Pub Date : 2026-01-16Epub Date: 2025-12-04DOI: 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.
{"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}
Pub Date : 2026-01-16Epub Date: 2026-01-15DOI: 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}
Pub Date : 2026-01-15DOI: 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}
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.
{"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}
Pub Date : 2026-01-15DOI: 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.
{"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}
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.
{"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}
Pub Date : 2026-01-14DOI: 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.
{"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}
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.
{"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}
Pub Date : 2026-01-13DOI: 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.
{"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}
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.
{"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}