{"title":"FTO downregulation-mediated m6A modification resulting in enhanced hepatocellular carcinoma invasion.","authors":"Cheng Zhou, Yong Zhang, Shi-Ming Shi, Dan Yin, Xue-Dong Li, Ying-Hong Shi, Jian Zhou, Zheng Wang, Qing Chen","doi":"10.1186/s13578-025-01395-w","DOIUrl":"https://doi.org/10.1186/s13578-025-01395-w","url":null,"abstract":"<p><strong>Background: </strong>Dysregulation of N6-methyladenosine (m6A) modifications has been implicated in various cancers, including hepatocellular carcinoma (HCC). This study aimed to elucidate the role of m6A modifications in HCC prognosis and the molecular mechanisms involved, particularly focusing on the demethylase FTO.</p><p><strong>Methods: </strong>We analyzed m6A expression in a cohort of 323 HCC patients using immunohistochemical (IHC) staining. The expression of m6A-related genes (FTO, ALKBH5, METTL3, METTL14) was evaluated by qRT-PCR in 120 paired HCC tissues. Further, we established HCC cell lines with altered FTO expression to assess its impact on cell proliferation, invasion, and metastasis through various in vitro assays and in vivo orthotopic HCC mouse models. Statistical analyses included Pearson chi-square test, Kaplan-Meier survival analysis, and both univariate and multivariate Cox regression analyses.</p><p><strong>Results: </strong>IHC staining revealed elevated m6A levels in HCC tissues compared to adjacent non-tumorous tissues, with 57.3% of HCC patients showing increased m6A expression. High m6A levels were correlated with poorer overall survival (OS) and recurrence-free survival (RFS) rates. FTO, a demethylase, was significantly downregulated in HCC tissues and cell lines, particularly in highly metastatic lines. Overexpression of FTO in HCC cells reduced proliferation, migration, and invasion, whereas FTO knockdown had the opposite effect. In vivo, FTO overexpression decreased tumor growth and metastasis. RNA-Seq analysis identified VEGFA as a key gene downregulated by FTO, implicating its role in angiogenesis and tumor progression.</p><p><strong>Conclusions: </strong>Our findings suggest that elevated m6A levels are associated with poor prognosis in HCC patients. FTO downregulation contributes to aberrant m6A modifications, promoting HCC progression and metastasis. FTO acts as a tumor suppressor by negatively regulating VEGFA expression, highlighting its potential as a therapeutic target for HCC treatment. These results highlight the significance of m6A modifications in HCC and provide a foundation for future research on targeted therapies.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"58"},"PeriodicalIF":6.1,"publicationDate":"2025-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12049069/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143989422","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}
Yongqi Shao, Yang Mei, Yixin Tan, Ming Yang, Haijing Wu
{"title":"The regulatory functions of G protein-coupled receptors signaling pathways in B cell differentiation and development contributing to autoimmune diseases.","authors":"Yongqi Shao, Yang Mei, Yixin Tan, Ming Yang, Haijing Wu","doi":"10.1186/s13578-025-01398-7","DOIUrl":"https://doi.org/10.1186/s13578-025-01398-7","url":null,"abstract":"<p><p>Autoimmune diseases are characterized by a dysfunction of the immune system. Disruptions in the balance of B-cell dynamics and the increase in auto-antibody levels are pivotal in the triggering of several autoimmune disorders. All of this is inextricably linked to the differentiation, development, migration, and functional regulation of B cells in the human immune response. G protein-coupled receptors (GPCR) are recognized as crucial targets in drug development and play pivotal roles in both B cell differentiation and the underlying mechanisms of autoimmune diseases. However, there has been an inadequate comprehension of how GPCR intricately modulate B cell development and impact the pathogenesis of autoimmune diseases. Ligands and functions of GPCR-chemokine receptors including CXCR3, CXCR4, CXCR5 and CCR7, lipid receptors including S1PR1-5, cannabinoid receptor CB2 as well as orphan GPCR including GPR132, GPR183, GPR174, and P2RY8 in B cell differentiation and development, will be elaborated in this review. The roles these GPCR play in mediating B cells in several autoimmune diseases will also be discussed. The elucidation of the multifaceted mechanisms controlled by GPCR not only enriches our comprehension of immune responses but also provides a promising avenue for therapeutic interventions in the domain of autoimmune disorders.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"57"},"PeriodicalIF":6.1,"publicationDate":"2025-04-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12042570/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143993032","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}
Fernando Gutiérrez Del Burgo, María Ángeles García-López, Tirso Pons, Enrique Vázquez de Luis, Carlos Martínez-A, Ricardo Villares
{"title":"The chromatin reader Dido3 is a regulator of the gene network that controls B cell differentiation.","authors":"Fernando Gutiérrez Del Burgo, María Ángeles García-López, Tirso Pons, Enrique Vázquez de Luis, Carlos Martínez-A, Ricardo Villares","doi":"10.1186/s13578-025-01394-x","DOIUrl":"https://doi.org/10.1186/s13578-025-01394-x","url":null,"abstract":"<p><p>The development of hematopoietic cell lineages is a highly complex process governed by a delicate interplay of various transcription factors. The expression of these factors is influenced, in part, by epigenetic signatures that define each stage of cell differentiation. In particular, the formation of B lymphocytes depends on the sequential silencing of stemness genes and the balanced expression of interdependent transcription factors, along with DNA rearrangement. We have investigated the impact of Dido3 deficiency, a protein involved in chromatin status readout, on B cell differentiation within the hematopoietic compartment of mice. Our findings revealed significant impairments in the successive stages of B cell development. The absence of Dido3 resulted in remarkable alterations in the expression of essential transcription factors and differentiation markers, which are crucial for orchestrating the differentiation process. Additionally, the somatic recombination process, responsible for generation of antigen receptor diversity, was also adversely affected. These observations highlight the vital role of epigenetic regulation, particularly the involvement of Dido3, in ensuring proper B cell differentiation. This study reveals new mechanisms underlying disruptive alterations, deepening our understanding of hematopoiesis and may potentially lead to insights that aid in the development of therapeutic interventions for disorders involving aberrant B cell development.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"56"},"PeriodicalIF":6.1,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12034202/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144055257","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}
Xu Yang, Lan Li, Wenzheng Qu, Xuejun Cheng, Jinyu Zhang, Yan Sun, Suxiao Liu, Guoping Peng, Rui Zheng, Xuekun Li
{"title":"Zmynd11 is essential for neurogenesis by coordinating H3K36me3 modification of Epha2 and PI3K signaling pathway.","authors":"Xu Yang, Lan Li, Wenzheng Qu, Xuejun Cheng, Jinyu Zhang, Yan Sun, Suxiao Liu, Guoping Peng, Rui Zheng, Xuekun Li","doi":"10.1186/s13578-025-01392-z","DOIUrl":"https://doi.org/10.1186/s13578-025-01392-z","url":null,"abstract":"<p><p>10p15.3 deletion syndrome is caused by the deficiency of MYND-type zinc finger domain-containing protein 11 (ZMYND11) and featured by global developmental delay, intellectual disability, behavioral abnormalities, etc. Although the roles of Zmynd11 is intensively studied in cancer, the function and associated mechanisms of Zmynd11 in neurodevelopment remain largely unknown. Here, we show that Zmynd11 displays abundant and dynamic expression pattern during embryonic neurodevelopment. Zmynd11 deficiency impairs embryonic neurogenesis and neurodevelopment in vitro and in vivo, and inhibits morphological maturation of neurons. Mechanistically, Zmynd11 deficiency leads to decreased Epha2 and disrupts PI3K signaling pathway. Under Zmynd11 deficient condition, H3K36me3 modification on Epha2 promoter abnormally increases and the binding of RNA polymerase II decreases. The restoration of PI3K signaling pathway by exogenous Epha2 can rescue aberrant neurogenesis induced by Zmynd11 depletion in vitro and in vivo. Collectively, our study reveals the essential function of Zmynd11 in neurogenesis via coordinating H3K36me3 modification of Epha2 and PI3K signaling pathway.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"55"},"PeriodicalIF":6.1,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12032794/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144055845","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}
Gabriele Sakalauskaite, Michael Weingartner, Sophie Ebert, Gina Boot, Thomas Bock, Julia Birk, Maria Tsachaki, John W Gallon, Salvatore Piscuoglio, Alex Odermatt
{"title":"A BioID-based approach uncovers the interactome of hexose-6-phosphate dehydrogenase in breast cancer cells and identifies anterior gradient protein 2 as an interacting partner.","authors":"Gabriele Sakalauskaite, Michael Weingartner, Sophie Ebert, Gina Boot, Thomas Bock, Julia Birk, Maria Tsachaki, John W Gallon, Salvatore Piscuoglio, Alex Odermatt","doi":"10.1186/s13578-025-01388-9","DOIUrl":"https://doi.org/10.1186/s13578-025-01388-9","url":null,"abstract":"<p><strong>Background: </strong>Hexose-6-phosphate dehydrogenase (H6PD) catalyzes the first two steps of the pentose-phosphate-pathway (PPP) within the endoplasmic reticulum, generating NADPH. H6PD modulates essential physiological processes, including energy and redox metabolism. Its sole reported interacting partner is 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), utilizing NADPH to reactivate glucocorticoids, linking energy status with hormonal response. Previous studies showed that loss of H6PD affects breast cancer cell properties, independent of 11β-HSD1. It remains unknown whether this is due to impaired concentrations of NADPH or PPP products downstream of H6PD. To gain insight into novel roles and pathways influenced by this enzyme, we aimed to assess the H6PD interactome.</p><p><strong>Results: </strong>We adapted the proximity-dependent Biotin Identification (BioID) method to identify novel H6PD interacting partners. First, we validated the method and confirmed the known interaction between H6PD and 11β-HSD1. Next, we constructed a triple-negative breast cancer MDA-MB-231 cell clone stably expressing a H6PD-biotin ligase fusion protein. Enriched biotinylated proteins were analyzed by mass-spectrometry and potential candidates assessed further by co-immunoprecipitation and functional assays. The resulting interactome revealed proteins of the calreticulin/calnexin cycle, unfolded-protein response (UPR) and chaperone activation pathways. Due to its known association with breast cancer, we examined the PDI Anterior gradient protein 2 (AGR2) as H6PD interacting partner. Gene set enrichment analysis revealed multiple overlapping pathways enriched in breast cancer tissues with relatively high H6PD and AGR2 expression. These included glycolysis, fatty acid metabolism, hypoxia, angiogenesis and epithelial to mesenchymal transition. Co-immunoprecipitation (Co-IP) from MCF7 cells confirmed a physical interaction between H6PD and AGR2. ARG2 knockdown in these cells increased H6PD protein levels but decreased activity. Coexpression with AGR2 in HEK-293 cells did not affect expression but enhanced H6PD activity.</p><p><strong>Conclusion: </strong>BioID was successfully applied in the endoplasmic reticulum to identify AGR2 as H6PD interactor. This was confirmed using Co-IP from MCF7 cells endogenously expressing both proteins. The results indicate that AGR2 controls H6PD protein expression and enhances its activity. Whether higher H6PD activity due to increased AGR2 expression promotes a more aggressive cancer cell phenotype, for example by altering energy metabolism, Ca<sup>2+</sup>-related processes or UPR and chaperone activation pathways, warrants further investigations.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"54"},"PeriodicalIF":6.1,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12032772/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144062598","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}
Chenchen Wang, Junshi Zhang, Yali Xu, Jiawei Zhao, Manman Qiu, Xingli Zhao, Guoqiang Li, Wentao Qiao, Juan Tan
{"title":"SAP30 deacetylates the Tas protein to inhibit PFV replication.","authors":"Chenchen Wang, Junshi Zhang, Yali Xu, Jiawei Zhao, Manman Qiu, Xingli Zhao, Guoqiang Li, Wentao Qiao, Juan Tan","doi":"10.1186/s13578-025-01400-2","DOIUrl":"https://doi.org/10.1186/s13578-025-01400-2","url":null,"abstract":"<p><strong>Background: </strong>Foamy viruses (FVs), a unique class of retroviruses, establish lifelong latent infections in the host without causing symptoms, contributing to the relatively slow progress in FV research. However, key mutations in FVs can result in severe consequences due to their broad cellular tropism, underscoring the importance of studying latent FV infections.</p><p><strong>Results: </strong>To identify new host proteins involved in the replication of prototype foamy virus (PFV), we previously infected the human fibrosarcoma cell line HT1080 with PFV and performed transcriptomic sequencing. The analysis revealed a significant upregulation of SAP30 mRNA levels following PFV infection. Further experiments demonstrated that PFV infection enhances SAP30 promoter activity via the Tas protein, leading to increased SAP30 mRNA and protein expression. Overexpression of SAP30 inhibited PFV replication, whereas knockdown of endogenous SAP30 enhanced PFV replication. Furthermore, SAP30 interacted with the Tas protein to induce its deacetylation, thereby suppressing Tas-mediated transactivation of the PFV LTR and IP promoters. The Sin3 interaction domain at the C-terminus of SAP30 was identified as the critical domain for inhibiting PFV transcription.</p><p><strong>Conclusions: </strong>Our findings suggest that SAP30 inhibits PFV replication by deacetylating the Tas protein, thereby disrupting its transcriptional activation function.</p><p><strong>Key words: </strong>prototype foamy virus; SAP30; Tas; transcription; deacetylation.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"53"},"PeriodicalIF":6.1,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12023400/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144024056","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}
Xiao-Lin Lin, Yi-Min Zhou, Ke Meng, Jia-Yi Yang, Han Zhang, Jin-Hua Lin, Hai-Yan Wu, Xiao-Yu Wang, Hui Zhao, Shan-Shan Feng, Kyu-Sang Park, Dong-Qing Cai, Li Zheng, Xu-Feng Qi
{"title":"CRISPR/Cas-mediated mRNA knockdown in the embryos of Xenopus tropicalis.","authors":"Xiao-Lin Lin, Yi-Min Zhou, Ke Meng, Jia-Yi Yang, Han Zhang, Jin-Hua Lin, Hai-Yan Wu, Xiao-Yu Wang, Hui Zhao, Shan-Shan Feng, Kyu-Sang Park, Dong-Qing Cai, Li Zheng, Xu-Feng Qi","doi":"10.1186/s13578-025-01397-8","DOIUrl":"https://doi.org/10.1186/s13578-025-01397-8","url":null,"abstract":"<p><p>The Xenopus tropicalis (Western clawed frog) is an important amphibian model for genetics, developmental and regenerative biology, due to its diploid genetic background and short generation time. CRISPR-Cas13 and CRISPR interference (CRISPRi) systems have recently been employed to suppress mRNA expression in many organisms such as yeast, plants, and mammalian cells. However, no systematic study of these two systems has been carried out in Xenopus tropicalis. Here, we show that CRISPRi rather than CRISPR-Cas13 is an effective and suitable approach to suppress specific mRNA transcription in Xenopus tropicalis embryos. We demonstrated that CRISPRi composed of dCas9 and KRAB-MeCP2 (dCas9-KM) can efficiently target exogenous and endogenous transcripts in Xenopus tropicalis embryos. Moreover, our data suggest that the new KRAB domain from ZIM3 protein (ZIM3-KRAB, ZIM3K) alone has a comparable transcript targeting capacity in Xenopus tropicalis embryos to the traditional fusion repressor KRAB-MeCP2 in which the KRAB domain from KOX1 protein. In conclusion, our results demonstrate that CRISPRi rather than CRISPR-Cas13 is an efficient knockdown platform to explore specific gene function in Xenopus tropicalis embryos.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"52"},"PeriodicalIF":6.1,"publicationDate":"2025-04-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12020200/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144054159","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}
Min Liu, De-Sheng Zou, Xue-Ying Zhang, De-Hua Wang
{"title":"Huddling behavior regulate adaptive thermogenesis in Brandt's voles (Lasiopodomys brandtii).","authors":"Min Liu, De-Sheng Zou, Xue-Ying Zhang, De-Hua Wang","doi":"10.1186/s13578-025-01391-0","DOIUrl":"https://doi.org/10.1186/s13578-025-01391-0","url":null,"abstract":"<p><strong>Background: </strong>Brown adipose tissue (BAT) is the main site of non-shivering thermogenesis (NST) in small mammals, playing an important role in maintaining body temperature and energy balance. Huddling is a behavioral strategy for small rodents to save energy and improve the survival under cold environments. However, the way of huddling behavior influence on hypothalamus, which regulate BAT thermogenesis in small mammals is rarely illustrated. We used male Brandt's voles (Lasiopodomys brandtii) to explore the possible regulation mechanisms in BAT thermogenesis by the way of cold acclimation and huddling behavior.</p><p><strong>Results: </strong>There is a strong relationship between huddling behavior and NST in BAT. The hypothalamus, which is impacted by huddling behavior, influences PPAR signaling pathway in the BAT, and induces thermogenesis through Calcium signaling pathway. PPAR pathway causes crosstalk among NF-κB signaling pathway, Thermogenesis and Fatty acid metabolism to perform functions for thermogenesis.</p><p><strong>Conclusions: </strong>The results suggest that huddling behavior can modulate adaptive thermogenesis in BAT. Cold acclimation and huddling had a synergistic effect on the regulation of thermogenic function, the hypothalamus mediates thermogenic changes in BAT induced by huddling behavior. In BAT, the specific pathway of thermogenesis is as follows: TRAF6-PPARγ-UCP1-SUCLG1.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"51"},"PeriodicalIF":6.1,"publicationDate":"2025-04-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12020165/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144002801","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}
Yanliang Jiang, Yongliang Zhao, Jie Deng, Xiaoyan Wu, Jian Li, Dong Guo, Ke Xu, Yali Qin, Mingzhou Chen
{"title":"Peptides targeting RAB11A-FIP2 complex inhibit HPIV3, RSV, and IAV replication as broad-spectrum antivirals.","authors":"Yanliang Jiang, Yongliang Zhao, Jie Deng, Xiaoyan Wu, Jian Li, Dong Guo, Ke Xu, Yali Qin, Mingzhou Chen","doi":"10.1186/s13578-025-01384-z","DOIUrl":"https://doi.org/10.1186/s13578-025-01384-z","url":null,"abstract":"<p><strong>Background: </strong>The cytoskeletal framework plays a critical role in the early stages of human parainfluenza virus type 3 (HPIV3) replication, including viral mRNA synthesis and translation. However, its contribution to later stages of infection, particularly in the context of RNA biology, is not well understood. This study focuses on the role of the cytoskeleton in viral nucleocapsid (vRNP, a ribonucleoprotein complex essential for RNA virus replication) transport, assembly, and budding, and explores the cooperative role of the small GTPase RAB11A and its effector RAB11 family interacting protein 2 (FIP2) in vRNP trafficking. These processes are crucial for respiratory RNA viruses like respiratory syncytial virus (RSV) and influenza A virus (IAV), highlighting the importance of RNA-protein interactions in viral pathogenesis.</p><p><strong>Results: </strong>Through the use of cytoskeleton-depolymerizing agents, the study identified actin microfilaments as indispensable for vRNP transport, viral assembly, and viral particle budding. It also revealed the importance of the RAB11A-FIP2 complex in these processes, which are critical for the intracellular trafficking of viral RNA. The development of peptides targeting the RAB11A-FIP2 complex led to the suppression of RAB11A function in infected cells, resulting in vRNP aggregation in the cytoplasm and reduced viral replication. The peptide YT-DRI showed strong broad-spectrum antiviral activity against HPIV3, RSV, and IAV in cellular and animal models and was effective against co-infections in vitro. The antiviral effects of YT-DRI were abolished upon deletion of RAB11A or core components of the RAB11A pathway.</p><p><strong>Conclusion: </strong>This work introduces a promising broad-spectrum antiviral strategy for respiratory tract infections by targeting the RAB11A-FIP2 complex, which regulates the transport and assembly of viral RNA. By disrupting this pathway, YT-DRI effectively inhibits the replication of multiple respiratory RNA viruses, including HPIV3, RSV, and IAV.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"50"},"PeriodicalIF":6.1,"publicationDate":"2025-04-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12013085/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144058290","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}
{"title":"Molecular mechanisms of endothelial-mesenchymal transition and its pathophysiological feature in cerebrovascular disease.","authors":"Huimin Jiang, Yifan Zhou, Weiyue Zhang, Hui Li, Wei Ma, Xunming Ji, Chen Zhou","doi":"10.1186/s13578-025-01393-y","DOIUrl":"https://doi.org/10.1186/s13578-025-01393-y","url":null,"abstract":"<p><p>The phenomenon of endothelial-mesenchymal transition (EndMT), a distinct subtype of epithelial-mesenchymal transition (EMT), has garnered significant attention from scholars. EndMT refers to the process whereby endothelial cells (ECs) transform into mesenchymal cells in response to various stimuli, resulting in the loss of their original characteristics. This process has diverse implications in both physiological and pathological states. Under physiological conditions, EndMT plays a crucial role in the development of the cardiovascular system. Conversely, under pathological conditions, EndMT has been identified as a pivotal factor in the development of cardiovascular diseases. Nonetheless, a comprehensive overview of EndMT in cerebrovascular disease is currently lacking. Here, we discuss the heterogeneity of EndMT occurrence and the regulatory factors involved in its development and analyze the feasibility of EndMT as a therapeutic target, aiming to provide a solid theoretical foundation and evidence to address diseases caused by pathological EndMT.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":"15 1","pages":"49"},"PeriodicalIF":6.1,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008988/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036270","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}