Cardiology is the management, diagnosis, treatment of disorders that relate to the Cardiovascular System. These conditions range from the very common to the very rare, spanning all ages and and can involve various organ systems.
- Relationship Between Coronary Artery Calcium Score and Vulnerability of Culprit Plaque Assessed by OCT in Patients With Established Coronary Artery Diseaseby Daichi Fujimoto Eisuke Usui Rocco Vergallo Daisuke Kinoshita Keishi Suzuki Takayuki Niida Marco Covani Iris McNulty Hang Lee Hiromasa Otake Junya Shite Maros Ferencik Damini Dey Tsunekazu Kakuta Ik-Kyung Jang Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston. (D.F., D.K., K.S., T.N., M.C., I.M., I.-K.J.) Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston. (H.L.) Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Japan (E.U., T.K.). Cardiothoracic and Vascular Department (DICATOV), Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy (R.V.). Department of Internal Medicine and Medical Specialties, Università di Genova, Italy (R.V.). Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe (H.O.). Division of Cardiovascular Medicine, Osaka Saiseikai Nakatsu Hospital, Japan (J.S.). Knight Cardiovascular Institute, Oregon Health and Science University, Portland (M.F.). Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA (D.D.). on December 20, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. BACKGROUND:Coronary artery calcium score (CACS) is widely used for risk stratification. However, in patients with established coronary artery disease, its clinical implication and relationship with plaque vulnerability are unclear. We sought to correlate the CACS and plaque vulnerability assessed by optical coherence tomography.METHODS:Patients with coronary artery disease who had CACS and optical coherence tomography before percutaneous coronary intervention were included. Patients were divided into 5 groups based on CACS: CACS of 0, 1 to 99, 100 to 399, 400 to 999, and ≥1000. Optical coherence tomography–derived vulnerable features in culprit plaque were compared between the groups.RESULTS:In 460 patients, the prevalence of lipid-rich plaque, macrophage, and cholesterol crystal significantly differed among the 5 groups, being lowest in the patients with a CACS of 0. The prevalence of thin-cap fibroatheroma tended to be lower in those with a CACS of 0. No significant difference in vulnerable features was observed between the 4 groups with CACS >0. In the 2-group comparison between the group with a CACS of 0 and the other 4 groups combined, the prevalence of lipid-rich plaque (60.5% versus 85.9%;P<0.001), macrophage (48.8% versus 74.1%;P<0.001), thin-cap fibroatheroma (16.3% versus 35.0%;P=0.013), and cholesterol crystal (11.6% versus 32.9%;P=0.004) was significantly lower in the patients with CACS of 0. CACS of 0 was independently negatively associated with lipid-rich plaque, macrophage, thin-cap fibroatheroma, and cholesterol crystal after adjustment for patient characteristics.CONCLUSIONS:Patients with a CACS of 0 have a significantly lower prevalence of vulnerable plaque features compared with those with CACS >0 in patients with established coronary artery disease.REGISTRATION:URL:https://www.clinicaltrials.gov; Unique identifier: NCT04523194.
- When Is Enough Enough? Additional Evidence of the Power of Zeroby Matthew J. Budoff Department of Medicine, Lundquist Institute at Harbor-UCLA, Torrance, CA, USA. on December 20, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Placental Pathology and Blood Pressure at Age 7: A Longitudinal Discordant Twin Analysisby Alexa A. Freedman Gregory E. Miller Andrew D. Franklin Lauren S. Keenan-Devlin Stephen E. Gilman Ann Borders Sadiya S. Khan Linda M. Ernst Department of Obstetrics and Gynecology, Endeavor Health (A.A.F.). Now with Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (A.A.F.). Institute for Policy Research, Northwestern University. (G.E.M.) Department of Psychology, Northwestern University. (G.E.M.) Department of Pediatrics, Endeavor Health, The University of Chicago Pritzker School of Medicine. (A.D.F.) Department of Pathology and Laboratory Medicine, Endeavor Health, The University of Chicago Pritzker School of Medicine. (L.M.E.) Department of Obstetrics and Gynecology, The University of Chicago Pritzker School of Medicine. (L.S.K.-D., A.B.) Social and Behavioral Sciences Branch, Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Northwestern University Feinberg School of Medicine, Chicago, IL. (S.E.G.). Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Northwestern University Feinberg School of Medicine, Chicago, IL. (S.E.G.). Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL. (A.B.) Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL. (S.S.K.) Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL. (S.S.K.) Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Endeavor Health, Evanston Hospital (A.B.). on December 19, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Evidence suggests that the intrauterine environment shapes offspring cardiovascular disease risk. Although placental dysfunction may be an important pathophysiologic pathway, numerous parental and pregnancy characteristics that influence offspring blood pressure are strong confounders of the mechanistic role of the placenta in observational analyses of singletons. Therefore, we leverage twin- and sibling-based comparison designs to determine whether placental pathology is associated with offspring blood pressure at age 7 while mitigating major sources of confounding.METHODS:Data are from pregnant participants and their offspring in the Collaborative Perinatal Project, a longitudinal pregnancy cohort conducted from 1959 to 1965 in the United States. After delivery, placentas were systematically examined for lesions indicative of maternal vascular malperfusion (MVM) and acute inflammation. Blood pressure was assessed at a follow-up research visit when the offspring were 7 years old. Linear fixed-effects models were used to estimate associations between within-twin or sibling discordance in placental pathology and differences in blood pressure at age 7.RESULTS:Overall, 193 twin pairs were eligible for inclusion, and 23.3% had placentas discordant for MVM. In a fixed-effect analysis, a twin with high-grade MVM had a higher systolic blood pressureZscore by 0.56 SDs than their co-twin without MVM (95% CI, 0.06–1.05) or a 5.7-mm Hg difference (95% CI, 0.6–10.8). Associations were consistent in a sensitivity analysis restricted to dichorionic twins and in a secondary analysis of 759 MVM-discordant sibling pairs. Acute placental inflammation was not associated with blood pressure at age 7.CONCLUSIONS:MVM in the placenta is associated with higher offspring blood pressure in mid-childhood, independent of parental and pregnancy characteristics that twins have in common. The findings support the role of the placenta and the intrauterine environment in the developmental origins of cardiovascular health.
- Heterogeneous Cardiac-Derived and Neural Crest–Derived Aortic Smooth Muscle Cells Exhibit Similar Transcriptional Changes After TGFβ Signaling Disruptionby Pengwei Ren Bo Jiang Abdulrahman Hassab Guangxin Li Wei Li Roland Assi George Tellides Department of Surgery (Cardiac), Yale School of Medicine, New Haven, CT. (P.R., B.J., A.H., G.L., W.L., R.A., G.T.) Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, CT. (R.A., G.T.) Now with Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province (B.J.). Now with Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Guangdong Province, China (G.L.). Now with Department of Vascular Surgery, Peking University People’s Hospital, Beijing, China (W.L.). Veterans Affairs Connecticut Healthcare System, West Haven, CT (R.A., G.T.). on December 19, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Smooth muscle cells (SMCs) of cardiac and neural crest origin contribute to the developing proximal aorta and are linked to disease propensity in adults.METHODS:We analyzed single-cell transcriptomes of aortic SMCs from adult mice to determine basal states and changes after disrupting TGFβ (transforming growth factor-β) signaling necessary for aortic homeostasis.RESULTS:A minority of Myh11 lineage-marked SMCs differentially expressed genes suggestive of embryological origin. Additional analyses in Nkx2-5 and Wnt1 lineage-marked SMCs derived from cardiac and neural crest progenitors, respectively, showed both lineages contributed to a major common cluster and each lineage to a minor distinct cluster. Common cluster SMCs extended from root to arch, cardiac subset cluster SMCs from root to ascending, and neural crest subset cluster SMCs were restricted to the arch. The neural crest subset cluster had greater expression of a subgroup of TGFβ-dependent genes. Nonetheless, conditional deletion of TGFβ receptors resulted in similar transcriptional changes among all SMC clusters. Several disease-associated transcriptional responses were comparable among SMC clusters in a mouse model of Marfan syndrome aortopathy, while many embryological markers of murine aortic SMCs were not detected in adult human aortas.CONCLUSIONS:There are multiple subtypes of cardiac-derived and neural crest–derived SMCs with shared or distinctive transcriptional profiles; neural crest subset cluster SMCs with increased expression of certain TGFβ-inducible genes are not spatially linked to the aortic root predisposed to aneurysms from aberrant TGFβ signaling; and loss of TGFβ responses after receptor deletion is uniform among SMC clusters.
- Pathways to Precision Medicine in Hypertrophic Cardiomyopathy: Opportunities and Challenges in Plasma Proteomicsby Usman A. Tahir Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. on December 19, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Two Hearts Beating in One Chest
- Rare Presentation and Treatment Strategy of Acute Myocardial Infarction in a Young Patientby El-Moatasem Gabr Amr Darwish Mohammed A. Chamsi-Pasha Houston Methodist DeBakey Heart and Vascular Institute, TX. on December 18, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Endothelial Cell TRPV4 Channels Turn to the Dark Side During Hypertensionby Alejandro Mata-Daboin Jonathan H. Jaggar Department of Physiology, University of Tennessee Health Science Center, Memphis. on December 18, 2024
Hypertension, Volume 82, Issue 1, Page 69-71, January 1, 2025. <br/>
- Editors and Editorial Boardon December 18, 2024
Hypertension, Volume 82, Issue 1, Page 1-2, January 1, 2025. <br/>
- Maternal Hypertension Aggravates Vascular Dysfunction After Injury in Male Adult Offspring Through Transgenerational Transmission of N6-Methyladenosineby Dezhong Zheng Jiayi Jiang Anna Shen Yixiang Zhong Yi Zhang Jiancheng Xiu Department of Cardiology, Southern Medical University, Guangzhou, China. (D.Z., J.X.) Department of Obstetrics and Gynecology, Southern Medical University, Guangzhou, China. (J.J., Y. Zhong, Y. Zhang) Nanfang Hospital, and Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China. (D.Z., A.S.) on December 17, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Whether maternal hypertension contributes to the enhanced susceptibility to vascular remodeling in adult offspring through epigenetic mechanisms remains unclear. We aimed to address this gap in the literature using a transgenerational mouse model.METHODS:Gestational hypertension was induced in pregnant mice using chronic angiotensin II infusion. Blood pressure was monitored using the tail-cuff method. Two months post-delivery, an N6-methyladenosine epitranscriptomic microarray analysis was performed on the carotid arteries of second-generation mice. A unilateral carotid artery injury model was used to study the postinjury vascular response in vivo. Furthermore, carotid ultrasonography, immunohistochemistry, and molecular biological parameters were assessed in adult offspring.RESULTS:Exposure to maternal hypertension decreased the birth weight of live pups and increased the fetal death rate. Compared with normal offspring, adult offspring with hypertension had wire-induced injury that led to greater vascular remodeling, which was associated with aggravated inflammation imbalance, fibrosis, and oxidative stress. In addition, aberrant N6-methyladenosine methylation, increased N6-methyladenosine levels, and increased METTL3 (methyltransferase-like 3) expression were detected in the vessels of offspring with hypertension. Maternal METTL3 deficiency increased the birth weight of live pups with hypertension, improved vascular dysfunction, and alleviated vascular inflammation in adult offspring with hypertension after injury.CONCLUSIONS:Maternal hypertension can induce transgenerational transmission of enhanced susceptibility to vascular remodeling, and the possible underlying mechanism is associated with altered METTL3-mediated N6-methyladenosine methylation. Therefore, this study reveals the role of epigenetic effects across generations and provides new insights into vascular remodeling causes.
- Use of Right Ventricular Free-Wall Strain in a Multivariable Estimate of Right Ventricular-Arterial Coupling in Pediatric Pulmonary Arterial Hypertensionby Charles T. Simpkin D. Dunbar Ivy Mark K. Friedberg Dale A. Burkett Division of Cardiology, Heart Institute, Children’s Hospital Colorado, Aurora (C.T.S., D.D.I., D.A.B.). University of Colorado - Anschutz Medical Campus, Aurora (C.T.S., D.D.I., D.A.B.). Division of Cardiology, Labatt Family Heart Center, Hospital for Sick Children, University of Toronto, Toronto, Canada (M.K.F.). on December 17, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e016882, December 1, 2024. BACKGROUND:Right ventricular-arterial coupling (RVAC) describes the relationship between right ventricular contractility and pulmonary vascular afterload. Noninvasive surrogates for RVAC using echocardiographic estimates of right ventricular function, such as tricuspid annular plane systolic excursion (TAPSE), have been shown to correlate with invasively measured RVAC and predict clinical outcomes in pediatric pulmonary arterial hypertension. However, given the limitations of TAPSE at accurately estimating right ventricular function in children, we hypothesized that a multivariable estimate of RVAC using right ventricular free-wall longitudinal strain (RVFW-LS) may perform better than those utilizing TAPSE at predicting clinical outcomes.METHODS:In all, 108 children from 2 institutions with pulmonary arterial hypertension underwent hemodynamic catheterization with simultaneous echocardiography. In a retrospective analysis, hybrid (echo and invasive) RVAC metrics included TAPSE/pulmonary vascular resistance (PVRi) and RVFW-LS/PVRi. Noninvasive echocardiographic metrics were TAPSE/echo-derived pulmonary artery systolic pressure (PASP) and RVFW-LS/PASP.RESULTS:RVFW-LS correlated with PVRi (r=0.315,P=0.01), though TAPSE did not (r=0.058,P=0.64). PVRi, PASP, and RVAC metrics declined in patients with worse World Health Organization Functional Class (n=108), while TAPSE and RVFW-LS did not. PVRi, PASP, RVFW-LS/PVRi, TAPSE/PVRi, and RVFW-LS/PASP predicted the outcome variable of transplant or death (area under the curve, 0.771 [P<0.001], 0.729 [P=0.004], 0.748 [P=0.002], 0.732 [P=0.009], and 0.714 [P=0.01], respectively), while TAPSE/PASP, RVFW-LS, and TAPSE did not (area under the curve, 0.671, 0.603, and 0.525, respectively). In patients without a history of repaired congenital heart disease (n=88), only RVFW-LS/PASP, PVRi, PASP, and RVFW-LS/PVRi predicted outcomes (area under the curve, 0.738 [P=0.002], 0.729 [P=0.01], 0.729 [P=0.01], and 0.729 [P=0.015], respectively).CONCLUSIONS:In the pediatric population, baseline PVRi and echo-estimated PASP were strongly associated with adverse clinical outcomes, but TAPSE and RVFW-LS were not. Estimates of RVAC utilizing RVFW-LS were superior to those utilizing TAPSE—however, only marginally additive to PASP and PVRi at predicting the adverse clinical outcome in patients without a history of repaired congenital heart disease.
- In This Issue of the Journalby Robert J. Gropler on December 17, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017863, December 1, 2024. <br/>
- Is Right Ventricular Strain the New TAPSE to Estimate Right Ventricular Arterial Coupling in Children?by Matthew Henry Lisa K. Hornberger Division of Pediatric Cardiology, Cardiology Program (M.H., L.K.H.), University of Alberta, Edmonton, Canada. Department of Pediatrics (M.H., L.K.H.), University of Alberta, Edmonton, Canada. Department of Obstetrics and Gynecology (L.K.H.), University of Alberta, Edmonton, Canada. on December 17, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017715, December 1, 2024. <br/>
- Baroreflex Sensitivity and Long-Term Dementia Risk in Older Adultsby Yuan Ma Yiwen Zhang Rikuta Hamaya Berend E. Westerhof Hossam A. Shaltout Maryam Kavousi Francesco Mattace-Raso Albert Hofman Frank J. Wolters Lewis A. Lipsitz M. Arfan Ikram Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Y.M., Y.Z., R.H., A.H.). Division of Preventive Medicine, Brigham and Women’s Hospital, Boston (R.H.). Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (B.E.W.). Westerhof Cardiovascular Research, Amstelveen, The Netherlands (B.E.W.). Hypertension and Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, NC (H.A.S.). Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands. (M.K., A.H., F.J.W., M.A.I.) Division of Geriatrics, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands. (F.M.-R.) Hebrew SeniorLife Hinda and Arthur Marcus Institute for Aging Research, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston (L.A.L.). on December 13, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Increased blood pressure (BP) variability is linked to dementia risk, but the relationship between baroreflex sensitivity (BRS), a fundamental mechanism for maintaining stable BP, and dementia risk is undetermined.METHODS:We tested the hypothesis that impaired BRS is associated with increased dementia risk in 1819 older adults (63% women; age, 71.0±6.3 years) from the community-based Rotterdam Study. Cardiac BRS was determined from a 5-minute beat-to-beat BP recording at supine rest between 1997 and 1999. Cardiac BRS measures the correlation between changes in consecutive beat-to-beat systolic BP and subsequent responses in heartbeat intervals, with a higher value indicating better BRS. The primary outcome was incident dementia ascertained from baseline through January 1, 2020; the secondary outcome was all-cause mortality.RESULTS:During a median follow-up of 14.8 years, 421 participants developed dementia. The association of cardiac BRS with dementia risk differed by antihypertensive medication use (Pinteraction=0.03) and was only observed in participants not taking antihypertensives. Specifically, in those not taking antihypertensive medication, reduced BRS was associated with a higher risk of dementia (adjusted hazard ratio comparing bottom versus top quintiles, 1.60 [95% CI, 1.07–2.40];Ptrend=0.02). Reduced BRS was also associated with an increased risk of death (corresponding hazard ratio, 1.76 [95% CI, 1.32–2.35]). The association remained after adjusting for average BP and BP variability.CONCLUSIONS:Impaired BRS partly explains hypertension-related brain damage and excessive dementia risk beyond conventional BP measures, making it a potential novel biomarker for early detection and prevention of dementia.
- Decreased Adipose Lipid Turnover Associates With Cardiometabolic Risk and the Metabolic Syndromeby Daniel P. Andersson Peter Arner Department of Medicine H7, C2:94 Karolinska Institutet, Stockholm, Sweden. Department of Endocrinology, C2:94 Karolinska University Hospital, Stockholm, Sweden. on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. BACKGROUND:Disturbed white adipose tissue function is important for cardiometabolic risk and metabolic syndrome (MetS). Whether this involves adipose lipid turnover (lipolysis and synthesis of triglycerides) is unknown and was presently investigated in subcutaneous adipose tissue, the body’s largest fat depot.METHODS:In cross-sectional studies in 78 subjects, adipose lipid age, representing overall lipid turnover (mobilization and storage), and lipid storage capacity were assessed by the incorporation of atmospheric14C into adipose lipids. Adipose lipid age from an algorithm of adipocyte lipolysis and clinical parameters was also determined in 185 subjects. Adult Treatment Panel III (ATPIII) scoring defined MetS (scores 3–5) or healthy (score 0). ANOVA or ANCOVA andttest were used for statistical comparison. Because there was no method interaction to determine lipid age, the 2 groups were combined.RESULTS:Lipid age increased by incremental ATPIII score (F=42;P<0.0001) and was 2-fold advanced in MetS (t=11.3;P<0.0001). The correlation with lipid age was independent of age, sex, body mass index, waist-to-hip ratio, sedentary lifestyle, absence of obesity, and adipose insulin resistance (F=10.7;P<0.0001). Lipid storage capacity was not related to the ATPIII score (F=1.0;P=0.44) or MetS (t=−0.9;P=0.35). Adipocyte lipolysis activation was decreased in MetS and inversely related to incremental ATPIII score, suggesting that decreased lipid mobilization is the major factor behind high lipid age in these conditions.CONCLUSIONS:Despite normal lipid assimilation capacity, abdominal subcutaneous adipose lipid turnover is decreased in MetS and high ATPIII score because of impaired ability to mobilize lipids involving low adipocyte lipolysis activation.
- Myocardial Hyperemia via Cardiomyocyte Catabolism of β-Hydroxybutyrateby Kara R. Gouwens Yibing Nong Ning Chen Emily B. Schulman-Geltzer Helen E. Collins Bradford G. Hill Matthew A. Nystoriak Department of Medicine, Division of Environmental Medicine, Center for Cardiometabolic Science, University of Louisville, KY. (K.R.G., Y.N., N.C., E.B.S.-G., H.E.C., B.G.H., M.A.N.) Department of Physiology, University of Louisville, KY. (K.R.G.) Now with Masonic Medical Research Institute, Utica, NY (M.A.N.). on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>
- Extracellular Vesicles From Preeclampsia Disrupt the Blood-Brain Barrier by Reducing CLDN5by Hermes Sandoval Belén Ibañez Moisés Contreras Felipe Troncoso Fidel O. Castro Diego Caamaño Lidice Mendez Estefanny Escudero-Guevara Francisco Nualart Hiten D. Mistry Lesia O. Kurlak Manu Vatish Jesenia Acurio Carlos Escudero Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (H.S., B.I., M.C., F.T., E.E.-G., J.A., C.E.). Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan, Chile (F.O.C., D.C., L.M.). Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile (F.N.). Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepción, Chile (F.N.). Division of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, United Kingdom (H.D.M.). Stroke Trials Unit (School of Medicine), University of Nottingham, United Kingdom (L.O.K.). Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, United Kingdom (M.V., C.E.). Group of Research and Innovation in Vascular Health, Chillan, Chile (C.E.). on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. BACKGROUND:The physiopathology of life-threatening cerebrovascular complications in preeclampsia is unknown. We investigated whether disruption of the blood-brain barrier, generated using circulating small extracellular vesicles (sEVs) from women with preeclampsia or placentae cultured under hypoxic conditions, impairs the expression of tight junction proteins, such as CLDN5 (claudin-5), mediated by VEGF (vascular endothelial growth factor), and activation of KDR (VEGFR2 [VEGF receptor 2]).METHODS:We perform a preclinical mechanistic study using sEVs isolated from plasma of pregnant women with normal pregnancy (sEVs-NP; n=9), sEVs isolated from plasma of women with preeclampsia (sEVs-PE; n=9), or sEVs isolated from placentas cultured in normoxia (sEVs-Nor; n=10) or sEVs isolated from placentas cultured in hypoxia (sEVs-Hyp; n=10). The integrity of the blood-brain barrier was evaluated using in vitro (human [hCMEC/D3] and mouse [BEND/3] brain endothelial cell lines) and in vivo (nonpregnant C57BL/6J mice [4–5 months old; n=13] injected with sEVs-Hyp) models.RESULTS:sEVs-PE and sEVs-Hyp reduced total and membrane-associated protein CLDN5 levels (P<0.05). These results were negated with sEVs-PE sonication. sEVs-Hyp injected into nonpregnant mice generated neurological deficits and blood-brain barrier disruption, specifically in the posterior area of the brain, associated with brain endothelial cell uptake of sEVs, sEVs-Hyp high extravasation, and reduction in CLDN5 levels in the brain cortex. Furthermore, sEVs-PE and sEVs-sHyp had higher VEGF levels than sEVs-NP and sEVs-Nor. Human brain endothelial cells exposed to sEVs-PE exhibited a reduction in the activation of KDR. Reduction in CLDN5 observed in cells treated with sEVs-Hyp was further enhanced in cells treated with KDR selective inhibitor.CONCLUSIONS:sEVs-PE disrupts the blood-brain barrier, an effect replicated with sEVs-Hyp, and involves reduced CLDN5 and elevated VEGF contained within these vesicles. However, our results do not support the participation of KDR activation in the downregulation of CLDN5 observed with sEVs-Hyp. These findings will improve our understanding of the pathophysiology of cerebrovascular alterations in women with preeclampsia.
- Looking Under the Hood at the Cytoskeletal Engine of Platelet Productionby Joseph E. Italiano Clementine Payne Roelof H. Bekendam Vascular Biology Program, Boston Children’s Hospital, MA (J.E.I., C.P., R.H.B.). Department of Surgery, Harvard Medical School, Boston, MA (J.E.I., C.P., R.H.B.). Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA (R.H.B.). on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>Blood platelets are anucleate cells essential for normal blood hemostasis. To maintain a normal platelet count of 150 000 to 400 000 per μL of blood, 1011platelets must be released each day from precursor cells called megakaryocytes. In this review, we aim to provide an overview of platelet production and evaluate the proposed mechanisms of platelet generation. We will discuss novel cytoskeletal mechanisms of platelet production, including microtubule and actin-based systems. We present new evidence that supports a cytoplasmic trigger for platelet production, discuss centrosome clustering as a new mechanism to trigger proplatelet production, and review new data supporting the bone marrow as the major location of platelet production.
- Semaglutide Improves Myocardial Perfusion and Performance in a Large Animal Model of Coronary Artery Diseaseby Christopher R. Stone Dwight D. Harris Mark Broadwin Meghamsh Kanuparthy Ju-Woo Nho Keertana Yalamanchili Jad Hamze M. Ruhul Abid Frank W. Sellke Department of Cardiothoracic Surgery, Brown University, Providence, RI. on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. BACKGROUND:Coronary artery disease is the leading cause of death worldwide. It imposes an enormous symptomatic burden on patients, leaving many with residual disease despite optimal procedural therapy and up to one-thirds with debilitating angina amenable neither to procedures, nor to current pharmacological options. Semaglutide (SEM), a GLP-1 (glucagon-like peptide 1) agonist originally approved for management of diabetes, has garnered substantial attention for its capacity to attenuate cardiovascular risk. Although subgroup analyses in patients indicate promise, studies explicitly designed to isolate the impact of SEM on the sequelae of coronary artery disease, independently of comorbid diabetes or obesity, are lacking.METHODS:Yorkshire swine (n=17) underwent placement of an ameroid constrictor around the left circumflex coronary artery to induce coronary artery disease. Oral SEM was initiated postoperatively at 1.5 mg and scaled up in 2 weeks to 3 mg in treatment animals (n=8) for a total of 5 weeks, while control animals (n=9) received no drug. All then underwent myocardial harvest with acquisition of perfusion and functional data using microsphere injection and pressure-volume loop catheterization. Immunoblotting, immunohistochemistry, and immunofluorescence were performed on the most ischemic myocardial segments for mechanistic elucidation.RESULTS:SEM animals exhibited improved left ventricular ejection fraction, both at rest and during rapid myocardial pacing (bothP<0.03), accompanied by increased perfusion to the most ischemic myocardial region at rest and during rapid pacing (bothP<0.03); reduced perivascular and interstitial fibrosis (bothP<0.03); and apoptosis (P=0.008). These changes were associated with increased activation of the endothelial-protective AMPK (AMP-activated protein kinase) pathway (P=0.005), coupled with downstream increases in eNOS (endothelial NO synthase;P=0.014).CONCLUSIONS:This study reveals the capacity of oral SEM to augment cardiac function in the chronically ischemic heart in a highly translational large animal model, likely through AMPK-mediated improvement in endothelial function and perfusion to the ischemic myocardium.
- DCBLD1 Modulates Angiogenesis by Regulation of the VEGFR-2 Endocytosis in Endothelial Cellsby Qi Feng Lingling Guo Chao Yu Xiaoning Liu Yanling Lin Chenyang Li Wenjun Zhang Yanhong Zong Weiwei Yang Yuehua Ma Runtao Wang Lijing Li Yunli Pei Huifang Wang Demin Liu Honglin Niu Mei Han Lei Nie Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China. (Q.F., L.G., C.Y., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.) The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China. (Q.F., L.G., C.Y., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.) Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, China. (Q.F., L.G., C.Y., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.) Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China. (Q.F., L.G., C.Y., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.) School of Nursing, Hebei Medical University, Shijiazhuang, China. (H.N.) Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China (D.L.). on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Unwanted angiogenesis is involved in the progression of various malignant tumors and cardiovascular diseases, and the factors that regulate angiogenesis are potential therapeutic targets. We tested the hypothesis that DCBLD1 (discoidin, CUB, and LCCL domain-containing protein 1) is a coreceptor of VEGFR-2 (vascular endothelial growth factor receptor-2) and modulates angiogenesis in endothelial cells.METHODS:A carotid artery ligation model and retinal angiogenesis assay were used to study angiogenesis using globe knockout or endothelial cell–specific conditionalDcbld1knockout mice in vivo. Immunoblotting, immunofluorescence staining, plasma membrane subfraction isolation, Coimmunoprecipitation, and mass spectrum assay were performed to clarify the molecular mechanisms.RESULTS:Loss ofDcbld1impaired VEGF (vascular endothelial growth factor) response and inhibited VEGF-induced endothelial cell proliferation and migration.Dcbld1deletion interfered with adult and developmental angiogenesis. Mechanistically, DCBLD1 bound to VEGFR-2 and regulated the formation of VEGFR-2 complex with negative regulators: protein tyrosine phosphatases, E3 ubiquitin ligases (Nedd4 and c-Cbl), and alsoDcbld1knockdown promoted lysosome-mediated VEGFR-2 degradation in endothelial cells.CONCLUSION:These findings demonstrated the essential role of endothelial DCBLD1 in regulating VEGF signaling and provided evidence that DCBLD1 promotes VEGF-induced angiogenesis by limiting the dephosphorylation, ubiquitination, and lysosome degradation after VEGFR-2 endocytosis. We proposed that endothelial DCBLD1 is a potential therapeutic target for ischemic cardiovascular diseases by the modulation of angiogenesis through regulation of the VEGFR-2 endocytosis.
- Sex Differences in Cardiovascular-Kidney-Metabolic Syndrome: 30-Year US Trends and Mortality Risksby Hongwei Ji Charumathi Sabanayagam Kunihiro Matsushita Ching-Yu Cheng Tyler Hyungtaek Rim Bin Sheng Huating Li Yih-Chung Tham Susan Cheng Tien-Yin Wong Tsinghua Medicine, Tsinghua University, Beijing, China (H.J., T.Y.W.). Department of Internal Medicine, Beijing Tsinghua Changgung Hospital, Beijing, China (H.J.). Singapore Eye Research Institute, Singapore National Eye Center (C.S., C.Y.C., T.H.R., Y.C.T., T.Y.W.). Ophthalmology and Visual Science Academic Clinical Program, Duke-NUS Medical School, Singapore (C.S., C.Y.C., Y.C.T.). Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.). Department of Computer Science and Engineering, Shanghai Jiao Tong University, China (B.S.). Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China (H.L.). Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (S.C.). on December 12, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. BACKGROUND:The American Heart Association recently published guidelines on how to clinically identify and categorize individuals with cardiovascular-kidney-metabolic (CKM) syndrome. The extent to which CKM syndrome prevalence and prognosis differ by sex remains unknown. This study aimed to examine the impact of sex on trends in prevalence over 30 years and the long-term prognosis of CKM syndrome in the United States.METHODS:We analyzed nationally representative National Health and Nutrition Examination Survey 1988 to 2018 data collected from 33 868 US adults (aged ≥20 years) who were under surveillance for all-cause mortality through December 31, 2019. We examined the sex-specific prevalence of CKM syndrome and sex-specific CKM associations with all-cause mortality.RESULTS:Of the 33 868 adults studied, the mean±SD age was 48.4±18.3 years with 52% women and 56% non-White. Overall prevalence of CKM syndrome increased steadily from 1988 to 2018 in both sexes, with a larger temporal rise in prevalent stage 3 CKM seen for males (from 18.9% to 22.4%) compared with women (from 13.9% to 15.2%). Over a median follow-up of 13.3 years, there were 8745 deaths. In the multivariable Cox regression analysis, worsening CKM severity was associated with all-cause mortality (P<0.001 for both sexes), with greater magnitudes of risk seen in women (hazards ratio, 1.24–3.33) compared with men (hazards ratio, 0.85–2.60) across all stages (likelihood ratio test χ2, 19.0;Pinteraction<0.001); results were similar for cardiovascular mortality (likelihood ratio test χ2, 22.3;Pinteraction<0.001).CONCLUSIONS:Women, compared with men, exhibited a lower prevalence of CKM stage 3 but experienced excess mortality risk across the spectrum of multisystem CKM dysfunction. These findings underscore the importance of identifying mechanisms underlying joint cardiovascular, kidney, and metabolic system pathophysiology to close a potentially widening sex disparities gap in multiorgan disease risk.
- Cardiac cGMP Regulation and Therapeutic Applicationsby Sumita Mishra Vivek Chander David A. Kass Center for Exercise Medicine Research, Fralin Biomedical Research Institute, Virginia Tech. (S.M., V.C.) Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech. (S.M.) Department of Human Nutrition, Foods, and Exercise, College of Life Sciences, Virginia Tech. (S.M.) Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke (S.M.). Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD. (D.A.K.) Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD. (D.A.K.) on December 11, 2024
Hypertension, Ahead of Print. <br/>cGMP plays a central role in cardiovascular regulation in health and disease. It is synthesized by NO or natriuretic peptide activated cyclases and hydrolyzed to 5′GMP by select members of the PDEs (phosphodiesterase) superfamily. The primary downstream effector is cGMP-dependent protein kinase, primarily cGK-1a also known as protein kinase G 1a in the heart and vasculature. cGMP signaling is controlled in intracellular nanodomains to regulate myocyte growth, survival, metabolism, protein homeostasis, G-protein–coupled receptor signaling, and other critical functions. The vascular effects of cGMP signaling have been dominated by its lowering of smooth muscle tone, but other cellular processes are also engaged. Localization of cyclases and corresponding PDEs within intracellular domains, along with their varying expression across different cell types, adds multiorgan complexity to cGMP signaling. This diversity can be leveraged therapeutically by targeting selective pathway components to impact some but not other cGMP signaling effects. Here, we review the generation and regulation of cGMP by PDEs and cyclases, focusing mainly on their role in cardiac physiology and pathophysiology. Current therapeutic uses of cGMP modulation and ongoing trials testing new potential applications are discussed.
- Characterizing the Origins of Primary Aldosteronismby Jenifer M. Brown Brooke Honzel Laura C. Tsai Julia Milks Yvonne Neibuhr Andrew J. Newman Michael Cherney David Stouffer Richard J. Auchus Anand Vaidya Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension (J.M.B., B.H., L.C.T., J.M., Y.N., A.J.N., A.V.). Division of Cardiovascular Medicine (J.M.B.). Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.M.B., B.H., L.C.T., J.M., Y.N., A.J.N., A.V.). Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor (M.C., D.S., R.J.A.). Endocrinology and Metabolism Section, LTC Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, MI (R.J.A.). on December 11, 2024
Hypertension, Ahead of Print. BACKGROUND:Renin-independent aldosterone production in normotensive people increases risk for developing hypertension. In parallel, normotensive adrenal glands frequently harbor aldosterone-producing micronodules with pathogenic somatic mutations known to induce primary aldosteronism (PA). A deeper understanding of these phenomena would inform the origins of PA and its role in hypertension pathogenesis.METHODS:Prospectively recruited normotensives underwent detailed characterization of PA features via the following: oral sodium suppression test to evaluate renin-independent aldosterone production, dexamethasone suppression and adrenocorticotropic hormone-stimulation tests to evaluate adrenocorticotropic hormone-mediated aldosterone production, and 24-hour ambulatory blood pressure monitoring. The magnitude of renin-independent aldosterone production was defined via using tertiles of 24-hour urinary aldosterone production during the oral sodium suppression test to create unbiased categorizations of the magnitude of PA. Serum aldosterone, serum 18-hybrid steroids, urine tetrahydroaldosterone (biomarkers of aldosterone synthase activity), urinary potassium, and blood pressure (biomarkers of mineralocorticoid receptor activation) were evaluated across tertiles.RESULTS:There was a spectrum of autonomous, nonsuppressible, and renin-independent production of aldosterone, 18-hybrid steroids, and 24-hour urinary tetrahydroaldosterone (P-trend <0.01). Correspondingly, there was a continuum of adrenocorticotropic hormone-mediated aldosterone production and 18-hybrid steroid production that also paralleled renin-independent aldosterone production. The spectrum of PA pathophysiology was associated with higher ambulatory daytime systolic BP (P-trend <0.05), even within the normotensive range, and greater urinary potassium excretion (P-trend <0.05), indicating a continuum of mineralocorticoid receptor activation.CONCLUSIONS:The pathophysiologic continuum of PA, characterized by renin-independent and adrenocorticotropic hormone-mediated aldosterone production, and enhanced aldosterone synthase and mineralocorticoid receptor activity, is evident in normotensive people. These findings provide mechanistic explanations to implicate PA in the pathogenesis of a substantial proportion of hypertension.
- Elevated Donor-Derived Cell-Free DNA Levels Are Associated With Reduced Myocardial Blood Flow but Not Angiographic Cardiac Allograft Vasculopathy: The EVIDENT Studyby Cathrine M. Moeller Daniel Oren Andrea Fernandez Valledor Gal Rubinstein Ersilia M. DeFilippis Salwa Rahman Yonatan Mehlman Elena M. Donald Dor Lotan Edward Lin Kyung T. Oh Sun H. Lee Jayant K. Raikhelkar Justin A. Fried David Majure Farhana Latif Gabriel T. Sayer Nir Uriel Kevin J. Clerkin Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center (C.M.M., D.O. A.F.V., G.R., E.M. DeFilippis, S.R., Y.M., E.M. Donald, D.L., E.F.L., K.T.O., S.H.L., J.K.R., J.A.F., F.L., G.T.S., N.U., K.J.C.). Greenberg Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Weill Cornell Medical College (D.M.). on December 10, 2024
Circulation: Heart Failure, Ahead of Print. BACKGROUND:Cardiac allograft vasculopathy (CAV) leads to impaired myocardial blood flow (MBF), increasing the risk of cardiovascular death or retransplant among heart transplantation (HT) recipients. Data on elevation in donor-derived cell-free DNA (dd-cfDNA) and CAV in the absence of rejection are mixed. We sought to test the hypothesis that CAV with reduced MBF (RMBF) is associated with elevated dd-cfDNA.METHODS:A retrospective review was conducted on HT recipients at a high-volume center who underwent dd-cfDNA testing between September 2019 and November 2022. Inclusion criteria included undergoing CAV screening with cardiac positron emission tomography scans and coronary angiograms. Patients were grouped by the presence of angiographic CAV diagnosis and MBF reserve evaluated through cardiac positron emission tomography. The latter was subdivided into normal MBF or RMBF, with RMBF defined as an MBF reserve ≤2. Elevated dd-cfDNA was defined as ≥0.12%.RESULTS:Two hundred fifty-six HT recipients were included (median age, 55 years; 27.6% female; median, 8 years [interquartile range (IQR), 5–14] post-HT). Ischemic etiology of heart failure was more prevalent in the RMBF group (36%) compared with the normal MBF group (20%;P=0.02). The prevalence and magnitude of a positive dd-cfDNA test with angiographic CAV (29%; median, 0.26% [IQR, 0.15%–0.62%]) were not significantly different from those without CAV (30%;P=0.94; median, 0.31% [IQR, 0.17%–0.71%];P=0.38). However, RMBF patients exhibited significantly higher dd-cfDNA prevalence and levels (51%; median, 0.81% [IQR, 0.48%–1.11%]) compared with normal MBF patients (27%;P<0.001; median, 0.25% [IQR, 0.15%–0.52%];P<0.001).CONCLUSIONS:HT recipients with angiographic CAV had similar dd-cfDNA levels and rates as those without. Notably, dd-cfDNA levels and rates were significantly elevated in patients with RMBF assessed by positron emission tomography compared with those with normal MBF.
- Insights From a 20-Year Follow-Up of the First Heart Transplant Recipient to Complete an Ironman Triathlonby Stephen J. Foulkes Rachel J. Skow Andre La Gerche Wayne J. Tymchak Mark J. Haykowsky Integrated Cardiovascular Exercise Physiology and Rehabilitation (iCARE) Lab, Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, AB, Canada (S.J.F., R.J.S., M.J.H.). Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada (W.J.T). Heart, Exercise and Research Trials (HEART) Lab, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia (S.J.F., A.L.G.). on December 9, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Cognitive and Procedural Competencies in the Cardiac Intensive Care Unitby Willard N. Applefeld Jacob C. Jentzer Saraschandra Vallabhajosyula Division of Cardiovascular Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC (W.N.A.). Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (J.C.J.). Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University and Brown University Health Cardiovascular Institute, Providence, RI (S.V.). on December 9, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Aerobic Capacity of Adults With Fontan Palliation: Disease-Specific Reference Values and Relationship to Outcomesby Alexander C. Egbe Ahmed E. Ali William R. Miranda Heidi M. Connolly Barry A. Borlaug Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN. on December 9, 2024
Circulation: Heart Failure, Ahead of Print. BACKGROUND:Patients with Fontan palliation have reduced aerobic capacity because of impaired cardiac, pulmonary, and skeletal muscle function. However, the assessment of aerobic capacity in this population still relies on comparisons with people without cardiovascular disease rather than comparison with the expected aerobic capacity of other Fontan patients. The purpose of this study was to determine the expected aerobic capacity of adults with Fontan palliation.METHODS:Adults with Fontan palliation who underwent a cardiopulmonary exercise test at Mayo Clinic (2003–2023) were stratified into quartiles based on the predicted peak oxygen consumption (VO2). We assessed the correlates of predicted peak VO2and the relationship between predicted peak VO2quartiles and cardiovascular outcomes (death/transplant).RESULTS:Of 323 patients (age, 29±9 years; 177 [55%] men), the median peak VO2was 19.1 (15.2–23.9) mL/kg per minute, and this corresponds to a predicted peak VO2of 51% (range, 19–88; interquartile range, 41–62). After multivariable adjustments, the correlates of predicted peak VO2were body mass index (β±SE, −2.61±0.95; 2.61% decrease in predicted peak VO2per 5 kg/m2increase in body mass index;P=0.009), systemic saturation (β±SE, 3.65±0.85; 3.65% increase in predicted peak VO2per 5% increase in oxygen saturation;P<0.001), and Fontan pressure (β±SE, −1.24±0.22; 1.24% decrease in predicted peak VO2per 1 mm Hg increase in Fontan pressures;P<0.001). There was a 47% increase in the risk for death/transplant from a higher predicted peak VO2quartile to the next lower quartile (adjusted hazard ratio, 1.47 [95% CI, 1.09–2.05];P=0.01).CONCLUSIONS:The results of the current study would help calibrate the interpretation of exercise test data in adults with Fontan palliation and improve risk stratification in this population. It also underscores the need to maintain normal Fontan hemodynamics and body weight, which are important determinants of aerobic capacity.
- Burden of Cardio-Cerebrovascular and Renal Diseases Attributable to Systolic Hypertension in France in 2021by Clémence Grave Christophe Bonaldi Laure Carcaillon-Bentata Amélie Gabet Jean-Michel Halimi Christophe Tzourio Yannick Béjot Marion J. Torres Philippe Gabriel Steg Isabelle Durand Zaleski Jacques Blacher Valérie Olié Direction des maladies non transmissibles, Santé publique France, Saint-Maurice, France (C.G., A.G., M.J.T., V.O.). Direction Appui, Traitements et Analyses de données, Santé publique France, Saint-Maurice, France (C.B.). University of Bordeaux, INSERM CIC-P 1401, France (L.C.-B.). Service de Néphrologie-hypertension, Dialyses, Transplantation Rénale, Hôpital Bretonneau et hôpital Clocheville, France (J.-M.H.). INSERM U1327, University de Tours, France (J.-M.H.). Université Bordeaux, Inserm, Bordeaux Population Health Research Center, France (C.T.). Service de neurologie, CHU Dijon Bourgogne, Université de Bourgogne, Dijon, France (Y.B.). Université Paris Cité, AP-HP Hôpital Bichat, and INSERM 1148, France (P.G.S.). Unité de recherche clinique en économie de la santé Hôtel Dieu, Paris, France (I.D.Z.). Centre de diagnostic et de thérapeutique, Hôtel Dieu, AP-HP, Université Paris Cité, France (J.B.). Equipe de Recherche en Epidemiologie Nutritionnelle, Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in epidemiology and Statistics, Bobigny, France (J.B.). on December 9, 2024
Hypertension, Ahead of Print. BACKGROUND:Hypertension is the most common chronic disease and a major modifiable risk factor for cardio-cerebrovascular and renal diseases. This study estimated the national burden of hypertension, defined as systolic blood pressure ≥140 mm Hg, on morbidity and mortality in 2021 in France.METHODS:For all diseases causally associated with hypertension (cardiovascular diseases, chronic kidney diseases, and dementia), the number and proportion of cases attributable to hypertension in adults aged ≥35 years were estimated using population attributable fractions. Age- and sex-specific population attributable fractions were computed using the distribution of hypertension in the French population. These population attributable fractions were applied to nationwide statistics for mortality, hospitalizations, disease prevalence, years of life lost, years of life lived with disability, and disability-adjusted years of life.RESULTS:The largest population attributable fractions were for ischemic heart disease and hemorrhagic stroke, with over 40% of cases attributable to hypertension. Overall, >385 000 patients were hospitalized due to hypertension, with 3.7 million hospitalizations and 6.2 million hospital days (all hospitalizations, including 3.4 million for chronic kidney disease) and including 390 000 overnight hospitalization. In 2021, >1.15 million individuals lived with ischemic heart disease attributable to hypertension, 1.26 million with chronic kidney diseases, and 358 033 with heart failure. Among 184 059 annual deaths from cardiovascular diseases, dementia, and chronic kidney diseases, 30% (55 280 deaths) were attributable to hypertension. Hypertension accounted for 8.5% of all deaths and 498 052 years of life lost.CONCLUSIONS:In France, despite near-universal health coverage and free health care access, the burden attributable to hypertension remains high.
- Clinical Management and Transplant Considerations in Pediatric Pulmonary Hypertension Due to Left Heart Disease: A Scientific Statement From the American Heart Associationby Rachel K. Hopper Georg Hansmann Seth A. Hollander Anne I. Dipchand Oscar van der Have Colleen Iler Cynthia Herrington Erika B. Rosenzweig Juan C. Alejos Karin Tran-Lundmark on December 9, 2024
Circulation: Heart Failure, Ahead of Print. <br/>Children with left heart disease are at risk for developing pulmonary hypertension, initially secondary to pulmonary venous hypertension that can progress to include elevated pulmonary vascular resistance, known as combined pre- and postcapillary pulmonary hypertension. Elevated pulmonary vascular resistance may pose a risk to the right ventricle of a newly transplanted heart because of increased afterload and is an important consideration for heart transplant eligibility. However, the epidemiology, pathophysiology, optimal diagnostic and treatment approaches, and thresholds for pulmonary vascular resistance in pulmonary hypertension associated with left heart disease remain unclear because of lack of evidence, particularly in pediatrics. The result is heterogeneity with respect to hemodynamic assessment, use of pulmonary vasodilator therapies, and heart transplant listing. This scientific statement aims to synthesize the available data and highlight areas of general consensus as well as important knowledge gaps.
- Association of T-Cell Phenotypes With Peri-Coronary Inflammation in People With and Without HIV and Without Cardiovascular Diseaseby Michael L. Freeman Mian B. Hossain Shana A.B. Burrowes Jean Jeudy Felisa Diaz-Mendez Sarah E. Mitchell Sumanth D. Prabhu Michael M. Lederman Shashwatee Bagchi Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH (M.L.F., M.M.L.). Department of Statistics, School of Community Health and Policy, Morgan State University, Baltimore, MD (M.B.H.). Section of Infectious Diseases, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, MA (S.A.B.B.). Evans Center for Implementation and Improvement Sciences, Boston University Chobanian and Avedisian School of Medicine, MA (S.A.B.B.). Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore (J.J.). Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD (F.D.-M., S.E.M.). Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO. (S.D.P.) Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO. (S.B.) on December 6, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>BACKGROUND:Persistent immune activation is linked to elevated cardiovascular diseases in people with HIV on antiretroviral therapy. The fat attenuation index (FAI) is a measure of peri-coronary inflammation that independently predicts cardiovascular disease risk in people without HIV. Whether FAI is associated with immune activation is unknown.METHODS:Peripheral blood T-cell activation and homing phenotypes were measured in people with HIV (n=58) and people without HIV (n=16) without known cardiovascular disease who underwent coronary computed tomography angiography and had FAI measurements. A cross-sectional analysis of an observational cohort was performed. The primary aim was to evaluate associations of T-cell activation and phenotypes with the outcome variables, FAI values of the right coronary artery and left anterior descending artery, which were assessed using multivariable regression models adjusted for age, natal sex, race, low-density lipoprotein cholesterol, body mass index, and use of lipid-lowering medication.RESULTS:T cells from people with HIV showed greater activation, as measured by cluster of differentiation (CD) 38/human leukocyte antigen – DR isotype coexpression on CD4 central memory and terminally-differentiated effector memory subsets and on CD8 effector memory (TEM), than did cells from people without HIV. Expression of the chemokine receptor C-C Chemokine Receptor 2 was reduced on CD4 central memory and TEM and CD8 TEM and terminally-differentiated effector memory subsets in people with HIV. Among all participants, PD-1 (programmed cell death 1) in CD8 central memory was associated with worsened peri-coronary inflammation of the right coronary artery, whereas perforin/granzyme B on CD8 TEM was associated with improved peri-coronary inflammation of the right coronary artery and left anterior descending artery in adjusted analyses. When accounting for HIV serostatus, CD38/human leukocyte antigen – DR isotype coexpression on CD8 central memory, TEM, and terminally-differentiated effector memory cells was associated with more peri-coronary inflammation of the left anterior descending artery.CONCLUSIONS:The associations between T-cell activation with FAI are novel and suggest that T-cell activation may be an important driver of peri-coronary inflammation, occurring at an early stage of atherosclerosis, even before the development of clinical disease.
- Sympathetic Response to 1-Leg Cycling Exercise Predicts Exercise Capacity in Patients With Heart Failure With Preserved Ejection Fractionby Mark B. Badrov Tomoyuki Tobushi Catherine F. Notarius Evan Keys Massimo Nardone David Z. Cherney Susanna Mak John S. Floras University Health Network and Sinai Health, Department of Medicine, University of Toronto, ON, Canada. (M.B.B., T.T., C.F.N., E.K., M.N., D.Z.C., S.M., J.S.F.) Faculty of Kinesiology and Physical Education (C.F.N.) Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.B.B., T.T., C.F.N., E.K., M.N., D.Z.C., J.S.F.). Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada (M.B.B., T.T., S.M., J.S.F.). on December 6, 2024
Circulation: Heart Failure, Ahead of Print. BACKGROUND:In heart failure, sympathetic excess and exercise intolerance impair quality of life. In heart failure with reduced ejection fraction, exercise stimulates a reflex increase in muscle sympathetic nerve activity (MSNA) that relates inversely to peak oxygen uptake (V̇O2peak). Whether similar sympathoexcitatory responses are present in heart failure with preserved EF (HFpEF) and relate to V̇O2peakare unknown.METHODS:In 13 patients with HFpEF (70±6 years), 17 comorbidity-matched controls (CMC; 67±8 years), and 18 healthy controls (65±8 years), we measured heart rate, blood pressure, and MSNA (microneurography) during (1) 7-minute baseline; (2) 2-minute isometric handgrip (40% maximal voluntary contraction) or rhythmic handgrip (50% and 30% maximal voluntary contraction) exercise, followed by 2-minute postexercise circulatory occlusion; and (3) 4-minute 1-leg cycling (2 minutes each at mild and moderate intensity). V̇O2peakwas obtained by open-circuit spirometry.RESULTS:Resting MSNA was higher and V̇O2peakwas lower in HFpEF versus CMCs and healthy controls (allP<0.05). During handgrip, MSNA increased in all groups (allP<0.05); in HFpEF, MSNA was greater than CMCs and healthy controls during HG and postexercise circulatory occlusion at 40% isometric handgrip (allP<0.05) and HG only at 50% and 30% rhythmic handgrip (allP<0.05). During cycling, MSNA (bursts·min−1) decreased during mild (−4±4;P=0.01) and moderate (−8±6;P<0.001) cycling in healthy controls, was unchanged during mild (+1±7;P=0.42) and moderate (+2±8;P=0.28) cycling in CMCs, yet increased in HFpEF during mild (+8±8;P<0.001) and moderate (+9±10;P<0.001) cycling. In HFpEF, the change in MSNA during moderate cycling related inversely to relative (r=−0.72;R2=0.51;P<0.01) and percent-predicted (r=−0.63;R2=0.39;P=0.03) V̇O2peak. No statistically significant relationships were detected in controls (P>0.05).CONCLUSIONS:In contrast to CMCs, patients with HFpEF exhibit augmented MSNA at rest and during exercise. The magnitude of such paradoxical sympathoexcitation during dynamic cycling relates inversely to V̇O2peak, consistent with a neurogenic, vasoconstrictor limit on exercise capacity in HFpEF.
- Orthostatic and Standing Hypertension and Risk of Cardiovascular Diseaseby Sean W. Dooley Fredrick Larbi Kwapong Hannah Col Ruth-Alma N. Turkson-Ocran Long H. Ngo Jennifer L. Cluett Kenneth J. Mukamal Lewis A. Lipsitz Mingyu Zhang Natalie R. Daya Elizabeth Selvin Pamela L. Lutsey Josef Coresh Beverly Gwen Windham Lynne Wagenknecht Stephen P. Juraschek Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (S.W.D., F.L.K., H.C., R.-A.N.T.-O., L.H.N., J.L.C., K.J.M., L.A.L., M.Z., S.P.J.). Harvard Medical School, Boston, MA (R.-A.N.T.-O., L.H.N., K.J.M., M.Z., S.P.J.). Hebrew SeniorLife Marcus Center (L.A.L.). Department of Epidemiology, Johns Hopkins University, Baltimore, MD (N.R.D., E.S.). University of Minnesota, Minneapolis (P.L.L.). New York University Grossman School of Medicine (J.C.). University of Mississippi, University (B.G.W.). Wake Forest University School of Medicine, Winston-Salem, NC (L.W.). on December 5, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Orthostatic hypertension is an emerging risk factor for adverse events. Recent consensus statements combine an increase in blood pressure upon standing with standing hypertension, but whether these 2 components have similar risk associations with cardiovascular disease (CVD) is unknown.METHODS:The ARIC study (Atherosclerosis Risk in Communities) measured supine and standing blood pressure during visit 1 (1987–1989). We defined systolic orthostatic increase (a rise in systolic blood pressure [SBP] ≥20 mm Hg, standing minus supine blood pressure) and elevated standing SBP (standing SBP ≥140 mm Hg) to examine the new consensus statement definition (rise in SBP ≥20 mm Hg and standing SBP ≥140 mm Hg). We used Cox regression to examine associations with incident coronary heart disease, heart failure, stroke, fatal coronary heart disease, and all-cause mortality.RESULTS:Of 11 369 participants (56% female; 25% Black adults; mean age, 54 years) without CVD at baseline, 1.8% had systolic orthostatic increases, 20.1% had standing SBP ≥140 mm Hg, and 1.3% had systolic orthostatic increases with standing SBP ≥140 mm Hg. During up to 30 years of follow-up, orthostatic increases were not significantly associated with any of the adverse outcomes of interest, while standing SBP ≥140 mm Hg was significantly associated with all end points. In joint models comparing systolic orthostatic increases and standing SBP ≥140 mm Hg, standing SBP ≥140 mm Hg was significantly associated with a higher risk of CVD, and associations differed significantly from systolic orthostatic increases.CONCLUSIONS:Unlike systolic orthostatic increases, standing SBP ≥140 mm Hg was strongly associated with CVD outcomes and death. These differences in CVD risk raise important concerns about combining systolic orthostatic increases and standing SBP ≥140 mm Hg in a consensus definition for orthostatic hypertension.
- Cross-Sex Hormone Therapy Is Associated With Loss of Circadian Rhythm in the Male Ratby Jordan H. Mallette Breland F. Crudup Adrian Oudomrath Speyrer Adam Z. Rawls Kathy Cockrell Alex T. Willis Kacey Davenport Licy L. Yanes Cardozo Noha M. Shawky Barbara T. Alexander Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson. (J.H.M., B.F.C., A.Z.R., K.C., A.T.W., B.T.A.) Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson. (K.D.) Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (L.L.Y.C., N.M.S.) School of Medicine, University of Mississippi Medical Center, Jackson. (A.O.S.) on December 5, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Transgender women are individuals born male but identify as female. Many transgender women undergo gender-affirming hormone therapy to alleviate the distress that can occur due to gender incongruence. For transgender women, gender-affirming hormone therapy includes 17β-estradiol (E2) combined with an antiandrogen therapy (AA) or surgical intervention. Numerous studies suggest that the risk of cardiovascular disease is elevated in transgender women; yet, the biological effects of gender-affirming hormone therapy on cardiovascular health are unknown. We hypothesize that a shift in the hormonal milieu versus natal sex in the male rat is associated with an increase in blood pressure at baseline and an enhanced responsiveness to a hypertensive challenge.METHODS:We developed clinically relevant models that mimic gender-affirming hormone therapy combination therapies utilized for the endocrine treatment of gender dysphoria in transgender women.RESULTS:Chronic E2 plus castration or the E2+antiandrogen spironolactone was associated with a significant reduction in lean mass and testosterone. At baseline, 24-hour mean arterial pressure did not differ in E2+castration or E2+antiandrogen therapy versus control, but circadian rhythm was disrupted. In response to chronic Ang II (angiotensin II; 200 ng/kg per minute), the Ang II-induced increase in blood pressure was attenuated in E2+castration compared with control, but the blood pressure response to Ang II was similar in E2+antiandrogen therapy versus control.CONCLUSIONS:Thus, these data indicate that the type of combination therapy utilized may exert differential effects on blood pressure and that disruption of circadian rhythm may be a contributory factor to the increased risk of adverse cardiovascular outcomes in transgender women exposed to high 17β-estradiol coupled to androgen suppression.
- microRNA and Hypertensionby Lishu He Yong Liu Michael E. Widlansky Alison J. Kriegel Qiongzi Qiu Mingyu Liang Department of Physiology, University of Arizona College of Medicine-Tucson (L.H., Y.L., Q.Q., M.L.). Department of Physiology, Medical College of Wisconsin, Milwaukee. (L.H., A.J.K.) Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee. (M.E.W.) on December 5, 2024
Hypertension, Ahead of Print. <br/>Several microRNAs (miRNAs) strongly influence blood pressure and the development of hypertension by modulating vascular, renal, and other physiological mechanisms. In addition, miRNAs may contribute to the genetic regulation of blood pressure. Future research should focus on investigating select miRNAs with potent physiological effects, understanding cellular context–dependent mechanisms conferring specificity to miRNA action, and integrating miRNAs as powerful modulators into the molecular system that underlies the regulation of blood pressure and the development of hypertension.
- Ang-(1-7) and ET-1 Interplay Through Mas and ETB Receptor Interaction Defines a Novel Vasoprotective Mechanismby Augusto C. Montezano Jithin Kuriakose Katie Y. Hood Yuan Yan Sin Livia L. Camargo Yoon Namkung Carlos H. Castro Robson A. Santos Rheure Alves-Lopes Gonzalo Tejeda Patricia Passaglia Sehrish Basheer Emily Gallen Jane E. Findlay Fazli R. Awan Stéphane A. Laporte Margaret R. MacLean George S. Baillie Rhian M. Touyz Research Institute of McGill University Health Centre, Canada (A.C.M., L.L.C., Y.N., S.A.L., R.M.T.). BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.). Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Federal University of Goiás, Brazil (C.H.C.). Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Federal University of Minas Gerais, Brazil. School of Medicine, University of Aberdeen, United Kingdom (R.A.S.). Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Brazil (R.A.-L.). Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering, Pakistan (P.P.). Department of Pharmacology and Therapeutics, McGill University, Canada. (S.B., F.R.A., S.A.L.) Department of Medicine, McGill University, Canada. (S.A.L., R.M.T.) Department of Family Medicine, McGill University, Canada. (M.R.M., R.M.T.) Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, United Kingdom. on December 5, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Ang-(1-7) (angiotensin (1-7)) via MasR (Mas receptor) opposes vaso-injurious actions of Ang II (angiotensin II) as shown in models of pulmonary hypertension. The underlying mechanisms remain unclear. We hypothesized cross talk between Ang-(1-7) and the protective arm of the ET-1 (endothelin-1) system involving MasR and ETBR (endothelin receptor type B).METHODS/RESULTS:To address this, we studied multiple models: in vivo, in a mouse model of ET-1–associated vascular injury (hypoxia-induced pulmonary hypertension); ex vivo, in isolated mouse arteries; and in vitro, in human endothelial cells. Pulmonary hypertension mice exhibited pulmonary vascular remodeling, endothelial dysfunction, and ET-1–induced hypercontractility. Ang-(1-7) treatment (14 days) ameliorated these effects and increased the expression of vascular ETBR. In human endothelial cells, Ang-(1-7)–induced activation of eNOS (endothelial NO synthase)/NO was attenuated by A779 (MasR antagonist) and BQ788 (ETBR antagonist). A779 inhibited ET-1–induced signaling. Coimmunoprecipitation and peptide array experiments demonstrated the interaction between MasR and ETBR. Binding sites for ETBR were mapped to MasR (amino acids 290–314). Binding sites for MasR on ETBR were identified (amino acids 176–200). Peptides that disrupt MasR:ETBR prevented Ang-(1-7) and ET-1 signaling. Using high-throughput screening, we identified compounds that enhance MasR:ETBR interaction, which we termed enhancers. Enhancers increased Ang-(1-7)–induced eNOS activity, NO production, and Ang-(1-7)–mediated vasorelaxation, and reduced contractile responses.CONCLUSIONS:We identify cross talk between Ang-(1-7) and ET-1 through MasR:ETBR interaction as a novel network that is vasoprotective. Promoting coactivity between these systems amplifies Ang-(1-7) signaling, increases ET-1/ETBR-mediated vascular actions, and attenuates the injurious effects of ET-1. Enhancing Ang-(1-7)/MasR:ET-1/ETBR signaling may have therapeutic potential in conditions associated with vascular damage.
- Epigenetic Upregulation of Carotid Body Angiotensin Signaling Increases Blood Pressureby Fengli Zhu Zhuqing Wang Kayla Davis Hayden McSwiggin Jekaterina Zyuzin Jie Liu Wei Yan Virender K. Rehan Nicholas Jendzjowsky The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.) Division of Metabolic Diseases and Translational Genomics, Harbor-UCLA Medical Center, Torrance. (W.Y.) Division of Neonatology, Harbor-UCLA Medical Center, Torrance. (V.K.R.) Division of Respiratory and Critical Care Medicine and Physiology, Harbor-UCLA Medical Center, Torrance. (N.J.) David Geffen School of Medicine, University of California, Los Angeles (W.Y., V.K.R., N.J.). on December 5, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Epigenetic changes can be shaped by a wide array of environmental cues, maternal health, and behaviors. One of the most detrimental behaviors to the developing fetus is nicotine exposure. Perinatal nicotine exposure remains a significant risk factor for cardiovascular health and, in particular, hypertension. Increased basal carotid body (CB) activity and excitation are significant contributors to hypertension. This study investigated the epigenetic changes to CB activity induced by perinatal nicotine exposure resulting in CB-mediated hypertension.METHODS:We used a rodent model of perinatal nicotine exposure and cell culture methods.RESULTS:We show that the AgtR1 (angiotensin II type 1 receptor) is upregulated in the carotid bodies of nicotine-exposed offspring. These changes were attributed to an upregulation of genetic promotion as DNA methylation of AgtR1 occurred within intron regions, exemplifying an upregulation of genetic transcription for these genes. Nicotine increased angiotensin signaling in vitro. CB reactivity to angiotensin was increased in perinatal nicotine-exposed offspring compared with control offspring. Furthermore, CB denervation reduced arterial pressure because of suppressed efferent sympathetic activity in perinatal nicotine-exposed offspring.CONCLUSIONS:Our data demonstrate that perinatal nicotine exposure adversely affects CB afferent sensing, which augments efferent sympathetic activity to increase vasoconstrictor signaling and induce hypertension. Targeting angiotensin signaling in the carotid bodies may provide a way to alleviate hypertension acquired by adverse maternal uterine environments in general and perinatal nicotine exposure in particular.
- Cost-Effectiveness of Intensive Blood Pressure Control in Youth With Chronic Kidney Diseaseby Carol L. Vincent Katherine A. Poehling Joseph Rigdon Christopher L. Schaich Andrew M. South Stephen M. Downs Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC. (C.L.V., K.A.P., A.M.S., S.M.D.) Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC. (J.R.) Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC. (C.L.S., A.M.S.) Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC. (K.A.P., A.M.S.) on December 5, 2024
Hypertension, Ahead of Print. BACKGROUND:Intensive blood pressure (BP) control in youth with chronic kidney disease (CKD) slows progression, delaying the need for kidney replacement therapy (KRT). Most youth with CKD have hypertension and BP control is difficult to achieve outside of controlled experimental settings. Implementing effective BP control strategies in this population may be cost-saving despite requiring additional resources. Our objective was to determine the economic and clinical impact of intensive versus usual care for BP management in youth with CKD in a microeconomic model.METHODS:We developed a decision tree from the US payer perspective to estimate the total costs and clinical effect of an intensified BP intervention over 5 years, modeled after the ESCAPE trial (Effect of Strict Blood Pressure Control and Angiotensin-Converting Enzyme [ACE] Inhibition on Progression of Chronic Renal Failure in Pediatric Patients) protocol. We compared this intervention to usual care in a hypothetical population of youth with mild-to-moderate CKD. Probabilities were informed by published literature; cost estimates were informed by publicly available data. Our outcomes were the net discounted cost of an intensive BP intervention, number needed to treat with the intervention to prevent 1 KRT episode, and incremental cost per KRT episode avoided.RESULTS:An intensive BP intervention, with a goal of an average 24-hour mean arterial pressure <50th percentile, improved outcomes with net cost savings of $9440 per participant over 5 years compared with usual care. To prevent 1 episode of KRT over 5 years, 13 participants need to receive intensive BP intervention.CONCLUSIONS:Routine use of the ESCAPE protocol for intensive BP control in youth with CKD could save overall costs for the payer and improve clinical outcomes.
- Platelet-Specific Deletion of TGF-β1 Impairs Septic Thrombosis in Miceby Yingying Li Huimin Jiang Xinyi Li Hui Zhu Yue Dai Jie Zhang Yueyue Sun Xiang Chu Wen Ju Mengdi Xu Zhenyu Li Lingyu Zeng Kailin Xu Jianlin Qiao Department of Hematology, Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China. on December 5, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Sepsis is featured as a systemic inflammation and thrombosis induced by infection. TGF-β (transforming growth factor-β) 1 is mainly secreted from platelets and plays a role in immune response and inflammation. Whether platelet-derived TGF-β1 participates in sepsis remains unclear. This study intends to investigate its role in sepsis in mice.METHODS:Platelet-specific TGF-β1 knockout mice received cecal ligation and puncture surgery to induce sepsis followed by the analysis of survival time, platelets number, pathology changes of lung and liver, liver function, the recruitment of platelets, neutrophils and monocytes, and neutrophil extracellular traps’ formation. In addition, TGF-β1 was administrated into platelet-specific TGF-β1 knockout mice to further evaluate its role in the pathogenesis of sepsis.RESULTS:TGF-β1 level was gradually increased in the lung during the progress of sepsis, and platelets are the major source of the elevated TGF-β1 level in the lung after sepsis. Deficiency of platelet-derived TGF-β1 prolonged the survival of sepsis mice, inhibited the drop of platelet number and bacterial growth, impaired the thrombus formation in the lung and liver, and improved liver function. In addition, platelet TGF-β1 deficiency also decreased the recruitment of neutrophils and monocytes to the lung and impaired neutrophil extracellular trap formation. However, the adoptive transfer of normal platelets to platelet-specific TGF-β1 knockout mice significantly reduced the number of circulating platelets, increased thrombosis in the lung and liver, and promoted the neutrophil extracellular trap formation.CONCLUSIONS:Deficiency of platelet-derived TGF-β1 inhibits septic thrombosis and prolongs survival time, indicating that it might be a novel therapeutic target for the treatment of sepsis.
- Clinical and Genomic Prediction of Coronary Artery Disease Subtypesby Lathan Liou Judit García-González Hei Man Wu Zhe Wang Clive J. Hoggart Amy R. Kontorovich Jason C. Kovacic Paul F. O’Reilly Department of Genetics and Genomic Sciences, Icahn School of Medicine, New York, NY. (L.L., J.G.-G., H.M.W., C.J.H., P.F.O.R.) Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine, New York, NY. (Z.W.) Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine, New York, NY. (A.R.K., J.C.K.) Cardiovascular Research Institute, Icahn School of Medicine, New York, NY. (A.R.K.) Biomedical Engineering and Imaging Institute, Icahn School of Medicine, New York, NY. (A.R.K.) The Institute for Genomic Health, Icahn School of Medicine, New York, NY. (A.R.K.) Department of Cardiology, St Vincent’s Hospital, Sydney, NSW, Australia (J.C.K.). Faculty of Medicine and Health, University of New South Wales, Sydney, Australia (J.C.K.). Victor Chang Cardiac Research Institute, Sydney, Australia (J.C.K.). on December 5, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Coronary artery disease (CAD) is a complex, heterogeneous disease with distinct etiological mechanisms. These different etiologies may give rise to multiple subtypes of CAD that could benefit from alternative preventions and treatments. However, so far, there have been no systematic efforts to predict CAD subtypes using clinical and genetic factors.METHODS:Here, we trained and applied statistical models incorporating clinical and genetic factors to predict CAD subtypes in 26 036 patients with CAD in the UK Biobank. We performed external validation of the UK Biobank models in the US-based All of Us cohort (8598 patients with CAD). Subtypes were defined as high versus normal LDL (low-density lipoprotein) levels, high versus normal Lpa (lipoprotein A) levels, ST-segment–elevation myocardial infarction versus non–ST-segment–elevation myocardial infarction, occlusive versus nonocclusive CAD, and stable versus unstable CAD. Clinical predictors included levels of ApoA, ApoB, HDL (high-density lipoprotein), triglycerides, and CRP (C-reactive protein). Genetic predictors were genome-wide and pathway-based polygenic risk scores (PRSs).RESULTS:Results showed that both clinical-only and genetic-only models can predict CAD subtypes, while combining clinical and genetic factors leads to greater predictive accuracy. Pathway-based PRSs had higher discriminatory power than genome-wide PRSs for the Lpa and LDL subtypes and provided insights into their etiologies. The 10-pathway PRS most predictive of the LDL subtype involved cholesterol metabolism. Pathway PRS models had poor generalizability to the All of Us cohort.CONCLUSIONS:In summary, we present the first systematic demonstration that CAD subtypes can be distinguished by clinical and genomic risk factors, which could have important implications for stratified cardiovascular medicine.
- Long Noncoding RNA Function in Smooth Muscle Cell Plasticity and Atherosclerosisby Lars Maegdefessel Francesca Fasolo Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany (L.M., F.F.). German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Berlin, Germany (L.M., F.F.). Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden (L.M.). on December 5, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>In the healthy mature artery, vascular cells, including endothelial cells, smooth muscle cells (SMCs), and fibroblasts are organized in different layers, performing specific functions. SMCs located in the media are in a differentiated state and exhibit a contractile phenotype. However, in response to vascular injury within the intima, stimuli from activated endothelial cells and recruited inflammatory cells reach SMCs and induce a series of remodeling events in them, known as phenotypic switching. Indeed, SMCs retain a certain degree of plasticity and are able to transdifferentiate into other cell types that are crucial for both the formation and development of atherosclerotic lesions. Because of their highly cell-specific expression profiles and their widely recognized contribution to physiological and disease-related biological processes, long noncoding RNAs have received increasing attention in atherosclerosis research. Dynamic fluctuations in their expression have been implicated in the regulation of SMC identity. Sophisticated technologies are now available to allow researchers to access single-cell transcriptomes and study long noncoding RNA function with unprecedented precision. Here, we discuss the state of the art of long noncoding RNAs regulation of SMC phenotypic switching, describing the methodologies used to approach this issue and evaluating the therapeutic perspectives of exploiting long noncoding RNAs as targets in atherosclerosis.
- Circadian Dysfunction in the Skeletal Muscle Impairs Limb Perfusion and Muscle Regeneration in Peripheral Artery Diseaseby Pei Zhu Calvin L. Chao Adam W.T. Steffeck Caitlyn Dang Noah X. Hamlish Eric M. Pfrender Bin Jiang Clara B. Peek Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL. (P.Z., A.W.T.S., N.X.H., E.M.P., C.B.P.) Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL. (P.Z., A.W.T.S., N.X.H., E.M.P., C.B.P.) Department of Surgery, Division of Vascular Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL. (C.L.C., C.D., B.J.) Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Chicago, IL (B.J.). on December 5, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Peripheral artery disease (PAD), caused by atherosclerosis, leads to limb ischemia, muscle damage, and impaired mobility in the lower extremities. Recent studies suggest that circadian rhythm disruptions can hinder vascular repair during ischemia, but the specific tissues involved and the impact on muscle health remain unclear. This study investigates the role of the skeletal muscle circadian clock in muscle adaptation to ischemic stress using a surgical mouse model of hindlimb ischemia.METHODS:We performed secondary analysis of publicly available RNA-sequencing data sets derived from patients with PAD to identify the differential expression of circadian-related genes in endothelial cells and ischemic limb skeletal muscles. We used mice with specific genetic loss of the circadian clock activator, BMAL1 (brain and muscle ARNT-like 1), in adult skeletal muscle tissues (Bmal1muscle).Bmal1musclemice and controls underwent femoral artery ligation surgery to induce hindlimb ischemia. Laser Doppler imaging was used to assess limb perfusion at various time points after the surgery. Muscle tissues were analyzed with RNA sequencing and histological examination to investigate PAD-related muscle pathologies. Additionally, we studied the role of BMAL1 in muscle fiber adaptation to hypoxia using RNA and assay for transposase-accessible chromatin with sequencing analyses in primary myotube culture model.RESULTS:Disrupted expression of circadian rhythm–related genes was observed in existing RNA-sequencing data sets from endothelial cells and ischemic limb skeletal muscles derived from patients with PAD. Genetic loss ofBmal1specifically in adult mouse skeletal muscle tissues delayed reperfusion recovery following induction of hindlimb ischemia. Histological examination of muscle tissues showed reduced regenerated myofiber number and a decreased proportion of type IIB fast-twitch myofibers inBmal1musclemouse muscles in the ischemic limbs but not in their contralateral nonischemic limbs. Transcriptomic analysis revealed abrogated metabolic, angiogenic, and myogenic pathways relevant to hypoxia adaptation inBmal1musclemouse muscles. These changes were corroborated inBmal1-deficient cultured primary myotubes cultured under hypoxic conditions.CONCLUSIONS:Circadian clock in skeletal muscle is crucial for the muscle’s response to hypoxia during hindlimb ischemia. Targeting the muscle circadian clock may have therapeutic potential for enhancing muscle response to reduced blood flow and promoting recovery in conditions such as PAD.
- SGK1-Mediated Vascular Smooth Muscle Cell Phenotypic Transformation Promotes Thoracic Aortic Dissection Progressionby Shuai Leng Haijie Li Pengfei Zhang Zhiqiao Dang Baowei Shao Shishan Xue Yansong Ning Xilong Teng Leilei Zhang Honglu Wang Na Li Fengquan Zhang Wenqian Yu Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.) Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China. (S.L., W.Y.) on December 5, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:The occurrence of thoracic aortic dissection (TAD) is closely related to the transformation of vascular smooth muscle cells (VSMCs) from a contractile to a synthetic phenotype. The role of SGK1 (serum- and glucocorticoid-regulated kinase 1) in VSMC phenotypic transformation and TAD occurrence is unclear.METHODS:Four-week-old male Sgk1F/F(Sgk1floxed) and Sgk1F/F;TaglnCre(smooth muscle cell–specificSgk1knockout) mice were administered β-aminopropionitrile monofumarate for 4 weeks to model TAD. The SGK1 inhibitor GSK650394 was administered daily via intraperitoneal injection to treat the mouse model of TAD. Immunopurification and mass spectrometry were used to identify proteins that interact with SGK1. Immunoprecipitation, immunofluorescence colocalization, and GST (glutathione S-transferase) pull-down were used to detect molecular interactions between SGK1 and SIRT6 (sirtuin 6). RNA-sequencing analysis was performed to evaluate changes in the SIRT6 transcriptome. Quantitative chromatin immunoprecipitation was used to determine the target genes regulated by SIRT6. Functional experiments were also conducted to investigate the role of SGK1-SIRT6-MMP9 (matrix metalloproteinase 9) in VSMC phenotypic transformation. The effect of SGK1 regulation on target genes was evaluated in human and mouse TAD samples.RESULTS:Sgk1F/F;TaglnCreor pharmacological blockade of Sgk1 inhibited the formation and rupture of β-aminopropionitrile monofumarate–induced TADs in mice and reduced the degradation of the ECM (extracellular matrix) in vessels. Mechanistically, SGK1 promoted the ubiquitination and degradation of SIRT6 by phosphorylating SIRT6 at Ser338, thereby reducing the expression of the SIRT6 protein. Furthermore, SIRT6 transcriptionally inhibits the expression of MMP9 through epigenetic modification, forming the SGK1-SIRT6-MMP9 regulatory axis, which participates in the ECM signaling pathway. Additionally, our data showed that the lack of SGK1-mediated inhibition of ECM degradation and VSMC phenotypic transformation is partially dependent on the regulatory effect of SIRT6-MMP9.CONCLUSIONS:These findings highlight the key role of SGK1 in the pathogenesis of TAD. A lack of SGK1 inhibits VSMC phenotypic transformation by regulating the SIRT6-MMP9 axis, providing insights into potential epigenetic strategies for TAD treatment.
- On-Treatment Change in d-Dimer Is Associated With Differential Outcomes Among Therapeutic Dose Heparin-Treated Noncritically Ill Patients Hospitalized for COVID-19
- Human Genetic Evidence to Inform Clinical Development of IL-6 Signaling Inhibition for Abdominal Aortic Aneurysmby Stephen Burgess Héléne T. Cronjé Emil deGoma Yung Chyung Dipender Gill MRC Biostatistics Unit, University of Cambridge, United Kingdom. (S.B., H.T.C.) Cardiovascular Epidemiology Unit, University of Cambridge, United Kingdom. (S.B.) Sequoia Genetics, London, United Kingdom (S.B., H.T.C., D.G.). Tourmaline Bio, New York, NY (E.d., Y.C.). on December 5, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Abdominal aortic aneurysm (AAA) represents a significant cause of mortality, yet no medical therapies have proven efficacious. The aim of the current study was to leverage human genetic evidence to inform clinical development of IL-6 (interleukin-6) signaling inhibition for the treatment of AAA.METHODS:Associations of rs2228145, a missense variant in theIL6Rgene region, are expressed per additional copy of the C allele, corresponding to the genetically predicted effect of IL-6 signaling inhibition. We consider genetic associations with AAA risk in the AAAgen consortium (39 221 cases and 1 086 107 controls) and UK Biobank (1963 cases and 365 680 controls). To validate against known effects of IL-6 signaling inhibition, we present associations with rheumatoid arthritis, polymyalgia rheumatica, and severe COVID-19. To explore mechanism specificity, we present associations with thoracic aortic aneurysm, intracranial aneurysm, and coronary artery disease. We further explored genetic associations in clinically relevant subgroups of the population.RESULTS:We observed strong genetic associations with AAA risk in the AAAgen consortium, UK Biobank, and FinnGen (odds ratios: 0.91 [95% CI, 0.90–0.92],P=4×10−30; 0.90 [95% CI, 0.84–0.96],P=0.001; and 0.86 [95% CI, 0.82–0.91],P=7×10−9, respectively). The association was similar for fatal AAA but with greater uncertainty due to the lower number of events. The association with AAA was of greater magnitude than associations with coronary artery disease and even rheumatological disorders for which IL-6 inhibitors have been approved. No strong associations were observed with thoracic aortic aneurysm or intracranial aneurysm. Associations attenuated toward the null in populations with concomitant rheumatological or connective tissue disease.CONCLUSIONS:Inhibition of IL-6 signaling is a promising strategy for treating AAA but not other types of aneurysmal disease. These findings serve to help inform clinical development of IL-6 signaling inhibition for AAA treatment.
- Left Ventricular Hypertrophy in Aortic Stenosis: Early Cell and Matrix Regression 2 Months Post-Aortic Valve Replacementby Jonathan Bennett George D. Thornton Christian Nitsche Francisco F. Gama Nikoo Aziminia Uzma Gul Abhishek Shetye Peter Kellman Rhodri H. Davies James C. Moon Thomas A. Treibel Institute of Cardiovascular Science, University College London, United Kingdom (J.B., G.D.T., C.N., N.A., R.H.D., J.C.M., T.A.T.). Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom (J.B., G.D.T., C.N., F.F.G., N.A., U.G., A.S., R.H.D., J.C.M., T.A.T.). Division of Cardiology, Medical University of Vienna, Austria (C.N.). National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (P.K.). on December 4, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017425, December 1, 2024. BACKGROUND:In aortic stenosis, the myocardium responds with left ventricular hypertrophy, which is characterized by increased left ventricular mass due to cellular hypertrophy and extracellular matrix expansion. Following aortic valve replacement (AVR), left ventricular hypertrophy regression occurs, but early cellular and extracellular dynamics are unknown.METHODS:Patients with severe symptomatic aortic stenosis undergoing surgical or transcatheter AVR were prospectively recruited. Pre- and early post-AVR cardiac magnetic resonance imaging assessed left ventricular remodeling, global longitudinal strain, and T1 mapping to determine extracellular volume fraction and volume of cellular and extracellular compartments.RESULTS:In all, 39 patients (aged 71.4±9.8 years, male 79%, aortic valve peak velocity 4.4±0.5 m/s) underwent cardiac magnetic resonance before and at median 7.7 weeks post-AVR. Left ventricular mass index reduced significantly by 15.4% (P<0.001*), primarily driven by cellular compartment regression (18.7%,P<0.001*), with a smaller reduction in the extracellular compartment (7.2%,P<0.001*). This unbalanced regression led to an apparent increase in extracellular volume fraction (27.4±3.1% to 30.2±2.8%;P<0.001*). Although there was no significant change in global longitudinal strain post-AVR, an increase in extracellular volume fraction was associated with worsening of global longitudinal strain (Pearson r=0.41,P=0.01). Mode of intervention (transcatheter versus surgical) did not influence the above myocardial parameters post-AVR (allP>0.05). The asterisk inPvalues indicates a statistical significance of <0.05.CONCLUSIONS:Within 8 weeks of AVR for aortic stenosis, substantial left ventricular hypertrophy regression occurs involving both cellular and extracellular compartments, demonstrating the early myocardial adaptability to afterload relief. Cellular compartment regression is greater than extracellular regression, leading to an apparent increase in extracellular volume fraction. Mode of intervention did not affect degree of reverse remodeling, indicating that both are effective at resulting beneficial changes post-AVR.REGISTRATION:URL:https://www.isrctn.com; Unique identifier: NCT04627987.
- Left Ventricular Hypertrophy Following Aortic Valve Replacement: Bruce Banner or the Hulk?by Timothy C. Wong Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, PA. University of Pittsburgh Medical Center (UPMC) Heart and Vascular Institute – Cardiovascular Magnetic Resonance and Hypertrophic Cardiomyopathy Centers, Pittsburgh, PA. on December 4, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017716, December 1, 2024. <br/>
- Boys With Duchenne Muscular Dystrophy Have Diastolic Dysfunction Based on CMRby Joseph R. Starnes Jeffrey G. Weiner Kristen George-Durrett Kimberly Crum Christopher C. Henderson M. Jay Campbell Katheryn Gambetta Kan N. Hor Nazia Husain Jennifer S. Li Frank J. Raucci Brian D. Soriano Christopher F. Spurney Larry W. Markham Jonathan H. Soslow Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN (J.R.S., J.G.W., K.G.-D., K.C., C.C.H., J.H.S.). Division of Pediatric Cardiology, Department of Pediatrics, Duke University Medical Center, Durham, NC (M.J.C., J.S.L.). Division of Cardiology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL (K.G., N.H.). Division of Cardiology, Department of Pediatrics, Nationwide Children’s Hospital, Ohio State University, Columbus (K.N.H.). Division of Cardiology, Department of Pediatrics, Children’s Hospital of Richmond at Virginia Commonwealth University (F.J.R.). Division of Cardiology, Department of Pediatrics, Seattle Children’s Hospital, WA (B.D.S.). Children’s National Heart Institute, Children’s National Hospital, Washington, DC (C.F.S.). Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health and Indiana University School of Medicine, Indianapolis (L.W.M.). on December 4, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017287, December 1, 2024. BACKGROUND:Cardiomyopathy is the leading cause of death in boys with Duchenne muscular dystrophy (DMD). While cardiac magnetic resonance (CMR) is routinely used to assess fibrosis and left ventricular (LV) ejection fraction, CMR measures of LV filling and ejection in DMD have not been reported.METHODS:Patients with DMD (n=179) and healthy controls (n=96) were prospectively enrolled and underwent CMR. The DMD cohort was followed clinically at multiple institutions, and clinical data were recorded. Standard volumes and functions were calculated, and LV filling and ejection curves were measured from baseline CMR. Multivariable linear regressions were used to compare ventricular filling and ejection measures between groups, adjusting for baseline differences. Cox regressions were used to evaluate the relationship between diastolic function measures and mortality in the DMD cohort.RESULTS:Patients with DMD had significantly smaller stature and ventricular volumes than healthy control patients (P<0.001). They had lower baseline LV ejection fraction (P<0.001), though most had normal systolic function. When adjusted for age, sex, heart rate, body surface area, and LV end-diastolic volume, patients with DMD had slower peak filling rates (P<0.001) and peak ejection rates (P<0.001), as well as slower time to peak ventricular ejection rate (P=0.011). When adjusted for heart rate, a lower peak ventricular ejection rate (P=0.007) and peak filling rate (P=0.033), normalized to LV end-diastolic volume, were associated with mortality in patients with DMD.CONCLUSIONS:Patients with DMD have significantly different baseline CMR filling and ejection indices compared with controls. Some filling indices are associated with mortality and may be useful prognostic measures. Further research is needed in larger cohorts to determine the prognostic value of these differences.
- No Time to Relax: Expanding CMR Utility in Duchene Muscular Dystrophyby Adarsh Katamreddy Ahmad Masri Division of Cardiology, Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland. on December 4, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017612, December 1, 2024. <br/>
- Identification of Circulating Proteins Associated With Blood Pressureby Siqi Xu Simin Wen Xizeng Zong Shifeng Wen Jianwei Zhu Weipeng Zheng Zhiqiang Wang Peihua Cao Zhijiang Liang Changhai Ding Yan Zhang Guangfeng Ruan Department of Rheumatology, Guangzhou First People’s Hospital, Guangzhou Medical University, China. (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.) Department of Orthopedics, Guangzhou First People’s Hospital, Guangzhou Medical University, China. (J.Z., W.Z.) Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.). Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China (Z.W., P.C., C.D., Y.Z.). Department of Public Health, Guangdong Women and Children Hospital, China (Z.L.). Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia (C.D.). on December 3, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Circulating proteins in blood are involved in various physiological processes, but their contributions to blood pressure regulation remain partially understood. In traditional observational studies, identifying circulating proteins causally associated with blood pressure is challenging because of potentially unmeasured confounding and possible reverse causality.METHODS:Two-sample Mendelian randomization analyses were conducted to estimate the causal effects of 2270 circulating proteins (data sourced from 8 genome-wide association studies) on diastolic blood pressure, systolic blood pressure, and pulse pressure. Colocalization analyses were then used to investigate whether the circulating proteins and blood pressure traits shared causal genetic variants. To further verify the findings, we subsequently performed Steiger filtering analyses, annotation of protein-altering variants, assessment of overlap between protein quantitative trait loci and expression quantitative trait loci, protein-protein interaction and functional enrichment analyses, and drug target evaluation. To provide more potential biomarkers, we further evaluated the epidemiological associations of 2923 circulating proteins with blood pressure and hypertension by cross-sectional and longitudinal analyses using individual data in the UK Biobank.RESULTS:Mendelian randomization and colocalization analyses identified 121 circulating proteins with putative causal effects on at least 1 blood pressure trait. Many of the identified proteins are enriched in the pathways relevant to blood pressure regulation, and a majority of these proteins are either known drug targets or druggable candidates.CONCLUSIONS:This study has uncovered numerous circulating proteins potentially causally associated with blood pressure, providing insights into the regulatory mechanisms of blood pressure and potential therapeutic targets to facilitate blood pressure management.
- ACE2, From the Kidney to SARS-CoV-2: Donald Seldin Award Lecture 2023by Daniel Batlle Luise Hassler Jan Wysocki Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL. on December 3, 2024
Hypertension, Ahead of Print. <br/>ACE2 (angiotensin-converting enzyme 2) is a monocarboxypeptidase that cleaves Ang II (angiotensin II) among other substrates. ACE2 is present in the cell membrane of many organs, most abundantly in epithelial cells of kidney proximal tubules and the small intestine, and also exists in soluble forms in plasma and body fluids. Membrane-bound ACE2 exerts a renoprotective action by metabolizing Ang II and therefore attenuating the undesirable actions of excess Ang II. Therefore, soluble ACE2, by downregulating this peptide, may exert a therapeutic action. Our laboratory has designed ACE2 truncates that pass the glomerular filtration barrier to target the kidney renin-angiotensin system directly and, therefore, compensate for loss of kidney membrane-bound ACE2. Membrane-bound ACE2 is also the essential receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Soluble ACE2 proteins have been studied as a way to intercept SARS-CoV-2 from binding to membrane-bound ACE2 and prevent cell entry of SARS-CoV-2 altogether. We bioengineered a soluble ACE2 protein, termed ACE2 618-DDC-ABD, with increased binding affinity for SARS-CoV-2 and prolonged duration of action, which, when administered intranasally, provides near-complete protection from lethality in k18hACE2 mice infected with different SARS-CoV-2 variants. The main advantage of soluble ACE2 proteins for the neutralization of SARS-CoV-2 is their immediate onset of action and universality for current and future emerging SARS-CoV-2 variants. It is notable that ACE2 is critically involved in 2 dissimilar functions: as a receptor for cell entry of many coronaviruses and as an enzyme in the metabolism of Ang II, and yet in both cases, it is a therapeutic target.
- Peli1 Deficiency in Macrophages Attenuates Pulmonary Hypertension by Enhancing Foxp1-Mediated Transcriptional Inhibition of IL-6by Donghai Lin Li Hu Dong Wei Yan Li Yanfang Yu Qiang Wang Xiaoxuan Sun Yueyao Shen Youjia Yu Kai Li Zhiwei Zhang Yue Cao Jiantao Li Yuehua Li David Fulton Jingyu Chen Jie Wang Huijie Huang Feng Chen Department of Forensic Medicine (D.L., L.H., Yan Li, Yanfang Yu, Y.S., Youjia Yu, K.L., Z.Z., Y.C., J.W., H.H., F.C.), Nanjing Medical University, China. Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine (L.H., J.L., Yuehua Li, J.W., F.C.), Nanjing Medical University, China. The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center (D.W., J.C., F.C.), Nanjing Medical University, China. Department of Rheumatology, First Affiliated Hospital of Nanjing Medical University, China (Q.W., X.S.). Vascular Biology Center, Medical College of Georgia at Augusta University, GA (D.F., F.C.). on December 2, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:The infiltration of macrophages into the lungs is a common characteristic of perivascular inflammation, contributing to vascular remodeling in pulmonary hypertension (PH). Peli1 (pellino E3 ubiquitin-protein ligase 1) plays a critical role in regulating the production of proinflammatory cytokines and the polarization of macrophages in various diseases. However, the role of Peli1 in PH remains to be investigated.METHODS:The expression and biological function of Peli1 were investigated in both human and experimental models of PH. Peli1-deficient mice and bone marrow transplant mice were utilized to explore the roles of Peli1 in macrophages in vivo. Proteomic analysis and molecular biology techniques were used to uncover the underlying mechanisms.RESULTS:The upregulation of Peli1 in the lungs and alveolar macrophages was observed in hypoxia-treated mice. Peli1 knockout mice and myeloid Peli1-deficient mice significantly ameliorated hypoxia-induced right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary vascular remodeling. Mechanistically, Peli1 facilitated the ubiquitination and subsequent proteasomal degradation of Foxp1 (forkhead box p1), thereby alleviating its suppression of IL (interleukin)-6 transcription and contributing to macrophage activation. Furthermore, myeloid Foxp1 deficiency partially eliminates the protective effect of myeloid Peli1 deficiency in hypoxia-induced PH mice.CONCLUSIONS:Our findings demonstrate that the Peli1-Foxp1-IL-6 pathway plays a crucial role in macrophage activation and recruitment during the development of PH, underscoring the potential of Peli1 as a therapeutic target for PH.
- Definitive Evidence for the Identification and Function of Renin-Expressing Cholinergic Neurons in the Nucleus Ambiguusby Éva M. Fekete Javier Gomez Mina Ghobrial Kathren Kaminski Patricia C. Muskus Carie R. Boychuk Ana Hantke Guixa Ibrahim Vazirabad Michelle Xie Azeez Ganiyu Daria Golosova Natalia M. Mathieu Yoko B. Wang Ko-Ting Lu Kelsey K. Wackman Daniel T. Brozoski Gary C. Mouradian Matthew R. Hodges Jeffrey L. Segar Justin L. Grobe Curt D. Sigmund Pablo Nakagawa Department of Physiology (E.M.F., J.G., M.G., K.K., P.C.M., A.H.G., I.V., M.X., A.G., D.G., N.M.M., K.-T.L., K.K.W., D.T.B., G.C.M., M.R.H., J.L.S., J.L.G., C.D.S., P.N.), Medical College of Wisconsin, Milwaukee. Cardiovascular Center (J.L.S., J.L.G., C.D.S., P.N.), Medical College of Wisconsin, Milwaukee. Neuroscience Research Center (J.L.G., C.D.S., P.N.), Medical College of Wisconsin, Milwaukee. Comprehensive Rodent Metabolic Phenotyping Core (J.L.G.), Medical College of Wisconsin, Milwaukee. Department of Pediatrics (J.L.S.), Medical College of Wisconsin, Milwaukee. Department of Biomedical Engineering (J.L.G.), Medical College of Wisconsin, Milwaukee. Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia (C.R.B., Y.B.W.). on December 2, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:The importance of the brain renin-angiotensin system in cardiovascular function is well accepted. However, not knowing the precise source of renin in the brain has been a limitation toward a complete understanding of how the brain renin-angiotensin system operates.METHODS:Highly sensitive in situ hybridization techniques and conditional knockout mice were used to address the location and function of renin in the brainstem.RESULTS:We identified novel renin-expressing cholinergic neurons in the nucleus ambiguus (NuAm), a major vagal cardioinhibitory center in the brainstem. The expression of renin-angiotensin system genes was relatively abundant in the NuAm, implying that angiotensin II might mediate an important regulatory role in this nucleus and other regions with neural connectivity to the NuAm. Then, we generated conditional knockout mice lacking the classical renin isoform (Ren-aChAT-KO), specifically in cholinergic neurons. Ablation of Ren-a in cholinergic neurons abrogated renin expression in the NuAm. Moreover, studies using radiotelemetry, heart rate variability analyses, and pharmacological approaches revealed that the parasympathetic nervous system is depressed in Ren-aChAT-KOmales while augmented in the Ren-aChAT-KOfemales. Subsequently, transcriptomic approaches were used to infer putative genes and signaling pathways regulated by renin within the NuAm.CONCLUSIONS:This study revealed that renin in cholinergic neurons plays a fundamental role in preserving autonomic balance and cardiovascular homeostasis in a sex-dependent manner. These findings define the NuAm as an endogenous, local source of renin with biological function and serve as conclusive evidence for the presence and functionality of the brain renin-angiotensin system.
- ATP2A3 in Primary Aldosteronism: Machine Learning-Based Discovery and Functional Validationby Zhuolun Sun Elisabeth Kemter Yingxian Pang Martin Bidlingmaier Eckhard Wolf Martin Reincke Tracy Ann Williams Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (Z.S., Y.P., M.B., M.R., T.A.W.). Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Germany (E.K., E.W.). on December 2, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Aldosterone-producing adenomas (APAs) are a common cause of primary aldosteronism that can lead to cardiovascular complications if left untreated. Machine learning-based bioinformatics approaches have emerged as powerful tools for identifying potential disease markers, gaining widespread recognition in biomedical research. We aimed to use machine learning to discover novel biomarkers of APAs to identify new pathophysiological mechanisms.METHODS:We applied 2 machine learning algorithms to published RNA sequencing data to identify APA feature genes. Validation was performed using APA tissue samples, spatial transcriptomics, pig adrenal glands, and in vitro assays in a human adrenocortical cell line.RESULTS:Machine learning identifiedATP2A3as a key feature gene in APA, and its upregulation in APAs compared with the adjacent cortex was confirmed by spatial transcriptomics. In human adrenocortical cells, angiotensin II treatment increasedATP2A3gene expression 9.15-fold. SilencingATP2A3decreased basalCYP11B2expression and aldosterone secretion by 3.51-fold and 1.46-fold, respectively, and by 1.77-fold and 1.94-fold under angiotensin II stimulation. Dietary sodium restriction in pigs significantly increasedATP2A3mRNA and protein levels. Spatial transcriptomics showed that APA cells exhibited higherATP2A3gene expression compared with all other adrenal cell types. The suppressive effect ofATP2A3silencing onCYP11B2expression was further enhanced by Ca2+inhibitors.CONCLUSIONS:TheATP2A3gene is highly expressed in APA and is a key regulator ofCYP11B2expression and aldosterone production.
- Unraveling Comorbidities Contribution to Cardiac Diastolic Dysfunction and Heart Failureby María Villalba-Orero Marina López-Olañeta Belén Campos-Olmo Daniel Jimenez-Carretero Lucía Sánchez Fátima Sánchez-Cabo Antonella Ausiello Rodrigo Cañas-Álvaro Emilio Camafeita Jesús Vázquez Pablo García-Pavía Domingo Pascual-Figal Enrique Lara-Pezzi Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (M.V.-O., M.L.-O., B.C.-O., D.J.-C., L.S., F.S.-C., A.A., R.C.-Á., E.C., J.V., P.G.-P., D.P.-F., E.L.-P.). Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Spain (M.V.-O.). Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain (M.V.-O., J.V., P.G.-P., D.P.-F., E.L.-P.). Biomedical Research Institute Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain (D.P.-F.). Medicine Department, University of Murcia, Spain (D.P.-F.). Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (P.G.-P.). Universidad Francisco de Vitoria, Madrid, Spain (P.G.-P.). on November 29, 2024
Circulation: Heart Failure, Ahead of Print. <br/>BACKGROUND:Heart failure with preserved ejection fraction (HFpEF) is a major public health problem characterized by multiple simultaneous comorbidities whose specific contribution is challenging to disentangle in humans, leading to a generalized therapeutic approach that may not account for the underlying pathology.METHODS:We followed distinct mouse models of major HFpEF comorbidities for 2.5 years to unveil their specific contribution to the syndrome.RESULTS:All comorbidities contributed to HFpEF through partially distinct routes. Aging alone resulted in HFpEF in old age, with delayed left ventricular relaxation and kidney fibrosis. Obesity induced a faster deterioration of relaxation associated with enlarged left ventricle and liver fibrosis. Hypertension caused delayed ventricular relaxation independent from structural changes that preceded left atrial dilatation linked to aortic stiffness and increased fibrosis in myocardium and kidney. Chronic intermittent hypoxia led to HFpEF and relaxation impairment associated with pulmonary hypertension. Hyperglycemia accelerated diastolic dysfunction and HFpEF onset associated with reduced arterial flow and left ventricular remodeling. Therefore, the pathological substrates contributing to HFpEF included cardiac and noncardiac alterations with differential features for each comorbidity. Critically, the characteristics linked to diastolic dysfunction and HFpEF across the various comorbidities agreed with phenogroups observed in human patients.CONCLUSIONS:The identification of time-dependent pathological features provides a comprehensive picture of HFpEF progression associated with each comorbidity.
- Hepatic Tissue Alterations in ST-Elevation Myocardial Infarction: Determinants and Prognostic Implicationsby Ivan Lechner Martin Reindl Sebastian von der Emde Alina Desheva Fritz Oberhollenzer Christina Tiller Magdalena Holzknecht Thomas Kremser Julian Faccini Can Gollmann-Tepeköylü Christian Kremser Agnes Mayr Axel Bauer Bernhard Metzler Sebastian J. Reinstadler University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria. University Clinic of Radiology (C.K., A.M.), Medical University of Innsbruck Innsbruck, Austria. University Clinic of Cardiac Surgery (C.G.-T.), Medical University of Innsbruck Innsbruck, Austria. on November 29, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017041, December 1, 2024. BACKGROUND:The presence and clinical significance of hepatic tissue alterations as assessed by cardiac magnetic resonance imaging in patients with ST-segment–elevation myocardial infarction (STEMI), are unclear. This study aimed to investigate associations of hepatic T1 patterns with myocardial tissue damage and clinical outcomes in patients suffering from STEMI.METHODS:We analyzed 485 patients with STEMI treated with percutaneous coronary intervention who were enrolled in the prospective MARINA STEMI study (Magnetic Resonance Imaging In Acute ST-Elevation Myocardial Infarction). Myocardial function and left and right ventricular (RV) infarct characteristics were assessed by cardiac magnetic resonance within the first week after STEMI. Native hepatic T1 times and extracellular volume were evaluated from standard cardiac T1 maps at baseline and 4 months thereafter.RESULTS:Median hepatic T1 times were 559 ms (interquartile range, 514–605) at baseline and decreased to 542 ms (interquartile range, 507–577) at 4 months (P<0.001). Hepatic T1 times at baseline were independently associated with female sex (β 0.116;P=0.008), hyperlipidemia (β −0.116;P=0.008), and myocardial tissue damage (infarct size: β 0.178;P<0.001; microvascular obstruction: β 0.193;P<0.001; RV infarction: β 0.161;P<0.001). Determinants of hepatic T1 times at 4 months were female sex (β 0.123;P=0.002), multivessel disease (β 0.121;P=0.002), N-terminal pro–B-type natriuretic peptide (β 0.101;P=0.010), RV infarction (β 0.501;P<0.001), and RV end-systolic volume index (β 0.087;P=0.031). Patients without a decrease exhibited a higher frequency of major adverse cardiovascular events (13% versus 5%;P=0.003). Hepatic T1 times at baseline (hazard ratio, 1.87 [95% CI, 1.40–2.50];P<0.001), 4 months (hazard ratio, 2.69 [95% CI, 2.15–3.36];P<0.001), and hepatic extracellular volume at 4 months (hazard ratio, 1.59 [95% CI, 1.33–1.90];P<0.001) were associated with major adverse cardiovascular events. After adjustment for univariable associates, only hepatic T1 times at 4 months were independently associated with adverse outcomes (hazard ratio, 2.86 [95% CI, 1.99–4.12];P<0.001).CONCLUSIONS:Hepatic tissue alterations determined by T1 mapping were associated with female sex, hyperlipidemia, multivessel disease, N-terminal pro–B-type natriuretic peptide, and left and RV myocardial tissue damage. These alterations can persist into the chronic phase after STEMI and indicate a worse clinical outcome.REGISTRATION:URL:https://www.clinicaltrials.gov; Unique identifier: NCT04113356.
- Is It Primetime for Finerenone in Heart Failure?by Neal M. Dixit Saul Schaefer Division of Cardiovascular Medicine, Department of Internal Medicine, UC Davis Health, Sacramento, CA. on November 29, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Hepatic Tissue Alterations in STEMI: New Insights Into the Prognostic Significance of Cardio-Hepatic Interplayby Théo Pezel Solenn Toupin MIRACL.ai Laboratory, Multimodality Imaging for Research and Analysis Core Laboratory and Artificial Intelligence, University Hospital of Lariboisiere (AP-HP), Paris, France (T.P., S.T.). Université Paris Cité, Department of Cardiology and Radiology, University Hospital of Lariboisiere, (Assistance Publique des Hôpitaux de Paris, AP-HP), France (T.P., S.T.). Inserm MASCOT – UMRS 942, University Hospital of Lariboisiere, Paris, France (T.P., S.T.). on November 29, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e017611, December 1, 2024. <br/>
- Abdominal Compared With Coronary Artery Calcification and Incident Cardiovascular Events and Mortality in Black Adultsby Temidayo A. Abe Fengxia Yan Titilope Olanipekun Michael Blaha Valery Effoe Ndausung Udongwo Robert J. Mentz Adebamike Oshunbade James G. Terry Jalal K. Ghali Wondwosen K. Yimer Chukwuemezie Kamanu Ifeoma Onuorah Michael Hall Anekwe Onwuanyi Adolfo Correa Melvin Echols Division of Cardiology, Department of Medicine (T.A.A.), Vanderbilt University Medical Center, Nashville, TN. Department of Radiology (J.G.T.), Vanderbilt University Medical Center, Nashville, TN. Division of Cardiology, Department of Medicine (F.Y., V.E., N.U., J.K.G., A. Onwuanyi), Morehouse School of Medicine, Atlanta, GA. Department of Medicine (F.Y., V.E., J.K.G., A. Onwuanyi), Morehouse School of Medicine, Atlanta, GA. Cardiovascular Research Institute (F.Y., V.E., J.K.G., A. Onwuanyi), Morehouse School of Medicine, Atlanta, GA. Department of Medicine, Brigham’s and Women, Harvard Medical School, Boston, MA (T.O.). Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD (M.B.). Division of Cardiology, Department of Medicine, Duke University School of Medicine, Duke Clinical Research Institute, Durham, NC (R.J.M.). Division of Internal Medicine, Department of Medicine, Tulane University, New Orleans, LA (A. Oshunbade). Department of Medicine, University of Mississippi Medical Center, Jackson (W.K.Y., M.H., A.C.). Division of Internal Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (C.K.). Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (I.O.). Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (M.E.). on November 27, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e016775, December 1, 2024. <br/>BACKGROUND:Black adults show heightened cardiovascular risk compared with other groups despite comparable or lower coronary artery calcium (CAC) scores, indicating potential cardiovascular risk underestimation by CAC. Abdominal aortic calcification (AAC), preceding CAC, may predict cardiovascular events better in Black adults who are prone to early atherosclerotic cardiovascular disease and excess events at low CAC scores.METHODS:We included 2551 participants from the JHS (Jackson Heart Study) visit 2 examination (2005–2008) without cardiovascular disease, followed through 2016. Cox regression estimated hazard ratios for incident cardiovascular events defined as a composite of myocardial infarction, stroke, heart failure, and all-cause mortality. The predictive value of CAC and AAC, when added to the American College of Cardiology/American Heart Association cardiovascular risk algorithm (pool cohort equation), was assessed.RESULTS:Mean age was 57±10 years; 66% were women. Over a follow-up period of 12.6 years, 287 (11.3%) cardiovascular events and 360 (14.1%) mortality cases were observed. Adjusting for demographic and clinical variables, each 2-fold increase in CAC and AAC was associated with cardiovascular events (CAC: hazard ratio, 1.10 [95% CI, 1.06–1.13]; AAC: hazard ratio, 1.10 [95% CI, 1.06–1.13]) and all-cause mortality CAC: hazard ratio, 1.04 [95% CI, 1.01–1.08]; AAC: hazard ratio, 1.05 [95% CI, 1.01–1.08]). The incremental value of CAC (0.17 [95% CI, 0.08–0.23]) and AAC (0.15 [95% CI, 0.07–0.29]) to the pool cohort equation by net reclassification index was comparable. Notably, 45% of participants with 0 CAC showed some degree of AAC. Each 2-fold increase in AAC was associated with an increased risk of cardiovascular events (hazard ratio, 1.07 [95% CI, 1.01–1.19]) but not all-cause mortality among participants with 0 CAC.CONCLUSIONS:AAC is comparable to CAC in predicting cardiovascular events and all-cause mortality among Black adults, potentially valuable when CAC is absent.
- Caregiving and Hypertension in Younger Black Women: The Jackson Heart Studyby Milla Arabadjian Yiwei Li Byron Jaeger Calvin L. Colvin Jolaade Kalinowski Miriam A. Miles Lenette M. Jones Jacquelyn Y. Taylor Kenneth Butler Paul Muntner Tanya M. Spruill Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola (M.A.). Department of Population Health, New York University Grossman School of Medicine (Y.L., T.M.S.). Department of Biostatistics, Wake Forest University, Winston-Salem, NC (B.J.). Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY (C.L.C.). Department of Human Development and Family Sciences, University of Connecticut, Storrs (J.K.). Heersink School of Medicine, University of Alabama at Birmingham. (M.A.M.) School of Public Health, University of Alabama at Birmingham. (P.M.) School of Nursing, University of Michigan, Ann Arbor (L.M.J.). Columbia University School of Nursing Center for Research on People of Color, New York, NY (J.Y.T.). Department of Medicine, University of Mississippi, Jackson (K.B.). Institute for Excellence in Health Equity, NYU Langone Health, New York, NY (T.M.S.). on November 27, 2024
Hypertension, Ahead of Print. BACKGROUND:Caregiving has been associated with high blood pressure in middle-aged and older women, but this relationship is understudied among younger Black women, a population at high risk for hypertension. We examined the associations of caregiving stress and caregiving for high-needs dependents with incident hypertension among reproductive-age women in the JHS (Jackson Heart Study), a cohort of community-dwelling Black adults.METHODS:We included 453 participants, aged 21 to 44 years, with blood pressure <140/90 mm Hg, and not taking antihypertensive medication at baseline (2000–2004). Caregiving stress over the past 12 months was assessed via a single item in the global perceived stress scale. Caregiving for a high-needs dependent status was assessed via a question on hours per week spent caregiving for children (≤5 years or disabled) or older adults. Incident hypertension was defined as systolic blood pressure ≥140 mm Hg, diastolic blood pressure ≥90 mm Hg, or self-report of taking antihypertensive medication at follow-up exams in 2005 to 2008 and 2009 to 2013.RESULTS:Over a median follow-up of 7.4 years, 43.5% of participants developed hypertension. Participants with moderate/high versus no/low caregiving stress had a higher incidence of hypertension (51.7% versus 40.6%). Higher caregiving stress was associated with incident hypertension after adjustment for sociodemographic and clinical factors, health behaviors, and depressive symptoms (hazard ratio, 1.39 [95% CI, 1.01–1.94]). Being a caregiver for a high-needs dependent was not associated with incident hypertension (adjusted hazard ratio, 0.88 [95% CI, 0.64–1.21]).CONCLUSIONS:Higher caregiving stress among reproductive-age Black women was associated with incident hypertension. Hypertension prevention approaches for this high-risk population may include caregiving stress management strategies.
- UBR1 Promotes Sex-Dependent ACE2 Ubiquitination in Hypertensionby Mona Elgazzaz Navya Lakkappa Clara Berdasco Uma Priya Mohan Anna Nuzzo Luke Restivo Alexa Martinez Amy Scarborough Jessie J. Guidry Srinivas Sriramula Jiaxi Xu Hisham Daoud Michelle A. Mendiola Plá Dawn E. Bowles Andreas M. Beyer Franck Mauvais-Jarvis Xinping Yue Catalin M. Filipeanu Eric Lazartigues Cardiovascular Center of Excellence (M.E., N.L., C.B., U.P.M., A.N., L.R., A.M., A.S., J.J.G., S.S., J.X., X.Y., C.M.F., E.L.), New Orleans, LA Departments of Pharmacology & Experimental Therapeutics (M.E., N.L., C.B., U.P.M., J.J.G., S.S., J.X., X.Y., E.L.), New Orleans, LA Medicine (E.L.), New Orleans, LA Neuroscience Center of Excellence (E.L.), New Orleans, LA Louisiana State University Health Sciences Center, New Orleans, LA (E.L.). Southeast Louisiana Veterans Health Care System, New Orleans, LA (M.E., N.L., C.B., U.P.M., F.M.-J., E.L.). Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt (M.E.). Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC (S.S.). Department of Physiology and Pathophysiology, Xi’an Jiaotong University, School of Medicine, China (J.X.). School of Computer and Cyber Sciences, Augusta University, GA (H.D.). Division of Surgical Sciences, Department of Surgery, Duke University, Durham, NC (M.A.M.P., D.E.B.). Department of Medicine, Medical College of Wisconsin, Milwaukee, WI (A.M.B.). Deming Department of Medicine, Tulane University, New Orleans, LA (F.M.-J.). on November 27, 2024
Hypertension, Volume 82, Issue 1, Page 84-95, January 1, 2025. <br/>BACKGROUND:Ang-II (angiotensin II) impairs the function of the antihypertensive enzyme ACE2 (angiotensin-converting enzyme 2) by promoting its internalization, ubiquitination, and degradation, thus contributing to hypertension. However, few ACE2 ubiquitination partners have been identified, and their role in hypertension remains unknown.METHODS:Proteomics and bioinformatic analyses were used to identify ACE2 ubiquitination partners in the brain, heart, and kidney of hypertensive C57BL6/J mice of both sexes. The interaction between UBR1 (ubiquitin protein ligase E3 component N-recognin) and ACE2 was validated in cells. Central and peripheral UBR1 knockdown was then performed in male mice to investigate its role in the maintenance of hypertension.RESULTS:Proteomics analysis of the hypothalamus identified UBR1 as a potential E3 (ubiquitin protein ligase) ligase promoting ACE2 ubiquitination. Enhanced UBR1 expression, associated with ACE2 reduction, was confirmed in various tissues from hypertensive male mice and human samples. Treatment of endothelial and smooth muscle cells with testosterone, but not 17β-estradiol, confirmed a sex-specific regulation of UBR1. In vivo silencing of UBR1 using chronic administration of small interference RNA resulted in the restoration of ACE2 levels in hypertensive male mice. A transient decrease in blood pressure after intracerebroventricular, but not systemic, infusion was also observed. Interestingly, UBR1 knockdown increased brain activation of Nedd4-2 (neural precursor cell expressed developmentally downregulated protein 4), an E3 ligase promoting ACE2 ubiquitination, and reduced expression of serum and glucocorticoid-regulated kinase 1, the kinase that inactivates Nedd4-2.CONCLUSIONS:These data demonstrate that UBR1 is a novel E3 ubiquitin ligase targeting ACE2 in hypertension. UBR1 and Nedd4-2 appear to work synergistically to ubiquitinate ACE2. Targeting these ubiquitin ligases may represent a novel strategy to restore ACE2 compensatory activity in hypertension.
- Eclipsed Mitral Regurgitation as an Early Sign of Myocardial Ischemia Due to Coronary Vasospasmby Cristina Garcia-Sebastian Ana García-Martín Ángel Sánchez-Recalde Paloma Remior-Pérez Luisa Salido-Tahoces Sara Fernandez-Santos Covadonga Fernández-Golfin José Luis Zamorano Cardiology Department, University Hospital Ramón y Cajal, Madrid, Spain (C.G.-S., A.G.-M., P.R.-P., L.S.-T., S.F.-S., C.F.-G.). Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain (C.G.-S., A.G.-M., Á.S.-R., P.R.-P., L.S.-T., S.F.-S., C.F.-G., J.L.Z.). on November 27, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- NETosis in Cardiovascular Disease: An Opportunity for Personalized Antithrombotic Treatments?by Constance C.F.M.J. Baaten Magdolna Nagy Henri M.H. Spronk Hugo Ten Cate Bas L.J.H. Kietselaer Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (C.C.F.M.J.B., M.N., H.M.H.S., H.T.C.). Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Germany (C.C.F.M.J.B.). Department of Internal Medicine, Maastricht University Medical Center+, the Netherlands (H.M.H.S., H.T.C.). Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (B.L.J.H.K.). on November 27, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2366-2370, December 1, 2024. <br/>
- SR-BI Models for Spontaneous Myocardial Infarction: High Unesterified/Total Cholesterol Ratio Not the Sole Piece of the Puzzleby Miranda Van Eck Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands. Pharmacy Leiden, the Netherlands. on November 27, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2489-2492, December 1, 2024. <br/>
- Elucidating VEGF Biology: A Journey of Discovery and Clinical Translationby Tommaso Mori Naresh Kumar R.N. Napoleone Ferrara Moores Cancer Center (T.M., N.K.R.N., N.F.), University of California San Diego, La Jolla. Department of Pathology (T.M., N.K.R.N., N.F.), University of California San Diego, La Jolla. Department of Ophthalmology (N.F.), University of California San Diego, La Jolla. on November 27, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2361-2365, December 1, 2024. <br/>
- Climber in the Cardiovascular System: Intravenous Leiomyomatosisby Zhengyang Han Suyun Hou Yang Chen Zhengguang Chen Zhenxing Sun Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., Z.S.). Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, China (Z.H., S.H., Y.C., Z.C.). on November 26, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Abnormalities in Coronary Microvasculature in Pulmonary Atresia With Intact Ventricular Septumby Annabelle Grace Binti Vincent Kenzo Ichimura Shoko Ichimura Aubrey Olive Alden Elizabeth Price Darren Salmi Ankit Kushwaha Edda Spiekerkoetter Sushma Reddy Department of Epidemiology and Clinical Research, Stanford University, Palo Alto, CA. (A.G.B.V.) Department of Pediatrics (Cardiology) and Cardiovascular Institute, Stanford University, Palo Alto, CA. (A.G.B.V., S.I., A.A., E.P., A.K., S.R.) Department of Medicine (Pulmonary Critical Care), Stanford University, Palo Alto, CA. (K.I., E.S.) Department of Pathology, Stanford University, Palo Alto, CA. (D.S.) on November 26, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Climate Change and Cardiovascular Disease: Who Is Vulnerable?by Haitham Khraishah Robert L. Ostergard Syed R. Nabi Donald De Alwis Barrak Alahmad Harrington Heart and Vascular Institute, University Hospitals at Case Western Reserve University, Cleveland, OH. (H.K.) Department of Medicine, University Hospitals at Case Western Reserve University, Cleveland, OH. (S.R.N.) Department of Political Science, University of Nevada, Reno (R.L.O.). University of Maryland School of Medicine, Baltimore (D.D.A.). Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA (B.A.). on November 26, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>Climate change involves a shift in earth’s climate indicators over extended periods of time due to human activity. Anthropogenic air pollution has resulted in trapping heat, contributing to global warming, which contributes to worsening air pollution through facilitating oxidizing of air constituents. It is becoming more evident that the effects of climate change, such as air pollution and ambient temperatures, are interconnected with each other and other environmental factors. While the relationship between climate change components and cardiovascular disease is well documented in the literature, their interaction with one another along with individuals’ biological and social risk factors is yet to be elucidated. In this review, we summarize that pathophysiological mechanisms by ambient temperatures directly affect cardiovascular health and describe the most vulnerable subgroups, defined by age, sex, race, and socioeconomic factors. Finally, we provide guidance on the importance of integrating climate, environmental, social, and health data into common platforms to inform researchers and policies.
- Tale of 2 Receptorsby Dudley K. Strickland Joanna M. Cooper Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore. (D.K.S., J.M.C.) Department of Physiology, University of Maryland School of Medicine, Baltimore. (D.K.S., J.M.C.) Department of Surgery, University of Maryland School of Medicine, Baltimore. (D.K.S.) on November 26, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>
- Disruption of PCSK9 Suppresses Inflammation and Attenuates Abdominal Aortic Aneurysm Formationby Zekun Peng Shuang-jie Lv Hong Chen Haojie Rao Ziyi Guo Qing Wan Jianfeng Yang Yuze Zhang De-Pei Liu Hou-zao Chen Miao Wang State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. (Z.P., H.C., H.R., Z.G., Q.W., J.Y., Y.Z., M.W.) State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. (S.-j.L., D.-P.L., H.-z.C.) Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. (M.W.) National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University (M.W.). on November 26, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Abdominal aortic aneurysm (AAA) is a chronic vascular inflammatory disease without effective medications. PCSK9 (proprotein convertase subtilisin/kexin 9), a serine protease from the proprotein convertase family, has recently been associated with AAA in human genome-wide association studies. However, its role in AAA is unknown.METHODS:Transcriptional and histological expression of PCSK9 was examined in AAA tissues and healthy controls. The impact of PCSK9 deletion and inhibition on AAA formation was assessed in mice with hyperlipidemia and Ang II (angiotensin II) overproduction. AAA lesion morphology was assessed by tissue staining. MMP (matrix metalloproteinase) activity was evaluated by gelatin zymography, and leukocyte-vessel wall interaction was monitored by intravital microscopy. RNA sequencing was used to characterize the downstream signaling of PCSK9.RESULTS:PCSK9 expression was upregulated and colocalized with macrophages in human and mouse AAAs.Pcsk9deletion attenuated AAA formation, improved survival, and decreased systemic inflammation, without altering circulating cholesterol levels.Pcsk9deficiency reduced aortic infiltration of macrophages and elastin degradation, without affecting vascular smooth muscle cell apoptosis and proliferation. Mechanistically, PCSK9 was essential in leukocyte-endothelium adhesion and expression of proinflammatory cytokines and MMP9 by macrophages. RNA sequencing of stimulated macrophages revealed thatPcsk9deficiency upregulated histone deacetylase SIRT1 (sirtuin-1) and suppressed NF-κB (nuclear factor-κB) inflammatory signaling. SIRT1 inhibition attenuated the proinflammatory actions of PCSK9. Furthermore, administration of PCSK9 small interfering RNA or antibody constrained AAA formation/progression and inhibited vascular inflammation.CONCLUSIONS:PCSK9 critically mediates macrophage inflammation and elastin degradation, promoting AAA formation. PCSK9 inhibitors bear a promise to curtail AAA, beyond being used as cholesterol-lowering drugs.
- PRKAG2 Syndrome Caused by a Novel Missense Variant Mimicked Sporadic Hypertrophic Cardiomyopathy Until Its Progression to Burned-Out Phaseby Takashi Hiruma Shunsuke Inoue Toshiyuki Ko Seitaro Nomura Ryo Abe Chie Bujo Junichi Ishida Norifumi Takeda Eisuke Amiya Masaru Hatano Hiroyuki Abe Hiroyuki Morita Minoru Ono Norihiko Takeda Issei Komuro Department of Cardiovascular Medicine (T.H., R.A., C.B., J.I., Norifumi Takeda, E.A., H.M., Norihiko Takeda), Graduate School of Medicine, University of Tokyo, Japan. Department of Frontier Cardiovascular Science (S.I., T.K., S.N., I.K.), Graduate School of Medicine, University of Tokyo, Japan. Department of Pathology (H.A.), Graduate School of Medicine, University of Tokyo, Japan. Department of Cardiovascular Surgery (M.O.), Graduate School of Medicine, University of Tokyo, Japan. Advanced Medical Center for Heart Failure, University of Tokyo Hospital, Japan (M.H.). International University of Health and Welfare, Tokyo, Japan (I.K.). on November 25, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012047, December 1, 2024. <br/>
- Mineralocorticoid Receptor Antagonists in Heart Failure: An Updateby João Pedro Ferreira Bertram Pitt Faiez Zannad Department of Physiology and Cardiothoracic Surgery, Cardiovascular R&D Centre – UnIC@RISE, Faculty of Medicine of the University of Porto, Portugal (J.P.F.). Heart Failure Clinic, Internal Medicine Department, Unidade Local de Saude de Gaia/Espinho, Gaia, Portugal (J.P.F.). Université de Lorraine, Inserm, Centre d’Investigation Clinique Plurithématique 1433, Nancy, France (J.P.F., F.Z.). University of Michigan School of Medicine, Ann Arbor (B.P.). on November 25, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011629, December 1, 2024. <br/>Spironolactone, a steroidal mineralocorticoid receptor antagonist (MRA), has been used to treat patients with heart failure (HF) for more than half a century. Spironolactone improved outcomes in patients with severely symptomatic HF with reduced ejection fraction, and later, eplerenone expanded the benefits to patients with mildly symptomatic HF with reduced ejection fraction and myocardial infarction complicated by HF. Spironolactone reduced HF events in some patients with HF with preserved ejection fraction, but the results were not generalizable to all patients with HF with preserved ejection fraction. More recently, the nonsteroidal MRA finerenone improved the HF outcomes of patients with HF with preserved ejection fraction, expanding the benefits previously seen among patients with diabetes and albuminuric chronic kidney disease. The use of MRAs has been limited due to excessive concern about hyperkalemia. Education about the limited true risk associated with hyperkalemia, and about how to predict, prevent, and manage hyperkalemia, may lead to wider acceptability and use of these agents. Several ongoing trials are testing steroidal and nonsteroidal MRAs in HF populations. In this review, we perform a critical appraisal of MRA use in HF populations and point toward future directions.
- Home Blood Pressure Measurements Are Not Performed According to Guidelines and Standardized Education Is Urgently Neededby Eleanor Clapham Dean S. Picone Samuel Carmichael George S. Stergiou Norm R.C. Campbell John Stevens Carol Batt Aletta E. Schutte Niamh Chapman Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Australia (E.C., D.S.P., S.C., J.S., C.B., N.C.). Faculty of Health and Medicine, School of Health Sciences, The University of Sydney, NSW, Australia (D.S.P., N.C.). Third Department of Medicine, Hypertension Center STRIDE-7, School of Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Greece (G.S.S.). Departments of Medicine, Community Health Sciences and Physiology and Pharmacology, University of Calgary, AB, Canada (N.R.C.C.). School of Population Health, The George Institute for Global Health, University of New South Wales, Sydney, Australia (A.E.S.). on November 25, 2024
Hypertension, Volume 82, Issue 1, Page 149-159, January 1, 2025. <br/>BACKGROUND:Patient education is needed to perform home blood pressure measurement (HBPM) according to blood pressure (BP) guidelines. It is not known how BP is measured at home and what education is provided, which was the aim of the study.METHODS:Mixed-methods study among Australian adults who perform HBPM (June to December 2023). Participants completed a 30-item online survey on whether they followed guideline recommendations and the education they received for HBPM. Phone interviews were conducted among a purposive sample to further explore survey topics.RESULTS:Participants (n=350) were middle-aged (58±16 years; 54% women), and most (n=250, 71%) had hypertension. Guideline recommendations for HBPM were not always followed by survey participants. Most participants measured BP seated (n=316, 90%) with the cuff fitted to a bare arm (n=269, 77%). Only 15% measured BP in the morning and evening (n=54) and 26% averaged the BP readings over 7 days (n=90). Interview participants (n=34) described measuring BP at “different times of the day after doing different things.” One-third of participants (n=112, 37%) received education for HBPM, which interview participants described as vague verbal instructions from health care practitioners. Participants who received education did not perform high-quality HBPM. Participants who did not receive education mimicked BP measurement methods of health care practitioners, “I do it the way I’ve seen them do it.”CONCLUSIONS:HBPM is not performed according to guideline recommendations, and adults who received ad hoc education did not perform high-quality HBPM. These findings highlight a need for effective education to support HBPM for clinical decision-making.
- Redefining Cardiac Antibody-Mediated Rejection With Donor-Specific Antibodies and Graft Dysfunctionby Jason F. Goldberg Xin Tian Ann Bon Yifei Xu Eleanor Gerhard Ruth Brower Moon Kyoo Jang Hyesik Kong Temesgen E. Andargie Woojin Park Samer S. Najjar Inna Tchoukina Keyur B. Shah Steven Hsu Maria E. Rodrigo Charles Marboe Gerald J. Berry Hannah A. Valantine Palak Shah Sean Agbor-Enoh Inova Schar Heart and Vascular, Falls Church, VA (J.F.G., P.S.). Inova L.J. Murphy Children’s Hospital, Falls Church, VA (J.F.G.). Genomic Research Alliance for Transplantation (J.F.G., X.T., R.B., M.K.J., H.K., T.E.A., S.S.N., I.T., K.B.S., S.H., M.E.R., C.M., G.J.B., H.A.V., P.S., S.A.-E.). Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Heath, Bethesda, MD (X.T., Y.X., R.B., M.K.J., H.K., T.E.A., W.P., S.A.-E.). Brown University, Providence, RI (A.B.). Weill Cornell Medical College, New York, NY (E.G.). MedStar Health, Baltimore, MD (S.S.N.). Virginia Commonwealth University, Richmond (I.T., K.B.S.). Johns Hopkins School of Medicine, Baltimore, MD (S.H., S.A.-E.). MedStar Washington Hospital Center, Washington, DC (M.E.R.). Columbia University Vagelos College of Physicians & Surgeons, New York, NY (C.M.). Stanford University School of Medicine, CA (G.B., H.V.). George Washington University School of Medicine, Washington, DC (P.S.). on November 25, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011592, December 1, 2024. BACKGROUND:Heart transplant recipients with donor-specific antibodies (DSAs) have an increased risk for antibody-mediated rejection. However, many patients with graft dysfunction and DSA do not have evidence of antibody-mediated rejection by endomyocardial biopsy (EMB).METHODS:Participants from this prospective, multicenter study underwent serial EMB, echocardiogram, DSA, and donor-derived cell-free DNA evaluations. Outcomes were defined as pAMR+ (pAMR≥1) or DSA+/left ventricle (LV) dysfunction (DSA presence+LVEF drop ≥10% to an LVEF≤50%). Cox regression evaluated the association between antibody-mediated rejection categories and death or sustained (for 3 months) reduction of LVEF to <50%.RESULTS:Two hundred sixteen patients (29% women, 39% Black race, median age 55 [interquartile range, 47–62] years) had 1488 EMB, 2792 DSA, 1821 echocardiograms, and 1190 donor-derived cell-free DNA evaluations. DSAs were present in 86 patients (40%). Fourteen patients had isolated pAMR+ episodes and 8 patients had isolated DSA+/LV dysfunction episodes; 2 patients had pAMR+ and then subsequently DSA+/LV dysfunction with pAMR+. Median %dd-cfDNA was significantly higher at diagnosis of pAMR+ (0.63% [interquartile range, 0.23–2.0];P=0.0002), or DSA+/LV dysfunction (0.40% [interquartile range, 0.36–1.24];P<0.0001), compared with patients without these outcomes (0.01% [interquartile range, 0.0001–0.10]). Both pAMR+ and DSA+/LV dysfunction were associated with long-term clinical outcome of death (n=18) or prolonged LV dysfunction (n=10): pAMR+ (hazard ratio, 2.8 [95% CI, 1.03–7.4];P=0.043); DSA+/LV dysfunction (hazard ratio, 26.2 [95% CI, 9.6–71.3];P<0.001); composite of both definitions (hazard ratio, 6.5 [95% CI, 2.9–14.3];P<0.001). Patients who developed pAMR+ or DSA+/LV dysfunction within the first 6 months of transplant were more likely to die within 3 years posttransplant (hazard ratio, 3.9 [95% CI, 1.03–14.6];P=0.031).CONCLUSIONS:Expanding the characterization of antibody-mediated rejection to include patients with DSA and concurrent allograft dysfunction identified DSA+ patients at risk for death and prolonged LV dysfunction.
- Mechanical Mitral Valve Avulsion and Mitral-Aortic Junction Pseudoaneurysm Following Surgical Treatment of Mitral Valve Aneurysm and Severe Aortic Insufficiency in a Patient With Spondyloarthropathy: A Rarity With a Possible Associationby Hongxia Ning Jiajing Ouyang Jun Yang Zongyi Xiu Tianxiang Gu Yang Bai Chunyan Ma Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning. (H.N., J.O., J.Y., Y.B., C.M.) Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, Liaoning. (Z.X., T.G.) Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China (H.N., J.O., J.Y., Y.B., C.M.). on November 25, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Antibody-Mediated Rejection: Beyond the Biopsyby April Stempien-Otero Elina Minami Department of Laboratory Medicine and Pathology (A.S.-O.), University of Washington School of Medicine, Seattle. Department of Medicine (A.S.-O., E.M.), University of Washington School of Medicine, Seattle. on November 25, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012438, December 1, 2024. <br/>
- Cerebral Blood Flow Dynamics in Neurogenic Orthostatic Hypotension: A Systematic Review and Meta-Analysisby Jacquie R. Baker Paul A. Beach Shaun I. Ranada Aishani Patel Jennifer Gewandter Can Ozan Tan Roy Freeman Satish R. Raj Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Alberta, Canada (J.R.B., S.I.R., A.P., S.R.R.) Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (J.R.B., S.I.R,. A.P., S.R.R). Department of Neurology, Emory University School of Medicine, Atlanta, GA (P.A.B.). Department of Anesthesiology and Perioperative Medicine, University of Rochester School of Medicine, NY (J.G.). Department of Electrical Engineering, Mathematics, and Computer Science, University of Twente, the Netherlands (C.O.T). Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (R.F.). on November 21, 2024
Hypertension, Volume 82, Issue 1, Page 106-117, January 1, 2025. BACKGROUND:Neurogenic orthostatic hypotension (nOH) causes pathological falls in standing blood pressure that may or may not be symptomatic. nOH also raises the risk of poor neurological outcomes irrespective of symptom presence, possibly reflecting subclinical cerebral hypoperfusion. Dynamic changes in cerebral blood flow velocity (CBFv) help infer how blood pressure fluctuations influence CBFv and cerebral autoregulation. Whether CBFv is impacted in nOH relative to related conditions without nOH and healthy controls (HC) remains unresolved. Whether nOH symptoms reflect greater CBFv falls is also unclear. This review aimed to compare CBFv between nOH and HC, nOH and disease-matched controls (eg, Parkinson disease±nOH), and between symptomatic and asymptomatic nOH.METHODS:Embase and MEDLINE were searched up to April 2024. Means, SDs, and sample sizes for supine and upright CBFv were extracted to generate standardized effect sizes (Hedge g). Random-effects modeling compared postintervention between-group effect sizes.RESULTS:Seventeen studies were included for review. Thirteen studies were suitable for meta-analysis comparing nOH to HC, 2 comparing disease-matched controls to nOH and to HC, and 3 for symptomatic comparisons. Compared with HC, nOH had larger drops in CBFv (Hedge g, −0.64 [95% CI, −0.85 to −0.44];P<0.001). CBFv falls between nOH and disease-matched controls were similar (P=0.17). Symptomatic nOH had larger CBFv drops (Hedge g, 0.84 [95% CI, 0.212–1.461];P=0.009) than asymptomatic nOH.CONCLUSIONS:nOH causes significant orthostatic reductions in CBFv compared with HC, and symptomatic patients experience greater falls in CBFv than asymptomatic patients. Recognizing the clinical implications of CBFv in nOH is crucial for mitigating adverse outcomes.
- Intravital Imaging of Disease Mechanisms in a Mouse Model of CCM Skin Lesionsby Michael M. Orlich Amanda Norrén Malavika Bimal Desai Annegret Holm Harry Kozakewich Hans Schoofs Taija Mäkinen Joyce Bischoff Konstantin Gaengel Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., A.N., M.B.D., H.S., T.M., K.G.). Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (M.M.O.). Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, MA. (A.H., J.B.) Department of Pathology, Boston Children’s Hospital, Harvard Medical School, MA. (H.K.) on November 21, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Cerebral cavernous malformation (CCM) is a disease characterized by vascular malformations that primarily develop in the brain. These malformations are prone to leak, and their rupture or thrombotic closure can cause life-threatening hemorrhages and strokes. Mouse models have been instrumental to study the disease, but most cause premature lethality, precluding the investigation of disease mechanisms through intravital microscopy. Current mouse models also do not recapitulate human CCM skin lesions.METHODS:Endothelial-specific deletion ofCcm3via systemic tamoxifen application at postnatal day 4 or 5 prolongs survival and induces vascular malformations in the mouse brain and ear skin. CCM skin lesions can also be induced by topic tamoxifen administration directly to the ear. The thin, flat morphology of the ear skin is ideal for intravital microscopy. Dextran dyes and platelet markers allow to study blood flow and blood clot formation, in living animals in real time.RESULTS:We report that human CCM skin lesions can be recapitulated in a mouse model and that skin lesions share hallmarks of CCM brain lesions. Intravital imaging reveals that CCM skin lesions are slow-flow malformations prone to thrombus formation.CONCLUSIONS:Intravital imaging of CCM skin lesions expands the toolkit of CCM research and allows longitudinal studies of lesion growth.
- Andexanet Alfa–Associated Heparin Resistance in Cardiac Surgery: Mechanism and In Vitro Perspectivesby Charlene V. Chabata Haixiang Yu Lei Ke James W. Frederiksen Prakash A. Patel Bruce A. Sullenger Nabil K. Thalji Department of Pharmacology and Cancer Biology, Duke University, Durham, NC. (C.V.C., H.Y., B.A.S.) Department of Surgery, Duke University, Durham, NC. (H.Y., J.W.F., B.A.S.) Department of Anesthesiology, Jefferson Abington Hospital, PA (P.A.P.). Division of Cardiothoracic Anesthesiology, Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (L.K., N.K.T.). on November 21, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Andexanet alfa (andexanet) is the only Food and Drug Administration–approved antidote for direct FXa (factor Xa) inhibitors but has been reported to cause resistance to unfractionated heparin (UFH). This has delayed anticoagulation for procedures requiring cardiopulmonary bypass. The mechanism, andexanet and UFH dose dependence, and thrombotic risk of andexanet-associated heparin resistance are unknown.METHODS:The effect of andexanet in vitro was determined using activated clotting times and thromboelastography. Ex vivo cardiopulmonary bypass circuits were used to determine whether andexanet impaired anticoagulation for extracorporeal circulation. Kinetics of AT (antithrombin) inhibition of FXa and thrombin were measured in the presence of andexanet. Equilibrium modeling and thrombin generation assay validation were used to predict the role of andexanet, AT, and UFH concentrations in andexanet-associated heparin resistance.RESULTS:Andexanet prevented UFH-mediated prolongation of activated clotting times and thromboelastography times. At lower concentrations of andexanet, heparin resistance could be overcome with suprapharmacologic doses of UFH, but not at higher andexanet concentrations. Andexanet rendered standard doses of UFH inadequate to prevent circuit thrombosis, and suprapharmacologic UFH doses were only partially able to overcome this. Scanning electron microscopy demonstrated coagulation activation in circuits. Andexanet prevented UFH enhancement of AT-mediated inhibition of FXa and thrombin. Equilibrium modeling and thrombin generation assay validation demonstrated that andexanet creates a triphasic equilibrium with UFH and AT: initial UFH unresponsiveness, normal UFH responsiveness when andexanet is depleted, and finally AT depletion. Sufficient cardiopulmonary bypass heparinization can only occur at low therapeutic andexanet doses and normal AT levels. Higher andexanet doses or AT deficiency may require high UFH doses and potentially AT supplementation.CONCLUSIONS:Andexanet causes heparin resistance due to redistribution of UFH-bound AT. If andexanet cannot be avoided before heparinization and direct thrombin inhibitors are undesirable, our in vitro study suggests excess UFH should be considered as a potential strategy before AT supplementation.
- Mechanisms and Screening for Atherosclerosis in Adults With Vasculitisby Michael J. Hendrickson Zachary S. Wallace Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston (M.J.H., Z.S.W.). Rheumatology and Allergy Clinical Epidemiology Research Center, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston (Z.S.W.). on November 21, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>Vascular inflammation is a hallmark of both primary systemic vasculitis and atherosclerosis. As such, cardiovascular events are common in patients with vasculitis and likely due to both direct vascular inflammation and accelerated atherosclerosis. Direct cardiac involvement is possible in all vasculitides, though more commonly described in Takayasu arteritis, polyarteritis nodosa, and eosinophilic granulomatosis with polyangiitis. Accelerated atherosclerosis has been described in Takayasu arteritis and antineutrophil cytoplasmic antibody–associated vasculitis, though there remains a paucity of data in other forms of vasculitis. Multiple screening and management approaches for cardiovascular risk in people with vasculitis have been proposed, though evidence-based guidelines are lacking. In this review, we discuss the latest evidence in epidemiology, mechanisms, and screening for atherosclerosis in patients with primary systemic vasculitides.
- Renal Proximal Tubule Cell-Specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed a Western Dietby Naofumi Amioka Michael K. Franklin Masayoshi Kukida Liyuan Zhu Jessica J. Moorleghen Deborah A. Howatt Yuriko Katsumata Adam E. Mullick Motoko Yanagita Michelle M. Martinez-Irizarry Ruben M. Sandoval Kenneth W. Dunn Hisashi Sawada Alan Daugherty Hong S. Lu Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.). Sanders-Brown Center on Aging (Y.K.). Department of Biostatistics (Y.K.). Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.). Ionis Pharmaceuticals, Carlsbad (A.E.M.). Department of Nephrology, Kyoto University Graduate School of Medicine (M.Y.). WPI Institute for the Advanced Study of Human Biology, Kyoto University, Japan (M.Y.). Department of Medicine, Indiana University, Indianapolis (M.M.M.-I., R.M.S., K.W.D.). on November 21, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Pharmacological inhibition of megalin (also known as LRP2 [low-density lipoprotein receptor-related protein-2]) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice.METHODS:FemaleLrp2f/f mice were bred with maleNdrg1-Cre ERT2+/0 mice to develop PTC-LRP2 +/+ and PTC-LRP2 −/− littermates. To study atherosclerosis, all mice were bred to an LDL (low-density lipoprotein) receptor −/− background and fed a Western diet to induce atherosclerosis.RESULTS:PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor −/− mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 −/− mice fed a Western diet but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 −/− mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not on hypercholesterolemia. In contrast, female PTC-LRP2 −/− mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished ALB (albumin) accumulation in PTCs within 10 days of Western diet feeding. RNA-sequencing analyses demonstrated the upregulation of inflammation-related pathways in the kidney.CONCLUSIONS:PTC-specific megalin deletion does not affect atherosclerosis but leads to tubulointerstitial nephritis in mice fed a Western diet, with severe pathologies in male mice.
- Response of Blood Pressure to Renal Denervation Is Not Associated With Genetic Variantsby Christian Delles Roland E. Schmieder Rónán Daly Dennis Kannenkeril Agnes Bosch Lucas Lauder Michael Kunz Michael Böhm Graham Hamilton Raphael S. Schmieder Axel Schmid Pawel Herzyk Felix Mahfoud School of Cardiovascular and Metabolic Health (C.D.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany. Glasgow Polyomics, Wolfson Wohl Cancer Research Centre (R.D., G.H., P.H.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany. University of Glasgow, UK. Department of Nephrology and Hypertension (R.E.S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany. Institute of Radiology (R.S.S., A.S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany. Department of Internal Medicine III, Cardiology, Angiology, Intensive Care Medicine, Saarland University Hospital, Homburg, Germany (L.L., M.K., M.B., F.M.). Department of Cardiology, University Heart Center Basel, University Hospital Basel, Switzerland (L.L., M.K., F.M.). Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge (F.M.). on November 21, 2024
Hypertension, Volume 82, Issue 1, Page 118-125, January 1, 2025. BACKGROUND:Renal denervation lowers blood pressure (BP) in patients with uncontrolled hypertension. We conducted an unbiased genomic screen to identify genetic variants that may associate with BP response to renal denervation (RDN).METHODS:Patients (n=268) with uncontrolled resistant hypertension (baseline BP, 166±21/90±15 mm Hg) who underwent endovascular RDN using the Symplicity catheter (Medtronic, Inc, Santa Rosa, CA) were included. Reduction in 24-hour ambulatory systolic BP was assessed at 6 months and divided into 2 groups: above and below the median response of 6.0 mm Hg, taking preintervention 24-hour ambulatory BP and regression to the mean into account. Whole exome sequencing assessing 249 669 variants, was conducted using Illumina NovaSeq technology read on a NovaSeq S4 Flow Cell device.RESULTS:We did not identify individual gene variants associated with BP response following RDN. These findings were confirmed after adjustment for sex and in a sensitivity analysis looking at tertiles of BP response. We also explored specific variants inAGT,ADD1, ADRB1,ADRB2, andSCNN1Athat have been proposed as potential candidate genes for response and found no association (allP>0.13). Gene ontology analysis of variants across the 2 responder groups highlighted differences in biologic processes such as cell adhesion and molecular function such as protein tyrosine kinase activity.CONCLUSIONS:The response to RDN, in terms of 24-hour BP reduction, was not associated with the genetic profile of patients with resistant hypertension. These data do not support the use of a genetic score to identify potential responders to RDN.
- Natural Language Processing to Adjudicate Heart Failure Hospitalizations in Global Clinical Trialsby Pablo M. Marti-Castellote Christopher Reeder Brian L. Claggett Pulkit Singh Emily S. Lau Shaan Khurshid Puneet Batra Steven A. Lubitz Mahnaz Maddah Orly Vardeny Eldrin F. Lewis Marc A. Pfeffer Pardeep S. Jhund Akshay S. Desai John J.V. McMurray Patrick T. Ellinor Jennifer E. Ho Scott D. Solomon Jonathan W. Cunningham Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Cardiovascular Disease Initiative, Broad Institute of Harvard University and the Massachusetts Institute of Technology, Cambridge, MA Cardiovascular Disease Initiative, Broad Institute of Harvard University and the Massachusetts Institute of Technology, Cambridge, MA; Data Sciences Platform, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA Cardiovascular Disease Initiative, Broad Institute of Harvard University and the Massachusetts Institute of Technology, Cambridge, MA; Division of Cardiology, Massachusetts General Hospital, Boston, MA Cardiovascular Disease Initiative, Broad Institute of Harvard University and the Massachusetts Institute of Technology, Cambridge, MA; Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA Minneapolis VA Hospital, University of Minnesota, Minneapolis, MN Stanford University School of Medicine, Palo Alto, CA British Heart Foundation Cardiovascular Research Centre, University of Glasgow, United Kingdom Cardiovascular Disease Initiative, Broad Institute of Harvard University and the Massachusetts Institute of Technology, Cambridge, MA; CardioVascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA on November 16, 2024
Circulation: Heart Failure, Ahead of Print. <br/>Background:Medical record review by a physician clinical events committee is the gold standard for identifying cardiovascular outcomes in clinical trials, but is labor-intensive and poorly reproducible. Automated outcome adjudication by artificial intelligence (AI) could enable larger and less expensive clinical trials, but has not been validated in global studies.Methods:We developed a novel model for automated AI-based heart failure adjudication (“HF-NLP”) using hospitalizations from three international clinical outcomes trials. This model was tested on potential heart failure hospitalizations from the DELIVER trial, a cardiovascular outcomes trial comparing dapagliflozin with placebo in 6063 patients with heart failure with mildly reduced or preserved ejection fraction. AI-based adjudications were compared with adjudications from a clinical events committee that followed FDA-based criteria.Results:AI-based adjudication agreed with the clinical events committee in 83% of events. A strategy of human review for events that the AI model deemed uncertain (16%) would have achieved 91% agreement with the clinical events committee while reducing adjudication workload by 84%. The estimated effect of dapagliflozin on heart failure hospitalization was nearly identical with AI-based adjudication (hazard ratio 0.76 [95% CI 0.66-0.88]) compared to clinical events committee adjudication (hazard ratio 0.77 [95% CI 0.67-0.89]). The AI model extracted symptoms, signs, and treatments of heart failure from each medical record in tabular format and quoted sentences documenting them.Conclusions:AI-based adjudication of clinical outcomes has the potential to improve the efficiency of global clinical trials while preserving accuracy and interpretability.
- Irisin Improves Preeclampsia by Promoting Embryo Implantation and Vascular Remodelingby Dawei Zhu Jie Huang Yujie Wu Lin Fan Yijun Liu Qianwen Zhang Li Li Jian Han Xinghui Liu Department of Obstetrics and Gynecology (D.Z., L.F., Y.L., Q.Z.), West China Second University Hospital, Sichuan University, Chengdu. Laboratory of the Key Perinatal Diseases (Y.W.), West China Second University Hospital, Sichuan University, Chengdu. Department of Gynaecology and Obstetrics, Daping Hospital, Army Medical University, Chongqing, China (J. Huang, L.L., J. Han). West China Second University Hospital, Chengdu, China (X.L.) on November 14, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Preeclampsia is a pregnancy-specific disorder with unclear pathogenesis. Irisin, a recently identified exercise-induced factor, significantly influences lipid metabolism and cardiovascular function. Nonetheless, its role in trophoblast development during human placentation and the related intracellular signaling pathways remain poorly understood.METHODS:We assessed peripheral blood irisin expression in early pregnancy among patients with preeclampsia and its correlation with key clinical indicators. In trophoblast cell lines and mice, we used exogenous irisin and viral knockdown to investigate functional changes. Phosphorylation-specific antibody arrays and dual-luciferase reporter assays were used to explore downstream molecular mechanisms, which were subsequently validated in trophoblast cell lines and relevant gene knockout mice.RESULTS:In early pregnancy, patients with preeclampsia exhibit decreased peripheral blood irisin levels, occurring earlier than traditional predictive markers, such as PLGF (placental growth factor) and sFlt-1 (soluble fms-like tyrosine kinase-1). Furthermore, irisin concentration is positively correlated with proteinuria and abnormal blood pressure during pregnancy. Exogenous irisin significantly enhanced trophoblast cell migration, invasion, and proliferation while inhibiting apoptosis. It also increased STAT (signal transducers and activators of transcription) 4 phosphorylation and its binding to the GLUT (glucose transporter)-3 promoter, resulting in elevated GLUT-3 expression and glucose uptake in trophoblast cells. In vivo, increased peripheral irisin promoted embryo implantation, vascular remodeling, and enhanced glucose uptake, whereas reduced irisin resulted in a preeclampsia-like phenotype characterized by elevated blood pressure, proteinuria, renal-placental dysfunction, adipose accumulation, and restricted fetal growth.CONCLUSIONS:Peripheral irisin improves preeclampsia by promoting embryo implantation and vascular remodeling through the activation of the STAT4/GLUT-3 pathway. Reduced peripheral irisin may contribute to preeclampsia-like pathologies. This study supports the advocacy for appropriate exercise during early pregnancy and provides new insights for preeclampsia prevention.
- Left Ventricular Hypertrophy in Women With a History of Preeclampsiaby Maria G. Hauge Peter Damm Klaus F. Kofoed Emma Louise Ries Møller Andrea G. Lopez Anne S. Ersbøll Marianne Johansen Per E. Sigvardsen Michael H.C. Pham Jens P. Goetze Andreas Fuchs Jørgen T. Kühl Børge G. Nordestgaard Lars V. Køber Finn Gustafsson Jesper J. Linde Department of Gynecology, Fertility and Obstetrics, Rigshospitalet (M.G.H., P.D., A.S.E., M.J.), Copenhagen University Hospital, Denmark. Department of Cardiology, The Heart Centre, Rigshospitalet (K.F.K., E.L.R.M., P.E.S., M.H.C.P., A.F., L.V.K., F.G., J.J.L.), Copenhagen University Hospital, Denmark. Department of Biomedical Sciences (J.P.G.), Copenhagen University Hospital, Denmark. Department of Radiology, The Diagnostic Center, Rigshospitalet (K.F.K.), Copenhagen University Hospital, Denmark. Department of Clinical Medicine, University of Copenhagen, Denmark (M.G.H., P.D., K.F.K., J.P.G., B.G.N., L.V.K., F.G.). Department of Clinical Biochemistry, Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark (B.G.N.). Department of Cardiology, Zealand University Hospital, Roskilde, Denmark (J.T.K.). Hospital Universitario de Puerto Real, Cádiz, Spain (A.G.L.). on November 14, 2024
Hypertension, Ahead of Print. BACKGROUND:As a hypertensive disorder of pregnancy, preeclampsia is associated with increased cardiovascular morbidity and mortality later in life. Since early signs of myocardial affection could indicate a higher risk of future cardiovascular disease manifestations, we investigated whether women with prior preeclampsia have a higher prevalence of left ventricular hypertrophy compared with women from the general population and to what extent chronic hypertension affects any potential difference.METHODS:In a cohort study, women aged 40 to 55 years with prior preeclampsia were compared with age- and parity-matched women from the general population. They underwent a research cardiac computed tomography, and the primary outcome was left ventricular hypertrophy, defined as a left ventricular mass index >30 g/m2.7.RESULTS:In 679 women with prior preeclampsia and 672 controls (median age, 47 years), we found a higher prevalence of left ventricular hypertrophy (14.0% versus 6.4%) in the preeclampsia group with an odds ratio of 1.62, 95% CI (1.07–2.46),P=0.024, median of 15 years (range, 0–28) after pregnancy, after adjustment for cardiovascular risk factors, including chronic hypertension. Left ventricular hypertrophy was more frequent among women with preeclampsia with (26.2% versus 15.6%) and without (5.5% versus 2.4%) chronic hypertension, and a mediation analysis showed that chronic hypertension explained 22% of the association between preeclampsia and left ventricular hypertrophy.CONCLUSIONS:Women with prior preeclampsia had a 2-fold higher prevalence of left ventricular hypertrophy compared with women from the general population, and preeclampsia was independently associated with left ventricular hypertrophy, regardless of the presence of cardiovascular risk factors, including chronic hypertension.REGISTRATION:URL:https://www.clinicalTrials.gov; Unique identifier: NCT03949829.
- VEGFC Overexpression in Kidney Progenitor Cells Is a Model of Renal Lymphangiectasiaby Michael D. Donnan Dilip K. Deb Vidhi Dalal Valentin David Daniele Procissi Susan E. Quaggin Northwestern University Feinberg School of Medicine, Chicago, IL. on November 14, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:Lymphangiogenesis is believed to be a protective response in the setting of multiple forms of kidney injury and mitigates the progression of interstitial fibrosis. To augment this protective response, promoting kidney lymphangiogenesis is being investigated as a potential treatment to slow the progression of kidney disease. As injury-related lymphangiogenesis is driven by signaling from the receptor VEGFR3 (vascular endothelial growth factor receptor 3) in response to the cognate growth factor VEGF (vascular endothelial growth factor)-C released by tubular epithelial cells, this signaling pathway is a candidate for future kidney therapeutics. However, the consequences to kidney development and function to targeting this signaling pathway remain poorly defined.METHODS:We generated a new mouse model expressingVegfcunder regulation of the nephron progenitor Six2Cre driver strain (Six2Vegfc). Mice underwent a detailed phenotypic evaluation. Whole kidneys were processed for histology and 3-dimensional imaging.RESULTS:Six2Vegfcmice had reduced body weight and kidney function compared with littermate controls.Six2Vegfckidneys demonstrated large peripelvic fluid-filled lesions with distortion of the pelvicalcyceal system which progressed in severity with age. Three-dimensional imaging showed a 3-fold increase in total cortical vascular density. Histology confirmed a substantial increase in LYVE1+ (lymphatic vessel endothelial hyaluronan receptor-1)/PDPN+ (podoplanin)/VEGFR3+ lymphatic capillaries extending alongside EMCN+ (endomucin) peritubular capillaries. There was no change in EMCN+ peritubular capillary density.CONCLUSIONS:Kidney lymphatic density was robustly increased in theSix2Vegfcmice. There were no changes in peritubular blood capillary density despite these endothelial cells also expressing VEGFR3. The model resulted in malformation of the lymphatic hilar plexus, resulting in severe hydronephrosis that resembled a human condition termed renal lymphangiectasia. This study defines the vascular consequences of augmenting VEGFC signaling during kidney development and provides new insight into human renal lymphatic malformations.
- Emerging Imaging Techniques for Atherosclerosis in Systemic Immune-Mediated Inflammatory Conditionsby George Markousis-Mavrogenis Muzzamal Habib Daniel M. Huck Florian Andre Henning Steen Monica Mukherjee Sophie I. Mavrogeni Brittany Weber Departments of Cardiology (G.M.-M., F.A., H.S.), Heidelberg University, Germany. Angiology (G.M.-M., F.A., H.S.), Heidelberg University, Germany. Pneumology (G.M.-M., F.A., H.S.), Heidelberg University, Germany. German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Germany (G.M.-M., F.A., H.S.). University Research Institute of Maternal and Child Health and Precision Medicine and UNESCO Chair in Adolescent Healthcare, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, Greece (G.M.-M., S.I.M.). Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (M.H., D.M.H., B.W.). Division of Cardiology, Johns Hopkins University, Baltimore, MD (M.M.). Onassis Cardiac Surgery Center, Athens, Greece (S.I.M.). University of Padova, Italy (S.I.M.). on November 14, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>Atherosclerosis affects patients with systemic immune-mediated inflammatory diseases at an increased rate compared with the general population. In recent years, our understanding of the pathophysiology of atherosclerosis has advanced considerably. Nevertheless, cardiovascular imaging modalities that can adequately assess the biological background of atherosclerosis have not reached widespread clinical adoption. Novel developments in cardiac imaging have the potential to enhance the diagnostic yield of these modalities further while providing essential insights into the anatomy, composition, and biology of atherosclerotic lesions. In this review, we highlight some of the latest developments in the field for the evaluation of atherosclerosis using advances in echocardiography, computed tomography, positron emission tomography, and cardiovascular magnetic resonance. Additionally, we discuss evidence specifically in patients with immune-mediated inflammatory diseases and outline unmet research needs for future development.
- Hidden Power: PAR4’s Role in Liver Damage From Acetaminophen Overdose in Miceby NaShea C. Kendrick Marvin T. Nieman Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH. on November 14, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>
- Reproducibility and Treatment Effect on Office and Ambulatory Pressure Relationby Giuseppe Mancia Rita Facchetti Fosca Quarti-Trevano Guido Grassi University Milano-Bicocca, Milan, Italy (G.M., R.F., G.G.). Department of Medicine, San Gerardo Hospital, University of Milano-Bicocca, Milan, Italy (F.Q.-T.). Clinica Medica, Department of Medicine and Surgery, University Milano-Bicocca, Italy (R.F., F.Q.-T., G.G.). on November 14, 2024
Hypertension, Volume 82, Issue 1, Page 126-135, January 1, 2025. BACKGROUND:In the absence of outcome-based ambulatory blood pressure (BP) trails hypertension guidelines provide 24-hour mean BP values corresponding to trial-validated office BP values. Data are shown for untreated and treated patients together, but whether corresponding ambulatory values are similar in untreated and treated hypertensives and reproducible at yearly measurements during treatment is undefined.METHODS:In 2397 patients of the ELSA (European Lacidipine Study on Atherosclerosis) and PHYLLIS (Plaque Hypertension Lipid-Lowering Italian Study) trials, we calculated the office and 24-hour BP relationship according to the linear regression model, with office systolic BP as the independent variable, at baseline and yearly during a 3-year treatment. Twenty-four hour BP values corresponding to clinically important office BP values (hypertension grading and treatment thresholds and targets) were calculated and compared with those provided by guidelines.RESULTS:Office and 24-hour systolic BP or diastolic BP always exhibited a significant linear relationship, with, however, limited Pearson correlation coefficients (never >0.44).The slopes of the relationship were superimposable between different years of treatment but always significantly less steep than the slope seen in untreated individuals. Compared with the guideline-provided corresponding values, 24-hour BP showed qualitative and quantitative differences; for example, it was considerably lower and higher than the guideline-corresponding values when office BP was in the high hypertension and low treatment target ranges, respectively.CONCLUSIONS:In treated patients with hypertension the slope of the office and 24-hour BP linear regression is reproducible over time. However, the slopes are steeper in untreated individuals, indicating that information on ambulatory BP values corresponding to office BP values can be more accurate if separately estimated in these 2 conditions.
- Exposure to Volatile Organic Compounds and Blood Pressure in NHANES 2011 to 2018by Katlyn E. McGraw Arce Domingo-Relloso Daniel W. Riggs Danielle N. Medgyesi Raghavee Neupane Jeanette A. Stingone Tiffany R. Sanchez Department of Environmental Health Science (K.E.M., A.D.-R., D.N.M., T.R.S.), Columbia University Mailman School of Public Health, New York, NY. Department of Epidemiology (J.A.S.), Columbia University Mailman School of Public Health, New York, NY. Christina Lee Brown Envirome Institute, University of Louisville, KY (D.W.R.). Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL (R.N.). on November 13, 2024
Hypertension, Volume 82, Issue 1, Page 136-148, January 1, 2025. <br/>BACKGROUND:Volatile organic compounds (VOCs) are ubiquitous environmental pollutants. Exposure to VOCs is associated with cardiovascular disease risk factors, including elevated blood pressure in susceptible populations. However, research in the general population, particularly among nonsmoking adults, is limited. We hypothesized that higher VOC exposure is associated with higher blood pressure and hypertension, among nonsmokers.METHODS:We included 4 cycles of data (2011–2018) of nonsmoking adults (n=4430) from the National Health and Nutrition Examination Survey. Urinary VOC metabolites were measured by ultraperformance liquid chromatography-mass spectrometry, adjusted for urine dilution, and log-transformed. We estimated mean differences in blood pressure using linear models and prevalence ratio of stage 2 hypertension using modified Poisson models with robust standard errors. Models were adjusted for age, sex, race and ethnicity, education, body mass index, estimated glomerular filtration rate, and National Health and Nutrition Examination Survey cycle.RESULTS:Participants were 54% female, with a median age of 48 years, 32.3% had hypertension, and 7.9% had diabetes. The mean differences (95% CI) in systolic blood pressure were 1.61 (0.07–3.15) and 2.46 (1.01–3.92) mm Hg when comparing the highest with the lowest quartile of urinary acrolein (N-acetyl-S-[2-carboxyethyl]-L-cysteine) and 1,3-butadiene (N-acetyl-S-[3,4-dihydroxybutyl]-L-cysteine) metabolites. The prevalence ratios for hypertension were 1.06 (95% CI, 1.02–1.09) and 1.05 (95% CI, 1.01–1.09) when comparing the highest with lowest quartiles of urinary acrolein (N-acetyl-S-[2-carboxyethyl]-L-cysteine) and 1,3-butadiene (N-acetyl-S-[3,4-dihydroxybutyl]-L-cysteine), respectively.CONCLUSIONS:Exposure to VOCs may be a relevant yet understudied environmental contributor to cardiovascular disease risk in the nonsmoking, US population.
- Mechanistic Insights Into Redox Damage of the Podocyte in Hypertensionby Daria V. Ilatovskaya Amanda Behr Alexander Staruschenko Gentzon Hall Oleg Palygin Department of Physiology, Medical College of Georgia (D.V.I.), Augusta University, Augusta, GA. Department of Medical Illustration, College of Allied Health Sciences (A.B.), Augusta University, Augusta, GA. Department of Molecular Pharmacology and Physiology (A.S.), University of South Florida, Tampa, FL. Hypertension and Kidney Research Center (A.S.), University of South Florida, Tampa, FL. James A. Haley Veterans’ Hospital, Tampa, FL (A.S.). Division of Nephrology, Department of Internal Medicine, Duke University School of Medicine, Durham, NC (G.H.). Division of Nephrology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC (G.H.). Division of Nephrology, Department of Medicine (O.P.), Medical University of South Carolina, Charleston, SC. Department of Regenerative Medicine and Cell Biology (O.P.), Medical University of South Carolina, Charleston, SC. on November 13, 2024
Hypertension, Volume 82, Issue 1, Page 14-25, January 1, 2025. <br/>Podocytes are specialized cells within the glomerular filtration barrier, which are crucial for maintaining glomerular structural integrity and convective ultrafiltration. Podocytes exhibit a unique arborized morphology with foot processes interfacing by slit diaphragms, ladder-like, multimolecular sieves, which provide size and charge selectivity for ultrafiltration and transmembrane signaling. Podocyte dysfunction, resulting from oxidative stress, dysregulated prosurvival signaling, or structural damage, can drive the development of proteinuria and glomerulosclerosis in hypertensive nephropathy. Functionally, podocyte injury leads to actin cytoskeleton rearrangements, foot process effacement, dysregulated slit diaphragm protein expression, and impaired ultrafiltration. Notably, the renin-angiotensin system plays a pivotal role in podocyte function, with beneficial AT2R (angiotensin receptor 2)-mediated nitric oxide (NO) signaling to counteract AT1R (angiotensin receptor 1)-driven calcium (Ca2+) influx and oxidative stress. Disruption of this balance contributes significantly to podocyte dysfunction and drives albuminuria, a marker of kidney damage and overall disease progression. Oxidative stress can also lead to sustained ion channel–mediated Ca2+influx and precipitate cytoskeletal disorganization. The complex interplay between GPCR (G-protein coupled receptor) signaling, ion channel activation, and redox injury pathways underscores the need for additional research aimed at identifying targeted therapies to protect podocytes and preserve glomerular function. Earlier detection of albuminuria and podocyte injury through routine noninvasive diagnostics will also be critical in populations at the highest risk for the development of hypertensive kidney disease. In this review, we highlight the established mechanisms of oxidative stress–mediated podocyte damage in proteinuric kidney diseases, with an emphasis on a hypertensive renal injury. We will also consider emerging therapies that have the potential to selectively protect podocytes from redox-related injury.
- A Medical Expert System for Intelligent Telemonitoring of Chronic Heart Failure Patients: Preliminary Validation and Perspectivesby Annamaria Vianello Martina Olivelli Massimiliano Donati Luca Fanucci Alessio Bechini Ilaria Petrucci Stefano Masi Department of Primary Care, ASL Toscana Nord-Ovest, Italy Department of Information Engineering, University of Pisa, Italy Department of Internal Medicine, University of Pisa, Italy on November 11, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Mavacamten: Real-World Experience from 22 Months of the Risk Evaluation and Mitigation Strategy (REMS) Programby Milind Y. Desai Dewey Seto Michael Cheung Sonia Afsari Niki Patel Arnaud Bastien Jeffrey Lockman Michele Coiro Matthew W. Martinez Hypertrophic Cardiomyopathy Center, Heart Vascular Thoracic Institute, Cleveland Clinic, Cleveland, OH Bristol Myers Squibb, Princeton, NJ Hypertrophic Cardiomyopathy Center, Morristown Medical Center, Atlantic Health System, Morristown, NJ on November 11, 2024
Circulation: Heart Failure, Ahead of Print. Background:Mavacamten is the only cardiac myosin inhibitor approved by the US FDA for treatment of symptomatic New York Heart Association class II-III obstructive hypertrophic cardiomyopathy (HCM) patients. Under the risk evaluation and mitigation strategy (REMS) program for mavacamten, patients are required to be monitored for development of systolic heart failure, and reduction of left ventricular ejection fraction (LVEF) to <50%. We report results from the mavacamten REMS database (28-Apr-2022 to 27-Feb-2024).Methods:Data on healthcare providers and pharmacy certification, patient monitoring (from Patient Status Forms, based partly on echocardiograms), and screening for drug interactions prior to each dispense were collected.Results:Of 6,299 patients who received ≥1 dose of mavacamten, 60.0% were women; 64.6% were >60 years of age. Of 5,573 patients with submitted Patient Status Forms, 256 (4.6%) developed LVEF <50% and 71 (1.3%) experienced heart failure requiring hospitalization (HFH). On the 29,111 status forms in these patients, each representing an assessment of an echocardiogram, LVEF <50% was reported on 276 (0.9%) and HFH was reported on 86 (0.3%). Of 1,929 patients with ≥1 year of treatment, 78 (4.0%) had an LVEF reduction to <50% and 4 (0.2%) experienced LVEF <50% + HFH but later resumed treatment. Of 3,228 patients initiated on 5 mg/day mavacamten and were treated for at least 6 months, 2,391 (74.1%) remained at 5 or 10 mg/day. At 3- and 6-months following mavacamten treatment initiation, 57.2% and 70.3%, respectively, demonstrated post-Valsalva LV outflow tract gradient <30 mmHg.Conclusions:We describe the feasibility and experience of the first 22 months of the REMS program for prescribing mavacamten in >6,000 symptomatic obstructive HCM patients. The need for temporary interruption for LVEF <50% was low, including for patients on therapy ≥1 year, with even fewer LVEF reductions associated with HFH.
- Stop Dreaming: Mavacamten REMS Data Are Hereby Ahmad Masri Neal K. Lakdawala Hypertrophic Cardiomyopathy Center, Division of Cardiology, School of Medicine, Oregon Health & Science University, Portland, OR Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA on November 11, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Genetics of Hypertension: Additive and Interactive Effectsby Zdenka Pausova Johanne Tremblay Pavel Hamet Centre hospitalier universitaire Sainte-Justine and Department of Pediatrics, University of Montreal, QC, Canada (Z.P.). Departments of Physiology and Nutritional Sciences, The Hospital for Sick Children, University of Toronto, ON, Canada (Z.P.). Centre de recherche du Centre hospitalier de l’Université de Montréal, QC, Canada (J.T., P.H.). on November 11, 2024
Hypertension, Volume 82, Issue 1, Page 3-7, January 1, 2025. <br/>
- Comprehensive Proteomics Profiling Identifies Circulating Biomarkers to Distinguish Hypertrophic Cardiomyopathy from Other Cardiomyopathies with Left Ventricular Hypertrophyby Keitaro Akita Mathew S. Maurer Albree Tower-Rader Michael A. Fifer Yuichi J. Shimada Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA on November 11, 2024
Circulation: Heart Failure, Ahead of Print. Background:Distinguishing hypertrophic cardiomyopathy (HCM) from other cardiomyopathies with left ventricular hypertrophy (LVH), such as hypertensive LVH, transthyretin amyloid cardiomyopathy (ATTR-CM), and aortic stenosis (AS), is sometimes challenging. Using plasma proteomics profiling, we aimed to identify circulating biomarkers and dysregulated signaling pathways specific to HCM.Methods:In this multicenter case-control study, plasma proteomics profiling was performed in cases with HCM and controls with hypertensive LVH, ATTR-CM, and AS. Two-thirds of patients enrolled earlier in each disease group were defined as the training set, and the remaining one-third as the test set. Protein concentrations in HCM were compared with those in hypertensive LVH (comparison 1), ATTR-CM (comparison 2), and AS (comparison 3). Candidate proteins that meet the following 2 criteria were selected: (1) Higher abundance in HCM throughout all 3 comparisons or lower abundance in HCM throughout all 3 comparisons with univariable P<0.05 and |log2(fold change)| >0.5 in both the training and test sets and (2) Independently associated with HCM with multivariable P<0.05 after adjusting for clinical parameters significantly different between HCM and controls. Using the selected candidate proteins, a logistic regression model to distinguish HCM from controls was developed in the training set and applied to the test set. Finally, pathway analysis was performed in each comparison using proteins with different abundance.Results:Overall, 4,979 proteins in 1,415 patients (HCM, n=879; hypertensive LVH, n=331; ATTR-CM, n=169; AS, n=36) were analyzed. Of those, 5 proteins were selected as candidate proteins. The logistic regression model with these 5 proteins had an area under the receiver-operating-characteristic curve of 0.86 (95% CI 0.82–0.89) in the test set. The MAPK and HIF-1 pathways were dysregulated in HCM throughout the 3 comparisons.Conclusions:This study identified circulating biomarkers that distinguish HCM from other cardiomyopathies with LVH independently from confounders and revealed signaling pathways associated with HCM.
- Social Determinants of Health and Disparities in Guideline-Directed Medical Therapy Optimization for Heart Failureby Joshua A. Jacobs Iyanuoluwa Ayodele Adam P. Bress Madeline R. Sterling Ambarish Pandey Catherine G. Derington Alexander R. Zheutlin Kevin S. Shah Stephen J. Greene Brooke Alhanti Rosalia Blanco Gregg C. Fonarow Intermountain Healthcare Department of Population Health Sciences (J.A.J., A.P.B., C.G.D.), Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City. Division of Cardiovascular Medicine, Department of Internal Medicine (K.S.S.), Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City. Department of Pharmacy, University of Utah Health, University of Utah, Salt Lake City (J.A.J.). Duke Clinical Research Institute (I.A., S.J.G., B.A., R.B.), Duke University School of Medicine, Durham, NC. Division of Cardiology (S.J.G.), Duke University School of Medicine, Durham, NC. Division of General Internal Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY (M.R.S.). University of Texas Southwestern Medical Center, Dallas (A.P.). Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL (A.R.Z.). Division of Cardiology, Ahmanson-UCLA Cardiomyopathy Center, University of California Los Angeles Medical Center (G.C.F.). on November 11, 2024
Circulation: Heart Failure, Ahead of Print. <br/>BACKGROUND:Fewer than 20% of eligible patients with heart failure with reduced ejection fraction receive all 4 pillars of guideline-directed medical therapy. Understanding disparities by race, ethnicity, sex, and adverse social determinants of health is necessary to equitably optimize quadruple therapy.METHODS:Utilizing the American Heart Association’s Get With The Guidelines-Heart Failure registry, we examined associations between race and ethnicity, sex, and adverse social determinants of health (insurance type and documented social need [any barrier to accessing health care]) with quadruple therapy optimization (QTO) in patients with heart failure with reduced ejection fraction hospitalized between July 1, 2021, and September 30, 2023, with complete medication data at discharge. We calculated adjusted mean differences (AMDs) in the discharge QTO score (range, 0%–100%) reflecting the proportion of eligible use of renin-angiotensin system inhibitors, β-blockers, mineralocorticoid receptor antagonists, and sodium-glucose cotransporter-2 inhibitors and compared across demographic and adverse social determinants of health groups.RESULTS:Among 82 637 patients (median age, 66 years; 32.5% female; 57.0% non-Hispanic White; 76.4% prior heart failure with reduced ejection fraction), the overall mean QTO score was 56.2% (SD, 25.5). After adjustment, compared with non-Hispanic White individuals, Black (AMD, 2.56 percentage points [95% CI, 2.16–2.96]) and Hispanic individuals (AMD, 0.71 percentage points [95% CI, 0.11–1.31]) had higher QTO scores. Females had higher QTO scores than males (AMD, 1.94 percentage points [95% CI, 1.58–2.31]). Patients with no insurance (AMD, −4.90 percentage points [−5.62 to −4.17]), Medicaid (AMD, −0.45 percentage points [−0.89 to −0.01]), and Medicare (AMD, −1.64 percentage points [−2.10 to −1.18]) had lower QTO scores versus private insurance. Those with an identified social need (n=24 651) had lower QTO scores than those without (AMD, −3.40 percentage points [95% CI, −4.10 to −2.71]).CONCLUSIONS:Disparities in QTO were most evident for patients with no insurance, Medicaid, Medicare, or potentially an identified social need. Future efforts should focus on reducing gaps to improve equitable guideline-directed medical therapy use.
- Discrepancy in the Diagnosis of Heart Failure With Preserved Ejection Fraction Between Supine Versus Upright Exercise Hemodynamic Testingby Marat Fudim Veraprapas Kittipibul Ashley Swavely Anna Gray Jeffrey Mikitka Erin Young Olivia Dobbin Matthew Radzom Jacqueline Fee Jeroen Molinger Brandy Patterson Giovanni Battista Perego Luigi P. Badano Gianfranco Parati Jean-Luc Vachiéry Michele Senni Ettore Lanzarone Fabio Previdi Stefano Paleari Claudia Baratto Sergio Caravita Department of Medicine (M.F., V.K., A.S., A.G., J. Mikitka, E.Y., O.D., M.R., J.F., J. Molinger, B.P.), Duke University Medical Center, Durham, NC. Department of Anesthesiology (J. Molinger), Duke University Medical Center, Durham, NC. Duke Clinical Research Institute, Durham, NC (M.F., V.K.). Institute of Heart Diseases, Wroclaw Medical University, Poland (M.F.). Department of Cardiology, Ospedale San Luca Istituto di Ricerca e Cura a Carattere Scientifico, Istituto Auxologico Italiano, Milan, Italy (G.B.P., L.P.B., G.P., C.B., S.C.). Department of Medicine and Surgery, University of Milano-Bicocca, Italy (L.P.B., G.P., M.S.). Department of Cardiology, Hôpital Universitaire de Bruxelles, Hôpital Erasme, Université Libre de Bruxelles, Belgium (J.-L.V.). Cardiovascular Department, Azienda Socio Sanitaria Territoriale, Papa Giovanni XXIII, Bergamo, Italy (M.S.). Department of Management, Information, and Production Engineering, University of Bergamo, Dalmine BG, Italy (E.L., F.P., S.P., S.C., C.B.). on November 8, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012020, December 1, 2024. BACKGROUND:Invasive exercise right heart catheterization is a gold standard in diagnosing heart failure with preserved ejection fraction (HFpEF). Body positions during the test influence hemodynamics. However, the discrepancy in HFpEF diagnosis between exercise testing in supine versus upright position is unknown.METHODS:We conducted a 2-center prospective study enrolling patients referred for exercise right heart catheterization for HFpEF. We performed a Supright protocol integrating submaximal supine bicycle ergometry (20 W) followed by maximal upright bicycle ergometry with a breath-by-breath oxygen analyzer. HFpEF hemodynamic criteria specific to testing positions were applied. Patients were considered to have concordant HFpEF if they met criteria in both positions or discordant HFpEF if they met criteria only in the supine position.RESULTS:Of 36 patients who met HFpEF criteria in supine position, 18 (50%) did not meet criteria in upright position (discordant HFpEF). Discordant HFpEF had less atrial fibrillation (0% versus 55%;P<0.001), lower left atrial volume (60±14 versus 77±21 mL;P=0.010), and lower H2FPEF score (2.1±1.3 versus 5.1±2.3;P<0.001). In supine position, pulmonary arterial wedge pressure was lower in discordant HFpEF at rest (15±4 versus 19±7 mm Hg;P=0.040). In upright position, pulmonary arterial wedge pressure was lower in discordant HFpEF both at rest (8±4 versus 14±6 mm Hg;P=0.002) and at peak exercise (14±4 versus 27±7 mm Hg;P<0.001). Pulmonary arterial wedge pressure/cardiac output slope was lower in discordant HFpEF (1.6±1.7 versus 3.6±2.9;P<0.001). Maximal workload (46±18 versus 49±24 W;P=0.59) or peak oxygen consumption (11.4±2.8 versus 12.9±3.4 mL/[kg·min];P=0.15) was similar between groups.CONCLUSIONS:Half of patients who met HFpEF criteria in the supine position did not meet the criteria in the upright position. Patients with a discordant HFpEF phenotype had less structural and hemodynamic abnormalities compared with those with concordant HFpEF. A Supright exercise right heart catheterization approach is feasible and merits further investigation to determine the clinical implications of discordant exercise hemodynamic findings in supine and upright positions.
- Myocardial Posttranscriptional Landscape in Peripartum Cardiomyopathyby Amy Li Bernard Fang Mengbo Li Yen Chin Koay Cassandra Malecki Benjamin Hunter Dylan Harney Cristobal G. dos Remedios Mark Larance John F. O’Sullivan Sean Lal School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW, Australia (A.L., B.F., Y.C.K., C.M., B.H., D.H., C.G.d.R., M. Larance, J.F.O., S.L.). Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia (A.L.). Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW (A.L.). Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia (M. Li). The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, NSW, Australia (C.M, J.F.O, S.L). on November 8, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011725, December 1, 2024. <br/>BACKGROUND:Pregnancy imposes significant cardiovascular adaptations, including progressive increases in plasma volume and cardiac output. For most women, this physiological adaptation resolves at the end of pregnancy, but some women develop pathological dilatation and ultimately heart failure late in pregnancy or in the postpartum period, manifesting as peripartum cardiomyopathy (PPCM). Despite the mortality risk of this form of heart failure, the molecular mechanisms underlying PPCM have not been extensively examined in human hearts.METHODs:Protein and metabolite profiles from left ventricular tissue of end-stage PPCM patients (N=6–7) were compared with dilated cardiomyopathy (DCM; N=5–6) and nonfailing donors (N=7–18) using unbiased quantitative mass spectrometry. All samples were derived from the Sydney Heart Bank. Data are available via ProteomeXchange with identifier PXD055986. Differential protein expression and metabolite abundance and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed.RESULTS:Proteomic analysis identified 2 proteins, SBSPON (somatomedin B and thrombospondin type 1 domain-containing protein precursor) and TNS3 (tensin 3), that were uniquely downregulated in PPCM. SBSPON, an extracellular matrix protein, and TNS3, involved in actin remodeling and cell signaling, may contribute to impaired tissue remodeling and fibrosis in PPCM. Metabolomic analysis revealed elevated levels of homogentisate and deoxycholate and reduced levels of lactate and alanine in PPCM, indicating disrupted metabolic pathways and glucose utilization. Both PPCM and DCM shared pathways related to inflammation, immune responses, and signal transduction. However, thyroid hormone signaling was notably reduced in PPCM, affecting contractility and calcium handling through altered expression of PLN (phospholamban) and Sarcoendoplasmic Reticulum Calcium ATPase (SERCA). Enhanced endoplasmic reticulum stress and altered endocytosis pathways in PPCM suggested additional mechanisms of energy metabolism disruption.CONCLUSIONS:The present study reveals unique posttranslational molecular features of the PPCM myocardium, which mediates cellular and metabolic remodeling, and holds promise as potential targets for therapeutic intervention.
- NETosis is an Important Component of Chronic Myocardial Inflammation in Patients With Heart Failureby Sawa Kostin Manfred Richter Florian Krizanic Benjamin Sasko Theodoros Kelesidis Nikolaos Pagonas Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany (S.K., N.P.). Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany (M.R.). Department of Internal Medicine and Cardiology, University Hospital Ruppin-Brandenburg, Medical School Theodor Fontane, Neuruppin, Germany (F.K., N.P.). Medical Department II, Marien Hospital Herne, Ruhr University Bochum, Germany (B.S.). University of Texas Southwestern Medical Center, Dallas (T.K.). on November 8, 2024
Circulation: Heart Failure, Ahead of Print. <br/>
- Inhibitors of the Interleukin-1 Receptor Accessory Protein Signaling: Another Asset in the Cardio-Immunology Toolboxby Stefano Toldo Guglielmo Gallone Antonio Abbate Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville (S.T., A.A.). Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G.). Department of Medical Sciences, University of Turin, Italy (G.G.). on November 8, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012244, December 1, 2024. <br/>
- Renal Effects of Combination Phosphodiesterase V Inhibition and Low-Dose B-Type Natriuretic Peptide in Acute Heart Failure: A Randomized Clinical Trialby Scott A. Hubers Sherry L. Benike Bradley K. Johnson Paul M. McKie Christopher Scott Horng H. Chen Department of Cardiovascular Medicine (S.A.H., P.M.M.K., H.H.C.), Mayo Clinic, Rochester, MN. Cardiorenal Research Laboratory (S.L.B., P.M.M., H.H.C.), Mayo Clinic, Rochester, MN. Quantitative Health Sciences (B.K.J., C.S.), Mayo Clinic, Rochester, MN. Minneapolis Heart Institute, United Hospital, St. Paul (S.A.H.). on November 8, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011761, December 1, 2024. <br/>BACKGROUND:Cardiorenal dysfunction with impaired cyclic GMP (cGMP) response is common in patients presenting with acute heart failure (HF). Type V phosphodiesterase (PDEV) is known to be upregulated in HF and may explain the dysfunction of renal response. The aim of this study was to determine whether B-type natriuretic peptide (BNP) alone or in combination with PDEV inhibition improves renal function and increases urinary sodium and cGMP excretion in acute HF.METHODS:This open-label study included 67 patients hospitalized with acute HF and renal dysfunction. Patients were randomized to standard care, low-dose intravenous BNP (0.005 µg/kg per minute), or combination BNP/PDEV inhibition with sildenafil (25 mg q12 hours) for 48 hours. The coprimary end points were the percent change in estimated glomerular filtration rate and blood urea nitrogen from baseline to 48 hours.RESULTS:Treatment with BNP and BNP/PDEV inhibitor significantly increased plasma cGMP at 24 hours (+25.6% [+9.8%, +84.7%] and +60.8% [+32.3%, +103.8%] for BNP and BNP/PDEV versus −13.5% [−29.1%, +14.2%] with standard care;P=0.001). However, there was no significant change in estimated glomerular filtration rate 0 (−10.8%, +12.7%) for standard care versus 0 (−15.3%, +11.8%) for the BNP group versus –8.8% (−14.3%, +8.3%) for the BNP/PDEV group (P=0.60) or blood urea nitrogen −1.4% (−10.7%, +12.0%) for standard care versus –5.9% (−14.6%, +9.4%) for the BNP group versus +6.9% (−5.3%, +18.8%) for the BNP/PDEV group (P=0.38) between groups. Hypotension was more common in the BNP/PDEV inhibitor group.CONCLUSIONS:BNP and combination BNP/PDEV inhibition increased plasma cGMP in patients with acute HF but did not improve renal function or urinary sodium/cGMP excretion. Our study does not support the use of intravenous low-dose BNP with or without PDEV inhibition to enhance renal function in patients admitted with acute HF.REGISTRATION:URL:https://www.clinicaltrials.gov; Unique identifier: NCT00972569.
- IL1RAP Blockade With a Monoclonal Antibody Reduces Cardiac Inflammation and Preserves Heart Function in Viral and Autoimmune Myocarditisby Diego A. Lema Gabriel Jakobsson Abdel Daoud David Elias Monica V. Talor Sara Rattik Caitríona Grönberg Hannah Kalinoski Elin Jaensson Gyllenbäck Nadan Wang David Liberg Alexandru Schiopu Daniela Čiháková Department of Pathology, School of Medicine (D.A.L., D.E., M.V.T., D.Č.), Johns Hopkins University, Baltimore, MD. W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health (A.D., H.K., D.Č.), Johns Hopkins University, Baltimore, MD. Department of Cardiology, School of Medicine (N.W.), Johns Hopkins University, Baltimore, MD. Department of Translational Medicine, Lund University, Malmö, Sweden (G.J., A.S.). Cantargia AB, Lund, Sweden (S.R., C.G., E.J.G., D.L.). Internal Medicine Clinic, Skåne University Hospital, Lund, Sweden (A.S.). “Nicolae Simionescu” Institute of Cellular Biology and Pathology, Bucharest, Romania (A.S.). on November 8, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011729, December 1, 2024. BACKGROUND:Currently, there are no therapies targeting specific pathogenic pathways in myocarditis. IL (interleukin)-1 blockade has shown promise in preclinical studies and case reports. We hypothesized that blockade of IL1RAP (IL-1 receptor accessory protein), a shared subunit of the IL-1, IL-33, and IL-36 receptors, could be more efficient than IL-1 blockade alone.METHODS:We induced coxsackievirus B3 (CVB3)–mediated or experimental autoimmune myocarditis (EAM) in BALB/c mice, followed by treatment with an Fc (fragment crystallizable)-modified mIgG2a mouse anti-mouse IL1RAP monoclonal antibody (mCAN10). Myocarditis severity and immune infiltration were assessed by histology and flow cytometry. Cardiac function was measured by echocardiography. We used spatial transcriptomics (Visium 10× Genomics) to compare the gene expression landscape in the hearts of mCAN10-treated versus control mice.RESULTS:IL1RAP blockade reduced CVB3 and EAM severity. In EAM, the treatment prevented deterioration of cardiac function, measured on day 42 post-disease induction (left ventricular ejection fraction: 56.5% versus 51.0% in isotype controls [P=0.002] and versus 51.4% in mice treated with anti-IL-1β antibodies alone [P=0.003]; n=10–11 mice per group). In the CVB3 model, mCAN10 did not impede viral clearance from the heart and significantly lowered the numbers of CD4+(cluster of differentiation 4) T cells (P=0.025), inflammatory Ly6C+CCR2+(lymphocyte antigen 6 complex, locus C/C-C motif chemokine receptor 2) monocytes (P=0.038), neutrophils (P=0.001) and eosinophils (P<0.001) infiltrating the myocardium. The spatial transcriptomic analysis revealed reduced canonical IL-1 signaling and chemokine expression in cardiac immune foci in CVB3-infected mice treated with IL1RAP blockade.CONCLUSIONS:Blocking IL1RAP reduces acute CVB3 myocarditis and EAM severity and preserves cardiac function in EAM. We conclude that IL1RAP blockade is a potential therapeutic strategy in viral and autoimmune myocarditis.
- Single Meandering Right Pulmonary Vein With Potential Systemic Arterial Fistula: Serial Cardiac MRI Assessment With 4D Flow Characterizationby Michael DiMaria Adam Dorfman Sowmya Balasubramanian Jimmy Lu Prachi Agarwal Swati Mody Aparna Joshi Anil Attili Department of Pediatrics, University of Michigan, C.S. Mott Children’s Hospital, Ann Arbor. (M.D., A.D., S.B., J.L.) Department of Radiology, University of Michigan, C.S. Mott Children’s Hospital, Ann Arbor. (P.A., S.M., A.J., A.A.) on November 7, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- m6A Modification of Profilin-1 in Vascular Smooth Muscle Cells Drives Phenotype Switching and Neointimal Hyperplasia via Activation of the p-ANXA2/STAT3 Pathwayby Xiao-Fei Gao Ai-Qun Chen Hao-Yue Tang Xiang-Quan Kong Huan Zhang Zhi-Mei Wang Wei Lu Li-Guo Wang Feng Wang Wen-Ying Zhou Yue Gu Guang-Feng Zuo Zhen Ge Jun-Jie Zhang Shao-Liang Chen Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China. on November 7, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2543-2559, December 1, 2024. <br/>BACKGROUND:In-stent restenosis is characterized by a significant reduction in lumen diameter within the stented segment, primarily attributed to excessive proliferation of vascular smooth muscle cells (VSMCs) and neointimal hyperplasia. PFN1 (profilin-1), an actin-sequestering protein extensively studied in amyotrophic lateral sclerosis, remains less explored in neointimal hyperplasia.METHODS:Utilizing single-cell RNA sequencing alongside data from in-stent restenosis patients and various experimental in-stent restenosis models (swine, rats, and mice), we investigated the role of PFN1 in promoting VSMC phenotype switching and neointimal hyperplasia.RESULTS:Single-cell RNA sequencing of stenotic rat carotid arteries revealed a critical role for PFN1 in neointimal hyperplasia, a finding corroborated in stented swine coronary arteries, in-stent restenosis patients, PFN1SMC-IKO(SMC-specific PFN1 knockout) mice, and PFN1 overexpressed mice. PFN1 deletion was shown to suppress VSMC phenotype switching and neointimal hyperplasia in PFN1SMC-IKOmice subjected to a wire-injured model. To elucidate the observed discordance in PFN1 mRNA and protein levels, we identified that METTL3 (N6-methyladenosine methyltransferase) and YTHDF3 (YTH N6-methyladenosine RNA binding protein F3; N6-methyladenosine–specific reader) enhance PFN1 translation efficiency in an N6-methyladenosine–dependent manner, confirmed through experiments involving METTL3 knockout and YTHDF3 knockout mice. Furthermore, PFN1 was mechanistically found to interact with the phosphorylation of ANXA2 (annexin A2) by recruiting Src (SRC proto-oncogene, nonreceptor tyrosine kinase), promoting the phosphorylation of STAT3 (signal transducer and activator of transcription 3), a typical transcription factor known to induce VSMC phenotype switching.CONCLUSIONS:This study unveils the significance of PFN1 N6-methyladenosine modification in VSMCs, demonstrating its role in promoting phenotype switching and neointimal hyperplasia through the activation of the p-ANXA2 (phospho-ANXA2)/STAT3 pathway.
- ApoE Receptor-2 R952Q Variant in Macrophages Elevates Soluble LRP1 to Potentiate Hyperlipidemia and Accelerate Atherosclerosis in Miceby Vanessa Turkson April Haller Anja Jaeschke David Y. Hui Department of Pharmacology and Systems Physiology (V.T.) Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.) on November 7, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:apoER2 (apolipoprotein E receptor-2) is a transmembrane receptor in the low-density lipoprotein receptor (LDLR) family with unique tissue expression. A single-nucleotide polymorphism that encodes the R952Q sequence variant has been associated with elevated plasma cholesterol levels and increased myocardial infarction risk in humans. The objective of this study was to delineate the mechanism underlying the association between the apoER2 R952Q variant and increased atherosclerosis risk.METHODS:An apoER2 R952Q mouse model was generated using a CRISPR/Cas9 strategy, intercrossed with LDLR knockout mice, followed by feeding a Western-type high-fat high-cholesterol diet for 16 weeks. Atherosclerosis was investigated by immunohistology. Plasma lipids and lipid distributions among the various lipoprotein classes were analyzed by colorimetric assay. Tissue-specific effects of the R952Q sequence variant on atherosclerosis were analyzed by bone marrow transplant studies. sLRP1 (soluble low-density lipoprotein receptor-related protein 1) was measured in plasma and conditioned media from bone marrow–derived macrophages by ELISA and GST-RAP (glutathione S-transferase-receptor-associated protein) pull-down, respectively. P38 MAPK (mitogen-activated protein kinase) phosphorylation in VLDL (very-low-density lipoprotein)–treated macrophages was determined by Western blot analysis.RESULTS:Consistent with observations in humans with this sequence variant, the apoER2 R952Q mutation exacerbated diet-induced hypercholesterolemia, via impediment of plasma triglyceride-rich lipoprotein clearance, to accelerate atherosclerosis in Western diet–fed LDLR knockout mice. Reciprocal bone marrow transplant experiments revealed that the apoER2 R952Q mutation in bone marrow–derived cells instead of non-bone marrow–derived cells was responsible for the increase in hypercholesterolemia and atherosclerosis. Additional data showed that the apoER2 R952Q mutation in macrophages promotes VLDL-induced LRP1 (low-density lipoprotein receptor-related protein 1) shedding in a p38 MAPK–dependent manner.CONCLUSIONS:The apoER2 R952Q mouse model recapitulates characteristics observed in human disease. The underlying mechanism is that the apoER2 R952Q mutation in macrophages exacerbates VLDL-stimulated sLRP1 production in a p38 MAPK–dependent manner, resulting in its competition with cell surface LRP1 to impede triglyceride-rich lipoprotein clearance, thereby resulting in increased hypercholesterolemia and accelerated atherosclerosis.
- Trem2/Tyrobp Signaling Protects Against Aortic Dissection and Rupture by Inhibiting Macrophage Activation in Miceby Zenghui Zhang Maoxiong Wu Lei Yao Weibin Zhou Xiao Liu Zhiteng Chen Ping Hua Leibo Xu Lei Lv Chiyu Liu Chunling Huang Sixu Chen Zhaoqi Huang Yuna Huang Jiaqi He Tingfeng Chen Jingfeng Wang Woliang Yuan Zhaoyu Liu Yangxin Chen Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.) Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.) Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.) Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.) Department of Cardio-Vascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. (P.H.) Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. (L.X.) Department of Cardiology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China (Z.Z.). Department of Cardiac and Vascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Yunnan, China (L.L.). on November 7, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:The development of aortic dissection (AD) is closely associated with inflammation. The Trem2 (triggering receptor expressed on myeloid cells 2)/Tyrobp (TYRO protein tyrosine kinase-binding protein) signaling pathway critically regulates innate immunity and has emerged as an important target in cardiovascular diseases; however, its role in AD remains unclear.METHODS:Transcriptome data from human and mouse ADs were used to perform differentially expressed gene–based protein-protein interaction network analyses.Tyrobpknockout (Tyrobp−/−), myeloid cell–specificTyrobp−/−(Tyrobpfl/flLyz2cre), andTrem2knockout (Trem2−/−) mice were given β-aminopropionitrile monofumarate in drinking water to induce AD. To dissect the role of macrophages inTyrobpdeficiency–mediated AD progression, macrophages were depleted using clodronate liposomes. Bulk and single-cell RNA sequencing, immunofluorescence staining, and quantitative real-time polymerase chain reaction were performed to assess inflammation and the underlying mechanisms of Tyrobp in AD.RESULTS:Network analysis identifiedTyrobpas a hub gene of AD, with elevated levels observed in both human and mouse ADs. Global deletion and myeloid cell–specific deficiency ofTyrobpin mice significantly increased AD incidence and exacerbated extracellular matrix degradation and macrophage infiltration within the aortic wall. Macrophage depletion mitigated the adverse effects ofTyrobpdeficiency on AD progression. Additionally,Tyrobpdeficiency enhanced TLR (Toll-like receptor)-4 signaling and macrophage activation, which were abrogated by TLR4 inhibitors. Furthermore, deletion of the Tyrobp-associated receptorTrem2significantly aggravated mouse AD development, whereas Trem2 agonist treatment conferred protection against AD.CONCLUSIONS:Our findings suggest a novel role for the Trem2/Tyrobp axis in AD development in mice. Enhancement of Trem2/Tyrobp signaling may represent a promising strategy for the prevention and treatment of AD. Future studies to clarify the role of Trem2/Tyrobp in human AD are warranted.
- Cure of Congenital Purpura Fulminans via Expression of Engineered Protein C Through Neonatal Genome Editing in Miceby Tomoki Togashi Nemekhbayar Baatartsogt Yasumitsu Nagao Yuji Kashiwakura Morisada Hayakawa Takafumi Hiramoto Takayuki Fujiwara Eriko Morishita Osamu Nureki Tsukasa Ohmori Department of Clinical Laboratory Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan (T.T., E.M.). Department of Biochemistry, Jichi Medical University School of Medicine, Tochigi, Japan (T.T., N.B., Y.K., M.H., T.H., T.O.). Center for Experimental Medicine (Y.N.), Jichi Medical University, Tochigi, Japan. Center for Gene Therapy Research (Y.K., M.H., T.O.), Jichi Medical University, Tochigi, Japan. Division of Cell and Molecular Medicine Center for Molecular Medicine (T.F.), Jichi Medical University, Tochigi, Japan. Department of Cardiovascular Medicine, The University of Tokyo Hospital, Japan (T.F.). Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Japan (O.N.). on November 7, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2616-2627, December 1, 2024. <br/>BACKGROUND:PC (protein C) is a plasma anticoagulant encoded byPROC; mutation in bothPROCalleles results in neonatal purpura fulminans—a fatal systemic thrombotic disorder. In the present study, we aimed to develop a genome editing treatment to cure congenital PC deficiency.METHODS:We generated an engineered APC (activated PC) to insert a furin-cleaving peptide sequence between light and heavy chains. The engineered PC was expressed in the liver of mice using an adeno-associated virus vector or CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat–associated 9)-mediated genome editing using an adeno-associated virus vector in vivo.RESULTS:The engineered PC could be released in its activated form and significantly prolonged the plasma coagulation time independent of the cofactor activity of PS (protein S) in vitro. The adeno-associated virus vector-mediated expression of the engineered PC, but not wild-type PC, prolonged coagulation time owing to the inhibition of activated coagulation FV (factor V) in a dose-dependent manner and abolished pathological thrombus formation in vivo in C57BL/6J mice. The insertion ofEGFP(enhanced green fluorescent protein) sequence conjugated with self-cleaving peptide sequence atAlblocus via neonatal in vivo genome editing using adeno-associated virus vector resulted in the expression of EGFP in 7% of liver cells, mainly via homology-directed repair, in mice. Finally, we succeeded in improving the survival of PC-deficient mice by expressing the engineered PC via neonatal genome editing in vivo.CONCLUSIONS:These results suggest that the expression of engineered PC via neonatal genome editing is a potential cure for severe congenital PC deficiency.
- Socioeconomic Disparities Are Associated With Delayed Access to Tafamidis in Transthyretin Cardiac Amyloidosisby Peter Miller Pierre Elias Andrew J. Einstein Mathew S. Maurer Gasmelseed Y. Ahmed Timothy J. Poterucha Department of Medicine (P.M., P.E., A.J.E., M.S.M., T.J.P.), Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York. Seymour, Paul, and Gloria Milstein Division of Cardiology, Department of Medicine (P.E., A.J.E., M.S.M., G.Y.A., T.J.P.), Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York. Department of Biomedical Informatics (P.E.), Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York. Department of Radiology (A.J.E.), Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York. on November 7, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012075, December 1, 2024. <br/>
- Rhythmic Contractions of Lymph Vessels and Lymph Flow Are Disrupted in Hypertensive Ratsby Soumiya Pal Ashim K. Bagchi David S. Henry Reid D. Landes Shengyu Mu Sung W. Rhee Nancy J. Rusch Amanda J. Stolarz Department of Pharmaceutical Sciences, College of Pharmacy (S.P., A.K.B., A.J.S.), University of Arkansas for Medical Sciences, Little Rock, AR. Department of Pharmacology and Toxicology, College of Medicine (D.S.H., S.M., S.W.R., N.J.R., A.J.S.), University of Arkansas for Medical Sciences, Little Rock, AR. Department of Biostatistics, College of Medicine (R.D.L.), University of Arkansas for Medical Sciences, Little Rock, AR. Now with Department of Biomedical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (S.W.R.). on November 6, 2024
Hypertension, Volume 82, Issue 1, Page 72-83, January 1, 2025. <br/>BACKGROUND:Hypertension increases the risk of lymphedema in patients with comorbidities, but whether hypertension directly compromises lymph vessel (LV) function and lymph flow is unclear. We compared the contractions of mesenteric LVs ex vivo and lymph flow in vivo between normotensive and Ang II (angiotensin II)-induced hypertensive rats and explored the ionic basis of contractile patterns. Key studies were recapitulated in spontaneously hypertensive rats and control Wistar-Kyoto rats.METHODS:Video microscopy continuously recorded the diameters of cannulated rat mesenteric LVs, and high-speed optical imaging estimated mesenteric lymph flow in vivo. Jess capillary Western electrophoresis evaluated expression levels of ion channel proteins.RESULTS:Isolated LVs from Ang II-induced hypertensive rats exhibited dysrhythmic contractions, whereas LVs from both Ang II-induced hypertensive rats and spontaneously hypertensive rats exhibited reduced diastolic diameters and cross-sectional flow. Mesenteric lymph flow in vivo was 2.9-fold lower in Ang II-induced hypertensive rats compared with normotensive rats. Surprisingly, the LVs from Ang II-induced hypertensive rats expressed fewer intact L-type Ca2+channel pore proteins and more modulatory cleaved C-terminal fragments. However, pharmacological block of voltage-gated K+channels but not other K+channel types in control LVs established the pattern of contractile dysfunction observed in hypertension. Jess capillary Western electrophoresis analysis confirmed a loss of Shaker-type KV1.2 channels in LVs from hypertensive rats.CONCLUSIONS:We provide initial evidence of lymphatic contractile dysfunction and compromised lymph flow in hypertensive rats, which may be caused by a loss of KV1.2 channels in the lymphatic muscle cells.
- Evolutionary Characteristics in Primary Aldosteronism Patientsby Yinjie Gao Yu Wang Yue Zhou Xiaoyan Chang Yushi Zhang Min Nie Anli Tong National Health Commission Key Laboratory of Endocrinology, Department of Endocrinology (Y.G., Y.W., Y. Zhou, M.N., A.T.), Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China. Department of Pathology (X.C.), Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China. Department of Urology (Y. Zhang), Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China. Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, China (Y.G.). on November 6, 2024
Hypertension, Volume 82, Issue 1, Page 96-105, January 1, 2025. <br/>BACKGROUND:Primary aldosteronism is predominantly caused by excessive aldosterone production from the adrenal cortex, and the aldosterone-producing structures could take many forms, like adenomas, nodules, micronodules, and so on. Most studies of primary aldosteronism were limited to the hotspot driver genes responsible for autonomous aldosterone production; however, the panoramic genetic architecture and genomic alterations of aldosterone-producing structures and their adjacent hyperplasia glands remain unknown.METHODS:In this study, whole-exome sequencing and transcriptome sequencing (RNA-seq) analyses were performed using functional nodules and matched hyperplasia tissues, which were microdissected guided by aldosterone synthase immunohistochemistry. Phylogenetic trees were constructed based on the shared and unique mutations, gene mutation spectrums, and clonal characteristics.RESULTS:The rates of mutations represented higher means of functional nodules than hyperplasia samples, and the little mutational overlap was shown between the 2 groups on phylogenetic trees. The mutations of the aldosterone driver gene (KCNJ5orCACNA1D) were only observed in functional nodules and indicated almost the largest values of cancer cell fraction. Moreover, the functional nodules also harbored some potential variants related to cell proliferation, which were not detected in hyperplasia tissues. Transcriptome analysis suggested that only 25.5% upregulated and 23.3% downregulated genes overlapped between functional nodules and hyperplasia tissues.CONCLUSIONS:This study demonstrated a genetic and transcriptome landscape of aldosterone-producing structures and adjacent hyperplasia glands in primary aldosteronism. The results indicated independent clonal origins on functional nodules and hyperplasia tissues, and little mutual evolutionary relationship was found on their phylogenetic trees.
- Acute Heart Failure Caused by Rupture of Sinus of Valsalva Into Right Atrium in a Patient With Possible Infective Endocarditis After Tricuspid Annuloplasty: A Misdirected Clinical Decision-Makingby Daiki Toyoshima Yasuhide Mochizuki Sunao Handa Daisuke Yokokawa Saori Chino Rumi Hachiya Hiroto Fukuoka Toshiro Shinke Division of Cardiology, Department of Internal Medicine, Showa University School of Medicine, Tokyo, Japan. on November 5, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011600, December 1, 2024. <br/>
- Protein Biomarkers of Adverse Clinical Features and Events in Sarcomeric Hypertrophic Cardiomyopathyby Usman A. Tahir Paul Kolm Raymond Y. Kwong Milind Y. Desai Sarahfaye F. Dolman Shuliang Deng Evan Appelbaum Patrice Desvigne-Nickens John P. DiMarco Gaurav Tiwari Matthias G. Friedrich Julissa H. Zelaya-Portillo Michael Jerosch-Herold Dong-Yun Kim Martin S. Maron Stefan K. Piechnik Jeanette Schulz-Menger Hugh Watkins William S. Weintraub Stefan Neubauer Christopher M. Kramer Petr Jarolim Robert E. Gerszten Carolyn Y. Ho Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (U.A.T., S.D., E.A., G.T., R.E.G.). Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (P.K., S.K.P., H.W., S.N.). Departments of Medicine, Radiology, and Pathology, Brigham and Women’s Hospital, Boston, MA (R.Y.K., M.J.-H., P.J., C.Y.H.). Heart, Vascular and Thoracic Institute, Cleveland Clinic, OH (M.Y.D). MedStar Heart and Vascular Institute, Washington, DC (S.F.D., J.H.Z.-P., W.S.W.). National Heart, Lung, and Blood Institute, Bethesda, MD (P.D.-N., D.-Y.K.). Cardiovascular Division, University of Virginia Health System, Charlottesville (J.P.D., C.M.K.). McGill University, Montreal, QC, Canada (M.G.F.). Lahey Hospital and Medical Center, Burlington, MA (M.S.M.). Charité Experimental Clinical Research Center and Helios Clinics Berlin-Buch, Germany (J.S.-M.). on November 5, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e011707, December 1, 2024. <br/>BACKGROUND:Hypertrophic cardiomyopathy (HCM) is a heterogeneous condition that can lead to atrial fibrillation, heart failure, and sudden cardiac death in many individuals but mild clinical impact in others. The mechanisms underlying this phenotypic heterogeneity are not well defined. The aim of this study was to use plasma proteomic profiling to help illuminate biomarkers that reflect or inform the heterogeneity observed in HCM.METHODS:The Olink antibody–based proteomic platform was used to measure plasma proteins in patients with genotype positive (sarcomeric) HCM participating in the HCM Registry. We assessed associations between plasma protein levels with clinical features, cardiac magnetic resonance imaging metrics, and the development of atrial fibrillation.RESULTS:We measured 275 proteins in 701 patients with sarcomeric HCM. There were associations between late gadolinium enhancement with proteins reflecting neurohormonal activation (NT-proBNP [N-terminal pro-B-type natriuretic peptide] and ACE2 [angiotensin-converting enzyme 2]). Metrics of left ventricular remodeling had novel associations with proteins involved in vascular development and homeostasis (vascular endothelial growth factor-D and TM [thrombomodulin]). Assessing clinical features, the European Society of Cardiology sudden cardiac death risk score was inversely associated with SCF (stem cell factor). Incident atrial fibrillation was associated with mediators of inflammation and fibrosis (MMP2 [matrix metalloproteinase 2] and SPON1 [spondin 1]).CONCLUSIONS:Proteomic profiling of sarcomeric HCM identified biomarkers associated with adverse imaging and clinical phenotypes. These circulating proteins are part of both established pathways, including neurohormonal activation and fibrosis, and less familiar pathways, including endothelial function and inflammatory proteins less well characterized in HCM. These findings highlight the value of plasma profiling to identify biomarkers of risk and to gain further insights into the pathophysiology of HCM.
- Adding a New Piece to the ASGR1 Puzzle: ANGPTL3by Itsaso Garcia-Arcos SUNY Downstate Health Sciences University, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Department of Cell Biology, Brooklyn, NY. on October 31, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2450-2452, December 1, 2024. <br/>
- Gut Microbiome Diversity and Composition Correlates With Time in the Therapeutic Range in Patients on Warfarin Treatment: A Pilot Study
- Whys and Wherefores of Coronary Arterial Positive Remodelingby Carlotta Onnis Renu Virmani Anna Madra Valentina Nardi Rodrigo Salgado Roberta Montisci Riccardo Cau Alberto Boi Amir Lerman Carlo N. De Cecco Peter Libby Luca Saba Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.). Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.). Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.). Department of Radiology, Antwerp University Hospital and Antwerp University Lier, Belgium (R.S.). Clinical Cardiology, Department of Medical Science and Public Health, University of Cagliari, Italy (R.M.). Department of Cardiology, Azienda Ospedaliera Brotzu, Cagliari, Italy (A.B.). Division of Cardiothoracic Imaging and Biomedical Informatics, Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA (C.N.D.C.). Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (P.L.). on October 31, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2416-2427, December 1, 2024. <br/>Positive remodeling (PR) is an atherosclerotic plaque feature defined as an increase in arterial caliber at the level of an atheroma, in response to increasing plaque burden. The mechanisms that lead to its formation are incompletely understood, but its role in coronary atherosclerosis has major clinical implications. Indeed, plaques with PR have elevated risk of provoking acute cardiac events. Hence, PR figures among the high-risk plaque features that cardiac imaging studies should report. This review aims to provide an overview of the current literature on coronary PR. It outlines the pathophysiology of PR, the different techniques used to assess its presence, and the imaging findings associated to PR, on both noninvasive and invasive studies. This review also summarizes clinical observations, trials, and studies, focused on the impact of PR on clinical outcome.
- PARP15 Is a Susceptibility Locus for Clarkson Disease (Monoclonal Gammopathy–Associated Systemic Capillary Leak Syndrome)by Eunice C. Chan Ararat J. Ablooglu Chandra C. Ghosh Abhishek Desai Niccole Schaible Xiuying Chen Ming Zhao M. Renee Olano Sundar Ganesan Justin B. Lack Ramaswamy Krishnan Samir M. Parikh Kirk M. Druey Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases (E.C.C., A.J.A., A.D., K.M.D.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD. Protein Chemistry Section (M.Z., M.R.O.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD. Biological Imaging Section (S.G.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD. NIAID Collaborative Bioinformatics Resource (J.B.L.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD. Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA (C.C.G., N.S., X.C., R.K.). Division of Nephrology, Departments of Internal Medicine and Pharmacology, University of Texas Southwestern Medical Center, Dallas (S.M.P.). on October 31, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2628-2646, December 1, 2024. <br/>BACKGROUND:Vascular leakage is a deadly complication of severe infections, ranging from bacterial sepsis to malaria. Worldwide, septicemia is among the top 10 causes of lethality because of the shock and multiorgan dysfunction that arise from the host vascular response. In the monoclonal gammopathy–associated capillary leak syndrome (MG-CLS), even otherwise mundane infections induce recurrent septic-like episodes of profound microvascular hyperpermeability and shock. There are no defined genetic risk factors for MG-CLS or effective treatments for acute crises.METHODS:We characterized predicted loss-of-function mutations in PARP15 (poly[ADP-ribose] polymerase 15), a protein of unknown function that is absent in mice, in patients with MG-CLS. We analyzed barrier function in PARP15-deficient vascular endothelial cells and vascular leakage in mice engineered to express wild-type or loss-of-function variant human PARP15.RESULTS:We discovered several loss-of-function PARP15 variants associated with MG-CLS. These mutations severely reduced PARP15 enzymatic function. The presence of the most frequently detected variant (G628R) correlated with clinical markers of severe vascular leakage. In human microvascular endothelial cells, PARP15 suppressed cytokine-induced barrier disruption by ADP-ribosylating the scaffold protein JIP3 (c-Jun N-terminal kinase-interacting protein 3) and inhibiting p38 MAP (mitogen-activated protein) kinase activation. Mice expressing enzymatically inactive human PARP15(G628R) were significantly more prone to inflammation-associated vascular leakage than mice expressing wild-type PARP15 in a p38-dependent fashion.CONCLUSIONS:PARP15represents a previously unrecognized genetic susceptibility factor for MG-CLS. PARP15-mediated ADP ribosylation is an essential and genetically determined mechanism of the human vascular response to inflammation.
- Immune-Mediated Inflammatory Diseases, Dyslipidemia, and Cardiovascular Risk: A Complex Interplayby Michael J. Wilkinson Michael D. Shapiro Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla (M.J.W.). Section on Cardiovascular Medicine, Center for Prevention of Cardiovascular Disease, Wake Forest University School of Medicine, Winston-Salem, NC (M.D.S.). on October 31, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2396-2406, December 1, 2024. <br/>Individuals with autoimmune inflammatory diseases, such as systemic lupus erythematosus, rheumatoid arthritis, and psoriasis, are at increased risk for cardiovascular disease. While these diseases share common features of systemic inflammation, the impact of individual autoimmune inflammatory conditions on circulating lipids and lipoproteins varies by specific disease, disease activity, and the immune-suppressing medications used to treat these conditions. A common feature observed in many autoimmune inflammatory diseases is the development of a proatherogenic dyslipidemic state, characterized by dysfunctional HDLs (high-density lipoproteins) and increased oxidation of LDLs (low-density lipoproteins). Various disease-modifying antirheumatic drugs also have complex and variable effects on lipids, and it is critical to take this into consideration when evaluating lipid-related risk in individuals with immune-mediated inflammatory conditions. This review aims to critically evaluate the current understanding of the relationship between immune-mediated inflammatory diseases and dyslipidemia, the underlying mechanisms contributing to atherogenesis, and the impact of various pharmacotherapies on lipid profiles and cardiovascular risk. We also discuss the role of lipid-lowering therapies, particularly statins, in managing residual risk in this high-risk population and explore the potential of emerging therapies with complementary anti-inflammatory and lipid-lowering effects.
- Effect of Acoramidis on Myocardial Structure and Function in Transthyretin Amyloid Cardiomyopathy: Insights From the ATTRibute-CM Cardiac Magnetic Resonance (CMR) Substudyby Yousuf Razvi Daniel P. Judge Ana Martinez-Naharro Adam Ioannou Lucia Venneri Rishi Patel Julian D. Gillmore Peter Kellman Laura Edwards Jorg Taubel Jing Du Jean-François Tamby Adam Castaño Suresh Siddhanti Leonid Katz Jonathan C. Fox Marianna Fontana Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Hospital, United Kingdom (Y.R., A.M.-N., A.I., L.V., R.P., J.D.G., M.F.). Medical University of South Carolina, Charleston (D.P.J.). National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (P.K.). Richmond Pharmacology, London, United Kingdom (L.E., J.T.). BridgeBio Pharma Inc, San Francisco, CA (J.D., J.-F.T., A.C., S.S., L.K., J.C.F.). on October 28, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012135, December 1, 2024. <br/>
- Pitfalls in the World of Evidence-Based Medicine: Should IABP Be en-DANGER-ed by the DanGer Shock Trial?by Arvind Bhimaraj Arthur R. Garan Manreet K. Kanwar Department of Cardiology, J.C. Walter Jr Transplant Center & DeBakey Heart and Vascular Center, Houston Methodist Hospital, TX (A.B.). Department of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (A.R.G.). Department of Cardiology, Cardiovascular Institute at Allegheny Health Network, Pittsburgh, PA (M.K.K.). on October 25, 2024
Circulation: Heart Failure, Volume 17, Issue 12, Page e012077, December 1, 2024. <br/>
- Single-Cell Meta-Analysis Uncovers the Pancreatic Endothelial Cell Transcriptomic Signature and Reveals a Key Role for NKX2-3 in PLVAP Expressionby Safwat T. Khan Neha Ahuja Sonia Taïb Shabana Vohra Ondine Cleaver Sara S. Nunes Institute of Biomedical Engineering (S.T.K., S.S.N.), University of Toronto, ON, Canada. Laboratory of Medicine and Pathobiology (S.S.N.), University of Toronto, ON, Canada. Heart and Stroke/Richard Lewar Centre of Excellence (S.S.N.), University of Toronto, ON, Canada. Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas (N.A., O.C.). Toronto General Hospital Research Institute (S.T.K., S.T., S.V., S.S.N.), University Health Network, ON, Canada. Ajmera Transplant Center (S.S.N.), University Health Network, ON, Canada. on October 24, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2596-2615, December 1, 2024. <br/>BACKGROUND:The pancreatic vasculature displays tissue-specific physiological and functional adaptations that support rapid insulin response by β-cells. However, the digestive enzymes have made it difficult to characterize pancreatic endothelial cells (ECs), resulting in the poor understanding of pancreatic EC specialization.METHODS:Available single-nuclei/single-cell RNA-sequencing data sets were mined to identify pancreatic EC-enriched signature genes and to develop an integrated atlas of human pancreatic ECs. We validated the findings using independent single-nuclei/single-cell RNA-sequencing data, bulk RNA-sequencing data of isolated ECs, spatial transcriptomics data, immunofluorescence, and RNAScope of selected markers. The NK2 homeobox 3 (NKX2-3) TF (transcription factor) was expressed in HUVECs via gene transfection, and the expression of pancreatic EC-enriched signature genes was assessed via RT-qPCR.RESULTS:We defined a pancreatic EC-enriched gene signature conserved across species and developmental stages that included genes involved in ECM (extracellular matrix) composition (COL15A1 and COL4A1), permeability and barrier function (PLVAP, EHD4, CAVIN3, HSPG2, ROBO4, HEG1, and CLEC14A), and key signaling pathways (S1P [sphingosine-1-phosphate], TGF-β [transforming growth factor-β], RHO/RAC GTPase [guanosine triphosphatase], PI3K/AKT [phosphoinositide 3-kinase/protein kinase B], and PDGF [platelet-derived growth factor]). The integrated atlas revealed the vascular hierarchy within the pancreas. We identified and validated a specialized islet capillary subpopulation characterized by genes involved in permeability (PLVAP and EHD4), immune-modulation (FABP5, HLA-C, and B2M), ECM composition (SPARC and SPARCL1), IGF (insulin-like growth factor) signaling (IGFBP7), and membrane transport (SLCO2A1, SLC2A3, and CD320). Importantly, we identified NKX2-3 as a key TF enriched in pancreatic ECs. DNA-binding motif analysis found NKX2-3 motifs in ≈40% of the signature genes. Induction of NKX2-3 in HUVECs promoted the expression of the islet capillary EC-enriched genes PLVAP and SPARCL1.CONCLUSIONS:We defined a validated transcriptomic signature of pancreatic ECs and uncovered their intratissue transcriptomic heterogeneity. We showed that NKX2-3 acts upstream of PLVAP and provided a single-cell online resource that can be further explored by the community:https://vasconcelos.shinyapps.io/pancreatic_endothelial/.
- TGF-β: A Wrench in the Gears of Arteriovenous Fistula Maturationby Mandy M.T. van Leent Raphaël Duivenvoorden BioMedical Engineering and Imaging Institute (M.M.T.v.L., R.D.), Icahn School of Medicine at Mount Sinai, New York. Cardiovascular Research Institute (M.M.T.v.L.), Icahn School of Medicine at Mount Sinai, New York. Department of Radiology (M.M.T.v.L), Icahn School of Medicine at Mount Sinai, New York. Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands (R.D.). on October 24, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2527-2529, December 1, 2024. <br/>
- A Sophisticated Model of Human Atherosclerosis on a Chipby Brandon J. Tefft Joint Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee. on October 24, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2473-2475, December 1, 2024. <br/>
- Tuning Into Immune Cell Responses of Chronic Stress With Intravital Microscopyby Matthias Nahrendorf Center for Systems Biology and Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA. Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA. Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA. Department of Internal Medicine I, University Hospital Würzburg, Germany. on October 24, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2507-2508, December 1, 2024. <br/>
- Impaired Renal Function With Higher Remnant Cholesterol Related to Risk of Atherosclerotic Cardiovascular Disease: A Cohort Studyby Daniel Elías-López Signe Vedel-Krogh Camilla Jannie Kobylecki Benjamin Nilsson Wadström Børge Grønne Nordestgaard Department of Clinical Biochemistry (D.E.-L., S.V.-K., C.J.K., B.N.W., B.G.N.), Copenhagen University Hospital, Herlev Gentofte, Denmark. The Copenhagen General Population Study (D.E.-L., S.V.-K., C.J.K., B.N.W., B.G.N.), Copenhagen University Hospital, Herlev Gentofte, Denmark. Department of Endocrinology and Metabolism and Research Center of Metabolic Diseases, National Institute of Medical Sciences and Nutrition Salvador Zubirán, México City, México (D.E.-L.). Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (S.V.-K., B.N.W., B.G.N.). on October 24, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2647-2658, December 1, 2024. BACKGROUND:Chronic kidney disease confers a high risk of atherosclerotic cardiovascular disease (ASCVD), partly due to hyperlipidemia. Although statins reduce the risk of ASCVD in chronic kidney disease, residual risk persists. We investigated whether higher remnant cholesterol is associated with an increased risk of ASCVD in statin users and nonusers with impaired renal function.METHODS:We included 107 925 individuals from CGPS (Copenhagen General Population Study) initiated in 2003 to 2015, of whom 10 427 had impaired renal function (estimated glomerular filtration rate, <60 mL/min per 1.73 m2). Remnant cholesterol was calculated from a standard lipid profile. ASCVD was myocardial infarction, coronary heart disease death, ischemic stroke, coronary artery bypass graft, or percutaneous coronary intervention extracted from Danish nationwide health registries from baseline through 2018; individuals with events before the start of follow-up were excluded from relevant analysis.RESULTS:In individuals with impaired renal function during up to 15 years of follow-up, 597 were diagnosed with myocardial infarction, 618 with ischemic stroke, and 1182 with ASCVD. In these individuals, a 1-mmol/L (39 mg/dL) higher remnant cholesterol level was associated with multivariable-adjusted hazard ratios of 1.22 (95% CI, 1.05–1.42) for myocardial infarction, 1.16 (95% CI, 0.97–1.38) for ischemic stroke, and 1.21 (95% CI, 1.08–1.36) for ASCVD. Corresponding hazard ratios for ASCVD were 1.40 (95% CI, 1.07–1.83) in statin users and 1.16 (95% CI, 1.01–1.34) in nonusers. Of the 1.36-fold excess risk of ASCVD in impaired versus normal renal function, elevated remnant cholesterol and elevated LDL (low-density lipoprotein) cholesterol explained 25% (95% CI, 2.5%–47%) and 0% in statin users and 8.3% (95% CI, 2.4%–14%) and 14% (95% CI, 6.4%–22%) in nonusers, respectively.CONCLUSIONS:Our results suggest that higher remnant cholesterol is a good marker of increased risk of ASCVD in individuals with impaired renal function, while higher LDL cholesterol may not be. Patients with chronic kidney disease who have high levels of remnant cholesterol are identifiable through higher non-HDL (high-density lipoprotein) cholesterol or apoB levels.
- Radiation-Induced Macrovessel/Microvessel Diseaseby Jun-ichi Abe Bryan G. Allen Andreas M. Beyer David Lewandowski Kranti A. Mapuskar Vikram Subramanian Michelle R. Tamplin Isabella M. Grumbach Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (J.-I.A.). Free Radical and Radiation Biology Program, Department of Radiation Oncology (B.G.A., K.A.M., I.M.G.), Carver College of Medicine, University of Iowa. Department of Internal Medicine, Abboud Cardiovascular Research Center (V.S., M.R.T., I.M.G.), Carver College of Medicine, University of Iowa. Department of Pharmacology and Physiology, Cardiovascular Center (A.M.B.), Medical College of Wisconsin, Milwaukee. Division of Cardiology/Cardiovascular Medicine, Department of Medicine (D.L.), Medical College of Wisconsin, Milwaukee. Iowa City VA Healthcare System (I.M.G.). on October 24, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2407-2415, December 1, 2024. <br/>Radiation therapy (RT) is a cornerstone in cancer treatment (used in 50% of cases), yet challenges persist because damage to normal tissue through direct impact of radiation or bystander effects is inevitable. Injury of macrovessels by RT manifests as obstructive disease, which is akin to atherosclerotic disease. Historically observed in coronary arteries of patients treated for breast cancer and lymphoma, it also affects patients receiving contemporary therapy for lung and chest cancers. Moreover, radiation at various sites can lead to peripheral vascular disease. An aspect of radiation-induced injury that has received little attention is microvascular injury, which typically results from damage to the endothelium and is considered the primary driver of RT-induced toxicity in the skin, kidney, and brain. This review delves into the clinical manifestations of RT-induced vascular disease, signaling pathways, cellular targets affected by radiation injury, and preclinical models of RT-induced vascular injury. The goal is to inspire the development of innovative strategies to prevent RT-related cardiovascular disease.
- Senescent Syncytiotrophoblast Secretion During Early Onset Preeclampsiaby Olivia Nonn Olivia Debnath Daniela S. Valdes Katja Sallinger Ali Kerim Secener Cornelius Fischer Sebastian Tiesmeyer Jose Nimo Thomas Kuenzer Juliane Ulrich Theresa Maxian Martin Knöfler Philipp Karau Hendrik Bartolomaeus Thomas Kroneis Alina Frolova Lena Neuper Nadine Haase Alexander Malt Niklas Müller-Bötticher Kristin Kräker Sarah Kedziora Désirée Forstner Roland Eils Ruth Schmidt-Ullrich Sandra Haider Stefan Verlohren Christina Stern Meryam Sugulle Stuart Jones Basky Thilaganathan Tu’uhevaha J. Kaitu’u-Lino Stephen Tong Berthold Huppertz Amin El-Heliebi Anne Cathrine Staff Fabian Coscia Dominik N. Müller Ralf Dechend Martin Gauster Naveed Ishaque Florian Herse Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.). Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.). Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.). German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.). Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria. (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.) Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria. (T. Kuenzer) Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.). Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.). Institute for Medical Systems Biology (BIMSB), Berlin, Germany (A.K.S., C.F.). Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (A.K.S.). Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.). Institute of Molecular Biology and Genetic of NASU, Kyiv, Ukraine (A.F.). Department of Obstetrics and Gynaecology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (S.V.). Department of Obstetrics and Gynaecology, University Hospital Graz, Medical University Graz, Austria (C.S.). Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway (M.S., A.C.S.). Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (M.S., A.C.S.). Clinical Biochemistry, King George’s Hospital, London, United Kingdom (S.J.). Fetal Medicine Unit, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom (B.T.). Department of Obstetrics and Gynaecology (Mercy Hospital for Women), The University of Melbourne, VIC, Australia (T.J.K.-L., S. Tong). Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany (R.D.). on October 23, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:Preeclampsia is a severe hypertensive disorder in pregnancy that causes preterm delivery, maternal and fetal morbidity, mortality, and life-long sequelae. Understanding the pathogenesis of preeclampsia is a critical first step toward protecting mother and child from this syndrome and increased risk of cardiovascular disease later in life. However, effective early predictive tests and therapies for preeclampsia are scarce.METHODS:To identify novel markers and signaling pathways for early onset preeclampsia, we profiled human maternal-fetal interface units (fetal villi and maternal decidua) from early onset preeclampsia and healthy controls using single-nucleus RNA sequencing combined with spatial transcriptomics. The placental syncytiotrophoblast is in direct contact with maternal blood and forms the barrier between fetal and maternal circulation.RESULTS:We identified different transcriptomic states of the endocrine syncytiotrophoblast nuclei with patterns of dysregulation associated with a senescence-associated secretory phenotype and a spatial dysregulation of senescence in the placental trophoblast layer. Elevated senescence markers were validated in placental tissues of clinical multicenter cohorts. Importantly, several secreted senescence-associated secretory phenotype factors were elevated in maternal blood already in the first trimester. We verified the secreted senescence markers, PAI-1 (plasminogen activator inhibitor 1) and activin A, as identified in our single-nucleus RNA sequencing model as predictive markers before clinical preeclampsia diagnosis.CONCLUSIONS:This indicates that increased syncytiotrophoblast senescence appears weeks before clinical manifestation of early onset preeclampsia, suggesting that the dysregulated preeclamptic placenta starts with higher cell maturation resulting in premature and increased senescence-associated secretory phenotype release. These senescence-associated secretory phenotype markers may serve as an additional early diagnostic tool for this syndrome.
- Increased TRPV4 Channel Expression Enhances and Impairs Blood Vessel Function in Hypertensionby Xun Zhang Charlotte Buckley Matthew D. Lee Christine Salaun Margaret MacDonald Calum Wilson John G. McCarron Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom. on October 23, 2024
Hypertension, Volume 82, Issue 1, Page 57-68, January 1, 2025. <br/>BACKGROUND:Endothelial cell TRPV4 (transient receptor potential vanilloid 4) channels provide a control point that is pivotal in regulating blood vessel diameter by mediating the Ca2+-dependent release of endothelial-derived vasoactive factors. In hypertension, TRPV4-mediated control of vascular function is disrupted, but the underlying mechanisms and precise physiological consequences remain controversial.METHODS:Here, using a comprehensive array of methodologies, endothelial TRPV4 channel function was examined in intact mesenteric resistance arteries from normotensive Wistar-Kyoto and spontaneously hypertensive rats.RESULTS:Our results show there is a notable shift in vascular reactivity in hypertension characterized by enhanced endothelium-dependent vasodilation at low levels of TRPV4 channel activation. However, at higher levels of TRPV4 activity, this vasodilatory response is reversed, contributing to the aberrant vascular tone observed in hypertension. The change in response, from dilation to constriction, was accompanied by a shift in intracellular Ca2+signaling modalities arising from TRPV4 activity. Oscillatory TRPV4-evoked IP3(inositol triphosphate)-mediated Ca2+release, which underlies dilation, decreased, while the contraction inducing sustained Ca2+rise, arising from TRPV4-mediated Ca2+influx, increased. Our findings also reveal that while the sensitivity of endothelial cell TRPV4 to activation was unchanged, expression of the channel is upregulated and IP3receptors are downregulated in hypertension.CONCLUSIONS:These data highlight the intricate interplay between endothelial TRPV4 channel expression, intracellular Ca2+signaling dynamics, and vascular reactivity. Moreover, the data support a new unifying hypothesis for the vascular impairment that accompanies hypertension. Specifically, endothelial cell TRPV4 channels play a dual role in modulating blood vessel function in hypertension.
- Nanoparticle-Directed Antioxidant Therapy Can Ameliorate Disease Progression in a Novel, Diet-Inducible Model of Coronary Artery Diseaseby Shi Su Zhifen Chen Qingen Ke Olivier Kocher Monty Krieger Peter M. Kang Cardiovascular Institute (S.S., Z.C., Q.K., P.M.K.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. Department of Pathology and Center for Vascular Biology Research (O.K.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. Department of Biology, Massachusetts Institute of Technology, Cambridge (M.K.). on October 17, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2476-2488, December 1, 2024. <br/>BACKGROUND:Oxidative stress plays a crucial role in the pathogenesis of coronary artery disease. In cardiovascular research using murine models, the generation and maintenance of models with robust coronary arterial atherosclerosis has been challenging.METHODS:We characterized a new mouse model in which the last 3 amino acids of the carboxyl terminus of the HDL (high-density lipoprotein) receptor (SR-B1 [scavenger receptor, class B, type 1]) were deleted in a low-density lipoprotein receptor knockout (LDLR−/−) mouse model (SR-B1ΔCT/LDLR−/−) fed an atherogenic diet. We also tested the therapeutic effects of an oxidative stress-targeted nanoparticle in atherogenic diet–fed SR-B1ΔCT/LDLR−/−mice.RESULTS:The SR-B1ΔCT/LDLR−/−mice fed an atherogenic diet had occlusive coronary artery atherosclerosis, impaired cardiac function, and a dramatically lower survival rate, compared with LDLR−/−mice fed the same diet. As SR-B1ΔCT/LDLR−/−mice do not exhibit female infertility or low pup yield, they are far easier and less costly to use than the previously described SR-B1–based models of coronary artery disease. We found that treatment with the targeted nanoparticles improved the cardiac functions and corrected hematologic abnormalities caused by the atherogenic diet in SR-B1ΔCT/LDLR−/−mice but did not alter the distinctive plasma lipid levels.CONCLUSIONS:The SR-B1ΔCT/LDLR−/−mice developed diet-inducible, fatal atherosclerotic coronary artery disease, which could be ameliorated by targeted nanoparticle therapy. Our study provides new tools for the development of cardiovascular therapies.
- Exploring Platelet Dysregulation in Congenital Heart Disease: Novel Findings From Mass Cytometryby Roxane Darbousset Division of Immunology, Boston Children’s Hospital, Harvard Medical School, MA. on October 17, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2540-2542, December 1, 2024. <br/>
- Thin Disguise: Workup of a Left Atrial Appendage Anomalyby Robyn Bryde B. Robbie Williams Matthew W. Martinez Department of Cardiovascular Medicine, Atlantic Health, Morristown Medical Center, NJ (R.B., M.W.M.). Department of Cardiovascular Medicine, Hypertrophic Cardiomyopathy Center, Emory Heart and Vascular Center, Atlanta, GA (B.R.W.). Sports Cardiology and Hypertrophic Cardiomyopathy, Morristown Medical Center, NJ (M.W.M.). on October 15, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Integrating Coronary Artery Assessment and Myocardial Late Enhancement Imaging With Photon-Counting Detector CT: Visualizing the Invisibleby Marie-Julie D.K. Lemmens Samuel Heuts Elham Bidar Joachim E. Wildberger Casper Mihl Martijn W. Smulders Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (MJ.D.K.L., S.H., E.B., J.E.W., C.M., M.W.S.). Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, the Netherlands. (MJ.D.K.L., J.E.W., C.M.) Department of Cardiology, Maastricht University Medical Center+, the Netherlands. (MJ.D.K.L., M.W.S.) Department of Cardiothoracic Surgery, Maastricht University Medical Center+, the Netherlands. (S.H., E.B.) on October 10, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Real-Time Imaging Assessment of Stress-Induced Vascular Inflammation Using Heartbeat-Synchronized Motion Compensationby Minseok A. Jang Joon Woo Song Ryeong Hyun Kim Dong Oh Kang Ungyo Kang Hyun Jung Kim Jin Hyuk Kim Eun Jin Park Ye Hee Park Bo-Hyung Lee Chi Kyung Kim Kyeongsoon Park Jin Won Kim Hongki Yoo Department of Mechanical Engineering, KAIST (Korea Advanced Institute of Science and Technology), Daejeon, Korea (M.A.J., U.K., H.Y.). Multimodal Imaging and Theranostics Laboratory, Cardiovascular Center (J.W.S., R.H.K., D.O.K., H.J.K., J.H.K., E.J.P., Y.H.P., J.W.K.), Korea University Guro Hospital, Seoul. Department of Neurology (B.-H.L., C.K.K.), Korea University Guro Hospital, Seoul. Department of Systems Biotechnology, Chung-Ang University, Anseong, Korea (K.P.). on October 10, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2493-2506, December 1, 2024. <br/>BACKGROUND:Chronic mental stress accelerates atherosclerosis through complicated neuroimmune pathways, needing for advanced imaging techniques to delineate underlying cellular mechanisms. While histopathology, ex vivo imaging, and snapshots of in vivo images offer promising evidence, they lack the ability to capture real-time visualization of blood cell dynamics within pulsatile arteries in longitudinal studies.METHODS:An electrically tunable lens was implemented in intravital optical microscopy, synchronizing the focal plane with heartbeats to follow artery movements. ApoE−/−mice underwent 2 weeks of restraint stress before baseline imaging followed by 2 weeks of stress exposure in the longitudinal imaging, while nonstressed mice remained undisturbed. The progression of vascular inflammation was assessed in the carotid arteries through intravital imaging and histological analyses.RESULTS:A 4-fold reduction of motion artifact, assessed by interframe SD, and an effective temporal resolution of 25.2 Hz were achieved in beating murine carotid arteries. Longitudinal intravital imaging showed chronic stress led to a 6.09-fold (P=0.017) increase in myeloid cell infiltration compared with nonstressed mice. After 3 weeks, we observed that chronic stress intensified vascular inflammation, increasing adhered myeloid cells by 2.45-fold (P=0.031), while no significant changes were noted in nonstressed mice. Microcirculation imaging revealed increased circulating, rolling, and adhered cells in stressed mice’s venules. Histological analysis of the carotid arteries confirmed the in vivo findings that stress augmented plaque area, myeloid cell and macrophage accumulation, and necrotic core volume while reducing fibrous cap thickness indicating accelerated plaque formation. We visualized the 3-dimensional structure of the carotid artery and 4-dimensional dynamics of the venules in the cremaster muscle.CONCLUSIONS:Dynamic focusing motion compensation intravital microscopy enabled subcellular resolution in vivo imaging of blood cell dynamics in beating arteries under chronic restraint stress in real time. This novel technique emphasizes the importance of advanced in vivo imaging for understanding cardiovascular disease.
- Novel Therapeutics and Upcoming Clinical Trials Targeting Inflammation in Cardiovascular Diseasesby Nicola Potere Aldo Bonaventura Antonio Abbate Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Italy (N.P.). Department of Internal Medicine, Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, ASST Sette Laghi Varese, Italy (A.B.). Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville (A.A.). on October 10, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2371-2395, December 1, 2024. <br/>Cardiovascular disease (CVD) remains a major health burden despite significant therapeutic advances accomplished over the last decades. It is widely and increasingly recognized that systemic inflammation not only represents a major cardiovascular risk and prognostic factor but also plays key pathogenic roles in CVD development and progression. Despite compelling preclinical evidence suggesting large potential of anti-inflammatory pharmacological interventions across numerous CVDs, clinical translation remains incomplete, mainly due to (1) yet undefined molecular signaling; (2) challenges of safety and efficacy profile of anti-inflammatory drugs; and (3) difficulties in identifying optimal patient candidates and responders to anti-inflammatory therapeutics, as well as optimal therapeutic windows. Randomized controlled trials demonstrated the safety/efficacy of canakinumab and colchicine in secondary cardiovascular prevention, providing confirmation for the involvement of a specific inflammatory pathway (NLRP3 [NACHT, LRR, and PYD domain-containing protein 3] inflammasome/IL [interleukin]-1β) in atherosclerotic CVD. Colchicine was recently approved by the US Food and Drug Administration for this indication. Diverse anti-inflammatory drugs targeting distinct inflammatory pathways are widely used for the management of other CVDs including myocarditis and pericarditis. Ongoing research efforts are directed to implementing anti-inflammatory therapeutic strategies across a growing number of CVDs, through repurposing of available anti-inflammatory drugs and development of novel anti-inflammatory compounds, which are herein concisely discussed. This review also summarizes the main characteristics and findings of completed and upcoming randomized controlled trials directly targeting inflammation in CVDs, and discusses major challenges and future perspectives in the exciting and constantly expanding landscape of cardioimmunology.
- ASGR1 Deficiency Inhibits Atherosclerosis in Western Diet–Fed ApoE−/− Mice by Regulating Lipoprotein Metabolism and Promoting Cholesterol Effluxby Yuyan Zhang Xinhai Jiang Weizhi Wang Lijuan Lei Ren Sheng Shunwang Li Jinque Luo Huan Liu Jing Zhang Xiaowan Han Yining Li Yuhao Zhang Chenyin Wang Shuyi Si Zheng-Gen Jin Yanni Xu State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.). Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.L., H.L., Z.-G.J.). on October 10, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2428-2449, December 1, 2024. <br/>BACKGROUND:Atherosclerosis is the most common cause of cardiovascular diseases. Clinical studies indicate that loss-of-function ASGR1 (asialoglycoprotein receptor 1) is significantly associated with lower plasma cholesterol levels and reduces cardiovascular disease risk. However, the effect of ASGR1 on atherosclerosis remains incompletely understood; whether inhibition of ASGR1 causes liver injury remains controversial. Here, we comprehensively investigated the effects and the underlying molecular mechanisms of ASGR1 deficiency and overexpression on atherosclerosis and liver injury in mice.METHODS:We engineeredAsgr1knockout mice (Asgr1−/−),Asgr1andApoEdouble-knockout mice (Asgr1−/−ApoE−/−), and ASGR1-overexpressing mice on anApoE−/−background and then fed them different diets to assess the role of ASGR1 in atherosclerosis and liver injury.RESULTS:After being fed a Western diet for 12 weeks,Asgr1−/−ApoE−/−mice exhibited significantly decreased atherosclerotic lesion areas in the aorta and aortic root sections, reduced plasma VLDL (very-low-density lipoprotein) cholesterol and LDL (low-density lipoprotein) cholesterol levels, decreased VLDL production, and increased fecal cholesterol contents. Conversely, ASGR1 overexpression inApoE−/−mice increased atherosclerotic lesions in the aorta and aortic root sections, augmented plasma VLDL cholesterol and LDL cholesterol levels and VLDL production, and decreased fecal cholesterol contents. Mechanistically, ASGR1 deficiency reduced VLDL production by inhibiting the expression of MTTP (microsomal triglyceride transfer protein) and ANGPTL3 (angiopoietin-like protein 3)/ANGPTL8 (angiopoietin-like protein 8) but increasing LPL (lipoprotein lipase) activity, increased LDL uptake by increasing LDLR (LDL receptor) expression, and promoted cholesterol efflux through increasing expression of LXRα (liver X receptor-α), ABCA1 (ATP-binding cassette subfamily A member 1), ABCG5 (ATP-binding cassette subfamily G member 5), and CYP7A1 (cytochrome P450 family 7 subfamily A member 1). These underlying alterations were confirmed in ASGR1-overexpressingApoE−/−mice. In addition, ASGR1 deficiency exacerbates liver injury in Western diet–inducedAsgr1−/−ApoE−/−mice and high-fat diet–induced but not normal laboratory diet–induced and high-fat and high-cholesterol diet–inducedAsgr1−/−mice, while its overexpression mitigates liver injury in Western diet–induced ASGR1-overexpressingApoE−/−mice.CONCLUSIONS:Inhibition of ASGR1 inhibits atherosclerosis in Western diet–fedApoE−/−mice, suggesting that inhibiting ASGR1 may serve as a novel therapeutic strategy to treat atherosclerosis and cardiovascular diseases.
- Immunoregulatory Macrophages Modify Local Pulmonary Immunity and Ameliorate Hypoxic Pulmonary Hypertensionby Angeles Fernandez-Gonzalez Amit Mukhia Janhavi Nadkarni Gareth R. Willis Monica Reis Kristjan Zhumka Sally Vitali Xianlan Liu Alexandra Galls S. Alex Mitsialis Stella Kourembanas Division of Newborn Medicine (A.F.-G., A.M., J.N., G.R.W., M.R., K.Z., S.V., X.L., A.G., S.A.M., S.K.), Boston Children’s Hospital, MA. Division of Critical Care Medicine, Department of Anesthesia, Perioperative, and Pain Medicine (S.V.), Boston Children’s Hospital, MA. Department of Pediatrics, Harvard Medical School, Boston, MA (A.F.-G., A.M., J.N., G.R.W., M.R., K.Z., X.L., A.G., S.A.M., S.K.). on October 10, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page e288-e303, December 1, 2024. <br/>BACKGROUND:Macrophages play a significant role in the onset and progression of vascular disease in pulmonary hypertension, and cell-based immunotherapies aimed at treating vascular remodeling are lacking. We aimed to evaluate the effect of pulmonary administration of macrophages modified to have an anti-inflammatory/proresolving phenotype in attenuating early pulmonary inflammation and progression of experimentally induced pulmonary hypertension.METHODS:Mouse bone marrow–derived macrophages were polarized in vitro to a regulatory (M2reg) phenotype. M2regprofile and anti-inflammatory capacity were assessed in vitro upon lipopolysaccharide/IFNγ (interferon-γ) restimulation, before their administration to 8- to 12-week-old mice. M2regprotective effect was evaluated at early (2–4 days) and late (4 weeks) time points during hypoxia (8.5% O2) exposure. Levels of inflammatory markers were quantified in alveolar macrophages and whole lung, while pulmonary hypertension development was ascertained by right ventricular systolic pressure (RVSP) and right ventricular hypertrophy measurements. Bronchoalveolar lavage from M2reg-transplanted hypoxic mice was collected and its inflammatory potential evaluated on naive bone marrow–derived macrophages.RESULTS:M2regmacrophages expressingTgfβ,Il10, andCd206demonstrated a stable anti-inflammatory phenotype in vitro, by downregulating the induction of proinflammatory cytokines and surface molecules (Cd86,Il6, andTnfα) upon a subsequent proinflammatory stimulus. A single dose of M2regsattenuated hypoxic monocytic recruitment and perivascular inflammation. Early hypoxic lung and alveolar macrophage inflammation leading to pulmonary hypertension development was significantly reduced, and, importantly, M2regsattenuated right ventricular hypertrophy, right ventricular systolic pressure, and vascular remodeling at 4 weeks post-treatment.CONCLUSIONS:Adoptive transfer of M2regshalts the recruitment of monocytes and modifies the hypoxic lung microenvironment, potentially changing the immunoreactivity of recruited macrophages and restoring normal immune functionality of the lung. These findings provide new mechanistic insights into the diverse role of macrophage phenotype on lung vascular homeostasis that can be explored as novel therapeutic targets.
- Lipoprotein Apheresis: Utility, Outcomes, and Implementation in Clinical Practice: A Scientific Statement From the American Heart Associationby Eugenia Gianos P. Barton Duell Peter P. Toth Patrick M. Moriarty Gilbert R. Thompson Eliot A. Brinton Lisa C. Hudgins Mary Nametka Kathleen H. Byrne Geetha Raghuveer Prashant Nedungadi Laurence S. Sperling on October 7, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page e304-e321, December 1, 2024. <br/>Despite the availability of multiple classes of lipoprotein-lowering medications, some high-risk patients have persistent hypercholesterolemia and may require nonpharmacologic therapy. Lipoprotein apheresis (LA) is a valuable but underused adjunctive therapeutic option for low-density lipoprotein cholesterol and lipoprotein(a) lowering, particularly in children and adults with familial hypercholesterolemia. In addition to lipid lowering, LA reduces serum levels of proinflammatory and prothrombotic factors, reduces blood viscosity, increases microvascular myocardial perfusion, and may provide beneficial effects on endothelial function. Multiple observational studies demonstrate strong evidence for improved cardiovascular outcomes with LA; however, use in the United States is limited to a fraction of its Food and Drug Administration–approved indications. In addition, there are limited data regarding LA benefit for refractory focal segmental glomerulosclerosis. In this scientific statement, we review the history of LA, mechanisms of action, cardiovascular and renal outcomes data, indications, and options for treatment.
- Acetaminophen Overdose Reveals PAR4 as a Low-Expressing but Potent Receptor on the Hepatic Endothelium in Miceby Rahul Rajala Audrey C.A. Cleuren Courtney T. Griffin Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (R.R., A.C.A.C., C.T.G.). Department of Cell Biology, University of Oklahoma Health Sciences Center (R.R., A.C.A.C., C.T.G.). Harold Hamm Diabetes Center, Oklahoma City, OK (R.R.). on October 3, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. <br/>BACKGROUND:The protease thrombin, which elicits multiple physiological and pathological effects on vascular endothelial cells (ECs), can signal through PARs (protease-activated receptors) 1 and 4. PAR1 is a high-affinity thrombin receptor known to signal on ECs, whereas PAR4 is a low-affinity thrombin receptor, and evidence for its expression and function on ECs is mixed. This study aims to exploit the high levels of thrombin generation and hepatic vascular dysfunction that occur during acetaminophen (APAP) overdose to determine (1) whether hepatic endothelial PAR4 is a functional receptor, and (2) the endothelial-specific functions for PAR1 and PAR4 in a high thrombin and pathological setting.METHODS:We generated mice with conditional deletion ofPar1/Par4in ECs and overdosed them with APAP. Hepatic vascular permeability, erythrocyte accumulation in the liver, thrombin generation, and liver function were assessed following overdose. Additionally, we investigated the expression levels of endothelial PARs and how they influence transcription in APAP-overdosed liver ECs using endothelial translating ribosome affinity purification followed by next-generation sequencing.RESULTS:We found that mice deficient in high-expressing endothelialPar1or low-expressingPar4had equivalent reductions in APAP-induced hepatic vascular instability, although mice deficient for both receptors had lower vascular permeability at an earlier timepoint after APAP overdose than either of the single mutants. Additionally, mice with loss of both endothelialPar1andPar4had reduced thrombin generation after APAP overdose, suggesting decreased hypercoagulability. Last, we found that endothelial PAR1—but not PAR4—can regulate transcription in hepatic ECs.CONCLUSIONS:Low-expressing PAR4 can react similarly to high-expressing PAR1 in APAP-overdosed hepatic ECs, demonstrating that PAR4 is a potent thrombin receptor. Additionally, these receptors are functionally redundant but act divergently in their expression and ability to influence transcription in hepatic ECs.
- CXCR3-CXCL11 Signaling Restricts Angiogenesis and Promotes Pericyte Recruitmentby Jihui Lee Megan E. Goeckel Allison Levitas Sarah Colijn Jimann Shin Anna Hindes Geonyoung Mun Zarek Burton Bharadwaj Chintalapati Ying Yin Javier Abello Lilianna Solnica-Krezel Amber N. Stratman Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO. Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO. Center of Regenerative Medicine (L.S.-K.), Washington University School of Medicine, St. Louis, MO. on October 3, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2577-2595, December 1, 2024. <br/>BACKGROUND:Endothelial cell (EC)–pericyte interactions are known to remodel in response to hemodynamic forces; yet there is a lack of mechanistic understanding of the signaling pathways that underlie these events. Here, we have identified a novel signaling network regulated by blood flow in ECs—the chemokine receptor CXCR3 (CXC motif chemokine receptor 3) and one of its ligands, CXCL11 (CXC motif chemokine ligand 11)—that delimits EC angiogenic potential and promotes pericyte recruitment to ECs during development.METHODS:We investigated the role of CXCR3 on vascular development using both 2- and 3-dimensional in vitro assays, to study EC-pericyte interactions and EC behavioral responses to blood flow. Additionally, genetic mutants and pharmacological modulators were used in zebrafish in vivo to study the impacts of CXCR3 loss and gain of function on vascular development.RESULTS:In vitro modeling of EC-pericyte interactions demonstrates that suppression of EC-specific CXCR3 signaling leads to loss of pericyte association with EC tubes. In vivo, phenotypic defects are particularly noted in the cranial vasculature, where we see a loss of pericyte association with ECs and expansion of the vasculature in zebrafish treated with the Cxcr3 inhibitor AMG487 or in homozygouscxcr3.1/3.2/3.3triple mutants. We also demonstrate that CXCR3-deficient ECs are more elongated, move more slowly, and have impaired EC-EC junctions compared with their control counterparts.CONCLUSIONS:Our results suggest that CXCR3 signaling in ECs helps promote vascular stabilization events during development by preventing EC overgrowth and promoting pericyte recruitment.
- lncRNA VELRP Modulates Pulmonary Arterial Smooth Muscle Cell Proliferation and Promotes Vascular Remodeling in Pulmonary Hypertensionby Cuilian Liu Jidong Chen Xingtao Huang Qinyi Xia Lei Yang Jiao Guo Jinglin Tian Jun Wang Yanqin Niu Li Li Deming Gou Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China. on October 3, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2560-2576, December 1, 2024. <br/>BACKGROUND:Pulmonary hypertension is a devastating vascular disorder characterized by extensive pulmonary vascular remodeling, ultimately leading to right ventricular failure and death. Activation of PDGF (platelet-derived growth factor) signaling promotes the hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs), thus contributing to the pulmonary vascular remodeling. However, the molecular mechanisms that govern hyperproliferation of PASMCs induced by PDGF remain largely unknown, including the contribution of long noncoding RNAs (lncRNAs). In this study, we aimed to identify a novel lncRNA regulated by PDGF implicated in PASMC proliferation in pulmonary vascular remodeling.METHODS:RNA-sequencing analysis was conducted to identify a novel lncRNA named vessel-enriched lncRNA regulated by PDGF-BB (platelet-derived growth factor-BB; VELRP). Functional investigations of VELRP were performed using knockdown and overexpression strategies along with RNA sequencing. Validation of the function and potential mechanisms of VELRP was performed through Western blot, RNA immunoprecipitation, and chromatin immunoprecipitation assays.RESULTS:We identified a novel vessel-enriched lncRNA with an increased response to PDGF-BB stimulus. VELRP was identified as an evolutionarily conserved RNA molecule. Modulation of VELRP in PASMCs significantly altered cell proliferation. Mechanistically, VELRP enhances trimethylation of H3K4 (histone H3 lysine 4) by interacting with WDR5 (WD repeat-containing protein 5), leading to increased expression of CDK (cyclin-dependent kinase) 1, CDK2, and CDK4 and consequent hyperproliferation of PASMCs. The pathological relevance of VELRP upregulation in pulmonary artery was confirmed using rat pulmonary hypertension models in vivo, as well as in PASMCs from patients with idiopathic pulmonary arterial hypertension. Specific knockdown of VELRP in smooth muscle cells using adeno-associated virus type 9 SM22α (smooth muscle protein 22α) promoter–shRNA-mediated silencing of VELRP resulted in a significant decrease in right ventricular systolic pressure and vascular remodeling in rat pulmonary hypertension model.CONCLUSIONS:VELRP, as an lncRNA upregulated by PDGF-BB, mediates PASMC proliferation via WDR5/CDK signaling. In vivo studies demonstrate that targeted intervention of VELRP in smooth muscle cells can prevent the development of pulmonary hypertension.
- Platypnea-Orthodeoxia Syndrome After Esophageal Dilation in a Patient With a Dilated Ascending Aortaby Matthew B. Saunders Matthew S. Michaleski Jeffrey Yim Miles Marchand John Jue David A. Wood Christina L. Luong Michael Y.C. Tsang Teresa S.M. Tsang Darwin F. Yeung Faculty of Medicine, University of British Columbia, Vancouver, Canada. (M.B.S.) Department of Medicine, University of British Columbia, Vancouver, Canada. (M.S.M.) Division of Cardiology, University of British Columbia, Vancouver, Canada. (J.Y., M.M., J.J., D.A.W., C.L.L., M.Y.C.T., T.S.M.T., D.F.Y.) on October 2, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Sudden Cardiac Arrest With Acute Myocardial Infarction Due to Myocardial Bridging and Hypertrophic Cardiomyopathy
- Beyond the Usual Suspects: Myocardial Infarction Triggered by Coronary Vasculitis in a Young Patient With Systemic Lupus Erythematosusby Maria Jose Santa Ana-Bayona Pavel Martinez-Dominguez Oscar Perez-Orpinel Enrique C. Guerra Nilda Espinola-Zavaleta Department of Nuclear Cardiology, National Institute of Cardiology Ignacio Chavez, Mexico City (M.J.S.A.-B., P.M.-D., E.C.G., N.E.-Z.). MD–PhD (PECEM) Program, School of Medicine, National Autonomous University of Mexico, Mexico City (E.C.G.). Department of Hemodynamics, National Institute of Cardiology Ignacio Chavez, Mexico City (O.P.-O.). on September 27, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Endothelial TGF-β Signaling Regulates Endothelial-Mesenchymal Transition During Arteriovenous Fistula Remodeling in Mice With Chronic Kidney Diseaseby Luis Gonzalez John Langford Carly Thaxton Bogdan Yatsula Kathleen A. Martin Julie Goodwin George Tellides Alan Dardik State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China (W.Z., C.S.). Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China (W.Z., X. Li, C.S.). Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT. Division of Cardiac Surgery, Department of Surgery (G.T.), Yale School of Medicine, New Haven, CT. Division of Vascular and Endovascular Surgery, Department of Surgery (A.D.), Yale School of Medicine, New Haven, CT. Department of Pediatrics (J.G.), Yale School of Medicine, New Haven, CT. Department of Cellular and Molecular Physiology (A.D.), Yale School of Medicine, New Haven, CT. Department of Medicine (Cardiovascular Medicine), Yale Cardiovascular Research Center (K.A.M.), Yale School of Medicine, New Haven, CT. Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O.). Surgical Service, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.). Vascular Biology Center, Medical College of Georgia at Augusta University (X. Long). on September 19, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2509-2526, December 1, 2024. <br/>BACKGROUND:Arteriovenous fistulae (AVF) are the preferred vascular access for hemodialysis in patients with end-stage kidney disease. Chronic kidney disease (CKD) is associated with endothelial injury, impaired AVF maturation, and reduced patency, as well as utilization. Because CKD is characterized by multiple pathophysiological processes that induce endothelial-to-mesenchymal transition (EndMT), we hypothesized that CKD promotes EndMT during venous remodeling and that disruption of endothelial TGF (transforming growth factor)-β signaling inhibits EndMT to prevent AVF failure even in the end-stage kidney disease environment.METHODS:The mouse 5/6 nephrectomy and aortocaval fistula models were used. CKD was created via 5/6 nephrectomy, with controls of no (0/6) or partial (3/6) nephrectomy in C57BL/6J mice. AVF were created in mice with knockdown of TGF-βR1/R2 (TGF-β receptors type 1/2) in either smooth muscle cells or endothelial cells. AVF diameters and patency were measured and confirmed by serial ultrasound examination. AVF, both murine and human, were examined using Western blot, histology, and immunofluorescence. Human and mouse endothelial cells were used for in vitro experiments.RESULTS:CKD accelerates TGF-β activation and promotes EndMT that is associated with increased AVF wall thickness and reduced patency in mice. Inhibition of TGF-β signaling in both endothelial cells and smooth muscle cells decreased smooth muscle cell proliferation in the AVF wall, attenuated EndMT, and was associated with reduced wall thickness, increased outward remodeling, and improved AVF patency. Human AVF also showed increased TGF-β signaling and EndMT.CONCLUSIONS:CKD promotes EndMT and reduces AVF patency. Inhibition of TGF-β signaling, especially disruption of endothelial cell–specific TGF-β signaling, attenuates EndMT and improves AVF patency in mouse AVF. Inhibition of EndMT may be a therapeutic approach of translational significance to improve AVF patency in human patients with CKD.
- Atherosclerosis on a Chip: A 3-Dimensional Microfluidic Model of Early Arterial Events in Human Plaquesby Ranganath Maringanti Christian G.M. van Dijk Elana M. Meijer Maarten M. Brandt Mingzi Li Vera P.C. Tiggeloven Merle M. Krebber Ihsan Chrifi Dirk J. Duncker Marianne C. Verhaar Caroline Cheng Department of Nephrology and Hypertension, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, the Netherlands (R.M., C.G.M.v.D., E.M.M., M.M.K., I.C., M.C.V., C.C.). Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (R.M., M.M.B., M.L., V.P.C.T., I.C., D.J.D., C.C.). on September 19, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2453-2472, December 1, 2024. <br/>BACKGROUND:Realistic reconstruction of the in vivo human atherosclerotic environment requires the coculture of different cell types arranged in atherosclerotic vessel–like structures with exposure to flow and circulating cells, presenting challenges for disease modeling. This study aimed to develop a 3-dimensional tubular microfluidic model with quadruple coculture of human aortic smooth muscle cells, human umbilical cord vein endothelial cells, and foam cells to recreate a complex human atherosclerotic vessel in vitro to study the effects of flow and circulating immune cells.METHODS:We developed a coculture protocol utilizing BFP (blue fluorescent protein)-labeled human aortic smooth muscle cells, GFP (green fluorescent protein)-labeled human umbilical cord vein endothelial cells, and THP-1 macrophage-derived, Dil-labeled oxidized LDL (low-density lipoprotein) foam cells within a fibrinogen/collagen I–based 3-dimensional ECM (extracellular matrix). Perfusion experiments were conducted for 24 hours on both atherosclerotic vessels and healthy vessels (BFP-labeled human aortic smooth muscle cells and GFP-labeled human umbilical cord vein endothelial cells without foam cells). Additionally, perfusion with circulating THP-1 monocytes was performed to observe cell extravasation and recruitment.RESULTS:The resulting vessels displayed early lesion morphology, with a layered composition including an endothelium and media, and foam cells accumulating in the subendothelial space. The layered wall composition of both atherosclerotic and healthy vessels remained stable under perfusion. Circulating THP-1 monocytes demonstrated cell extravasation into the atherosclerotic vessel wall and recruitment to the foam cell core. The qPCR (quantitative polymerase chain reaction) analysis indicated increased expression of atherosclerosis markers in the atherosclerotic vessels and adaptation of vascular smooth muscle cell migration in response to flow and the plaque microenvironment, compared with control vessels.CONCLUSIONS:The human 3-dimensional atherosclerosis model demonstrated stability under perfusion and allowed for the observation of immune cell behavior, providing a valuable tool for the atherosclerosis research field.
- To Harmonize or to Hinder … Do We Need 2 Sets of European Hypertension Guidelines in 2024?by Aletta E. Schutte Garry L.R. Jennings School of Population Health, University of New South Wales, Sydney, Australia (A.E.S.). The George Institute for Global Health, Sydney, New South Wales, Australia (A.E.S.). Hypertension in Africa Research Team, MRC Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa (A.E.S.). Faculty of Medicine and Health Sciences, University of Sydney, New South Wales, Australia (G.L.R.J.). School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia (G.L.R.J.). on September 16, 2024
Hypertension, Volume 82, Issue 1, Page 8-10, January 1, 2025. <br/>
- What Is New in the ESC Hypertension Guideline?by Jordana B. Cohen Renal-Electrolyte and Hypertension Division and Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia. on September 16, 2024
Hypertension, Volume 82, Issue 1, Page 11-13, January 1, 2025. <br/>
- Intracardiac Echocardiography in Alterra Prestent Constrained by the Native Pulmonary Valve Leafletby Rupesh Kumar Natarajan Neha Ahluwalia Thomas Fagan Division of Pediatric Cardiology, Children’s Hospital of Michigan, Detroit (R.K.N., N.A., T.F.) Division of Pediatric Cardiology, Department of Pediatrics, West Virginia University, Morgantown (R.K.N.). on September 10, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- Coronary Artery Embolism Secondary to Q Fever Endocarditisby Minggang Zhou Li Chen Yong He Department of Cardiology (M.Z., Y.H.), West China Hospital, Sichuan University, Chengdu. Laboratory of Cardiovascular Diseases (L.C.), West China Hospital, Sichuan University, Chengdu. on August 29, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e016839, December 1, 2024. <br/>
- Carcinoid Pulmonary Valve Stenosis: Multimodality Imaging and Transcatheter Valve Implant With Prestenting Technique
- Chronic Subepicardial Hematoma: A Multimodality Imaging Diagnostic Approach After Elective Percutaneous Transluminal Coronary Angioplastyby Maria Riasat Sakul Sakul Vikram Agarwal Department of Cardiology, Mount Sinai Morningside, Icahn School of Medicine at Mount Sinai, New York, NY. on August 23, 2024
Circulation: Cardiovascular Imaging, Ahead of Print. <br/>
- High-Dimensional Single-Cell Mass Cytometry Demonstrates Differential Platelet Functional Phenotypes in Infants With Congenital Heart Diseaseby Sean X. Gu Brian S. Marcus Vivian W. Gu Adarsh P. Varghese John Hwa E. Vincent S. Faustino Department of Laboratory Medicine (S.X.G.), Yale University School of Medicine, New Haven, CT. Department of Pediatrics (B.S.M., E.V.S.F.), Yale University School of Medicine, New Haven, CT. Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center (V.W.G., A.P.V., J.H.), Yale University School of Medicine, New Haven, CT. on August 22, 2024
Arteriosclerosis, Thrombosis, and Vascular Biology, Volume 44, Issue 12, Page 2530-2539, December 1, 2024. <br/>BACKGROUND:Congenital heart disease (CHD) is a group of complex heart defects associated with hematologic abnormalities, including increased risk of thrombotic and bleeding events. Past studies have observed evidence of platelet hyperreactivity, while other studies showed decreased platelet activation in patients with CHD. The goal of this study was to develop a mass spectrometry approach to characterize single platelets in infants with CHD and identify potential etiology for such discrepant results.METHODS:We enrolled 19 infants with CHD along with 21 non-CHD controls at Yale New Haven Children’s Heart Center. A single-cell high-dimensional mass cytometry method was developed to quantitatively interrogate platelet surface markers in whole blood. Additionally, plasma cytokine analysis was performed through a multiplexed panel of 52 vascular and inflammatory markers to assess for platelet releasates.RESULTS:We found that infants with CHD had significant differences in platelet activation and functional markers by mass cytometry compared with non-CHD controls. Based on cell surface markers, we classified the platelets into 8 subpopulations (P0 to P7). Distinct subpopulations of platelets (P1, P4, and P5) exhibiting decreased aggregatory phenotype but altered secretory phenotypes were also identified and found to be more abundant in the blood of infants with CHD. Electron microscopy identified increased proportion of hypogranular platelets in CHD. Moreover, cytokine analysis demonstrated an overall increase in plasma cytokines and biomarkers in CHD, including IL (interleukin)-6, IL-8, IL-27, RANTES (regulated upon activation, normal T cell expressed and secreted), and VWF (von Willebrand factor), which are expressed in platelet granules and can be released upon activation.CONCLUSIONS:We developed a robust mass cytometry approach to identify platelet phenotypic heterogeneity. Infants with CHD had alterations in distinct subpopulations of platelets with overall reduced aggregatory phenotype and secretory dysfunction. These findings suggest that platelets in infants with CHD may be exhausted due to persistent stimulation and may explain both bleeding and thrombotic vascular complications associated with CHD.
- Multimodality Imaging of a Rare Extranodal Rosai-Dorfman Disease Involving Great Vesselsby Rui Wang Ziming Zhang Jing Zhang Guohua Wang Li Zhang Mingxing Xie Department of Ultrasound Medicine (R.W., Z.Z., J.Z., L.Z., M.X.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. Department of Cardiovascular Surgery (G.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, China (R.W., Z.Z., J.Z., L.Z., M.X.). Hubei Key Laboratory of Molecular Imaging, Wuhan, China (R.W., Z.Z., J.Z., L.Z., M.X.). on August 22, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e016746, December 1, 2024. <br/>
- Hypertensive Disorders of Pregnancy and Brain Health in Midlife: The CARDIA Studyby Xiaqing Jiang Pamela J. Schreiner Erica P. Gunderson Kristine Yaffe Department of Psychiatry and Behavioral Sciences, University of California San Francisco. (X.J., K.Y.) Department of Neurology, Epidemiology and Biostatistics, University of California San Francisco. (K.Y.) Division of Epidemiology and Community Health, University of Minnesota, Minneapolis (P.J.S.). Division of Research, Kaiser Permanente Northern California, Oakland (E.P.G.). Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (E.P.G.). San Francisco VA Health Care System, CA (K.Y.). on August 20, 2024
Hypertension, Ahead of Print. <br/>BACKGROUND:To understand the role of hypertensive disorders of pregnancy (HDP), including preeclampsia and gestational hypertension (GH), in brain health earlier in life, we investigated the association of HDP with midlife cognition and brain health.METHODS:We studied a prospective cohort of women, baseline age 18 to 30 years, who were assessed at study years 25 and 30 with a cognitive battery and a subset with brain magnetic resonance imaging. A history of HDP was defined based on self-report. We conducted linear regression to assess the association of a history of preeclampsia, GH, or no HDP with cognition and brain magnetic resonance imaging white matter hyperintensities.RESULTS:Among 1441 women (mean age, 55.2±3.6 years), 202 reported preeclampsia and 112 reported GH. GH was associated with worse cognitive performance: global cognition (mean score, 23.2 versus 24.0;P=0.018), processing speed (67.5 versus 71.3;P=0.01), verbal fluency (29.5 versus 31.1;P=0.033), and a trend for executive function (24.3 versus 22.6;P=0.09), after multivariable adjustment. GH was associated with a greater 5-year decline in processing speed (mean change, −4.9 versus −2.7;P=0.049) and executive function (−1.7 versus 0.3;P=0.047); preeclampsia was associated with a greater 5-year decline on delayed verbal memory (−0.3 versus 0.1;P=0.041). GH and preeclampsia were associated with greater white matter hyperintensities in the parietal and frontal lobes, respectively.CONCLUSIONS:GH and preeclampsia are associated with cognition and white matter hyperintensities during midlife, with differences in cognitive domains and brain lobes. Women with HDP may need to be closely monitored for adverse brain outcomes starting in midlife.
- Vanishing Left Atrial Mass in a Middle-aged Woman: Spontaneous Intramural Left Atrial Hematoma in Isolated Atrial Amyloidosisby Midori Makino Keishi Moriwaki Naoki Fujimoto Yosuke Kirii Hana Mizutani Tomoyuki Goto Masaki Ishida Ryuji Okamoto Kyoko Imanaka-Yoshida Kaoru Dohi Department of Cardiology and Nephrology (M.M., K.M., N.F., Y.K., H.M., R.O., K.D.), Mie University Graduate School of Medicine, Tsu, Japan Department of Radiology (M.I.), Mie University Graduate School of Medicine, Tsu, Japan Department of Pathology and Matrix Biology (K.I.-Y.), Mie University Graduate School of Medicine, Tsu, Japan Department of Cardiology, Suzuka General Hospital, Suzuka, Japan (M.M.). Department of Cardiovascular Surgery, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan (T.G.). on July 31, 2024
Circulation: Cardiovascular Imaging, Volume 17, Issue 12, Page e016905, December 1, 2024. <br/>
- Transforming Hypertension Diagnosis and Management in The Era of Artificial Intelligence: A 2023 National Heart, Lung, and Blood Institute (NHLBI) Workshop Reportby Daichi Shimbo Rashmee U. Shah Marwah Abdalla Ritu Agarwal Faraz S. Ahmad Gabriel Anaya Zachi I. Attia Sheana Bull Alexander R. Chang Yvonne Commodore-Mensah Keith Ferdinand Kensaku Kawamoto Rohan Khera Jane Leopold James Luo Sonya Makhni Bobak J. Mortazavi Young S. Oh Lucia C. Savage Erica S. Spatz George Stergiou Mintu P. Turakhia Paul K. Whelton Clyde W. Yancy Erin Iturriaga Department of Medicine, Columbia University Irving Medical Center, New York, NY (D.S., M.A.). Division of Cardiovascular Medicine (R.U.S.), University of Utah School of Medicine, Salt Lake City. Department of Biomedical Informatics (K.K.), University of Utah School of Medicine, Salt Lake City. Center for Digital Health and Artificial Intelligence, Johns Hopkins Carey Business School, Baltimore, MD (R.A.). Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (F.S.A.). Division of Cardiovascular Sciences, National Institutes of Health, National Heart, Lung and Blood Institute, Bethesda, MD (G.A., J.L., Y.S.O., E.I.). Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Z.I.A.). Department of Community and Behavioral Health, Colorado School of Public Health, Aurora (S.B.). Departments of Nephrology and Population Health Sciences, Geisinger, Danville, PA (A.R.C.). Johns Hopkins School of Nursing and Bloomberg School of Public Health, Department of Epidemiology, Baltimore, MD (Y.C.-M.). John W. Deming Department of Medicine (K.F.), Tulane University School of Medicine, New Orleans, LA. Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine (P.K.W.), Tulane University School of Medicine, New Orleans, LA. Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT (R.K., E.S.S.). Center for Outcomes Research and Evaluation, Yale New Haven Hospital, CT (R.K., E.S.S.). Section of Health Informatics, Department of Biostatistics (R.K.), Yale University School of Public Health, New Haven, CT Department of Epidemiology (Chronic Diseases) (E.S.S.), Yale University School of Public Health, New Haven, CT Yale University School of Public Health, New Haven, CT (R.K.). Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.L.). Department of Medicine, University of Chicago Medicine and Biological Sciences Division, Chicago (S.M.). Department of Computer Science & Engineering, Texas A&M University, College Station (B.J.M.). Yale School of Medicine, Yale University, New Haven, CT (B.J.M.). Chief Privacy & Regulatory Officer, Omada Health, Inc, San Francisco, CA (L.C.S.). Hypertension Center STRIDE-7, National and Kapodistrian University of Athens, School of Medicine, Third Department of Medicine, Sotiria Hospital, Greece (G.S.). Stanford University School of Medicine (Cardiovascular Medicine), CA (M.P.T.). Division of Cardiology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL (C.W.Y.). on July 16, 2024
Hypertension, Volume 82, Issue 1, Page 36-45, January 1, 2025. <br/>Hypertension is among the most important risk factors for cardiovascular disease, chronic kidney disease, and dementia. The artificial intelligence (AI) field is advancing quickly, and there has been little discussion on how AI could be leveraged for improving the diagnosis and management of hypertension. AI technologies, including machine learning tools, could alter the way we diagnose and manage hypertension, with potential impacts for improving individual and population health. The development of successful AI tools in public health and health care systems requires diverse types of expertise with collaborative relationships between clinicians, engineers, and data scientists. Unbiased data sources, management, and analyses remain a foundational challenge. From a diagnostic standpoint, machine learning tools may improve the measurement of blood pressure and be useful in the prediction of incident hypertension. To advance the management of hypertension, machine learning tools may be useful to find personalized treatments for patients using analytics to predict response to antihypertension medications and the risk for hypertension-related complications. However, there are real-world implementation challenges to using AI tools in hypertension. Herein, we summarize key findings from a diverse group of stakeholders who participated in a workshop held by the National Heart, Lung, and Blood Institute in March 2023. Workshop participants presented information on communication gaps between clinical medicine, data science, and engineering in health care; novel approaches to estimating BP, hypertension risk, and BP control; and real-world implementation challenges and issues.
- Brief Review and Primer of Key Terminology for Artificial Intelligence and Machine Learning in Hypertensionby Patrick Dunn Asif Ali Akash P. Patel Srikanta Banerjee American Heart Association, Center for Health Technology & Innovation, Dallas, TX (P.D.). University of Texas Health Science Center, Houston (A.A.). University of Texas at Austin, Dell Medical School (A.P.). School of Health Sciences and Public Policy, Walden University, Minneapolis, MN (S.B.). on July 16, 2024
Hypertension, Volume 82, Issue 1, Page 26-35, January 1, 2025. <br/>Recent breakthroughs in artificial intelligence (AI) have caught the attention of many fields, including health care. The vision for AI is that a computer model can process information and provide output that is indistinguishable from that of a human and, in specific repetitive tasks, outperform a human’s capability. The 2 critical underlying technologies in AI are used for supervised and unsupervised machine learning. Machine learning uses neural networks and deep learning modeled after the human brain from structured or unstructured data sets to learn, make decisions, and continuously improve the model. Natural language processing, used for supervised learning, is understanding, interpreting, and generating information using human language in chatbots and generative and conversational AI. These breakthroughs result from increased computing power and access to large data sets, setting the stage for releasing large language models, such as ChatGPT and others, and new imaging models using computer vision. Hypertension management involves using blood pressure and other biometric data from connected devices and generative AI to communicate with patients and health care professionals. AI can potentially improve hypertension diagnosis and treatment through remote patient monitoring and digital therapeutics.
- Artificial Intelligence–Derived Risk Prediction: A Novel Risk Calculator Using Office and Ambulatory Blood Pressureby Pedro Guimarães Andreas Keller Michael Böhm Lucas Lauder Tobias Fehlmann Luis M. Ruilope Ernest Vinyoles Manuel Gorostidi Julián Segura Gema Ruiz-Hurtado Natalie Staplin Bryan Williams Alejandro de la Sierra Felix Mahfoud Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany (P.G., A.K., T.F.). University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Institute for Nuclear Sciences Applied to Health, Portugal (P.G.). Department of Neurology and Neurological Sciences, Stanford University, CA (A.K.). Department of Internal Medicine III, Cardiology, Angiology, Intensive Care Medicine, Universitätsklinikum des Saarlandes, Saarland University, Homburg/Saar, Germany (M.B., L.L., F.M.). Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, (L.M.R., J.S., G.R.-H.), Hospital Universitario 12 de Octubre, Madrid, Spain. Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (L.M.R., G.R.-H.), Hospital Universitario 12 de Octubre, Madrid, Spain. Faculty of Sport Sciences, European University of Madrid, Spain (L.M.R.). La Mina Primary Care Center, University of Barcelona, Spain (E.V.). IDIAP Jordi Gol, Barcelona, Spain (E.V.). Department of Nephrology, Hospital Universitario Central de Asturias, REDinREN, Oviedo, Spain (M.G.). Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom (N.S.). University College London (UCL), Institute of Cardiovascular Science, National Institute for Health Research, UCL Hospitals Biomedical Research Centre, United Kingdom (B.W.). Department of Internal Medicine, Hospital Universitario Mútua Terrasa, Universidad de Barcelona, Spain (A.d.l.S.). Harvard-MIT Biomedical Engineering Center, Institute for Medical Engineering and Science, MIT, Cambrigde, MA (F.M.). Department of Cardiology, University Heart Center, Basel University Hospital, Petersgraben 4, 4031 Basel (F.M.). on April 25, 2024
Hypertension, Volume 82, Issue 1, Page 46-56, January 1, 2025. <br/>BACKGROUND:Quantification of total cardiovascular risk is essential for individualizing hypertension treatment. This study aimed to develop and validate a novel, machine-learning–derived model to predict cardiovascular mortality risk using office blood pressure (OBP) and ambulatory blood pressure (ABP).METHODS:The performance of the novel risk score was compared with existing risk scores, and the possibility of predicting ABP phenotypes utilizing clinical variables was assessed. Using data from 59 124 patients enrolled in the Spanish ABP Monitoring registry, machine-learning approaches (logistic regression, gradient-boosted decision trees, and deep neural networks) and stepwise forward feature selection were used.RESULTS:For the prediction of cardiovascular mortality, deep neural networks yielded the highest clinical performance. The novel mortality prediction models using OBP and ABP outperformed other risk scores. The area under the curve achieved by the novel approach, already when using OBP variables, was significantly higher when compared with the area under the curve of the Framingham risk score, Systemic Coronary Risk Estimation 2, and Atherosclerotic Cardiovascular Disease score. However, the prediction of cardiovascular mortality with ABP instead of OBP data significantly increased the area under the curve (0.870 versus 0.865;P=3.61×10−28), accuracy, and specificity, respectively. The prediction of ABP phenotypes (ie, white-coat, ambulatory, and masked hypertension) using clinical characteristics was limited.CONCLUSIONS:The receiver operating characteristic curves for cardiovascular mortality using ABP and OBP with deep neural network models outperformed all other risk metrics, indicating the potential for improving current risk scores by applying state-of-the-art machine learning approaches. The prediction of cardiovascular mortality using ABP data led to a significant increase in area under the curve and performance metrics.