首页 > 最新文献

International immunopharmacology最新文献

英文 中文
Genetic variants in PD-1 and its ligands, gene–gene and gene–environment interactions in allergic rhinitis 变应性鼻炎中PD-1及其配体的遗传变异、基因与基因及基因与环境的相互作用。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113912
Ruo-Xi Chen , Zheng Luan , Chong Shen , Meng-Di Dai , Chang-Yu Qiu , Xin-Jie Zhu , Qing-Zhao Zhang , Mei-Ping Lu , Lei Cheng

Background

The etiology of allergic rhinitis (AR), in which genetic and environmental factors are closely intertwined, has not yet been completely clarified. Programmed cell death 1 (PD-1) and its ligands (PD-L1 and PD-L2) regulate the immune and inflammatory responses during the development of immune-related and atopic diseases. To clarify the associations of genetic variants in PD-1, PD-L1 and PD-L2 with susceptibility to AR, gene–gene and gene–environment interactions were investigated.

Methods

A total of 452 AR patients and 495 controls were enrolled in this hospital-based case-control study. Eight single nucleotide polymorphisms (SNPs) in the PDCD1, PDCD1LG1 and PDCD1LG2 genes were genotyped. The correlations between SNPs and AR incidence, as well as gene–gene and gene–environment interactions were explored. Differentially expressed genes were screened by the Limma package in two Gene Expression Omnibus (GEO) datasets of AR patients. Expression qualitative trait locus (eQTL) analysis was performed via the Genotype-Tissue Expression (GTEx) database.

Results

The rs2297136 (A/G) in PDCD1LG1 was associated with a significantly increased risk of AR, whereas the PDCD1LG2 rs16923189 G allele was associated with a reduced risk of AR. In the subgroups according to AR-related phenotypes, the rs2297136 G allele increased, while the rs16923189 G allele reduced AR risk. Gene–gene interactions and gene–environment interactions (e.g., PDCD1LG1 polymorphisms with factors such as smoke, main road and cooking fumes) were verified in AR patients, but they were not significant after Bonferroni correction.

Conclusion

PDCD1LG1 rs2297136 and PDCD1LG2 rs16923189 are associated with susceptibility to AR in this Chinese population. The PD-1/PD-L1 and PD-1/PD-L2 signaling pathways may regulate gene–gene and gene–environment interactions in the pathogenesis of AR.
背景:变应性鼻炎(AR)的病因学尚未完全明确,遗传和环境因素密切相关。程序性细胞死亡1 (PD-1)及其配体(PD-L1和PD-L2)在免疫相关疾病和特应性疾病的发展过程中调节免疫和炎症反应。为了阐明PD-1、PD-L1和PD-L2基因变异与AR易感性的关系,研究了基因-基因和基因-环境相互作用。方法:共纳入452例AR患者和495名对照者,进行以医院为基础的病例对照研究。对PDCD1、PDCD1LG1和PDCD1LG2基因的8个单核苷酸多态性(snp)进行基因分型。探讨了snp与AR发病率的相关性,以及基因-基因和基因-环境的相互作用。采用Limma包在两个AR患者基因表达综合(GEO)数据集中筛选差异表达基因。通过基因型-组织表达(GTEx)数据库进行表达质量性状位点(eQTL)分析。结果:PDCD1LG1基因中的rs2297136 (A/G)与AR风险显著升高相关,而PDCD1LG2基因中的rs16923189 G等位基因与AR风险降低相关。在AR相关表型的亚组中,rs2297136 G等位基因升高,而rs16923189 G等位基因降低AR风险。在AR患者中证实了基因-基因相互作用和基因-环境相互作用(如PDCD1LG1多态性与烟雾、主干道和烹饪油烟等因素的相互作用),但经Bonferroni校正后不显著。结论:PDCD1LG1 rs2297136和PDCD1LG2 rs16923189与中国人群AR易感性相关。PD-1/PD-L1和PD-1/PD-L2信号通路可能调控AR发病过程中基因-基因和基因-环境相互作用。
{"title":"Genetic variants in PD-1 and its ligands, gene–gene and gene–environment interactions in allergic rhinitis","authors":"Ruo-Xi Chen ,&nbsp;Zheng Luan ,&nbsp;Chong Shen ,&nbsp;Meng-Di Dai ,&nbsp;Chang-Yu Qiu ,&nbsp;Xin-Jie Zhu ,&nbsp;Qing-Zhao Zhang ,&nbsp;Mei-Ping Lu ,&nbsp;Lei Cheng","doi":"10.1016/j.intimp.2024.113912","DOIUrl":"10.1016/j.intimp.2024.113912","url":null,"abstract":"<div><h3>Background</h3><div>The etiology of allergic rhinitis (AR), in which genetic and environmental factors are closely intertwined, has not yet been completely clarified. Programmed cell death 1 (PD-1) and its ligands (PD-L1 and PD-L2) regulate the immune and inflammatory responses during the development of immune-related and atopic diseases. To clarify the associations of genetic variants in PD-1, PD-L1 and PD-L2 with susceptibility to AR, gene–gene and gene–environment interactions were investigated.</div></div><div><h3>Methods</h3><div>A total of 452 AR patients and 495 controls were enrolled in this hospital-based case-control study. Eight single nucleotide polymorphisms (SNPs) in the <em>PDCD1</em>, <em>PDCD1LG1</em> and <em>PDCD1LG2</em> genes were genotyped. The correlations between SNPs and AR incidence, as well as gene–gene and gene–environment interactions were explored. Differentially expressed genes were screened by the Limma package in two Gene Expression Omnibus (GEO) datasets of AR patients. Expression qualitative trait locus (eQTL) analysis was performed via the Genotype-Tissue Expression (GTEx) database.</div></div><div><h3>Results</h3><div>The rs2297136 (A/G) in <em>PDCD1LG1</em> was associated with a significantly increased risk of AR, whereas the <em>PDCD1LG2</em> rs16923189 G allele was associated with a reduced risk of AR. In the subgroups according to AR-related phenotypes, the rs2297136 G allele increased, while the rs16923189 G allele reduced AR risk. Gene–gene interactions and gene–environment interactions (e.g., <em>PDCD1LG1</em> polymorphisms with factors such as smoke, main road and cooking fumes) were verified in AR patients, but they were not significant after Bonferroni correction.</div></div><div><h3>Conclusion</h3><div><em>PDCD1LG1</em> rs2297136 and <em>PDCD1LG2</em> rs16923189 are associated with susceptibility to AR in this Chinese population. The PD-1/PD-L1 and PD-1/PD-L2 signaling pathways may regulate gene–gene and gene–environment interactions in the pathogenesis of AR.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113912"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142964533","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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 GSDMD inhibits pyroptosis in psoriasis by blocking the NOD-like receptor signaling pathway. GSDMD的下调通过阻断nod样受体信号通路抑制银屑病的焦亡。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 Epub Date: 2025-01-07 DOI: 10.1016/j.intimp.2025.114036
Shuqin Lai, Huaqing Chen, Xiaojuan Ji, Wenjie Zhu, Zhiwei Wu, Shan Huang, Chunli Lin, Tao Yang, Zhaolin Zeng, Longnian Li

Background: Psoriasis is a chronic inflammatory skin disease regulated by autoimmunity, and pyroptosis plays an important role in this condition. This research sought to examine the function and potential molecular pathway of Gasdermin D (GSDMD) in psoriasis.

Methods: GSDMD expression was examined by immunohistochemistry in biopsied skin tissues from patients with psoriasis. Pyroptosis-related genes and inflammatory factors were quantified using qRT-PCR and ELISA, respectively. HaCaT cells were treated with M5 cytokines to develop an in vitro psoriasis model, while imiquimod (IMQ) was administered to construct an in vivo psoriasis model. To counteract the inhibition of the NOD-like receptor (NLR) pathway caused by GSDMD knockdown, the pathway activator M-TriDAP was employed.

Results: In the lesional skin tissues of patients with psoriasis, GSDMD expression was highly expressed. The levels of pro-pyroptosis mediators were increased, whereas the level of anti-inflammatory factor was lowered. GSDMD knockdown and disulfiram treatment inhibited pyroptosis and promoted apoptosis in M5-induced HaCaT cells. In the IMQ-induced psoriasis-like mouse model, GSDMD knockdown suppressed pyroptosis and improved skin lesion severity, alleviating erythema, epidermal thickness, and inflammatory cell infiltration. Mechanistically, GSDMD knockdown inhibited the NLR pathway, accompanied by reduced protein levels of NLRP3, NOD1, NOD2, and PYCARD. NLR pathway activator, M-TriDAP treatment significantly reversed the effects of GSDMD knockdown on psoriasis progression.

Conclusions: Knockdown of GSDMD inhibits pyroptosis in psoriasis by blocking the NLR signaling pathway, presenting a novel potential strategy for psoriasis treatment.

背景:银屑病是一种由自身免疫调节的慢性炎症性皮肤病,其中焦腐在银屑病中起重要作用。本研究旨在探讨GSDMD在银屑病中的功能和潜在的分子途径。方法:采用免疫组化方法检测银屑病患者皮肤组织中GSDMD的表达。分别采用qRT-PCR和ELISA法对热释热相关基因和炎症因子进行定量分析。用M5细胞因子处理HaCaT细胞建立银屑病体外模型,用咪喹莫特(IMQ)建立银屑病体内模型。为了抵消GSDMD敲低引起的nod样受体(NLR)通路的抑制,我们使用了通路激活剂M-TriDAP。结果:GSDMD在银屑病患者皮损组织中高表达。促焦亡介质水平升高,抗炎因子水平降低。GSDMD敲除和双硫仑处理抑制m5诱导的HaCaT细胞焦亡,促进细胞凋亡。在imq诱导的银屑病样小鼠模型中,GSDMD敲低抑制了焦下垂,改善了皮肤病变严重程度,减轻了红斑、表皮厚度和炎症细胞浸润。在机制上,GSDMD敲低抑制NLR通路,同时NLRP3、NOD1、NOD2和PYCARD蛋白水平降低。NLR通路激活剂M-TriDAP治疗显著逆转GSDMD敲除对银屑病进展的影响。结论:敲低GSDMD通过阻断NLR信号通路抑制银屑病热下垂,为银屑病治疗提供了一种新的潜在策略。
{"title":"Knockdown of GSDMD inhibits pyroptosis in psoriasis by blocking the NOD-like receptor signaling pathway.","authors":"Shuqin Lai, Huaqing Chen, Xiaojuan Ji, Wenjie Zhu, Zhiwei Wu, Shan Huang, Chunli Lin, Tao Yang, Zhaolin Zeng, Longnian Li","doi":"10.1016/j.intimp.2025.114036","DOIUrl":"10.1016/j.intimp.2025.114036","url":null,"abstract":"<p><strong>Background: </strong>Psoriasis is a chronic inflammatory skin disease regulated by autoimmunity, and pyroptosis plays an important role in this condition. This research sought to examine the function and potential molecular pathway of Gasdermin D (GSDMD) in psoriasis.</p><p><strong>Methods: </strong>GSDMD expression was examined by immunohistochemistry in biopsied skin tissues from patients with psoriasis. Pyroptosis-related genes and inflammatory factors were quantified using qRT-PCR and ELISA, respectively. HaCaT cells were treated with M5 cytokines to develop an in vitro psoriasis model, while imiquimod (IMQ) was administered to construct an in vivo psoriasis model. To counteract the inhibition of the NOD-like receptor (NLR) pathway caused by GSDMD knockdown, the pathway activator M-TriDAP was employed.</p><p><strong>Results: </strong>In the lesional skin tissues of patients with psoriasis, GSDMD expression was highly expressed. The levels of pro-pyroptosis mediators were increased, whereas the level of anti-inflammatory factor was lowered. GSDMD knockdown and disulfiram treatment inhibited pyroptosis and promoted apoptosis in M5-induced HaCaT cells. In the IMQ-induced psoriasis-like mouse model, GSDMD knockdown suppressed pyroptosis and improved skin lesion severity, alleviating erythema, epidermal thickness, and inflammatory cell infiltration. Mechanistically, GSDMD knockdown inhibited the NLR pathway, accompanied by reduced protein levels of NLRP3, NOD1, NOD2, and PYCARD. NLR pathway activator, M-TriDAP treatment significantly reversed the effects of GSDMD knockdown on psoriasis progression.</p><p><strong>Conclusions: </strong>Knockdown of GSDMD inhibits pyroptosis in psoriasis by blocking the NLR signaling pathway, presenting a novel potential strategy for psoriasis treatment.</p>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"114036"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948310","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Remimazolam inhibits apoptosis of endothelial and epithelial cells by activating the PI3K/AKT pathway in acute lung injury 雷马唑仑通过激活PI3K/AKT通路抑制急性肺损伤中内皮细胞和上皮细胞凋亡。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113949
Ruohan Li , Chuchu Zhang , Jiajia Ren , Guorong Deng , Ya Gao , Xiaoming Gao , Jiamei Li , Jingjing Zhang , Xi Xu , Xuting Jin , Xiaochuang Wang , Gang Wang

Background

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are significant burdens on global health. Remimazolam (REM), a novel sedative, has shown potential in its anti-inflammatory effects. However, a lack of evidence currently hinders our ability to determine if REM can improve ALI/ARDS.

Methods

We initially evaluated REM’s impact on lung injury in a lipopolysaccharide (LPS)–induced ALI mouse model. Subsequently, a network pharmacology (NP) strategy and ribonucleic acid–sequencing (RNA-seq) technique were used to investigate the potential molecular mechanisms underlying REM’s action against ALI. Finally, we carried out in vivo and in vitro experiments to validate our findings on these mechanisms.

Results

REM effectively mitigated lung injury in the mouse model. NP and RNA-seq analyses revealed significant enrichment of apoptosis-related pathways. Both in vivo and in vitro experiments revealed that REM significantly reduced levels of cleaved cysteine–aspartic acid–specific protease/proteinases 7 and 3 (cleaved Caspases-7 and −3) and cytochrome c (Cyt c) while enhancing the B-cell lymphoma 2 (Bcl-2)/Bcl-2–like protein 4 (Bax) ratio and phosphorylated protein kinase B (P-AKT) levels in lung tissue, endothelial cells, and epithelial cells. Furthermore, in vitro experiments confirmed that inhibiting the phosphoinositide 3-kinase (PI3K)/AKT pathway with LY294002 weakened REM’s antiapoptotic effects. In addition, pretreatment with PK11195 (the ligand of 18-kDa translocator protein [TSPO]) attenuated REM’s upregulation of the PI3K/AKT pathway and antiapoptotic effect in LPS-induced endothelial cells.

Conclusions

This study presents novel findings elucidating the beneficial effect of REM in ALI. This effect can be attributed to REM’s ability to inhibit apoptosis by activating of the PI3K/AKT pathway in endothelial and epithelial cells. Additionally, REM targeted TSPO to regulate this pathway in endothelial cells. These results suggested a potential protective role for REM in ALI/ARDS management.
背景:急性肺损伤(ALI)和急性呼吸窘迫综合征(ARDS)是全球健康的重大负担。雷马唑仑(Remimazolam, REM)是一种新型的镇静剂,具有潜在的抗炎作用。然而,目前缺乏证据阻碍了我们确定REM是否可以改善ALI/ARDS。方法:我们在脂多糖(LPS)诱导的ALI小鼠模型中初步评估REM对肺损伤的影响。随后,利用网络药理学(NP)策略和核糖核酸测序(RNA-seq)技术研究REM对ALI作用的潜在分子机制。最后,我们进行了体内和体外实验来验证我们对这些机制的发现。结果:快速眼动可有效减轻小鼠肺损伤。NP和RNA-seq分析显示凋亡相关通路显著富集。体内和体外实验均显示,REM显著降低了肺组织、内皮细胞和上皮细胞中裂解型半胱氨酸-天冬氨酸特异性蛋白酶/蛋白酶7和蛋白酶3(裂解型caspase -7和-3)和细胞色素c (Cyt c)水平,同时提高了B细胞淋巴瘤2 (Bcl-2)/Bcl-2样蛋白4 (Bax)比率和磷酸化蛋白激酶B (P-AKT)水平。此外,体外实验证实LY294002抑制磷酸肌苷3-激酶(PI3K)/AKT通路减弱REM的抗凋亡作用。此外,在lps诱导的内皮细胞中,用PK11195 (18kda转运蛋白[TSPO]的配体)预处理可减弱REM对PI3K/AKT通路的上调和抗凋亡作用。结论:本研究提出了新的发现,阐明了快速眼动在急性脑损伤中的有益作用。这种作用可归因于REM通过激活内皮细胞和上皮细胞的PI3K/AKT通路抑制细胞凋亡的能力。此外,REM靶向TSPO调节内皮细胞的这一途径。这些结果提示REM在ALI/ARDS治疗中具有潜在的保护作用。
{"title":"Remimazolam inhibits apoptosis of endothelial and epithelial cells by activating the PI3K/AKT pathway in acute lung injury","authors":"Ruohan Li ,&nbsp;Chuchu Zhang ,&nbsp;Jiajia Ren ,&nbsp;Guorong Deng ,&nbsp;Ya Gao ,&nbsp;Xiaoming Gao ,&nbsp;Jiamei Li ,&nbsp;Jingjing Zhang ,&nbsp;Xi Xu ,&nbsp;Xuting Jin ,&nbsp;Xiaochuang Wang ,&nbsp;Gang Wang","doi":"10.1016/j.intimp.2024.113949","DOIUrl":"10.1016/j.intimp.2024.113949","url":null,"abstract":"<div><h3>Background</h3><div>Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are significant burdens on global health. Remimazolam (REM), a novel sedative, has shown potential in its anti-inflammatory effects. However, a lack of evidence currently hinders our ability to determine if REM can improve ALI/ARDS.</div></div><div><h3>Methods</h3><div>We initially evaluated REM’s impact on lung injury in a lipopolysaccharide (LPS)–induced ALI mouse model. Subsequently, a network pharmacology (NP) strategy and ribonucleic acid–sequencing (RNA-seq) technique were used to investigate the potential molecular mechanisms underlying REM’s action against ALI. Finally, we carried out <em>in vivo</em> and <em>in vitro</em> experiments to validate our findings on these mechanisms.</div></div><div><h3>Results</h3><div>REM effectively mitigated lung injury in the mouse model. NP and RNA-seq analyses revealed significant enrichment of apoptosis-related pathways. Both <em>in vivo</em> and <em>in vitro</em> experiments revealed that REM significantly reduced levels of cleaved cysteine–aspartic acid–specific protease/proteinases 7 and 3 (cleaved Caspases-7 and −3) and cytochrome <em>c</em> (Cyt c) while enhancing the B-cell lymphoma 2 (Bcl-2)/Bcl-2–like protein 4 (Bax) ratio and phosphorylated protein kinase B (P-AKT) levels in lung tissue, endothelial cells, and epithelial cells. Furthermore, <em>in vitro</em> experiments confirmed that inhibiting the phosphoinositide 3-kinase (PI3K)/AKT pathway with LY294002 weakened REM’s antiapoptotic effects. In addition, pretreatment with PK11195 (the ligand of 18-kDa translocator protein [TSPO]) attenuated REM’s upregulation of the PI3K/AKT pathway and antiapoptotic effect in LPS-induced endothelial cells.</div></div><div><h3>Conclusions</h3><div>This study presents novel findings elucidating the beneficial effect of REM in ALI. This effect can be attributed to REM’s ability to inhibit apoptosis by activating of the PI3K/AKT pathway in endothelial and epithelial cells. Additionally, REM targeted TSPO to regulate this pathway in endothelial cells. These results suggested a potential protective role for REM in ALI/ARDS management.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113949"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142914545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Clinical implications of human Parvovirus B19 infection on autoimmunity and autoimmune diseases 人细小病毒B19感染对自身免疫和自身免疫性疾病的临床意义。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113960
Chih-Chen Tzang , Liang-Yun Chi , Chen-Yu Lee , Zi-Yi Chang , Chiao-An Luo , Yan-Hua Chen , Tzu-An Lin , Liang-Chien Yu , Yo-Rong Chen , Bor-Show Tzang , Tsai-Ching Hsu
Parvovirus B19 (B19V) is a human pathogen from the Parvoviridae family that primarily targets and replicates in erythroid progenitor cells (EPCs). While its symptoms are typically self-limiting in healthy individuals, B19V can cause or exacerbate autoimmune diseases in vulnerable patients. This review integrates the involvement of B19V in the development and worsening of several autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), hematological disorders (thalassemia, anemia, and thrombocytopenia), vasculitis, antiphospholipid syndrome (APS), dermatological disease (systemic sclerosis, psoriasis), autoimmune thyroid disease, myocarditis, and myasthenia gravis, and autoinflammatory disease of adult-onset Still’s disease (AOSD). B19V contributes to autoimmunity and autoimmune disease onset and progression through mechanisms such as molecular mimicry, immune system disruption, and chronic infection. By summarizing findings from in vitro experiments, clinical case studies, seroprevalence data, and biopsy results, this review highlights the critical connection between B19V and autoimmune disease development. Recognizing the role of B19V in the early diagnosis and management of these conditions is essential, as its presence may influence the disease course and severity. Greater awareness among healthcare professionals and the public is necessary to address the impact of B19V, leading to more accurate diagnoses and better-informed treatment approaches for autoimmune diseases linked to the virus.
细小病毒B19 (B19V)是细小病毒科的一种人类病原体,主要以红系祖细胞(EPCs)为靶点并进行复制。虽然其症状在健康个体中通常是自限性的,但B19V可能导致或加剧脆弱患者的自身免疫性疾病。本综述整合了B19V在几种自身免疫性疾病的发展和恶化中的作用,包括系统性红斑狼疮(SLE)、类风湿性关节炎(RA)、青少年特发性关节炎(JIA)、血液系统疾病(地中海贫血、贫血和血小板减少症)、血管炎、抗磷脂综合征(APS)、皮肤病(系统性硬化症、牛皮癣)、自身免疫性甲状腺疾病、心肌炎和重症肌无力。成人发病斯蒂尔氏病(AOSD)的自身炎症性疾病。B19V通过分子模仿、免疫系统破坏和慢性感染等机制促进自身免疫和自身免疫性疾病的发生和进展。通过总结体外实验、临床病例研究、血清阳性率数据和活检结果,本综述强调了B19V与自身免疫性疾病发展之间的重要联系。认识到B19V在这些疾病的早期诊断和管理中的作用是必不可少的,因为它的存在可能影响疾病的进程和严重程度。医疗保健专业人员和公众有必要提高对B19V影响的认识,从而更准确地诊断和更好地了解与该病毒有关的自身免疫性疾病的治疗方法。
{"title":"Clinical implications of human Parvovirus B19 infection on autoimmunity and autoimmune diseases","authors":"Chih-Chen Tzang ,&nbsp;Liang-Yun Chi ,&nbsp;Chen-Yu Lee ,&nbsp;Zi-Yi Chang ,&nbsp;Chiao-An Luo ,&nbsp;Yan-Hua Chen ,&nbsp;Tzu-An Lin ,&nbsp;Liang-Chien Yu ,&nbsp;Yo-Rong Chen ,&nbsp;Bor-Show Tzang ,&nbsp;Tsai-Ching Hsu","doi":"10.1016/j.intimp.2024.113960","DOIUrl":"10.1016/j.intimp.2024.113960","url":null,"abstract":"<div><div>Parvovirus B19 (B19V) is a human pathogen from the Parvoviridae family that primarily targets and replicates in erythroid progenitor cells (EPCs). While its symptoms are typically self-limiting in healthy individuals, B19V can cause or exacerbate autoimmune diseases in vulnerable patients. This review integrates the involvement of B19V in the development and worsening of several autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), hematological disorders (thalassemia, anemia, and thrombocytopenia), vasculitis, antiphospholipid syndrome (APS), dermatological disease (systemic sclerosis, psoriasis), autoimmune thyroid disease, myocarditis, and myasthenia gravis, and autoinflammatory disease of adult-onset Still’s disease (AOSD). B19V contributes to autoimmunity and autoimmune disease onset and progression through mechanisms such as molecular mimicry, immune system disruption, and chronic infection. By summarizing findings from in vitro experiments, clinical case studies, seroprevalence data, and biopsy results, this review highlights the critical connection between B19V and autoimmune disease development. Recognizing the role of B19V in the early diagnosis and management of these conditions is essential, as its presence may influence the disease course and severity. Greater awareness among healthcare professionals and the public is necessary to address the impact of B19V, leading to more accurate diagnoses and better-informed treatment approaches for autoimmune diseases linked to the virus.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113960"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142921678","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Aerosol inhalation of rhIL-10 improves acute lung injury in mice by affecting pulmonary neutrophil phenotypes through neutrophil-platelet aggregates 气溶胶吸入rhIL-10通过中性粒细胞-血小板聚集影响肺中性粒细胞表型,改善小鼠急性肺损伤。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113948
Huan Qin , Jiangang Wang , Luyuan Bai , Huiqin Ding , Hailing Ding , Fengyi Zhang , Yantao Han
This study investigates the therapeutic effects of recombinant human IL-10 (rhIL-10) administered via aerosol inhalation in acute lung injury (ALI), with a particular focus on neutrophils. It explores how rhIL-10, in the presence of platelets, modulates neutrophil polarization to ameliorate acute lung injury. Initially, the ALI model established in mice demonstrated that aerosol inhalation of rhIL-10 significantly mitigated the cytokine storm in the lungs, reduced pulmonary edema, and alleviated histopathological damage to lung tissue. Additionally, rhIL-10 administration was found to decrease neutrophil infiltration and platelet activation in the lungs of mice, inhibiting the formation of platelet-neutrophil aggregates (PNAs) and promoting the differentiation of neutrophils toward an anti-inflammatory phenotype in the presence of platelets. Subsequently, primary neutrophils and platelets were isolated from mouse bone marrow and blood to explore the underlying mechanisms. The results indicated that rhIL-10 promotes the expression of the signal transducer and activator of transcription 3 (STAT3) and the suppressor of cytokine signaling 3 (SOCS3) in neutrophils while inhibiting the activation of the nuclear factor kappa B (NF-κB) and the NF-κB inhibitor (IκB), which in turn enhances CD40 expression. This interaction facilitates the formation of PNAs and influences neutrophil phenotype differentiation. Furthermore, the application of the STAT3 phosphorylation inhibitor Stattic and CD40 antibody in vivo provided further validation of this potential mechanism. In conclusion, these results indicate that aerosol inhalation of rhIL-10 effectively ameliorates ALI. The underlying mechanism may involve the modulation of the neutrophil STAT/SOCS-IκB/NF-κB-CD40 signaling pathway, promoting interactions between neutrophils and platelets that facilitate the differentiation of neutrophils toward an anti-inflammatory phenotype.
本研究探讨了重组人IL-10 (rhIL-10)通过气溶胶吸入治疗急性肺损伤(ALI)的效果,特别关注中性粒细胞。它探讨了在血小板存在下rhIL-10如何调节中性粒细胞极化以改善急性肺损伤。最初,在小鼠中建立的ALI模型表明,气溶胶吸入rhIL-10可显著减轻肺部细胞因子风暴,减轻肺水肿,减轻肺组织的组织病理学损伤。此外,研究发现rhIL-10可以减少小鼠肺部的中性粒细胞浸润和血小板活化,抑制血小板-中性粒细胞聚集体(PNAs)的形成,并在血小板存在的情况下促进中性粒细胞向抗炎表型分化。随后,从小鼠骨髓和血液中分离出原代中性粒细胞和血小板,以探索其潜在的机制。结果表明,rhIL-10可促进中性粒细胞中信号转导和转录激活因子3 (STAT3)和细胞因子信号传导抑制因子3 (SOCS3)的表达,抑制核因子κB (NF-κB)和NF-κB抑制剂(i -κB)的激活,从而增强CD40的表达。这种相互作用促进PNAs的形成并影响中性粒细胞表型分化。此外,STAT3磷酸化抑制剂statstatic和CD40抗体在体内的应用进一步验证了这一潜在机制。综上所述,这些结果表明,气溶胶吸入rhIL-10可有效改善ALI。其潜在机制可能涉及中性粒细胞STAT/SOCS-IκB/NF-κB-CD40信号通路的调节,促进中性粒细胞和血小板之间的相互作用,从而促进中性粒细胞向抗炎表型分化。
{"title":"Aerosol inhalation of rhIL-10 improves acute lung injury in mice by affecting pulmonary neutrophil phenotypes through neutrophil-platelet aggregates","authors":"Huan Qin ,&nbsp;Jiangang Wang ,&nbsp;Luyuan Bai ,&nbsp;Huiqin Ding ,&nbsp;Hailing Ding ,&nbsp;Fengyi Zhang ,&nbsp;Yantao Han","doi":"10.1016/j.intimp.2024.113948","DOIUrl":"10.1016/j.intimp.2024.113948","url":null,"abstract":"<div><div>This study investigates the therapeutic effects of recombinant human IL-10 (rhIL-10) administered via aerosol inhalation in acute lung injury (ALI), with a particular focus on neutrophils. It explores how rhIL-10, in the presence of platelets, modulates neutrophil polarization to ameliorate acute lung injury. Initially, the ALI model established in mice demonstrated that aerosol inhalation of rhIL-10 significantly mitigated the cytokine storm in the lungs, reduced pulmonary edema, and alleviated histopathological damage to lung tissue. Additionally, rhIL-10 administration was found to decrease neutrophil infiltration and platelet activation in the lungs of mice, inhibiting the formation of platelet-neutrophil aggregates (PNAs) and promoting the differentiation of neutrophils toward an anti-inflammatory phenotype in the presence of platelets. Subsequently, primary neutrophils and platelets were isolated from mouse bone marrow and blood to explore the underlying mechanisms. The results indicated that rhIL-10 promotes the expression of the signal transducer and activator of transcription 3 (STAT3) and the suppressor of cytokine signaling 3 (SOCS3) in neutrophils while inhibiting the activation of the nuclear factor kappa B (NF-κB) and the NF-κB inhibitor (IκB), which in turn enhances CD40 expression. This interaction facilitates the formation of PNAs and influences neutrophil phenotype differentiation. Furthermore, the application of the STAT3 phosphorylation inhibitor Stattic and CD40 antibody in vivo provided further validation of this potential mechanism. In conclusion, these results indicate that aerosol inhalation of rhIL-10 effectively ameliorates ALI. The underlying mechanism may involve the modulation of the neutrophil STAT/SOCS-IκB/NF-κB-CD40 signaling pathway, promoting interactions between neutrophils and platelets that facilitate the differentiation of neutrophils toward an anti-inflammatory phenotype.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113948"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948482","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Role of COL5A1 in lung squamous cell Carcinoma: Prognostic Implications and therapeutic potential COL5A1在肺鳞癌中的作用:预后意义和治疗潜力。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113977
Chengjuan Zhang , Bo Wang , Tingjie Wang , Chi Yan , Jing Yuan , Peng Li , Bin Ma , Tao Wang , Benling Xu , Ruihua Bai , Xiance Tang , Youwei Shi , Minqing Wu , Tianqi Lei , Wenhao Xu , Ning Li , Yongjun Guo

Background

Lung squamous cell carcinoma (LUSC) is a significant health concern, characterized by a lack of specific therapies and limited treatment options for patients in advanced stages. This study aims to identify key molecules of prognostic importance in LUSC and provide an experimental foundation for their potential therapeutic applications.

Methods

Immune-related transcriptome expression analysis was performed on LUSC samples using the NanoString digital gene analysis system to develop a prognostic transcriptomic signature. This was followed by validation within the LUSC cohort database, and the immune properties and cellular functions of the critical molecule were examined through molecular biology experiments.

Results

Advanced nCounter analysis revealed significant differences in the numbers of T cells, cytotoxic cells, B cells, and CD45+ and CD8+ T cells between the OS1 (short-term survival) group and the OS2 (long-term survival) group. A comparison of the differences in tumor immune-related pathways between the two groups revealed that signaling pathways such as the PI3K-AKT, NF-kappaB signaling, Notch signaling, angiogenesis, matrix remodeling, and metastasis pathways were activated in the OS1 subgroup, and DNA damage repair and lymphatic chamber signaling pathways were activated in the OS2 subgroup. We analyzed and compared differentially expressed mRNAs with high expression levels in the OS1 and stage IV groups. Collagen type V alpha 1 (COL5A1) was found to be associated with the prognosis of LUSC. Phenotypic analysis revealed that COL5A1 knockdown inhibited the proliferation, migration, and invasion of SKMES1 cells. Locating COL5A1 was shown to be expressed in CAFs, T cells, and EPI cells through single-cell omics analysis.

Conclusion

COL5A1 plays a crucial role in tumor progression, indicating that COL5A1 inhibitors may represent a promising therapeutic strategy for the treatment of LUSC.
背景:肺鳞状细胞癌(LUSC)是一个重要的健康问题,其特点是缺乏特异性治疗方法,晚期患者的治疗选择有限。本研究旨在确定在LUSC中具有预后重要性的关键分子,并为其潜在的治疗应用提供实验基础。方法:使用NanoString数字基因分析系统对LUSC样本进行免疫相关转录组表达分析,以建立预后转录组特征。随后在LUSC队列数据库中进行验证,并通过分子生物学实验检查关键分子的免疫特性和细胞功能。结果:高级nCounter分析显示,OS1(短期生存)组和OS2(长期生存)组在T细胞、细胞毒性细胞、B细胞、CD45+和CD8+ T细胞数量上存在显著差异。比较两组肿瘤免疫相关通路的差异发现,OS1亚组激活了PI3K-AKT、NF-kappaB、Notch、血管生成、基质重塑和转移等信号通路,OS2亚组激活了DNA损伤修复和淋巴室信号通路。我们分析并比较了OS1和IV期组中高表达的差异mrna。发现V α 1型胶原蛋白(COL5A1)与LUSC的预后相关。表型分析显示COL5A1敲低抑制SKMES1细胞的增殖、迁移和侵袭。通过单细胞组学分析发现COL5A1在CAFs、T细胞和EPI细胞中表达。结论:COL5A1在肿瘤进展中起着至关重要的作用,表明COL5A1抑制剂可能是治疗LUSC的一种有前景的治疗策略。
{"title":"Role of COL5A1 in lung squamous cell Carcinoma: Prognostic Implications and therapeutic potential","authors":"Chengjuan Zhang ,&nbsp;Bo Wang ,&nbsp;Tingjie Wang ,&nbsp;Chi Yan ,&nbsp;Jing Yuan ,&nbsp;Peng Li ,&nbsp;Bin Ma ,&nbsp;Tao Wang ,&nbsp;Benling Xu ,&nbsp;Ruihua Bai ,&nbsp;Xiance Tang ,&nbsp;Youwei Shi ,&nbsp;Minqing Wu ,&nbsp;Tianqi Lei ,&nbsp;Wenhao Xu ,&nbsp;Ning Li ,&nbsp;Yongjun Guo","doi":"10.1016/j.intimp.2024.113977","DOIUrl":"10.1016/j.intimp.2024.113977","url":null,"abstract":"<div><h3>Background</h3><div>Lung squamous cell carcinoma (LUSC) is a significant health concern, characterized by a lack of specific therapies and limited treatment options for patients in advanced stages. This study aims to identify key molecules of prognostic importance in LUSC and provide an experimental foundation for their potential therapeutic applications.</div></div><div><h3>Methods</h3><div>Immune-related transcriptome expression analysis was performed on LUSC samples using the NanoString digital gene analysis system to develop a prognostic transcriptomic signature. This was followed by validation within the LUSC cohort database, and the immune properties and cellular functions of the critical molecule were examined through molecular biology experiments.</div></div><div><h3>Results</h3><div>Advanced nCounter analysis revealed significant differences in the numbers of T cells, cytotoxic cells, B cells, and CD45<sup>+</sup> and CD8<sup>+</sup> T cells between the OS1 (short-term survival) group and the OS2 (long-term survival) group. A comparison of the differences in tumor immune-related pathways between the two groups revealed that signaling pathways such as the PI3K-AKT, NF-kappaB signaling, Notch signaling, angiogenesis, matrix remodeling, and metastasis pathways were activated in the OS1 subgroup, and DNA damage repair and lymphatic chamber signaling pathways were activated in the OS2 subgroup. We analyzed and compared differentially expressed mRNAs with high expression levels in the OS1 and stage IV groups. Collagen type V alpha 1 (COL5A1) was found to be associated with the prognosis of LUSC. Phenotypic analysis revealed that COL5A1 knockdown inhibited the proliferation, migration, and invasion of SKMES1 cells. Locating COL5A1 was shown to be expressed in CAFs, T cells, and EPI cells through single-cell omics analysis.</div></div><div><h3>Conclusion</h3><div>COL5A1 plays a crucial role in tumor progression, indicating that COL5A1 inhibitors may represent a promising therapeutic strategy for the treatment of LUSC.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113977"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142925992","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Rhnull blood group caused by novel base deletion and comprehensive pedigree analysis 新型碱基缺失致Rhnull血型及综合家系分析。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113993
Zhu Xiaoli , Qi Xi , Gao Hongjun , Zhu Ziqing , Sha Yuxuan , Qin Yi , Li Anming , Zhu Jianfeng , Sha Yayun , Han Junling , Gao Lingbao

Objective

The objective of this study was to rigorously investigate and elucidate the genetic mechanisms underlying the formation of the RHnull blood group in a specific case and to systematically analyse the RH blood group genes among the family members of the proband.

Methods

Serological methods were used to determine the RH blood group phenotype of the proband. To elucidate the underlying genetic mechanism responsible for the RHnull phenotype, a comprehensive approach was undertaken, including RHCE genotyping, sequencing of RHD and RHCE genes, and exon sequencing of RHAG. For a comparative analysis, the same methodologies were applied to two family members of the proband.

Results

The genotype of the proband was determined as CcDEe. Subsequent RHAG exon sequencing analysis revealed a homozygous frameshift mutation in exon 5. Specifically, a nucleotide deletion at position c.732 in RHAG resulted in an amino acid substitution from phenylalanine to serine, followed by a frameshift and premature termination at codon 245 (p.Phe245Serfs*16). This mutation was confirmed as a novel genetic variant in the NCBI database.
Furthermore, serological findings, genotyping results, and RHAG exon sequencing data obtained from the proband’s sister were identical to those of the proband. In contrast, the proband’s son exhibited a serological phenotype of CCDee with a corresponding genotyping result for CCDee. RHAG exon sequencing of the son revealed a heterozygous frameshift mutation, which was consistent with the findings observed in the proband.

Conclusion

A novel mutation, specifically c.732delC, was identified in RHAG. The RHnull phenotype observed in this subject was attributed to a homozygous frameshift mutation in this gene. This mutation results in a truncated and nonfunctional RHAG protein, which subsequently disrupts the expression of other RH antigens on the cell membrane. Therefore, the serological phenotype associated with this genetic anomaly was classified as RHnull.
目的:本研究的目的是严格调查和阐明在特定情况下RH血型形成的遗传机制,并系统地分析先证家庭成员中的RH血型基因。方法:采用血清学方法测定先证者RH血型表型。为了阐明RHnull表型的潜在遗传机制,研究人员采用了一种综合的方法,包括RHCE基因分型、RHD和RHCE基因测序以及RHAG的外显子测序。为了进行比较分析,将相同的方法应用于先证者的两个家庭成员。结果:先证者基因型确定为CcDEe。随后的RHAG外显子测序分析显示外显子5有纯合移码突变。具体来说,在RHAG中c.732位的核苷酸缺失导致苯丙氨酸的氨基酸替换为丝氨酸,随后在密码子245处发生移码和过早终止(p.Phe245Serfs*16)。该突变在NCBI数据库中被确认为一种新的遗传变异。此外,从先证者的姐妹获得的血清学结果、基因分型结果和RHAG外显子测序数据与先证者相同。相比之下,先证者的儿子表现出CCDee的血清学表型和相应的CCDee基因分型结果。儿子的RHAG外显子测序显示一个杂合移码突变,这与先证者观察到的结果一致。结论:在RHAG中发现了一种新的突变,特别是c.732delC。在这个对象中观察到的RHnull表型归因于该基因的纯合移码突变。这种突变导致截断和无功能的RHAG蛋白,随后破坏细胞膜上其他RH抗原的表达。因此,与该遗传异常相关的血清学表型被归类为RHnull。
{"title":"Rhnull blood group caused by novel base deletion and comprehensive pedigree analysis","authors":"Zhu Xiaoli ,&nbsp;Qi Xi ,&nbsp;Gao Hongjun ,&nbsp;Zhu Ziqing ,&nbsp;Sha Yuxuan ,&nbsp;Qin Yi ,&nbsp;Li Anming ,&nbsp;Zhu Jianfeng ,&nbsp;Sha Yayun ,&nbsp;Han Junling ,&nbsp;Gao Lingbao","doi":"10.1016/j.intimp.2024.113993","DOIUrl":"10.1016/j.intimp.2024.113993","url":null,"abstract":"<div><h3>Objective</h3><div>The objective of this study was to rigorously investigate and elucidate the genetic mechanisms underlying the formation of the RH<sub>null</sub> blood group in a specific case and to systematically analyse the RH blood group genes among the family members of the proband.</div></div><div><h3>Methods</h3><div>Serological methods were used to determine the RH blood group phenotype of the proband. To elucidate the underlying genetic mechanism responsible for the RH<sub>null</sub> phenotype, a comprehensive approach was undertaken, including RHCE genotyping, sequencing of RHD and RHCE genes, and exon sequencing of RHAG. For a comparative analysis, the same methodologies were applied to two family members of the proband.</div></div><div><h3>Results</h3><div>The genotype of the proband was determined as <em>CcDEe</em>. Subsequent RHAG exon sequencing analysis revealed a homozygous frameshift mutation in exon 5. Specifically, a nucleotide deletion at position c.732 in RHAG resulted in an amino acid substitution from phenylalanine to serine, followed by a frameshift and premature termination at codon 245 (p.Phe245Serfs*16). This mutation was confirmed as a novel genetic variant in the NCBI database.</div><div>Furthermore, serological findings, genotyping results, and RHAG exon sequencing data obtained from the proband’s sister were identical to those of the proband. In contrast, the proband’s son exhibited a serological phenotype of CCDee with a corresponding genotyping result for <em>CCDee.</em> RHAG exon sequencing of the son revealed a heterozygous frameshift mutation, which was consistent with the findings observed in the proband.</div></div><div><h3>Conclusion</h3><div>A novel mutation, specifically c.732delC, was identified in RHAG. The RH<sub>null</sub> phenotype observed in this subject was attributed to a homozygous frameshift mutation in this gene. This mutation results in a truncated and nonfunctional RHAG protein, which subsequently disrupts the expression of other RH antigens on the cell membrane. Therefore, the serological phenotype associated with this genetic anomaly was classified as RH<sub>null</sub>.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113993"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142925983","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
FcγRI plays a pro-inflammatory role in the immune response to Chlamydia respiratory infection by upregulating dendritic cell-related genes fc γ - ri通过上调树突状细胞相关基因在衣原体呼吸道感染的免疫应答中发挥促炎作用。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113943
Ruoyuan Sun , Jinxi Yu , Zeyang Zou , Shuaini Yang , Yuqing Tuo , Lu Tan , Hong Zhang , Longhao Sun , Hong Bai

Background

FcγRI, a pivotal cell surface receptor, is implicated in diverse immune responses and is ubiquitously expressed on numerous immune cells. However, its role in intracellular bacterial infections remains understudied.

Methods

Wild-type (WT) and FcγRI knockout (FcγRI-KO) mice were inoculated intranasally with a specific dose of C. muridarum. Lung tissues were harvested for transcriptome sequencing, and flow cytometry was employed to validate bioinformatics immune infiltration analysis. Differentially expressed DC-associated genes were subjected to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses to elucidate their functions during infection. A PPI network was constructed to pinpoint crucial genes, and qPCR was utilized to confirm their expression changes. Additionally, we compared body weight, lung Chlamydia load, and pathological alterations between WT and FcγRI-KO mice post-infection to assess the effect of FcγRI on inflammation via gene regulation. Lastly, an mRNA-miRNA-lncRNA network was formulated to further probe the molecular mechanisms of FcγRI in C. muridarum infection.

Results

Post-C. muridarum infection, FcγRI-KO mice exhibited a notable decrease in DC infiltration and maturation, along with downregulated co-stimulatory molecules (CD40, CD80, CD86) in lung tissues. Differential gene analysis identified 26 differentially expressed DC-related genes implicated in DC proliferation, migration, and inflammatory responses. KEGG analysis revealed their close association with key immune pathways. The PPI network delineated two modules, with the top six genes in the pivotal cluster 1 (Ccl4, Il6, Ccl3, Ptgs2, Il 1α, Il7) being significantly downregulated in FcγRI-KO mice. A ceRNA network encompassing 12 miRNAs and 37 lncRNAs regulating four key genes (Ptgs2, Il1α, Il6, Il7) was also constructed.

Conclusions

In C. muridarum respiratory infections, FcγRI facilitates DC infiltration and maturation, upregulates six pro-inflammatory genes (Ccl4, Il6, Ccl3, Ptgs2, Il1α, Il7), and exhibits a pro-inflammatory role. A key ceRNA network was formulated to unravel the underlying molecular mechanisms.
背景:fc γ - ri是一种关键的细胞表面受体,参与多种免疫反应,在许多免疫细胞中普遍表达。然而,其在细胞内细菌感染中的作用仍未得到充分研究。方法:以野生型(WT)和fc γ γ ri敲除型(FcγRI- ko)小鼠为研究对象,经鼻灌胃接种一定剂量的毒舌草。采集肺组织进行转录组测序,流式细胞术验证生物信息学免疫浸润分析。对差异表达的dc相关基因进行基因本体和京都基因与基因组百科分析,以阐明其在感染过程中的功能。构建PPI网络以确定关键基因,并利用qPCR确认其表达变化。此外,我们比较了WT和fc γ γ - ri - ko小鼠感染后的体重、肺衣原体负荷和病理变化,以评估fc γ - ri通过基因调控对炎症的影响。最后,我们构建了mRNA-miRNA-lncRNA网络,进一步探讨了fc γ - ri在muridarum感染中的分子机制。结果:Post-C。感染后,FcγRI-KO小鼠DC浸润和成熟明显减少,肺组织中共刺激分子(CD40, CD80, CD86)下调。差异基因分析确定了26个差异表达的DC相关基因,这些基因与DC增殖、迁移和炎症反应有关。KEGG分析显示它们与关键免疫通路密切相关。PPI网络描绘了两个模块,关键簇1中的前六个基因(Ccl4, Il6, Ccl3, Ptgs2, Il 1α, Il7)在FcγRI-KO小鼠中显著下调。构建了一个包含12个mirna和37个lncrna的ceRNA网络,调控4个关键基因(Ptgs2, Il1α, Il6, Il7)。结论:在muridarum呼吸道感染中,FcγRI促进DC浸润和成熟,上调6个促炎基因(Ccl4、Il6、Ccl3、Ptgs2、Il1α、Il7),并表现出促炎作用。制定了一个关键的ceRNA网络来揭示潜在的分子机制。
{"title":"FcγRI plays a pro-inflammatory role in the immune response to Chlamydia respiratory infection by upregulating dendritic cell-related genes","authors":"Ruoyuan Sun ,&nbsp;Jinxi Yu ,&nbsp;Zeyang Zou ,&nbsp;Shuaini Yang ,&nbsp;Yuqing Tuo ,&nbsp;Lu Tan ,&nbsp;Hong Zhang ,&nbsp;Longhao Sun ,&nbsp;Hong Bai","doi":"10.1016/j.intimp.2024.113943","DOIUrl":"10.1016/j.intimp.2024.113943","url":null,"abstract":"<div><h3>Background</h3><div>FcγRI, a pivotal cell surface receptor, is implicated in diverse immune responses and is ubiquitously expressed on numerous immune cells. However, its role in intracellular bacterial infections remains understudied.</div></div><div><h3>Methods</h3><div>Wild-type (WT) and FcγRI knockout (<em>FcγRI</em>-KO) mice were inoculated intranasally with a specific dose of <em>C. muridarum</em>. Lung tissues were harvested for transcriptome sequencing, and flow cytometry was employed to validate bioinformatics immune infiltration analysis. Differentially expressed DC-associated genes were subjected to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses to elucidate their functions during infection. A PPI network was constructed to pinpoint crucial genes, and qPCR was utilized to confirm their expression changes. Additionally, we compared body weight, lung Chlamydia load, and pathological alterations between WT and <em>FcγRI</em>-KO mice post-infection to assess the effect of <em>FcγRI</em> on inflammation via gene regulation. Lastly, an mRNA-miRNA-lncRNA network was formulated to further probe the molecular mechanisms of FcγRI in <em>C. muridarum</em> infection.</div></div><div><h3>Results</h3><div>Post-<em>C. muridarum</em> infection, <em>FcγRI</em>-KO mice exhibited a notable decrease in DC infiltration and maturation, along with downregulated co-stimulatory molecules (CD40, CD80, CD86) in lung tissues. Differential gene analysis identified 26 differentially expressed DC-related genes implicated in DC proliferation, migration, and inflammatory responses. KEGG analysis revealed their close association with key immune pathways. The PPI network delineated two modules, with the top six genes in the pivotal cluster 1 (<em>Ccl4, Il6, Ccl3, Ptgs2, Il 1α, Il7</em>) being significantly downregulated in <em>FcγRI</em>-KO mice. A ceRNA network encompassing 12 miRNAs and 37 lncRNAs regulating four key genes (<em>Ptgs2, Il1α, Il6, Il7</em>) was also constructed.</div></div><div><h3>Conclusions</h3><div>In <em>C. muridarum</em> respiratory infections, FcγRI facilitates DC infiltration and maturation, upregulates six pro-inflammatory genes (<em>Ccl4, Il6, Ccl3, Ptgs2, Il1α, Il7</em>), and exhibits a pro-inflammatory role. A key ceRNA network was formulated to unravel the underlying molecular mechanisms.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113943"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927231","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Leptin, NK cells, and the weight of immunity: Insights into obesity 瘦素,NK细胞和免疫的重量:对肥胖的见解。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 DOI: 10.1016/j.intimp.2024.113992
Arthur Gomes de Andrade , Shayenne Eduarda Ramos Vanderley , Lorrane de Farias Marques , Fernanda Silva Almeida , Luiz Henrique Agra Cavalcante-Silva , Tatjana Souza Lima Keesen
Obesity is a chronic inflammatory disease that affects more than 1 billion people worldwide and is associated with various metabolic and physiological dysfunctions, directly impacting the dynamics of the immune response, partly due to elevated leptin levels. Leptin is an important peptide hormone that regulates neuroendocrine function and energy homeostasis, with its blood levels reflecting energy reserves, fat mass, or energy deprivation. This hormone also plays a fundamental role in regulating immune function, including the activity of NK cells, which are essential components in antiviral and antitumor activity. In obese individuals, leptin resistance is commonly established, however, NK cells and other immune components remain responsive to this hormone. So far, leptin has demonstrated paradoxical activities of these cells, often associated with a dysfunctional profile when associated with obesity. The excessive fat is usually related to metabolic remodeling in NK cells, resulting in compromised antitumor responses due to reduced cytotoxic capacity and decreased expression of cytokines important for these defense mechanisms, such as IFN-γ. Therefore, this review approaches a better understanding of the immunoendocrine interactions between leptin and NK cells in the context of obesity.
肥胖是一种慢性炎症性疾病,影响全球超过10亿人,与各种代谢和生理功能障碍有关,直接影响免疫反应的动态,部分原因是瘦素水平升高。瘦素是一种重要的肽激素,调节神经内分泌功能和能量稳态,其血液水平反映能量储备、脂肪量或能量剥夺。这种激素在调节免疫功能,包括NK细胞的活性方面也起着重要作用,NK细胞是抗病毒和抗肿瘤活性的重要组成部分。在肥胖个体中,瘦素抵抗是普遍存在的,然而,NK细胞和其他免疫成分仍然对这种激素有反应。到目前为止,瘦素已经证明了这些细胞的矛盾活动,当与肥胖有关时,往往与功能失调有关。过多的脂肪通常与NK细胞的代谢重塑有关,由于细胞毒能力降低和对这些防御机制重要的细胞因子(如IFN-γ)表达减少,导致抗肿瘤反应受损。因此,本综述旨在更好地了解肥胖背景下瘦素和NK细胞之间的免疫内分泌相互作用。
{"title":"Leptin, NK cells, and the weight of immunity: Insights into obesity","authors":"Arthur Gomes de Andrade ,&nbsp;Shayenne Eduarda Ramos Vanderley ,&nbsp;Lorrane de Farias Marques ,&nbsp;Fernanda Silva Almeida ,&nbsp;Luiz Henrique Agra Cavalcante-Silva ,&nbsp;Tatjana Souza Lima Keesen","doi":"10.1016/j.intimp.2024.113992","DOIUrl":"10.1016/j.intimp.2024.113992","url":null,"abstract":"<div><div>Obesity is a chronic inflammatory disease that affects more than 1 billion people worldwide and is associated with various metabolic and physiological dysfunctions, directly impacting the dynamics of the immune response, partly due to elevated leptin levels. Leptin is an important peptide hormone that regulates neuroendocrine function and energy homeostasis, with its blood levels reflecting energy reserves, fat mass, or energy deprivation. This hormone also plays a fundamental role in regulating immune function, including the activity of NK cells, which are essential components in antiviral and antitumor activity. In obese individuals, leptin resistance is commonly established, however, NK cells and other immune components remain responsive to this hormone. So far, leptin has demonstrated paradoxical activities of these cells, often associated with a dysfunctional profile when associated with obesity. The excessive fat is usually related to metabolic remodeling in NK cells, resulting in compromised antitumor responses due to reduced cytotoxic capacity and decreased expression of cytokines important for these defense mechanisms, such as IFN-γ. Therefore, this review approaches a better understanding of the immunoendocrine interactions between leptin and NK cells in the context of obesity.</div></div>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"Article 113992"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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 tryptophan metabolite kynurenic acid ameliorates septic colonic injury through activation of the PPARγ signaling pathway. 色氨酸代谢物犬尿酸通过激活PPARγ信号通路改善败血性结肠损伤。
IF 4.8 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-06 Epub Date: 2024-12-31 DOI: 10.1016/j.intimp.2024.113651
Fei Wang, Meng Zhang, Liping Yin, Ziyang Zhou, Ziyao Peng, Wenweiran Li, Hui Chen, Guohong Yu, Jianguo Tang

Sepsis is the leading cause of death among critically ill patients in clinical practice, making it urgent to reduce its incidence and mortality rates. In sepsis, macrophage dysfunction often worsens and complicates the condition. M1 and M2 macrophages, two distinct types, contribute to pro-inflammatory and anti-inflammatory effects, respectively. An imbalance between them is a major cause of sepsis. The aim of this study was to explore the potential of a differential metabolite between M1 and M2 macrophages in mitigating septic colonic injury via multiomics in combination with clinical data and animal experiments. Using nontargeted metabolomics analysis, we found that Kynurenic acid (KYNA), a metabolite of tryptophan metabolism, was significantly upregulated in the supernatant of M2 macrophages. Furthermore, we discovered that the level of KYNA was significantly decreased in sepsis in both human and mouse serum and was negatively correlated with inflammatory factor levels. In vivo experiments demonstrated that KYNA can effectively alleviate septic colon injury and reduce inflammatory factor levels in mice, indicating that KYNA plays a very important protective role in sepsis. Mechanistically, KYNA promotes the transition of M1 macrophages to M2 macrophages by inhibiting the NF-κB signaling pathway and alleviates septic colonic injury through the PPARγ/NF-κB axis. This article reveals that KYNA, a differentially abundant metabolite between M1 and M2 macrophages, can become a new strategy for alleviating septic colon injury.

脓毒症是临床上危重症患者死亡的主要原因,降低脓毒症的发病率和死亡率迫在眉睫。在败血症中,巨噬细胞功能障碍经常恶化并使病情复杂化。M1和M2巨噬细胞是两种不同的类型,分别具有促炎和抗炎作用。两者之间的失衡是导致败血症的主要原因。本研究的目的是通过多组学方法,结合临床数据和动物实验,探索M1和M2巨噬细胞之间的差异代谢物在减轻败血性结肠损伤中的潜力。通过非靶向代谢组学分析,我们发现色氨酸代谢的代谢物KYNA在M2巨噬细胞的上清液中显著上调。此外,我们发现在脓毒症中,人和小鼠血清中KYNA水平显著降低,并与炎症因子水平呈负相关。体内实验表明,KYNA能有效减轻小鼠脓毒症结肠损伤,降低炎症因子水平,提示KYNA在脓毒症中具有非常重要的保护作用。机制上,KYNA通过抑制NF-κB信号通路促进M1型巨噬细胞向M2型巨噬细胞转变,并通过PPARγ/NF-κB轴减轻败血性结肠损伤。本文揭示了巨噬细胞M1和M2之间差异丰富的代谢物KYNA可能成为缓解脓毒性结肠损伤的新策略。
{"title":"The tryptophan metabolite kynurenic acid ameliorates septic colonic injury through activation of the PPARγ signaling pathway.","authors":"Fei Wang, Meng Zhang, Liping Yin, Ziyang Zhou, Ziyao Peng, Wenweiran Li, Hui Chen, Guohong Yu, Jianguo Tang","doi":"10.1016/j.intimp.2024.113651","DOIUrl":"10.1016/j.intimp.2024.113651","url":null,"abstract":"<p><p>Sepsis is the leading cause of death among critically ill patients in clinical practice, making it urgent to reduce its incidence and mortality rates. In sepsis, macrophage dysfunction often worsens and complicates the condition. M1 and M2 macrophages, two distinct types, contribute to pro-inflammatory and anti-inflammatory effects, respectively. An imbalance between them is a major cause of sepsis. The aim of this study was to explore the potential of a differential metabolite between M1 and M2 macrophages in mitigating septic colonic injury via multiomics in combination with clinical data and animal experiments. Using nontargeted metabolomics analysis, we found that Kynurenic acid (KYNA), a metabolite of tryptophan metabolism, was significantly upregulated in the supernatant of M2 macrophages. Furthermore, we discovered that the level of KYNA was significantly decreased in sepsis in both human and mouse serum and was negatively correlated with inflammatory factor levels. In vivo experiments demonstrated that KYNA can effectively alleviate septic colon injury and reduce inflammatory factor levels in mice, indicating that KYNA plays a very important protective role in sepsis. Mechanistically, KYNA promotes the transition of M1 macrophages to M2 macrophages by inhibiting the NF-κB signaling pathway and alleviates septic colonic injury through the PPARγ/NF-κB axis. This article reveals that KYNA, a differentially abundant metabolite between M1 and M2 macrophages, can become a new strategy for alleviating septic colon injury.</p>","PeriodicalId":13859,"journal":{"name":"International immunopharmacology","volume":"147 ","pages":"113651"},"PeriodicalIF":4.8,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142914564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
International immunopharmacology
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