首页 > 最新文献

Apoptosis最新文献

英文 中文
Progenitor CD8+ T cells and hyper-Treg crosstalk: a driver of immune checkpoint inhibitor resistance in esophageal squamous cell carcinoma 祖细胞CD8+ T细胞和超treg串扰:食管鳞状细胞癌免疫检查点抑制剂耐药的驱动因素
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-12 DOI: 10.1007/s10495-025-02256-0
Suning Huang, Jingwei Yan, Wenqi Liu, Baijun Li

Despite the crucial role of antigen presentation in immune checkpoint inhibitor (ICI) efficacy, its contribution and the mechanism in esophageal squamous cell carcinoma (ESCC) remain unclear, representing a key knowledge gap in overcoming immune escape in its immunotherapy. This study explores how tumor antigen presentation and intercellular interactions in the tumor microenvironment (TME) drive ICI resistance using single-cell RNA sequencing (scRNA-seq). Publicly available scRNA-seq data from 24 ESCC patients treated with chemotherapy and ICIs were analyzed. Cell clustering, transcription factor regulation, cell–cell communication analysis, and KEGG/GO enrichment analyses were used to examine malignant cell heterogeneity, the relationship between antigen-presenting cells and ICI responses, and cell–cell interactions influencing anti-tumor response. Spatial relationships were validated through multiplex immunofluorescence. Malignant cells were classified by enrichment analyses into cNMF_1, cNMF_2, cNMF_3, and cNMF_4, with cNMF_4 showing antigen-presenting traits. Based on ICI response groups, cell–cell communication analysis revealed that in poor responders, the antigen presentation ability of tumors induced by treatment was enhanced, and mainly enriched in the MHC-I pathway. The crosstalk between Progenitor CD8+ Tex and hyper-Treg in the TME drove ICI resistance. Hyper-Treg likely regulated CD8+ T activation through the CLEC2C-KLRB1 axis, forming an inhibitory cell interaction network dominated by hyper-Treg, resulting in an overall strong immune suppression state of the TME in this population. The antigen-presenting malignant epithelial cells of ESCC exhibit significant interactions with various T cells in the TME. ICI resistance is closely associated with the crosstalk between progenitor CD8+ Tex and hyper-Treg, representing a promising target for personalized ESCC therapy.

尽管抗原呈递在免疫检查点抑制剂(ICI)的疗效中起着至关重要的作用,但其在食管鳞状细胞癌(ESCC)中的作用及其机制尚不清楚,这是其免疫治疗中克服免疫逃逸的关键知识空白。本研究利用单细胞RNA测序(scRNA-seq)探索肿瘤抗原呈递和肿瘤微环境(TME)中的细胞间相互作用如何驱动ICI耐药性。对24名接受化疗和ICIs治疗的ESCC患者公开获得的scRNA-seq数据进行了分析。细胞聚类、转录因子调控、细胞-细胞通讯分析和KEGG/GO富集分析用于检测恶性细胞异质性、抗原呈递细胞与ICI反应之间的关系以及影响抗肿瘤反应的细胞-细胞相互作用。空间关系通过多重免疫荧光验证。通过富集分析将恶性肿瘤细胞分为cNMF_1、cNMF_2、cNMF_3和cNMF_4, cNMF_4表现出抗原提呈特征。基于ICI应答组,细胞-细胞通讯分析显示,在不良应答者中,治疗诱导的肿瘤抗原呈递能力增强,且主要富集于MHC-I通路。TME中祖细胞CD8+ Tex和hyper-Treg之间的串扰驱动了ICI耐药性。Hyper-Treg可能通过CLEC2C-KLRB1轴调控CD8+ T活化,形成以Hyper-Treg为主的抑制性细胞相互作用网络,导致该人群TME整体处于较强的免疫抑制状态。ESCC的抗原呈递恶性上皮细胞与TME中的各种T细胞表现出显著的相互作用。ICI耐药性与前体细胞CD8+ Tex和hyper-Treg之间的串音密切相关,代表了个性化ESCC治疗的一个有希望的靶点。
{"title":"Progenitor CD8+ T cells and hyper-Treg crosstalk: a driver of immune checkpoint inhibitor resistance in esophageal squamous cell carcinoma","authors":"Suning Huang,&nbsp;Jingwei Yan,&nbsp;Wenqi Liu,&nbsp;Baijun Li","doi":"10.1007/s10495-025-02256-0","DOIUrl":"10.1007/s10495-025-02256-0","url":null,"abstract":"<div><p>Despite the crucial role of antigen presentation in immune checkpoint inhibitor (ICI) efficacy, its contribution and the mechanism in esophageal squamous cell carcinoma (ESCC) remain unclear, representing a key knowledge gap in overcoming immune escape in its immunotherapy. This study explores how tumor antigen presentation and intercellular interactions in the tumor microenvironment (TME) drive ICI resistance using single-cell RNA sequencing (scRNA-seq). Publicly available scRNA-seq data from 24 ESCC patients treated with chemotherapy and ICIs were analyzed. Cell clustering, transcription factor regulation, cell–cell communication analysis, and KEGG/GO enrichment analyses were used to examine malignant cell heterogeneity, the relationship between antigen-presenting cells and ICI responses, and cell–cell interactions influencing anti-tumor response. Spatial relationships were validated through multiplex immunofluorescence. Malignant cells were classified by enrichment analyses into cNMF_1, cNMF_2, cNMF_3, and cNMF_4, with cNMF_4 showing antigen-presenting traits. Based on ICI response groups, cell–cell communication analysis revealed that in poor responders, the antigen presentation ability of tumors induced by treatment was enhanced, and mainly enriched in the MHC-I pathway. The crosstalk between Progenitor CD8<sup>+</sup> Tex and hyper-Treg in the TME drove ICI resistance. Hyper-Treg likely regulated CD8<sup>+</sup> T activation through the CLEC2C-KLRB1 axis, forming an inhibitory cell interaction network dominated by hyper-Treg, resulting in an overall strong immune suppression state of the TME in this population. The antigen-presenting malignant epithelial cells of ESCC exhibit significant interactions with various T cells in the TME. ICI resistance is closely associated with the crosstalk between progenitor CD8<sup>+</sup> Tex and hyper-Treg, representing a promising target for personalized ESCC therapy.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951368","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CSN5 overexpression promotes the integral progression of cervical cancer by enhancing ENO3-mediated glycolysis CSN5过表达通过增强eno3介导的糖酵解促进宫颈癌的整体进展。
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-12 DOI: 10.1007/s10495-025-02246-2
Zhu Cao, Shizhou Yang, Jian Zou, Ye Jin, Yanan Zhang, Tingting Chen, Yuanming Shen

Cervical cancer (CC) remains a significant global health challenge. A deeper understanding of the molecular mechanisms driving CC progression is crucial for developing improved therapeutic strategies. CSN5 is vital in cell functions and cancer, but its role in CC is unclear. This study aims to explore CSN5’s role and its target gene in CC progression, assessing their potential as therapeutic targets. We employed an integrated approach combining bioinformatics analysis, proteomic profiling, and in vitro and in vivo functional assays. Immunohistochemistry was used to analyze CSN5 and ENO3 expression in CC and normal tissues. CSN5 upregulation was associated with advanced clinical stage and poor differentiation. Furthermore, CSN5 overexpression enhanced cellular proliferation and glycolytic metabolism but, paradoxically, suppressed migration, invasion, and the epithelial-mesenchymal transition (EMT). These pro-tumorigenic effects were confirmed in vivo, and the glycolytic inhibitor 2-DG was found to reverse the phenotypes induced by CSN5. Protein sequencing highlighted ENO3’s role in CSN5-mediated tumorigenesis, regulating EMT and glycolysis by stabilizing its ubiquitination degradation through CSN5. Crucially, silencing ENO3 attenuated the oncogenic effects of CSN5 both in vitro and in vivo. Our findings unveil a novel mechanistic paradigm in which CSN5 promotes CC progression by co-opting ENO3 to enhance glycolytic flux while concurrently suppressing cell motility. This study not only deepens the understanding of CC pathogenesis but also identifies the CSN5-ENO3 axis as a promising target for novel therapeutic interventions.

Graphical abstract

宫颈癌(CC)仍然是一个重大的全球卫生挑战。更深入地了解驱动CC进展的分子机制对于制定改进的治疗策略至关重要。CSN5在细胞功能和癌症中至关重要,但其在CC中的作用尚不清楚。本研究旨在探讨CSN5及其靶基因在CC进展中的作用,评估其作为治疗靶点的潜力。我们采用了结合生物信息学分析、蛋白质组学分析以及体外和体内功能分析的综合方法。免疫组化检测CSN5和ENO3在CC和正常组织中的表达。CSN5上调与临床分期较晚、分化较差相关。此外,CSN5过表达增强了细胞增殖和糖酵解代谢,但矛盾的是,它抑制了迁移、侵袭和上皮-间质转化(EMT)。这些促肿瘤作用在体内得到了证实,并且发现糖酵解抑制剂2-DG可以逆转CSN5诱导的表型。蛋白测序显示,ENO3在CSN5介导的肿瘤发生、通过稳定其泛素化降解来调节EMT和糖酵解中发挥作用。至关重要的是,在体外和体内,沉默ENO3都能减弱CSN5的致癌作用。我们的发现揭示了一种新的机制范式,其中CSN5通过协同选择ENO3来增强糖酵解通量,同时抑制细胞运动,从而促进CC进展。这项研究不仅加深了对CC发病机制的理解,而且还确定了CSN5-ENO3轴作为新的治疗干预措施的有希望的靶点。
{"title":"CSN5 overexpression promotes the integral progression of cervical cancer by enhancing ENO3-mediated glycolysis","authors":"Zhu Cao,&nbsp;Shizhou Yang,&nbsp;Jian Zou,&nbsp;Ye Jin,&nbsp;Yanan Zhang,&nbsp;Tingting Chen,&nbsp;Yuanming Shen","doi":"10.1007/s10495-025-02246-2","DOIUrl":"10.1007/s10495-025-02246-2","url":null,"abstract":"<div><p>Cervical cancer (CC) remains a significant global health challenge. A deeper understanding of the molecular mechanisms driving CC progression is crucial for developing improved therapeutic strategies. CSN5 is vital in cell functions and cancer, but its role in CC is unclear. This study aims to explore CSN5’s role and its target gene in CC progression, assessing their potential as therapeutic targets. We employed an integrated approach combining bioinformatics analysis, proteomic profiling, and in vitro and in vivo functional assays. Immunohistochemistry was used to analyze CSN5 and ENO3 expression in CC and normal tissues. CSN5 upregulation was associated with advanced clinical stage and poor differentiation. Furthermore, CSN5 overexpression enhanced cellular proliferation and glycolytic metabolism but, paradoxically, suppressed migration, invasion, and the epithelial-mesenchymal transition (EMT). These pro-tumorigenic effects were confirmed in vivo, and the glycolytic inhibitor 2-DG was found to reverse the phenotypes induced by CSN5. Protein sequencing highlighted ENO3’s role in CSN5-mediated tumorigenesis, regulating EMT and glycolysis by stabilizing its ubiquitination degradation through CSN5. Crucially, silencing ENO3 attenuated the oncogenic effects of CSN5 both in vitro and in vivo. Our findings unveil a novel mechanistic paradigm in which CSN5 promotes CC progression by co-opting ENO3 to enhance glycolytic flux while concurrently suppressing cell motility. This study not only deepens the understanding of CC pathogenesis but also identifies the CSN5-ENO3 axis as a promising target for novel therapeutic interventions.</p><h3>Graphical abstract</h3><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12795886/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951380","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
Advances in the research of gasdermin-dependent pyroptosis mechanisms and therapeutic targets in inflammatory diseases 炎性疾病中气垫胶依赖性焦亡机制及治疗靶点的研究进展。
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-12 DOI: 10.1007/s10495-025-02250-6
Chuanyu Shen, Yiming Zhang, Chuqiao Wei, Hanchi Wang, Yanmin Zhou

Inflammation, especially invasive bacterial inflammation, has always been a hot topic in medical research. With the emergence of superbugs and highly virulent viruses, current treatment methods are gradually revealing areas of deficiency. As an inflammatory form of cell death, the concept of pyroptosis offers new possibilities for the treatment of inflammation. Pyroptosis is typically triggered by inflammasomes and executed by the Gasdermin (GSDM) family. It is characterized by membrane perforation, cell swelling, and the release of cellular contents such as pro-inflammatory factors. Pyroptosis plays a crucial role in host immune defense, inflammatory diseases, and tumor regulation. In this review, we comprehensively explored each member of the GSDM family and its activation pathways, reviewed the cellular consequences of GSDM activation, and the role of GSDM-mediated pyroptosis in inflammatory diseases such as sepsis. We also provided a detailed account of the latest advancements in inflammation control targeting GSDM, aiming to offer new perspectives for precise therapies targeting pyroptosis.

Graphical abstract

炎症,尤其是侵袭性细菌炎症,一直是医学研究的热点。随着超级细菌和高毒力病毒的出现,目前的治疗方法逐渐暴露出不足之处。作为细胞死亡的一种炎症形式,焦亡的概念为炎症的治疗提供了新的可能性。焦亡通常由炎性小体触发,由Gasdermin (GSDM)家族执行。它的特点是膜穿孔,细胞肿胀,细胞内容物如促炎因子的释放。焦亡在宿主免疫防御、炎症疾病和肿瘤调节中起着至关重要的作用。在这篇综述中,我们全面探讨了GSDM家族的每个成员及其激活途径,综述了GSDM激活的细胞后果,以及GSDM介导的焦亡在脓毒症等炎症性疾病中的作用。我们还详细介绍了针对GSDM的炎症控制的最新进展,旨在为针对焦亡的精确治疗提供新的视角。
{"title":"Advances in the research of gasdermin-dependent pyroptosis mechanisms and therapeutic targets in inflammatory diseases","authors":"Chuanyu Shen,&nbsp;Yiming Zhang,&nbsp;Chuqiao Wei,&nbsp;Hanchi Wang,&nbsp;Yanmin Zhou","doi":"10.1007/s10495-025-02250-6","DOIUrl":"10.1007/s10495-025-02250-6","url":null,"abstract":"<div><p>Inflammation, especially invasive bacterial inflammation, has always been a hot topic in medical research. With the emergence of superbugs and highly virulent viruses, current treatment methods are gradually revealing areas of deficiency. As an inflammatory form of cell death, the concept of pyroptosis offers new possibilities for the treatment of inflammation. Pyroptosis is typically triggered by inflammasomes and executed by the Gasdermin (GSDM) family. It is characterized by membrane perforation, cell swelling, and the release of cellular contents such as pro-inflammatory factors. Pyroptosis plays a crucial role in host immune defense, inflammatory diseases, and tumor regulation. In this review, we comprehensively explored each member of the GSDM family and its activation pathways, reviewed the cellular consequences of GSDM activation, and the role of GSDM-mediated pyroptosis in inflammatory diseases such as sepsis. We also provided a detailed account of the latest advancements in inflammation control targeting GSDM, aiming to offer new perspectives for precise therapies targeting pyroptosis.</p><h3>Graphical abstract</h3><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Propofol attenuates angiogenesis by activating endoplasmic reticulum stress to suppress TFAP2C-driven VEGFA transcription 异丙酚通过激活内质网应激来抑制tfap2c驱动的VEGFA转录,从而减弱血管生成。
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-12 DOI: 10.1007/s10495-025-02214-w
Fan Yang, Yi Liu, Hui Li, Xue Shang, Qing Hua, Yun Zhu, Beibei Tao, Zhirong Sun

During anesthesia, significant hemodynamic changes often alter the vascular microenvironment and affecting endothelial cell behavior. Propofol, a commonly used intravenous anesthetic, has been widely studied for its role in tumor angiogenesis through tumor cell–derived VEGF–mediated endothelial interactions. However, its direct effects on endothelial cell–mediated angiogenesis in non-malignant diseases such as diabetic retinopathy, diabetic nephropathy, and coronary heart disease remain unclear. To address this gap, we examined the effects of propofol on VEGFA-mediated angiogenesis in vitro and in vivo. Mechanistically, propofol triggers endoplasmic reticulum stress by promoting phosphorylation of PERK and its downstream effector eIF2α, leading to suppressed translation of TFAP2C—a transcription factor critical for endothelial function. Further analysis revealed that TFAP2C directly binds to the VEGFA promoter to activate its transcription, thereby facilitating VEGFA/VEGFR2-dependent angiogenesis. Together, these findings not only broaden the understanding of propofol’s pharmacological profile, but also identify TFAP2C as a novel transcriptional regulator of VEGFA, offering new perspectives for therapeutic targeting of VEGFA-mediated angiogenesis.

Graphical abstract

Schematic representation of the mechanism of propofol inhibition of VEGFA/VEGFR2-associated angiogenesis via the PERK/eIF2α/TFAP2C axis.

在麻醉过程中,显著的血流动力学变化经常改变血管微环境并影响内皮细胞的行为。异丙酚是一种常用的静脉麻醉剂,它通过肿瘤细胞源性vegf介导的内皮相互作用在肿瘤血管生成中的作用已被广泛研究。然而,其对非恶性疾病(如糖尿病视网膜病变、糖尿病肾病和冠心病)内皮细胞介导的血管生成的直接影响尚不清楚。为了解决这一差距,我们在体外和体内研究了异丙酚对vegf介导的血管生成的影响。在机制上,异丙酚通过促进PERK及其下游效应物eIF2α的磷酸化引发内质网应激,从而抑制tfap2c的翻译,tfap2c是内皮功能的关键转录因子。进一步分析发现,TFAP2C直接结合VEGFA启动子激活其转录,从而促进VEGFA/ vegfr2依赖性血管生成。总之,这些发现不仅拓宽了对异丙酚药理学特征的理解,而且还确定了TFAP2C是一种新的VEGFA转录调节剂,为靶向治疗VEGFA介导的血管生成提供了新的视角。
{"title":"Propofol attenuates angiogenesis by activating endoplasmic reticulum stress to suppress TFAP2C-driven VEGFA transcription","authors":"Fan Yang,&nbsp;Yi Liu,&nbsp;Hui Li,&nbsp;Xue Shang,&nbsp;Qing Hua,&nbsp;Yun Zhu,&nbsp;Beibei Tao,&nbsp;Zhirong Sun","doi":"10.1007/s10495-025-02214-w","DOIUrl":"10.1007/s10495-025-02214-w","url":null,"abstract":"<div><p>During anesthesia, significant hemodynamic changes often alter the vascular microenvironment and affecting endothelial cell behavior. Propofol, a commonly used intravenous anesthetic, has been widely studied for its role in tumor angiogenesis through tumor cell–derived VEGF–mediated endothelial interactions. However, its direct effects on endothelial cell–mediated angiogenesis in non-malignant diseases such as diabetic retinopathy, diabetic nephropathy, and coronary heart disease remain unclear. To address this gap, we examined the effects of propofol on VEGFA-mediated angiogenesis in vitro and in vivo. Mechanistically, propofol triggers endoplasmic reticulum stress by promoting phosphorylation of PERK and its downstream effector eIF2α, leading to suppressed translation of TFAP2C—a transcription factor critical for endothelial function. Further analysis revealed that TFAP2C directly binds to the VEGFA promoter to activate its transcription, thereby facilitating VEGFA/VEGFR2-dependent angiogenesis. Together, these findings not only broaden the understanding of propofol’s pharmacological profile, but also identify TFAP2C as a novel transcriptional regulator of VEGFA, offering new perspectives for therapeutic targeting of VEGFA-mediated angiogenesis.</p><h3>Graphical abstract</h3><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div><p>Schematic representation of the mechanism of propofol inhibition of VEGFA/VEGFR2-associated angiogenesis via the PERK/eIF2α/TFAP2C axis.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12795951/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951324","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 emerging role of cholesterol metabolism in gynecologic cancer development and therapy 胆固醇代谢在妇科癌症发展和治疗中的新作用
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1007/s10495-025-02200-2
Hejing Liu, Yujia Zhou, Jiamin Zhang, Liqing Miao, Qianqian Wu, He Zhu, Shuya Pan, Xueqiong Zhu

The significance of cholesterol metabolism in cancer is a topic of renewed interest. Cholesterol is an essential factor for mammal cells, for it is not only involved in constituting the cell membrane, but also serves as a precursor to steroid hormones and bile acids. Numerous studies have provided increasing evidence of its high relevance to cancer progression. Targeting cholesterol metabolism by using cholesterol metabolism inhibitors has offered another therapeutic strategy for reversing drug resistance in tumors. Here, the regulatory process of cholesterol homeostasis under normal physiological conditions was introduced. Then, the mechanism by which cholesterol metabolism disorder caused gynecologic cancer development and therapy resistance was summarized. Finally, the therapeutic strategies targeting cholesterol metabolism were also discussed in this review.

胆固醇代谢在癌症中的意义是一个重新引起人们兴趣的话题。胆固醇对哺乳动物的细胞来说是一个必不可少的因素,因为它不仅参与构成细胞膜,而且还是类固醇激素和胆汁酸的前体。大量的研究提供了越来越多的证据表明它与癌症进展高度相关。利用胆固醇代谢抑制剂靶向胆固醇代谢为逆转肿瘤耐药提供了另一种治疗策略。本文介绍了正常生理条件下胆固醇稳态的调节过程。综述了胆固醇代谢紊乱导致妇科肿瘤发生及耐药的机制。最后,本文还讨论了针对胆固醇代谢的治疗策略。
{"title":"The emerging role of cholesterol metabolism in gynecologic cancer development and therapy","authors":"Hejing Liu,&nbsp;Yujia Zhou,&nbsp;Jiamin Zhang,&nbsp;Liqing Miao,&nbsp;Qianqian Wu,&nbsp;He Zhu,&nbsp;Shuya Pan,&nbsp;Xueqiong Zhu","doi":"10.1007/s10495-025-02200-2","DOIUrl":"10.1007/s10495-025-02200-2","url":null,"abstract":"<div><p>The significance of cholesterol metabolism in cancer is a topic of renewed interest. Cholesterol is an essential factor for mammal cells, for it is not only involved in constituting the cell membrane, but also serves as a precursor to steroid hormones and bile acids. Numerous studies have provided increasing evidence of its high relevance to cancer progression. Targeting cholesterol metabolism by using cholesterol metabolism inhibitors has offered another therapeutic strategy for reversing drug resistance in tumors. Here, the regulatory process of cholesterol homeostasis under normal physiological conditions was introduced. Then, the mechanism by which cholesterol metabolism disorder caused gynecologic cancer development and therapy resistance was summarized. Finally, the therapeutic strategies targeting cholesterol metabolism were also discussed in this review.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s10495-025-02200-2.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145930645","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
Cross-regulation of autophagy and pyroptosis: a new perspective on the inflammatory microenvironment of atherosclerosis 自噬与焦亡的交叉调控:动脉粥样硬化炎症微环境的新视角
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1007/s10495-025-02221-x
Yu-Mei Gan, Nai-Shen Qin, Jun Ouyang, Yu-Ming Tang, Jia-Hui Qin, Shu-Min Wei, Hui Wang, Jiang-Nan Huang

The pathogenesis of atherosclerosis (AS) is a chronic disease marked by inflammation, and there are intimate associations with various forms of programmed cell death (PCD). Recently, the mechanisms of pyroptosis and autophagy in AS have attracted much attention. Pyroptosis is a form of PCD mediated by inflammasomes, which worsens local inflammatory responses by releasing proinflammatory factors (e.g., IL-18 and IL-1β) and favors plaque instability and thrombosis. Autophagy is a process that helps to keep cells healthy by breaking down damaged cell structures and abnormal proteins. Mitophagy, a specialized form of autophagy, is of major importance to redox homeostasis and the regulation of inflammation. However, the dysregulation of autophagy may disturb the cellular homeostasis, which then accelerates the progression of AS. Studies have found a complex mutual regulation between pyroptosis and autophagy. Autophagy can block the occurrence of pyroptosis by degrading the components of such as NLRP3. The inflammatory mediators released during pyroptosis may cause the disorder of autophagy, which aggravates the cell death and inflammatory response. The disorder of autophagy will also promote pyroptosis’ occurrence and progress. Both of them play a vital role in AS. This study is mainly focused on clarifying the relationship and molecular mechanism between pyroptosis and autophagy in the context of AS. These findings pave the way for new avenues for understanding its pathogenesis and potentially therapeutic decision-making.

动脉粥样硬化(AS)的发病机制是一种以炎症为特征的慢性疾病,与多种形式的程序性细胞死亡(PCD)密切相关。近年来,AS细胞的自噬和焦亡机制备受关注。焦亡是炎症小体介导的PCD的一种形式,通过释放促炎因子(如IL-18和IL-1β)加重局部炎症反应,有利于斑块不稳定和血栓形成。自噬是一种通过分解受损细胞结构和异常蛋白质来帮助保持细胞健康的过程。自噬是自噬的一种特殊形式,对氧化还原稳态和炎症调节具有重要意义。然而,自噬的失调可能会扰乱细胞稳态,从而加速AS的进展。研究发现焦亡与自噬之间存在复杂的相互调节关系。自噬可以通过降解NLRP3等成分来阻断焦亡的发生。焦亡过程中释放的炎症介质可引起细胞自噬紊乱,加重细胞死亡和炎症反应。自噬的紊乱也会促进焦亡的发生和发展。两者在AS中都起着至关重要的作用。本研究的重点是阐明AS背景下焦亡与自噬之间的关系及分子机制。这些发现为了解其发病机制和潜在的治疗决策开辟了新的途径。
{"title":"Cross-regulation of autophagy and pyroptosis: a new perspective on the inflammatory microenvironment of atherosclerosis","authors":"Yu-Mei Gan,&nbsp;Nai-Shen Qin,&nbsp;Jun Ouyang,&nbsp;Yu-Ming Tang,&nbsp;Jia-Hui Qin,&nbsp;Shu-Min Wei,&nbsp;Hui Wang,&nbsp;Jiang-Nan Huang","doi":"10.1007/s10495-025-02221-x","DOIUrl":"10.1007/s10495-025-02221-x","url":null,"abstract":"<div><p>The pathogenesis of atherosclerosis (AS) is a chronic disease marked by inflammation, and there are intimate associations with various forms of programmed cell death (PCD). Recently, the mechanisms of pyroptosis and autophagy in AS have attracted much attention. Pyroptosis is a form of PCD mediated by inflammasomes, which worsens local inflammatory responses by releasing proinflammatory factors (e.g., IL-18 and IL-1β) and favors plaque instability and thrombosis. Autophagy is a process that helps to keep cells healthy by breaking down damaged cell structures and abnormal proteins. Mitophagy, a specialized form of autophagy, is of major importance to redox homeostasis and the regulation of inflammation. However, the dysregulation of autophagy may disturb the cellular homeostasis, which then accelerates the progression of AS. Studies have found a complex mutual regulation between pyroptosis and autophagy. Autophagy can block the occurrence of pyroptosis by degrading the components of such as NLRP3. The inflammatory mediators released during pyroptosis may cause the disorder of autophagy, which aggravates the cell death and inflammatory response. The disorder of autophagy will also promote pyroptosis’ occurrence and progress. Both of them play a vital role in AS. This study is mainly focused on clarifying the relationship and molecular mechanism between pyroptosis and autophagy in the context of AS. These findings pave the way for new avenues for understanding its pathogenesis and potentially therapeutic decision-making.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145930369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell death-associated genes as novel diagnostic biomarkers for autism spectrum disorder 细胞死亡相关基因作为自闭症谱系障碍新的诊断生物标志物
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1007/s10495-025-02201-1
Chanhua Li, Lijuan Wei, Wanling Chen, Guanghui Ran, Lili Liu, Zhongyi Li, Anhua Song, Meiliang Liu, Dongping Huang, Kun Tang, Xiaoyun Zeng, Lijun Wang

Autism spectrum disorder (ASD) involves neuroinflammation and dysregulated neuronal death but lacks objective diagnostic biomarkers. This study investigated whether cell death could serve as a molecular basis for ASD diagnosis. We identified cell death-associated genes (CDGs) in peripheral blood mononuclear cells between ASD and control groups and performed functional enrichment, protein–protein interaction, immune characteristics, and non-negative matrix factorization clustering analyses. Key genes were further selected using 6 machine learning algorithms and weighted gene co-expression network analysis to construct a diagnostic model, which was subsequently validated using external human blood samples and mice prefrontal cortex samples. We explored microRNAs (miRNAs), transcription factors (TFs), and drugs potentially interacted with the key genes. Fourteen CDGs and 4 out of 22 types of immune cells were differentially expressed between ASD and controls in GSE18123. These genes were enriched in pathways such as neuron apoptosis and RAGE receptor binding. Among 6 machine learning models, K-Nearest Neighbors (KNN) model exhibited the best performance. Six key genes were selected as the most important hub genes and used to construct the diagnostic model. The model showed AUCs of 0.722 in GSE42133, 0.781 in our local samples, and 0.889 in mice prefrontal cortex samples. A total of 172 miRNAs, 111 TFs, and 98 drugs were found to interact with these key genes. The expression patterns of CDGs in the peripheral blood of children with ASD demonstrate potential diagnostic value. Further studies are warranted to validate their diagnostic performance in real-world settings and to elucidate the role of cell death dysregulation in ASD.

自闭症谱系障碍(ASD)涉及神经炎症和失调的神经元死亡,但缺乏客观的诊断生物标志物。本研究探讨细胞死亡是否可以作为ASD诊断的分子基础。我们在ASD组和对照组之间的外周血单个核细胞中鉴定了细胞死亡相关基因(CDGs),并进行了功能富集、蛋白-蛋白相互作用、免疫特性和非阴性基质因子聚类分析。通过6种机器学习算法和加权基因共表达网络分析选择关键基因,构建诊断模型,随后使用外部人血样本和小鼠前额皮质样本进行验证。我们探索了microRNAs (miRNAs)、转录因子(TFs)和可能与关键基因相互作用的药物。在GSE18123中,14种CDGs和22种免疫细胞中的4种在ASD和对照组中存在差异表达。这些基因在神经元凋亡和RAGE受体结合等途径中富集。在6种机器学习模型中,k近邻(KNN)模型表现出最好的性能。选择6个关键基因作为最重要的枢纽基因,构建诊断模型。该模型显示GSE42133的auc为0.722,我们局部样本的auc为0.781,小鼠前额皮质样本的auc为0.889。共发现172种mirna、111种tf和98种药物与这些关键基因相互作用。CDGs在ASD患儿外周血中的表达模式显示出潜在的诊断价值。需要进一步的研究来验证它们在现实环境中的诊断性能,并阐明细胞死亡失调在ASD中的作用。
{"title":"Cell death-associated genes as novel diagnostic biomarkers for autism spectrum disorder","authors":"Chanhua Li,&nbsp;Lijuan Wei,&nbsp;Wanling Chen,&nbsp;Guanghui Ran,&nbsp;Lili Liu,&nbsp;Zhongyi Li,&nbsp;Anhua Song,&nbsp;Meiliang Liu,&nbsp;Dongping Huang,&nbsp;Kun Tang,&nbsp;Xiaoyun Zeng,&nbsp;Lijun Wang","doi":"10.1007/s10495-025-02201-1","DOIUrl":"10.1007/s10495-025-02201-1","url":null,"abstract":"<div><p>Autism spectrum disorder (ASD) involves neuroinflammation and dysregulated neuronal death but lacks objective diagnostic biomarkers. This study investigated whether cell death could serve as a molecular basis for ASD diagnosis. We identified cell death-associated genes (CDGs) in peripheral blood mononuclear cells between ASD and control groups and performed functional enrichment, protein–protein interaction, immune characteristics, and non-negative matrix factorization clustering analyses. Key genes were further selected using 6 machine learning algorithms and weighted gene co-expression network analysis to construct a diagnostic model, which was subsequently validated using external human blood samples and mice prefrontal cortex samples. We explored microRNAs (miRNAs), transcription factors (TFs), and drugs potentially interacted with the key genes. Fourteen CDGs and 4 out of 22 types of immune cells were differentially expressed between ASD and controls in GSE18123. These genes were enriched in pathways such as neuron apoptosis and RAGE receptor binding. Among 6 machine learning models, K-Nearest Neighbors (KNN) model exhibited the best performance. Six key genes were selected as the most important hub genes and used to construct the diagnostic model. The model showed AUCs of 0.722 in GSE42133, 0.781 in our local samples, and 0.889 in mice prefrontal cortex samples. A total of 172 miRNAs, 111 TFs, and 98 drugs were found to interact with these key genes. The expression patterns of CDGs in the peripheral blood of children with ASD demonstrate potential diagnostic value. Further studies are warranted to validate their diagnostic performance in real-world settings and to elucidate the role of cell death dysregulation in ASD.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145930761","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phillyrin promotes flap survival by mitigating pyroptosis through the TLR4/NF-κB/NLRP3 signaling pathway philyrin通过TLR4/NF-κB/NLRP3信号通路减轻皮瓣焦亡,从而促进皮瓣存活
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1007/s10495-025-02248-0
Jiapeng Deng, Wenrui Kang, An Wang, Jialong Yang, Kaitao Wang, Lekai Quan, Hebin Pan, Weilong Song, Panshen Xu, Dingsheng Lin

Background

Flap transplantation is a widely used for wound reconstruction but continues to carry a substantial risk of postoperative necrosis. We evaluated the therapeutic effect of phillyrin for improving flap survival and elucidated its underlying pharmacological mechanisms.

Methods

In vivo, a McFarlane flap model was established on the dorsal skin of rats. Animals were assigned to four groups: control, low-dose (17.5 mg/kg/day, intraperitoneal [i.p.]), medium-dose (35 mg/kg/day, i.p.), and high-dose (70 mg/kg/day, i.p.) phillyrin. At 7 days postoperatively, we evaluated flap survival, blood perfusion, oxidative stress, cytokine expression, toll-like receptor 4/nuclear factor κB/NOD-like receptor protein 3 (TLR4/NF-κB/NLRP3) axis activity, and pyroptosis. In vitro, human umbilical vein endothelial cells were treated with hypoxia/reoxygenation or lipopolysaccharide/nigericin models, to investigate the protective effects of phillyrin against inflammation-related pyroptotic injury.

Results

Phillyrin improved flap survival, significantly enhanced local blood perfusion, reduced neutrophil infiltration, and mitigated oxidative stress. Its mechanism of action is closely associated with the inhibition of the TLR4/NF-κB/NLRP3 axis: phillyrin significantly downregulated pathway activity, reduced cytokines secretion, and decreased the expression of pyroptosis-related effector proteins. In vitro, phillyrin demonstrated anti-inflammatory and anti-pyroptotic cytoprotective effects in H/R and inflammatory injury models by modulating the TLR4/NF-κB/NLRP3 axis.

Conclusion

Phillyrin improves flap survival by inhibiting the TLR4/NF-κB/NLRP3 signaling pathway, suppressing excessive inflammation and alleviating ischemia-reperfusion injury.

Graphical abstract

背景:皮瓣移植是一种广泛应用于伤口重建的方法,但仍然存在术后坏死的巨大风险。我们评估了连翘苷提高皮瓣存活率的疗效,并阐明了其潜在的药理学机制。方法在体内建立大鼠背侧皮肤麦克法兰皮瓣模型。实验动物分为4组:对照组、低剂量组(17.5 mg/kg/d)、腹腔注射组(腹腔注射组)。])、中剂量(35mg /kg/天,静脉滴注)和高剂量(70mg /kg/天,静脉滴注)。术后7天,我们评估皮瓣存活、血液灌注、氧化应激、细胞因子表达、toll样受体4/核因子κB/ nod样受体蛋白3 (TLR4/NF-κB/NLRP3)轴活性和焦亡情况。在体外实验中,采用缺氧/再氧化或脂多糖/尼日利亚菌素模型处理人脐静脉内皮细胞,探讨连翘素对炎症相关焦亡损伤的保护作用。结果甘草苷提高皮瓣存活率,显著增强局部血流灌注,减少中性粒细胞浸润,减轻氧化应激。其作用机制与抑制TLR4/NF-κB/NLRP3轴密切相关:茶树苷显著下调通路活性,减少细胞因子分泌,降低热作用相关效应蛋白的表达。在体外,连翘苷通过调节TLR4/NF-κB/NLRP3轴在H/R和炎症损伤模型中表现出抗炎和抗焦亡的细胞保护作用。结论连根霉素通过抑制TLR4/NF-κB/NLRP3信号通路,抑制过度炎症反应,减轻缺血再灌注损伤,提高皮瓣存活。图形抽象
{"title":"Phillyrin promotes flap survival by mitigating pyroptosis through the TLR4/NF-κB/NLRP3 signaling pathway","authors":"Jiapeng Deng,&nbsp;Wenrui Kang,&nbsp;An Wang,&nbsp;Jialong Yang,&nbsp;Kaitao Wang,&nbsp;Lekai Quan,&nbsp;Hebin Pan,&nbsp;Weilong Song,&nbsp;Panshen Xu,&nbsp;Dingsheng Lin","doi":"10.1007/s10495-025-02248-0","DOIUrl":"10.1007/s10495-025-02248-0","url":null,"abstract":"<div><h3>Background</h3><p>Flap transplantation is a widely used for wound reconstruction but continues to carry a substantial risk of postoperative necrosis. We evaluated the therapeutic effect of phillyrin for improving flap survival and elucidated its underlying pharmacological mechanisms.</p><h3>Methods</h3><p>In vivo, a McFarlane flap model was established on the dorsal skin of rats. Animals were assigned to four groups: control, low-dose (17.5 mg/kg/day, intraperitoneal [i.p.]), medium-dose (35 mg/kg/day, i.p.), and high-dose (70 mg/kg/day, i.p.) phillyrin. At 7 days postoperatively, we evaluated flap survival, blood perfusion, oxidative stress, cytokine expression, toll-like receptor 4/nuclear factor κB/NOD-like receptor protein 3 (TLR4/NF-κB/NLRP3) axis activity, and pyroptosis. In vitro, human umbilical vein endothelial cells were treated with hypoxia/reoxygenation or lipopolysaccharide/nigericin models, to investigate the protective effects of phillyrin against inflammation-related pyroptotic injury.</p><h3>Results</h3><p>Phillyrin improved flap survival, significantly enhanced local blood perfusion, reduced neutrophil infiltration, and mitigated oxidative stress. Its mechanism of action is closely associated with the inhibition of the TLR4/NF-κB/NLRP3 axis: phillyrin significantly downregulated pathway activity, reduced cytokines secretion, and decreased the expression of pyroptosis-related effector proteins. In vitro, phillyrin demonstrated anti-inflammatory and anti-pyroptotic cytoprotective effects in H/R and inflammatory injury models by modulating the TLR4/NF-κB/NLRP3 axis.</p><h3>Conclusion</h3><p>Phillyrin improves flap survival by inhibiting the TLR4/NF-κB/NLRP3 signaling pathway, suppressing excessive inflammation and alleviating ischemia-reperfusion injury.</p><h3>Graphical abstract</h3><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145930666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reparative effects of VCAM-1 high-performance MSC-derived exosomes on aged diabetic cardiomyocyte injury: a focus on ferroptosis suppression VCAM-1高性能msc来源外显体对老年糖尿病心肌细胞损伤的修复作用:对铁下垂抑制的关注
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1007/s10495-025-02224-8
Xiaoyang Yin, Yimeng Wei, Yu Liu, Gang Chen, Jing Chen, Jie Cheng, Yongming Liu, Hulai Wei

Cardiac dysfunction in elderly diabetes, due to superimposition of age-related myocardial senescence and diabetes-induced injury, lacks effective therapeutic strategies. Ferroptosis may be a key mechanism underlying cardiomyocyte injury in diabetic cardiomyopathy. Mesenchymal stem cells (MSCs) and their exosomes show potential for repairing cardiomyocytes, restoring cardiac function, improving insulin sensitivity, and mitigating diabetes-related complications, but their mechanisms and relationship with ferroptosis remain unclear. The present study aimed to investigate reparative effects and ferroptosis-mediated mechanism of exosomes derived from VCAM-1 high-performance MSCs on aged diabetic cardiomyocyte injury. High-glucose-damaged senescent cardiomyocyte and aged rat model of diabetic cardiomyopathy were established and treated with VCAM-1⁺-UC-MSCs or -derived exosomes. Assessments of cell phenotypes, RNA sequencing, cardiac function, and markers of senescence and ferroptosis revealed significant mitochondrial damage, iron-ion accumulation, reactive oxygen species (ROS), and cardiac troponin (c-TnT) elevation in the damaged myocardial cells and rat heart tissues, along with weakened cardiac function and pronounced senescence and ferroptosis features, and activation of Ras/Raf/MEK/ERK/c-FOS pathway. VCAM-1⁺ MSCs or exosome administration significantly alleviated these effects, and improved cardiac function. Notably, the reparative effect of VCAM-1⁺-UC-MSCs-derived exosomes was superior to that of conventional MSCs-derived exosomes. In conclusion, VCAM-1⁺-UC-MSCs-derived exosomes attenuate cardiomyocyte ferroptosis by suppressing Ras/Raf/MEK/ERK/c-FOS pathway, thereby ameliorating myocardial injury resulting from superimposition of aging-caused myocardial senescence and diabetes-induced damage in elderly diabetic cardiomyopathy. This may lay a foundation for identifying potential prevention and treatment strategies and targets of MSCs and -derived exosomes on myocardial injury.

老年糖尿病心功能障碍,由于与年龄相关的心肌衰老和糖尿病引起的损伤叠加,缺乏有效的治疗策略。铁下垂可能是糖尿病性心肌病心肌细胞损伤的重要机制。间充质干细胞(MSCs)及其外泌体显示出修复心肌细胞、恢复心功能、改善胰岛素敏感性和减轻糖尿病相关并发症的潜力,但其机制和与铁凋亡的关系尚不清楚。本研究旨在探讨VCAM-1高性能MSCs衍生的外泌体对老年糖尿病心肌细胞损伤的修复作用和凋亡介导的机制。建立高糖损伤的衰老心肌细胞和衰老大鼠糖尿病性心肌病模型,并用VCAM-1 + -UC-MSCs或衍生外泌体治疗。细胞表型、RNA测序、心功能、衰老和铁下垂标志物的评估显示,受损心肌细胞和大鼠心脏组织中存在明显的线粒体损伤、铁离子积累、活性氧(ROS)和心肌肌钙蛋白(c-TnT)升高,同时心功能减弱、明显的衰老和铁下垂特征,以及Ras/Raf/MEK/ERK/c-FOS通路的激活。VCAM-1 + MSCs或外泌体给药可明显缓解这些影响,并改善心功能。值得注意的是,VCAM-1⁺- uc - msc衍生外泌体的修复效果优于常规的msc衍生外泌体。综上所述,VCAM-1⁺- uc - msc来源的外泌体通过抑制Ras/Raf/MEK/ERK/c-FOS通路减轻心肌细胞铁凋亡,从而改善老年糖尿病性心肌病中因衰老引起的心肌衰老和糖尿病引起的损伤叠加而造成的心肌损伤。这可能为确定MSCs及其衍生外泌体对心肌损伤的潜在防治策略和靶点奠定基础。
{"title":"Reparative effects of VCAM-1 high-performance MSC-derived exosomes on aged diabetic cardiomyocyte injury: a focus on ferroptosis suppression","authors":"Xiaoyang Yin,&nbsp;Yimeng Wei,&nbsp;Yu Liu,&nbsp;Gang Chen,&nbsp;Jing Chen,&nbsp;Jie Cheng,&nbsp;Yongming Liu,&nbsp;Hulai Wei","doi":"10.1007/s10495-025-02224-8","DOIUrl":"10.1007/s10495-025-02224-8","url":null,"abstract":"<div><p>Cardiac dysfunction in elderly diabetes, due to superimposition of age-related myocardial senescence and diabetes-induced injury, lacks effective therapeutic strategies. Ferroptosis may be a key mechanism underlying cardiomyocyte injury in diabetic cardiomyopathy. Mesenchymal stem cells (MSCs) and their exosomes show potential for repairing cardiomyocytes, restoring cardiac function, improving insulin sensitivity, and mitigating diabetes-related complications, but their mechanisms and relationship with ferroptosis remain unclear. The present study aimed to investigate reparative effects and ferroptosis-mediated mechanism of exosomes derived from VCAM-1 high-performance MSCs on aged diabetic cardiomyocyte injury. High-glucose-damaged senescent cardiomyocyte and aged rat model of diabetic cardiomyopathy were established and treated with VCAM-1⁺-UC-MSCs or -derived exosomes. Assessments of cell phenotypes, RNA sequencing, cardiac function, and markers of senescence and ferroptosis revealed significant mitochondrial damage, iron-ion accumulation, reactive oxygen species (ROS), and cardiac troponin (c-TnT) elevation in the damaged myocardial cells and rat heart tissues, along with weakened cardiac function and pronounced senescence and ferroptosis features, and activation of Ras/Raf/MEK/ERK/c-FOS pathway. VCAM-1⁺ MSCs or exosome administration significantly alleviated these effects, and improved cardiac function. Notably, the reparative effect of VCAM-1⁺-UC-MSCs-derived exosomes was superior to that of conventional MSCs-derived exosomes. In conclusion, VCAM-1⁺-UC-MSCs-derived exosomes attenuate cardiomyocyte ferroptosis by suppressing Ras/Raf/MEK/ERK/c-FOS pathway, thereby ameliorating myocardial injury resulting from superimposition of aging-caused myocardial senescence and diabetes-induced damage in elderly diabetic cardiomyopathy. This may lay a foundation for identifying potential prevention and treatment strategies and targets of MSCs and -derived exosomes on myocardial injury.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145930638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Suppression of endothelial cell PANoptosis: qingyi decoction targets the TMAO-HMGB1 pathway to mitigate lung injury in acute pancreatitis rats 抑制内皮细胞PANoptosis:清胰汤通过TMAO-HMGB1通路减轻急性胰腺炎大鼠肺损伤
IF 8.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1007/s10495-025-02212-y
Jie Liu, Peng Ge, Zhenxuan Sun, Yalan Luo, Lu Xun, Huijuan Li, Xinyu Luo, Xuetao Zhang, Boliang Pei, Yinan Cao, Guanyu Wang, Hailong Chen

Trimethylamine N-oxide (TMAO) is linked to the onset and progression of acute pancreatitis (AP). Qingyi decoction (QYD) is widely used in Chinese hospitals for treating AP and its complications. This study aimed to explore the role and mechanism of QYD in alleviating severe acute pancreatitis-associated lung injury (SAP-ALI) by the modulation of the gut microbiota metabolite TMAO. The composition of the QYD was analysed using LC–MS/MS. A lung injury model in rats with SAP was created with a retrograde injection of 5% sodium taurocholate (STC, 50 mg/kg) into the pancreaticobiliary duct. The effects of exogenous TMAO, 3,3-dimethyl-l-butanol (DMB), and QYD on pulmonary microvascular endothelial barrier damage in SAP rats were assessed by ELISA, haematoxylin and eosin staining, transmission electron microscopy, RT‒qPCR, immunohistochemistry, and Western blot analysis. An in vitro model of TMAO-induced damage in HUVECs and a monocyte‒endothelial cell coculture system were created. RNA sequencing (RNA-seq) was employed to determine the impact of TMAO on the gene expression profile of endothelial cells. RT‒qPCR, Western blotting, flow cytometry, TUNEL assays, PI staining, and immunofluorescence were performed to assess whether TMAO induces endothelial cell PANoptosis and the effects of QYD on this process. The critical role of high mobility group box 1 (HMGB1) was investigated using a rescue experiment. The results demonstrated that QYD significantly alleviated pancreatic inflammation and lung injury in SAP rats, reduced TMAO levels, and effectively inhibited HMGB1-mediated pulmonary endothelial dysfunction during SAP. RNA-seq, scRNA-seq analysis and in vitro experiments confirmed that TMAO induced PANoptosis in endothelial cells. Mechanistically, QYD may exert protective effects on SAP-ALI by suppressing the TMAO-HMGB1 pathway that mediates endothelial cell PANoptosis, suggesting a potential therapeutic strategy for SAP.

三甲胺n -氧化物(TMAO)与急性胰腺炎(AP)的发生和进展有关。清胰汤在我国医院广泛应用于治疗急性胰腺炎及其并发症。本研究旨在探讨芪黄酮通过调节肠道菌群代谢物氧化三甲胺(TMAO)减轻严重急性胰腺炎相关肺损伤(SAP-ALI)的作用及机制。采用LC-MS /MS对其组成进行分析。采用胰胆管逆行注射5%牛磺胆酸钠(STC, 50 mg/kg)建立SAP大鼠肺损伤模型。采用ELISA、血红素和伊红染色、透射电镜、RT-qPCR、免疫组织化学和Western blot分析外源性TMAO、3,3-二甲基丁醇(DMB)和QYD对SAP大鼠肺微血管内皮屏障损伤的影响。建立了tmao诱导HUVECs损伤的体外模型和单核细胞-内皮细胞共培养系统。采用RNA测序(RNA-seq)技术检测氧化三甲胺对内皮细胞基因表达谱的影响。采用RT-qPCR、Western blotting、流式细胞术、TUNEL、PI染色、免疫荧光等方法观察TMAO是否诱导内皮细胞PANoptosis以及QYD对这一过程的影响。采用救援实验研究了高迁移率组盒1 (HMGB1)的关键作用。结果表明,芪黄酮可显著减轻SAP大鼠胰腺炎症和肺损伤,降低TMAO水平,有效抑制SAP过程中hmgb1介导的肺内皮功能障碍。RNA-seq、scRNA-seq分析及体外实验证实,TMAO可诱导内皮细胞PANoptosis。在机制上,QYD可能通过抑制介导内皮细胞PANoptosis的TMAO-HMGB1通路对SAP- ali发挥保护作用,提示SAP的潜在治疗策略。
{"title":"Suppression of endothelial cell PANoptosis: qingyi decoction targets the TMAO-HMGB1 pathway to mitigate lung injury in acute pancreatitis rats","authors":"Jie Liu,&nbsp;Peng Ge,&nbsp;Zhenxuan Sun,&nbsp;Yalan Luo,&nbsp;Lu Xun,&nbsp;Huijuan Li,&nbsp;Xinyu Luo,&nbsp;Xuetao Zhang,&nbsp;Boliang Pei,&nbsp;Yinan Cao,&nbsp;Guanyu Wang,&nbsp;Hailong Chen","doi":"10.1007/s10495-025-02212-y","DOIUrl":"10.1007/s10495-025-02212-y","url":null,"abstract":"<div><p>Trimethylamine N-oxide (TMAO) is linked to the onset and progression of acute pancreatitis (AP). Qingyi decoction (QYD) is widely used in Chinese hospitals for treating AP and its complications. This study aimed to explore the role and mechanism of QYD in alleviating severe acute pancreatitis-associated lung injury (SAP-ALI) by the modulation of the gut microbiota metabolite TMAO. The composition of the QYD was analysed using LC–MS/MS. A lung injury model in rats with SAP was created with a retrograde injection of 5% sodium taurocholate (STC, 50 mg/kg) into the pancreaticobiliary duct. The effects of exogenous TMAO, 3,3-dimethyl-l-butanol (DMB), and QYD on pulmonary microvascular endothelial barrier damage in SAP rats were assessed by ELISA, haematoxylin and eosin staining, transmission electron microscopy, RT‒qPCR, immunohistochemistry, and Western blot analysis. An in vitro model of TMAO-induced damage in HUVECs and a monocyte‒endothelial cell coculture system were created. RNA sequencing (RNA-seq) was employed to determine the impact of TMAO on the gene expression profile of endothelial cells. RT‒qPCR, Western blotting, flow cytometry, TUNEL assays, PI staining, and immunofluorescence were performed to assess whether TMAO induces endothelial cell PANoptosis and the effects of QYD on this process. The critical role of high mobility group box 1 (HMGB1) was investigated using a rescue experiment. The results demonstrated that QYD significantly alleviated pancreatic inflammation and lung injury in SAP rats, reduced TMAO levels, and effectively inhibited HMGB1-mediated pulmonary endothelial dysfunction during SAP. RNA-seq, scRNA-seq analysis and in vitro experiments confirmed that TMAO induced PANoptosis in endothelial cells. Mechanistically, QYD may exert protective effects on SAP-ALI by suppressing the TMAO-HMGB1 pathway that mediates endothelial cell PANoptosis, suggesting a potential therapeutic strategy for SAP.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"31 1","pages":""},"PeriodicalIF":8.1,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145930669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Apoptosis
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1