首页 > 最新文献

Inflammation Research最新文献

英文 中文
APOE4 impairs autophagy and Aβ clearance by microglial cells. APOE4 会影响小胶质细胞的自噬和 Aβ 清除。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-04-01 DOI: 10.1007/s00011-025-02016-5
Rawan Bassal, Maria Rivkin-Natan, Alon Rabinovich, Daniel Moris Michaelson, Dan Frenkel, Ronit Pinkas-Kramarski

Alzheimer's disease (AD) is a predominant form of dementia in elderly. In sporadic AD and in families with higher risk of AD, correlation with apolipoprotein E4 (APOE) allele expression has been found. How APOE4 induces its pathological effects is still unclear. Several studies indicate that autophagy, a major degradation pathway trough the lysosome, may be compromised in AD. Here we studied, the effects of APOE isoforms expression in microglia cells. By using an in-situ model, the clearance of Aβ plaques from brain sections of transgenic 5xFAD mice by the APOE expressing microglia was examined. The results show that APOE4 microglia has Impairment In clearance of insoluble Aβ plaques as compared to APOE3 and APOE2 microglia. Furthermore, APOE4 affect the uptake of soluble Aβ. We found that microglia expressing APOE4 exhibit reduced autophagic flux as compared to those expressing APOE3. The autophagy inhibitor chloroquine also blocked Aβ plaque uptake in APOE3 expressing cells. Furthermore, we found that APOE4 expressing microglia have altered mitochondrial dynamics protein expression, mitochondrial morphology and mitochondrial activity compared to those expressing APOE2, and APOE3. Rapamycin treatment corrected Mitochondrial Membrane Potential in APOE4-expressing cells. Taken together, these findings suggest that APOE4 impairs the activation of autophagy, mitophagy, and Aβ clearance and that autophagy-inducing treatments, such as rapamycin, can enhance autophagy and mitochondrial functions in APOE4 expressing microglia. Our results reveal a direct link between APOE4 to autophagy activity in microglia, suggesting that the pathological effects of APOE4 could be counteracted by pharmacological treatments inducing autophagy, such as rapamycin.

{"title":"APOE4 impairs autophagy and Aβ clearance by microglial cells.","authors":"Rawan Bassal, Maria Rivkin-Natan, Alon Rabinovich, Daniel Moris Michaelson, Dan Frenkel, Ronit Pinkas-Kramarski","doi":"10.1007/s00011-025-02016-5","DOIUrl":"10.1007/s00011-025-02016-5","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a predominant form of dementia in elderly. In sporadic AD and in families with higher risk of AD, correlation with apolipoprotein E4 (APOE) allele expression has been found. How APOE4 induces its pathological effects is still unclear. Several studies indicate that autophagy, a major degradation pathway trough the lysosome, may be compromised in AD. Here we studied, the effects of APOE isoforms expression in microglia cells. By using an in-situ model, the clearance of Aβ plaques from brain sections of transgenic 5xFAD mice by the APOE expressing microglia was examined. The results show that APOE4 microglia has Impairment In clearance of insoluble Aβ plaques as compared to APOE3 and APOE2 microglia. Furthermore, APOE4 affect the uptake of soluble Aβ. We found that microglia expressing APOE4 exhibit reduced autophagic flux as compared to those expressing APOE3. The autophagy inhibitor chloroquine also blocked Aβ plaque uptake in APOE3 expressing cells. Furthermore, we found that APOE4 expressing microglia have altered mitochondrial dynamics protein expression, mitochondrial morphology and mitochondrial activity compared to those expressing APOE2, and APOE3. Rapamycin treatment corrected Mitochondrial Membrane Potential in APOE4-expressing cells. Taken together, these findings suggest that APOE4 impairs the activation of autophagy, mitophagy, and Aβ clearance and that autophagy-inducing treatments, such as rapamycin, can enhance autophagy and mitochondrial functions in APOE4 expressing microglia. Our results reveal a direct link between APOE4 to autophagy activity in microglia, suggesting that the pathological effects of APOE4 could be counteracted by pharmacological treatments inducing autophagy, such as rapamycin.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"61"},"PeriodicalIF":4.8,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143752530","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
Identification of inflammatory protein biomarkers for predicting the different subtype of adult with tuberculosis: an Olink proteomic study.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-04-01 DOI: 10.1007/s00011-025-02020-9
Yunlin Song, Buzukela Abuduaini, Xinting Yang, Jiyuan Zhang, Guirong Wang, Xiaobo Lu

Objective: This study aimed to identify the potential inflammatory molecular biomarkers that could be utilized for the early prediction of different subtypes of tuberculosis (TB) in adults.

Methods: Plasma samples were obtained from a cohort of adults diagnosed with 48 cases of active TB, including drug-susceptible TB (S-TB, n = 28), multidrug-resistant TB (R-TB, n = 20), latent TB infection (LTBI, n = 20), as well as a control group of healthy individuals without any infection (HC, n = 20). The expression level of 92 inflammatory-related proteins was detected by using the high-throughput Olink proteomics platform.

Results: There were 47 inflammatory proteins showing a significant difference (p < 0.05) among TB, LTBI, and HC groups, and 7 of them differed significantly between HC and LTBI groups, 43 proteins differed considerably between LTBI and TB groups, and overall, CXCL10 and TGF-alpha proteins differed substantially among the three groups which could be used as potential diagnostic biomarkers. Furthermore, SCF demonstrates remarkable discriminatory power in distinguishing TB from LTBI, with an area under the curve (AUC) score of 0.920. SLAMF1 has emerged as the top predictor for distinguishing Sputum Culture-Negative from positive tuberculosis cases, with an AUC of 0.779. The Correlation analyses showed various relationships among co-differentiated proteins. In LTBI versus HC, TGF-alpha and CXCL10 had a strong positive correlation. In non-severe versus severe TB, CXCL10 and CXCL9, as well as TNF and CCL3, were strongly positively correlated, while IL-6 and SCF had a negative correlation. These co-differentiated proteins were found to be enriched in various biological processes and molecular functions related to immune regulation and signaling pathways, such as the p53 signaling pathway, the TNF signaling pathway, and the NF-kappa B signaling pathway, highlighting the complex interplay of these proteins in the immune response to TB infection.

Conclusion: Inflammation-related proteins exhibited distinct expression profiles in various conditions of TB. These proteins are intercorrelated and involve the pathogenesis of tuberculosis by activating diverse immune cells and promoting the secretion of pro-inflammatory cytokines. Their functions influence cellular phenotypes, which play a crucial regulatory role in the interaction between the host and Mycobacterium tuberculosis. These findings suggest that these proteins are potential disease prevention and treatment targets.

{"title":"Identification of inflammatory protein biomarkers for predicting the different subtype of adult with tuberculosis: an Olink proteomic study.","authors":"Yunlin Song, Buzukela Abuduaini, Xinting Yang, Jiyuan Zhang, Guirong Wang, Xiaobo Lu","doi":"10.1007/s00011-025-02020-9","DOIUrl":"10.1007/s00011-025-02020-9","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to identify the potential inflammatory molecular biomarkers that could be utilized for the early prediction of different subtypes of tuberculosis (TB) in adults.</p><p><strong>Methods: </strong>Plasma samples were obtained from a cohort of adults diagnosed with 48 cases of active TB, including drug-susceptible TB (S-TB, n = 28), multidrug-resistant TB (R-TB, n = 20), latent TB infection (LTBI, n = 20), as well as a control group of healthy individuals without any infection (HC, n = 20). The expression level of 92 inflammatory-related proteins was detected by using the high-throughput Olink proteomics platform.</p><p><strong>Results: </strong>There were 47 inflammatory proteins showing a significant difference (p < 0.05) among TB, LTBI, and HC groups, and 7 of them differed significantly between HC and LTBI groups, 43 proteins differed considerably between LTBI and TB groups, and overall, CXCL10 and TGF-alpha proteins differed substantially among the three groups which could be used as potential diagnostic biomarkers. Furthermore, SCF demonstrates remarkable discriminatory power in distinguishing TB from LTBI, with an area under the curve (AUC) score of 0.920. SLAMF1 has emerged as the top predictor for distinguishing Sputum Culture-Negative from positive tuberculosis cases, with an AUC of 0.779. The Correlation analyses showed various relationships among co-differentiated proteins. In LTBI versus HC, TGF-alpha and CXCL10 had a strong positive correlation. In non-severe versus severe TB, CXCL10 and CXCL9, as well as TNF and CCL3, were strongly positively correlated, while IL-6 and SCF had a negative correlation. These co-differentiated proteins were found to be enriched in various biological processes and molecular functions related to immune regulation and signaling pathways, such as the p53 signaling pathway, the TNF signaling pathway, and the NF-kappa B signaling pathway, highlighting the complex interplay of these proteins in the immune response to TB infection.</p><p><strong>Conclusion: </strong>Inflammation-related proteins exhibited distinct expression profiles in various conditions of TB. These proteins are intercorrelated and involve the pathogenesis of tuberculosis by activating diverse immune cells and promoting the secretion of pro-inflammatory cytokines. Their functions influence cellular phenotypes, which play a crucial regulatory role in the interaction between the host and Mycobacterium tuberculosis. These findings suggest that these proteins are potential disease prevention and treatment targets.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"60"},"PeriodicalIF":4.8,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143752535","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
Unique signatures of airway and systemic immunity in severe COVID-19 patients infected with alpha to Omicron SARS-CoV-2 variants of concern.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-28 DOI: 10.1007/s00011-025-02018-3
Geovane Marques-Ferreira, Alice Aparecida Lourenço, Ana Carolina Campi-Azevedo, Felipe Alves Clarindo, André Felipe Leal Bernardes, Ludmila Oliveira Lamounier, Natalia Rocha Guimaraes, Talita Emile Ribeiro Adelino, Felipe Campos de Melo Iani, Daniel Assis Santos, Vanessa Caroline Randi Magalhães, Camila Pacheco Silveira Martins da Mata, Erik Vinicius de Sousa Reis, Thaís de Fátima Silva Moraes, Letícia Gomes-de-Pontes, Flávio Guimarães da Fonseca, Andréa Teixeira-Carvalho, Mayra Gonçalves Menegueti, Maria Auxiliadora-Martins, Paulo Henrique Ribeiro Amaral, Juan Carlos González Pérez, Olindo Assis Martins-Filho, Jordana Grazziela Coelho-Dos-Reis

In this study, systemic and localized immunity induced by SARS-CoV-2 variants of concern or interest (VOC/VOI) was investigated. For that, serum and tracheal aspirate soluble chemokines, pro-inflammatory/regulatory cytokines, and growth factors were measured in severe COVID-19 patients under mechanical ventilation upon infection with different SARS-CoV-2 variants, namely Alpha, Gamma, Zeta, Delta and Omicron. Increased levels of soluble mediators were observed in serum from severe COVID-19 patients regardless of the variant. In tracheal aspirate samples, the patients infected with the Gamma, Zeta, Delta, and Omicron variants exhibited reduced levels of inflammatory cytokines when compared to those infected with the Alpha variant. The trend of lower cytokine levels was also observed in the serum of patients across these variants, except for the Delta variant. By using network analysis and cytokine storm signatures, the data confirmed that severe COVID-19 induced by different variants have a completely divergent pattern of connectivity in serum samples as well as tracheal aspirates. Patients infected with variants at later time points in the pandemic such as Omicron exhibited networks of weak central architecture in serum samples as compared to tracheal aspirates, with lower number of neighborhood connections and clusters of pro-inflammatory and regulatory cytokines. By and large, this study points out to important systemic and local divergences and to loss of airway localized immunity in severe COVID-19 patients infected with SARS-CoV-2 variants, which brings insight into understanding host responses and viral escape vis-à-vis the virus mutations and evolution.

本研究调查了 SARS-CoV-2 变异体(VOC/VOI)诱导的全身和局部免疫。为此,研究人员测量了机械通气下的严重 COVID-19 患者在感染不同 SARS-CoV-2 变体(即 Alpha、Gamma、Zeta、Delta 和 Omicron)后的血清和气管吸出物中的可溶性趋化因子、促炎/调节细胞因子和生长因子。在重症 COVID-19 患者的血清中观察到可溶性介质水平升高,与变异体无关。在气管吸出物样本中,感染了 Gamma、Zeta、Delta 和 Omicron 变种的患者与感染了 Alpha 变种的患者相比,炎症细胞因子水平有所降低。除Delta变异体外,其他变异体患者血清中的细胞因子水平也呈下降趋势。通过使用网络分析和细胞因子风暴特征,数据证实不同变异体诱发的严重COVID-19在血清样本和气管吸出物中具有完全不同的连接模式。与气管抽吸物相比,感染大流行后期变异体(如 Omicron)的患者在血清样本中表现出弱中心结构的网络,邻域连接和促炎与调节细胞因子集群的数量较少。总的来说,这项研究指出了感染 SARS-CoV-2 变异株的严重 COVID-19 患者在全身和局部方面的重要差异,以及气道局部免疫力的丧失,这有助于了解病毒变异和进化过程中宿主的反应和病毒逃逸。
{"title":"Unique signatures of airway and systemic immunity in severe COVID-19 patients infected with alpha to Omicron SARS-CoV-2 variants of concern.","authors":"Geovane Marques-Ferreira, Alice Aparecida Lourenço, Ana Carolina Campi-Azevedo, Felipe Alves Clarindo, André Felipe Leal Bernardes, Ludmila Oliveira Lamounier, Natalia Rocha Guimaraes, Talita Emile Ribeiro Adelino, Felipe Campos de Melo Iani, Daniel Assis Santos, Vanessa Caroline Randi Magalhães, Camila Pacheco Silveira Martins da Mata, Erik Vinicius de Sousa Reis, Thaís de Fátima Silva Moraes, Letícia Gomes-de-Pontes, Flávio Guimarães da Fonseca, Andréa Teixeira-Carvalho, Mayra Gonçalves Menegueti, Maria Auxiliadora-Martins, Paulo Henrique Ribeiro Amaral, Juan Carlos González Pérez, Olindo Assis Martins-Filho, Jordana Grazziela Coelho-Dos-Reis","doi":"10.1007/s00011-025-02018-3","DOIUrl":"https://doi.org/10.1007/s00011-025-02018-3","url":null,"abstract":"<p><p>In this study, systemic and localized immunity induced by SARS-CoV-2 variants of concern or interest (VOC/VOI) was investigated. For that, serum and tracheal aspirate soluble chemokines, pro-inflammatory/regulatory cytokines, and growth factors were measured in severe COVID-19 patients under mechanical ventilation upon infection with different SARS-CoV-2 variants, namely Alpha, Gamma, Zeta, Delta and Omicron. Increased levels of soluble mediators were observed in serum from severe COVID-19 patients regardless of the variant. In tracheal aspirate samples, the patients infected with the Gamma, Zeta, Delta, and Omicron variants exhibited reduced levels of inflammatory cytokines when compared to those infected with the Alpha variant. The trend of lower cytokine levels was also observed in the serum of patients across these variants, except for the Delta variant. By using network analysis and cytokine storm signatures, the data confirmed that severe COVID-19 induced by different variants have a completely divergent pattern of connectivity in serum samples as well as tracheal aspirates. Patients infected with variants at later time points in the pandemic such as Omicron exhibited networks of weak central architecture in serum samples as compared to tracheal aspirates, with lower number of neighborhood connections and clusters of pro-inflammatory and regulatory cytokines. By and large, this study points out to important systemic and local divergences and to loss of airway localized immunity in severe COVID-19 patients infected with SARS-CoV-2 variants, which brings insight into understanding host responses and viral escape vis-à-vis the virus mutations and evolution.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"57"},"PeriodicalIF":4.8,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143735799","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
Inflammatory pain and electroacupuncture: how the P2X3 receptor can help modulate inflammation-a review of current literature.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-28 DOI: 10.1007/s00011-025-02023-6
Jardel Cristiano Ecco, Adinei Abadio Soares, Keroli Eloiza Tessaro da Silva, Vinicius Ansolin, Guilherme Vinício Sousa Silva, Débora Tavares Resende E Silva

Aim: Inflammatory pain arises from tissue stress or injury and is initiated by signaling molecules that stimulate the immune and nervous systems. Evidence suggests that purinergic signaling pathways can modulate pain and inflammation through the activation of P1 and P2 purinergic receptors, such as the P2X3 receptor, which are stimulated by extracellular molecules like adenosine triphosphate (ATP). Electroacupuncture (EA) exhibits precise mechanisms that modulate inflammatory pain through the activation of the P2X3 receptor.

Objective: This review analyzed evidence regarding the role of electroacupuncture and the purinergic system, particularly the P2X3 receptor, in modulating inflammation and pain.

Materials and methods: A search for the most relevant articles available in the SciVerse Scopus and MEDLINE/PubMed databases was conducted for publications from 1995 to 2024. Articles were initially selected by reading the title, abstract, and main text, respectively.

Results: It was found that the P2X3 receptor, as well as the molecules activating purinergic receptors, such as ATP and adenosine, have the potential to regulate pain and inflammation. Additionally, EA can modulate the purinergic system in an anti-inflammatory response. EA may stimulate analgesia mainly through the conversion of ATP to adenosine, a crucial molecule in pain control.

Conclusion: The purinergic system directly influences inflammatory pain and controls inflammation. In this context, EA has the potential to orchestrate this system to control pain and inflammation.

目的:炎性疼痛源于组织应激或损伤,由刺激免疫和神经系统的信号分子引发。有证据表明,嘌呤能信号通路可通过激活 P1 和 P2 嘌呤能受体(如 P2X3 受体)来调节疼痛和炎症。电针(EA)显示出通过激活 P2X3 受体调节炎性疼痛的精确机制:本综述分析了有关电针和嘌呤能系统(尤其是 P2X3 受体)在调节炎症和疼痛中作用的证据:对 SciVerse Scopus 和 MEDLINE/PubMed 数据库中 1995 年至 2024 年发表的最相关文章进行了检索。分别通过阅读标题、摘要和正文初步筛选出文章:结果:研究发现,P2X3 受体以及激活嘌呤能受体的分子(如 ATP 和腺苷)具有调节疼痛和炎症的潜力。此外,EA 还能在抗炎反应中调节嘌呤能系统。EA 可能主要通过将 ATP 转化为腺苷(一种控制疼痛的重要分子)来刺激镇痛:结论:嘌呤能系统直接影响炎性疼痛并控制炎症。结论:嘌呤能系统直接影响炎症性疼痛并控制炎症。在这种情况下,EA 具有协调该系统控制疼痛和炎症的潜力。
{"title":"Inflammatory pain and electroacupuncture: how the P2X3 receptor can help modulate inflammation-a review of current literature.","authors":"Jardel Cristiano Ecco, Adinei Abadio Soares, Keroli Eloiza Tessaro da Silva, Vinicius Ansolin, Guilherme Vinício Sousa Silva, Débora Tavares Resende E Silva","doi":"10.1007/s00011-025-02023-6","DOIUrl":"https://doi.org/10.1007/s00011-025-02023-6","url":null,"abstract":"<p><strong>Aim: </strong>Inflammatory pain arises from tissue stress or injury and is initiated by signaling molecules that stimulate the immune and nervous systems. Evidence suggests that purinergic signaling pathways can modulate pain and inflammation through the activation of P1 and P2 purinergic receptors, such as the P2X3 receptor, which are stimulated by extracellular molecules like adenosine triphosphate (ATP). Electroacupuncture (EA) exhibits precise mechanisms that modulate inflammatory pain through the activation of the P2X3 receptor.</p><p><strong>Objective: </strong>This review analyzed evidence regarding the role of electroacupuncture and the purinergic system, particularly the P2X3 receptor, in modulating inflammation and pain.</p><p><strong>Materials and methods: </strong>A search for the most relevant articles available in the SciVerse Scopus and MEDLINE/PubMed databases was conducted for publications from 1995 to 2024. Articles were initially selected by reading the title, abstract, and main text, respectively.</p><p><strong>Results: </strong>It was found that the P2X3 receptor, as well as the molecules activating purinergic receptors, such as ATP and adenosine, have the potential to regulate pain and inflammation. Additionally, EA can modulate the purinergic system in an anti-inflammatory response. EA may stimulate analgesia mainly through the conversion of ATP to adenosine, a crucial molecule in pain control.</p><p><strong>Conclusion: </strong>The purinergic system directly influences inflammatory pain and controls inflammation. In this context, EA has the potential to orchestrate this system to control pain and inflammation.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"58"},"PeriodicalIF":4.8,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143735797","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
Luteolin modulates liver macrophage subtype polarization and play protective role in sepsis induced acute hepatic injury.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-28 DOI: 10.1007/s00011-025-02026-3
Liangyong Deng, Qiulei Yu, Gang Kuang, Liuyang Wang, Jing Fan, Lin Ye

Background: Luteolin has an anti-inflammatory effect, but the mechanism has not been elucidated in sepsis-induced acute hepatic injury (AHI). The purpose of this study was to investigate the effects and potential mechanisms of luteolin on sepsis-induced AHI.

Methods: In this study, we utilized both wild-type (WT) mice and Toll-like receptor 4 (TLR4)-deficient (TLR4-/-) mice alongside RAW264.7 cells. We constructed a CLP-induced AHI mouse model to study the effects of luteolin on liver inflammation, survival and liver macrophage subtypes in mice. In addition, we extracted mouse serum, mouse bone marrow-derived macrophages (BMDMs) and liver tissue and analysed the effects of luteolin on macrophage polarization subtypes and downstream inflammatory cytokines by flow cytometry, ELISA, Western blotting (WB) and qPCR. To further verify the effect of luteolin on macrophage polarization and explore the possible potential mechanism, we used a CLP-induced AHI mouse model and LPS-stimulated RAW 264.7 macrophages to assess the effect of luteolin on macrophage polarization; the expression of TNF-α and IL-10 in the cell culture supernatant; and the expression of iNOS, ARG-1, NF-κB (P65), p-P65 and MyD88 by flow cytometry, ELISA, immunohistochemistry and Western blotting.

Results: We found that luteolin reduced liver injury and inflammatory response and improved the survival rate of mice. Luteolin modulated the macrophage subtype proportion, promoted the change of macrophages from a proinflammatory M1 phenotype to an anti-inflammatory M2 phenotype, and reduced the inflammatory response both in vivo and in vitro. Moreover, luteolin reduced the expression of NF-κB (p-P65), TLR4 and MyD88. By integrating the predictions from network pharmacology with the in vitro and in vivo experimental results, it was determined that the mechanism by which luteolin alleviates sepsis-induced acute hepatic injury is closely related to the TLR4/MyD88/NF-κB pathway.

Conclusions: The results of this study suggest that luteolin helps alleviate liver injury, reduces the expression of proinflammatory cytokines and promotes the expression of anti-inflammatory factors in sepsis-induced acute hepatic injury. This effect may be related to the regulation of macrophage polarization by luteolin through the TLR4/MyD88/NF-κB signalling pathway.

背景:叶黄素具有抗炎作用,但其在脓毒症诱导的急性肝损伤(AHI)中的作用机制尚未阐明。本研究旨在探讨木犀草素对败血症诱导的急性肝损伤的影响和潜在机制:在本研究中,我们利用野生型(WT)小鼠和Toll样受体4(TLR4)缺陷型(TLR4-/-)小鼠以及RAW264.7细胞。我们构建了一个CLP诱导的AHI小鼠模型,以研究木犀草素对小鼠肝脏炎症、存活率和肝脏巨噬细胞亚型的影响。此外,我们还提取了小鼠血清、小鼠骨髓巨噬细胞(BMDMs)和肝组织,并通过流式细胞术、ELISA、Western 印迹(WB)和 qPCR 分析了叶黄素对巨噬细胞极化亚型和下游炎症细胞因子的影响。为了进一步验证木犀草素对巨噬细胞极化的影响并探索其可能的潜在机制,我们使用CLP诱导的AHI小鼠模型和LPS刺激的RAW 264.7巨噬细胞,通过流式细胞术、ELISA、免疫组织化学和Western印迹等方法评估了叶黄素对巨噬细胞极化的影响;细胞培养上清液中TNF-α和IL-10的表达;以及iNOS、ARG-1、NF-κB (P65)、p-P65和MyD88的表达:结果:我们发现木犀草素能减轻肝损伤和炎症反应,提高小鼠的存活率。叶黄素能调节巨噬细胞亚型比例,促进巨噬细胞从促炎的M1表型转变为抗炎的M2表型,减轻体内和体外的炎症反应。此外,木犀草素还能降低 NF-κB (p-P65)、TLR4 和 MyD88 的表达。通过将网络药理学的预测与体内外实验结果相结合,确定了叶黄素缓解败血症诱导的急性肝损伤的机制与 TLR4/MyD88/NF-κB通路密切相关:本研究结果表明,在脓毒症诱导的急性肝损伤中,叶黄素有助于缓解肝损伤,减少促炎细胞因子的表达,促进抗炎因子的表达。这种作用可能与叶黄素通过TLR4/MyD88/NF-κB信号通路调节巨噬细胞极化有关。
{"title":"Luteolin modulates liver macrophage subtype polarization and play protective role in sepsis induced acute hepatic injury.","authors":"Liangyong Deng, Qiulei Yu, Gang Kuang, Liuyang Wang, Jing Fan, Lin Ye","doi":"10.1007/s00011-025-02026-3","DOIUrl":"https://doi.org/10.1007/s00011-025-02026-3","url":null,"abstract":"<p><strong>Background: </strong>Luteolin has an anti-inflammatory effect, but the mechanism has not been elucidated in sepsis-induced acute hepatic injury (AHI). The purpose of this study was to investigate the effects and potential mechanisms of luteolin on sepsis-induced AHI.</p><p><strong>Methods: </strong>In this study, we utilized both wild-type (WT) mice and Toll-like receptor 4 (TLR4)-deficient (TLR4<sup>-/-</sup>) mice alongside RAW264.7 cells. We constructed a CLP-induced AHI mouse model to study the effects of luteolin on liver inflammation, survival and liver macrophage subtypes in mice. In addition, we extracted mouse serum, mouse bone marrow-derived macrophages (BMDMs) and liver tissue and analysed the effects of luteolin on macrophage polarization subtypes and downstream inflammatory cytokines by flow cytometry, ELISA, Western blotting (WB) and qPCR. To further verify the effect of luteolin on macrophage polarization and explore the possible potential mechanism, we used a CLP-induced AHI mouse model and LPS-stimulated RAW 264.7 macrophages to assess the effect of luteolin on macrophage polarization; the expression of TNF-α and IL-10 in the cell culture supernatant; and the expression of iNOS, ARG-1, NF-κB (P65), p-P65 and MyD88 by flow cytometry, ELISA, immunohistochemistry and Western blotting.</p><p><strong>Results: </strong>We found that luteolin reduced liver injury and inflammatory response and improved the survival rate of mice. Luteolin modulated the macrophage subtype proportion, promoted the change of macrophages from a proinflammatory M1 phenotype to an anti-inflammatory M2 phenotype, and reduced the inflammatory response both in vivo and in vitro. Moreover, luteolin reduced the expression of NF-κB (p-P65), TLR4 and MyD88. By integrating the predictions from network pharmacology with the in vitro and in vivo experimental results, it was determined that the mechanism by which luteolin alleviates sepsis-induced acute hepatic injury is closely related to the TLR4/MyD88/NF-κB pathway.</p><p><strong>Conclusions: </strong>The results of this study suggest that luteolin helps alleviate liver injury, reduces the expression of proinflammatory cytokines and promotes the expression of anti-inflammatory factors in sepsis-induced acute hepatic injury. This effect may be related to the regulation of macrophage polarization by luteolin through the TLR4/MyD88/NF-κB signalling pathway.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"59"},"PeriodicalIF":4.8,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143735798","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
Roquin-1 interaction with Regnase-1 inhibits the progression of rheumatoid arthritis via suppressing FGF2 expression and NF-κB pathway. Roquin-1 与 Regnase-1 相互作用,通过抑制 FGF2 的表达和 NF-κB 通路抑制类风湿性关节炎的进展。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-18 DOI: 10.1007/s00011-025-02012-9
Hui Wang, Zizheng Tang, Kangqi Xie, Tiantian Hao, Gang Su

Objective: This study aimed to explore the effect of Roquin-1 on rheumatoid arthritis (RA) and its potential mechanisms.

Methods: Firstly, we used TNF-α to stimulate fibroblast-like synoviocytes (FLSs) to establish an in vitro model of RA. Moreover, a rat model of RA was established with bovine type II collagen and complete Freund's adjuvant. EdU and transwell assays were applied for evaluating the proliferation and migration of FLSs. The multiple mRNA and proteins expressions in FLSs and rats synovial tissues were measured using qRT-PCR, ELISA, western blot, immunohistochemistry staining and immunofluorescence staining. Double immunofluorescence staining and co-IP assay were used to validate the protein interaction between Roquin-1 and Regnase-1. Additionally, cycloheximide (CHX) chase assay was applied for assessing the degradation of fibroblast growth factor 2 (FGF2). Besides, the state of synovial hyperplasia and articular cartilage were also evaluated using HE and Safranin O/Fast Green staining.

Results: The mRNA and protein expressions of Roquin-1 were significantly reduced in TNF-α-stimulated FLSs and the synovial tissues of RA rats. Roquin-1 interacted with Regnase-1 to promote FGF2 degradation and further inhibit the proliferation, migration and inflammation response in TNF-α-stimulated FLSs. Moreover, we also demonstrated that Roquin-1 interacted with Regnase-1 to inhibit NF-κB pathway via suppressing FGF2 expression in TNF-α-stimulated FLSs. In addition, Roquin-1 suppressed inflammatory response in RA rats.

Conclusion: Our findings demonstrated that Roquin-1 could interact with Regnase-1 to inhibit the progression of RA via suppressing FGF2 expression and NF-κB pathway.

{"title":"Roquin-1 interaction with Regnase-1 inhibits the progression of rheumatoid arthritis via suppressing FGF2 expression and NF-κB pathway.","authors":"Hui Wang, Zizheng Tang, Kangqi Xie, Tiantian Hao, Gang Su","doi":"10.1007/s00011-025-02012-9","DOIUrl":"https://doi.org/10.1007/s00011-025-02012-9","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to explore the effect of Roquin-1 on rheumatoid arthritis (RA) and its potential mechanisms.</p><p><strong>Methods: </strong>Firstly, we used TNF-α to stimulate fibroblast-like synoviocytes (FLSs) to establish an in vitro model of RA. Moreover, a rat model of RA was established with bovine type II collagen and complete Freund's adjuvant. EdU and transwell assays were applied for evaluating the proliferation and migration of FLSs. The multiple mRNA and proteins expressions in FLSs and rats synovial tissues were measured using qRT-PCR, ELISA, western blot, immunohistochemistry staining and immunofluorescence staining. Double immunofluorescence staining and co-IP assay were used to validate the protein interaction between Roquin-1 and Regnase-1. Additionally, cycloheximide (CHX) chase assay was applied for assessing the degradation of fibroblast growth factor 2 (FGF2). Besides, the state of synovial hyperplasia and articular cartilage were also evaluated using HE and Safranin O/Fast Green staining.</p><p><strong>Results: </strong>The mRNA and protein expressions of Roquin-1 were significantly reduced in TNF-α-stimulated FLSs and the synovial tissues of RA rats. Roquin-1 interacted with Regnase-1 to promote FGF2 degradation and further inhibit the proliferation, migration and inflammation response in TNF-α-stimulated FLSs. Moreover, we also demonstrated that Roquin-1 interacted with Regnase-1 to inhibit NF-κB pathway via suppressing FGF2 expression in TNF-α-stimulated FLSs. In addition, Roquin-1 suppressed inflammatory response in RA rats.</p><p><strong>Conclusion: </strong>Our findings demonstrated that Roquin-1 could interact with Regnase-1 to inhibit the progression of RA via suppressing FGF2 expression and NF-κB pathway.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"55"},"PeriodicalIF":4.8,"publicationDate":"2025-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143648476","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
Mediation of macrophage M1 polarization dynamics change by ubiquitin-autophagy-pathway regulated NLRP3 inflammasomes in PD-1 inhibitor-related myocardial inflammatory injury.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-18 DOI: 10.1007/s00011-024-01979-1
Yanxin Chen, Yuxi Luo, Yunwei Liu, Xingpeng Qiu, Daya Luo, Anwen Liu

Objective and design: Immune checkpoint inhibitors (ICIs)-induced cardiac injury is a life-threatening immune-related adverse events (irAEs). However, the current understanding of its pathogenesis and therapeutic options is relatively limited. We aimed to provide a comprehensive overview of the myocardial inflammatory injury process and underlying mechanisms associated with ICIs.

Material or subjects: We conducted a descriptive analysis of lung cancer patients with ICIs-induced myocarditis at our institution and validated our clinical findings using single-cell sequencing (scRNA-seq) data. Furthermore, we established animal and cellular models to investigate the underlying mechanisms.

Results: Our findings revealed that lung cancer patients with ICIs-induced myocarditis exhibit an early increase in peripheral blood monocytes, which migrate to the heart and differentiate into macrophages, ultimately leading to myocardial inflammation. Mechanistically, programmed death-1 (PD-1) inhibition triggers myocardial inflammation through the activation of the signal transducer and activator of transcription 1 (STAT1)/nuclear factor kappa-B (NF-κB)/oligomerization domain (NOD)-like receptor thermal protein domain-associated protein 3 (NLRP3) signaling pathway and drives the polarization of macrophages towards the M1 phenotype. However, the ubiquitin (Ub)-autophagy pathway degrades NLRP3 inflammasomes, resulting in the gradual resolution of inflammation and the transition of M1 macrophages to the M2 phenotype. Finally, mouse experiments confirmed that NLRP3 inhibition using MCC950 effectively alleviates myocardial inflammatory injury.

Conclusions: We recommend monitoring fluctuations in peripheral blood monocyte counts in lung cancer patients undergoing ICIs treatment. Additionally, MCC950 holds potential as a therapeutic agent for ICIs-induced cardiac inflammation injury.

{"title":"Mediation of macrophage M1 polarization dynamics change by ubiquitin-autophagy-pathway regulated NLRP3 inflammasomes in PD-1 inhibitor-related myocardial inflammatory injury.","authors":"Yanxin Chen, Yuxi Luo, Yunwei Liu, Xingpeng Qiu, Daya Luo, Anwen Liu","doi":"10.1007/s00011-024-01979-1","DOIUrl":"https://doi.org/10.1007/s00011-024-01979-1","url":null,"abstract":"<p><strong>Objective and design: </strong>Immune checkpoint inhibitors (ICIs)-induced cardiac injury is a life-threatening immune-related adverse events (irAEs). However, the current understanding of its pathogenesis and therapeutic options is relatively limited. We aimed to provide a comprehensive overview of the myocardial inflammatory injury process and underlying mechanisms associated with ICIs.</p><p><strong>Material or subjects: </strong>We conducted a descriptive analysis of lung cancer patients with ICIs-induced myocarditis at our institution and validated our clinical findings using single-cell sequencing (scRNA-seq) data. Furthermore, we established animal and cellular models to investigate the underlying mechanisms.</p><p><strong>Results: </strong>Our findings revealed that lung cancer patients with ICIs-induced myocarditis exhibit an early increase in peripheral blood monocytes, which migrate to the heart and differentiate into macrophages, ultimately leading to myocardial inflammation. Mechanistically, programmed death-1 (PD-1) inhibition triggers myocardial inflammation through the activation of the signal transducer and activator of transcription 1 (STAT1)/nuclear factor kappa-B (NF-κB)/oligomerization domain (NOD)-like receptor thermal protein domain-associated protein 3 (NLRP3) signaling pathway and drives the polarization of macrophages towards the M1 phenotype. However, the ubiquitin (Ub)-autophagy pathway degrades NLRP3 inflammasomes, resulting in the gradual resolution of inflammation and the transition of M1 macrophages to the M2 phenotype. Finally, mouse experiments confirmed that NLRP3 inhibition using MCC950 effectively alleviates myocardial inflammatory injury.</p><p><strong>Conclusions: </strong>We recommend monitoring fluctuations in peripheral blood monocyte counts in lung cancer patients undergoing ICIs treatment. Additionally, MCC950 holds potential as a therapeutic agent for ICIs-induced cardiac inflammation injury.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"56"},"PeriodicalIF":4.8,"publicationDate":"2025-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143648440","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 RNA-binding protein KSRP reduces asthma-like characteristics in a murine model.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-17 DOI: 10.1007/s00011-025-02024-5
Kim-Alicia Palzer, Vanessa Bolduan, Jelena Lakus, Ingrid Tubbe, Evelyn Montermann, Björn E Clausen, Matthias Bros, Andrea Pautz

Background and objective: Asthma is a chronic inflammatory disease characterized by dysregulated cytokine expression. The RNA-binding protein KSRP reduces the expression of several pro-inflammatory mediators. Therefore, we investigated whether KSRP modulates Th2-associated immune responses in vivo in an ovalbumin-induced (OVA) allergic asthma model in C57BL/6 KSRP-deficient mice (KSRP-/-).

Methods: Asthma severity in OVA-immunized wild type or KSRP-/- mice was determined by airway hyperresponsiveness (AHR), structural changes of lung tissue, and OVA-specific antibody production. Cytokine expression in bronchoalveolar lavage fluid (BALF) was measured by Cytometric Bead Array (CBA) analysis. Cellular signaling pathways involved in KSRP-mediated effects in asthma pathogenesis were analyzed in vitro in cell culture models using specific inhibitors.

Results: KSRP deficiency exacerbates OVA-induced allergic asthma compared to wild type mice, as indicated by increased AHR, more severe lung damage, goblet cell hyperplasia and increased OVA-specific antibody production. CBA analyses confirmed, that KSRP deficiency enhances IL-4, IL-5 and IL-13 production in BALF. The effect of KSRP on Th2-associated cytokine expression appears to be mediated by modulation of the STAT6 and NFAT signaling pathway rather than by inhibiting the stability of cytokine-encoding mRNA species.

Conclusion: Our data demonstrate that KSRP dampens Th2 immune cell activity and therefore seems to be important for the pathogenesis of Th2-mediated diseases.

{"title":"The RNA-binding protein KSRP reduces asthma-like characteristics in a murine model.","authors":"Kim-Alicia Palzer, Vanessa Bolduan, Jelena Lakus, Ingrid Tubbe, Evelyn Montermann, Björn E Clausen, Matthias Bros, Andrea Pautz","doi":"10.1007/s00011-025-02024-5","DOIUrl":"10.1007/s00011-025-02024-5","url":null,"abstract":"<p><strong>Background and objective: </strong>Asthma is a chronic inflammatory disease characterized by dysregulated cytokine expression. The RNA-binding protein KSRP reduces the expression of several pro-inflammatory mediators. Therefore, we investigated whether KSRP modulates Th2-associated immune responses in vivo in an ovalbumin-induced (OVA) allergic asthma model in C57BL/6 KSRP-deficient mice (KSRP<sup>-/-</sup>).</p><p><strong>Methods: </strong>Asthma severity in OVA-immunized wild type or KSRP<sup>-/-</sup> mice was determined by airway hyperresponsiveness (AHR), structural changes of lung tissue, and OVA-specific antibody production. Cytokine expression in bronchoalveolar lavage fluid (BALF) was measured by Cytometric Bead Array (CBA) analysis. Cellular signaling pathways involved in KSRP-mediated effects in asthma pathogenesis were analyzed in vitro in cell culture models using specific inhibitors.</p><p><strong>Results: </strong>KSRP deficiency exacerbates OVA-induced allergic asthma compared to wild type mice, as indicated by increased AHR, more severe lung damage, goblet cell hyperplasia and increased OVA-specific antibody production. CBA analyses confirmed, that KSRP deficiency enhances IL-4, IL-5 and IL-13 production in BALF. The effect of KSRP on Th2-associated cytokine expression appears to be mediated by modulation of the STAT6 and NFAT signaling pathway rather than by inhibiting the stability of cytokine-encoding mRNA species.</p><p><strong>Conclusion: </strong>Our data demonstrate that KSRP dampens Th2 immune cell activity and therefore seems to be important for the pathogenesis of Th2-mediated diseases.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"54"},"PeriodicalIF":4.8,"publicationDate":"2025-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11914311/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143647424","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
Vitamin D3 suppresses NLRP3 inflammasome pathway and enhances steroid sensitivity in a neutrophilic steroid hyporesponsive asthma mouse model.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-14 DOI: 10.1007/s00011-025-02009-4
Bariaa Khalil, Narjes Saheb Sharif-Askari, Balachandar Selvakumar, Bushra Mdkhana, Ibrahim Hachim, Adel Zakri, Jennifer Hundt, Qutayba Hamid, Rabih Halwani

Objective: Severe steroid hyporesponsive asthma is a heterogeneous group of chronic inflammatory diseases characterized by irreversible airflow limitation, hyperresponsiveness, inflammation, and remodelling of the airways. Severe asthmatics account for more than 60% of asthma-related healthcare cost worldwide given they are hyporesponsive to corticosteroids and due to the absence of targeted treatment specifically for the T helper-17 (Th-17) high endotype. Hence, there is a clear unmet need to investigate other treatment options to control patients' symptoms. The role of the NLRP3 inflammasome pathway has been highlighted in the literature to contribute to disease pathogenesis and severity. Interestingly, vitamin D3 is an important regulator of the NLRP3 inflammasome pathway.

Methods: Using house dust mite (HDM) and lipopolysaccharide (LPS), we induced a neutrophilic steroid hyporesponsive asthma mouse model to investigate the effect of vitamin D3 on downregulating the NLRP3 inflammasome pathway and enhancing steroid sensitivity.

Results: We showed that calcitriol, the active form of vitamin D3, could downregulate the NLRP3 inflammasome pathway. This was associated with a significant reduction in airway hyperresponsiveness, IL-17 release, neutrophil infiltration, and mucus secretion. Further, calcitriol enhanced steroid sensitivity by inhibiting the expression of GR-β. Mechanistically, calcitriol targeted the NLRP3 inflammasome to ubiquitination.

Conclusions: Our research highlights the potential use of calcitriol as a low cost and accessible supplement to ameliorate airway inflammation during severe steroid hyporesponsive asthma.

{"title":"Vitamin D3 suppresses NLRP3 inflammasome pathway and enhances steroid sensitivity in a neutrophilic steroid hyporesponsive asthma mouse model.","authors":"Bariaa Khalil, Narjes Saheb Sharif-Askari, Balachandar Selvakumar, Bushra Mdkhana, Ibrahim Hachim, Adel Zakri, Jennifer Hundt, Qutayba Hamid, Rabih Halwani","doi":"10.1007/s00011-025-02009-4","DOIUrl":"https://doi.org/10.1007/s00011-025-02009-4","url":null,"abstract":"<p><strong>Objective: </strong>Severe steroid hyporesponsive asthma is a heterogeneous group of chronic inflammatory diseases characterized by irreversible airflow limitation, hyperresponsiveness, inflammation, and remodelling of the airways. Severe asthmatics account for more than 60% of asthma-related healthcare cost worldwide given they are hyporesponsive to corticosteroids and due to the absence of targeted treatment specifically for the T helper-17 (Th-17) high endotype. Hence, there is a clear unmet need to investigate other treatment options to control patients' symptoms. The role of the NLRP3 inflammasome pathway has been highlighted in the literature to contribute to disease pathogenesis and severity. Interestingly, vitamin D3 is an important regulator of the NLRP3 inflammasome pathway.</p><p><strong>Methods: </strong>Using house dust mite (HDM) and lipopolysaccharide (LPS), we induced a neutrophilic steroid hyporesponsive asthma mouse model to investigate the effect of vitamin D3 on downregulating the NLRP3 inflammasome pathway and enhancing steroid sensitivity.</p><p><strong>Results: </strong>We showed that calcitriol, the active form of vitamin D3, could downregulate the NLRP3 inflammasome pathway. This was associated with a significant reduction in airway hyperresponsiveness, IL-17 release, neutrophil infiltration, and mucus secretion. Further, calcitriol enhanced steroid sensitivity by inhibiting the expression of GR-β. Mechanistically, calcitriol targeted the NLRP3 inflammasome to ubiquitination.</p><p><strong>Conclusions: </strong>Our research highlights the potential use of calcitriol as a low cost and accessible supplement to ameliorate airway inflammation during severe steroid hyporesponsive asthma.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"51"},"PeriodicalIF":4.8,"publicationDate":"2025-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143624383","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
Dog allergen-induced asthma in mice: a relevant model of T2low severe asthma with airway remodelling.
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-14 DOI: 10.1007/s00011-025-02004-9
Victor Margelidon-Cozzolino, Joanne Balsamelli, Julie Carrard, Saliha Ait Yahia, Marie-Hélène Gevaert, Silvia Demoulin-Alexikova, Muriel Pichavant, Anne Tsicopoulos, Cécile Chenivesse, Stéphanie Lejeune, Patricia de Nadai

Objective and design: Airway remodelling (AR) is a disabling phenomenon in patients with severe asthma, yet suitable models are lacking. We previously developed a dog allergen-induced murine asthma model characterized by T2low Th17-driven neutrophilic airway inflammation and AR. To assess its relevance to human AR associated with T2low severe asthma, a condition characterised by poor response to inhaled steroids, we tested the steroid sensitivity of the key features of this model.

Material: Asthma was induced in C57BL/6 J mice by intranasal sensitization, followed by a three-week challenge with dog allergen.

Treatment: Daily intraperitoneal 1 mg kg-1 dexamethasone was administrated during the last week of challenge.

Methods: We measured airway resistances in response to methacholine, cellular inflammation in bronchoalveolar lavage, lung cytokines, and quantified AR features, in response to dexamethasone.

Results: Dexamethasone-treated mice showed persistent airway hyperresponsiveness, neutrophilic inflammation, and Il17a overexpression, whereas Il22 expression was abrogated. Pathological AR features, including mucus hyperproduction, subepithelial fibrosis and smooth muscle hypertrophy were not eliminated by dexamethasone.

Conclusions: Our dog allergen-induced murine model of asthma mirrors the steroid-insensitive traits of human severe T2low asthma with AR, making it a relevant tool for identifying novel therapeutic targets in this orphan asthma subset.

{"title":"Dog allergen-induced asthma in mice: a relevant model of T2<sup>low</sup> severe asthma with airway remodelling.","authors":"Victor Margelidon-Cozzolino, Joanne Balsamelli, Julie Carrard, Saliha Ait Yahia, Marie-Hélène Gevaert, Silvia Demoulin-Alexikova, Muriel Pichavant, Anne Tsicopoulos, Cécile Chenivesse, Stéphanie Lejeune, Patricia de Nadai","doi":"10.1007/s00011-025-02004-9","DOIUrl":"10.1007/s00011-025-02004-9","url":null,"abstract":"<p><strong>Objective and design: </strong>Airway remodelling (AR) is a disabling phenomenon in patients with severe asthma, yet suitable models are lacking. We previously developed a dog allergen-induced murine asthma model characterized by T2<sup>low</sup> Th17-driven neutrophilic airway inflammation and AR. To assess its relevance to human AR associated with T2<sup>low</sup> severe asthma, a condition characterised by poor response to inhaled steroids, we tested the steroid sensitivity of the key features of this model.</p><p><strong>Material: </strong>Asthma was induced in C57BL/6 J mice by intranasal sensitization, followed by a three-week challenge with dog allergen.</p><p><strong>Treatment: </strong>Daily intraperitoneal 1 mg kg<sup>-1</sup> dexamethasone was administrated during the last week of challenge.</p><p><strong>Methods: </strong>We measured airway resistances in response to methacholine, cellular inflammation in bronchoalveolar lavage, lung cytokines, and quantified AR features, in response to dexamethasone.</p><p><strong>Results: </strong>Dexamethasone-treated mice showed persistent airway hyperresponsiveness, neutrophilic inflammation, and Il17a overexpression, whereas Il22 expression was abrogated. Pathological AR features, including mucus hyperproduction, subepithelial fibrosis and smooth muscle hypertrophy were not eliminated by dexamethasone.</p><p><strong>Conclusions: </strong>Our dog allergen-induced murine model of asthma mirrors the steroid-insensitive traits of human severe T2<sup>low</sup> asthma with AR, making it a relevant tool for identifying novel therapeutic targets in this orphan asthma subset.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"52"},"PeriodicalIF":4.8,"publicationDate":"2025-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11906515/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143624372","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
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:481959085
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