首页 > 最新文献

Circulation research最新文献

英文 中文
CPT1a Expression Is a Critical Cardioprotective Response to Pathological Stress That Enables Rescue by Gene Transfer. CPT1a表达的增加是对病理性应激的关键心脏保护反应,抑制重塑基因程序并通过基因转移实现拯救。
IF 16.2 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-16 Epub Date: 2025-12-04 DOI: 10.1161/CIRCRESAHA.125.327403
Andrew N Carley, Santosh K Maurya, Chandan K Maurya, Yang Wang, Amy Webb, Azariyas A Challa, Tatiana Gromova, Thomas M Vondriska, Zhentao Zhang, Hua Zhu, Ahlke Heydemann, Kenneth C Bedi, Christos P Kyriakopoulos, Craig H Selzman, Stavros G Drakos, Kenneth B Margulies, E Douglas Lewandowski

Background: CPT1 (carnitine palmitoyltransferase 1) is a rate-limiting enzyme for long-chain fatty acid oxidation. In adult hearts, CPT1b predominates, while CPT1a is coexpressed at lower levels. Pathological stress on the heart induces CPT1a expression, coinciding with a reduction in fatty acid oxidation, yet the role of CPT1a in pathological remodeling is unknown.

Methods: CPT1 isoform expression was assayed in the myocardium of patients with heart failure with nonischemic cardiomyopathy and a preclinical mouse model of heart failure. Mice were subjected to afterload stress via transverse aortic constriction (TAC) or sham surgery (sham) with cardiac-specific CPT1a knockdown or cardiac-specific, adeno-associated virus serotype 9 (AAV9)-mediated CPT1a overexpression (AAV9.cTnT [cardiac troponin T].Cpt1a) versus empty virus or PBS infusions as controls. MicroRNA 370, known to suppress hepatic CPT1a, was assayed and overexpressed to determine if microRNA 370 regulates cardiac CPT1a expression.

Results: CPT1a protein was elevated and microRNA 370 reduced in the myocardium of male and female patients with nonischemic cardiomyopathy, as well as in failing mouse hearts. AAV9-mediated microRNA 370 overexpression in mouse hearts suppressed CPT1a expression and attenuated the response of CPT1a to TAC. Preventing CPT1a upregulation in response to TAC in cardiac-specific CPT1a knockout mice exacerbated adverse remodeling, severe dysfunction, and increased mortality. In contrast, CPT1a overexpression (2.8-fold) attenuated impaired ejection fraction (by 54%) versus control TAC hearts (P<0.05). Delivery of AAV9.cTnT.Cpt1a 4 weeks after TAC surgery led to significant rescue of ejection fraction and mitigated the exacerbated dysfunction of cardiac-specific CPT1a knockout mice TAC hearts. RNA-seq revealed a novel function of CPT1a in suppressing hypertrophic, profibrotic, and cell death gene programs in both sham and TAC hearts, irrespective of changes in fatty acid oxidation, with reduced histone acetylation.

Conclusions: The effects of CPT1a in the heart extend beyond fatty acid oxidation including noncanonical regulation of gene programs. CPT1a upregulation occurs in nonischemic cardiomyopathy and is a critical cardioprotective adaptation to pathological stress.

背景:CPT1(肉碱棕榈酰转移酶1)是长链脂肪酸氧化的限速酶。在成人心脏中,CPT1b占主导地位,而CPT1a在较低水平上共表达。心脏的病理性应激诱导CPT1a表达,与脂肪酸氧化减少相一致,但CPT1a在病理性重塑中的作用尚不清楚。方法:检测CPT1亚型在心力衰竭合并非缺血性心肌病患者和临床前心力衰竭小鼠心肌中的表达。通过主动脉横缩术(TAC)或假手术(sham),小鼠受到心脏特异性CPT1a敲低或心脏特异性、腺相关病毒血清型9介导的CPT1a过表达(腺相关病毒血清型9)的负荷后应激。心肌肌钙蛋白T。Cpt1a)对照空病毒或PBS输注作为对照。已知抑制肝脏CPT1a的MicroRNA 370被检测并过表达,以确定MicroRNA 370是否调节心脏CPT1a的表达。结果:在男性和女性非缺血性心肌病患者以及衰竭小鼠心肌中,CPT1a蛋白升高,microRNA 370减少。腺相关病毒介导的microRNA 370在小鼠心脏中的过表达抑制了CPT1a的表达,并减弱了CPT1a对TAC的反应。在心脏特异性CPT1a敲除小鼠中,防止CPT1a对TAC的上调会加剧不良重构、严重功能障碍和死亡率增加。相比之下,与对照组相比,CPT1a过表达(2.8倍)减少了54%的射血分数(p结论:CPT1a在心脏中的作用超出了脂肪酸氧化,包括基因程序的非规范调节。CPT1a上调发生在非缺血性心肌病中,是对病理性应激的关键心脏保护适应。
{"title":"CPT1a Expression Is a Critical Cardioprotective Response to Pathological Stress That Enables Rescue by Gene Transfer.","authors":"Andrew N Carley, Santosh K Maurya, Chandan K Maurya, Yang Wang, Amy Webb, Azariyas A Challa, Tatiana Gromova, Thomas M Vondriska, Zhentao Zhang, Hua Zhu, Ahlke Heydemann, Kenneth C Bedi, Christos P Kyriakopoulos, Craig H Selzman, Stavros G Drakos, Kenneth B Margulies, E Douglas Lewandowski","doi":"10.1161/CIRCRESAHA.125.327403","DOIUrl":"10.1161/CIRCRESAHA.125.327403","url":null,"abstract":"<p><strong>Background: </strong>CPT1 (carnitine palmitoyltransferase 1) is a rate-limiting enzyme for long-chain fatty acid oxidation. In adult hearts, CPT1b predominates, while CPT1a is coexpressed at lower levels. Pathological stress on the heart induces CPT1a expression, coinciding with a reduction in fatty acid oxidation, yet the role of CPT1a in pathological remodeling is unknown.</p><p><strong>Methods: </strong>CPT1 isoform expression was assayed in the myocardium of patients with heart failure with nonischemic cardiomyopathy and a preclinical mouse model of heart failure. Mice were subjected to afterload stress via transverse aortic constriction (TAC) or sham surgery (sham) with cardiac-specific CPT1a knockdown or cardiac-specific, adeno-associated virus serotype 9 (AAV9)-mediated CPT1a overexpression (AAV9.cTnT [cardiac troponin T].Cpt1a) versus empty virus or PBS infusions as controls. MicroRNA 370, known to suppress hepatic CPT1a, was assayed and overexpressed to determine if microRNA 370 regulates cardiac CPT1a expression.</p><p><strong>Results: </strong>CPT1a protein was elevated and microRNA 370 reduced in the myocardium of male and female patients with nonischemic cardiomyopathy, as well as in failing mouse hearts. AAV9-mediated microRNA 370 overexpression in mouse hearts suppressed CPT1a expression and attenuated the response of CPT1a to TAC. Preventing CPT1a upregulation in response to TAC in cardiac-specific CPT1a knockout mice exacerbated adverse remodeling, severe dysfunction, and increased mortality. In contrast, CPT1a overexpression (2.8-fold) attenuated impaired ejection fraction (by 54%) versus control TAC hearts (<i>P</i><0.05). Delivery of AAV9.cTnT.Cpt1a 4 weeks after TAC surgery led to significant rescue of ejection fraction and mitigated the exacerbated dysfunction of cardiac-specific CPT1a knockout mice TAC hearts. RNA-seq revealed a novel function of CPT1a in suppressing hypertrophic, profibrotic, and cell death gene programs in both sham and TAC hearts, irrespective of changes in fatty acid oxidation, with reduced histone acetylation.</p><p><strong>Conclusions: </strong>The effects of CPT1a in the heart extend beyond fatty acid oxidation including noncanonical regulation of gene programs. CPT1a upregulation occurs in nonischemic cardiomyopathy and is a critical cardioprotective adaptation to pathological stress.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":"e327403"},"PeriodicalIF":16.2,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145667357","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
Meet the First Authors. 认识第一作者。
IF 16.2 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-16 Epub Date: 2026-01-15 DOI: 10.1161/RES.0000000000000744
{"title":"Meet the First Authors.","authors":"","doi":"10.1161/RES.0000000000000744","DOIUrl":"https://doi.org/10.1161/RES.0000000000000744","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"138 2","pages":"e000744"},"PeriodicalIF":16.2,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145984439","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
Fatty Acid Transport at the Heart of Metabolic Adaptation. 脂肪酸转运在代谢适应中的核心作用。
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-15 DOI: 10.1161/circresaha.125.327929
Anja Karlstaedt
{"title":"Fatty Acid Transport at the Heart of Metabolic Adaptation.","authors":"Anja Karlstaedt","doi":"10.1161/circresaha.125.327929","DOIUrl":"https://doi.org/10.1161/circresaha.125.327929","url":null,"abstract":"","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"10 1","pages":"e327929"},"PeriodicalIF":20.1,"publicationDate":"2026-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971795","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
Grhl3 Downregulation Facilitates ECM Adaptation for Fibroblast to iCM Commitment. Grhl3下调促进成纤维细胞向iCM承诺的ECM适应
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-15 DOI: 10.1161/circresaha.125.327726
Xin Wu,Lanbing Liu,Yuanru Huang,Yi Ling,Fang Luo,Dongyu Gu,Mengxin Liu,Zhenhua Jia,Zhangyi Yu,Xiangjie Kong,Hong Ma,Yanggan Wang,Li Wang
BACKGROUNDDirect cardiac reprogramming offers a promising therapeutic strategy for heart regeneration by converting endogenous fibroblasts to functional induced cardiomyocytes (iCMs) that integrate into the myocardium to restore heart structure and function. While ECM (extracellular matrix) plays critical roles in cardiac disease and repair, the dynamic changes and transcriptional regulation underlying ECM remodeling during reprogramming remain poorly understood.METHODSWe investigated ECM dynamics during iCM reprogramming using integrated transcriptomic, proteomic, and epigenetic analyses, focusing on cell type-specific ECM components. A loss-of-function screen was used to identify critical ECM components and regulators, including Itga8 (integrin alpha-8) and Grhl3 (grainyhead-like protein 3 homolog), respectively, as reprogramming barriers. Mechanistic studies integrated RNA sequencing, mass spectrometry, and Cleavage Under Targets and Tagmentation to define Grhl3-dependent regulation. Functional outcomes were evaluated in vitro using decellularized ECM and in vivo using a myocardial infarction model with genetic lineage tracing.RESULTSCardiac reprogramming induced dynamic ECM remodeling, with significant changes in collagen, fibrillar proteins, and integrins. Itga8 was identified as a pivotal ECM component that restricts iCM conversion via the TGF-β (transforming growth factor-β)/SMAD pathway. Grhl3 emerged as a key transcriptional regulator for ECM components, including Itga8. ECM derived from Grhl3-deficient fibroblasts enhanced iCM induction, while Grhl3 depletion also reduced fibroblast activation and increased cellular plasticity. These effects synergized with TF (transcription factor)-mediated reprogramming to improve iCM efficiency, structural organization, and functional maturation. In vivo, removing Grhl3 enhanced fibroblast-to-cardiomyocyte conversion, reduced scar formation, and improved cardiac function after myocardial infarction.CONCLUSIONSOur findings establish ECM adaptation as a critical determinant of cardiac reprogramming and identify Grhl3 as a promising therapeutic target to advance myocardial repair strategies.
背景:直接心脏重编程为心脏再生提供了一种很有前景的治疗策略,通过将内源性成纤维细胞转化为功能诱导心肌细胞(iCMs),并整合到心肌中以恢复心脏结构和功能。虽然ECM(细胞外基质)在心脏疾病和修复中起着关键作用,但在重编程过程中ECM重塑的动态变化和转录调控仍然知之甚少。方法我们使用整合转录组学、蛋白质组学和表观遗传学分析来研究iCM重编程过程中的ECM动力学,重点研究细胞类型特异性ECM成分。功能丧失筛选用于鉴定关键的ECM成分和调节因子,包括Itga8(整合素α -8)和Grhl3(颗粒状头样蛋白3同源物),分别作为重编程障碍。机制研究整合了RNA测序、质谱分析、靶下切割和标记来定义grhl3依赖性调控。在体外使用去细胞化ECM评估功能结果,在体内使用具有遗传谱系追踪的心肌梗死模型评估功能结果。结果心肌重编程诱导动态ECM重构,胶原蛋白、纤维蛋白和整合素发生显著变化。Itga8被鉴定为通过TGF-β(转化生长因子-β)/SMAD途径限制iCM转化的关键ECM成分。Grhl3是ECM成分(包括Itga8)的关键转录调控因子。来自Grhl3缺失的成纤维细胞的ECM增强了iCM的诱导,而Grhl3缺失也降低了成纤维细胞的激活并增加了细胞的可塑性。这些效应与TF(转录因子)介导的重编程协同作用,提高iCM的效率、结构组织和功能成熟。在体内,去除Grhl3增强了成纤维细胞到心肌细胞的转化,减少了疤痕的形成,改善了心肌梗死后的心功能。结论我们的研究结果表明,ECM适应性是心脏重编程的关键决定因素,并确定Grhl3是推进心肌修复策略的有希望的治疗靶点。
{"title":"Grhl3 Downregulation Facilitates ECM Adaptation for Fibroblast to iCM Commitment.","authors":"Xin Wu,Lanbing Liu,Yuanru Huang,Yi Ling,Fang Luo,Dongyu Gu,Mengxin Liu,Zhenhua Jia,Zhangyi Yu,Xiangjie Kong,Hong Ma,Yanggan Wang,Li Wang","doi":"10.1161/circresaha.125.327726","DOIUrl":"https://doi.org/10.1161/circresaha.125.327726","url":null,"abstract":"BACKGROUNDDirect cardiac reprogramming offers a promising therapeutic strategy for heart regeneration by converting endogenous fibroblasts to functional induced cardiomyocytes (iCMs) that integrate into the myocardium to restore heart structure and function. While ECM (extracellular matrix) plays critical roles in cardiac disease and repair, the dynamic changes and transcriptional regulation underlying ECM remodeling during reprogramming remain poorly understood.METHODSWe investigated ECM dynamics during iCM reprogramming using integrated transcriptomic, proteomic, and epigenetic analyses, focusing on cell type-specific ECM components. A loss-of-function screen was used to identify critical ECM components and regulators, including Itga8 (integrin alpha-8) and Grhl3 (grainyhead-like protein 3 homolog), respectively, as reprogramming barriers. Mechanistic studies integrated RNA sequencing, mass spectrometry, and Cleavage Under Targets and Tagmentation to define Grhl3-dependent regulation. Functional outcomes were evaluated in vitro using decellularized ECM and in vivo using a myocardial infarction model with genetic lineage tracing.RESULTSCardiac reprogramming induced dynamic ECM remodeling, with significant changes in collagen, fibrillar proteins, and integrins. Itga8 was identified as a pivotal ECM component that restricts iCM conversion via the TGF-β (transforming growth factor-β)/SMAD pathway. Grhl3 emerged as a key transcriptional regulator for ECM components, including Itga8. ECM derived from Grhl3-deficient fibroblasts enhanced iCM induction, while Grhl3 depletion also reduced fibroblast activation and increased cellular plasticity. These effects synergized with TF (transcription factor)-mediated reprogramming to improve iCM efficiency, structural organization, and functional maturation. In vivo, removing Grhl3 enhanced fibroblast-to-cardiomyocyte conversion, reduced scar formation, and improved cardiac function after myocardial infarction.CONCLUSIONSOur findings establish ECM adaptation as a critical determinant of cardiac reprogramming and identify Grhl3 as a promising therapeutic target to advance myocardial repair strategies.","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"39 1","pages":""},"PeriodicalIF":20.1,"publicationDate":"2026-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145968383","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
A Road Map to Understanding Cardiovascular Disease in Diabetes: From the AHA Strategically Focused Research Network in Cardiometabolic Health and Type 2 Diabetes. 了解糖尿病心血管疾病的路线图:来自美国心脏协会心脏代谢健康和2型糖尿病战略重点研究网络
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-15 DOI: 10.1161/circresaha.125.325798
E Dale Abel,Rexford S Ahima,Ethan J Anderson,David D Berg,Jeffrey S Berger,Saumya Das,Mark W Feinberg,Edward A Fisher,Michael S Garshick,Chiara Giannarelli,Ira J Goldberg,Naomi M Hamburg,Sangwon F Kim,Filipe A Moura,Chiadi E Ndumele,Jonathan D Newman,Marc S Sabatine,Elizabeth Selvin,Ravi Shah
Despite major advances in medical therapies and prevention strategies, the risk of cardiovascular complications in patients with both type I and type II diabetes remains substantially elevated. In 2019, the American Heart Association sought applications for a Strategically Focused Research Network on Cardiometabolic Health and Type 2 Diabetes. In 2020, 4 centers were named, including Brigham and Women's Hospital, Johns Hopkins University, New York University, and the University of Iowa. These centers performed basic, translational, and clinical studies to provide insights to explain the over 2-fold risk of cardiovascular complications in diabetes. Clinical studies and studies in cells and animals aimed to uncover new mechanisms responsible for disease development. Studies using human populations sought to uncover new biomarkers to prognosticate risk. In this review, we discuss several key issues and current and developing methods to understand why diabetes drives atherosclerotic cardiovascular disease and heart failure. Both human data and experimental models are considered. We integrate a review of these topics with work from the Strategically Focused Research Network and conclude with suggestions for identifying novel risk factors and future experimental research.
尽管在医学治疗和预防策略方面取得了重大进展,但I型和II型糖尿病患者心血管并发症的风险仍然很高。2019年,美国心脏协会为心脏代谢健康和2型糖尿病战略重点研究网络寻求申请。2020年,有4个中心被命名,包括布里格姆妇女医院、约翰霍普金斯大学、纽约大学和爱荷华大学。这些中心进行了基础、转化和临床研究,以解释糖尿病患者心血管并发症超过2倍的风险。临床研究以及对细胞和动物的研究旨在揭示疾病发展的新机制。利用人类群体进行的研究试图发现新的生物标志物来预测风险。在这篇综述中,我们讨论了几个关键问题和当前和发展的方法来理解为什么糖尿病驱动动脉粥样硬化性心血管疾病和心力衰竭。同时考虑了人类数据和实验模型。我们将这些主题的回顾与战略重点研究网络的工作结合起来,最后提出了确定新的风险因素和未来实验研究的建议。
{"title":"A Road Map to Understanding Cardiovascular Disease in Diabetes: From the AHA Strategically Focused Research Network in Cardiometabolic Health and Type 2 Diabetes.","authors":"E Dale Abel,Rexford S Ahima,Ethan J Anderson,David D Berg,Jeffrey S Berger,Saumya Das,Mark W Feinberg,Edward A Fisher,Michael S Garshick,Chiara Giannarelli,Ira J Goldberg,Naomi M Hamburg,Sangwon F Kim,Filipe A Moura,Chiadi E Ndumele,Jonathan D Newman,Marc S Sabatine,Elizabeth Selvin,Ravi Shah","doi":"10.1161/circresaha.125.325798","DOIUrl":"https://doi.org/10.1161/circresaha.125.325798","url":null,"abstract":"Despite major advances in medical therapies and prevention strategies, the risk of cardiovascular complications in patients with both type I and type II diabetes remains substantially elevated. In 2019, the American Heart Association sought applications for a Strategically Focused Research Network on Cardiometabolic Health and Type 2 Diabetes. In 2020, 4 centers were named, including Brigham and Women's Hospital, Johns Hopkins University, New York University, and the University of Iowa. These centers performed basic, translational, and clinical studies to provide insights to explain the over 2-fold risk of cardiovascular complications in diabetes. Clinical studies and studies in cells and animals aimed to uncover new mechanisms responsible for disease development. Studies using human populations sought to uncover new biomarkers to prognosticate risk. In this review, we discuss several key issues and current and developing methods to understand why diabetes drives atherosclerotic cardiovascular disease and heart failure. Both human data and experimental models are considered. We integrate a review of these topics with work from the Strategically Focused Research Network and conclude with suggestions for identifying novel risk factors and future experimental research.","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"100 1","pages":"e325798"},"PeriodicalIF":20.1,"publicationDate":"2026-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971835","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
Circulating CD34+ Fibroblast Progenitors Engaged in Heart Fibrosis of Allograft. 循环CD34+成纤维细胞祖细胞参与同种异体移植心脏纤维化。
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-14 DOI: 10.1161/circresaha.125.326558
Xiaotong Sun,Ting Wang,Hui Gong,Yichao Qiu,Yuesheng Zhang,Mengjia Chen,Jianing Xue,Guoguo Ye,Rong Mou,Peng Teng,Weidong Li,Ting Chen,Li Zhang,Xiaogang Guo,Wei Mao,Haige Zhao,Liang Ma,Qingbo Xu
BACKGROUNDFibrosis is one of the major causes of cardiac allograft malfunction and is mainly driven by fibroblasts. However, the role of recipient-derived cells in generating allograft fibroblasts and the underlying mechanisms remain to be explored.METHODSWe analyzed human heart allograft samples and used murine transplant models (C57BL/6J, Cd34-CreERT2; R26-tdTomato, mRFP mice, Rosa26-iDTR, Postn-CreERT2; R26-tdTomato, double-tdTomato, and immunodeficient mice with BALB/c donors). Human progenitor cells were cultivated from blood. Single-cell RNA sequencing, Western blotting, quantitative polymerase chain reaction, and immunohistochemistry, whole-mount staining with 3-dimensional reconstruction, and in vivo/in vitro experiments were applied to characterize allograft cellular composition and communication.RESULTSSingle-cell RNA sequencing was introduced to delineate the allograft cell atlas of patients and mice. Y chromosome analysis identified that recipient-derived cells contributed to allograft fibroblasts in both patients and murine models. Combining the genetic cell lineage tracing technique, we found that recipient-derived CD34+ cells could give rise to activated fibroblasts. Bone marrow transplantation and parabiosis models revealed that the recipient's circulating non-bone marrow Cd34+ cells could generate allograft fibroblasts. Human CD34+ cells could differentiate into fibroblasts both in vivo and in vitro. CD34+ fibroblast progenitors were recruited by CXCL12-ACKR3 and MIF-ACKR3 interactions and differentiated via the TGFβ (transforming growth factor beta)/GFPT2 (glutamine-fructose-6-phosphate transaminase 2)/SMAD2/4 axis. Ablation of recipient Cd34+ cells reduced activated fibroblasts and alleviated allograft fibrosis.CONCLUSIONSWe identify circulating CD34+ cells as a novel source of fibroblast progenitors that contribute to cardiac allograft fibrosis, suggesting that targeting recipient CD34+ cells could be a novel therapeutic potential for treating cardiac fibrosis after heart transplantation.
背景:纤维化是同种异体心脏移植功能障碍的主要原因之一,主要由成纤维细胞驱动。然而,受体来源的细胞在产生同种异体移植成纤维细胞中的作用及其潜在机制仍有待探索。方法采用小鼠移植模型(C57BL/6J、Cd34-CreERT2、R26-tdTomato、mRFP小鼠、Rosa26-iDTR、Postn-CreERT2、R26-tdTomato、双tdtomato和BALB/c供体免疫缺陷小鼠)分析同种异体心脏移植样本。人类祖细胞是从血液中培养出来的。采用单细胞RNA测序、Western blotting、定量聚合酶链反应、免疫组织化学、三维重建全贴装染色和体内/体外实验来表征同种异体移植物的细胞组成和通讯。结果采用单细胞RNA测序技术绘制了患者和小鼠的同种异体移植细胞图谱。Y染色体分析发现,在患者和小鼠模型中,受体来源的细胞对同种异体移植成纤维细胞都有贡献。结合遗传细胞谱系追踪技术,我们发现受体来源的CD34+细胞可以产生活化的成纤维细胞。骨髓移植和异种共生模型显示,受体循环的非骨髓Cd34+细胞可以生成同种异体移植成纤维细胞。人CD34+细胞在体内和体外均可向成纤维细胞分化。CD34+成纤维细胞祖细胞通过CXCL12-ACKR3和MIF-ACKR3相互作用募集,并通过TGFβ(转化生长因子β)/GFPT2(谷氨酰胺-果糖-6-磷酸转氨酶2)/SMAD2/4轴分化。消融受体Cd34+细胞可减少活化的成纤维细胞,减轻同种异体移植物纤维化。结论:我们发现循环CD34+细胞是导致同种异体心脏移植纤维化的成纤维细胞祖细胞的新来源,这表明靶向受体CD34+细胞可能是治疗心脏移植后心脏纤维化的一种新的治疗潜力。
{"title":"Circulating CD34+ Fibroblast Progenitors Engaged in Heart Fibrosis of Allograft.","authors":"Xiaotong Sun,Ting Wang,Hui Gong,Yichao Qiu,Yuesheng Zhang,Mengjia Chen,Jianing Xue,Guoguo Ye,Rong Mou,Peng Teng,Weidong Li,Ting Chen,Li Zhang,Xiaogang Guo,Wei Mao,Haige Zhao,Liang Ma,Qingbo Xu","doi":"10.1161/circresaha.125.326558","DOIUrl":"https://doi.org/10.1161/circresaha.125.326558","url":null,"abstract":"BACKGROUNDFibrosis is one of the major causes of cardiac allograft malfunction and is mainly driven by fibroblasts. However, the role of recipient-derived cells in generating allograft fibroblasts and the underlying mechanisms remain to be explored.METHODSWe analyzed human heart allograft samples and used murine transplant models (C57BL/6J, Cd34-CreERT2; R26-tdTomato, mRFP mice, Rosa26-iDTR, Postn-CreERT2; R26-tdTomato, double-tdTomato, and immunodeficient mice with BALB/c donors). Human progenitor cells were cultivated from blood. Single-cell RNA sequencing, Western blotting, quantitative polymerase chain reaction, and immunohistochemistry, whole-mount staining with 3-dimensional reconstruction, and in vivo/in vitro experiments were applied to characterize allograft cellular composition and communication.RESULTSSingle-cell RNA sequencing was introduced to delineate the allograft cell atlas of patients and mice. Y chromosome analysis identified that recipient-derived cells contributed to allograft fibroblasts in both patients and murine models. Combining the genetic cell lineage tracing technique, we found that recipient-derived CD34+ cells could give rise to activated fibroblasts. Bone marrow transplantation and parabiosis models revealed that the recipient's circulating non-bone marrow Cd34+ cells could generate allograft fibroblasts. Human CD34+ cells could differentiate into fibroblasts both in vivo and in vitro. CD34+ fibroblast progenitors were recruited by CXCL12-ACKR3 and MIF-ACKR3 interactions and differentiated via the TGFβ (transforming growth factor beta)/GFPT2 (glutamine-fructose-6-phosphate transaminase 2)/SMAD2/4 axis. Ablation of recipient Cd34+ cells reduced activated fibroblasts and alleviated allograft fibrosis.CONCLUSIONSWe identify circulating CD34+ cells as a novel source of fibroblast progenitors that contribute to cardiac allograft fibrosis, suggesting that targeting recipient CD34+ cells could be a novel therapeutic potential for treating cardiac fibrosis after heart transplantation.","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"37 1","pages":""},"PeriodicalIF":20.1,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145961454","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
1-Phosphatidylinositol 3-Phosphate 5-Kinase Inhibition by Apilimod Promotes an Adipocyte-Like Vascular Smooth Muscle Cell Phenotype and Prevents Arterial Calcification. Apilimod抑制1-磷脂酰肌醇3-磷酸5激酶促进脂肪细胞样血管平滑肌细胞表型并防止动脉钙化。
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-14 DOI: 10.1161/circresaha.125.326772
Nicolas Hense,Andrea Gogels,Bilal Mir,Emiel P C van der Vorst,Guillaume Falgayrac,Mara Terliesner,Isabel Goncalves,Ljubica Matic,Andreas Edsfeldt,Jiangming Sun,Ulf Hedin,Sikander Hayat,Jessica Thiel,Maria Alejandra Ramirez-Torres,Mirna Barsoum,Alexander Rauch,Martina Rauner,Alexander Gombert,Christian Preisinger,Heidi Noels,Nikolaus Marx,Claudia Goettsch
BACKGROUNDCardiovascular calcification is a significant predictor and contributor to cardiovascular diseases. Vascular smooth muscle cell (SMC)-derived extracellular vesicles (EVs) play a crucial role in microcalcification formation. EVs can originate from the endosomal system, and PIKFYVE (1-phosphatidylinositol 3-phosphate 5-kinase), a lipid kinase, plays a key role in endomembrane maturation. We hypothesize that PIKFYVE inhibition will modulate EV cargo and, thereby, arterial calcification.METHODSHuman coronary artery SMCs were cultured in osteogenic media to induce calcification. PIKFYVE inhibition was achieved using the pharmacological inhibitor apilimod and siRNA. We characterized the SMC phenotype and EVs through proteomics, transcriptomics, and kinomics. Ldlr-deficient mice fed a high-fat, high-cholesterol diet received apilimod for 5 weeks.RESULTSCalcified human arteries and SMCs exhibited increased PIKFYVE protein expression compared with controls. In calcifying SMCs, phosphatidylinositol 3-phosphate levels were reduced but restored by apilimod. Apilimod prevented matrix mineralization and collagen deposition in calcifying SMCs, accompanied by reduced procollagen 1A1 secretion. Apilimod inhibited TNAP (tissue-nonspecific alkaline phosphatase) at mRNA, protein, and activity levels. EVs released from apilimod-treated calcifying SMCs exhibited lower mineral cargo, reduced aggregation potential, and diminished TNAP cargo. Phenotypic omics analyses revealed that apilimod induced a shift toward an adipocyte-like SMC phenotype, marked by upregulation of adipogenic TFs (transcription factors), fatty acid metabolism genes, and increased fatty acid uptake. Reactivation of YAP (Yes-associated protein)-TEAD (transcriptional enhancer factor) signaling partially reversed these phenotypic changes. In vivo, apilimod reduced vascular calcification and plaque TNAP activity but increased plaque lipid accumulation.CONCLUSIONSPharmacological inhibition of PIKFYVE disrupts YAP-TEAD signaling, thereby reducing arterial calcification by limiting the calcification potential of EVs and suppressing osteogenic SMC programming, but also induces an adipocyte-like SMC phenotype with lipid accumulation. These findings emphasize the need to consider SMC phenotypic plasticity and potential adverse effects when developing therapeutic strategies for arterial calcification.
背景:心血管钙化是心血管疾病的重要预测因子和因素。血管平滑肌细胞(SMC)衍生的细胞外囊泡(EVs)在微钙化形成中起着至关重要的作用。EVs可以起源于内体系统,PIKFYVE(1-磷脂酰肌醇3-磷酸5激酶)是一种脂质激酶,在内膜成熟中起关键作用。我们假设抑制PIKFYVE会调节EV货物,从而调节动脉钙化。方法在成骨培养基中培养人冠状动脉SMCs,诱导其钙化。使用药物抑制剂apilimod和siRNA实现PIKFYVE抑制。我们通过蛋白质组学、转录组学和基因组学对SMC表型和ev进行了表征。低密度脂蛋白缺陷小鼠喂食高脂肪、高胆固醇饮食,给予阿匹利莫德5周。结果与对照组相比,钙化的人动脉和SMCs中PIKFYVE蛋白表达增加。在钙化的SMCs中,磷脂酰肌醇3-磷酸水平降低,但阿利莫德使其恢复。Apilimod阻止钙化SMCs的基质矿化和胶原沉积,同时减少前胶原1A1分泌。Apilimod在mRNA、蛋白和活性水平上抑制TNAP(组织非特异性碱性磷酸酶)。经apilimod处理的钙化SMCs释放的ev表现出较低的矿物载货量、降低的聚集电位和减少的TNAP载货量。表型组学分析显示,apilimod诱导向脂肪细胞样SMC表型转变,其特征是脂肪生成tf(转录因子)、脂肪酸代谢基因的上调,以及脂肪酸摄取的增加。YAP (yes相关蛋白)-TEAD(转录增强因子)信号的再激活部分逆转了这些表型变化。在体内,apilimod降低了血管钙化和斑块TNAP活性,但增加了斑块脂质积累。结论药理学抑制PIKFYVE可破坏YAP-TEAD信号,从而通过限制内皮细胞的钙化电位和抑制成骨SMC编程来减少动脉钙化,但也可诱导脂肪细胞样SMC表型伴脂质积累。这些发现强调在制定动脉钙化治疗策略时需要考虑SMC表型可塑性和潜在的不良影响。
{"title":"1-Phosphatidylinositol 3-Phosphate 5-Kinase Inhibition by Apilimod Promotes an Adipocyte-Like Vascular Smooth Muscle Cell Phenotype and Prevents Arterial Calcification.","authors":"Nicolas Hense,Andrea Gogels,Bilal Mir,Emiel P C van der Vorst,Guillaume Falgayrac,Mara Terliesner,Isabel Goncalves,Ljubica Matic,Andreas Edsfeldt,Jiangming Sun,Ulf Hedin,Sikander Hayat,Jessica Thiel,Maria Alejandra Ramirez-Torres,Mirna Barsoum,Alexander Rauch,Martina Rauner,Alexander Gombert,Christian Preisinger,Heidi Noels,Nikolaus Marx,Claudia Goettsch","doi":"10.1161/circresaha.125.326772","DOIUrl":"https://doi.org/10.1161/circresaha.125.326772","url":null,"abstract":"BACKGROUNDCardiovascular calcification is a significant predictor and contributor to cardiovascular diseases. Vascular smooth muscle cell (SMC)-derived extracellular vesicles (EVs) play a crucial role in microcalcification formation. EVs can originate from the endosomal system, and PIKFYVE (1-phosphatidylinositol 3-phosphate 5-kinase), a lipid kinase, plays a key role in endomembrane maturation. We hypothesize that PIKFYVE inhibition will modulate EV cargo and, thereby, arterial calcification.METHODSHuman coronary artery SMCs were cultured in osteogenic media to induce calcification. PIKFYVE inhibition was achieved using the pharmacological inhibitor apilimod and siRNA. We characterized the SMC phenotype and EVs through proteomics, transcriptomics, and kinomics. Ldlr-deficient mice fed a high-fat, high-cholesterol diet received apilimod for 5 weeks.RESULTSCalcified human arteries and SMCs exhibited increased PIKFYVE protein expression compared with controls. In calcifying SMCs, phosphatidylinositol 3-phosphate levels were reduced but restored by apilimod. Apilimod prevented matrix mineralization and collagen deposition in calcifying SMCs, accompanied by reduced procollagen 1A1 secretion. Apilimod inhibited TNAP (tissue-nonspecific alkaline phosphatase) at mRNA, protein, and activity levels. EVs released from apilimod-treated calcifying SMCs exhibited lower mineral cargo, reduced aggregation potential, and diminished TNAP cargo. Phenotypic omics analyses revealed that apilimod induced a shift toward an adipocyte-like SMC phenotype, marked by upregulation of adipogenic TFs (transcription factors), fatty acid metabolism genes, and increased fatty acid uptake. Reactivation of YAP (Yes-associated protein)-TEAD (transcriptional enhancer factor) signaling partially reversed these phenotypic changes. In vivo, apilimod reduced vascular calcification and plaque TNAP activity but increased plaque lipid accumulation.CONCLUSIONSPharmacological inhibition of PIKFYVE disrupts YAP-TEAD signaling, thereby reducing arterial calcification by limiting the calcification potential of EVs and suppressing osteogenic SMC programming, but also induces an adipocyte-like SMC phenotype with lipid accumulation. These findings emphasize the need to consider SMC phenotypic plasticity and potential adverse effects when developing therapeutic strategies for arterial calcification.","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"55 1","pages":""},"PeriodicalIF":20.1,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971796","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
SWAP70 Promotes Atherosclerosis Via Endothelial CAV1 Nuclear Translocation. SWAP70通过内皮细胞CAV1核易位促进动脉粥样硬化。
IF 20.1 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-14 DOI: 10.1161/circresaha.125.327048
Tianyu Gao,Xinxin Li,Wei Zhang,Yuzhou Yang,Yiying Liu,Fengchao Liang,Haocheng Lu,Laiyuan Wang,Bo Bai,Dongfeng Gu
BACKGROUNDAtherosclerosis, the leading cause of coronary artery disease, is initiated and exacerbated by disturbed blood flow and chronic endothelial inflammation. SWAP70 (Switch-associated protein 70), a multifunctional signaling adaptor, has been genetically linked to coronary artery disease susceptibility via the risk allele rs10840293. However, its precise role in atherogenesis remains poorly understood.METHODSWe employed both endothelial cell-specific Swap70 overexpression and knockout mouse models, alongside lentiviral overexpression and siRNA-mediated SWAP70 knockdown in human umbilical vein endothelial cells, to investigate the functional role of SWAP70 in vascular inflammation and plaque development. In vitro assays subjected human umbilical vein endothelial cells to oscillatory shear stress or proinflammatory cytokines, followed by evaluation of adhesion molecule and chemokine expression. Mechanistic studies were performed using coimmunoprecipitation, proximity ligation assay, mimetic peptide interference, RNA sequencing, and ChIP-qPCR analyses.RESULTSSWAP70 expression was significantly upregulated in human atherosclerotic plaques and in human umbilical vein endothelial cells exposed to oscillatory shear stress compared with laminar shear stress. On oscillatory shear stress stimulation, SWAP70 bound to the scaffolding domain of CAV1 (caveolin-1) to facilitate its nuclear translocation, thereby enhancing transcription of key inflammatory mediators, including adhesion molecules and chemokines. In vitro, SWAP70 knockdown suppressed oscillatory shear stress and TNF-α (tumor necrosis factor-α)-induced proinflammatory gene expression. In vivo, endothelial-specific deletion of Swap70 attenuated high-fat diet-induced atherosclerotic lesion formation, reduced vascular inflammation, and improved plaque stability. Conversely, overexpression of Swap70 amplified inflammatory responses and worsened atherogenic outcomes.CONCLUSIONSOur findings identify SWAP70 as a mechano-responsive regulator of endothelial inflammation and atherosclerosis, acting through a novel mechanism involving CAV1 nuclear translocation. Targeting the SWAP70-CAV1 signaling axis represents a promising therapeutic strategy for mitigating vascular inflammation and attenuating the progression of atherosclerotic cardiovascular disease.
背景:动脉硬化是冠状动脉疾病的主要原因,它是由血流紊乱和慢性内皮炎症引发和加剧的。SWAP70(开关相关蛋白70)是一种多功能信号适配器,通过风险等位基因rs10840293与冠状动脉疾病易感性有遗传联系。然而,其在动脉粥样硬化发生中的确切作用仍然知之甚少。方法采用内皮细胞特异性Swap70过表达和敲除小鼠模型,以及慢病毒过表达和sirna介导的Swap70敲除人脐静脉内皮细胞,研究Swap70在血管炎症和斑块形成中的功能作用。体外实验将人脐静脉内皮细胞置于振荡剪切应力或促炎细胞因子的作用下,然后评估粘附分子和趋化因子的表达。机制研究采用共免疫沉淀、邻近连接试验、模拟肽干扰、RNA测序和ChIP-qPCR分析。结果与层流剪切应力相比,swap70在振荡剪切应力下的人动脉粥样硬化斑块和脐静脉内皮细胞中的表达明显上调。在振荡剪切应力刺激下,SWAP70与CAV1 (caveolin-1)的支架结构域结合,促进其核易位,从而增强关键炎症介质的转录,包括粘附分子和趋化因子。在体外,SWAP70敲低抑制振荡剪切应力和TNF-α(肿瘤坏死因子-α)诱导的促炎基因表达。在体内,内皮特异性缺失Swap70可减轻高脂肪饮食诱导的动脉粥样硬化病变形成,减少血管炎症,并改善斑块稳定性。相反,Swap70的过表达放大了炎症反应,恶化了动脉粥样硬化的结果。我们的研究结果表明SWAP70是内皮炎症和动脉粥样硬化的机械响应性调节剂,通过涉及CAV1核易位的新机制起作用。靶向SWAP70-CAV1信号轴是缓解血管炎症和减缓动脉粥样硬化性心血管疾病进展的一种有希望的治疗策略。
{"title":"SWAP70 Promotes Atherosclerosis Via Endothelial CAV1 Nuclear Translocation.","authors":"Tianyu Gao,Xinxin Li,Wei Zhang,Yuzhou Yang,Yiying Liu,Fengchao Liang,Haocheng Lu,Laiyuan Wang,Bo Bai,Dongfeng Gu","doi":"10.1161/circresaha.125.327048","DOIUrl":"https://doi.org/10.1161/circresaha.125.327048","url":null,"abstract":"BACKGROUNDAtherosclerosis, the leading cause of coronary artery disease, is initiated and exacerbated by disturbed blood flow and chronic endothelial inflammation. SWAP70 (Switch-associated protein 70), a multifunctional signaling adaptor, has been genetically linked to coronary artery disease susceptibility via the risk allele rs10840293. However, its precise role in atherogenesis remains poorly understood.METHODSWe employed both endothelial cell-specific Swap70 overexpression and knockout mouse models, alongside lentiviral overexpression and siRNA-mediated SWAP70 knockdown in human umbilical vein endothelial cells, to investigate the functional role of SWAP70 in vascular inflammation and plaque development. In vitro assays subjected human umbilical vein endothelial cells to oscillatory shear stress or proinflammatory cytokines, followed by evaluation of adhesion molecule and chemokine expression. Mechanistic studies were performed using coimmunoprecipitation, proximity ligation assay, mimetic peptide interference, RNA sequencing, and ChIP-qPCR analyses.RESULTSSWAP70 expression was significantly upregulated in human atherosclerotic plaques and in human umbilical vein endothelial cells exposed to oscillatory shear stress compared with laminar shear stress. On oscillatory shear stress stimulation, SWAP70 bound to the scaffolding domain of CAV1 (caveolin-1) to facilitate its nuclear translocation, thereby enhancing transcription of key inflammatory mediators, including adhesion molecules and chemokines. In vitro, SWAP70 knockdown suppressed oscillatory shear stress and TNF-α (tumor necrosis factor-α)-induced proinflammatory gene expression. In vivo, endothelial-specific deletion of Swap70 attenuated high-fat diet-induced atherosclerotic lesion formation, reduced vascular inflammation, and improved plaque stability. Conversely, overexpression of Swap70 amplified inflammatory responses and worsened atherogenic outcomes.CONCLUSIONSOur findings identify SWAP70 as a mechano-responsive regulator of endothelial inflammation and atherosclerosis, acting through a novel mechanism involving CAV1 nuclear translocation. Targeting the SWAP70-CAV1 signaling axis represents a promising therapeutic strategy for mitigating vascular inflammation and attenuating the progression of atherosclerotic cardiovascular disease.","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":"2 1","pages":""},"PeriodicalIF":20.1,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145961451","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
Control of Blood Pressure Variability Across Behavioral States by Brainstem Adrenergic Neurons. 脑干肾上腺素能神经元对行为状态下血压变异性的控制。
IF 16.2 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-13 DOI: 10.1161/CIRCRESAHA.125.326792
George M P R Souza, Harsha Thakkalapally, Faye E Berry, Leah F Wisniewski, Ulrich M Atongazi, Daniel S Stornetta, Stephen B G Abbott

Background: Short-term blood pressure (BP) variability is increasingly recognized as an independent predictor of cardiovascular and cerebrovascular risks, yet the central neural mechanisms that govern this variability, particularly across behavioral states, remain poorly defined.

Methods: We investigated the role of rostral ventrolateral medulla C1 (RVLMC1) neurons in short-term BP regulation during sleep-wake transitions and physical activity in freely behaving rats. Genetically targeted fiber photometry was used to record RVLMC1 neuronal activity across behavioral states. The contribution of feedback from the arterial baroreflex to the activity of RVLMC1 neurons was assessed using sinoaortic denervation. Selective genetic ablation of RVLMC1 neurons was performed to determine their role in BP regulation.

Results: RVLMC1 neurons exhibited state-dependent activity, with rapid activation during arousal from nonrapid eye movement sleep, sustained activity during rapid eye movement sleep, and further recruitment during physical activity. Baroreflex input contributed to the modulation of RVLMC1 neuron activity by pharmacological manipulations of BP and transitions from nonrapid eye movement sleep to rapid eye movement sleep. Selective ablation of RVLMC1 neurons did not alter mean BP but resulted in marked BP instability during arousal and movement.

Conclusions: RVLMC1 neurons stabilize BP during changes in the behavioral state by integrating arousal-related central drive with baroreceptor feedback. Disruption of these neurons leads to increased short-term BP variability despite preserved mean BP, providing a potential neural mechanism underlying pathological BP instability.

背景:短期血压(BP)变异性越来越被认为是心脑血管风险的独立预测因素,然而控制这种变异性的中枢神经机制,特别是跨行为状态的中枢神经机制,仍然不明确。方法:研究自由行为大鼠的吻侧腹外侧髓质C1 (RVLMC1)神经元在睡眠-觉醒转换和身体活动期间短期血压调节中的作用。基因靶向纤维光度法用于记录RVLMC1神经元在行为状态下的活动。动脉压力反射对RVLMC1神经元活动的反馈作用通过窦主动脉去神经支配进行评估。选择性基因消融RVLMC1神经元以确定其在血压调节中的作用。结果:RVLMC1神经元表现出状态依赖性活动,在非快速眼动睡眠唤醒时快速激活,在快速眼动睡眠期间持续活动,在身体活动期间进一步募集。通过药理学操作血压和从非快速眼动睡眠到快速眼动睡眠的转换,bar反射输入有助于RVLMC1神经元活动的调节。选择性消融RVLMC1神经元不会改变平均血压,但会导致觉醒和运动期间明显的血压不稳定。结论:RVLMC1神经元通过整合唤醒相关中枢驱动和压力感受器反馈,在行为状态变化时稳定BP。尽管平均血压保持不变,但这些神经元的破坏导致短期血压变异性增加,这为病理性血压不稳定提供了潜在的神经机制。
{"title":"Control of Blood Pressure Variability Across Behavioral States by Brainstem Adrenergic Neurons.","authors":"George M P R Souza, Harsha Thakkalapally, Faye E Berry, Leah F Wisniewski, Ulrich M Atongazi, Daniel S Stornetta, Stephen B G Abbott","doi":"10.1161/CIRCRESAHA.125.326792","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.125.326792","url":null,"abstract":"<p><strong>Background: </strong>Short-term blood pressure (BP) variability is increasingly recognized as an independent predictor of cardiovascular and cerebrovascular risks, yet the central neural mechanisms that govern this variability, particularly across behavioral states, remain poorly defined.</p><p><strong>Methods: </strong>We investigated the role of rostral ventrolateral medulla C1 (RVLM<sup>C1</sup>) neurons in short-term BP regulation during sleep-wake transitions and physical activity in freely behaving rats. Genetically targeted fiber photometry was used to record RVLM<sup>C1</sup> neuronal activity across behavioral states. The contribution of feedback from the arterial baroreflex to the activity of RVLM<sup>C1</sup> neurons was assessed using sinoaortic denervation. Selective genetic ablation of RVLM<sup>C1</sup> neurons was performed to determine their role in BP regulation.</p><p><strong>Results: </strong>RVLM<sup>C1</sup> neurons exhibited state-dependent activity, with rapid activation during arousal from nonrapid eye movement sleep, sustained activity during rapid eye movement sleep, and further recruitment during physical activity. Baroreflex input contributed to the modulation of RVLM<sup>C1</sup> neuron activity by pharmacological manipulations of BP and transitions from nonrapid eye movement sleep to rapid eye movement sleep. Selective ablation of RVLM<sup>C1</sup> neurons did not alter mean BP but resulted in marked BP instability during arousal and movement.</p><p><strong>Conclusions: </strong>RVLM<sup>C1</sup> neurons stabilize BP during changes in the behavioral state by integrating arousal-related central drive with baroreceptor feedback. Disruption of these neurons leads to increased short-term BP variability despite preserved mean BP, providing a potential neural mechanism underlying pathological BP instability.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":""},"PeriodicalIF":16.2,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145959059","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
Neratinib, a Clinical Drug Against Breast Cancer, Protects Against Vascular Inflammation and Atherosclerosis. Neratinib是一种抗乳腺癌的临床药物,可以预防血管炎症和动脉粥样硬化。
IF 16.2 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Pub Date : 2026-01-09 DOI: 10.1161/CIRCRESAHA.125.326508
Fan-Shun Zhang, Chenyang He, Yanjun Yin, Zhihua Wang, Xiumei Wu, Danielle Kamato, Ruixue Leng, Jiang-Yun Luo, Jianping Weng, Suowen Xu

Background: Atherosclerosis commences with endothelial dysfunction and the retention of cholesterol within the vessel wall, followed by a chronic inflammatory response. Lowering LDL-C (low-density lipoprotein-cholesterol; such as statins and PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) is the mainstay of current treatment for patients with atherosclerotic cardiovascular diseases, but residual inflammatory risk remains high.

Methods: To address this pressing challenge, we used connectivity map screening of Food and Drug Administration-approved drugs, using perturbational data sets obtained from TNF-α (tumor necrosis factor-α) and IL (interleukin)-1β-stimulated human endothelial cells. Male and female Ldlr-/- mouse models were used to evaluate the in vivo antiatherosclerotic effect of the hit compound identified.

Results: This screening endeavor allows us to identify neratinib, a clinical drug against breast cancer, as the hit compound with broad anti-inflammatory actions in endothelial cells. Further studies reveal that neratinib inhibited endothelial cell inflammation elicited by 3 different proinflammatory stimuli (TNF-α, IL-1β, and lipopolysaccharide). Intriguingly, the anti-inflammatory effect of neratinib was independent of its classical target HER2 (human epidermal growth factor receptor 2)/ERBB2 inhibition. Further mechanistic investigation revealed that neratinib directly binds to ASK1 (apoptosis signal-regulating kinase 1) and suppresses ASK1 activation. Importantly, in both male and female Ldlr-/- mice, treatment with neratinib decreased the plaque burden, reduced the necrotic core size, and mitigated lesional macrophage infiltration. Of translational impact, we observed that neratinib, in conjunction with the use of rosuvastatin (a standard lipid-lowering drug), produced superior antiatherosclerotic effects compared with statin monotherapy. Olink proteomics study pinpointed that combination treatment alleviated inflammation-related cytokines/chemokines in the serum from Ldlr-/- mice.

Conclusions: Taken together, these findings support the concept that neratinib could be tested as a repurposed drug for vascular inflammation and atherosclerosis, thereby streamlining efforts to translate preclinical discoveries to clinical testing in humans.

背景:动脉粥样硬化始于内皮功能障碍和胆固醇在血管壁内的滞留,随后是慢性炎症反应。降低LDL-C(低密度脂蛋白-胆固醇,如他汀类药物和PCSK9[蛋白转化酶枯草杆菌素/ keexin 9型]抑制剂)是目前动脉粥样硬化性心血管疾病患者的主要治疗方法,但残余炎症风险仍然很高。方法:为了解决这一紧迫的挑战,我们使用从TNF-α(肿瘤坏死因子-α)和IL(白细胞介素)-1β刺激的人内皮细胞中获得的扰动数据集,对美国食品和药物管理局批准的药物进行连接图筛选。使用雄性和雌性Ldlr-/-小鼠模型来评估所鉴定的hit化合物的体内抗动脉粥样硬化作用。结果:这项筛选工作使我们能够确定奈拉替尼,一种抗乳腺癌的临床药物,作为对内皮细胞具有广泛抗炎作用的hit化合物。进一步的研究表明,neratinib可以抑制3种不同的促炎刺激(TNF-α, IL-1β和脂多糖)引起的内皮细胞炎症。有趣的是,neratinib的抗炎作用独立于其经典靶点HER2(人表皮生长因子受体2)/ERBB2抑制。进一步的机制研究表明,neratinib直接结合ASK1(凋亡信号调节激酶1)并抑制ASK1的激活。重要的是,在雄性和雌性Ldlr-/-小鼠中,neratinib治疗降低了斑块负担,减少了坏死核心的大小,减轻了病变巨噬细胞的浸润。在转化影响方面,我们观察到奈拉替尼联合瑞舒伐他汀(一种标准降脂药物)与他汀类药物单药治疗相比,产生了更好的抗动脉粥样硬化效果。Olink蛋白质组学研究指出,联合治疗可减轻Ldlr-/-小鼠血清中炎症相关的细胞因子/趋化因子。结论:综上所述,这些发现支持了neratinib可以作为血管炎症和动脉粥样硬化的再用途药物进行测试的概念,从而简化了将临床前发现转化为人体临床试验的努力。
{"title":"Neratinib, a Clinical Drug Against Breast Cancer, Protects Against Vascular Inflammation and Atherosclerosis.","authors":"Fan-Shun Zhang, Chenyang He, Yanjun Yin, Zhihua Wang, Xiumei Wu, Danielle Kamato, Ruixue Leng, Jiang-Yun Luo, Jianping Weng, Suowen Xu","doi":"10.1161/CIRCRESAHA.125.326508","DOIUrl":"https://doi.org/10.1161/CIRCRESAHA.125.326508","url":null,"abstract":"<p><strong>Background: </strong>Atherosclerosis commences with endothelial dysfunction and the retention of cholesterol within the vessel wall, followed by a chronic inflammatory response. Lowering LDL-C (low-density lipoprotein-cholesterol; such as statins and PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) is the mainstay of current treatment for patients with atherosclerotic cardiovascular diseases, but residual inflammatory risk remains high.</p><p><strong>Methods: </strong>To address this pressing challenge, we used connectivity map screening of Food and Drug Administration-approved drugs, using perturbational data sets obtained from TNF-α (tumor necrosis factor-α) and IL (interleukin)-1β-stimulated human endothelial cells. Male and female <i>Ldlr</i><sup>-/-</sup> mouse models were used to evaluate the in vivo antiatherosclerotic effect of the hit compound identified.</p><p><strong>Results: </strong>This screening endeavor allows us to identify neratinib, a clinical drug against breast cancer, as the hit compound with broad anti-inflammatory actions in endothelial cells. Further studies reveal that neratinib inhibited endothelial cell inflammation elicited by 3 different proinflammatory stimuli (TNF-α, IL-1β, and lipopolysaccharide). Intriguingly, the anti-inflammatory effect of neratinib was independent of its classical target HER2 (human epidermal growth factor receptor 2)/ERBB2 inhibition. Further mechanistic investigation revealed that neratinib directly binds to ASK1 (apoptosis signal-regulating kinase 1) and suppresses ASK1 activation. Importantly, in both male and female <i>Ldlr</i><sup>-/-</sup> mice, treatment with neratinib decreased the plaque burden, reduced the necrotic core size, and mitigated lesional macrophage infiltration. Of translational impact, we observed that neratinib, in conjunction with the use of rosuvastatin (a standard lipid-lowering drug), produced superior antiatherosclerotic effects compared with statin monotherapy. Olink proteomics study pinpointed that combination treatment alleviated inflammation-related cytokines/chemokines in the serum from <i>Ldlr</i><sup>-/-</sup> mice.</p><p><strong>Conclusions: </strong>Taken together, these findings support the concept that neratinib could be tested as a repurposed drug for vascular inflammation and atherosclerosis, thereby streamlining efforts to translate preclinical discoveries to clinical testing in humans.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":" ","pages":""},"PeriodicalIF":16.2,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932248","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:604180095
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