activation of caspase-1 and GSDMD, as well as elevated secretion of IL-1β. Treatment with DSF significantly attenuated GSDMD-mediated pyroptosis triggered by Aqp5 deficiency in the regenerating liver. Furthermore, the administration of NAC to Aqp5-deficient mice resulted in a reduction in the expression levels of NLRP3, the activity levels of caspase-1 and GSDMD, as well as the release of IL-1β. Our findings indicate that the deficiency of Aqp5 facilitates GSDMD activation through the production of ROS. The suppression of ROS or inhibition of GSDMD significantly alleviates the damage and pyroptosis observed in Aqp5-deficient regenerative liver.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111602"},"PeriodicalIF":4.4,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000991","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 : 2025-01-13DOI: 10.1016/j.cellsig.2025.111599
Zucheng Luo , Shaoluan Zheng , Jiaqi Liu , Fazhi Qi
{"title":"Corrigendum to “The role of α7nAchR and PD-L1 in neuroimmune regulation of keloid treatment” [Cellular Signalling, Volume 121 (2024), Article 111275]","authors":"Zucheng Luo , Shaoluan Zheng , Jiaqi Liu , Fazhi Qi","doi":"10.1016/j.cellsig.2025.111599","DOIUrl":"10.1016/j.cellsig.2025.111599","url":null,"abstract":"","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111599"},"PeriodicalIF":4.4,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982241","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 : 2025-01-11DOI: 10.1016/j.cellsig.2025.111603
Fengyi Deng , Ping Zhang , Huaiyun Li , Xingyu Fan , Yijun Du , Xing Zhong , Nuojin Wang , Meiwen He , Yue Wang , Tianrong Pan
Diabetic kidney disease (DKD), a microvascular complication of diabetes mellitus, represents a significant clinical challenge. This study investigated the reno-protective effects of dulaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1 RA) widely used in the management of diabetes, and aimed to elucidate its underlying mechanisms. Mice with db/db and db/m genotypes were allocated into four experimental groups and treated with either dulaglutide or a saline control for 10 weeks. Following the treatment period, biological samples were collected for comprehensive analysis. Serum and urinary creatinine levels were measured using a creatinine assay, while urinary protein concentrations were quantified via ELISA. Histopathological kidney damage was assessed through hematoxylin and eosin (HE) staining, with glomerular lesions evaluated using periodic acid-Schiff (PAS) staining. Inflammatory markers, ferroptosis-related indicators, and fibrosis in kidney tissues were further analyzed through PCR, Western blot (WB), immunohistochemistry (IHC), and transmission electron microscopy (TEM). Consistent with prior findings, this research demonstrated that dulaglutide improves renal function and mitigates pathological kidney damage in db/db mice. Treatment with dulaglutide significantly reduced mRNA expression of ferroptosis-related markers, including ACSL4, SLC7A11, and Ptgs2, alongside a decrease in 4-HNE levels in kidney tissues. Furthermore, dulaglutide downregulated ACSL4 protein levels and upregulated GPX4 protein expression, thereby ameliorating mitochondrial damage in renal tubular cells. In addition to these effects, dulaglutide alleviated kidney inflammation and fibrosis in db/db mice, with concomitant suppression of P-STAT3 and P-ERK expression. Collectively, these findings underscore dulaglutide's reno-protective effects in DKD, mediated through the inhibition of inflammation, improvement in renal fibrosis and ferroptosis, and modulation of P-STAT3 and P-ERK signaling pathways.
糖尿病肾病(DKD)是糖尿病的微血管并发症,是一个重大的临床挑战。本研究探讨了胰高血糖素样肽-1受体激动剂(GLP-1 RA)在糖尿病治疗中广泛应用的肾保护作用,并旨在阐明其潜在机制。将db/db和db/m基因型小鼠分为4个实验组,分别给予杜拉鲁肽或生理盐水对照治疗10 周。治疗期结束后,采集生物样本进行综合分析。采用肌酐测定法测定血清和尿肌酐水平,通过ELISA测定尿蛋白浓度。组织病理学肾损害通过苏木精和伊红(HE)染色评估,肾小球病变采用周期性酸-希夫(PAS)染色评估。通过PCR、Western blot (WB)、免疫组化(IHC)、透射电镜(TEM)等方法进一步分析肾脏组织的炎症标志物、凋亡相关指标及纤维化情况。与先前的研究结果一致,本研究表明杜拉鲁肽可以改善db/db小鼠的肾功能并减轻病理性肾损伤。杜拉鲁肽治疗显著降低了铁中毒相关标志物的mRNA表达,包括ACSL4、SLC7A11和Ptgs2,同时降低了肾组织中4-HNE的水平。此外,杜拉鲁肽下调ACSL4蛋白水平,上调GPX4蛋白表达,从而改善肾小管细胞线粒体损伤。除了这些作用外,杜拉鲁肽还能减轻db/db小鼠的肾脏炎症和纤维化,同时抑制P-STAT3和P-ERK的表达。总的来说,这些发现强调了杜拉鲁肽在DKD中的肾保护作用,通过抑制炎症、改善肾纤维化和铁下垂以及调节P-STAT3和P-ERK信号通路来介导。
{"title":"Effect of the glucagon-like peptide-1 receptor agonists dulaglutide on kidney outcomes in db/db mice","authors":"Fengyi Deng , Ping Zhang , Huaiyun Li , Xingyu Fan , Yijun Du , Xing Zhong , Nuojin Wang , Meiwen He , Yue Wang , Tianrong Pan","doi":"10.1016/j.cellsig.2025.111603","DOIUrl":"10.1016/j.cellsig.2025.111603","url":null,"abstract":"<div><div>Diabetic kidney disease (DKD), a microvascular complication of diabetes mellitus, represents a significant clinical challenge. This study investigated the reno-protective effects of dulaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1 RA) widely used in the management of diabetes, and aimed to elucidate its underlying mechanisms. Mice with db/db and db/m genotypes were allocated into four experimental groups and treated with either dulaglutide or a saline control for 10 weeks. Following the treatment period, biological samples were collected for comprehensive analysis. Serum and urinary creatinine levels were measured using a creatinine assay, while urinary protein concentrations were quantified via ELISA. Histopathological kidney damage was assessed through hematoxylin and eosin (HE) staining, with glomerular lesions evaluated using periodic acid-Schiff (PAS) staining. Inflammatory markers, ferroptosis-related indicators, and fibrosis in kidney tissues were further analyzed through PCR, Western blot (WB), immunohistochemistry (IHC), and transmission electron microscopy (TEM). Consistent with prior findings, this research demonstrated that dulaglutide improves renal function and mitigates pathological kidney damage in db/db mice. Treatment with dulaglutide significantly reduced mRNA expression of ferroptosis-related markers, including ACSL4, SLC7A11, and Ptgs2, alongside a decrease in 4-HNE levels in kidney tissues. Furthermore, dulaglutide downregulated ACSL4 protein levels and upregulated GPX4 protein expression, thereby ameliorating mitochondrial damage in renal tubular cells. In addition to these effects, dulaglutide alleviated kidney inflammation and fibrosis in db/db mice, with concomitant suppression of P-STAT3 and P-ERK expression. Collectively, these findings underscore dulaglutide's reno-protective effects in DKD, mediated through the inhibition of inflammation, improvement in renal fibrosis and ferroptosis, and modulation of P-STAT3 and P-ERK signaling pathways.</div></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":"127 ","pages":"Article 111603"},"PeriodicalIF":4.4,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142977881","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}