巨噬细胞迁移抑制因子促进血栓性炎症并预测主动脉狭窄的快速进展

IF 7.4 1区 医学 Q1 HEMATOLOGY Arteriosclerosis, Thrombosis, and Vascular Biology Pub Date : 2024-09-01 Epub Date: 2024-07-11 DOI:10.1161/ATVBAHA.124.321000
Karin Anne Lydia Mueller, Carolin Langnau, Tobias Harm, Manuel Sigle, Kristina Mott, Michal Droppa, Oliver Borst, Anne-Katrin Rohlfing, Sarah Gekeler, Manina Günter, Nora Goebel, Ulrich F W Franke, Medhat Radwan, Christian Schlensak, Henrik Janning, Sophia Scheuermann, Christian M Seitz, Dominik Rath, Klaus-Peter Kreisselmeier, Tatsiana Castor, Iris Irmgard Mueller, Harald Schulze, Stella E Autenrieth, Meinrad Paul Gawaz
{"title":"巨噬细胞迁移抑制因子促进血栓性炎症并预测主动脉狭窄的快速进展","authors":"Karin Anne Lydia Mueller, Carolin Langnau, Tobias Harm, Manuel Sigle, Kristina Mott, Michal Droppa, Oliver Borst, Anne-Katrin Rohlfing, Sarah Gekeler, Manina Günter, Nora Goebel, Ulrich F W Franke, Medhat Radwan, Christian Schlensak, Henrik Janning, Sophia Scheuermann, Christian M Seitz, Dominik Rath, Klaus-Peter Kreisselmeier, Tatsiana Castor, Iris Irmgard Mueller, Harald Schulze, Stella E Autenrieth, Meinrad Paul Gawaz","doi":"10.1161/ATVBAHA.124.321000","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Aortic stenosis (AS) is driven by progressive inflammatory and fibrocalcific processes regulated by circulating inflammatory and valve resident endothelial and interstitial cells. The impact of platelets, platelet-derived mediators, and platelet-monocyte interactions on the acceleration of local valvular inflammation and mineralization is presently unknown.</p><p><strong>Methods: </strong>We prospectively enrolled 475 consecutive patients with severe symptomatic AS undergoing aortic valve replacement. Clinical workup included repetitive echocardiography, analysis of platelets, monocytes, chemokine profiling, aortic valve tissue samples for immunohistochemistry, and gene expression analysis.</p><p><strong>Results: </strong>The patients were classified as fast-progressive AS by the median ∆Vmax of 0.45 m/s per year determined by echocardiography. Immunohistological aortic valve analysis revealed enhanced cellularity in fast-progressive AS (slow- versus fast-progressive AS; median [interquartile range], 247 [142.3-504] versus 717.5 [360.5-1234]; <i>P</i><0.001) with less calcification (calcification area, mm<sup>2</sup>: 33.74 [27.82-41.86] versus 20.54 [13.52-33.41]; <i>P</i><0.001). MIF (macrophage migration inhibitory factor)-associated gene expression was significantly enhanced in fast-progressive AS accompanied by significantly elevated MIF plasma levels (mean±SEM; 6877±379.1 versus 9959±749.1; <i>P</i><0.001), increased platelet activation, and decreased intracellular MIF expression indicating enhanced MIF release upon platelet activation (CD62P, %: median [interquartile range], 16.8 [11.58-23.8] versus 20.55 [12.48-32.28], <i>P</i>=0.005; MIF, %: 4.85 [1.48-9.75] versus 2.3 [0.78-5.9], <i>P</i><0.001). Regression analysis confirmed that MIF-associated biomarkers are strongly associated with an accelerated course of AS.</p><p><strong>Conclusions: </strong>Our findings suggest a key role for platelet-derived MIF and its interplay with circulating and valve resident monocytes/macrophages in local and systemic thromboinflammation during accelerated AS. MIF-based biomarkers predict an accelerated course of AS and represent a novel pharmacological target to attenuate progression of AS.</p>","PeriodicalId":8401,"journal":{"name":"Arteriosclerosis, Thrombosis, and Vascular Biology","volume":null,"pages":null},"PeriodicalIF":7.4000,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11335082/pdf/","citationCount":"0","resultStr":"{\"title\":\"Macrophage Migration Inhibitory Factor Promotes Thromboinflammation and Predicts Fast Progression of Aortic Stenosis.\",\"authors\":\"Karin Anne Lydia Mueller, Carolin Langnau, Tobias Harm, Manuel Sigle, Kristina Mott, Michal Droppa, Oliver Borst, Anne-Katrin Rohlfing, Sarah Gekeler, Manina Günter, Nora Goebel, Ulrich F W Franke, Medhat Radwan, Christian Schlensak, Henrik Janning, Sophia Scheuermann, Christian M Seitz, Dominik Rath, Klaus-Peter Kreisselmeier, Tatsiana Castor, Iris Irmgard Mueller, Harald Schulze, Stella E Autenrieth, Meinrad Paul Gawaz\",\"doi\":\"10.1161/ATVBAHA.124.321000\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Aortic stenosis (AS) is driven by progressive inflammatory and fibrocalcific processes regulated by circulating inflammatory and valve resident endothelial and interstitial cells. The impact of platelets, platelet-derived mediators, and platelet-monocyte interactions on the acceleration of local valvular inflammation and mineralization is presently unknown.</p><p><strong>Methods: </strong>We prospectively enrolled 475 consecutive patients with severe symptomatic AS undergoing aortic valve replacement. Clinical workup included repetitive echocardiography, analysis of platelets, monocytes, chemokine profiling, aortic valve tissue samples for immunohistochemistry, and gene expression analysis.</p><p><strong>Results: </strong>The patients were classified as fast-progressive AS by the median ∆Vmax of 0.45 m/s per year determined by echocardiography. Immunohistological aortic valve analysis revealed enhanced cellularity in fast-progressive AS (slow- versus fast-progressive AS; median [interquartile range], 247 [142.3-504] versus 717.5 [360.5-1234]; <i>P</i><0.001) with less calcification (calcification area, mm<sup>2</sup>: 33.74 [27.82-41.86] versus 20.54 [13.52-33.41]; <i>P</i><0.001). MIF (macrophage migration inhibitory factor)-associated gene expression was significantly enhanced in fast-progressive AS accompanied by significantly elevated MIF plasma levels (mean±SEM; 6877±379.1 versus 9959±749.1; <i>P</i><0.001), increased platelet activation, and decreased intracellular MIF expression indicating enhanced MIF release upon platelet activation (CD62P, %: median [interquartile range], 16.8 [11.58-23.8] versus 20.55 [12.48-32.28], <i>P</i>=0.005; MIF, %: 4.85 [1.48-9.75] versus 2.3 [0.78-5.9], <i>P</i><0.001). Regression analysis confirmed that MIF-associated biomarkers are strongly associated with an accelerated course of AS.</p><p><strong>Conclusions: </strong>Our findings suggest a key role for platelet-derived MIF and its interplay with circulating and valve resident monocytes/macrophages in local and systemic thromboinflammation during accelerated AS. MIF-based biomarkers predict an accelerated course of AS and represent a novel pharmacological target to attenuate progression of AS.</p>\",\"PeriodicalId\":8401,\"journal\":{\"name\":\"Arteriosclerosis, Thrombosis, and Vascular Biology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":7.4000,\"publicationDate\":\"2024-09-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11335082/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Arteriosclerosis, Thrombosis, and Vascular Biology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1161/ATVBAHA.124.321000\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/7/11 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q1\",\"JCRName\":\"HEMATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Arteriosclerosis, Thrombosis, and Vascular Biology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1161/ATVBAHA.124.321000","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/7/11 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:主动脉瓣狭窄(AS)是由循环炎症细胞和瓣膜内皮细胞及间质细胞调节的渐进性炎症和纤维钙化过程驱动的。目前尚不清楚血小板、血小板衍生介质以及血小板-单核细胞相互作用对加速局部瓣膜炎症和矿化的影响:我们对 475 例接受主动脉瓣置换术的严重症状性 AS 患者进行了前瞻性登记。临床检查包括重复超声心动图检查、血小板和单核细胞分析、趋化因子分析、主动脉瓣组织样本免疫组化和基因表达分析:根据超声心动图测定的中位ΔVmax(每年0.45米/秒),患者被归类为快速进展型强直性脊柱炎。免疫组织学主动脉瓣分析显示,快速进展型强直性脊柱炎的细胞性增强(慢进展型强直性脊柱炎与快速进展型强直性脊柱炎的中位数[四分位间范围],247 [142.3-504]对717.5[360.5-1234];P2:33.74[27.82-41.86]对20.54[13.52-33.41];PPP=0.005;MIF,%:4.85[1.48-9.75]对2.3[0.78-5.9],PConclusions:我们的研究结果表明,血小板衍生的 MIF 及其与循环和瓣膜驻留单核细胞/巨噬细胞的相互作用在加速 AS 期间的局部和全身血栓性炎症中起着关键作用。以 MIF 为基础的生物标志物可预测强直性脊柱炎的加速过程,并代表了一种可减轻强直性脊柱炎进展的新型药物靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Macrophage Migration Inhibitory Factor Promotes Thromboinflammation and Predicts Fast Progression of Aortic Stenosis.

Background: Aortic stenosis (AS) is driven by progressive inflammatory and fibrocalcific processes regulated by circulating inflammatory and valve resident endothelial and interstitial cells. The impact of platelets, platelet-derived mediators, and platelet-monocyte interactions on the acceleration of local valvular inflammation and mineralization is presently unknown.

Methods: We prospectively enrolled 475 consecutive patients with severe symptomatic AS undergoing aortic valve replacement. Clinical workup included repetitive echocardiography, analysis of platelets, monocytes, chemokine profiling, aortic valve tissue samples for immunohistochemistry, and gene expression analysis.

Results: The patients were classified as fast-progressive AS by the median ∆Vmax of 0.45 m/s per year determined by echocardiography. Immunohistological aortic valve analysis revealed enhanced cellularity in fast-progressive AS (slow- versus fast-progressive AS; median [interquartile range], 247 [142.3-504] versus 717.5 [360.5-1234]; P<0.001) with less calcification (calcification area, mm2: 33.74 [27.82-41.86] versus 20.54 [13.52-33.41]; P<0.001). MIF (macrophage migration inhibitory factor)-associated gene expression was significantly enhanced in fast-progressive AS accompanied by significantly elevated MIF plasma levels (mean±SEM; 6877±379.1 versus 9959±749.1; P<0.001), increased platelet activation, and decreased intracellular MIF expression indicating enhanced MIF release upon platelet activation (CD62P, %: median [interquartile range], 16.8 [11.58-23.8] versus 20.55 [12.48-32.28], P=0.005; MIF, %: 4.85 [1.48-9.75] versus 2.3 [0.78-5.9], P<0.001). Regression analysis confirmed that MIF-associated biomarkers are strongly associated with an accelerated course of AS.

Conclusions: Our findings suggest a key role for platelet-derived MIF and its interplay with circulating and valve resident monocytes/macrophages in local and systemic thromboinflammation during accelerated AS. MIF-based biomarkers predict an accelerated course of AS and represent a novel pharmacological target to attenuate progression of AS.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
15.60
自引率
2.30%
发文量
337
审稿时长
2-4 weeks
期刊介绍: The journal "Arteriosclerosis, Thrombosis, and Vascular Biology" (ATVB) is a scientific publication that focuses on the fields of vascular biology, atherosclerosis, and thrombosis. It is a peer-reviewed journal that publishes original research articles, reviews, and other scholarly content related to these areas. The journal is published by the American Heart Association (AHA) and the American Stroke Association (ASA). The journal was published bi-monthly until January 1992, after which it transitioned to a monthly publication schedule. The journal is aimed at a professional audience, including academic cardiologists, vascular biologists, physiologists, pharmacologists and hematologists.
期刊最新文献
Associations Between Flavonoid Intake and Subclinical Atherosclerosis: The MESA. Ethnic Diversity and Distinctive Features of Familial Versus Multifactorial Chylomicronemia Syndrome: Insights From the UK FCS National Registry. Spatial Transcriptomic Approach to Understanding Coronary Atherosclerotic Plaque Stability. Institutional Support for the Career Advancement of Women Faculty in Science and Academic Medicine: Successes, Challenges, and Future Directions. IL-1β Induces LDL Transcytosis by a Novel Pathway Involving LDLR and Rab27a.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
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
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
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