Pub Date : 2025-01-30DOI: 10.1016/j.jare.2025.01.050
Kexiong Liu, Luyao Zhang, Xiaoling Xu, Mengyao Song, Haiquan Ding, Linli Xiao, Junhui Wen, Chunmei Zhou, Jiahua Bai, Yan Liu
Introduction
In modern sows, lactational high weight loss (HWL), caused by the large litter size and inadequate feed intake, has a negative effect on follicular development after weaning, resulting in poor reproductive performance in the subsequent parity. However, the underlying mechanism remains unclear.
Objectives
This research aimed to explore the mechanism that sows HWL during lactation damages follicular development and attempt to improve the reproductive function by treating with butyrate.
Method
Four multiparous sister sows were chosen to build a HWL model for lactating sows through feed restriction during the final week of a 21-day lactation. Spatially transcriptomics (ST) and tissue immunofluorescent staining were then utilized for the antral follicles in the ovarian surface to search for differentially expressed genes and proteins among different cell types. Subsequently, the mouse assay, including immunofluorescent staining, transmission electron microscopy, hormone detection and western blot, were conducted to verify the findings in sows and investigate the effect of butyrate on the follicular development in HWL mice.
Results
Based on the transcriptomic analysis, differentially expressed genes in granulosa cells, theca cells, and ovarian stromal cells were examined. The findings revealed that HWL disturbs the mitochondrial electron transport chain and steroidogenesis in all three cell types by downregulating the expression of NDUFB3, SDHB, CYCS, COX8A and CYP19A1, as well as upregulating the expression of STAR, CYP11A1 and CYP17A1. Furthermore, results from mouse assays demonstrated that HWL causes apoptosis and alters sex hormone secretion by impairing mitochondrial function and disordering the expression of steroidogenesis key enzymes in ovarian cells, while these effects were partially mitigated by butyrate.
Conclusion
The mitochondrial dysfunction and abnormal steroidogenesis induced by HWL during lactation in ovarian cells harm the follicular development of weaning sows, which could be alleviated by butyrate.
{"title":"Lactational high weight loss impairs follicular development by causing mitochondrial dysfunction of ovarian cells in sows and mitigated by butyrate supplement","authors":"Kexiong Liu, Luyao Zhang, Xiaoling Xu, Mengyao Song, Haiquan Ding, Linli Xiao, Junhui Wen, Chunmei Zhou, Jiahua Bai, Yan Liu","doi":"10.1016/j.jare.2025.01.050","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.050","url":null,"abstract":"<h3>Introduction</h3>In modern sows, lactational high weight loss (HWL), caused by the large litter size and inadequate feed intake, has a negative effect on follicular development after weaning, resulting in poor reproductive performance in the subsequent parity. However, the underlying mechanism remains unclear.<h3>Objectives</h3>This research aimed to explore the mechanism that sows HWL during lactation damages follicular development and attempt to improve the reproductive function by treating with butyrate.<h3>Method</h3>Four multiparous sister sows were chosen to build a HWL model for lactating sows through feed restriction during the final week of a 21-day lactation. Spatially transcriptomics (ST) and tissue immunofluorescent staining were then utilized for the antral follicles in the ovarian surface to search for differentially expressed genes and proteins among different cell types. Subsequently, the mouse assay, including immunofluorescent staining, transmission electron microscopy, hormone detection and western blot, were conducted to verify the findings in sows and investigate the effect of butyrate on the follicular development in HWL mice.<h3>Results</h3>Based on the transcriptomic analysis, differentially expressed genes in granulosa cells, theca cells, and ovarian stromal cells were examined. The findings revealed that HWL disturbs the mitochondrial electron transport chain and steroidogenesis in all three cell types by downregulating the expression of <em>NDUFB3</em>, <em>SDHB</em>, <em>CYCS</em>, <em>COX8A</em> and <em>CYP19A1</em>, as well as upregulating the expression of <em>STAR</em>, <em>CYP11A1</em> and <em>CYP17A1</em>. Furthermore, results from mouse assays demonstrated that HWL causes apoptosis and alters sex hormone secretion by impairing mitochondrial function and disordering the expression of steroidogenesis key enzymes in ovarian cells, while these effects were partially mitigated by butyrate.<h3>Conclusion</h3>The mitochondrial dysfunction and abnormal steroidogenesis induced by HWL during lactation in ovarian cells harm the follicular development of weaning sows, which could be alleviated by butyrate.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"60 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143072131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-30DOI: 10.1016/j.jare.2025.01.046
Pengju Yu, Mi Zhou, Deshui Yu, Zhongchi Zhang, Shuting Ye, Yifa Yu, Xianyun Sun, Shaojie Li, Chengcheng Hu
Introduction
The synthesis and regulation of ergosterol are vital for fungal growth and stress adaptation. While ergosterol-mediated feedback regulation is a recognized mechanism controlling sterol biosynthesis in fungi, prior research suggests the presence of additional regulatory mechanisms. However, the specifics of the alternative regulatory mechanisms have not been systematically investigated.
Objectives
We proposed that a regulatory network is likely to discern disturbances in sterol biosynthesis and trigger responses accordingly. This study aimed to validate the hypothesis and investigate the regulatory mechanisms.
Methods
Quantitative Real-time PCR and HPLC-MS/MS were used to explore and compare the regulation of sterol biosynthesis in different fungi. Key transcription factors involved in the alternative regulatory mechanism in Neurospora crassa were identified by phenotypic profiling of a transcription factor mutant library. ChIP-qPCR, fluorescence confocal imaging, RNA sequencing, and gene set enrichment analysis (GSEA) reveal the mechanism of each transcription factor.
Results
Unlike the canonical ergosterol-mediated feedback regulation in fungi like C. neoformans, our study demonstrated that the inhibitions of ergosterol biosynthesis at specific steps triggered distinct transcriptional responses of erg genes in fungi, including N. crassa and Aspergillus fumigatus. In N. crassa, the responses were orchestrated by different transcription factors. Specifically, the inhibition of ERG24 and ERG2 activated transcription factors SAH-2 and AtrR, resulting in the upregulation of erg24, erg2, erg25, and erg3. Furthermore, the inhibition of ERG11/CYP51 activated transcription factor NcSR, leading to the upregulation of erg11 and erg6. Phenotypic profiles of mutants of various N. crassa erg genes and the aforementioned transcription factors implied that the targeted regulation of ergosterol biosynthesis could fortify fungal viability within complex habitats.
Conclusion
Our study reveals a novel regulatory mechanism in fungi: targeted upregulation of specific sterol biosynthesis genes in response to given perturbations in ergosterol biosynthesis, exhibiting a higher degree of precision and sophistication in sterol biosynthesis regulation.
{"title":"Targeted regulation of sterol biosynthesis genes according to perturbations in ergosterol biosynthesis in fungi","authors":"Pengju Yu, Mi Zhou, Deshui Yu, Zhongchi Zhang, Shuting Ye, Yifa Yu, Xianyun Sun, Shaojie Li, Chengcheng Hu","doi":"10.1016/j.jare.2025.01.046","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.046","url":null,"abstract":"<h3>Introduction</h3>The synthesis and regulation of ergosterol are vital for fungal growth and stress adaptation. While ergosterol-mediated feedback regulation is a recognized mechanism controlling sterol biosynthesis in fungi, prior research suggests the presence of additional regulatory mechanisms. However, the specifics of the alternative regulatory mechanisms have not been systematically investigated.<h3>Objectives</h3>We proposed that a regulatory network is likely to discern disturbances in sterol biosynthesis and trigger responses accordingly. This study aimed to validate the hypothesis and investigate the regulatory mechanisms.<h3>Methods</h3>Quantitative Real-time PCR and HPLC-MS/MS were used to explore and compare the regulation of sterol biosynthesis in different fungi. Key transcription factors involved in the alternative regulatory mechanism in <em>Neurospora crassa</em> were identified by phenotypic profiling of a transcription factor mutant library. ChIP-qPCR, fluorescence confocal imaging, RNA sequencing, and gene set enrichment analysis (GSEA) reveal the mechanism of each transcription factor.<h3>Results</h3>Unlike the canonical ergosterol-mediated feedback regulation in fungi like <em>C. neoformans</em>, our study demonstrated that the inhibitions of ergosterol biosynthesis at specific steps triggered distinct transcriptional responses of <em>erg</em> genes in fungi, including <em>N. crassa</em> and <em>Aspergillus fumigatus</em>. In <em>N. crassa</em>, the responses were orchestrated by different transcription factors. Specifically, the inhibition of ERG24 and ERG2 activated transcription factors SAH-2 and AtrR, resulting in the upregulation of <em>erg24</em>, <em>erg2</em>, <em>erg25,</em> and <em>erg3</em>. Furthermore, the inhibition of ERG11/CYP51 activated transcription factor NcSR, leading to the upregulation of <em>erg11</em> and <em>erg6</em>. Phenotypic profiles of mutants of various <em>N. crassa erg</em> genes and the aforementioned transcription factors implied that the targeted regulation of ergosterol biosynthesis could fortify fungal viability within complex habitats.<h3>Conclusion</h3>Our study reveals a novel regulatory mechanism in fungi: targeted upregulation of specific sterol biosynthesis genes in response to given perturbations in ergosterol biosynthesis, exhibiting a higher degree of precision and sophistication in sterol biosynthesis regulation.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"6 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143071954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Osteoarthritis (OA), a widespread degenerative joint disease, predominantly affects individuals from middle age onwards, exhibiting non-inflammatory characteristics. OA leads to the gradual deterioration of articular cartilage and subchondral bone, causing pain and reduced mobility. The risk of OA increases with age, making it a critical health concern for seniors. Despite significant research efforts and various therapeutic approaches, the precise causes of OA remain unclear.
Aim of review
This paper provides a thorough examination of OA characteristics, pathogenic mechanisms at various levels, and personalized treatment strategies for different OA stages. The review aims to enhance understanding of disease mechanisms and establish a theoretical framework for developing more effective therapeutic interventions.
Key scientific concepts of review
This review systematically examines OA through multiple perspectives, integrating current knowledge of clinical presentation, pathological mechanisms, and associated signaling pathways. It assesses diagnostic methods and reviews both pharmacological and surgical treatments for OA, as well as emerging tissue engineering approaches to manage the disease. While therapeutic strategies such as exercise, anti-inflammatory drugs, and surgical interventions are employed to manage symptoms and modify joint structure, none have been able to effectively halt OA’s advancement or achieve long-lasting symptom relief. Tissue engineering strategies, such as cell-seeded scaffolds, supportive matrices, and growth factor delivery, have emerged as promising approaches for cartilage repair and OA treatment. To combat the debilitating effects of OA, it is crucial to investigate the molecular basis of its pathogenesis and seek out innovative therapeutic targets for more potent preventive and treatment strategies.
{"title":"Advances in the pathology and treatment of osteoarthritis","authors":"Xueliang Peng, Xuanning Chen, Yifan Zhang, Zhichao Tian, Meihua Wang, Zhuoyue Chen","doi":"10.1016/j.jare.2025.01.053","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.053","url":null,"abstract":"<h3>Background</h3>Osteoarthritis (OA), a widespread degenerative joint disease, predominantly affects individuals from middle age onwards, exhibiting non-inflammatory characteristics. OA leads to the gradual deterioration of articular cartilage and subchondral bone, causing pain and reduced mobility. The risk of OA increases with age, making it a critical health concern for seniors. Despite significant research efforts and various therapeutic approaches, the precise causes of OA remain unclear.<h3>Aim of review</h3>This paper provides a thorough examination of OA characteristics, pathogenic mechanisms at various levels, and personalized treatment strategies for different OA stages. The review aims to enhance understanding of disease mechanisms and establish a theoretical framework for developing more effective therapeutic interventions.<h3>Key scientific concepts of review</h3>This review systematically examines OA through multiple perspectives, integrating current knowledge of clinical presentation, pathological mechanisms, and associated signaling pathways. It assesses diagnostic methods and reviews both pharmacological and surgical treatments for OA, as well as emerging tissue engineering approaches to manage the disease. While therapeutic strategies such as exercise, anti-inflammatory drugs, and surgical interventions are employed to manage symptoms and modify joint structure, none have been able to effectively halt OA’s advancement or achieve long-lasting symptom relief. Tissue engineering strategies, such as cell-seeded scaffolds, supportive matrices, and growth factor delivery, have emerged as promising approaches for cartilage repair and OA treatment. To combat the debilitating effects of OA, it is crucial to investigate the molecular basis of its pathogenesis and seek out innovative therapeutic targets for more potent preventive and treatment strategies.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"58 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143057153","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-30DOI: 10.1016/j.jare.2025.01.049
Baolian Fan, Zhongju Ji, Min Zhu, Yidu Chen, Jincai Liang, Yu Li, Runxiang Yi, Chenxu Liu, Lijun Wang, Ningwei Shi, Tingxing Yang, Ruoshi Huang, Lu Yang, Aijia Ji, Zhongqiu Liu, Lixin Duan
Introduction
Timosaponins, as steroidal saponins, are the primary active constituents and quality biomarkers in Anemarrhena asphodeloides Bunge. Despite their significance, the biosynthetic pathways of timosaponins have not been thoroughly investigated.
Objectives
This study aims to delineate the biosynthetic pathway of timosaponins in A. asphodeloides, elucidate the catalytic mechanisms of the key cycloartenol synthase (CAS), and investigate the antifungal properties of timosaponins.
Methods
Genes were cloned from A. asphodeloides and heterologous expressed in yeast, tobacco or bacillus coli. Site-directed mutagenesis and molecular docking were used to elucidate the catalytic mechanism of CAS. Antifungal assays were conducted to evaluate the antifungal activities of timosaponins.
Results
In this study, we elucidated the biochemical functions of seven genes involved in timosaponins biosynthesis in A. asphodeloides. Among three candidate OSC genes, AaOSCR12 was identified as the gene encoding cycloartenol synthase, which is responsible for the skeleton cyclization in timosaponin biosynthesis. Six residues (257H, 369N, 448T, 507V, 558P, 616Y) were identified as the critical catalytic active sites of CAS (AaOSCR12). Sterol methyltransferase (AaSMT1) and sterol side-chain reductase (AaSSR2) were found to be the subsequent enzymes and the branching points leading to phytosterol and cholesterol biosynthesis, respectively. Two oxide reductase genes, AaCYP90B27 and AaCYP90B2, were responsible for post-modification of cholesterol, serving as a precursor of timosaponins. A key 26-O-β-glucosidase (AaF26G1) was identified as facilitating the conversion of furostanol-type timosaponins into spirostanol-type timosaponins. Antifungal assays revealed that spirostanol-type timosaponin AⅢ exhibits superior antifungal activity compared to furostanol-type timosaponin BⅡ, potentially linked to plant defense mechanisms involving AaF26G1.
Conclusion
This study utilized a multi-chassis cross-identification strategy, revealing key enzymes in the timosaponin biosynthetic pathway and offering novel insights into plant defense mechanisms against microbial pathogens.
{"title":"Elucidation of the biosynthetic pathways of timosaponins reveals the antifungal mechanisms in Anemarrhena asphodeloides","authors":"Baolian Fan, Zhongju Ji, Min Zhu, Yidu Chen, Jincai Liang, Yu Li, Runxiang Yi, Chenxu Liu, Lijun Wang, Ningwei Shi, Tingxing Yang, Ruoshi Huang, Lu Yang, Aijia Ji, Zhongqiu Liu, Lixin Duan","doi":"10.1016/j.jare.2025.01.049","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.049","url":null,"abstract":"<h3>Introduction</h3>Timosaponins, as steroidal saponins, are the primary active constituents and quality biomarkers in <em>Anemarrhena asphodeloides</em> Bunge. Despite their significance, the biosynthetic pathways of timosaponins have not been thoroughly investigated.<h3>Objectives</h3>This study aims to delineate the biosynthetic pathway of timosaponins in <em>A. asphodeloides</em>, elucidate the catalytic mechanisms of the key cycloartenol synthase (CAS), and investigate the antifungal properties of timosaponins.<h3>Methods</h3>Genes were cloned from <em>A. asphodeloides</em> and heterologous expressed in yeast, tobacco or bacillus coli. Site-directed mutagenesis and molecular docking were used to elucidate the catalytic mechanism of CAS. Antifungal assays were conducted to evaluate the antifungal activities of timosaponins.<h3>Results</h3>In this study, we elucidated the biochemical functions of seven genes involved in timosaponins biosynthesis in <em>A. asphodeloides</em>. Among three candidate <em>OSC</em> genes, <em>AaOSCR12</em> was identified as the gene encoding cycloartenol synthase, which is responsible for the skeleton cyclization in timosaponin biosynthesis. Six residues (257H, 369N, 448T, 507V, 558P, 616Y) were identified as the critical catalytic active sites of CAS (AaOSCR12). Sterol methyltransferase (AaSMT1) and sterol side-chain reductase (AaSSR2) were found to be the subsequent enzymes and the branching points leading to phytosterol and cholesterol biosynthesis, respectively. Two oxide reductase genes, <em>AaCYP90B27</em> and <em>AaCYP90B2</em>, were responsible for post-modification of cholesterol, serving as a precursor of timosaponins. A key 26-O-<em>β</em>-glucosidase (AaF26G1) was identified as facilitating the conversion of furostanol-type timosaponins into spirostanol-type timosaponins. Antifungal assays revealed that spirostanol-type timosaponin AⅢ exhibits superior antifungal activity compared to furostanol-type timosaponin BⅡ, potentially linked to plant defense mechanisms involving AaF26G1.<h3>Conclusion</h3>This study utilized a multi-chassis cross-identification strategy, revealing key enzymes in the timosaponin biosynthetic pathway and offering novel insights into plant defense mechanisms against microbial pathogens.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"12 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143056798","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-28DOI: 10.1016/j.jare.2025.01.051
Yan Li, Mengyi Zhu, Penghui Yang, Daoyang Chen, Dongmei Zhou, Yinghui Ren, Zimu Zhang, Chuangdong Ruan, Yurong Da, Rongxin Zhang
Introduction
Although it is believed that chronic inflammatory and degenerative diseases of the central nervous system are mediated by autoimmune Th17 cells, the underlying mechanisms remain largely unexplored. Recent studies and our research have revealed that Sp3 was blocked in multiple sclerosis (MS) patients and experimental autoimmune encephalomyelitis (EAE). However, it remained unclear why it is silent and how it regulates Th17 cell differentiation in MS.
Objectives
This study aimed to explore the impact of Sp3 on Th17 cell-mediated EAE and the underlying mechanism.
Methods
The effect of Sp3 on the clinical symptoms of EAE was evaluated by scoring, histochemistry, and fast blue (FB) techniques, scRNA-seq data analysis, flow cytometry, ELISA, PCR, WB, immunofluorescence and reporter gene techniques were used to explore the molecular mechanism of Sp3 regulating Th17 cell differentiation.
Results
Injection of overexpression Sp3 lentivirus could significantly ameliorate the EAE progress and clinical symptoms and prevent the polarization of Th1 and Th17 cells both in vivo and in vitro. We confirmed the occurrence of EAE in Sp3+/+CD4Cre mice and Sp3+/- knockout mice. Furthermore, we identified Sp3 as a target of miR-223, which is found to be upregulated in the blood of MS patients, as well as in EAE and Th17 cells. Moreover, knockdown of miR-223 led to a marked improvement in EAE symptoms and a suppression of Th1 and Th17 cell polarization in vivo and in vitro. Mechanistically, Sp3 significantly suppressed RORγt expression and the phosphorylation of Stat3 and Smad2/3 by directly upregulating Socs3. Interestingly, Socs3 was found to regulate Sp3 expression in response to TGF-β1 via a feedback loop. Moreover, Socs3 modulated phospho-Smad2/3 by binding to and degrading the transforming growth factor-β receptor II (TβRII).
Conclusion
Thus, our study suggests a novel mechanism involving miR-223/Sp3/Socs3/TGF-β signaling as a potential therapeutic strategy for targeting Th17 cells in immunotherapy.
{"title":"Sp3 ameliorated experimental autoimmune encephalomyelitis by triggering Socs3 in Th17 cells","authors":"Yan Li, Mengyi Zhu, Penghui Yang, Daoyang Chen, Dongmei Zhou, Yinghui Ren, Zimu Zhang, Chuangdong Ruan, Yurong Da, Rongxin Zhang","doi":"10.1016/j.jare.2025.01.051","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.051","url":null,"abstract":"<h3>Introduction</h3>Although it is believed that chronic inflammatory and degenerative diseases of the central nervous system are mediated by autoimmune Th17 cells, the underlying mechanisms remain largely unexplored. Recent studies and our research have revealed that Sp3 was blocked in multiple sclerosis (MS) patients and experimental autoimmune encephalomyelitis (EAE). However, it remained unclear why it is silent and how it regulates Th17 cell differentiation in MS.<h3>Objectives</h3>This study aimed to explore the impact of Sp3 on Th17 cell-mediated EAE and the underlying mechanism.<h3>Methods</h3>The effect of Sp3 on the clinical symptoms of EAE was evaluated by scoring, histochemistry, and fast blue (FB) techniques, scRNA-seq data analysis, flow cytometry, ELISA, PCR, WB, immunofluorescence and reporter gene techniques were used to explore the molecular mechanism of Sp3 regulating Th17 cell differentiation.<h3>Results</h3>Injection of overexpression Sp3 lentivirus could significantly ameliorate the EAE progress and clinical symptoms and prevent the polarization of Th1 and Th17 cells both <em>in vivo</em> and <em>in vitro</em>. We confirmed the occurrence of EAE in Sp3<sup>+/+CD4Cre</sup> mice and Sp3<sup>+/-</sup> knockout mice. Furthermore, we identified Sp3 as a target of miR-223, which is found to be upregulated in the blood of MS patients, as well as in EAE and Th17 cells. Moreover, knockdown of miR-223 led to a marked improvement in EAE symptoms and a suppression of Th1 and Th17 cell polarization <em>in vivo</em> and <em>in vitro</em>. Mechanistically, Sp3 significantly suppressed RORγt expression and the phosphorylation of Stat3 and Smad2/3 by directly upregulating Socs3. Interestingly, Socs3 was found to regulate Sp3 expression in response to TGF-β1 via a feedback loop. Moreover, Socs3 modulated phospho-Smad2/3 by binding to and degrading the transforming growth factor-β receptor II (TβRII).<h3>Conclusion</h3>Thus, our study suggests a novel mechanism involving miR-223/Sp3/Socs3/TGF-β signaling as a potential therapeutic strategy for targeting Th17 cells in immunotherapy.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"148 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143050398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-27DOI: 10.1016/j.jare.2025.01.045
Anca C. Parau, Senem Büyüksungur, Guanhao Li, Quan Liu, Ernesto Badillo, Laura Blum, Jürgen Schmidt, Iulian Pana, Catalin Vitelaru, Ioana M. Marinescu, Mihaela Dinu, Michael Smuglov, Christina Schmuttermaier, Tugba E. Tanir, Harald Klüter, Nesrin Hasirci, Julia Kzhyshkowska, Alina Vladescu Dragomir
Introduction
Chronic inflammation leading to implant failure present major challenges in orthopedics, dentistry, and reconstructive surgery. Titanium alloys, while widely used, often provoke inflammatory complications. Zinc-doped calcium phosphate (CaP) coatings offer potential to enhance implant integration by improving corrosion resistance, bioactivity, and immunocompatibility.
Objectives
The objective of the study was to develope novel coating composition based on zinc-doped CaP coatings on Ti64 alloy implants that for the first time combines improved corrosion resistance, antibacterial properties and principally improved compatibly with the innate immunity primarily due to the proper programming of resident tissue macrophages to promote long-term implant acceptance.
Methods
Ti64 substrates were coated with CaP and Zn-doped CaP using the microarc oxidation (MAO) technique. The adhesion between substrate and coatings are investigated by the progressive scratch test. The corrosion resistance and bioactivity were evaluated through electrochemical tests and simulated body fluid immersion. RNA sequencing was used to assess inflammatory responses in human primary macrophages. Antibacterial efficacy was tested against Escherichia coli and Staphylococcus aureus.
Results
Zn addition significantly increased the adhesion of the coatings to Ti64 alloy, doubling up the critical load (from 4 N to 11 N). Zn-doped CaP coatings demonstrated enhanced corrosion resistance and increased biomineralization. RNA sequencing revealed that Zn addition suppressed inflammatory and promoted tolerogenic macrophage programming. Most pronounced effects was compensatory effect Zi on the CaP-supressed oxidative phosphorylation and lysosomal pathways in healing macrophages, and by upregulation of metallothioneins. Zn-doped coatings also exhibited superior antibacterial efficacy, reducing E. coli and S. aureus colonization by 99 % and 90 %, respectively.
Conclusion
Zinc-doped CaP coatings on Ti64 implants significantly improved corrosion resistance, bioactivity, and antibacterial performance. We developed advanced multifunctional biomaterial equipped with beneficial anti-inflammatory and tissue integrative programming of innate immunity providing principal advantages for the long-term implant integration and reducing the implant failure risks.
{"title":"Zn-doped CaP coating equips Ti implants with corrosion resistance, biomineralization, antibacterial and immunotolerant activities","authors":"Anca C. Parau, Senem Büyüksungur, Guanhao Li, Quan Liu, Ernesto Badillo, Laura Blum, Jürgen Schmidt, Iulian Pana, Catalin Vitelaru, Ioana M. Marinescu, Mihaela Dinu, Michael Smuglov, Christina Schmuttermaier, Tugba E. Tanir, Harald Klüter, Nesrin Hasirci, Julia Kzhyshkowska, Alina Vladescu Dragomir","doi":"10.1016/j.jare.2025.01.045","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.045","url":null,"abstract":"<h3>Introduction</h3>Chronic inflammation leading to implant failure present major challenges in orthopedics, dentistry, and reconstructive surgery. Titanium alloys, while widely used, often provoke inflammatory complications. Zinc-doped calcium phosphate (CaP) coatings offer potential to enhance implant integration by improving corrosion resistance, bioactivity, and immunocompatibility.<h3>Objectives</h3>The objective of the study was to develope novel coating composition based on zinc-doped CaP coatings on Ti64 alloy implants that for the first time combines improved corrosion resistance, antibacterial properties and principally improved compatibly with the innate immunity primarily due to the proper programming of resident tissue macrophages to promote long-term implant acceptance.<h3>Methods</h3>Ti64 substrates were coated with CaP and Zn-doped CaP using the microarc oxidation (MAO) technique. The adhesion between substrate and coatings are investigated by the progressive scratch test. The corrosion resistance and bioactivity were evaluated through electrochemical tests and simulated body fluid immersion. RNA sequencing was used to assess inflammatory responses in human primary macrophages. Antibacterial efficacy was tested against <em>Escherichia coli</em> and <em>Staphylococcus aureus</em>.<h3>Results</h3>Zn addition significantly increased the adhesion of the coatings to Ti64 alloy, doubling up the critical load (from 4 N to 11 N). Zn-doped CaP coatings demonstrated enhanced corrosion resistance and increased biomineralization. RNA sequencing revealed that Zn addition suppressed inflammatory and promoted tolerogenic macrophage programming. Most pronounced effects was compensatory effect Zi on the CaP-supressed oxidative phosphorylation and lysosomal pathways in healing macrophages, and by upregulation of metallothioneins. Zn-doped coatings also exhibited superior antibacterial efficacy, reducing <em>E. coli</em> and <em>S. aureus</em> colonization by 99 % and 90 %, respectively.<h3>Conclusion</h3>Zinc-doped CaP coatings on Ti64 implants significantly improved corrosion resistance, bioactivity, and antibacterial performance. We developed advanced multifunctional biomaterial equipped with beneficial anti-inflammatory and tissue integrative programming of innate immunity providing principal advantages for the long-term implant integration and reducing the implant failure risks.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"64 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143050459","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Developing strategies to improve the therapeutic efficacy of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) in autoimmune diseases have garnered increased attention.
Objectives
To evaluate whether rapamycin-induced autophagy within the systemic lupus erythematosus (SLE) inflammatory microenvironment (Rapa-SLE) augments the therapeutic effects of MSC-derived EVs in SLE.
Methods
The therapeutic potential of the resulting EVs (Rapa-SLE-EV) was assessed in MRL/lpr mice. Rapa-SLE-EVs were compared with EVs derived from MSCs from MSCs cultured with EV-depleted fetal bovine serum (FBS-EV), EVs from MSCs cultured with rapamycin-treated FBS (Rapa-FBS-EV), and EVs exposed to SLE serum without rapamycin (SLE-EV). The autoimmune response, renal function, and pathological damage were assessed among the mouse groups. Additionally, mechanistic investigations into the role of the anti-inflammatory protein IDO1 within the EVs.
Results
Interaction with the SLE inflammatory microenvironment triggered autophagy in MSCs, which was further enhanced by rapamycin treatment. Rapa-SLE-EV administration significantly ameliorated the autoimmune response and renal damage in MRL/lpr mice, outperforming other MSC-EV groups. This treatment mitigated key manifestations of SLE, including reduced autoantibody levels, as well as splenomegaly, and lymphadenopathy. Furthermore, Rapa-SLE-EV demonstrated superior suppression of plasma inflammatory cytokines, preserved renal function, mitigated pathological damage, and reduced glomerular immune complex deposition. Mechanistically, Rapa-SLE-EV exhibits exceptional inhibitory effects on SLE-B cell function, benefited by the high expression of the anti-inflammatory protein IDO1, which was confirmed to enter SLE-B cells through EVs.
Conclusions
We developed a novel strategy to improve the therapeutic efficacy of MSC-EVs in SLE and confirmed that the immunomodulatory function of the MSC-EVs is enhanced through autophagic activation and interaction with the SLE serum microenvironment, a process likely benefited by the high expression of IDO1.
{"title":"Autophagy activation within inflammatory microenvironment improved the therapeutic effect of MSC-Derived extracellular Vesicle in SLE","authors":"Shuzhen Liao, Fengbiao Guo, Zengzhi Xiao, Haiyan Xiao, Quan-ren Pan, Yugan Guo, Jiaxuan Chen, Xi Wang, Shuting Wang, Haimin Huang, Lawei Yang, Hua-feng Liu, Qingjun Pan","doi":"10.1016/j.jare.2025.01.044","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.044","url":null,"abstract":"<h3>Introduction</h3>Developing strategies to improve the therapeutic efficacy of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) in autoimmune diseases have garnered increased attention.<h3>Objectives</h3>To evaluate whether rapamycin-induced autophagy within the systemic lupus erythematosus (SLE) inflammatory microenvironment (Rapa-SLE) augments the therapeutic effects of MSC-derived EVs in SLE.<h3>Methods</h3>The therapeutic potential of the resulting EVs (Rapa-SLE-EV) was assessed in MRL/lpr mice. Rapa-SLE-EVs were compared with EVs derived from MSCs from MSCs cultured with EV-depleted fetal bovine serum (FBS-EV), EVs from MSCs cultured with rapamycin-treated FBS (Rapa-FBS-EV), and EVs exposed to SLE serum without rapamycin (SLE-EV). The autoimmune response, renal function, and pathological damage were assessed among the mouse groups. Additionally, mechanistic investigations into the role of the anti-inflammatory protein IDO1 within the EVs.<h3>Results</h3>Interaction with the SLE inflammatory microenvironment triggered autophagy in MSCs, which was further enhanced by rapamycin treatment. Rapa-SLE-EV administration significantly ameliorated the autoimmune response and renal damage in MRL/lpr mice, outperforming other MSC-EV groups. This treatment mitigated key manifestations of SLE, including reduced autoantibody levels, as well as splenomegaly, and lymphadenopathy. Furthermore, Rapa-SLE-EV demonstrated superior suppression of plasma inflammatory cytokines, preserved renal function, mitigated pathological damage, and reduced glomerular immune complex deposition. Mechanistically, Rapa-SLE-EV exhibits exceptional inhibitory effects on SLE-B cell function, benefited by the high expression of the anti-inflammatory protein IDO1, which was confirmed to enter SLE-B cells through EVs.<h3>Conclusions</h3>We developed a novel strategy to improve the therapeutic efficacy of MSC-EVs in SLE and confirmed that the immunomodulatory function of the MSC-EVs is enhanced through autophagic activation and interaction with the SLE serum microenvironment, a process likely benefited by the high expression of IDO1.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"25 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143050460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Extrachromosomal circular DNA (eccDNA) plays significant roles in cancer progression and prognosis. However, it remains unclear whether cell-free eccDNA, considered more stable than linear DNA, possesses cancer-specific genomic features. Furthermore, the biogenesis and function of eccDNAs are not yet fully understood.
Objectives
This study aims to characterize the genomic landscape of urinary cell-free eccDNAs in prostate cancer (PCa) and non-cancer (NC) individuals, elucidate their biogenesis and PCa-specific genomic features, and investigate their roles in PCa progression.
Methods
We conducted urine Circle-seq for 21 PCa patients and 16 NC individuals, performed integrated analysis with other omics datasets, and finally validated the function of eccDNA by in vitro transfection and RNA-seq.
Results
We pioneered the profiling of urinary cell-free eccDNAs landscape in PCa and uncovered a high association between eccDNA generation and active chromatin status as well as gene transcription. Double strand breaks and R-loops, which preferentially occur in active genomic sites and cause genome instability, can promote eccDNA generation. Genome instability frequently results in genomic mutations, and our study further established a link between eccDNA generation and oncogenic mutations. Additionally, genes specifically exhibiting high eccDNA generation frequency (HFGs) in PCa contributed to PCa progression and were associated with poorer survival outcomes in PCa patients. Finally, we demonstrated that eccDNAs derived from PCa-specific HFGs, in contrast to intergenic eccDNAs, could suppress PCa cell proliferation and migration, which was independent of their host gene expression.
Conclusion
Our study illustrated the biogenesis of eccDNAs from DSBs in active genes, revealed PCa-specific eccDNA features and suggested new mechanisms underlying eccDNA function.
{"title":"The urinary eccDNA landscape in prostate cancer reveals associations with genome instability and vital roles in cancer progression","authors":"Shengcai Chen, Zhimin Zhou, Yangchen Ye, Zhen You, Qi Lv, Yu Dong, Jindan Luo, Liang Gong, Yanfen Zhu","doi":"10.1016/j.jare.2025.01.039","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.039","url":null,"abstract":"<h3>Introduction</h3>Extrachromosomal circular DNA (eccDNA) plays significant roles in cancer progression and prognosis. However, it remains unclear whether cell-free eccDNA, considered more stable than linear DNA, possesses cancer-specific genomic features. Furthermore, the biogenesis and function of eccDNAs are not yet fully understood.<h3>Objectives</h3>This study aims to characterize the genomic landscape of urinary cell-free eccDNAs in prostate cancer (PCa) and non-cancer (NC) individuals, elucidate their biogenesis and PCa-specific genomic features, and investigate their roles in PCa progression.<h3>Methods</h3>We conducted urine Circle-seq for 21 PCa patients and 16 NC individuals, performed integrated analysis with other omics datasets, and finally validated the function of eccDNA by <em>in vitro</em> transfection and RNA-seq.<h3>Results</h3>We pioneered the profiling of urinary cell-free eccDNAs landscape in PCa and uncovered a high association between eccDNA generation and active chromatin status as well as gene transcription. Double strand breaks and R-loops, which preferentially occur in active genomic sites and cause genome instability, can promote eccDNA generation. Genome instability frequently results in genomic mutations, and our study further established a link between eccDNA generation and oncogenic mutations. Additionally, genes specifically exhibiting high eccDNA generation frequency (HFGs) in PCa contributed to PCa progression and were associated with poorer survival outcomes in PCa patients. Finally, we demonstrated that eccDNAs derived from PCa-specific HFGs, in contrast to intergenic eccDNAs, could suppress PCa cell proliferation and migration, which was independent of their host gene expression.<h3>Conclusion</h3>Our study illustrated the biogenesis of eccDNAs from DSBs in active genes, revealed PCa-specific eccDNA features and suggested new mechanisms underlying eccDNA function.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"27 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143044817","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Spinal cord injury (SCI) is a severe central nervous system disorder with limited treatment options. While autophagy plays a protective role in neural repair, its regulatory mechanisms in SCI remain unclear. Actin-like protein 6A (Actl6a) influences cell fate and neural development, yet its specific role in SCI repair is not well understood. This study investigates Actl6a’s function in regulating autophagy and apoptosis via the transcription factor Sox2 in SCI.
Objectives
This study aims to determine if Actl6a promotes neural survival post-SCI by regulating autophagy-related genes Atg5 and Atg7 through Sox2. It also examines how the demethylase Fto modulates Actl6a mRNA stability via m6A methylation.
Methods
In vitro experiments were conducted using primary neurons and HT-22 hippocampal cells exposed to hydrogen peroxide (H2O2)-induced oxidative stress. Actl6a expression was manipulated by knockdown or overexpression. For in vivo studies, a rat SCI model was established with AAV-Actl6a injected at the injury site to induce Actl6a overexpression. Autophagy and apoptosis markers were analyzed using immunofluorescence, Western blotting, and qPCR. Additionally, m6A dot blot and RNA immunoprecipitation (RIP) assays were performed to assess Fto’s role in regulating Actl6a mRNA methylation and stability.
Results
Actl6a expression significantly decreased after SCI, resulting in increased apoptosis. Overexpressing Actl6a enhanced autophagy, reduced apoptosis, and improved neurological function in SCI models. Mechanistically, Actl6a and Sox2 collaboratively upregulated Atg5 and Atg7 expression, promoting autophagy. Fto’s modulation of Actl6a mRNA stability via m6A demethylation further influenced autophagy and apoptosis.
Conclusion
Actl6a, through interaction with Sox2, plays a critical role in modulating autophagy and reducing apoptosis in SCI, with Fto’s m6A modification affecting Actl6a stability. This Fto/Actl6a/Sox2 axis is a promising therapeutic target for SCI repair.
{"title":"Actl6a regulates autophagy via Sox2-dependent Atg5 and Atg7 expression to inhibit apoptosis in spinal cord injury","authors":"Jian Hao, Yubiao Yang, Li Xie, Zhenhan Li, Boyuan Ma, Bitao Wang, Jinyu Chen, Zhi Zeng, Xianhu Zhou","doi":"10.1016/j.jare.2025.01.038","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.038","url":null,"abstract":"<h3>Introduction</h3>Spinal cord injury (SCI) is a severe central nervous system disorder with limited treatment options. While autophagy plays a protective role in neural repair, its regulatory mechanisms in SCI remain unclear. Actin-like protein 6A (Actl6a) influences cell fate and neural development, yet its specific role in SCI repair is not well understood. This study investigates Actl6a’s function in regulating autophagy and apoptosis via the transcription factor Sox2 in SCI.<h3>Objectives</h3>This study aims to determine if Actl6a promotes neural survival post-SCI by regulating autophagy-related genes Atg5 and Atg7 through Sox2. It also examines how the demethylase Fto modulates Actl6a mRNA stability via m6A methylation.<h3>Methods</h3>In vitro experiments were conducted using primary neurons and HT-22 hippocampal cells exposed to hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>)-induced oxidative stress. Actl6a expression was manipulated by knockdown or overexpression. For in vivo studies, a rat SCI model was established with AAV-Actl6a injected at the injury site to induce Actl6a overexpression. Autophagy and apoptosis markers were analyzed using immunofluorescence, Western blotting, and qPCR. Additionally, m6A dot blot and RNA immunoprecipitation (RIP) assays were performed to assess Fto’s role in regulating Actl6a mRNA methylation and stability.<h3>Results</h3>Actl6a expression significantly decreased after SCI, resulting in increased apoptosis. Overexpressing Actl6a enhanced autophagy, reduced apoptosis, and improved neurological function in SCI models. Mechanistically, Actl6a and Sox2 collaboratively upregulated Atg5 and Atg7 expression, promoting autophagy. Fto’s modulation of Actl6a mRNA stability via m6A demethylation further influenced autophagy and apoptosis.<h3>Conclusion</h3>Actl6a, through interaction with Sox2, plays a critical role in modulating autophagy and reducing apoptosis in SCI, with Fto’s m6A modification affecting Actl6a stability. This Fto/Actl6a/Sox2 axis is a promising therapeutic target for SCI repair.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"19 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143035069","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-25DOI: 10.1016/j.jare.2025.01.040
Longchen Yu, Qining Guo, Yaping Li, Mai Mao, Zhenping Liu, Tingting Li, Lei Wang, Xin Zhang
Introduction
Pancreatic cancer (PC) cannot currently be completely cured and has a poor prognosis. Necroptosis is a distinct form of regulated cell death that differs from both necrosis and apoptosis. Understanding the role of necroptosis during PC progression would open new avenues for targeted therapy.
Objectives
The purpose of this study is to examine the impact of necroptosis on the progression of PC and related mechanisms.
Methods
RNA sequencing was performed to identify necroptosis-related genes that are differentially expressed in PC tissues. The biological functions of CHMP4C and its necroptosis effects were determined in vitro and in vivo. RNA immunoprecipitation, MeRIP-qPCR, Co-immunoprecipitation assays were conducted to evaluate the interaction among CHMP4C, YBX1 and caspase-8 mRNA. Extracellular vesicles were isolated using the differential ultracentrifugation method. The expression of CHMP4C, p-MLKL and CD117 were detected on a PC tissue microarray using multiplex immunofluorescence staining.
Results
CHMP4C was significantly overexpressed in PC cells and tissues. It promoted cell growth and suppressed necroptosis of PC cells in both in vivo and in vitro settings. Mechanistically, CHMP4C interacted with YBX1 to mediate m5C modification of caspase-8 mRNA, resulting in increased caspase-8 expression and inhibition of RIPK1/RIPK3/MLKL pathway phosphorylation. Furthermore, CHMP4C promoted extracellular exocytosis of p-MLKL to further suppress necroptosis. Additionally, PC cells used CHMP4C within extracellular vesicles to recruit and stimulate mast cells (MCs), which in turn promoted PC cell proliferation. In PC tissues, the expression of CHMP4C showed a negative correlation with p-MLKL and a positive association with CD117. High expression levels of CHMP4C in patients were associated with poorer overall survival outcomes.
Conclusions
CHMP4C promotes PC progression by inhibiting necroptosis, which has potential as a biomarker and therapeutic target in PC.
{"title":"CHMP4C promotes pancreatic cancer progression by inhibiting necroptosis via the RIPK1/RIPK3/MLKL pathway","authors":"Longchen Yu, Qining Guo, Yaping Li, Mai Mao, Zhenping Liu, Tingting Li, Lei Wang, Xin Zhang","doi":"10.1016/j.jare.2025.01.040","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.040","url":null,"abstract":"<h3>Introduction</h3>Pancreatic cancer (PC) cannot currently be completely cured and has a poor prognosis. Necroptosis is a distinct form of regulated cell death that differs from both necrosis and apoptosis. Understanding the role of necroptosis during PC progression would open new avenues for targeted therapy.<h3>Objectives</h3>The purpose of this study is to examine the impact of necroptosis on the progression of PC and related mechanisms.<h3>Methods</h3>RNA sequencing was performed to identify necroptosis-related genes that are differentially expressed in PC tissues. The biological functions of CHMP4C and its necroptosis effects were determined <em>in vitro</em> and <em>in vivo</em>. RNA immunoprecipitation, MeRIP-qPCR, Co-immunoprecipitation assays were conducted to evaluate the interaction among CHMP4C, YBX1 and caspase-8 mRNA. Extracellular vesicles were isolated using the differential ultracentrifugation method. The expression of CHMP4C, p-MLKL and CD117 were detected on a PC tissue microarray using multiplex immunofluorescence staining.<h3>Results</h3>CHMP4C was significantly overexpressed in PC cells and tissues. It promoted cell growth and suppressed necroptosis of PC cells in both <em>in vivo</em> and <em>in vitro</em> settings. Mechanistically, CHMP4C interacted with YBX1 to mediate m<sup>5</sup>C modification of caspase-8 mRNA, resulting in increased caspase-8 expression and inhibition of RIPK1/RIPK3/MLKL pathway phosphorylation. Furthermore, CHMP4C promoted extracellular exocytosis of p-MLKL to further suppress necroptosis. Additionally, PC cells used CHMP4C within extracellular vesicles to recruit and stimulate mast cells (MCs), which in turn promoted PC cell proliferation. In PC tissues, the expression of CHMP4C showed a negative correlation with p-MLKL and a positive association with CD117. High expression levels of CHMP4C in patients were associated with poorer overall survival outcomes.<h3>Conclusions</h3>CHMP4C promotes PC progression by inhibiting necroptosis, which has potential as a biomarker and therapeutic target in PC.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"38 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031135","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}