Cell Communication and Signaling最新文献

筛选
英文 中文
Comparative evaluation of methods for isolating extracellular vesicles from ICC cell culture supernatants: Insights into proteomic and glycomic analysis. 从ICC细胞培养上清液中分离细胞外囊泡方法的比较评价:蛋白质组学和糖组学分析的见解。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-29 DOI: 10.1186/s12964-025-02207-x
Linlin Wu, Jiao Wei, Yueping Zhan, Xiao Xiao, Xuewen Xu, Chenjun Huang, Tian Long, Yueyue Li, Bin Fu, Mengmeng Wang, Chunfang Gao
{"title":"Comparative evaluation of methods for isolating extracellular vesicles from ICC cell culture supernatants: Insights into proteomic and glycomic analysis.","authors":"Linlin Wu, Jiao Wei, Yueping Zhan, Xiao Xiao, Xuewen Xu, Chenjun Huang, Tian Long, Yueyue Li, Bin Fu, Mengmeng Wang, Chunfang Gao","doi":"10.1186/s12964-025-02207-x","DOIUrl":"https://doi.org/10.1186/s12964-025-02207-x","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs) are nanoscale structures involved in intercellular communication and play a key role in cancer pathology. Intrahepatic cholangiocarcinoma (ICC) is a highly invasive malignancy marked by abnormal sialylated glycosylation. Analyzing proteins and glycans in EVs provides insights into ICC molecular subtyping and mechanisms. Optimizing EV isolation methods for ICC-derived EVs enables comprehensive proteomic and glycomic analysis.</p><p><strong>Methods: </strong>We systematically evaluated five EV isolation methods-Ultracentrifugation (UC), exoEasy, Total Exosome Isolation (TEI), EVtrap, and ÄKTA-by analyzing the biophysical properties, proteomic profiles, and glycomic structures of EVs. Subsequently, we applied TMT-based quantitative proteome and light/heavy methylamine labeling for the quantification of sialylated N-glycan linkage isomers to investigate alterations in proteins and N-glycans within EVs secreted by HuCCT1 and HCCC-9810 cells with overexpressing ST6 β‑galactoside α2,6‑sialyltransferase 1 (ST6GAL1).</p><p><strong>Results: </strong>By evaluating the biophysical properties, proteome, and N-glycome of EVs extracted using five different methods, UC was identified as the optimal approach for this study, as it offered a balance between operational complexity, cost-effectiveness, and the preservation of EVs activity. In this study, a total of 1,928 high-confidence proteins and over 84 high-confidence glycans were quantified. EVs secreted by HuCCT1 and HCCC-9810 cells overexpressing ST6GAL1 exhibited consistent upregulation of 16 proteins, consistent downregulation of 10 proteins, as well as consistent upregulation of 3 glycans and consistent downregulation of 3 glycans.</p><p><strong>Conclusions: </strong>Quantitative proteomic and glycomic analysis of ICC-derived EVs revealed that ST6GAL1 overexpression led to significant alterations in proteins involved in cancer cell adhesion and glycosylation pathways, along with specific changes in N-glycan structures. Notably, these modifications extended beyond α2,6-sialylation, suggesting that interactions between glycosyltransferases and glycans may drive these alterations.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"207"},"PeriodicalIF":8.2,"publicationDate":"2025-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12042569/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144043013","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}
引用次数: 0
Inhibition of adenosine/A2A receptor signaling suppresses dermal fibrosis by enhancing fatty acid oxidation. 抑制腺苷/A2A受体信号通过增强脂肪酸氧化抑制真皮纤维化。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-29 DOI: 10.1186/s12964-025-02210-2
Xiaoye Zhang, Jinjian Sun, Jia Guo, Xiaoru Hu, Junyu Zhou, Xiaoyun Xie, Xiang Chen, Hui Luo, Hong Liu, Yuzi Tian
{"title":"Inhibition of adenosine/A2A receptor signaling suppresses dermal fibrosis by enhancing fatty acid oxidation.","authors":"Xiaoye Zhang, Jinjian Sun, Jia Guo, Xiaoru Hu, Junyu Zhou, Xiaoyun Xie, Xiang Chen, Hui Luo, Hong Liu, Yuzi Tian","doi":"10.1186/s12964-025-02210-2","DOIUrl":"https://doi.org/10.1186/s12964-025-02210-2","url":null,"abstract":"<p><strong>Background: </strong>Skin fibrosis presents a major challenge for clinicians treating conditions like systemic sclerosis (SSc) due to its progressive course and limited treatment options. While the role of metabolism in fibrosis has gained increasing attention, the crucial alterations of metabolic pathway and the underlying signaling of metabolic interconnections in regulating SSc-related skin fibrosis remain largely elusive.</p><p><strong>Methods: </strong>Metabolomic analysis was performed on plasma samples from 35 SSc patients to identify metabolic alterations. In bleomycin (BLM)- and hypochlorous acid (HOCL)-induced skin fibrosis mouse models, we assessed the impact of global A2a receptor knockout on skin fibrosis. Single-cell RNA sequencing of mouse skin was utilized to investigate the role of A2A in fibroblasts during fibrotic challenge. Human dermal fibroblasts were used in in vitro experiments, employing RNA sequencing and Seahorse assays, to assess the relationship between A2A signaling and fatty acid oxidation (FAO). Finally, fibroblast-specific conditional A2a knockout mice were used to test the effects of specifically targeting A2A in dermal fibroblasts.</p><p><strong>Results: </strong>Adenosine-centered nucleotide metabolism was elevated in the plasma of SSc patients. Mechanistically, by stimulating dermal fibroblasts with key pathogenic cytokines associated with SSc, we observed significant changes in adenosine receptor A2A expression in response to IL-1β. Immunofluorescence revealed upregulation of A2A expression in dermal fibroblasts of SSc patients. Further, global A2a knockout significantly attenuated skin fibrosis in both BLM- and HOCL-induced skin fibrosis mouse models. Single-cell RNA sequencing of mouse skin revealed significant alterations in fatty acid metabolism in fibroblasts from A2a-deficient mice following fibrotic challenge. RNA sequencing, Seahorse assays and in vitro experiments showed that A2A inhibition promotes FAO by upregulating CPT1A expression via suppressing CREB phosphorylation, alleviating fibrosis in human primary dermal fibroblasts. Furthermore, targeted intervention of A2a specifically in fibroblasts improves outcomes and increases CPT1A expression in BLM-induced skin fibrosis mouse model.</p><p><strong>Conclusion: </strong>Our study highlights the crucial interplay between adenosine metabolism-A2A receptor axis and FAO in SSc-associated skin fibrosis, suggesting that targeting the adenosine receptor A2A-FAO metabolic axis offers a promising therapeutic strategy for skin fibrosis.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"206"},"PeriodicalIF":8.2,"publicationDate":"2025-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12042438/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144033668","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}
引用次数: 0
LAMA4+ CD90+ eCAFs provide immunosuppressive microenvironment for liver cancer through induction of CD8+ T cell senescence. LAMA4+ CD90+ eCAFs通过诱导CD8+ T细胞衰老为肝癌提供免疫抑制微环境。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-28 DOI: 10.1186/s12964-025-02162-7
Jianlei Zhang, Zhihui Li, Qiong Zhang, Wen Ma, Weina Fan, Jing Dong, Jingjie Tian, Hongfan Liao, Junzhe Guo, Yabing Cao, Jiang Yin, Guopei Zheng, Nan Li
{"title":"LAMA4<sup>+</sup> CD90<sup>+</sup> eCAFs provide immunosuppressive microenvironment for liver cancer through induction of CD8<sup>+</sup> T cell senescence.","authors":"Jianlei Zhang, Zhihui Li, Qiong Zhang, Wen Ma, Weina Fan, Jing Dong, Jingjie Tian, Hongfan Liao, Junzhe Guo, Yabing Cao, Jiang Yin, Guopei Zheng, Nan Li","doi":"10.1186/s12964-025-02162-7","DOIUrl":"https://doi.org/10.1186/s12964-025-02162-7","url":null,"abstract":"<p><p>Despite significant advances in cancer biology research and treatment, clinical outcomes for patients with liver cancer remain unsatisfactory. The biological and molecular mechanisms underlying the bidirectional signaling between tumor cells and the tumor microenvironment (TME), which promotes tumor progression in the liver, remain to be elucidated. Fibroblasts are crucial regulators of tumor progression and response to therapy; however, our understanding of their roles remains limited. Here, we integrated single-cell RNA sequencing and spatial transcriptomic data of pan-liver cancers to characterize the different subtypes of cancer-associated fibroblasts (CAFs). siRNA transfection was used for knockdown the expression of LAMA4. Western blot assay was used for gene expression analysis. Flow cytometry was used to detect proliferation, toxicity and cytolytic capacity of CD8<sup>+</sup> T cells. To establish a spontaneous murine hepatocellular carcinoma (HCC) model, a combined DEN and CCL4 approach was performed. Notably, we identified CD90<sup>+</sup> extracellular matrix CAFs (eCAFs) associated with poor prognosis. These CD90<sup>+</sup> eCAFs, located distal to the tumor nest, overlapped with the distribution of CD8<sup>+</sup> T cells. Functional experiments demonstrated that CD90<sup>+</sup> eCAFs recruited CD8<sup>+</sup> T cells and inhibited their function through secretion of LAMA4. Further investigation revealed that LAMA4 induced the CD8<sup>+</sup> T cell senescence through a DNA damage signaling pathway mediated by the receptor ITGA6. In a mouse model of spontaneous HCC, targeting LAMA4 can inhibit the progression of malignant transformation and synergize with anti-PD-1 therapy. Our study reveals the function of specific CAFs subtypes and highlights the importance of interactions with the immune system.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"203"},"PeriodicalIF":8.2,"publicationDate":"2025-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12036274/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144054532","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}
引用次数: 0
ERK2-mediated phosphorylation of ZEB1 at S322 enhances PD-L1 expression and EMT, leading to pancreatic cancer progression. erk2介导的ZEB1在S322位点的磷酸化增强了PD-L1表达和EMT,导致胰腺癌进展。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-28 DOI: 10.1186/s12964-025-02182-3
Mi Kyung Park, Hye Ja Lee, Jee Young Sung, Hyun Jung Byun, Hyun Ji Kim, Eun Ji Kim, Tuan Minh Nguyen, Gyeoung Jin Kang, Seung Hyun Oh, Jae Gal Shim, Ho Lee, Ki Taek Nam, Yong Yun Kim, Seung Bae Rho, Sang Gun Kim, Chang Hoon Lee
{"title":"ERK2-mediated phosphorylation of ZEB1 at S322 enhances PD-L1 expression and EMT, leading to pancreatic cancer progression.","authors":"Mi Kyung Park, Hye Ja Lee, Jee Young Sung, Hyun Jung Byun, Hyun Ji Kim, Eun Ji Kim, Tuan Minh Nguyen, Gyeoung Jin Kang, Seung Hyun Oh, Jae Gal Shim, Ho Lee, Ki Taek Nam, Yong Yun Kim, Seung Bae Rho, Sang Gun Kim, Chang Hoon Lee","doi":"10.1186/s12964-025-02182-3","DOIUrl":"https://doi.org/10.1186/s12964-025-02182-3","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic cancer is the fourth leading cause of cancer-related deaths. Epithelial-mesenchymal transition (EMT) drives aggressive behaviour and unfavourable outcomes in this disease. The zinc finger E-box-binding homeobox 1 (ZEB1) transcription factor is pivotal in orchestrating EMT, promoting tumor cell mobility, metastasis, and immune evasion through phosphorylation events. However, the precise mechanisms underlying individual phosphorylation sites and their relationship with ZEB1's functions in vivo remain inadequately understood.</p><p><strong>Methods: </strong>We assessed EMT using various techniques, including reverse transcription-quantitative polymerase chain reaction (RT-qPCR), immunoblotting, microscopy, migration, and invasion assays. ZEB1 knockdown was achieved via short hairpin RNA (shRNA), while plasmid transfection facilitated the overexpression of ZEB1, extracellular signal-regulated kinase 1 (ERK1), and extracellular signal-regulated kinase 2 (ERK2). Co-immunoprecipitation and kinase assays were used to examine the interaction between ZEB1 and ERK1/2. PANC-1 and HPAC cells were transplanted in an orthotopic mouse model for in vivo analysis.</p><p><strong>Results: </strong>Sphingosylphosphorylcholine (SPC) induced EMT in PANC-1 and HPAC cells in a dose- and time-dependent manner through the phosphorylation and nuclear translocation of ZEB1. Notably, ERK2 interacted with ZEB1 and catalysed the phosphorylation of serine 322 (S322) within the ZEB1 molecule. Disrupting S322 phosphorylation hindered ZEB1's nuclear translocation, leading to reduced programmed death-ligand 1 (PD-L1) expression and suppressed migration and invasion of pancreatic cancer cells. Furthermore, in an orthotopic mouse model, implantation of S322 phosphorylation-deficient (shZEB1/S322A) pancreatic cancer cells suppressed tumour formation and metastasis. We developed a phosphoS322 detection antibody, which validated ZEB1 phosphorylation in pancreatic cancer cells and tissue samples from patients with pancreatic cancer.</p><p><strong>Conclusion: </strong>SPC induces ZEB1 phosphorylation, with ERK2, rather than ERK1, targeting the S322 site. Inhibiting S322 phosphorylation mitigates EMT, PD-L1 expression, and progression of pancreatic cancer. The phosphoS322 detection antibody might be a valuable tool for predicting pancreatic cancer prognosis. These findings implicate ERK2 as a potential therapeutic target for pancreatic cancer and highlight phosphoZEB1 as a valuable prognostic marker.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"204"},"PeriodicalIF":8.2,"publicationDate":"2025-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12036305/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144060896","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}
引用次数: 0
Correction: A network-based approach to overcome BCR::ABL1-independent resistance in chronic myeloid leukemia. 纠正:一种基于网络的方法来克服慢性髓系白血病中BCR:: abl1非依赖性耐药。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-28 DOI: 10.1186/s12964-025-02206-y
Valeria Bica, Veronica Venafra, Giorgia Massacci, Simone Graziosi, Sara Gualdi, Gessica Minnella, Federica Sorà, Patrizia Chiusolo, Maria Elsa Brunetti, Gennaro Napolitano, Massimo Breccia, Dimitrios Mougiakakos, Martin Böttcher, Thomas Fischer, Livia Perfetto, Francesca Sacco
{"title":"Correction: A network-based approach to overcome BCR::ABL1-independent resistance in chronic myeloid leukemia.","authors":"Valeria Bica, Veronica Venafra, Giorgia Massacci, Simone Graziosi, Sara Gualdi, Gessica Minnella, Federica Sorà, Patrizia Chiusolo, Maria Elsa Brunetti, Gennaro Napolitano, Massimo Breccia, Dimitrios Mougiakakos, Martin Böttcher, Thomas Fischer, Livia Perfetto, Francesca Sacco","doi":"10.1186/s12964-025-02206-y","DOIUrl":"https://doi.org/10.1186/s12964-025-02206-y","url":null,"abstract":"","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"205"},"PeriodicalIF":8.2,"publicationDate":"2025-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12036233/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144058406","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}
引用次数: 0
N-terminal truncation of STAT1 transcription factor causes CD3- and CD20-negative non-Hodgkin lymphoma through upregulation of STAT3-mediated oncogenic functions. STAT1转录因子n端截断通过上调stat3介导的致癌功能导致CD3-和cd20阴性非霍奇金淋巴瘤。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-26 DOI: 10.1186/s12964-025-02183-2
Sana Mumtaz Sheikh, Julia Staab, Martina Bleyer, Aleksandar Ivetic, Fred Lühder, Oliver Wirths, Thomas Meyer
{"title":"N-terminal truncation of STAT1 transcription factor causes CD3- and CD20-negative non-Hodgkin lymphoma through upregulation of STAT3-mediated oncogenic functions.","authors":"Sana Mumtaz Sheikh, Julia Staab, Martina Bleyer, Aleksandar Ivetic, Fred Lühder, Oliver Wirths, Thomas Meyer","doi":"10.1186/s12964-025-02183-2","DOIUrl":"https://doi.org/10.1186/s12964-025-02183-2","url":null,"abstract":"<p><p>The cytokine-driven transcription factor STAT1 (signal transducer and activator of transcription 1) executes anti-microbial and pro-apoptotic functions, and loss-of-function mutations are associated with increased susceptibility to various infections and the development of tumors. A targeted mutation in mice expressing an N-terminally truncated STAT1 protein (STAT1-ΔN) typically develops splenomegaly in animals older than 6 months due to the formation of splenic non-Hodgkin lymphomas. The expression of the STAT1-ΔN variant resulted in the disruption of normal spleen architecture by malignant CD3- and CD20-negative tumor cells, which stained positively for both tyrosine-phosphorylated STAT1 and STAT3. Immunoblotting of lysates from isolated tumor cells revealed the cytokine-independent hyperphosphorylation of both STAT proteins, whereas the expression level of NF-κB was significantly reduced. Gel-shift assays showed that the DNA-binding activity of STAT1-ΔN was increased compared to the wild-type protein. This elevated level of tyrosine-phosphorylated STAT1-ΔN did not further increase upon stimulation of isolated tumor cells with either interferon-γ (IFNγ), lipopolysaccharide (LPS), or the combination of both. Since the truncation mutant was unable to accumulate in the nucleus upon cytokine stimulation, real-time PCR data from tumor tissue as well as from isolated, IFNγ/LPS-treated lymphoma cells demonstrated significantly reduced STAT1-regulated target gene expression despite its observed hyperphosphorylation. The nuclear import defect of tyrosine-phosphorylated STAT1-ΔN was associated with an elevated tyrosine-phosphorylation level of its antagonistic homolog STAT3, which is a known oncogene. These data demonstrate that the lack of STAT1 nuclear accumulation interferes with the functional balance between the two STAT proteins and, thereby, promotes the formation of phospho-STAT3-expressing CD3<sup>-/-</sup> CD20<sup>-/-</sup> non-Hodgkin lymphomas in the spleens of the diseased animals.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"201"},"PeriodicalIF":8.2,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12034123/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143998164","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}
引用次数: 0
ER-phagy mediates the anti-tumoral synergism between HDAC inhibition and chemotherapy. er吞噬介导HDAC抑制与化疗之间的抗肿瘤协同作用。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-26 DOI: 10.1186/s12964-025-02198-9
Felix J Gössl, Pierfrancesco Polo, Frederik Helmprobst, André Menzenbach, Alexander Visekruna, Thomas M Gress, Till Adhikary, Matthias Lauth
{"title":"ER-phagy mediates the anti-tumoral synergism between HDAC inhibition and chemotherapy.","authors":"Felix J Gössl, Pierfrancesco Polo, Frederik Helmprobst, André Menzenbach, Alexander Visekruna, Thomas M Gress, Till Adhikary, Matthias Lauth","doi":"10.1186/s12964-025-02198-9","DOIUrl":"https://doi.org/10.1186/s12964-025-02198-9","url":null,"abstract":"<p><strong>Background: </strong>Histone deacetylase inhibitors (HDACi) are clinically approved drugs for the treatment of hematological malignancies synergizing with chemotherapy. However, despite the long history of HDACi, the mechanistic underpinnings of this synergism have remained unclear.</p><p><strong>Methods: </strong>Using transmission electron microscopy, we identified autophagy and ER-stress in HDACi-treated cells. We quantified ER-phagy and ER-stress with reporter systems by using 3D-deconvolution microscopy and flow cytometry. We complemented these data with qPCR and Western blot results. Apoptosis rates were assessed using a caspase assay and flow cytometry, and large public datasets were utilized.</p><p><strong>Results: </strong>HDAC blockade results in specific upregulation of the selective autophagy receptor FAM134B (RETREG1) and the induction of ER-phagy. Combined with the chemotherapeutic drug Gemcitabine, this results in subsequent elevated ER-stress levels and apoptosis. Inhibiting the distinct ER-stress branches fully rescues this process. Broadening the scope of these findings, certain non-HDAC-inhibitory and clinically approved compounds like Loperamide and Nelfinavir are able to induce FAM134B and could hence constitute novel Gemcitabine-synergizing molecules. Additionally, pancreatic cancer patients with high FAM134B expression have significantly longer survival rates under chemotherapy.</p><p><strong>Conclusion: </strong>In summary, we provide mechanistic evidence for ER-phagy playing a hitherto unknown central role in the clinical synergy between HDACi and chemotherapy.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"202"},"PeriodicalIF":8.2,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12034116/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144053394","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}
引用次数: 0
The Roxadustat (FG-4592) ameliorates tubulointerstitial fibrosis by promoting intact FGF23 cleavage. Roxadustat (FG-4592)通过促进完整的FGF23切割改善小管间质纤维化。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-25 DOI: 10.1186/s12964-025-02175-2
Jing Wang, Zuo-Lin Li, Yan Zhou, Zhong-Tang Li, Yan Tu, Xin-Hui Hu, Jin-Hua Zhu, Bi-Cheng Liu, Hong Liu
{"title":"The Roxadustat (FG-4592) ameliorates tubulointerstitial fibrosis by promoting intact FGF23 cleavage.","authors":"Jing Wang, Zuo-Lin Li, Yan Zhou, Zhong-Tang Li, Yan Tu, Xin-Hui Hu, Jin-Hua Zhu, Bi-Cheng Liu, Hong Liu","doi":"10.1186/s12964-025-02175-2","DOIUrl":"https://doi.org/10.1186/s12964-025-02175-2","url":null,"abstract":"<p><strong>Background: </strong>Hypoxia-inducible factor prolyl hydroxylase inhibitor (HIF-PHI) represents a novel therapeutic approach for renal anemia, a prevalent complication of chronic kidney disease (CKD). However, the effects of HIF-PHI on renal functional outcomes remain poorly characterized. Here, the potential effects of FG-4592, an orally administered HIF-PHI, on renal fibrosis were explored systematically.</p><p><strong>Methods: </strong>In this study, a CKD rat model was established through subtotal 5/6 nephrectomy. Rats were administered either FG-4592 or vehicle control via oral gavage three times weekly for 12 consecutive weeks. Additionally, recombinant FGF23 was continuously delivered via subcutaneously implanted Alzet osmotic minipumps for 28 days.</p><p><strong>Results: </strong>Interestingly, we found that CKD-induced anemia was significantly ameliorated in CKD rats with FG-4592 treatment. Meanwhile, markedly alleviated histopathological changes and renal tubulointerstitial fibrosis (TIF) were observed in rats with FG-4592 administration. Notably, serum levels of intact FGF23 (iFGF23) were significantly reduced following FG-4592 administration in CKD rats. This finding was subsequently validated in CKD patients receiving Roxadustat therapy. Mechanistically, we illustrated that inhibition of the iFGF23-WNT5A pathway was the exact mechanism by which FG-4592 ameliorated TIF. Further, we also demonstrated that transcriptional activation of Furin enzyme was the exact molecular mechanism for FG-4592-mediated iFGF23 cleavage.</p><p><strong>Conclusions: </strong>FG-4592 attenuates TIF through Furin-mediated proteolytic cleavage of iFGF23. These findings provide novel mechanistic insights into HIF-PHI-mediated renal protection and establish a theoretical framework for clinical translation.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"200"},"PeriodicalIF":8.2,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12032739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144059050","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}
引用次数: 0
Epigenetic modulation of the NLRP6 inflammasome sensor as a therapeutic modality to reduce necroptosis-driven gastrointestinal mucosal dysfunction in HIV/SIV infection. NLRP6炎症小体传感器的表观遗传学调节作为一种治疗方式来减少HIV/SIV感染中坏死驱动的胃肠道粘膜功能障碍。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-25 DOI: 10.1186/s12964-025-02193-0
Lakmini S Premadasa, Marina McDew-White, Luis Romero, Beverly Gondo, Jade A Drawec, Binhua Ling, Chioma M Okeoma, Mahesh Mohan
{"title":"Epigenetic modulation of the NLRP6 inflammasome sensor as a therapeutic modality to reduce necroptosis-driven gastrointestinal mucosal dysfunction in HIV/SIV infection.","authors":"Lakmini S Premadasa, Marina McDew-White, Luis Romero, Beverly Gondo, Jade A Drawec, Binhua Ling, Chioma M Okeoma, Mahesh Mohan","doi":"10.1186/s12964-025-02193-0","DOIUrl":"https://doi.org/10.1186/s12964-025-02193-0","url":null,"abstract":"<p><strong>Background: </strong>Gastrointestinal (GI) disease/dysfunction persists in people living with HIV (PLWH) receiving suppressive combination anti-retroviral therapy (ART) leading to epithelial barrier breakdown, microbial translocation and systemic inflammation that can drive non-AIDS associated comorbidities. Although epigenetic mechanisms are predicted to drive GI dysfunction, they remain unknown and unaddressed in HIV/SIV infection. The present study investigated genome-wide changes in DNA methylation, and gene expression exclusively in colon epithelial cells (CE) in response to simian immunodeficiency virus infection (SIV) and long-term low-dose delta-9-tetrahydrocannabinol (THC).</p><p><strong>Methods: </strong>Using reduced-representation bisulfite sequencing, we characterized DNA methylation changes in colonic epithelium (CE) of uninfected controls (n=5) and SIV-infected rhesus macaques (RMs) administered vehicle (VEH/SIV; n=7) or THC (THC/SIV; n=6). Intact jejunum resection segments (~5cm) were collected from sixteen ART treated SIV-infected RMs [(VEH/SIV/ART; n=8) and (THC/SIV/ART; n=8)] to confirm protein expression data identified in the colon of ART-naïve SIV-infected RMs. Transcriptomics data was used to confirm expression of differentially methylated genes. Protein expression of differentially methylated genes and their downstream targets was assessed using Immunofluorescence followed by HALO quantification.</p><p><strong>Results: </strong>SIV infection in ART-naïve RMs induced marked hypomethylation throughout promoter-associated CpG islands (paCGIs) in genes related to inflammatory response (NLRP6, cGAS), cellular adhesion (PCDH17, CDH7) and proliferation (WIF1, SFRP1, TERT, and HAND2) in CEs. Moreover, low-dose THC reduced NLRP6 protein expression in CE by hypermethylating the NLRP6 paCGI and blocked polyI:C induced NLRP6 upregulation in vitro. In ART suppressed SIV-infected RMs, significant NLRP6 protein upregulation during acute infection was unaffected by long-term ART administration during chronic infection despite successful plasma and tissue viral suppression. In this group, NLRP6 protein upregulation was associated with significantly increased expression of necroptosis-driving proteins; phosphorylated-RIPK3(Ser199), phosphorylated-MLKL(Thr357/Ser358), and HMGB1. Most strikingly, adding ART to THC-treated SIV-infected RMs effectively reduced NLRP6 and necroptosis-driving protein expression to pre-infection levels.</p><p><strong>Conclusions: </strong>We conclude that DNA hypomethylation-assisted NLRP6 upregulation can lead to its constitutively high expression resulting in the activation of necroptosis signaling via the RIPK3/p-MLKL pathway that can eventually drive intestinal epithelial loss/death. From a clinical standpoint, low-dose phytocannabinoids in combination with ART could safely and successfully reduce/reverse persistent GI inflammatory responses via modulating DNA methylation.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"199"},"PeriodicalIF":8.2,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12023470/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144045194","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}
引用次数: 0
Neurodegenerative disease-associated microRNAs acting as signaling molecules modulate CNS neuron structure and viability. 神经退行性疾病相关的microrna作为信号分子调节中枢神经系统神经元的结构和活力。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2025-04-24 DOI: 10.1186/s12964-025-02199-8
Victor Kumbol, Andranik Ivanov, Hugo McGurran, Jutta Schüler, Yuanyuan Zhai, Katarzyna Ludwik, Lukas Hinkelmann, Mariam Brehm, Christina Krüger, Judit Küchler, Thomas Wallach, Markus Höltje, Dieter Beule, Harald Stachelscheid, Seija Lehnardt
{"title":"Neurodegenerative disease-associated microRNAs acting as signaling molecules modulate CNS neuron structure and viability.","authors":"Victor Kumbol, Andranik Ivanov, Hugo McGurran, Jutta Schüler, Yuanyuan Zhai, Katarzyna Ludwik, Lukas Hinkelmann, Mariam Brehm, Christina Krüger, Judit Küchler, Thomas Wallach, Markus Höltje, Dieter Beule, Harald Stachelscheid, Seija Lehnardt","doi":"10.1186/s12964-025-02199-8","DOIUrl":"https://doi.org/10.1186/s12964-025-02199-8","url":null,"abstract":"<p><strong>Background: </strong>Dysregulation of microRNA (miRNA) expression in the brain is a common feature of neurodegenerative diseases. Beyond their conventional role in regulating gene expression at the post-transcriptional level, certain miRNAs can act extracellularly as signaling molecules. Our study elucidates the identity of such miRNA species serving as ligands for membrane receptors expressed in central nervous system (CNS) neurons and the impact of such miRNAs on neurons in the context of neurodegenerative disease.</p><p><strong>Methods: </strong>We combined a machine learning approach with the analysis of disease-associated miRNA databases to predict Alzheimer's disease (AD)-associated miRNAs as potential signaling molecules for single-stranded RNA-sensing Toll-like receptors (TLRs) 7 and 8. TLR-expressing HEK-Blue reporter cells, primary murine microglia, and human THP-1 macrophages were used to validate the AD miRNAs as ligands for human and mouse TLR7 and/or TLR8. Interaction between mouse cortical neurons and extracellularly applied AD miRNAs was analyzed by live cell imaging and confocal microscopy. Transcriptome changes in cortical neurons exposed to AD miRNAs were assessed by RNAseq and RT-qPCR. The extracellular AD miRNAs' effects on CNS neuron structure were investigated in cell cultures of murine primary cortical neurons and iPSC-derived human cortical neurons by immunocytochemistry. We employed a mouse model of intrathecal injection to assess effects of AD miRNAs acting as signaling molecules on neurons in vivo.</p><p><strong>Results: </strong>We identified the AD-associated miRNAs miR-124-5p, miR-92a-1-5p, miR-9-5p, and miR-501-3p as novel endogenous ligands for TLR7 and/or TLR8. These miRNAs being extracellularly stable and active were taken up by murine cortical neurons via endocytosis and induced changes in neuronal inflammation-, proliferation-, and apoptosis-related gene expression. Exposure of both murine and human cortical neurons to the AD-associated miRNAs led to alterations of dendrite and axon structure, synapse protein expression, and cell viability in a sequence-dependent fashion. Extracellular introduction of the AD miRNAs into the cerebrospinal fluid of mice resulted in both changes in neuronal structure and synapses, and neuronal loss in the cerebral cortex. Most of the observed extracellular miRNA-induced effects on cortical neurons involved TLR7/8 signaling.</p><p><strong>Conclusion: </strong>Neurodegenerative disease-associated miRNAs in extracellular form act as signaling molecules for CNS neurons including human cortical neurons, thereby modulating their structure and viability.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"196"},"PeriodicalIF":8.2,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12020182/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144060425","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}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信