首页 > 最新文献

Circulation research最新文献

英文 中文
Meet the First Authors. 认识第一作者
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-09-12 DOI: 10.1161/RES.0000000000000692
{"title":"Meet the First Authors.","authors":"","doi":"10.1161/RES.0000000000000692","DOIUrl":"https://doi.org/10.1161/RES.0000000000000692","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"135 7","pages":"706-707"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142281156","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
Targeting Cyclophilin A in the Cardiac Microenvironment Preserves Heart Function and Structure in Failing Hearts. 靶向心脏微环境中的嗜环素 A 可保护衰竭心脏的功能和结构
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-08-14 DOI: 10.1161/CIRCRESAHA.124.324812
Manuel Sigle, Anne-Katrin Rohlfing, Melanie Cruz Santos, Timo Kopp, Konstantin Krutzke, Vincent Gidlund, Ferdinand Kollotzek, Julia Marzi, Saskia von Ungern-Sternberg, Antti Poso, Mathias Heikenwälder, Katja Schenke-Layland, Peter Seizer, Julia Möllmann, Nikolaus Marx, Robert Feil, Susanne Feil, Robert Lukowski, Oliver Borst, Tilman E Schäffer, Karin Anne Lydia Müller, Meinrad P Gawaz, David Heinzmann

Background: Cardiac hypertrophy is characterized by remodeling of the myocardium, which involves alterations in the ECM (extracellular matrix) and cardiomyocyte structure. These alterations critically contribute to impaired contractility and relaxation, ultimately leading to heart failure. Emerging evidence implicates that extracellular signaling molecules are critically involved in the pathogenesis of cardiac hypertrophy and remodeling. The immunophilin CyPA (cyclophilin A) has been identified as a potential culprit. In this study, we aimed to unravel the interplay between eCyPA (extracellular CyPA) and myocardial dysfunction and evaluate the therapeutic potential of inhibiting its extracellular accumulation to improve heart function.

Methods: Employing a multidisciplinary approach encompassing in silico, in vitro, in vivo, and ex vivo experiments we studied a mouse model of cardiac hypertrophy and human heart specimen to decipher the interaction of CyPA and the cardiac microenvironment in highly relevant pre-/clinical settings. Myocardial expression of CyPA (immunohistology) and the inflammatory transcriptome (NanoString) was analyzed in human cardiac tissue derived from patients with nonischemic, noninflammatory congestive heart failure (n=187). These analyses were paralleled by a mouse model of Ang (angiotensin) II-induced heart failure, which was assessed by functional (echocardiography), structural (immunohistology, atomic force microscopy), and biomolecular (Raman spectroscopy) analyses. The effect of inhibiting eCyPA in the cardiac microenvironment was evaluated using a newly developed neutralizing anti-eCyPA monoclonal antibody.

Results: We observed a significant accumulation of eCyPA in both human and murine-failing hearts. Importantly, higher eCyPA expression was associated with poor clinical outcomes in patients (P=0.043) and contractile dysfunction in mice (Pearson correlation coefficient, -0.73). Further, myocardial expression of eCyPA was critically associated with an increase in myocardial hypertrophy, inflammation, fibrosis, stiffness, and cardiac dysfunction in vivo. Antibody-based inhibition of eCyPA prevented (Ang II)-induced myocardial remodeling and dysfunction in mice.

Conclusions: Our study provides strong evidence of the pathogenic role of eCyPA in remodeling, myocardial stiffening, and dysfunction in heart failure. The findings suggest that antibody-based inhibition of eCyPA may offer a novel therapeutic strategy for nonischemic heart failure. Further research is needed to evaluate the translational potential of these interventions in human patients with cardiac hypertrophy.

背景:心肌肥大的特点是心肌重塑,其中涉及 ECM(细胞外基质)和心肌细胞结构的改变。这些改变严重影响了心肌的收缩和松弛功能,最终导致心力衰竭。新的证据表明,细胞外信号分子与心肌肥厚和重塑的发病机制密切相关。免疫噬菌素 CyPA(环噬菌素 A)已被确定为潜在的罪魁祸首。在这项研究中,我们旨在揭示 eCyPA(细胞外 CyPA)与心肌功能障碍之间的相互作用,并评估抑制其细胞外积累以改善心脏功能的治疗潜力:方法:我们采用了一种多学科方法,包括硅学、体外、体内和体外实验,研究了小鼠心脏肥大模型和人类心脏标本,以在高度相关的临床前/临床环境中破解 CyPA 与心脏微环境的相互作用。在非缺血性、非炎症性充血性心力衰竭患者(187 人)的人体心脏组织中分析了 CyPA 的心肌表达(免疫组织学)和炎症转录组(NanoString)。与这些分析同时进行的还有 Ang(血管紧张素)II 诱导的心力衰竭小鼠模型,该模型通过功能(超声心动图)、结构(免疫组织学、原子力显微镜)和生物分子(拉曼光谱)分析进行了评估。使用新开发的中和性抗 eCyPA 单克隆抗体评估了抑制心脏微环境中 eCyPA 的效果:结果:我们观察到 eCyPA 在人类和小鼠衰竭心脏中都有明显积累。重要的是,较高的 eCyPA 表达与患者的不良临床预后(P=0.043)和小鼠的收缩功能障碍(皮尔逊相关系数,-0.73)有关。此外,eCyPA 在心肌中的表达与心肌肥厚、炎症、纤维化、僵硬度和体内心脏功能障碍的增加密切相关。基于抗体的 eCyPA 抑制剂可防止(Ang II)诱导的小鼠心肌重塑和功能障碍:我们的研究为 eCyPA 在心衰的重塑、心肌僵化和功能障碍中的致病作用提供了有力证据。研究结果表明,基于抗体的 eCyPA 抑制可能为非缺血性心力衰竭提供一种新的治疗策略。要评估这些干预措施在人类心肌肥厚患者中的转化潜力,还需要进一步的研究。
{"title":"Targeting Cyclophilin A in the Cardiac Microenvironment Preserves Heart Function and Structure in Failing Hearts.","authors":"Manuel Sigle, Anne-Katrin Rohlfing, Melanie Cruz Santos, Timo Kopp, Konstantin Krutzke, Vincent Gidlund, Ferdinand Kollotzek, Julia Marzi, Saskia von Ungern-Sternberg, Antti Poso, Mathias Heikenwälder, Katja Schenke-Layland, Peter Seizer, Julia Möllmann, Nikolaus Marx, Robert Feil, Susanne Feil, Robert Lukowski, Oliver Borst, Tilman E Schäffer, Karin Anne Lydia Müller, Meinrad P Gawaz, David Heinzmann","doi":"10.1161/CIRCRESAHA.124.324812","DOIUrl":"10.1161/CIRCRESAHA.124.324812","url":null,"abstract":"<p><strong>Background: </strong>Cardiac hypertrophy is characterized by remodeling of the myocardium, which involves alterations in the ECM (extracellular matrix) and cardiomyocyte structure. These alterations critically contribute to impaired contractility and relaxation, ultimately leading to heart failure. Emerging evidence implicates that extracellular signaling molecules are critically involved in the pathogenesis of cardiac hypertrophy and remodeling. The immunophilin CyPA (cyclophilin A) has been identified as a potential culprit. In this study, we aimed to unravel the interplay between eCyPA (extracellular CyPA) and myocardial dysfunction and evaluate the therapeutic potential of inhibiting its extracellular accumulation to improve heart function.</p><p><strong>Methods: </strong>Employing a multidisciplinary approach encompassing in silico, in vitro, in vivo, and ex vivo experiments we studied a mouse model of cardiac hypertrophy and human heart specimen to decipher the interaction of CyPA and the cardiac microenvironment in highly relevant pre-/clinical settings. Myocardial expression of CyPA (immunohistology) and the inflammatory transcriptome (NanoString) was analyzed in human cardiac tissue derived from patients with nonischemic, noninflammatory congestive heart failure (n=187). These analyses were paralleled by a mouse model of Ang (angiotensin) II-induced heart failure, which was assessed by functional (echocardiography), structural (immunohistology, atomic force microscopy), and biomolecular (Raman spectroscopy) analyses. The effect of inhibiting eCyPA in the cardiac microenvironment was evaluated using a newly developed neutralizing anti-eCyPA monoclonal antibody.</p><p><strong>Results: </strong>We observed a significant accumulation of eCyPA in both human and murine-failing hearts. Importantly, higher eCyPA expression was associated with poor clinical outcomes in patients (<i>P</i>=0.043) and contractile dysfunction in mice (Pearson correlation coefficient, -0.73). Further, myocardial expression of eCyPA was critically associated with an increase in myocardial hypertrophy, inflammation, fibrosis, stiffness, and cardiac dysfunction in vivo. Antibody-based inhibition of eCyPA prevented (Ang II)-induced myocardial remodeling and dysfunction in mice.</p><p><strong>Conclusions: </strong>Our study provides strong evidence of the pathogenic role of eCyPA in remodeling, myocardial stiffening, and dysfunction in heart failure. The findings suggest that antibody-based inhibition of eCyPA may offer a novel therapeutic strategy for nonischemic heart failure. Further research is needed to evaluate the translational potential of these interventions in human patients with cardiac hypertrophy.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":"758-773"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141975201","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
Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. 微粒子介导的凋亡素递送可改善心肌梗死后小鼠的心功能
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-08-15 DOI: 10.1161/CIRCRESAHA.124.324608
Ling Tang, Huiliang Qiu, Bing Xu, Yajuan Su, Verah Nyarige, Pengsheng Li, Houjia Chen, Brady Killham, Jun Liao, Henderson Adam, Aaron Yang, Alexander Yu, Michelle Jang, Michael Rubart, Jingwei Xie, Wuqiang Zhu

Background: Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI).

Methods: A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot.

Results: The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression.

Conclusions: Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.

背景:凋亡素是一种内源性前肽,可调节心脏稳态和各种生理过程。研究表明,静脉注射可改善心力衰竭患者的心脏收缩力。然而,由于其半衰期较短,因此无法研究其对左心室重塑的长期影响。在此,我们旨在研究微颗粒介导的凋亡素缓释是否能改善心肌梗死(MI)小鼠的心脏功能和左心室重塑:方法:将含凋亡素的微颗粒嵌入纤维蛋白凝胶支架中,制成心脏补片。通过永久结扎成年 C57BL/6J 小鼠的左前降支冠状动脉诱发心肌梗死,然后在心肌梗死后立即(急性心肌梗死)或 28 天(慢性心肌梗死)放置心外膜贴片。这项研究包括四组,即假性心肌梗死、心肌梗死加空微粒包埋贴片处理和心肌梗死加含凋亡素微粒包埋贴片处理。心功能通过经胸超声心动图进行评估。心肌细胞形态、凋亡和心脏纤维化通过组织学进行评估。通过 RNA 测序、定量聚合酶链式反应和 Western 印迹确定心脏保护途径:结果:内源性凋亡素的水平在诱导心肌梗死后的前 7 天大幅降低,到第 28 天已恢复正常。包裹在聚(乳酸-共聚-乙醇酸)微粒中的凋亡磷-13可持续释放长达28天。在急性和慢性心肌梗死模型中,使用含有凋亡素的微粒包埋贴片可抑制心脏肥大,缩小瘢痕大小,从而改善心脏功能。细胞和分子分析数据显示,芹菜素通过阻止转化生长因子-β介导的Smad2/3活化和下游坏死基因表达,抑制了心脏成纤维细胞的活化和增殖:结论:聚乳酸-共聚乙醇酸微粒延长了凋亡磷在小鼠心脏中的释放时间。心外膜输送含凋亡素的微颗粒包埋贴片可保护小鼠免受急性和慢性心肌梗死诱发的心功能障碍的影响,抑制心脏纤维化并改善左心室重塑。
{"title":"Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice.","authors":"Ling Tang, Huiliang Qiu, Bing Xu, Yajuan Su, Verah Nyarige, Pengsheng Li, Houjia Chen, Brady Killham, Jun Liao, Henderson Adam, Aaron Yang, Alexander Yu, Michelle Jang, Michael Rubart, Jingwei Xie, Wuqiang Zhu","doi":"10.1161/CIRCRESAHA.124.324608","DOIUrl":"10.1161/CIRCRESAHA.124.324608","url":null,"abstract":"<p><strong>Background: </strong>Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI).</p><p><strong>Methods: </strong>A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot.</p><p><strong>Results: </strong>The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression.</p><p><strong>Conclusions: </strong>Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":"777-798"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392624/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141981845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Oxysterol Sensing Through GPR183 Triggers Endothelial Senescence in Hypertension. 通过 GPR183 感知氧杂环醇引发高血压的内皮衰老
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-08-23 DOI: 10.1161/CIRCRESAHA.124.324722
Qingqing Chu, Yujia Li, Jichao Wu, Yanjiao Gao, Xiangyun Guo, Jing Li, Hang Lv, Min Liu, Wei Tang, Peng Zhan, Tao Zhang, Huili Hu, Hong Liu, Jinpeng Sun, Xiaojie Wang, Fan Yi

Background: Despite endothelial dysfunction being an initial step in the development of hypertension and associated cardiovascular/renal injuries, effective therapeutic strategies to prevent endothelial dysfunction are still lacking. GPR183 (G protein-coupled receptor 183), a recently identified G protein-coupled receptor for oxysterols and hydroxylated metabolites of cholesterol, has pleiotropic roles in lipid metabolism and immune responses. However, the role of GPR183 in the regulation of endothelial function remains unknown.

Methods: Endothelial-specific GPR183 knockout mice were generated and used to examine the role of GPR183 in endothelial senescence by establishing 2 independent hypertension models: desoxycorticosterone acetate/salt-induced and Ang II (angiotensin II)-induced hypertensive mice. Echocardiography, transmission electron microscopy, blood pressure measurement, vasorelaxation response experiments, flow cytometry analysis, and chromatin immunoprecipitation analysis were performed in this study.

Results: Endothelial GPR183 was significantly induced in hypertensive mice, which was further confirmed in renal biopsies from subjects with hypertensive nephropathy. Endothelial-specific deficiency of GPR183 markedly alleviated cardiovascular and renal injuries in hypertensive mice. Moreover, we found that GPR183 regulated endothelial senescence in both hypertensive mice and aged mice. Mechanistically, GPR183 disrupted circadian signaling by inhibiting PER1 (period circadian regulator 1) expression, thereby facilitating endothelial senescence and dysfunction through the cAMP (cyclic adenosine monophosphate)/PKA (protein kinase A)/CREB (cAMP-response element binding protein) signaling pathway. Importantly, pharmacological inhibition of the oxysterol-GPR183 axis by NIBR189 or clotrimazole ameliorated endothelial senescence and cardiovascular/renal injuries in hypertensive mice.

Conclusions: This study discovers a previously unrecognized role of GPR183 in promoting endothelial senescence. Pharmacological targeting of GPR183 may be an innovative therapeutic strategy for hypertension and its associated complications.

背景:尽管内皮功能障碍是高血压和相关心血管/肾损伤发生的第一步,但目前仍缺乏预防内皮功能障碍的有效治疗策略。GPR183(G蛋白偶联受体183)是最近发现的一种G蛋白偶联受体,用于氧化甾醇和胆固醇的羟化代谢产物,在脂质代谢和免疫反应中具有多方面的作用。然而,GPR183 在调节内皮功能方面的作用仍然未知:方法:通过建立两个独立的高血压模型:醋酸去氧皮质酮/盐诱导的高血压小鼠和血管紧张素 II(Ang II)诱导的高血压小鼠,产生了内皮特异性 GPR183 基因敲除小鼠,用于研究 GPR183 在内皮衰老中的作用。该研究进行了超声心动图、透射电子显微镜、血压测量、血管舒张反应实验、流式细胞仪分析和染色质免疫沉淀分析:结果:高血压小鼠的内皮细胞 GPR183 被显著诱导,这在高血压肾病患者的肾活检中得到了进一步证实。内皮特异性 GPR183 的缺乏明显减轻了高血压小鼠的心血管和肾脏损伤。此外,我们还发现 GPR183 可调节高血压小鼠和老龄小鼠的内皮衰老。从机制上讲,GPR183 通过抑制 PER1(周期 1)的表达破坏了昼夜节律信号传导,从而通过 cAMP/PKA(蛋白激酶 A)/CREB(cAMP-反应元件结合蛋白)信号传导途径促进了内皮衰老和功能障碍。重要的是,NIBR189或克霉唑对氧杂环醇-GPR183轴的药理抑制可改善高血压小鼠的内皮衰老和心血管/肾损伤:本研究发现了 GPR183 在促进内皮衰老中的作用,而这一作用此前尚未被认识。以 GPR183 为药理靶点可能是治疗高血压及其相关并发症的一种创新策略。
{"title":"Oxysterol Sensing Through GPR183 Triggers Endothelial Senescence in Hypertension.","authors":"Qingqing Chu, Yujia Li, Jichao Wu, Yanjiao Gao, Xiangyun Guo, Jing Li, Hang Lv, Min Liu, Wei Tang, Peng Zhan, Tao Zhang, Huili Hu, Hong Liu, Jinpeng Sun, Xiaojie Wang, Fan Yi","doi":"10.1161/CIRCRESAHA.124.324722","DOIUrl":"10.1161/CIRCRESAHA.124.324722","url":null,"abstract":"<p><strong>Background: </strong>Despite endothelial dysfunction being an initial step in the development of hypertension and associated cardiovascular/renal injuries, effective therapeutic strategies to prevent endothelial dysfunction are still lacking. GPR183 (G protein-coupled receptor 183), a recently identified G protein-coupled receptor for oxysterols and hydroxylated metabolites of cholesterol, has pleiotropic roles in lipid metabolism and immune responses. However, the role of GPR183 in the regulation of endothelial function remains unknown.</p><p><strong>Methods: </strong>Endothelial-specific GPR183 knockout mice were generated and used to examine the role of GPR183 in endothelial senescence by establishing 2 independent hypertension models: desoxycorticosterone acetate/salt-induced and Ang II (angiotensin II)-induced hypertensive mice. Echocardiography, transmission electron microscopy, blood pressure measurement, vasorelaxation response experiments, flow cytometry analysis, and chromatin immunoprecipitation analysis were performed in this study.</p><p><strong>Results: </strong>Endothelial GPR183 was significantly induced in hypertensive mice, which was further confirmed in renal biopsies from subjects with hypertensive nephropathy. Endothelial-specific deficiency of GPR183 markedly alleviated cardiovascular and renal injuries in hypertensive mice. Moreover, we found that GPR183 regulated endothelial senescence in both hypertensive mice and aged mice. Mechanistically, GPR183 disrupted circadian signaling by inhibiting PER1 (period circadian regulator 1) expression, thereby facilitating endothelial senescence and dysfunction through the cAMP (cyclic adenosine monophosphate)/PKA (protein kinase A)/CREB (cAMP-response element binding protein) signaling pathway. Importantly, pharmacological inhibition of the oxysterol-GPR183 axis by NIBR189 or clotrimazole ameliorated endothelial senescence and cardiovascular/renal injuries in hypertensive mice.</p><p><strong>Conclusions: </strong>This study discovers a previously unrecognized role of GPR183 in promoting endothelial senescence. Pharmacological targeting of GPR183 may be an innovative therapeutic strategy for hypertension and its associated complications.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":"708-721"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142035419","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
Restoring Atrial T-Tubules Augments Systolic Ca Upon Recovery From Heart Failure. 恢复心房 T 型微管可增强心力衰竭患者恢复后的收缩压。
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-08-14 DOI: 10.1161/CIRCRESAHA.124.324601
Jessica L Caldwell, Jessica D Clarke, Charlotte E R Smith, Christian Pinali, Callum J Quinn, Charles M Pearman, Aiste Adomaviciene, Emma J Radcliffe, Amy Watkins, Margaux A Horn, Elizabeth F Bode, George W P Madders, Mark Eisner, David A Eisner, Andrew W Trafford, Katharine M Dibb

Background: Transverse (t)-tubules drive the rapid and synchronous Ca2+ rise in cardiac myocytes. The virtual complete atrial t-tubule loss in heart failure (HF) decreases Ca2+ release. It is unknown if or how atrial t-tubules can be restored and how this affects systolic Ca2+.

Methods: HF was induced in sheep by rapid ventricular pacing and recovered following termination of rapid pacing. Serial block-face scanning electron microscopy and confocal imaging were used to study t-tubule ultrastructure. Function was assessed using patch clamp, Ca2+, and confocal imaging. Candidate proteins involved in atrial t-tubule recovery were identified by western blot and expressed in rat neonatal ventricular myocytes to determine if they altered t-tubule structure.

Results: Atrial t-tubules were lost in HF but reappeared following recovery from HF. Recovered t-tubules were disordered, adopting distinct morphologies with increased t-tubule length and branching. T-tubule disorder was associated with mitochondrial disorder. Recovered t-tubules were functional, triggering Ca2+ release in the cell interior. Systolic Ca2+, ICa-L, sarcoplasmic reticulum Ca2+ content, and sarcoendoplasmic reticulum Ca2+ ATPase function were restored following recovery from HF. Confocal microscopy showed fragmentation of ryanodine receptor staining and movement away from the z-line in HF, which was reversed following recovery from HF. Acute detubulation, to remove recovered t-tubules, confirmed their key role in restoration of the systolic Ca2+ transient, the rate of Ca2+ removal, and the peak L-type Ca2+ current. The abundance of telethonin and myotubularin decreased during HF and increased during recovery. Transfection with these proteins altered the density and structure of tubules in neonatal myocytes. Myotubularin had a greater effect, increasing tubule length and branching, replicating that seen in the recovery atria.

Conclusions: We show that recovery from HF restores atrial t-tubules, and this promotes recovery of ICa-L, sarcoplasmic reticulum Ca2+ content, and systolic Ca2+. We demonstrate an important role for myotubularin in t-tubule restoration. Our findings reveal a new and viable therapeutic strategy.

背景:横向(t)微管驱动心肌细胞中 Ca2+ 快速同步上升。心力衰竭(HF)患者实际上完全丧失了心房的 t 型微管,从而减少了 Ca2+ 的释放。方法:通过快速心室起搏诱发绵羊心力衰竭,并在快速起搏终止后恢复。使用序列块面扫描电子显微镜和共聚焦成像技术研究 t 型微管的超微结构。使用膜片钳、Ca2+和共聚焦成像技术评估其功能。通过 Western 印迹鉴定了参与心房 t 型微管恢复的候选蛋白,并在大鼠新生心室肌细胞中表达了这些蛋白,以确定它们是否改变了 t 型微管的结构:结果:心房颤动导致心房t-微管缺失,但在心房颤动恢复后又重新出现。恢复后的t-微管结构紊乱,形态各异,t-微管长度和分支增加。t-微管紊乱与线粒体紊乱有关。恢复的t-微管具有功能性,可触发细胞内部的Ca2+释放。高频恢复后,收缩期Ca2+、ICa-L、肌浆网Ca2+含量和SERCA功能均得到恢复。共聚焦显微镜显示,在高房颤动中,雷诺丁受体染色破碎并偏离z线,而在高房颤动恢复后,这种情况被逆转。急性去管法去除恢复的t-管,证实了它们在恢复收缩期Ca2+瞬态、Ca2+去除率和L型Ca2+电流峰值中的关键作用。telethonin 和 myotubularin 的丰度在高频过程中降低,而在恢复过程中升高。转染这些蛋白后,新生儿心肌细胞小管的密度和结构发生了改变。Myotubularin的作用更大,它增加了小管的长度和分支,复制了在恢复期心房中看到的情况:我们的研究表明,心房颤动的恢复可恢复心房的 t 型小管,从而促进 ICa-L、肌质网 Ca2+ 含量和收缩期 Ca2+ 的恢复。我们证明了肌球蛋白在微管恢复中的重要作用。我们的发现揭示了一种新的可行治疗策略。
{"title":"Restoring Atrial T-Tubules Augments Systolic Ca Upon Recovery From Heart Failure.","authors":"Jessica L Caldwell, Jessica D Clarke, Charlotte E R Smith, Christian Pinali, Callum J Quinn, Charles M Pearman, Aiste Adomaviciene, Emma J Radcliffe, Amy Watkins, Margaux A Horn, Elizabeth F Bode, George W P Madders, Mark Eisner, David A Eisner, Andrew W Trafford, Katharine M Dibb","doi":"10.1161/CIRCRESAHA.124.324601","DOIUrl":"10.1161/CIRCRESAHA.124.324601","url":null,"abstract":"<p><strong>Background: </strong>Transverse (t)-tubules drive the rapid and synchronous Ca<sup>2+</sup> rise in cardiac myocytes. The virtual complete atrial t-tubule loss in heart failure (HF) decreases Ca<sup>2+</sup> release. It is unknown if or how atrial t-tubules can be restored and how this affects systolic Ca<sup>2+</sup>.</p><p><strong>Methods: </strong>HF was induced in sheep by rapid ventricular pacing and recovered following termination of rapid pacing. Serial block-face scanning electron microscopy and confocal imaging were used to study t-tubule ultrastructure. Function was assessed using patch clamp, Ca<sup>2+</sup>, and confocal imaging. Candidate proteins involved in atrial t-tubule recovery were identified by western blot and expressed in rat neonatal ventricular myocytes to determine if they altered t-tubule structure.</p><p><strong>Results: </strong>Atrial t-tubules were lost in HF but reappeared following recovery from HF. Recovered t-tubules were disordered, adopting distinct morphologies with increased t-tubule length and branching. T-tubule disorder was associated with mitochondrial disorder. Recovered t-tubules were functional, triggering Ca<sup>2+</sup> release in the cell interior. Systolic Ca<sup>2+</sup>, <i>I</i><sub>Ca-L</sub>, sarcoplasmic reticulum Ca<sup>2+</sup> content, and sarcoendoplasmic reticulum Ca<sup>2+</sup> ATPase function were restored following recovery from HF. Confocal microscopy showed fragmentation of ryanodine receptor staining and movement away from the z-line in HF, which was reversed following recovery from HF. Acute detubulation, to remove recovered t-tubules, confirmed their key role in restoration of the systolic Ca<sup>2+</sup> transient, the rate of Ca<sup>2+</sup> removal, and the peak L-type Ca<sup>2+</sup> current. The abundance of telethonin and myotubularin decreased during HF and increased during recovery. Transfection with these proteins altered the density and structure of tubules in neonatal myocytes. Myotubularin had a greater effect, increasing tubule length and branching, replicating that seen in the recovery atria.</p><p><strong>Conclusions: </strong>We show that recovery from HF restores atrial t-tubules, and this promotes recovery of <i>I</i><sub>Ca-L</sub>, sarcoplasmic reticulum Ca<sup>2+</sup> content, and systolic Ca<sup>2+</sup>. We demonstrate an important role for myotubularin in t-tubule restoration. Our findings reveal a new and viable therapeutic strategy.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":"739-754"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392124/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141975200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Targeting the IKs Channel PKA Phosphorylation Axis to Restore Its Function in High-Risk LQT1 Variants. 以 IKs 通道 PKA 磷酸化轴为靶点恢复高风险 LQT1 变异的功能
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-08-21 DOI: 10.1161/CIRCRESAHA.124.325009
Ling Zhong, Zhenzhen Yan, Dexiang Jiang, Kuo-Chan Weng, Yue Ouyang, Hangyu Zhang, Xiaoqing Lin, Chenxin Xiao, Huaiyu Yang, Jing Yao, Xinjiang Kang, Changhe Wang, Chen Huang, Bing Shen, Sookja Kim Chung, Zhi-Hong Jiang, Wandi Zhu, Erwin Neher, Jonathan R Silva, Panpan Hou

Background: The KCNQ1+KCNE1 (IKs) potassium channel plays a crucial role in cardiac adaptation to stress, in which β-adrenergic stimulation phosphorylates the IKs channel through the cyclic adenosine monophosphate (cAMP)/PKA (protein kinase A) pathway. Phosphorylation increases the channel current and accelerates repolarization to adapt to an increased heart rate. Variants in KCNQ1 can cause long-QT syndrome type 1 (LQT1), and those with defective cAMP effects predispose patients to the highest risk of cardiac arrest and sudden death. However, the molecular connection between IKs channel phosphorylation and channel function, as well as why high-risk LQT1 mutations lose cAMP sensitivity, remain unclear.

Methods: Regular patch clamp and voltage clamp fluorometry techniques were utilized to record pore opening and voltage sensor movement of wild-type and mutant KCNQ1/IKs channels. The clinical phenotypic penetrance of each LQT1 mutation was analyzed as a metric for assessing their clinical risk. The patient-specific-induced pluripotent stem-cell model was used to test mechanistic findings in physiological conditions.

Results: By systematically elucidating mechanisms of a series of LQT1 variants that lack cAMP sensitivity, we identified molecular determinants of IKs channel regulation by phosphorylation. These key residues are distributed across the N-terminus of KCNQ1 extending to the central pore region of IKs. We refer to this pattern as the IKs channel PKA phosphorylation axis. Next, by examining LQT1 variants from clinical databases containing 10 579 LQT1 carriers, we found that the distribution of the most high-penetrance LQT1 variants extends across the IKs channel PKA phosphorylation axis, demonstrating its clinical relevance. Furthermore, we found that a small molecule, ML277, which binds at the center of the phosphorylation axis, rescues the defective cAMP effects of multiple high-risk LQT1 variants. This finding was then tested in high-risk patient-specific induced pluripotent stem cell-derived cardiomyocytes, where ML277 remarkably alleviates the beating abnormalities.

Conclusions: Our findings not only elucidate the molecular mechanism of PKA-dependent IKs channel phosphorylation but also provide an effective antiarrhythmic strategy for patients with high-risk LQT1 variants.

背景:KCNQ1+KCNE1(IKs)钾通道在心脏适应应激过程中起着至关重要的作用,β-肾上腺素能刺激通过环磷酸腺苷(cAMP)/PKA(蛋白激酶 A)途径使 IKs 通道磷酸化。磷酸化会增加通道电流并加速再极化,以适应心率的增加。KCNQ1 的变异可导致 1 型长 QT 综合征(LQT1),而具有 cAMP 作用缺陷的患者发生心脏骤停和猝死的风险最高。然而,IKs 通道磷酸化与通道功能之间的分子联系以及高风险 LQT1 突变失去 cAMP 敏感性的原因仍不清楚:方法:利用常规膜片钳和电压钳荧光测定技术记录野生型和突变型 KCNQ1/IKs 通道的孔开放和电压传感器运动。分析了每种 LQT1 突变的临床表型穿透性,以此作为评估其临床风险的指标。患者特异性诱导多能干细胞模型用于在生理条件下检验机理发现:结果:通过系统阐明一系列缺乏 cAMP 敏感性的 LQT1 变异的机制,我们确定了通过磷酸化调控 IKs 通道的分子决定因素。这些关键残基分布在 KCNQ1 的 N 端,一直延伸到 IKs 的中心孔区域。我们将这种模式称为 IKs 通道 PKA 磷酸化轴。接下来,通过研究临床数据库中包含的 10 579 个 LQT1 基因携带者的 LQT1 变异,我们发现大多数高隐匿性 LQT1 变异的分布横跨 IKs 通道 PKA 磷酸化轴,证明了其临床相关性。此外,我们还发现,一种结合在磷酸化轴中心的小分子 ML277 能挽救多种高风险 LQT1 变异的 cAMP 效应缺陷。这一发现随后在高风险患者特异性诱导多能干细胞衍生的心肌细胞中进行了测试,ML277显著缓解了心肌细胞的跳动异常:我们的研究结果不仅阐明了 PKA 依赖性 IKs 通道磷酸化的分子机制,还为高危 LQT1 变异患者提供了有效的抗心律失常策略。
{"title":"Targeting the I<sub>Ks</sub> Channel PKA Phosphorylation Axis to Restore Its Function in High-Risk LQT1 Variants.","authors":"Ling Zhong, Zhenzhen Yan, Dexiang Jiang, Kuo-Chan Weng, Yue Ouyang, Hangyu Zhang, Xiaoqing Lin, Chenxin Xiao, Huaiyu Yang, Jing Yao, Xinjiang Kang, Changhe Wang, Chen Huang, Bing Shen, Sookja Kim Chung, Zhi-Hong Jiang, Wandi Zhu, Erwin Neher, Jonathan R Silva, Panpan Hou","doi":"10.1161/CIRCRESAHA.124.325009","DOIUrl":"10.1161/CIRCRESAHA.124.325009","url":null,"abstract":"<p><strong>Background: </strong>The KCNQ1+KCNE1 (I<sub>Ks</sub>) potassium channel plays a crucial role in cardiac adaptation to stress, in which β-adrenergic stimulation phosphorylates the I<sub>Ks</sub> channel through the cyclic adenosine monophosphate (cAMP)/PKA (protein kinase A) pathway. Phosphorylation increases the channel current and accelerates repolarization to adapt to an increased heart rate. Variants in KCNQ1 can cause long-QT syndrome type 1 (LQT1), and those with defective cAMP effects predispose patients to the highest risk of cardiac arrest and sudden death. However, the molecular connection between I<sub>Ks</sub> channel phosphorylation and channel function, as well as why high-risk LQT1 mutations lose cAMP sensitivity, remain unclear.</p><p><strong>Methods: </strong>Regular patch clamp and voltage clamp fluorometry techniques were utilized to record pore opening and voltage sensor movement of wild-type and mutant KCNQ1/I<sub>Ks</sub> channels. The clinical phenotypic penetrance of each LQT1 mutation was analyzed as a metric for assessing their clinical risk. The patient-specific-induced pluripotent stem-cell model was used to test mechanistic findings in physiological conditions.</p><p><strong>Results: </strong>By systematically elucidating mechanisms of a series of LQT1 variants that lack cAMP sensitivity, we identified molecular determinants of I<sub>Ks</sub> channel regulation by phosphorylation. These key residues are distributed across the N-terminus of KCNQ1 extending to the central pore region of I<sub>Ks</sub>. We refer to this pattern as the I<sub>Ks</sub> channel PKA phosphorylation axis. Next, by examining LQT1 variants from clinical databases containing 10 579 LQT1 carriers, we found that the distribution of the most high-penetrance LQT1 variants extends across the I<sub>Ks</sub> channel PKA phosphorylation axis, demonstrating its clinical relevance. Furthermore, we found that a small molecule, ML277, which binds at the center of the phosphorylation axis, rescues the defective cAMP effects of multiple high-risk LQT1 variants. This finding was then tested in high-risk patient-specific induced pluripotent stem cell-derived cardiomyocytes, where ML277 remarkably alleviates the beating abnormalities.</p><p><strong>Conclusions: </strong>Our findings not only elucidate the molecular mechanism of PKA-dependent I<sub>Ks</sub> channel phosphorylation but also provide an effective antiarrhythmic strategy for patients with high-risk LQT1 variants.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":"722-738"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392204/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008352","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
In This Issue. 本期内容
IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2024-09-13 Epub Date: 2024-09-12 DOI: 10.1161/RES.0000000000000691
{"title":"In This Issue.","authors":"","doi":"10.1161/RES.0000000000000691","DOIUrl":"https://doi.org/10.1161/RES.0000000000000691","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"135 7","pages":"705"},"PeriodicalIF":16.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142307225","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
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":"52 1","pages":""},"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":"3 1","pages":"755-757"},"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":"4 1","pages":"774-776"},"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
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