首页 > 最新文献

Circulation research最新文献

英文 中文
Myeloid-Specific JAK2 Contributes to Inflammation and Salt Sensitivity of Blood Pressure. 髓系特异性 JAK2 对炎症和血压的盐敏感性有贡献
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-12 DOI: 10.1161/circresaha.124.323595
Mohammad Saleem,Luul A Aden,Ashley Pitzer Mutchler,Chitra Basu,Lale A Ertuglu,Quanhu Sheng,Niki Penner,Anna R Hemnes,Jennifer H Park,Jeanne A Ishimwe,Cheryl L Laffer,Fernando Elijovich,Celestine N Wanjalla,Nestor de la Visitacion,Paul D Kastner,Claude F Albritton,Taseer Ahmad,Alexandria P Haynes,Justin Yu,Meghan K Graber,Sharia Yasmin,Kay-Uwe Wagner,Peter P Sayeski,Antonis K Hatzopoulos,Eric R Gamazon,Alexander G Bick,Thomas R Kleyman,Annet Kirabo
BACKGROUNDSalt sensitivity of blood pressure (SSBP), characterized by acute changes in blood pressure with changes in dietary sodium intake, is an independent risk factor for cardiovascular disease and mortality in people with and without hypertension. We previously found that elevated sodium concentration activates antigen-presenting cells (APCs), resulting in high blood pressure, but the mechanisms are unknown. Here, we hypothesized that APC-specific JAK2 (Janus kinase 2) through STAT3 (signal transducer and activator of transcription 3) and SMAD3 (small mothers against decapentaplegic homolog 3) contributes to SSBP.METHODWe performed bulk or single-cell transcriptomic analyses following in vitro monocytes exposed to high salt and in vivo high sodium treatment in humans using a rigorous salt-loading/depletion protocol to phenotype SSBP. We also used a myeloid cell-specific CD11c+ JAK2 knockout mouse model and measured blood pressure with radiotelemetry after N-omega-nitro-L-arginine-methyl ester and a high salt diet treatment. We used flow cytometry for immunophenotyping and measuring cytokine levels. Fluorescence in situ hybridization and immunohistochemistry were performed to spatially visualize the kidney's immune cells and cytokine levels. Echocardiography was performed to assess cardiac function.RESULTSWe found that high salt treatment upregulates gene expression of the JAK/STAT/SMAD pathway while downregulating inhibitors of this pathway, such as suppression of cytokine signaling and cytokine-inducible SH2, in human monocytes. Expression of the JAK2 pathway genes mirrored changes in blood pressure after salt loading and depletion in salt-sensitive but not salt-resistant humans. Ablation of JAK2, specifically in CD11c+ APCs, attenuated salt-induced hypertension in mice with SSBP. Mechanistically, we found that SMAD3 acted downstream of JAK2 and STAT3, leading to increased production of highly reactive isolevuglandins and proinflammatory cytokine IL (interleukin)-6 in renal APCs, which activate T cells and increase production of IL-17A, IL-6, and TNF-α (tumor necrosis factor-alpha).CONCLUSIONSOur findings reveal the APC JAK2 signaling pathway as a potential target for the diagnosis and treatment of SSBP in humans.
背景盐敏感性血压(SSBP)的特点是血压随膳食钠摄入量的变化而发生急性变化,它是高血压患者和非高血压患者心血管疾病和死亡的独立危险因素。我们之前发现,钠浓度升高会激活抗原递呈细胞(APCs),从而导致高血压,但其机制尚不清楚。在此,我们假设抗原递呈细胞特异性 JAK2(Janus 激酶 2)通过 STAT3(信号转导和转录激活因子 3)和 SMAD3(抗截瘫小母细胞同源物 3)对 SSBP 起作用。我们还使用了髓细胞特异性 CD11c+ JAK2 基因敲除小鼠模型,并在 N-欧米伽-硝基-L-精氨酸甲酯和高盐饮食处理后使用放射性遥测测量血压。我们使用流式细胞术进行免疫分型并测量细胞因子水平。荧光原位杂交和免疫组化技术可在空间上观察肾脏的免疫细胞和细胞因子水平。结果我们发现,高盐处理会上调人单核细胞中 JAK/STAT/SMAD 通路的基因表达,同时下调该通路的抑制因子,如抑制细胞因子信号转导和细胞因子诱导的 SH2。JAK2 通路基因的表达反映了对盐敏感而不耐盐的人在盐负荷和盐耗尽后血压的变化。JAK2的消减,特别是在CD11c+ APCs中的消减,减轻了盐诱导的SSBP小鼠高血压。从机理上讲,我们发现 SMAD3 作用于 JAK2 和 STAT3 的下游,导致肾脏 APC 中高反应性异芦荟素和促炎细胞因子 IL(白细胞介素)-6 的产生增加,从而激活 T 细胞并增加 IL-17A、IL-6 和 TNF-α(肿瘤坏死因子-α)的产生。
{"title":"Myeloid-Specific JAK2 Contributes to Inflammation and Salt Sensitivity of Blood Pressure.","authors":"Mohammad Saleem,Luul A Aden,Ashley Pitzer Mutchler,Chitra Basu,Lale A Ertuglu,Quanhu Sheng,Niki Penner,Anna R Hemnes,Jennifer H Park,Jeanne A Ishimwe,Cheryl L Laffer,Fernando Elijovich,Celestine N Wanjalla,Nestor de la Visitacion,Paul D Kastner,Claude F Albritton,Taseer Ahmad,Alexandria P Haynes,Justin Yu,Meghan K Graber,Sharia Yasmin,Kay-Uwe Wagner,Peter P Sayeski,Antonis K Hatzopoulos,Eric R Gamazon,Alexander G Bick,Thomas R Kleyman,Annet Kirabo","doi":"10.1161/circresaha.124.323595","DOIUrl":"https://doi.org/10.1161/circresaha.124.323595","url":null,"abstract":"BACKGROUNDSalt sensitivity of blood pressure (SSBP), characterized by acute changes in blood pressure with changes in dietary sodium intake, is an independent risk factor for cardiovascular disease and mortality in people with and without hypertension. We previously found that elevated sodium concentration activates antigen-presenting cells (APCs), resulting in high blood pressure, but the mechanisms are unknown. Here, we hypothesized that APC-specific JAK2 (Janus kinase 2) through STAT3 (signal transducer and activator of transcription 3) and SMAD3 (small mothers against decapentaplegic homolog 3) contributes to SSBP.METHODWe performed bulk or single-cell transcriptomic analyses following in vitro monocytes exposed to high salt and in vivo high sodium treatment in humans using a rigorous salt-loading/depletion protocol to phenotype SSBP. We also used a myeloid cell-specific CD11c+ JAK2 knockout mouse model and measured blood pressure with radiotelemetry after N-omega-nitro-L-arginine-methyl ester and a high salt diet treatment. We used flow cytometry for immunophenotyping and measuring cytokine levels. Fluorescence in situ hybridization and immunohistochemistry were performed to spatially visualize the kidney's immune cells and cytokine levels. Echocardiography was performed to assess cardiac function.RESULTSWe found that high salt treatment upregulates gene expression of the JAK/STAT/SMAD pathway while downregulating inhibitors of this pathway, such as suppression of cytokine signaling and cytokine-inducible SH2, in human monocytes. Expression of the JAK2 pathway genes mirrored changes in blood pressure after salt loading and depletion in salt-sensitive but not salt-resistant humans. Ablation of JAK2, specifically in CD11c+ APCs, attenuated salt-induced hypertension in mice with SSBP. Mechanistically, we found that SMAD3 acted downstream of JAK2 and STAT3, leading to increased production of highly reactive isolevuglandins and proinflammatory cytokine IL (interleukin)-6 in renal APCs, which activate T cells and increase production of IL-17A, IL-6, and TNF-α (tumor necrosis factor-alpha).CONCLUSIONSOur findings reveal the APC JAK2 signaling pathway as a potential target for the diagnosis and treatment of SSBP in humans.","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142174627","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
On the Mend: Atrial Tubulogenesis After Tachypacing-Induced Heart Failure. 亡羊补牢:快速起搏诱发心力衰竭后的心房管生成。
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-12 DOI: 10.1161/circresaha.124.325211
Michelle L Munro,Luis A Gonano
{"title":"On the Mend: Atrial Tubulogenesis After Tachypacing-Induced Heart Failure.","authors":"Michelle L Munro,Luis A Gonano","doi":"10.1161/circresaha.124.325211","DOIUrl":"https://doi.org/10.1161/circresaha.124.325211","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142231286","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Cyclophilin(g) a Knowledge Gap in Heart Failure Pathogenesis. 心力衰竭发病机制中的一个知识空白点--环纤蛋白(g)
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-12 DOI: 10.1161/circresaha.124.325132
Marion Delaunay,Zegeye H Jebessa,Timothy A McKinsey
{"title":"Cyclophilin(g) a Knowledge Gap in Heart Failure Pathogenesis.","authors":"Marion Delaunay,Zegeye H Jebessa,Timothy A McKinsey","doi":"10.1161/circresaha.124.325132","DOIUrl":"https://doi.org/10.1161/circresaha.124.325132","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142231283","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Patching-Up Post-MI Hearts With Microparticle-Delivered Apelin. 用微粒递送的 Apelin 修补心肌梗死后的心脏
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-12 DOI: 10.1161/circresaha.124.325194
Vladislav Leonov,Timothy J Kamp
{"title":"Patching-Up Post-MI Hearts With Microparticle-Delivered Apelin.","authors":"Vladislav Leonov,Timothy J Kamp","doi":"10.1161/circresaha.124.325194","DOIUrl":"https://doi.org/10.1161/circresaha.124.325194","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142231279","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Experimental TET2 Clonal Hematopoiesis Predisposes to Renal Hypertension Through an Inflammasome-Mediated Mechanism. 实验性 TET2 克隆造血通过炎症体介导的机制易导致肾性高血压
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-05 DOI: 10.1161/CIRCRESAHA.124.324492
Ariel H Polizio, Lucila Marino, Kyung-Duk Min, Yoshimitsu Yura, Luca Rolauer, Jesse D Cochran, Megan A Evans, Eunbee Park, Heather Doviak, Emiri Miura-Yura, Miranda E Good, Abigail G Wolpe, Maria Grandoch, Brant Isakson, Kenneth Walsh

Background: Hypertension incidence increases with age and represents one of the most prevalent risk factors for cardiovascular disease. Clonal events in the hematopoietic system resulting from somatic mutations in driver genes are prevalent in elderly individuals who lack overt hematologic disorders. This condition is referred to as age-related clonal hematopoiesis (CH), and it is a newly recognized risk factor for cardiovascular disease. It is not known whether CH and hypertension in the elderly are causally related and, if so, what are the mechanistic features.

Methods and results: A murine model of adoptive bone marrow transplantation was employed to examine the interplay between Tet2 (ten-eleven translocation methylcytosine dioxygenase 2) CH and hypertension. In this model, a subpressor dose of Ang II (angiotensin II) resulted in elevated systolic and diastolic blood pressure as early as 1 day after the challenge. These conditions led to the expansion of Tet2-deficient proinflammatory monocytes and bone marrow progenitor populations. Tet2-deficiency promoted renal CCL5 chemokine expression and macrophage infiltration into the kidney. Consistent with macrophage involvement, Tet2-deficiency in myeloid cells promoted hypertension when mice were treated with a subpressor dose of Ang II. The hematopoietic Tet2-/- condition led to sodium retention, renal inflammasome activation, and elevated levels of IL (interleukin)-1β and IL-18. Analysis of the sodium transporters indicated NCC (Na+-Cl- cotransporter) and NKCC2 activation at residues Thr53 and Ser105, respectively. Administration of the NLRP3 inflammasome inhibitor MCC950 reversed the hypertensive state, sodium retention, and renal transporter activation.

Conclusions: Tet2-mediated CH sensitizes mice to a hypertensive stimulus. Mechanistically, the expansion of hematopoietic Tet2-deficient cells promotes hypertension due to elevated renal immune cell infiltration and activation of the NLRP3 inflammasome, with consequences on sodium retention. These data indicate that carriers of TET2 CH could be at elevated risk for the development of hypertension and that immune modulators could be useful in treating hypertension in this patient population.

背景:高血压发病率随年龄增长而增加,是心血管疾病最普遍的危险因素之一。在没有明显血液病的老年人中,由于驱动基因的体细胞突变而导致的造血系统克隆事件非常普遍。这种情况被称为与年龄相关的克隆性造血(CH),是一种新近被确认的心血管疾病风险因素。目前尚不清楚老年克隆性造血和高血压是否有因果关系,如果有,其机理特征是什么:方法和结果:我们采用了一种小鼠收养性骨髓移植模型来研究 Tet2(十-十一易位甲基胞嘧啶二氧酶 2)CH 与高血压之间的相互作用。在该模型中,亚压剂量的 Ang II(血管紧张素 II)导致收缩压和舒张压升高,最早出现在挑战后 1 天。这些条件导致 Tet2 缺陷的促炎单核细胞和骨髓祖细胞扩增。Tet2缺陷促进了肾脏CCL5趋化因子的表达和巨噬细胞向肾脏的浸润。与巨噬细胞的参与相一致,当小鼠接受亚抑制剂量的 Ang II 治疗时,骨髓细胞中的 Tet2 缺失会促进高血压。造血Tet2-/-条件导致钠潴留、肾脏炎症小体激活以及IL(白细胞介素)-1β和IL-18水平升高。对钠转运体的分析表明,NCC(Na+-Cl- 共转运体)和 NKCC2 分别在 Thr53 和 Ser105 残基处被激活。服用 NLRP3 炎性体抑制剂 MCC950 逆转了高血压状态、钠潴留和肾转运体激活:结论:Tet2 介导的 CH 使小鼠对高血压刺激敏感。从机理上讲,造血Tet2缺陷细胞的扩增会促进肾脏免疫细胞浸润和NLRP3炎性体的激活,从而导致高血压,并对钠潴留产生影响。这些数据表明,TET2 CH携带者罹患高血压的风险可能会升高,免疫调节剂可能有助于治疗这类患者的高血压。
{"title":"Experimental TET2 Clonal Hematopoiesis Predisposes to Renal Hypertension Through an Inflammasome-Mediated Mechanism.","authors":"Ariel H Polizio, Lucila Marino, Kyung-Duk Min, Yoshimitsu Yura, Luca Rolauer, Jesse D Cochran, Megan A Evans, Eunbee Park, Heather Doviak, Emiri Miura-Yura, Miranda E Good, Abigail G Wolpe, Maria Grandoch, Brant Isakson, Kenneth Walsh","doi":"10.1161/CIRCRESAHA.124.324492","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.124.324492","url":null,"abstract":"<p><strong>Background: </strong>Hypertension incidence increases with age and represents one of the most prevalent risk factors for cardiovascular disease. Clonal events in the hematopoietic system resulting from somatic mutations in driver genes are prevalent in elderly individuals who lack overt hematologic disorders. This condition is referred to as age-related clonal hematopoiesis (CH), and it is a newly recognized risk factor for cardiovascular disease. It is not known whether CH and hypertension in the elderly are causally related and, if so, what are the mechanistic features.</p><p><strong>Methods and results: </strong>A murine model of adoptive bone marrow transplantation was employed to examine the interplay between Tet2 (ten-eleven translocation methylcytosine dioxygenase 2) CH and hypertension. In this model, a subpressor dose of Ang II (angiotensin II) resulted in elevated systolic and diastolic blood pressure as early as 1 day after the challenge. These conditions led to the expansion of Tet2-deficient proinflammatory monocytes and bone marrow progenitor populations. Tet2-deficiency promoted renal CCL5 chemokine expression and macrophage infiltration into the kidney. Consistent with macrophage involvement, Tet2-deficiency in myeloid cells promoted hypertension when mice were treated with a subpressor dose of Ang II. The hematopoietic Tet2<sup>-</sup><sup>/-</sup> condition led to sodium retention, renal inflammasome activation, and elevated levels of IL (interleukin)-1β and IL-18. Analysis of the sodium transporters indicated NCC (Na<sup>+</sup>-Cl<sup>-</sup> cotransporter) and NKCC2 activation at residues Thr53 and Ser105, respectively. Administration of the NLRP3 inflammasome inhibitor MCC950 reversed the hypertensive state, sodium retention, and renal transporter activation.</p><p><strong>Conclusions: </strong>Tet2-mediated CH sensitizes mice to a hypertensive stimulus. Mechanistically, the expansion of hematopoietic Tet2-deficient cells promotes hypertension due to elevated renal immune cell infiltration and activation of the NLRP3 inflammasome, with consequences on sodium retention. These data indicate that carriers of TET2 CH could be at elevated risk for the development of hypertension and that immune modulators could be useful in treating hypertension in this patient population.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142132008","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Macrophage-Expressed Coagulation Factor 7 Promotes Adverse Cardiac Remodeling. 巨噬细胞表达的凝血因子 7 促进不良心脏重塑
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-05 DOI: 10.1161/CIRCRESAHA.123.324114
Venkata Garlapati, Qi Luo, Jens Posma, Melania Aluia, Than Son Nguyen, Kristin Grunz, Michael Molitor, Stefanie Finger, Gregory Harms, Tobias Bopp, Wolfram Ruf, Philip Wenzel

Background: Excess fibrotic remodeling causes cardiac dysfunction in ischemic heart disease, driven by MAP (mitogen-activated protein) kinase-dependent TGF-ß1 (transforming growth factor-ß1) activation by coagulation signaling of myeloid cells. How coagulation-inflammatory circuits can be specifically targeted to achieve beneficial macrophage reprogramming after myocardial infarction (MI) is not completely understood.

Methods: Mice with permanent ligation of the left anterior descending artery were used to model nonreperfused MI and analyzed by single-cell RNA sequencing, protein expression changes, confocal microscopy, and longitudinal monitoring of recovery. We probed the role of the tissue factor (TF)-factor 7 (F7)-integrin ß1-PAR2 (protease-activated receptor 2) signaling complex by utilizing genetic mouse models and pharmacological intervention.

Results: Cleavage-insensitive PAR2R38E and myeloid cell integrin ß1-deficient mice had improved cardiac function after MI compared with controls. Proximity ligation assays of monocytic cells demonstrated that colocalization of F7 with integrin ß1 was diminished in monocyte/macrophage F7-deficient mice after MI. Compared with controls, F7fl/fl CX3CR1Cre mice showed reduced TGF-ß1 and MAP kinase activation, as well as cardiac dysfunction after MI, despite unaltered overall recruitment of myeloid cells. Single-cell mRNA sequencing of CD45 (cluster of differentiation 45)+ cells 3 and 7 days after MI uncovered a trajectory from recruited monocytes to inflammatory TF+/F7+/TREM (triggered receptor expressed on myeloid cells) 1+ macrophages. As early as 7 days after MI, macrophage F7 deletion led to an expansion of reparative Olfml (olfactomedin) 3+ macrophages and, conversely, to a reduction of TF+/F7+/TREM1+ macrophages, which were also reduced in PAR2R38E mice. Short-term treatment from days 1 to 5 after nonreperfused MI with a monoclonal antibody inhibiting the macrophage TF-F7-PAR2 signaling complex without anticoagulant activity improved cardiac dysfunction, decreased excess fibrosis, attenuated vascular endothelial dysfunction, and increased survival 28 days after MI.

Conclusions: Extravascular TF-F7-PAR2 complex signaling drives inflammatory macrophage polarization in ischemic heart disease. Targeting this signaling complex for specific therapeutic macrophage reprogramming following MI attenuates cardiac fibrosis and improves cardiovascular function.

背景:在缺血性心脏病中,过多的纤维重塑会导致心脏功能障碍,其驱动因素是骨髓细胞凝血信号的MAP(丝裂原活化蛋白)激酶依赖性TGF-ß1(转化生长因子-ß1)激活。心肌梗死(MI)后如何有针对性地针对凝血-炎症回路进行有益的巨噬细胞重编程,目前还不完全清楚:方法:用永久性结扎左前降支动脉的小鼠建立非再灌注心肌梗死模型,并通过单细胞RNA测序、蛋白质表达变化、共聚焦显微镜和纵向恢复监测进行分析。我们利用遗传小鼠模型和药物干预,探究了组织因子(TF)-因子7(F7)-整合素ß1-PAR2(蛋白酶激活受体2)信号复合体的作用:结果:与对照组相比,对裂解不敏感的PAR2R38E和骨髓细胞整合素ß1缺陷小鼠在心肌梗死后的心脏功能有所改善。单核细胞的邻近接合试验表明,缺失单核细胞/巨噬细胞 F7 的小鼠在心肌梗死后 F7 与整合素 ß1 的共定位减少。与对照组相比,F7fl/fl CX3CR1Cre小鼠的TGF-ß1和MAP激酶活化以及心肌梗死后的心脏功能障碍均有所降低,尽管髓系细胞的整体招募没有改变。心肌梗死后3天和7天,CD45(分化簇45)+细胞的单细胞mRNA测序发现了从招募的单核细胞到炎性TF+/F7+/TREM(髓样细胞上表达的触发受体)1+巨噬细胞的轨迹。早在心肌梗死后 7 天,巨噬细胞 F7 缺失就导致了修复性 Olfml(嗅探素)3+ 巨噬细胞的扩增,反之,TF+/F7+/TREM1+ 巨噬细胞的减少,PAR2R38E 小鼠的巨噬细胞也减少了。在非再灌注心肌梗死后的第1至5天,使用一种抑制巨噬细胞TF-F7-PAR2信号复合物的单克隆抗体进行短期治疗,但不具有抗凝活性,这种抗体可改善心功能障碍,减少过度纤维化,减轻血管内皮功能障碍,并提高心肌梗死后28天的存活率:结论:血管外 TF-F7-PAR2 复合物信号驱动缺血性心脏病中巨噬细胞的炎症极化。结论:血管外 TF-F7-PAR2 复合物信号驱动缺血性心脏病中的炎性巨噬细胞极化,针对这一信号复合物进行特定的治疗性巨噬细胞重编程可减轻心肌梗死后的心脏纤维化并改善心血管功能。
{"title":"Macrophage-Expressed Coagulation Factor 7 Promotes Adverse Cardiac Remodeling.","authors":"Venkata Garlapati, Qi Luo, Jens Posma, Melania Aluia, Than Son Nguyen, Kristin Grunz, Michael Molitor, Stefanie Finger, Gregory Harms, Tobias Bopp, Wolfram Ruf, Philip Wenzel","doi":"10.1161/CIRCRESAHA.123.324114","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.123.324114","url":null,"abstract":"<p><strong>Background: </strong>Excess fibrotic remodeling causes cardiac dysfunction in ischemic heart disease, driven by MAP (mitogen-activated protein) kinase-dependent TGF-ß1 (transforming growth factor-ß1) activation by coagulation signaling of myeloid cells. How coagulation-inflammatory circuits can be specifically targeted to achieve beneficial macrophage reprogramming after myocardial infarction (MI) is not completely understood.</p><p><strong>Methods: </strong>Mice with permanent ligation of the left anterior descending artery were used to model nonreperfused MI and analyzed by single-cell RNA sequencing, protein expression changes, confocal microscopy, and longitudinal monitoring of recovery. We probed the role of the tissue factor (TF)-factor 7 (F7)-integrin ß1-PAR2 (protease-activated receptor 2) signaling complex by utilizing genetic mouse models and pharmacological intervention.</p><p><strong>Results: </strong>Cleavage-insensitive PAR2<sup>R38E</sup> and myeloid cell integrin ß1-deficient mice had improved cardiac function after MI compared with controls. Proximity ligation assays of monocytic cells demonstrated that colocalization of F7 with integrin ß1 was diminished in monocyte/macrophage F7-deficient mice after MI. Compared with controls, F7<sup>fl/fl</sup> CX3CR1<sup>Cre</sup> mice showed reduced TGF-ß1 and MAP kinase activation, as well as cardiac dysfunction after MI, despite unaltered overall recruitment of myeloid cells. Single-cell mRNA sequencing of CD45 (cluster of differentiation 45)<sup>+</sup> cells 3 and 7 days after MI uncovered a trajectory from recruited monocytes to inflammatory TF<sup>+</sup>/F7<sup>+</sup>/TREM (triggered receptor expressed on myeloid cells) 1<sup>+</sup> macrophages. As early as 7 days after MI, macrophage F7 deletion led to an expansion of reparative Olfml (olfactomedin) 3<sup>+</sup> macrophages and, conversely, to a reduction of TF<sup>+</sup>/F7<sup>+</sup>/TREM1<sup>+</sup> macrophages, which were also reduced in PAR2<sup>R38E</sup> mice. Short-term treatment from days 1 to 5 after nonreperfused MI with a monoclonal antibody inhibiting the macrophage TF-F7-PAR2 signaling complex without anticoagulant activity improved cardiac dysfunction, decreased excess fibrosis, attenuated vascular endothelial dysfunction, and increased survival 28 days after MI.</p><p><strong>Conclusions: </strong>Extravascular TF-F7-PAR2 complex signaling drives inflammatory macrophage polarization in ischemic heart disease. Targeting this signaling complex for specific therapeutic macrophage reprogramming following MI attenuates cardiac fibrosis and improves cardiovascular function.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142132009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
EPAS1 Attenuates Atherosclerosis Initiation at Disturbed Flow Sites Through Endothelial Fatty Acid Uptake. EPAS1 通过内皮脂肪酸的摄取减轻动脉粥样硬化在紊乱血流部位的发生。
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-05 DOI: 10.1161/CIRCRESAHA.123.324054
Daniela Pirri, Siyu Tian, Blanca Tardajos-Ayllon, Sophie Irving, Francesco Donati, Scott P Allen, Tadanori Mammoto, Gemma Vilahur, Lida Kabir, Jane Bennett, Yasmin Rasool, Charis Pericleous, Guianfranco Mazzei, Liam McAllan, William R Scott, Thomas Koestler, Urs Zingg, Graeme Birdsey, Clint Miller, Torsten Schenkel, Emily V Chambers, Mark Dunning, Jovana Serbanovic-Canic, Francesco Botrè, Akiko Mammoto, Suowen Xu, Elena Osto, Weiping Han, Maria Fragiadaki, Paul C Evans

Background: Atherosclerotic plaques form unevenly due to disturbed blood flow, causing localized endothelial cell (EC) dysfunction. Obesity exacerbates this process, but the underlying molecular mechanisms are unclear. The transcription factor EPAS1 (HIF2A) has regulatory roles in endothelium, but its involvement in atherosclerosis remains unexplored. This study investigates the potential interplay between EPAS1, obesity, and atherosclerosis.

Methods: Responses to shear stress were analyzed using cultured porcine aortic EC exposed to flow in vitro coupled with metabolic and molecular analyses and by en face immunostaining of murine aortic EC exposed to disturbed flow in vivo. Obesity and dyslipidemia were induced in mice via exposure to a high-fat diet or through Leptin gene deletion. The role of Epas1 in atherosclerosis was evaluated by inducible endothelial Epas1 deletion, followed by hypercholesterolemia induction (adeno-associated virus-PCSK9 [proprotein convertase subtilisin/kexin type 9]; high-fat diet).

Results: En face staining revealed EPAS1 enrichment at sites of disturbed blood flow that are prone to atherosclerosis initiation. Obese mice exhibited substantial reduction in endothelial EPAS1 expression. Sulforaphane, a compound with known atheroprotective effects, restored EPAS1 expression and concurrently reduced plasma triglyceride levels in obese mice. Consistently, triglyceride derivatives (free fatty acids) suppressed EPAS1 in cultured EC by upregulating the negative regulator PHD2. Clinical observations revealed that reduced serum EPAS1 correlated with increased endothelial PHD2 and PHD3 in obese individuals. Functionally, endothelial EPAS1 deletion increased lesion formation in hypercholesterolemic mice, indicating an atheroprotective function. Mechanistic insights revealed that EPAS1 protects arteries by maintaining endothelial proliferation by positively regulating the expression of the fatty acid-handling molecules CD36 and LIPG to increase fatty acid beta-oxidation.

Conclusions: Endothelial EPAS1 attenuates atherosclerosis at sites of disturbed flow by maintaining EC proliferation via fatty acid uptake and metabolism. This endothelial repair pathway is inhibited in obesity, suggesting a novel triglyceride-PHD2 modulation pathway suppressing EPAS1 expression. These findings have implications for therapeutic strategies addressing vascular dysfunction in obesity.

背景:动脉粥样硬化斑块因血流紊乱而形成不均,导致局部内皮细胞(EC)功能障碍。肥胖会加剧这一过程,但其潜在的分子机制尚不清楚。转录因子 EPAS1(HIF2A)在内皮中具有调节作用,但其在动脉粥样硬化中的参与作用仍未得到探讨。本研究探讨了 EPAS1、肥胖和动脉粥样硬化之间的潜在相互作用:方法:使用体外暴露于流动的培养猪主动脉 EC,结合代谢和分子分析,并通过对体内暴露于紊乱流动的鼠主动脉 EC 进行正面免疫染色,分析其对剪切应力的反应。通过高脂饮食或Leptin基因缺失诱导小鼠肥胖和血脂异常。通过诱导性内皮 Epas1 基因缺失和高胆固醇血症诱导(腺相关病毒-PCSK9 [proprotein convertase subtilisin/kexin type 9]; 高脂饮食)评估 Epas1 在动脉粥样硬化中的作用:正面染色显示,EPAS1富集在容易引发动脉粥样硬化的血流紊乱部位。肥胖小鼠的内皮 EPAS1 表达量大幅减少。已知具有动脉粥样硬化保护作用的化合物 Sulforaphane 可恢复 EPAS1 的表达,同时降低肥胖小鼠的血浆甘油三酯水平。同样,甘油三酯衍生物(游离脂肪酸)通过上调负调控因子 PHD2 抑制了培养 EC 中的 EPAS1。临床观察显示,血清 EPAS1 的降低与肥胖者内皮 PHD2 和 PHD3 的升高相关。从功能上讲,内皮 EPAS1 基因缺失会增加高胆固醇血症小鼠的病变形成,表明其具有动脉粥样硬化保护功能。机理研究发现,EPAS1通过正向调节脂肪酸处理分子CD36和LIPG的表达以增加脂肪酸β-氧化,从而维持内皮增殖,从而保护动脉:内皮 EPAS1 通过脂肪酸摄取和代谢维持内皮细胞增殖,从而减轻血流紊乱部位的动脉粥样硬化。这一内皮修复途径在肥胖症中受到抑制,表明一种新型甘油三酯-PHD2调节途径抑制了EPAS1的表达。这些发现对解决肥胖症血管功能障碍的治疗策略具有重要意义。
{"title":"EPAS1 Attenuates Atherosclerosis Initiation at Disturbed Flow Sites Through Endothelial Fatty Acid Uptake.","authors":"Daniela Pirri, Siyu Tian, Blanca Tardajos-Ayllon, Sophie Irving, Francesco Donati, Scott P Allen, Tadanori Mammoto, Gemma Vilahur, Lida Kabir, Jane Bennett, Yasmin Rasool, Charis Pericleous, Guianfranco Mazzei, Liam McAllan, William R Scott, Thomas Koestler, Urs Zingg, Graeme Birdsey, Clint Miller, Torsten Schenkel, Emily V Chambers, Mark Dunning, Jovana Serbanovic-Canic, Francesco Botrè, Akiko Mammoto, Suowen Xu, Elena Osto, Weiping Han, Maria Fragiadaki, Paul C Evans","doi":"10.1161/CIRCRESAHA.123.324054","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.123.324054","url":null,"abstract":"<p><strong>Background: </strong>Atherosclerotic plaques form unevenly due to disturbed blood flow, causing localized endothelial cell (EC) dysfunction. Obesity exacerbates this process, but the underlying molecular mechanisms are unclear. The transcription factor EPAS1 (HIF2A) has regulatory roles in endothelium, but its involvement in atherosclerosis remains unexplored. This study investigates the potential interplay between EPAS1, obesity, and atherosclerosis.</p><p><strong>Methods: </strong>Responses to shear stress were analyzed using cultured porcine aortic EC exposed to flow in vitro coupled with metabolic and molecular analyses and by en face immunostaining of murine aortic EC exposed to disturbed flow in vivo. Obesity and dyslipidemia were induced in mice via exposure to a high-fat diet or through Leptin gene deletion. The role of <i>Epas1</i> in atherosclerosis was evaluated by inducible endothelial <i>Epas1</i> deletion, followed by hypercholesterolemia induction (adeno-associated virus-PCSK9 [proprotein convertase subtilisin/kexin type 9]; high-fat diet).</p><p><strong>Results: </strong>En face staining revealed EPAS1 enrichment at sites of disturbed blood flow that are prone to atherosclerosis initiation. Obese mice exhibited substantial reduction in endothelial EPAS1 expression. Sulforaphane, a compound with known atheroprotective effects, restored EPAS1 expression and concurrently reduced plasma triglyceride levels in obese mice. Consistently, triglyceride derivatives (free fatty acids) suppressed EPAS1 in cultured EC by upregulating the negative regulator PHD2. Clinical observations revealed that reduced serum EPAS1 correlated with increased endothelial PHD2 and PHD3 in obese individuals. Functionally, endothelial EPAS1 deletion increased lesion formation in hypercholesterolemic mice, indicating an atheroprotective function. Mechanistic insights revealed that EPAS1 protects arteries by maintaining endothelial proliferation by positively regulating the expression of the fatty acid-handling molecules CD36 and LIPG to increase fatty acid beta-oxidation.</p><p><strong>Conclusions: </strong>Endothelial EPAS1 attenuates atherosclerosis at sites of disturbed flow by maintaining EC proliferation via fatty acid uptake and metabolism. This endothelial repair pathway is inhibited in obesity, suggesting a novel triglyceride-PHD2 modulation pathway suppressing EPAS1 expression. These findings have implications for therapeutic strategies addressing vascular dysfunction in obesity.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142132007","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Crotonylation of NAE1 Modulates Cardiac Hypertrophy via Gelsolin Neddylation. NAE1 的 Crotonylation 通过 Gelsolin Neddylation 调节心肌肥大。
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-04 DOI: 10.1161/CIRCRESAHA.124.324733
Jie Ju, Kai Wang, Fang Liu, Cui-Yun Liu, Yun-Hong Wang, Shao-Chong Wang, Lu-Yu Zhou, Xin-Min Li, Yu-Qin Wang, Xin-Zhe Chen, Rui-Feng Li, Shi-Jun Xu, Chen Chen, Mei-Hua Zhang, Su-Min Yang, Jin-Wei Tian, Kun Wang
<p><strong>Background: </strong>Cardiac hypertrophy and its associated remodeling are among the leading causes of heart failure. Lysine crotonylation is a recently discovered posttranslational modification whose role in cardiac hypertrophy remains largely unknown. NAE1 (NEDD8-activating enzyme E1 regulatory subunit) is mainly involved in the neddylation modification of protein targets. However, the function of crotonylated NAE1 has not been defined. This study aims to elucidate the effects and mechanisms of NAE1 crotonylation on cardiac hypertrophy.</p><p><strong>Methods: </strong>Crotonylation levels were detected in both human and mouse subjects with cardiac hypertrophy through immunoprecipitation and Western blot assays. TMT-labeled quantitative lysine crotonylome analysis was performed to identify the crotonylated proteins in a mouse cardiac hypertrophic model induced by transverse aortic constriction. We generated NAE1 knock-in mice carrying a crotonylation-defective lysine to arginine K238R (lysine to arginine mutation at site 238) mutation (NAE1 K238R) and NAE1 knock-in mice expressing a crotonylation-mimicking lysine to glutamine K238Q (lysine to glutamine mutation at site 238) mutation (NAE1 K238Q) to assess the functional role of crotonylation of NAE1 at K238 in pathological cardiac hypertrophy. Furthermore, we combined coimmunoprecipitation, mass spectrometry, and dot blot analysis that was followed by multiple molecular biological methodologies to identify the target GSN (gelsolin) and corresponding molecular events contributing to the function of NAE1 K238 crotonylation.</p><p><strong>Results: </strong>The crotonylation level of NAE1 was increased in mice and patients with cardiac hypertrophy. Quantitative crotonylomics analysis revealed that K238 was the main crotonylation site of NAE1. Loss of K238 crotonylation in NAE1 K238R knock-in mice attenuated cardiac hypertrophy and restored the heart function, while hypercrotonylation mimic in NAE1 K238Q knock-in mice significantly enhanced transverse aortic constriction-induced pathological hypertrophic response, leading to impaired cardiac structure and function. The recombinant adenoviral vector carrying NAE1 K238R mutant attenuated, while the K238Q mutant aggravated Ang II (angiotensin II)-induced hypertrophy. Mechanistically, we identified GSN as a direct target of NAE1. K238 crotonylation of NAE1 promoted GSN neddylation and, thus, enhanced its protein stability and expression. NAE1 crotonylation-dependent increase of GSN promoted actin-severing activity, which resulted in adverse cytoskeletal remodeling and progression of pathological hypertrophy.</p><p><strong>Conclusions: </strong>Our findings provide new insights into the previously unrecognized role of crotonylation on nonhistone proteins during cardiac hypertrophy. We found that K238 crotonylation of NAE1 plays an essential role in mediating cardiac hypertrophy through GSN neddylation, which provides potential novel therapeutic
背景:心脏肥大及其相关重塑是导致心力衰竭的主要原因之一。赖氨酸巴豆酰化是最近发现的一种翻译后修饰,其在心肌肥厚中的作用在很大程度上仍然未知。NAE1(NEDD8-激活酶 E1 调控亚基)主要参与蛋白质靶点的奈德基化修饰。然而,巴豆酰化的 NAE1 的功能尚未明确。本研究旨在阐明NAE1巴豆酰化对心脏肥大的影响和机制:方法:通过免疫沉淀和Western印迹检测人和小鼠心肌肥厚患者的巴豆酰化水平。在横向主动脉收缩诱导的小鼠心脏肥大模型中,进行了TMT标记的赖氨酸巴豆酰化定量分析,以确定巴豆酰化的蛋白质。我们产生了携带赖氨酸至精氨酸K238R(位点238上的赖氨酸至精氨酸突变)突变的NAE1基因敲入小鼠(NAE1 K238R)和表达模拟赖氨酸至谷氨酸突变的NAE1基因敲入小鼠(NAE1 K238R)。我们还研究了表达模拟赖氨酸至谷氨酰胺 K238Q(位点 238 上的赖氨酸至谷氨酰胺突变)突变(NAE1 K238Q)的 NAE1 基因敲入小鼠,以评估 NAE1 K238 上的巴豆酰化在病理性心肌肥厚中的功能作用。此外,我们还结合免疫沉淀、质谱分析和点印迹分析等多种分子生物学方法,鉴定了目标GSN(凝胶溶素)和导致NAE1 K238巴豆酰化功能的相应分子事件:结果:在小鼠和心肌肥厚患者体内,NAE1的巴豆酰化水平升高。定量巴豆酰组学分析表明,K238是NAE1的主要巴豆酰化位点。NAE1 K238R基因敲入小鼠的K238巴豆酰化缺失可减轻心脏肥大并恢复心脏功能,而NAE1 K238Q基因敲入小鼠的高巴豆酰化模拟可显著增强横主动脉收缩诱导的病理性肥大反应,导致心脏结构和功能受损。携带NAE1 K238R突变体的重组腺病毒载体减轻了Ang II(血管紧张素II)诱导的肥厚,而K238Q突变体则加重了Ang II诱导的肥厚。从机理上讲,我们发现GSN是NAE1的直接靶标。NAE1的K238巴豆酰化促进了GSN的尼达基化,从而增强了其蛋白的稳定性和表达。NAE1巴豆酰化依赖的GSN增加促进了肌动蛋白的分裂活性,从而导致了不良的细胞骨架重塑和病理性肥大的进展:我们的研究结果为我们提供了新的视角,使我们了解到巴豆酰化在心脏肥大过程中对非组蛋白的作用。我们发现,NAE1的K238巴豆酰化在通过GSN内酰化介导心肌肥大中起着至关重要的作用,这为病理性肥大和心脏重塑提供了潜在的新治疗靶点。
{"title":"Crotonylation of NAE1 Modulates Cardiac Hypertrophy via Gelsolin Neddylation.","authors":"Jie Ju, Kai Wang, Fang Liu, Cui-Yun Liu, Yun-Hong Wang, Shao-Chong Wang, Lu-Yu Zhou, Xin-Min Li, Yu-Qin Wang, Xin-Zhe Chen, Rui-Feng Li, Shi-Jun Xu, Chen Chen, Mei-Hua Zhang, Su-Min Yang, Jin-Wei Tian, Kun Wang","doi":"10.1161/CIRCRESAHA.124.324733","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.124.324733","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Cardiac hypertrophy and its associated remodeling are among the leading causes of heart failure. Lysine crotonylation is a recently discovered posttranslational modification whose role in cardiac hypertrophy remains largely unknown. NAE1 (NEDD8-activating enzyme E1 regulatory subunit) is mainly involved in the neddylation modification of protein targets. However, the function of crotonylated NAE1 has not been defined. This study aims to elucidate the effects and mechanisms of NAE1 crotonylation on cardiac hypertrophy.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;Crotonylation levels were detected in both human and mouse subjects with cardiac hypertrophy through immunoprecipitation and Western blot assays. TMT-labeled quantitative lysine crotonylome analysis was performed to identify the crotonylated proteins in a mouse cardiac hypertrophic model induced by transverse aortic constriction. We generated NAE1 knock-in mice carrying a crotonylation-defective lysine to arginine K238R (lysine to arginine mutation at site 238) mutation (NAE1 K238R) and NAE1 knock-in mice expressing a crotonylation-mimicking lysine to glutamine K238Q (lysine to glutamine mutation at site 238) mutation (NAE1 K238Q) to assess the functional role of crotonylation of NAE1 at K238 in pathological cardiac hypertrophy. Furthermore, we combined coimmunoprecipitation, mass spectrometry, and dot blot analysis that was followed by multiple molecular biological methodologies to identify the target GSN (gelsolin) and corresponding molecular events contributing to the function of NAE1 K238 crotonylation.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;The crotonylation level of NAE1 was increased in mice and patients with cardiac hypertrophy. Quantitative crotonylomics analysis revealed that K238 was the main crotonylation site of NAE1. Loss of K238 crotonylation in NAE1 K238R knock-in mice attenuated cardiac hypertrophy and restored the heart function, while hypercrotonylation mimic in NAE1 K238Q knock-in mice significantly enhanced transverse aortic constriction-induced pathological hypertrophic response, leading to impaired cardiac structure and function. The recombinant adenoviral vector carrying NAE1 K238R mutant attenuated, while the K238Q mutant aggravated Ang II (angiotensin II)-induced hypertrophy. Mechanistically, we identified GSN as a direct target of NAE1. K238 crotonylation of NAE1 promoted GSN neddylation and, thus, enhanced its protein stability and expression. NAE1 crotonylation-dependent increase of GSN promoted actin-severing activity, which resulted in adverse cytoskeletal remodeling and progression of pathological hypertrophy.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;Our findings provide new insights into the previously unrecognized role of crotonylation on nonhistone proteins during cardiac hypertrophy. We found that K238 crotonylation of NAE1 plays an essential role in mediating cardiac hypertrophy through GSN neddylation, which provides potential novel therapeutic","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Monocytes Reprogrammed by 4-PBA Potently Contribute to the Resolution of Inflammation and Atherosclerosis. 经 4-PBA 重编程的单核细胞可有效缓解炎症和动脉粥样硬化。
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-03 DOI: 10.1161/CIRCRESAHA.124.325023
Shuo Geng, Ran Lu, Yao Zhang, Yajun Wu, Ling Xie, Blake Caldwell, Kisha Pradhan, Ziyue Yi, Jacqueline Hou, Feng Xu, Xian Chen, Liwu Li

Background: Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis.

Methods: Systems analyses integrating single-cell RNA sequencing and complementary immunologic approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming were assessed by integrated biochemical and genetic approaches. The intercellular propagation of homeostasis resolution was evaluated by coculture assays with donor monocytes trained by 4-PBA and recipient naive monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high-fat diet-fed ApoE-/- mouse model with IP 4-PBA administration. Furthermore, the selective efficacy of 4-PBA-trained monocytes was examined by IV transfusion of ex vivo trained monocytes by 4-PBA into recipient high-fat diet-fed ApoE-/- mice.

Results: In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 (intercellular adhesion molecule 1) via reducing peroxisome stress and attenuating SYK (spleen tyrosine kinase)-mTOR (mammalian target of rapamycin) signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ (peroxisome proliferator-activated receptor γ) neddylation mediated by TOLLIP (toll-interacting protein). 4-PBA-trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA-trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo.

Conclusions: Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.

背景:由炎症单核细胞引发的慢性炎症是动脉粥样硬化发病机制的基础。然而,目前还没有针对单核细胞有效解决慢性低度炎症的方法。小分子化合物 4-苯基丁酸(4-PBA)在减轻动脉粥样硬化方面具有广泛的抗炎作用。选择性递送 4-PBA 重编程单核细胞可能具有新的潜力,为治疗动脉粥样硬化提供靶向和精准治疗:方法:结合单细胞 RNA 测序和互补免疫学方法进行系统分析,确定了 4-PBA 训练的重编程单核细胞的关键分辨特征和定义标记。综合生化和遗传方法评估了单核细胞重编程的分子机制。通过与经 4-PBA 训练的供体单核细胞和受体天真单核细胞进行共培养实验,评估了细胞间平衡解析的传播。在高脂饮食喂养的载脂蛋白E-/-小鼠模型中,通过IP 4-PBA给药,评估了4-PBA在体内解决单核细胞问题和预防动脉粥样硬化的效果。此外,还通过向接受高脂饮食的载脂蛋白E-/-小鼠静脉注射4-PBA训练的单核细胞,检测了4-PBA训练的单核细胞的选择性功效:结果:在这项研究中,我们发现单核细胞可被 4-PBA 有效地重塑为一种以粘附性降低和抗炎介质 CD24 表达增强为特征的免疫溶解状态。从机理上讲,4-PBA 通过降低过氧化物酶体应激和减弱 SYK(脾脏酪氨酸激酶)-mTOR(哺乳动物雷帕霉素靶标)信号传导,减少了 ICAM-1(细胞间粘附分子 1)的表达。与此同时,4-PBA 通过促进由 TOLLIP(通行费互作蛋白)介导的 PPARγ(过氧化物酶体增殖激活受体γ)内酰化,增强了分解介质 CD24 的表达。经过 4-PBA 训练的单核细胞可通过 CD24 向邻近的单核细胞有效传播抗炎活性。我们的数据进一步证明,4-PBA 训练的单核细胞在体内给药时可有效减少动脉粥样硬化的发病机制:我们的研究描述了一种稳健有效的方法来生成抗动脉粥样硬化单核细胞,描述了靶向单核细胞重编程的新机制,并提供了一种基于提供重编程抗动脉粥样硬化单核细胞的精准疗法。
{"title":"Monocytes Reprogrammed by 4-PBA Potently Contribute to the Resolution of Inflammation and Atherosclerosis.","authors":"Shuo Geng, Ran Lu, Yao Zhang, Yajun Wu, Ling Xie, Blake Caldwell, Kisha Pradhan, Ziyue Yi, Jacqueline Hou, Feng Xu, Xian Chen, Liwu Li","doi":"10.1161/CIRCRESAHA.124.325023","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.124.325023","url":null,"abstract":"<p><strong>Background: </strong>Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis.</p><p><strong>Methods: </strong>Systems analyses integrating single-cell RNA sequencing and complementary immunologic approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming were assessed by integrated biochemical and genetic approaches. The intercellular propagation of homeostasis resolution was evaluated by coculture assays with donor monocytes trained by 4-PBA and recipient naive monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high-fat diet-fed <i>ApoE</i><sup><i>-/-</i></sup> mouse model with IP 4-PBA administration. Furthermore, the selective efficacy of 4-PBA-trained monocytes was examined by IV transfusion of ex vivo trained monocytes by 4-PBA into recipient high-fat diet-fed <i>ApoE</i><sup><i>-/-</i></sup> mice.</p><p><strong>Results: </strong>In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 (intercellular adhesion molecule 1) via reducing peroxisome stress and attenuating SYK (spleen tyrosine kinase)-mTOR (mammalian target of rapamycin) signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ (peroxisome proliferator-activated receptor γ) neddylation mediated by TOLLIP (toll-interacting protein). 4-PBA-trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA-trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo.</p><p><strong>Conclusions: </strong>Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142119127","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
AMPK Attenuation of β-Adrenergic Receptor-Induced Cardiac Injury via Phosphorylation of β-Arrestin-1-ser330. AMPK通过β-阿瑞斯汀-1-ser330的磷酸化减轻β-肾上腺素能受体诱导的心脏损伤
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-08-30 Epub Date: 2024-07-31 DOI: 10.1161/CIRCRESAHA.124.324762
Mingming Zhao, Ning Cao, Huijun Gu, Jiachao Xu, Wenli Xu, Di Zhang, Tong-You Wade Wei, Kang Wang, Ruiping Guo, Hongtu Cui, Xiaofeng Wang, Xin Guo, Zhiyuan Li, Kangmin He, Zijian Li, Youyi Zhang, John Y-J Shyy, Erdan Dong, Han Xiao

Background: β-adrenergic receptor (β-AR) overactivation is a major pathological cue associated with cardiac injury and diseases. AMPK (AMP-activated protein kinase), a conserved energy sensor, regulates energy metabolism and is cardioprotective. However, whether AMPK exerts cardioprotective effects via regulating the signaling pathway downstream of β-AR remains unclear.

Methods: Using immunoprecipitation, mass spectrometry, site-specific mutation, in vitro kinase assay, and in vivo animal studies, we determined whether AMPK phosphorylates β-arrestin-1 at serine (Ser) 330. Wild-type mice and mice with site-specific mutagenesis (S330A knock-in [KI]/S330D KI) were subcutaneously injected with the β-AR agonist isoproterenol (5 mg/kg) to evaluate the causality between β-adrenergic insult and β-arrestin-1 Ser330 phosphorylation. Cardiac transcriptomics was used to identify changes in gene expression from β-arrestin-1-S330A/S330D mutation and β-adrenergic insult.

Results: Metformin could decrease cAMP/PKA (protein kinase A) signaling induced by isoproterenol. AMPK bound to β-arrestin-1 and phosphorylated Ser330 with the highest phosphorylated mass spectrometry score. AMPK activation promoted β-arrestin-1 Ser330 phosphorylation in vitro and in vivo. Neonatal mouse cardiomyocytes overexpressing β-arrestin-1-S330D (active form) inhibited the β-AR/cAMP/PKA axis by increasing PDE (phosphodiesterase) 4 expression and activity. Cardiac transcriptomics revealed that the differentially expressed genes between isoproterenol-treated S330A KI and S330D KI mice were mainly involved in immune processes and inflammatory response. β-arrestin-1 Ser330 phosphorylation inhibited isoproterenol-induced reactive oxygen species production and NLRP3 (NOD-like receptor protein 3) inflammasome activation in neonatal mouse cardiomyocytes. In S330D KI mice, the β-AR-activated cAMP/PKA pathways were attenuated, leading to repressed inflammasome activation, reduced expression of proinflammatory cytokines, and mitigated macrophage infiltration. Compared with S330A KI mice, S330D KI mice showed diminished cardiac fibrosis and improved cardiac function upon isoproterenol exposure. However, the cardiac protection exerted by AMPK was abolished in S330A KI mice.

Conclusions: AMPK phosphorylation of β-arrestin-1 Ser330 potentiated PDE4 expression and activity, thereby inhibiting β-AR/cAMP/PKA activation. Subsequently, β-arrestin-1 Ser330 phosphorylation blocks β-AR-induced cardiac inflammasome activation and remodeling.

背景:β-肾上腺素能受体(β-AR)过度激活是与心脏损伤和疾病相关的主要病理线索。AMPK(AMP 激活蛋白激酶)是一种保守的能量传感器,可调节能量代谢并具有心脏保护作用。然而,AMPK是否通过调节β-AR下游的信号通路来发挥心脏保护作用仍不清楚:方法:我们利用免疫沉淀、质谱分析、位点特异性突变、体外激酶测定和体内动物实验,确定了 AMPK 是否会使β-arrestin-1 在丝氨酸(Ser)330 处磷酸化。给野生型小鼠和位点特异性突变(S330A基因敲入[KI]/S330D KI)小鼠皮下注射β-AR激动剂异丙肾上腺素(5 mg/kg),以评估β-肾上腺素能损伤与β-arrestin-1 Ser330磷酸化之间的因果关系。心脏转录组学用于确定β-arrestin-1-S330A/S330D突变和β-肾上腺素损伤引起的基因表达变化:结果:二甲双胍可降低异丙肾上腺素诱导的cAMP/PKA(蛋白激酶A)信号传导。AMPK与β-arrestin-1结合并磷酸化Ser330,其磷酸化质谱得分最高。在体外和体内,AMPK的激活促进了β-arrestin-1 Ser330的磷酸化。过表达β-arrestin-1-S330D(活性形式)的新生小鼠心肌细胞通过增加PDE(磷酸二酯酶)4的表达和活性来抑制β-AR/cAMP/PKA轴。心脏转录组学显示,异丙肾上腺素处理的 S330A KI 和 S330D KI 小鼠之间表达不同的基因主要涉及免疫过程和炎症反应。在新生小鼠心肌细胞中,β-arrestin-1 Ser330磷酸化抑制了异丙托品醇诱导的活性氧产生和NLRP3(NOD样受体蛋白3)炎性组的激活。在 S330D KI 小鼠中,β-AR 激活的 cAMP/PKA 通路被削弱,导致炎症小体激活被抑制,促炎细胞因子表达减少,巨噬细胞浸润减轻。与 S330A KI 小鼠相比,S330D KI 小鼠在接触异丙肾上腺素后,心脏纤维化减轻,心脏功能改善。然而,在 S330A KI 小鼠中,AMPK 对心脏的保护作用消失了:结论:AMPK 对 β-arrestin-1 Ser330 的磷酸化增强了 PDE4 的表达和活性,从而抑制了 β-AR/cAMP/PKA 的激活。随后,β-arrestin-1 Ser330 磷酸化阻断了 β-AR 诱导的心脏炎症小体激活和重塑。
{"title":"AMPK Attenuation of β-Adrenergic Receptor-Induced Cardiac Injury via Phosphorylation of β-Arrestin-1-ser330.","authors":"Mingming Zhao, Ning Cao, Huijun Gu, Jiachao Xu, Wenli Xu, Di Zhang, Tong-You Wade Wei, Kang Wang, Ruiping Guo, Hongtu Cui, Xiaofeng Wang, Xin Guo, Zhiyuan Li, Kangmin He, Zijian Li, Youyi Zhang, John Y-J Shyy, Erdan Dong, Han Xiao","doi":"10.1161/CIRCRESAHA.124.324762","DOIUrl":"10.1161/CIRCRESAHA.124.324762","url":null,"abstract":"<p><strong>Background: </strong>β-adrenergic receptor (β-AR) overactivation is a major pathological cue associated with cardiac injury and diseases. AMPK (AMP-activated protein kinase), a conserved energy sensor, regulates energy metabolism and is cardioprotective. However, whether AMPK exerts cardioprotective effects via regulating the signaling pathway downstream of β-AR remains unclear.</p><p><strong>Methods: </strong>Using immunoprecipitation, mass spectrometry, site-specific mutation, in vitro kinase assay, and in vivo animal studies, we determined whether AMPK phosphorylates β-arrestin-1 at serine (Ser) 330. Wild-type mice and mice with site-specific mutagenesis (S330A knock-in [KI]/S330D KI) were subcutaneously injected with the β-AR agonist isoproterenol (5 mg/kg) to evaluate the causality between β-adrenergic insult and β-arrestin-1 Ser330 phosphorylation. Cardiac transcriptomics was used to identify changes in gene expression from β-arrestin-1-S330A/S330D mutation and β-adrenergic insult.</p><p><strong>Results: </strong>Metformin could decrease cAMP/PKA (protein kinase A) signaling induced by isoproterenol. AMPK bound to β-arrestin-1 and phosphorylated Ser330 with the highest phosphorylated mass spectrometry score. AMPK activation promoted β-arrestin-1 Ser330 phosphorylation in vitro and in vivo. Neonatal mouse cardiomyocytes overexpressing β-arrestin-1-S330D (active form) inhibited the β-AR/cAMP/PKA axis by increasing PDE (phosphodiesterase) 4 expression and activity. Cardiac transcriptomics revealed that the differentially expressed genes between isoproterenol-treated S330A KI and S330D KI mice were mainly involved in immune processes and inflammatory response. β-arrestin-1 Ser330 phosphorylation inhibited isoproterenol-induced reactive oxygen species production and NLRP3 (NOD-like receptor protein 3) inflammasome activation in neonatal mouse cardiomyocytes. In S330D KI mice, the β-AR-activated cAMP/PKA pathways were attenuated, leading to repressed inflammasome activation, reduced expression of proinflammatory cytokines, and mitigated macrophage infiltration. Compared with S330A KI mice, S330D KI mice showed diminished cardiac fibrosis and improved cardiac function upon isoproterenol exposure. However, the cardiac protection exerted by AMPK was abolished in S330A KI mice.</p><p><strong>Conclusions: </strong>AMPK phosphorylation of β-arrestin-1 Ser330 potentiated PDE4 expression and activity, thereby inhibiting β-AR/cAMP/PKA activation. Subsequently, β-arrestin-1 Ser330 phosphorylation blocks β-AR-induced cardiac inflammasome activation and remodeling.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141855001","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Circulation 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