首页 > 最新文献

Inflammation Research最新文献

英文 中文
Erianin protects chondrocytes against IL-1β-induced oxidative stress and ferroptosis by activating GPX4/STING signaling in osteoarthritis. Erianin通过激活骨关节炎中的GPX4/STING信号通路,保护软骨细胞免受il -1β诱导的氧化应激和铁凋亡。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-14 DOI: 10.1007/s00011-025-02162-w
Cuiyu Li, Wei Jian, Shuai Lu, Yun Wang, Chao Fang

Background: Erianin (Eri) has been known for its analgesic and antipyretic properties. This research focuses on impact of Eri on chondrocyte viability, inflammatory cytokine production, extracellular matrix (ECM) degradation, and ferroptosis, which are key factors in cartilage diseases.

Methods: The mouse model of osteoarthritis (OA) was induced by destabilization of medial meniscus (DMM). Chondrocytes were treated with different concentrations of Eri and exposed to IL-1β to simulate disease conditions. The chondrocytes were induced to undergo ferroptosis using erastin (Era), and ferroptosis was inhibited by Fer-1. This was done to form an intervention control group in combination with Era and to explore the synergistic effect. The effects of Eri on cell viability, proliferation, inflammatory responses, ECM degradation, and ferroptosis were assessed using CCK-8 analysis, EDU assay, Western blot, immunofluorescence, ROS staining, and flow cytometry. The Cellular Thermal Shift Assay (CETSA) was also employed to confirm the direct binding and thermal stability of GPX4 and STING in the presence of Eri.

Results: The findings indicate that Eri does not exhibit cytotoxic effects at certain concentrations and can actually enhance chondrocyte proliferation and viability. It also reduces the production of inflammatory cytokines and ECM degradation products, suggesting a protective role against cartilage damage. Furthermore, Eri was found to inhibit ferroptosis in chondrocytes, potentially through the activation of the GPX4/STING signaling pathway. Molecular docking combined with CETSA confirmed that Eri enhances the thermal stability of GPX4 and STING, indicating a stabilizing effect on this key enzyme. In the DMM mouse model, Eri significantly alleviated cartilage degeneration and improved chondrocyte function, as evidenced by reduced osteophyte formation and subchondral bone sclerosis. Eri can act independently or in combination with the ferroptosis inducer erastin (Era) and the ferroptosis inhibitor Ferrostatin-1 (Fer-1). By inhibiting lipid peroxidation, regulating cell proliferation and extracellular matrix degradation, it exerts an intervention effect on IL-1β-induced ferroptosis of chondrocytes. Moreover, when used in combination with Fer-1, it has a synergistic enhancing effect in reversing ferroptosis-related damage.

Conclusions: Eri demonstrates promising therapeutic potential in the treatment of OA by inhibiting chondrocyte ferroptosis and protecting against ECM degradation and inflammatory responses.

背景:鸢尾素(Erianin, Eri)以其镇痛和解热特性而闻名。本研究的重点是Eri对软骨细胞活力、炎症细胞因子产生、细胞外基质(ECM)降解和铁下垂的影响,这是软骨疾病的关键因素。方法:采用内侧半月板失稳法(DMM)诱导小鼠骨关节炎(OA)模型。软骨细胞用不同浓度的Eri处理并暴露于IL-1β以模拟疾病状况。用erastin (Era)诱导软骨细胞发生铁下垂,fe -1抑制铁下垂。本研究拟与Era联合组成干预对照组,探讨其协同效应。通过CCK-8分析、EDU测定、Western blot、免疫荧光、ROS染色和流式细胞术评估Eri对细胞活力、增殖、炎症反应、ECM降解和铁下垂的影响。细胞热移实验(CETSA)也证实了GPX4和STING在Eri存在下的直接结合和热稳定性。结果:Eri在一定浓度下不表现细胞毒作用,反而能增强软骨细胞的增殖和活力。它还减少炎症细胞因子和ECM降解产物的产生,表明对软骨损伤具有保护作用。此外,研究发现Eri可能通过激活GPX4/STING信号通路来抑制软骨细胞的铁下垂。与CETSA结合的分子对接证实,Eri增强了GPX4和STING的热稳定性,表明对这一关键酶具有稳定作用。在DMM小鼠模型中,Eri可显著缓解软骨退变,改善软骨细胞功能,减少骨赘形成和软骨下骨硬化。Eri可以单独或与铁下垂诱导剂erastin (Era)和铁下垂抑制剂Ferrostatin-1 (Fer-1)联合作用。通过抑制脂质过氧化,调节细胞增殖和细胞外基质降解,对il -1β诱导的软骨细胞铁凋亡有干预作用。此外,当与fe -1联合使用时,它在逆转铁中毒相关损伤方面具有协同增强作用。结论:Eri通过抑制软骨细胞铁下垂和防止ECM降解和炎症反应,在OA治疗中显示出良好的治疗潜力。
{"title":"Erianin protects chondrocytes against IL-1β-induced oxidative stress and ferroptosis by activating GPX4/STING signaling in osteoarthritis.","authors":"Cuiyu Li, Wei Jian, Shuai Lu, Yun Wang, Chao Fang","doi":"10.1007/s00011-025-02162-w","DOIUrl":"https://doi.org/10.1007/s00011-025-02162-w","url":null,"abstract":"<p><strong>Background: </strong>Erianin (Eri) has been known for its analgesic and antipyretic properties. This research focuses on impact of Eri on chondrocyte viability, inflammatory cytokine production, extracellular matrix (ECM) degradation, and ferroptosis, which are key factors in cartilage diseases.</p><p><strong>Methods: </strong>The mouse model of osteoarthritis (OA) was induced by destabilization of medial meniscus (DMM). Chondrocytes were treated with different concentrations of Eri and exposed to IL-1β to simulate disease conditions. The chondrocytes were induced to undergo ferroptosis using erastin (Era), and ferroptosis was inhibited by Fer-1. This was done to form an intervention control group in combination with Era and to explore the synergistic effect. The effects of Eri on cell viability, proliferation, inflammatory responses, ECM degradation, and ferroptosis were assessed using CCK-8 analysis, EDU assay, Western blot, immunofluorescence, ROS staining, and flow cytometry. The Cellular Thermal Shift Assay (CETSA) was also employed to confirm the direct binding and thermal stability of GPX4 and STING in the presence of Eri.</p><p><strong>Results: </strong>The findings indicate that Eri does not exhibit cytotoxic effects at certain concentrations and can actually enhance chondrocyte proliferation and viability. It also reduces the production of inflammatory cytokines and ECM degradation products, suggesting a protective role against cartilage damage. Furthermore, Eri was found to inhibit ferroptosis in chondrocytes, potentially through the activation of the GPX4/STING signaling pathway. Molecular docking combined with CETSA confirmed that Eri enhances the thermal stability of GPX4 and STING, indicating a stabilizing effect on this key enzyme. In the DMM mouse model, Eri significantly alleviated cartilage degeneration and improved chondrocyte function, as evidenced by reduced osteophyte formation and subchondral bone sclerosis. Eri can act independently or in combination with the ferroptosis inducer erastin (Era) and the ferroptosis inhibitor Ferrostatin-1 (Fer-1). By inhibiting lipid peroxidation, regulating cell proliferation and extracellular matrix degradation, it exerts an intervention effect on IL-1β-induced ferroptosis of chondrocytes. Moreover, when used in combination with Fer-1, it has a synergistic enhancing effect in reversing ferroptosis-related damage.</p><p><strong>Conclusions: </strong>Eri demonstrates promising therapeutic potential in the treatment of OA by inhibiting chondrocyte ferroptosis and protecting against ECM degradation and inflammatory responses.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"19"},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145966034","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Machine learning and multi-omics integration identifies immunological predictors and mechanistic insights in autoimmune encephalitis. 机器学习和多组学整合识别自身免疫性脑炎的免疫学预测因子和机制见解。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-14 DOI: 10.1007/s00011-025-02180-8
Junshuang Guo, Yizhou Zou

Objective: To develop an interpretable prognostic prediction model for autoimmune encephalitis (AE) using immunological indicators and to investigate the potential role of nucleophosmin (NPM1) in disease pathogenesis through multi-omics approaches.

Methods: We enrolled patients diagnosed with antibody-positive AE and analyzed a broad panel of immunological indicators. Prognostic prediction models were developed using eight machine learning algorithms and validated in an independent cohort. Model interpretability was enhanced through SHapley Additive exPlanations (SHAP) analysis. We further evaluated the therapeutic potential of protein A immunoadsorption (PAIA) in reducing pathogenic antibodies. Building upon these clinical and immunological findings, we sought to investigate the underlying mechanisms by exploring the role of nucleophosmin (NPM1). To this end, we integrated single-cell RNA sequencing and spatial transcriptomics in an experimental autoimmune encephalomyelitis (EAE) model and conducted a phenome-wide association study (PheWAS) to assess its safety as a potential therapeutic target candidate.

Results: Six key immunological indicators were identified for model construction: cerebrospinal fluid /serum IgG quotient (QIgG), lymphocyte count, double negative T cell count, double positive T cell count, NK cell count, and T cell percentage. The RF, XGBoost, and LGBM models demonstrated high predictive performance, with AUC values of 0.978, 0.917, and 0.900, and accuracies of 0.940, 0.916, and 0.831, respectively. Anti-NMDAR antibody titers in cerebrospinal fluid decreased (from 1:3.2 to 1:1) following PAIA treatment in a single patient. Cell communication analysis revealed enhanced intercellular signaling in the high-Npm1 expression group, particularly involving the PSAP pathway. Spatial transcriptomics confirmed upregulated Npm1 expression in EAE lesions. PheWAS indicated no significant off-target effects associated with NPM1.

Conclusion: This study provides an interpretable prognostic framework for AE, presents preliminary evidence for PAIA, and nominates NPM1 as a potential mechanistic player in disease pathogenesis. Its suitability as a potential therapeutic target requires further safety validation, despite the absence of significant signals in the preliminary PheWAS.

目的:利用免疫学指标建立可解释的自身免疫性脑炎(AE)预后预测模型,并通过多组学方法探讨核磷蛋白(NPM1)在疾病发病机制中的潜在作用。方法:我们招募了被诊断为抗体阳性AE的患者,并分析了广泛的免疫指标。使用八种机器学习算法开发了预后预测模型,并在独立队列中进行了验证。通过SHapley加性解释(SHAP)分析增强了模型的可解释性。我们进一步评估了蛋白A免疫吸附(PAIA)在降低致病性抗体方面的治疗潜力。在这些临床和免疫学发现的基础上,我们试图通过探索核磷蛋白(NPM1)的作用来研究其潜在机制。为此,我们在实验性自身免疫性脑脊髓炎(EAE)模型中整合了单细胞RNA测序和空间转录组学,并进行了全现象关联研究(PheWAS),以评估其作为潜在治疗靶点的安全性。结果:确定了构建模型的6个关键免疫学指标:脑脊液/血清IgG商(QIgG)、淋巴细胞计数、双阴性T细胞计数、双阳性T细胞计数、NK细胞计数、T细胞百分比。RF、XGBoost和LGBM模型的AUC分别为0.978、0.917和0.900,准确率分别为0.940、0.916和0.831。单例患者接受PAIA治疗后,脑脊液中抗nmdar抗体滴度下降(从1:3.2降至1:1)。细胞通讯分析显示,npm1高表达组的细胞间信号传导增强,特别是涉及PSAP途径。空间转录组学证实了EAE病变中Npm1的表达上调。PheWAS未显示与NPM1相关的显著脱靶效应。结论:本研究为AE提供了一个可解释的预后框架,为PAIA提供了初步证据,并表明NPM1可能在AE的发病机制中发挥作用。尽管在初步的PheWAS中没有明显的信号,但它作为潜在治疗靶点的适用性需要进一步的安全性验证。
{"title":"Machine learning and multi-omics integration identifies immunological predictors and mechanistic insights in autoimmune encephalitis.","authors":"Junshuang Guo, Yizhou Zou","doi":"10.1007/s00011-025-02180-8","DOIUrl":"https://doi.org/10.1007/s00011-025-02180-8","url":null,"abstract":"<p><strong>Objective: </strong>To develop an interpretable prognostic prediction model for autoimmune encephalitis (AE) using immunological indicators and to investigate the potential role of nucleophosmin (NPM1) in disease pathogenesis through multi-omics approaches.</p><p><strong>Methods: </strong>We enrolled patients diagnosed with antibody-positive AE and analyzed a broad panel of immunological indicators. Prognostic prediction models were developed using eight machine learning algorithms and validated in an independent cohort. Model interpretability was enhanced through SHapley Additive exPlanations (SHAP) analysis. We further evaluated the therapeutic potential of protein A immunoadsorption (PAIA) in reducing pathogenic antibodies. Building upon these clinical and immunological findings, we sought to investigate the underlying mechanisms by exploring the role of nucleophosmin (NPM1). To this end, we integrated single-cell RNA sequencing and spatial transcriptomics in an experimental autoimmune encephalomyelitis (EAE) model and conducted a phenome-wide association study (PheWAS) to assess its safety as a potential therapeutic target candidate.</p><p><strong>Results: </strong>Six key immunological indicators were identified for model construction: cerebrospinal fluid /serum IgG quotient (QIgG), lymphocyte count, double negative T cell count, double positive T cell count, NK cell count, and T cell percentage. The RF, XGBoost, and LGBM models demonstrated high predictive performance, with AUC values of 0.978, 0.917, and 0.900, and accuracies of 0.940, 0.916, and 0.831, respectively. Anti-NMDAR antibody titers in cerebrospinal fluid decreased (from 1:3.2 to 1:1) following PAIA treatment in a single patient. Cell communication analysis revealed enhanced intercellular signaling in the high-Npm1 expression group, particularly involving the PSAP pathway. Spatial transcriptomics confirmed upregulated Npm1 expression in EAE lesions. PheWAS indicated no significant off-target effects associated with NPM1.</p><p><strong>Conclusion: </strong>This study provides an interpretable prognostic framework for AE, presents preliminary evidence for PAIA, and nominates NPM1 as a potential mechanistic player in disease pathogenesis. Its suitability as a potential therapeutic target requires further safety validation, despite the absence of significant signals in the preliminary PheWAS.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"18"},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145966087","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cumulative environmental exposures adversely impact social behaviour and are associated with dysregulation of genes and proteins involved in epigenetic, ribosomal, and immune regulation in male mice. 在雄性小鼠中,累积的环境暴露会对社会行为产生不利影响,并与参与表观遗传、核糖体和免疫调节的基因和蛋白质失调有关。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02152-y
Morgan C Bucknor, Brooke A Keating, Velda X Han, Brian S Gloss, Pinki Dey, Nader Aryamanesh, Lee L Marshall, Mark E Graham, Ruwani Dissanayake, Xianzhong Lau, Shrujna Patel, Stela P Petkova, Peter Valtchev, Anand Gururajan, Russell C Dale, Markus J Hofer

Objective: This study investigated how cumulative environmental exposures influence offspring behaviour and inflammation-related molecular signatures in the brain and peripheral immune system.

Methods: A novel "triple-hit" mouse model was developed using C57Bl/6JAusB mice (N = 70), combining preconceptual social stress, antenatal high-fat diet, and a postnatal immune challenge (poly(I:C), 10 mg/kg). At 12 weeks, offspring underwent behavioural tests relevant to neurodevelopmental disorders (NDDs), including the Elevated Plus Maze, 3-Chamber Social Preference, Self-Grooming, and Marble Burying. A composite NDD-risk index was calculated. Single-cell RNA sequencing (scRNA-seq) and bulk proteomics were performed on male triple-hit offspring to identify differentially expressed genes and proteins associated with inflammatory pathways.

Results: Male triple-hit offspring showed elevated NDD-related behavioural risk and social deficits, not observed in females. scRNA-seq revealed altered inflammatory and ribosomal pathways in brain glia and peripheral immune cells. Proteomic analysis showed decreased abundance of proteins involved in inflammation, translation, chromatin remodelling, and synaptic function in both brain and blood.

Conclusion: Combined environmental stressors may drive male-specific behavioural and inflammatory changes relevant to NDDs. The identification of overlapping inflammatory signatures in brain and peripheral immune cells supports a role for shared immune mechanisms in brain-immune axis dysfunction. However, these pathway-level findings should be interpreted as preliminary hypotheses and warrant independent validation to confirm their mechanistic significance.

目的:本研究探讨了累积环境暴露如何影响后代行为以及大脑和外周免疫系统中与炎症相关的分子特征。方法:采用C57Bl/6JAusB小鼠(N = 70),结合孕前社会应激、产前高脂饮食和产后免疫刺激(poly(I:C), 10 mg/kg),建立新型“三重打击”小鼠模型。12周时,幼崽接受了与神经发育障碍(ndd)相关的行为测试,包括高台迷宫、三室社会偏好、自我梳理和大理石埋葬。计算ndd风险综合指数。对雄性三击后代进行单细胞RNA测序(scRNA-seq)和大量蛋白质组学,以鉴定与炎症途径相关的差异表达基因和蛋白质。结果:雄性三击后代表现出更高的ndd相关行为风险和社会缺陷,而在雌性中没有观察到。scRNA-seq揭示了脑胶质细胞和外周免疫细胞中炎症和核糖体通路的改变。蛋白质组学分析显示,在大脑和血液中,参与炎症、翻译、染色质重塑和突触功能的蛋白质丰度降低。结论:综合环境压力因素可能驱动与ndd相关的男性特异性行为和炎症变化。大脑和外周免疫细胞中重叠炎症特征的识别支持了共享免疫机制在脑免疫轴功能障碍中的作用。然而,这些通路水平的发现应该被解释为初步假设,并保证独立验证以确认其机制意义。
{"title":"Cumulative environmental exposures adversely impact social behaviour and are associated with dysregulation of genes and proteins involved in epigenetic, ribosomal, and immune regulation in male mice.","authors":"Morgan C Bucknor, Brooke A Keating, Velda X Han, Brian S Gloss, Pinki Dey, Nader Aryamanesh, Lee L Marshall, Mark E Graham, Ruwani Dissanayake, Xianzhong Lau, Shrujna Patel, Stela P Petkova, Peter Valtchev, Anand Gururajan, Russell C Dale, Markus J Hofer","doi":"10.1007/s00011-025-02152-y","DOIUrl":"10.1007/s00011-025-02152-y","url":null,"abstract":"<p><strong>Objective: </strong>This study investigated how cumulative environmental exposures influence offspring behaviour and inflammation-related molecular signatures in the brain and peripheral immune system.</p><p><strong>Methods: </strong>A novel \"triple-hit\" mouse model was developed using C57Bl/6JAusB mice (N = 70), combining preconceptual social stress, antenatal high-fat diet, and a postnatal immune challenge (poly(I:C), 10 mg/kg). At 12 weeks, offspring underwent behavioural tests relevant to neurodevelopmental disorders (NDDs), including the Elevated Plus Maze, 3-Chamber Social Preference, Self-Grooming, and Marble Burying. A composite NDD-risk index was calculated. Single-cell RNA sequencing (scRNA-seq) and bulk proteomics were performed on male triple-hit offspring to identify differentially expressed genes and proteins associated with inflammatory pathways.</p><p><strong>Results: </strong>Male triple-hit offspring showed elevated NDD-related behavioural risk and social deficits, not observed in females. scRNA-seq revealed altered inflammatory and ribosomal pathways in brain glia and peripheral immune cells. Proteomic analysis showed decreased abundance of proteins involved in inflammation, translation, chromatin remodelling, and synaptic function in both brain and blood.</p><p><strong>Conclusion: </strong>Combined environmental stressors may drive male-specific behavioural and inflammatory changes relevant to NDDs. The identification of overlapping inflammatory signatures in brain and peripheral immune cells supports a role for shared immune mechanisms in brain-immune axis dysfunction. However, these pathway-level findings should be interpreted as preliminary hypotheses and warrant independent validation to confirm their mechanistic significance.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"17"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783315/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
N4BP1 acts as a potent negative regulator of IL-17 signaling by blocking the translation of Act1 mRNA. N4BP1通过阻断Act1 mRNA的翻译,作为IL-17信号传导的有效负调控因子。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02159-5
Yanli Li, Zhuolong Xu, Xiaoyu Zhang, Zijian Zhang, Changsheng Ji, Xiaohong Guo, Jie Zhang, Peida Feng, Renfang Mao, Zhaoxiu Liu, Yang Lu, Yihui Fan

Background: Interleukin 17 (IL-17) is a primary pathogenic cytokine, and antibodies blocking its function are clinically approved for treating psoriasis. Although Act1 (TRAF3IP2) is an essential multifunctional adaptor in IL-17 signaling, its regulatory mechanisms remain poorly understood. In this study, the role of endoribonuclease N4BP1 in regulating the IL-17 signaling pathway was characterized.

Methods: N4BP1 was knocked out in both in vivo and in vitro experimental models to detect alterations in the IL-17 signaling pathway. Moreover, the specific mechanism by which N4BP1 exerts its regulatory effect was explored by examining the stability, degradation rate, transcription and translation rate of key proteins.

Results: N4BP1 deficiency markedly enhanced IL-17-induced expression of proinflammatory mediators, including CXCL1, CCL20, and MMP9. Unexpectedly, the mRNA stability of CXCL1, CCL20, and MMP9 was largely unaffected by N4BP1 knockout. Further investigation revealed that N4BP1-deficient cells exhibited elevated MAPK phosphorylation, particularly of p38. Pharmacological inhibition of p38 substantially reduced CXCL1, CCL20, and MMP9 levels in N4BP1-deficient cells. This hyperactivation of MAPKs was attributed to an increased protein level of Act1 in N4BP1-deficient cells. Silencing of Act1 with shRNAs in N4BP1-deficient cells greatly diminished the upregulation of CXCL1, CCL20 and MMP9. The elevated Act1 protein level in N4BP1-deficient cells was not due to enhanced Act1 mRNA stability. Instead, polysome profiling demonstrated a pronounced enrichment of Act1 mRNA in the translationally active polysome fraction in N4BP1-deficient cells. In vivo, under pathological stimuli such as IMQ or aging, N4BP1-deficient mice exhibited increased Act1 protein, MAPK phosphorylation, and increased expression of IL-17 downstream genes, including CXCL1, CCL20, and MMP9. Pharmacological inhibition of Act1 ameliorates IMQ-induced skin damage, with a more pronounced therapeutic effect observed in N4BP1 KO mice.

Conclusions: These findings collectively establish that N4BP1 is a potent negative regulator of IL-17 signaling that suppresses the translation of Act1 mRNA.

背景:白细胞介素17 (IL-17)是一种原发性致病细胞因子,阻断其功能的抗体已被临床批准用于治疗银屑病。尽管Act1 (TRAF3IP2)在IL-17信号传导中是一个重要的多功能适配器,但其调控机制仍然知之甚少。本研究表征了核糖核酸内切酶N4BP1在调节IL-17信号通路中的作用。方法:在体内和体外实验模型中敲除N4BP1,检测IL-17信号通路的变化。此外,通过检测关键蛋白的稳定性、降解率、转录和翻译率,探索N4BP1发挥其调控作用的具体机制。结果:N4BP1缺乏显著增强il -17诱导的促炎介质的表达,包括CXCL1、CCL20和MMP9。出乎意料的是,CXCL1、CCL20和MMP9的mRNA稳定性在很大程度上不受N4BP1敲除的影响。进一步的研究表明,n4bp1缺陷细胞表现出MAPK磷酸化升高,尤其是p38。药理抑制p38可显著降低n4bp1缺陷细胞中CXCL1、CCL20和MMP9的水平。这种MAPKs的过度激活归因于n4bp1缺陷细胞中Act1蛋白水平的升高。在n4bp1缺陷细胞中,用shrna沉默Act1可显著降低CXCL1、CCL20和MMP9的上调。n4bp1缺陷细胞中Act1蛋白水平升高并非由于Act1 mRNA稳定性增强所致。相反,在n4bp1缺乏的细胞中,多聚体分析显示,在翻译活性多聚体部分中,Act1 mRNA显著富集。在体内,在IMQ或衰老等病理刺激下,n4bp1缺陷小鼠表现出Act1蛋白、MAPK磷酸化增加,IL-17下游基因CXCL1、CCL20和MMP9表达增加。药理抑制Act1可改善imq诱导的皮肤损伤,在N4BP1 KO小鼠中观察到更明显的治疗效果。结论:这些发现共同证明N4BP1是IL-17信号的有效负调控因子,可抑制Act1 mRNA的翻译。
{"title":"N4BP1 acts as a potent negative regulator of IL-17 signaling by blocking the translation of Act1 mRNA.","authors":"Yanli Li, Zhuolong Xu, Xiaoyu Zhang, Zijian Zhang, Changsheng Ji, Xiaohong Guo, Jie Zhang, Peida Feng, Renfang Mao, Zhaoxiu Liu, Yang Lu, Yihui Fan","doi":"10.1007/s00011-025-02159-5","DOIUrl":"https://doi.org/10.1007/s00011-025-02159-5","url":null,"abstract":"<p><strong>Background: </strong>Interleukin 17 (IL-17) is a primary pathogenic cytokine, and antibodies blocking its function are clinically approved for treating psoriasis. Although Act1 (TRAF3IP2) is an essential multifunctional adaptor in IL-17 signaling, its regulatory mechanisms remain poorly understood. In this study, the role of endoribonuclease N4BP1 in regulating the IL-17 signaling pathway was characterized.</p><p><strong>Methods: </strong>N4BP1 was knocked out in both in vivo and in vitro experimental models to detect alterations in the IL-17 signaling pathway. Moreover, the specific mechanism by which N4BP1 exerts its regulatory effect was explored by examining the stability, degradation rate, transcription and translation rate of key proteins.</p><p><strong>Results: </strong>N4BP1 deficiency markedly enhanced IL-17-induced expression of proinflammatory mediators, including CXCL1, CCL20, and MMP9. Unexpectedly, the mRNA stability of CXCL1, CCL20, and MMP9 was largely unaffected by N4BP1 knockout. Further investigation revealed that N4BP1-deficient cells exhibited elevated MAPK phosphorylation, particularly of p38. Pharmacological inhibition of p38 substantially reduced CXCL1, CCL20, and MMP9 levels in N4BP1-deficient cells. This hyperactivation of MAPKs was attributed to an increased protein level of Act1 in N4BP1-deficient cells. Silencing of Act1 with shRNAs in N4BP1-deficient cells greatly diminished the upregulation of CXCL1, CCL20 and MMP9. The elevated Act1 protein level in N4BP1-deficient cells was not due to enhanced Act1 mRNA stability. Instead, polysome profiling demonstrated a pronounced enrichment of Act1 mRNA in the translationally active polysome fraction in N4BP1-deficient cells. In vivo, under pathological stimuli such as IMQ or aging, N4BP1-deficient mice exhibited increased Act1 protein, MAPK phosphorylation, and increased expression of IL-17 downstream genes, including CXCL1, CCL20, and MMP9. Pharmacological inhibition of Act1 ameliorates IMQ-induced skin damage, with a more pronounced therapeutic effect observed in N4BP1 KO mice.</p><p><strong>Conclusions: </strong>These findings collectively establish that N4BP1 is a potent negative regulator of IL-17 signaling that suppresses the translation of Act1 mRNA.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"14"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932967","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of pyroptosis during HIV infection. 热亡在HIV感染中的作用。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02164-8
Mahmoud M Yaseen, Nizar M Abuharfeil, Homa Darmani, Mohammed N Shatnawi

Background: Pyroptosis, a proinflammatory form of programmed cell death, has emerged as a central driver of chronic inflammation, CD4⁺ T cell depletion, and non-AIDS comorbidities in HIV infection. This review synthesizes current evidence on the molecular mechanisms and pathological consequences of pyroptosis in HIV.

Methods: We conducted a comprehensive analysis of the literature, examining the molecular pathways of pyroptosis triggered by abortive HIV infection, the roles of specific inflammasomes (e.g., AIM2, NLRP3, CARD8) and viral proteins, and the subsequent amplification of inflammation through cytokine release and gut barrier dysfunction.

Results: Abortive infection in resting CD4⁺ T cells generates cytosolic viral DNA, activating inflammasomes (primarily AIM2/IFI16) and executing pyroptosis via GSDMD. This process initiates a vicious cycle of immune activation, mucosal damage, microbial translocation, and systemic inflammation, leading to CD4⁺ T cell loss, reservoir persistence, and end-organ damage. Therapeutic targeting of key nodes (e.g., caspase-1, NLRP3, GSDMD) shows promise in preclinical models.

Conclusion: Pyroptosis is a critical pathological engine in HIV, linking viral infection to chronic immunodeficiency and comorbidities. Adjunctive therapies targeting this pathway may reduce inflammation, preserve immune function, and support strategies toward a functional cure.

背景:焦亡是程序性细胞死亡的一种促炎形式,已经成为HIV感染中慢性炎症、CD4 + T细胞耗竭和非艾滋病合并症的主要驱动因素。本文综述了目前关于HIV病毒焦亡的分子机制和病理后果的证据。方法:综合文献分析,探讨流产性HIV感染引发热亡的分子途径,特异性炎症小体(如AIM2、NLRP3、CARD8)和病毒蛋白的作用,以及随后通过细胞因子释放和肠道屏障功能障碍放大炎症。结果:静止CD4 + T细胞的流产感染产生胞质病毒DNA,激活炎症小体(主要是AIM2/IFI16),并通过GSDMD进行焦亡。这一过程启动了免疫激活、粘膜损伤、微生物易位和全身性炎症的恶性循环,导致CD4 + T细胞丢失、储层持续存在和终末器官损伤。关键节点(如caspase-1, NLRP3, GSDMD)的治疗靶向在临床前模型中显示出希望。结论:焦亡是HIV的一个关键病理引擎,将病毒感染与慢性免疫缺陷和合并症联系起来。针对这一途径的辅助治疗可能会减少炎症,保持免疫功能,并支持功能性治愈的策略。
{"title":"The role of pyroptosis during HIV infection.","authors":"Mahmoud M Yaseen, Nizar M Abuharfeil, Homa Darmani, Mohammed N Shatnawi","doi":"10.1007/s00011-025-02164-8","DOIUrl":"https://doi.org/10.1007/s00011-025-02164-8","url":null,"abstract":"<p><strong>Background: </strong>Pyroptosis, a proinflammatory form of programmed cell death, has emerged as a central driver of chronic inflammation, CD4⁺ T cell depletion, and non-AIDS comorbidities in HIV infection. This review synthesizes current evidence on the molecular mechanisms and pathological consequences of pyroptosis in HIV.</p><p><strong>Methods: </strong>We conducted a comprehensive analysis of the literature, examining the molecular pathways of pyroptosis triggered by abortive HIV infection, the roles of specific inflammasomes (e.g., AIM2, NLRP3, CARD8) and viral proteins, and the subsequent amplification of inflammation through cytokine release and gut barrier dysfunction.</p><p><strong>Results: </strong>Abortive infection in resting CD4⁺ T cells generates cytosolic viral DNA, activating inflammasomes (primarily AIM2/IFI16) and executing pyroptosis via GSDMD. This process initiates a vicious cycle of immune activation, mucosal damage, microbial translocation, and systemic inflammation, leading to CD4⁺ T cell loss, reservoir persistence, and end-organ damage. Therapeutic targeting of key nodes (e.g., caspase-1, NLRP3, GSDMD) shows promise in preclinical models.</p><p><strong>Conclusion: </strong>Pyroptosis is a critical pathological engine in HIV, linking viral infection to chronic immunodeficiency and comorbidities. Adjunctive therapies targeting this pathway may reduce inflammation, preserve immune function, and support strategies toward a functional cure.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"12"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting neuroinflammation in neurodegenerative disorders: the emerging potential of semaglutide. 靶向神经退行性疾病的神经炎症:西马鲁肽的新潜力。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02166-6
Vito Evola, Mayur S Parmar

Background: Chronic neuroinflammation is increasingly recognized not as a secondary effect but as a primary driver of neurodegenerative disease progression. In conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Huntington's disease (HD), and Lewy body dementia (LBD), dysregulated glial activity, marked by sustained microglial and astrocytic activation, initiates a cascade of cytokine release, oxidative stress, and impaired neuronal support. This review synthesizes recent advances in understanding these shared inflammatory processes, emphasizing how glia-centric pathology shapes disease-specific trajectories and therapeutic responses.

Findings: Within this framework, we evaluate the therapeutic potential of semaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA) with emerging neuroprotective properties. Preclinical studies suggest that semaglutide can suppress pro-inflammatory signaling, mitigate oxidative injury, and enhance key anti-inflammatory and neuroprotective pathways that restore trophic support and cellular resilience. We also examine real-world evidence and emerging human clinical trial data, which recently demonstrated that semaglutide rapidly modulates AD pathology by significantly reducing cerebrospinal fluid (CSF) levels of p-tau, t-tau, and neurogranin, and promoting a less inflammatory CD8+T-cell signature. In addition to reduction in neuroinflammation marker, YKL-40. While subsequent large-scale Phase 3 trials in early AD did not meet primary cognitive endpoints (CDR-SB) despite favorable biomarker modulation.

Conclusion: Positioning semaglutide as a therapeutic option targeting neuroinflammation-mediated neuropathology, this review underscores its potential for repurposing as a disease-modifying therapy across diverse neurodegenerative disorders and highlights the urgent need for targeted trials in MS, ALS, FTD, HD, and LBD-conditions that remain without effective immunomodulatory treatments despite clear inflammatory origins. However, while direct CSF measurements confirm limited but measurable BBB penetration, the clinical translation of its effects remains a key challenge.

背景:慢性神经炎症越来越被认为不是次要的影响,而是神经退行性疾病进展的主要驱动因素。在阿尔茨海默病(AD)、帕金森病(PD)、多发性硬化症(MS)、肌萎缩性侧索硬化症(ALS)、额颞叶痴呆(FTD)、亨廷顿氏病(HD)和路易体痴呆(LBD)等疾病中,神经胶质活性失调,以持续的小胶质细胞和星形胶质细胞激活为标志,引发细胞因子释放、氧化应激和神经元支持受损的级联反应。这篇综述综合了了解这些共同炎症过程的最新进展,强调了以胶质细胞为中心的病理如何塑造疾病特异性轨迹和治疗反应。研究结果:在这个框架内,我们评估了semaglutide的治疗潜力,semaglutide是一种具有新兴神经保护特性的胰高血糖素样肽-1受体激动剂(GLP-1RA)。临床前研究表明,西马鲁肽可以抑制促炎信号,减轻氧化损伤,增强关键的抗炎和神经保护通路,恢复营养支持和细胞弹性。我们还研究了现实世界的证据和新兴的人类临床试验数据,这些数据最近表明,semaglutide通过显著降低脑脊液(CSF)中p-tau、t-tau和神经颗粒蛋白的水平,并促进炎症性较低的CD8+ t细胞特征,快速调节AD病理。除了降低神经炎症标志物YKL-40。而随后针对早期阿尔茨海默病的大规模3期试验,尽管有有利的生物标志物调节,但并未达到主要认知终点(CDR-SB)。结论:本综述将semaglutide定位为针对神经炎症介导的神经病理学的治疗选择,强调了其作为多种神经退行性疾病的疾病改善治疗的潜力,并强调了在MS, ALS, FTD, HD和lbd -尽管炎症起源明确,但仍没有有效免疫调节治疗的疾病中进行靶向试验的迫切需要。然而,虽然直接脑脊液测量证实有限但可测量的血脑屏障穿透,但其效果的临床翻译仍然是一个关键挑战。
{"title":"Targeting neuroinflammation in neurodegenerative disorders: the emerging potential of semaglutide.","authors":"Vito Evola, Mayur S Parmar","doi":"10.1007/s00011-025-02166-6","DOIUrl":"https://doi.org/10.1007/s00011-025-02166-6","url":null,"abstract":"<p><strong>Background: </strong>Chronic neuroinflammation is increasingly recognized not as a secondary effect but as a primary driver of neurodegenerative disease progression. In conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Huntington's disease (HD), and Lewy body dementia (LBD), dysregulated glial activity, marked by sustained microglial and astrocytic activation, initiates a cascade of cytokine release, oxidative stress, and impaired neuronal support. This review synthesizes recent advances in understanding these shared inflammatory processes, emphasizing how glia-centric pathology shapes disease-specific trajectories and therapeutic responses.</p><p><strong>Findings: </strong>Within this framework, we evaluate the therapeutic potential of semaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA) with emerging neuroprotective properties. Preclinical studies suggest that semaglutide can suppress pro-inflammatory signaling, mitigate oxidative injury, and enhance key anti-inflammatory and neuroprotective pathways that restore trophic support and cellular resilience. We also examine real-world evidence and emerging human clinical trial data, which recently demonstrated that semaglutide rapidly modulates AD pathology by significantly reducing cerebrospinal fluid (CSF) levels of p-tau, t-tau, and neurogranin, and promoting a less inflammatory CD8<sup>+</sup>T-cell signature. In addition to reduction in neuroinflammation marker, YKL-40. While subsequent large-scale Phase 3 trials in early AD did not meet primary cognitive endpoints (CDR-SB) despite favorable biomarker modulation.</p><p><strong>Conclusion: </strong>Positioning semaglutide as a therapeutic option targeting neuroinflammation-mediated neuropathology, this review underscores its potential for repurposing as a disease-modifying therapy across diverse neurodegenerative disorders and highlights the urgent need for targeted trials in MS, ALS, FTD, HD, and LBD-conditions that remain without effective immunomodulatory treatments despite clear inflammatory origins. However, while direct CSF measurements confirm limited but measurable BBB penetration, the clinical translation of its effects remains a key challenge.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"13"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932997","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Knockdown of caspase-activated DNase and B-cell lymphoma 2 inhibits cell proliferation and drug resistance in TP53-mutant multiple myeloma. caspase激活的DNase和b细胞淋巴瘤2的敲低抑制tp53突变型多发性骨髓瘤细胞增殖和耐药。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02151-z
Faqing Tian, Jinxing Wang, Pengwei Zhang, Jian Jiang, Xiaohui Cheng, Juheng Li, Meiqin Tang, Jiaoyang Fan, Pu Yan, Guoxin Zhao
{"title":"Knockdown of caspase-activated DNase and B-cell lymphoma 2 inhibits cell proliferation and drug resistance in TP53-mutant multiple myeloma.","authors":"Faqing Tian, Jinxing Wang, Pengwei Zhang, Jian Jiang, Xiaohui Cheng, Juheng Li, Meiqin Tang, Jiaoyang Fan, Pu Yan, Guoxin Zhao","doi":"10.1007/s00011-025-02151-z","DOIUrl":"https://doi.org/10.1007/s00011-025-02151-z","url":null,"abstract":"","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"16"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dual-function cytokines as modulators of autophagy: reprogramming inflammatory resolution in severe COVID-19. 双功能细胞因子作为自噬的调节剂:重编程重症COVID-19的炎症消退
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02169-3
Sohrab Khan, Ping Tang, Pingchang Yang, Jing Li, Haiqiang Wu

Background: Acute respiratory distress syndrome (ARDS) and systemic immune-mediated damage are two of the severe COVID-19 outcomes that are primarily caused by cytokine storms triggered by dysregulated immune responses. The limited benefits of current immunosuppressive treatments highlight the need for mechanistic understanding to direct focused interventions.

Objective: The dual functions of cytokines in controlling autophagy during SARS-CoV-2 infection are examined in this review, along with the potential for autophagy modulation to limit hyperinflammation and restore immune homeostasis.

Key findings: Emerging evidence suggests that autophagy critically modulates the balance between pro- and anti-inflammatory cytokines in COVID-19. Through anti-inflammatory feedback mechanisms, cytokines contribute to resolution while promoting inflammation in the early stages. The IRE1α-XBP1 axis is activated by SARS-CoV-2-induced endoplasmic reticulum stress, which increases cytokine production and modifies autophagic flux. Concurrently, extracellular vesicles containing cytokines, damage-associated molecular patterns, and viral components are released as secretory autophagy reroutes cytoplasmic cargo toward multivesicular bodies and amphisomes, increasing paracrine immune activation. Suppressed degradative autophagy and increased secretory autophagy-mediated inflammatory signaling are the hallmarks of this pathological state.

Conclusions: In severe COVID-19, targeted autophagy restoration is a promising therapeutic approach to restore immune responses, reduce excessive inflammation, and encourage the resolution of cytokine storms. Restoring immune homeostasis through more targeted immunointerventions may be made possible by modifying autophagy pathways.

背景:急性呼吸窘迫综合征(ARDS)和全身免疫介导的损伤是COVID-19的两种严重结局,主要由免疫反应失调引发的细胞因子风暴引起。当前免疫抑制治疗的有限益处突出了对机制的理解以指导重点干预的必要性。目的:本综述探讨了细胞因子在SARS-CoV-2感染过程中控制自噬的双重功能,以及调节自噬以限制过度炎症和恢复免疫稳态的潜力。新发现的证据表明,自噬在COVID-19中对促炎性细胞因子和抗炎性细胞因子之间的平衡起关键调节作用。通过抗炎反馈机制,细胞因子在早期促进炎症的同时有助于解决问题。IRE1α-XBP1轴被sars - cov -2诱导的内质网应激激活,增加细胞因子的产生,改变自噬通量。同时,含有细胞因子、损伤相关分子模式和病毒成分的细胞外囊泡被释放,因为分泌性自噬将细胞质货物转向多泡体和两体,增加旁分泌免疫激活。抑制降解性自噬和增加分泌性自噬介导的炎症信号是这种病理状态的标志。结论:在重症COVID-19中,靶向自噬修复是一种很有希望的治疗方法,可以恢复免疫反应,减少过度炎症,并促进细胞因子风暴的解决。通过更有针对性的免疫干预,可以通过修改自噬途径来恢复免疫稳态。
{"title":"Dual-function cytokines as modulators of autophagy: reprogramming inflammatory resolution in severe COVID-19.","authors":"Sohrab Khan, Ping Tang, Pingchang Yang, Jing Li, Haiqiang Wu","doi":"10.1007/s00011-025-02169-3","DOIUrl":"https://doi.org/10.1007/s00011-025-02169-3","url":null,"abstract":"<p><strong>Background: </strong>Acute respiratory distress syndrome (ARDS) and systemic immune-mediated damage are two of the severe COVID-19 outcomes that are primarily caused by cytokine storms triggered by dysregulated immune responses. The limited benefits of current immunosuppressive treatments highlight the need for mechanistic understanding to direct focused interventions.</p><p><strong>Objective: </strong>The dual functions of cytokines in controlling autophagy during SARS-CoV-2 infection are examined in this review, along with the potential for autophagy modulation to limit hyperinflammation and restore immune homeostasis.</p><p><strong>Key findings: </strong>Emerging evidence suggests that autophagy critically modulates the balance between pro- and anti-inflammatory cytokines in COVID-19. Through anti-inflammatory feedback mechanisms, cytokines contribute to resolution while promoting inflammation in the early stages. The IRE1α-XBP1 axis is activated by SARS-CoV-2-induced endoplasmic reticulum stress, which increases cytokine production and modifies autophagic flux. Concurrently, extracellular vesicles containing cytokines, damage-associated molecular patterns, and viral components are released as secretory autophagy reroutes cytoplasmic cargo toward multivesicular bodies and amphisomes, increasing paracrine immune activation. Suppressed degradative autophagy and increased secretory autophagy-mediated inflammatory signaling are the hallmarks of this pathological state.</p><p><strong>Conclusions: </strong>In severe COVID-19, targeted autophagy restoration is a promising therapeutic approach to restore immune responses, reduce excessive inflammation, and encourage the resolution of cytokine storms. Restoring immune homeostasis through more targeted immunointerventions may be made possible by modifying autophagy pathways.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"11"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933037","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparison of cancer risks associated with JAK inhibitors and TNF inhibitors treatment in patients with rheumatoid arthritis: a systematic review and meta-analysis of real-world cohort studies. 类风湿关节炎患者JAK抑制剂和TNF抑制剂治疗相关癌症风险的比较:现实世界队列研究的系统回顾和荟萃分析
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-08 DOI: 10.1007/s00011-025-02170-w
Fu-Yu Yang, Yu-Chang Liu, Min-You Wu, Yao-Cheng Wu, Cheng-Hsien Hung

Introduction: Potential increased cancer risk associated with janus kinase inhibitors (JAKi) compared with anti-tumor necrosis factor inhibitors (TNFi) in patients with rheumatoid arthritis (RA) remains a concern. Published cohort studies have reported conflicting results, and the discrepancies between randomized trials and real-world data remain unclear. We conducted this systematic review and meta-analysis to assess this association.

Material/methods: We systematically searched PubMed, Embase and Cochrane Library for cohort studies up to January 31, 2025, comparing JAKi with TNFi and reporting cancer outcomes in RA patients. The primary outcome was overall cancer risk, and secondary outcomes included site-specific cancers. Pooled hazard ratios (HR) with 95% confidence intervals (CI) were calculated using a random-effects meta-analysis. Subgroup and sensitivity analyses were conducted to explore potential sources of heterogeneity. The certainty of evidence (CoE) were assessed using the GRADE framework.

Results: We included 5 cohort studies with 137,640 RA patients. Compared to TNFi, JAKi did not increase the risk of overall cancers (pooled HR: 1.06, 95% CI: 0.81-1.37; CoE: very low). Regarding secondary outcomes, JAKi was not linked to most cancers but increased the risk of non-melanoma skin cancer (NMSC) (HR: 1.21, 95% CI: 1.03-1.41; CoE: very low). The finding was consistent across multiple subgroup and sensitivity analyses.

Conclusion: This meta-analysis found no increase in overall cancer risk with JAKi compared to TNFi, but identified an increased risk of NMSC, suggesting the need for regular dermatologic surveillance.

在类风湿关节炎(RA)患者中,与抗肿瘤坏死因子抑制剂(TNFi)相比,janus激酶抑制剂(JAKi)的潜在癌症风险增加仍然是一个值得关注的问题。已发表的队列研究报告了相互矛盾的结果,随机试验与实际数据之间的差异仍不清楚。我们进行了系统回顾和荟萃分析来评估这种关联。材料/方法:我们系统地检索PubMed、Embase和Cochrane Library,检索截至2025年1月31日的队列研究,比较JAKi和TNFi,并报告RA患者的癌症结局。主要结果是总体癌症风险,次要结果包括部位特异性癌症。采用随机效应荟萃分析计算合并风险比(HR)和95%置信区间(CI)。进行亚组分析和敏感性分析以探索异质性的潜在来源。使用GRADE框架评估证据的确定性(CoE)。结果:我们纳入了5项队列研究,共137,640例RA患者。与TNFi相比,JAKi没有增加总体癌症的风险(合并HR: 1.06, 95% CI: 0.81-1.37; CoE:非常低)。关于次要结局,JAKi与大多数癌症无关,但增加了非黑色素瘤皮肤癌(NMSC)的风险(HR: 1.21, 95% CI: 1.03-1.41; CoE:非常低)。这一发现在多个亚组和敏感性分析中是一致的。结论:该荟萃分析发现,与TNFi相比,JAKi的总体癌症风险没有增加,但确定了NMSC的风险增加,这表明需要定期进行皮肤病学监测。
{"title":"Comparison of cancer risks associated with JAK inhibitors and TNF inhibitors treatment in patients with rheumatoid arthritis: a systematic review and meta-analysis of real-world cohort studies.","authors":"Fu-Yu Yang, Yu-Chang Liu, Min-You Wu, Yao-Cheng Wu, Cheng-Hsien Hung","doi":"10.1007/s00011-025-02170-w","DOIUrl":"10.1007/s00011-025-02170-w","url":null,"abstract":"<p><strong>Introduction: </strong>Potential increased cancer risk associated with janus kinase inhibitors (JAKi) compared with anti-tumor necrosis factor inhibitors (TNFi) in patients with rheumatoid arthritis (RA) remains a concern. Published cohort studies have reported conflicting results, and the discrepancies between randomized trials and real-world data remain unclear. We conducted this systematic review and meta-analysis to assess this association.</p><p><strong>Material/methods: </strong>We systematically searched PubMed, Embase and Cochrane Library for cohort studies up to January 31, 2025, comparing JAKi with TNFi and reporting cancer outcomes in RA patients. The primary outcome was overall cancer risk, and secondary outcomes included site-specific cancers. Pooled hazard ratios (HR) with 95% confidence intervals (CI) were calculated using a random-effects meta-analysis. Subgroup and sensitivity analyses were conducted to explore potential sources of heterogeneity. The certainty of evidence (CoE) were assessed using the GRADE framework.</p><p><strong>Results: </strong>We included 5 cohort studies with 137,640 RA patients. Compared to TNFi, JAKi did not increase the risk of overall cancers (pooled HR: 1.06, 95% CI: 0.81-1.37; CoE: very low). Regarding secondary outcomes, JAKi was not linked to most cancers but increased the risk of non-melanoma skin cancer (NMSC) (HR: 1.21, 95% CI: 1.03-1.41; CoE: very low). The finding was consistent across multiple subgroup and sensitivity analyses.</p><p><strong>Conclusion: </strong>This meta-analysis found no increase in overall cancer risk with JAKi compared to TNFi, but identified an increased risk of NMSC, suggesting the need for regular dermatologic surveillance.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"15"},"PeriodicalIF":5.4,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932900","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The pivotal role and intervention strategies of BMAL1 mediated circadian clock dysregulation in intervertebral disc degeneration. BMAL1介导的生物钟失调在椎间盘退变中的关键作用和干预策略。
IF 5.4 3区 医学 Q2 CELL BIOLOGY Pub Date : 2026-01-07 DOI: 10.1007/s00011-025-02167-5
Jingwen Chen, Yingjin Luo, Feilong Li, Yuheng He, Yang Zhou, Li Peng, Yong Liu, Zongchao Liu, Chao Song, Zhenglong Wang

Background: Intervertebral disc degeneration (IVDD), a primary cause of chronic low back pain, involves extracellular matrix (ECM) degradation and nucleus pulposus cell apoptosis. While traditionally linked to mechanical stress, inflammation, oxidative stress, and metabolic dysfunction, emerging evidence positions circadian clock disruption as the central hub integrating these factors. Under physiological conditions, the core clock genes BMAL1/CLOCK regulate ECM homeostasis.

Results: In IVDD, however, this rhythm is disrupted: abnormal mechanical stress inhibits BMAL1 via the RhoA/ROCK pathway; inflammation (e.g., IL-1β) suppresses BMAL1 transcription through NF-κB, creating a vicious cycle; aging-related oxidative stress and ferroptosis are exacerbated by BMAL1 epigenetic silencing; and metabolic disorders promote NLRP3 inflammasome activation via mTORC1-mediated autophagy suppression and miR-155-dependent BMAL1 mRNA decay. Consequently, circadian dysregulation accelerates ECM breakdown and mitochondrial apoptosis. This synthesis establishes a novel 'circadian-centric' model of IVDD, unifying multifactorial pathogenesis under the framework of rhythm disruption. It reveals precise pathways such as RhoA/BMAL1/ECM, bridging key mechanistic gaps. Therapeutically, this model advocates a paradigm shift from symptomatic management to circadian rhythm reconstruction. Potential strategies include restoring BMAL1 rhythmicity to reverse ECM catabolism, targeting the circadian-inflammatory axis (e.g., melatonin, IL-1β antagonists) to concurrently mitigate inflammation and oxidative damage, and employing chrono-therapeutic interventions such as timed mechanical loading or nighttime drug administration.

Conclusion: This review provides a foundational rationale for developing chrono-precise diagnostics and treatments, aiming to redefine IVDD management toward endogenous rhythm restoration.

背景:椎间盘退变(IVDD)是慢性腰痛的主要原因,涉及细胞外基质(ECM)降解和髓核细胞凋亡。虽然传统上与机械应力、炎症、氧化应激和代谢功能障碍有关,但新出现的证据表明,生物钟紊乱是整合这些因素的中心枢纽。生理条件下,核心时钟基因BMAL1/ clock调节ECM内稳态。结果:然而,在IVDD中,这种节律被打乱:异常的机械应力通过RhoA/ROCK途径抑制BMAL1;炎症(如IL-1β)通过NF-κB抑制BMAL1的转录,形成恶性循环;BMAL1的表观遗传沉默加剧了衰老相关的氧化应激和铁凋亡;代谢紊乱通过mtorc1介导的自噬抑制和mir -155依赖的BMAL1 mRNA衰变促进NLRP3炎性体活化。因此,昼夜节律失调加速了ECM分解和线粒体凋亡。这种综合建立了一种新的“以昼夜节律为中心”的IVDD模型,在节律紊乱的框架下统一了多因素发病机制。它揭示了RhoA/BMAL1/ECM等精确通路,弥合了关键的机制差距。在治疗上,该模型提倡从症状管理到昼夜节律重建的范式转变。潜在的策略包括恢复BMAL1节律性以逆转ECM分解代谢,靶向昼夜炎症轴(如褪黑激素、IL-1β拮抗剂)同时减轻炎症和氧化损伤,以及采用时间治疗干预措施,如定时机械负荷或夜间给药。结论:本综述为开发时间精确诊断和治疗提供了基础理论依据,旨在重新定义内源性心律恢复的IVDD管理。
{"title":"The pivotal role and intervention strategies of BMAL1 mediated circadian clock dysregulation in intervertebral disc degeneration.","authors":"Jingwen Chen, Yingjin Luo, Feilong Li, Yuheng He, Yang Zhou, Li Peng, Yong Liu, Zongchao Liu, Chao Song, Zhenglong Wang","doi":"10.1007/s00011-025-02167-5","DOIUrl":"https://doi.org/10.1007/s00011-025-02167-5","url":null,"abstract":"<p><strong>Background: </strong>Intervertebral disc degeneration (IVDD), a primary cause of chronic low back pain, involves extracellular matrix (ECM) degradation and nucleus pulposus cell apoptosis. While traditionally linked to mechanical stress, inflammation, oxidative stress, and metabolic dysfunction, emerging evidence positions circadian clock disruption as the central hub integrating these factors. Under physiological conditions, the core clock genes BMAL1/CLOCK regulate ECM homeostasis.</p><p><strong>Results: </strong>In IVDD, however, this rhythm is disrupted: abnormal mechanical stress inhibits BMAL1 via the RhoA/ROCK pathway; inflammation (e.g., IL-1β) suppresses BMAL1 transcription through NF-κB, creating a vicious cycle; aging-related oxidative stress and ferroptosis are exacerbated by BMAL1 epigenetic silencing; and metabolic disorders promote NLRP3 inflammasome activation via mTORC1-mediated autophagy suppression and miR-155-dependent BMAL1 mRNA decay. Consequently, circadian dysregulation accelerates ECM breakdown and mitochondrial apoptosis. This synthesis establishes a novel 'circadian-centric' model of IVDD, unifying multifactorial pathogenesis under the framework of rhythm disruption. It reveals precise pathways such as RhoA/BMAL1/ECM, bridging key mechanistic gaps. Therapeutically, this model advocates a paradigm shift from symptomatic management to circadian rhythm reconstruction. Potential strategies include restoring BMAL1 rhythmicity to reverse ECM catabolism, targeting the circadian-inflammatory axis (e.g., melatonin, IL-1β antagonists) to concurrently mitigate inflammation and oxidative damage, and employing chrono-therapeutic interventions such as timed mechanical loading or nighttime drug administration.</p><p><strong>Conclusion: </strong>This review provides a foundational rationale for developing chrono-precise diagnostics and treatments, aiming to redefine IVDD management toward endogenous rhythm restoration.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"75 1","pages":"10"},"PeriodicalIF":5.4,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145911425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","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
Inflammation Research
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