51
|
Wang Y, Bi L, Li Q, Wang Q, Lv T, Zhang P. Remnant cholesterol inflammatory index and its association with all-cause and cause-specific mortality in middle-aged and elderly populations: evidence from US and Chinese national population surveys. Lipids Health Dis 2025; 24:155. [PMID: 40275392 PMCID: PMC12020154 DOI: 10.1186/s12944-025-02580-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND The remnant cholesterol inflammatory index (RCII) is a novel metric that combines remnant cholesterol and high-sensitivity C-reactive protein, reflecting the metabolic and inflammatory risk. This study investigates the association between RCII and long-term risks of all-cause and cause-specific mortality in middle-aged and elderly populations in the US and China. METHOD We analyzed data from the National Health and Nutrition Examination Survey (NHANES) and the China Health and Retirement Longitudinal Study (CHARLS), including 7,565 and 12,932 participants aged 45 years and older, respectively. The participants were categorized into quartiles based on natural log-transformed RCII (lnRCII) values. Kaplan-Meier survival analysis, Cox proportional hazards models, restricted cubic splines (RCS) and mediation analysis were used to examine the relationship between lnRCII and mortality outcomes, adjusting for potential covariates. RESULT The mean age of the participants was 59.90 ± 10.44 years (NHANES) and 58.64 ± 9.78 years (CHARLS), with 53.28% and 52.50% female, respectively. Kaplan-Meier survival analysis showed that higher lnRCII quartiles (≥ 0.79 in NHANES, ≥ -0.13 in CHARLS) were significantly associated with increased all-cause mortality risk (p < 0.001). Each standard deviation (SD) increase in lnRCII corresponded to a higher risk of all-cause mortality, and the hazard ratios (HRs) and 95% confidence interval (CI) were 1.29 (95% CI: 1.21-1.36) in NHANES and 1.26 (95% CI: 1.15-1.38) in CHARLS. In NHANES, lnRCII was also associated with elevated risks of cardiovascular mortality (HR = 1.21, 95% CI: 1.08-1.35) and cancer mortality (HR = 1.30, 95% CI: 1.09-1.55). RCS analysis indicated a J-shaped relationship between lnRCII and both all-cause and cardiovascular mortality, and a linear association with cancer mortality. Mediation analysis showed that systolic blood pressure and fasting plasma glucose partially mediated these associations. Subgroup analyses suggested a stronger association between lnRCII and all-cause mortality in middle-aged US participants (p for interaction = 0.010). CONCLUSIONS Elevated RCII levels are significantly associated with increased all-cause mortality risk middle-aged and elderly populations in both the US and China. In the US population, RCII is also associated with increased risks of cardiovascular and cancer mortality. By integrating metabolic and inflammatory risk factors, RCII may serve as a valuable tool for mortality risk stratification and clinical decision-making.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Lei Bi
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Qing Li
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Qiuyu Wang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Tingting Lv
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China.
| | - Ping Zhang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China.
| |
Collapse
|
52
|
Glenn AJ, Tessier AJ, Kavanagh ME, Morgan GA, Clish CB, Salas-Salvado J, Malik VS, Hanley AJ, Bazinet RP, Comelli EM, El-Sohemy A, Liu S, Boucher BA, Kendall CWC, Jenkins DJA, Hu FB, Sievenpiper JL. Metabolomic profiling of a cholesterol lowering plant-based diet from two randomized controlled feeding trials. Eur J Clin Nutr 2025:10.1038/s41430-025-01625-x. [PMID: 40263496 DOI: 10.1038/s41430-025-01625-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Objective biomarkers of diet, such as metabolomics, may improve dietary assessment and provide additional insight into how diet influences disease risk. The portfolio diet, a cholesterol-lowering plant-based diet, is recommended for lowering low-density lipoprotein cholesterol (LDL-C). This diet is low in saturated fat and includes nuts, plant protein (legumes), viscous fiber, and phytosterols. OBJECTIVE We examined metabolomic profiles in response to the portfolio diet in two randomized controlled trials (RCTs), where all foods were provided to the participants, compared to a control vegetarian diet and the same control diet with a statin. METHODS The first RCT included 34 adults (age 58.4 ± 8.6 y) and the second RCT included 25 adults (age 61.0 ± 9.6 y), all with high LDL-C (>4.1 mmol/L). Plasma samples were obtained at baseline, week 2, and week 4 in both RCTs for metabolomics analysis using liquid chromatography-tandem mass spectrometry. Linear mixed models were used to examine effects of the interventions on the metabolites in each RCT, applying a Bonferroni correction. RESULTS Of 496 known metabolites, 145 and 63 metabolites significantly changed within the portfolio diet interventions in the first and second RCT, respectively. The majority were glycerophosphocholines (32%), triacylglycerols (20%), glycerophosphoethanolamines (14%), sphingomyelins (8%), and amino acids and peptides (8%) in the first RCT, and glycerophosphocholines (48%), glycerophosphoethanolamines (17%), and amino acids and peptides (8%) in the second RCT. Fifty-two metabolites were consistently changed in the same direction with the portfolio diet intervention across both RCTs, after Bonferroni correction. CONCLUSIONS Many of these metabolites likely reflect the plant-based nature, low saturated fat content, and cholesterol-lowering effects of the diet, such as increased N2-acetylornithine, L-pipecolic acid, lenticin, and decreased C18:0 lipids and cholesteryl esters. Further research is needed to validate these metabolites as biomarkers of a plant-based dietary pattern.
Collapse
Affiliation(s)
- Andrea J Glenn
- Department of Nutrition and Food Studies, New York University, New York, NY, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada.
| | - Anne-Julie Tessier
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- Montreal Heart Institute, Montreal, QC, Canada
- Institut de Valorisation des Données (IVADO), Montreal, QC, Canada
| | - Meaghan E Kavanagh
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
| | - Gloria A Morgan
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
- School of Nutrition, Toronto Metropolitan University, Toronto, ON, Canada
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jordi Salas-Salvado
- CIBER Fisiopatologıa de la Obesidad y Nutricion (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institut d'Investigacions Sanitàries Pere i Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Vasanti S Malik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Simin Liu
- Department of Epidemiology and Biostatistics, Joe C. Wen School of Population & Public Health, UC Irvine, Irvine, CA, USA
- Center for Global Cardiometabolic Health & Nutrition (CGCHN), Mary & Steve Wen Division of Cardiology, Department of Medicine, School of Medicine, UC Irvine, Irvine, CA, USA
- Division of Endocrinology Department of Medicine, and Division of Cardiothoracic Surgery Department of Surgery, Warren Alpert School of Medicine and Rhode Island Hospital, Providence, RI, USA
- Department of Epidemiology, Brown University, School of Public Health, Providence, RI, USA
| | - Beatrice A Boucher
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Cyril W C Kendall
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - David J A Jenkins
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - John L Sievenpiper
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada.
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada.
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
53
|
Jiménez MDCFF. Plant-Based Diet and Erectile Dysfunction: A Narrative Review. J Nutr 2025:S0022-3166(25)00229-9. [PMID: 40274235 DOI: 10.1016/j.tjnut.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Evidence shows that the consumption of plant foods, particularly those in their whole form (fruits, vegetables, whole grains, nuts, seeds, and legumes from which no edible part has been removed), improves cardiometabolic risk factors and is associated with reduced risk of cardiovascular diseases (CVDs), diabetes, cancer, chronic kidney disease and mortality compared with animal (meat, fish, eggs and dairy) and nonwhole plant foods (sugar-sweetened beverages, refined grains, etc.). Erectile dysfunction (ED) is considered a strong predictor of CVD. The underlying defect in arteriogenic ED is endothelial dysfunction. A plant-based diet focused on whole plant foods could enhance penile erection as it improves endothelial function through various mechanisms. First, it provides nitrates, L-arginine, and L-citrulline, substrates for nitric oxide production. In addition, this diet lowers low-density lipoprotein cholesterol, trimethylamine N-oxide, postprandial triglycerides, advanced glycation end product, inflammation, and vasoconstrictors levels, contributing to higher nitric oxide concentrations, increased endothelial progenitor cells preservation and decreased arterial stiffness. This review explores the epidemiological evidence of a plant-based diet emphasizing whole plant foods on ED and the potential biological pathways involved.
Collapse
|
54
|
Luo Y, Liu J, Qu P, Han S, Li X, Wang Y, Su X, Zeng J, Li J, Deng S, Liang Q, Hou L, Cheng P. The crosstalk of breast cancer and ischemic heart disease. Cell Death Discov 2025; 11:185. [PMID: 40251177 PMCID: PMC12008236 DOI: 10.1038/s41420-025-02428-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
In recent years, the continuous optimization of anti-tumor therapy has greatly improved the cancer-specific survival rate for patients with breast cancer (BC). The prevention and treatment of breast cancer-related heart diseases have become a new breakthrough in improving the long-term survival for BC patient. The cardiac damages caused by BC treatment are increasingly prominent among BC patients, of which ischemic heart disease (IHD) is the most prominent. Besides, the systemic inflammatory response activated by tumor microenvironment c an induce and exacerbate IHD and increase the risk of myocardial infarction (MI). Conversely, IHD can also exert detrimental effects on tumors. MI not only increases the risk of BC, but also induces specialized immune cell to BC and accelerates the progression of BC. Meanwhile, the treatment of IHD can also promote BC metastasis and transition to more aggressive phenotypes. Although BC and IHD are diseases of two independent systems, their crosstalk increases the difficulty of anti-cancer treatment and IHD management, which reduces the survival for both diseases. Therefore, this review mainly explores the mutual influence and underlying mechanisms between BC and IHD, aiming to provide insights for improving the long-term survival for patients with BC or IHD.
Collapse
Affiliation(s)
- Yunbo Luo
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Jun Liu
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Peng Qu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
| | - Shiqi Han
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Xue Li
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
| | - Yali Wang
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Xiaohan Su
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Jiao Zeng
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Jinsui Li
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Shishan Deng
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Qi Liang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China.
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China.
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China.
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, 610072, P.R. China.
| |
Collapse
|
55
|
Imannezhad M, Kamrani F, Shariatikia A, Nasrollahi M, Mahaki H, Rezaee A, Moohebati M, Shahri SHH, Darroudi S. Association of atherogenic indices and triglyceride-total cholesterol-body weight index (TCBI) with severity of stenosis in patients undergoing angiography: a case-control study. BMC Res Notes 2025; 18:180. [PMID: 40247425 PMCID: PMC12004764 DOI: 10.1186/s13104-025-07203-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/26/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVES Coronary artery disease, caused by atherosclerosis, necessitates assessing plaque formation risk using indices like the atherogenic index of plasma (AIP), Castelli's risk indexes (CRI-I and CRI-II), the atherogenic coefficient (AC), and the triglyceride-total cholesterol-body weight index (TCBI). Although TCBI primarily assesses mortality risk, its relationship with stenosis severity is unclear. Utilizing data from a prior study, a case-control analysis was conducted on 1,187 subjects, which included 781 patients who underwent coronary angiography and 406 healthy controls. The indices were compared across varying degrees of arterial blockages. RESULTS AIP significantly correlated with stenosis severity in women, increasing the risk of three-vessel stenosis by 2.5 times. AC raised the risk of single-vessel stenosis in men by 2.7 times. CRI-I and CRI-II showed a positive relationship with arterial stenosis in women, with CRI-I increasing the risk of two and three-vessel blockages by 21.9% and 22.4%, respectively. A one-unit increase in CRI-II raised the risk by 33.1% for two arteries and 25.3% for three. In conclusion, AIP, CRI-I, and CRI-II in women, along with AC in men, correlated with arterial stenosis severity, while TCBI did not. Further research is needed to determine which index is most effective in predicting CAD risk.
Collapse
Affiliation(s)
- Mobina Imannezhad
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzam Kamrani
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shariatikia
- Department of Cardiovascular, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Nasrollahi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Hanie Mahaki
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran
| | - Ali Rezaee
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohsen Moohebati
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran
| | | | - Susan Darroudi
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran.
| |
Collapse
|
56
|
Shen H, Huang A, Wang B, Huang X, Chen R. Clinical value of Lp-PLA2, LDL-C, HDL-C, hs-CRP, leukocyte, FPG and HbA1c in type 2 diabetes mellitus patients with acute ischemic stroke. Front Endocrinol (Lausanne) 2025; 16:1546961. [PMID: 40313488 PMCID: PMC12043486 DOI: 10.3389/fendo.2025.1546961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Background To evaluate the clinical significance of human plasma lipoprotein-associated phospholipase A2 (Lp-PLA2), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), high-sensitivity C-reactive protein (hs-CRP), leukocyte count, fasting plasma glucose (FPG), and glycated hemoglobin (HbA1c) in patients with Type 2 diabetes mellitus (T2DM) and acute ischemic stroke (AIS). Methods A total of 155 T2DM patients with AIS, admitted to the Second Affiliated Hospital of Shantou University Medical College between October 2023 and October 2024, were included in the stroke group. Additionally, 86 T2DM subjects from the same period were included in the T2DM control group. Serum levels of Lp-PLA2, LDL-C, HDL-C, hs-CRP, leukocyte count, FPG, and HbA1c were compared between the T2DM-AIS group and the T2DM control group, as well as among T2DM-AIS patients with different infarct sizes and degrees of neurological impairment. The clinical value of the above indexes in the diagnosis of T2DM with AIS was analyzed by receiver operating characteristic curve (ROC curve). Results Serum levels of Lp-PLA2 (142.9 [115.8, 178.3] ng/L), LDL-C (3.4 [2.6, 4.2] mmol/L), hs-CRP (3.6 [1.5, 11.2] mg/L), leukocytes (8.0 [6.8, 10.3] × 10^9/L), FPG (10.2 [7.5, 14.4] mmol/L), and HbA1c (10.2 [7.5, 14.4] %) were significantly higher in the T2DM-AIS group compared to the T2DM control group (Lp-PLA2: 102.1 [76.6, 121.9] ng/L, LDL-C: 3.2 [2.4, 3.7] mmol/L, hs-CRP: 2.4 [0.9, 5.2] mg/L, leukocytes: 7.0 [6.1, 8.1] × 10^9/L, FPG: 7.4 [6.1, 10.3] mmol/L, HbA1c: 7.0 [6.4, 8.2] %). In contrast, serum HDL-C levels (1.1 [0.9, 1.3] mmol/L) were significantly lower than those in the control group (1.3 [1.1, 1.5] mmol/L), which correlated with larger infarct size and greater neurological injury. ROC curve analysis indicated that the combined use of seven tests had an AUC of 0.906, with a sensitivity of 77.40% and a specificity of 95.30%. Conclusion Monitoring serum levels of Lp-PLA2, LDL-C, HDL-C, hs-CRP, leukocyte count, FPG, and HbA1c provides a comprehensive assessment of cerebral infarction in T2DM patients with AIS and serves as auxiliary indicators for evaluating disease severity.
Collapse
Affiliation(s)
- Huimin Shen
- Department of Brain Center, People’s Hospital of Chenghai District, Shantou, Guangdong, China
| | - Anyan Huang
- Department of Health Care, Shantou Maternal and Child Health Hospital, Shantou, Guangdong, China
| | - Bingxin Wang
- Department of Brain Center, People’s Hospital of Chenghai District, Shantou, Guangdong, China
| | - Xiaohua Huang
- Department of Pediatrics, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ruiqian Chen
- Department of Information, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
57
|
Yang Y, Huang L, Gu Y, Wang Z, Liu S, Chen Q, Ning W, Hong G. Predicting cerebral infarction and transient ischemic attack in healthy individuals and those with dysmetabolism: a machine learning approach combined with routine blood tests. Sci Rep 2025; 15:13044. [PMID: 40240412 PMCID: PMC12003726 DOI: 10.1038/s41598-025-94682-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Ischemic cerebral infarction is the most prevalent type of stroke, causing significant disability and death worldwide. Transient ischemic attack (TIA) is a strong predictor of subsequent stroke. Individuals with dysmetabolism, such as hypertension, hypercholesterolemia, and diabetes, are at increased risk for cerebral infarction (CI) and TIA. In resource-limited settings, diagnosing CI and TIA can be particularly difficult due to a lack of advanced imaging and specialized expertise. Therefore, we aim to develop a simple, convenient, blood-based approach that could assist clinicians in diagnosing CI and TIA, especially in regions where advanced imaging or stroke-specific expertise is limited. All study subjects were patients admitted to the First Hospital of Xiamen University and healthy check-up populations between January 2018 and September 2023. This study employed five machine learning methods alongside 21 blood routine indicators, 30 blood biochemical indicators, age, and gender to construct predictive models for CI and TIA in both healthy individuals and those with dysmetabolism. The Area Under the Receiver Operating Characteristic (ROC) Curve (AUC) served as the metric to assess the comprehensive predictive capability of the models. Subsequently, the SHAP package was employed for model interpretation. Extreme Gradient Boosting (XGBoost) outperforms other models in all predictive models. In the models predicting CI and TIA among healthy, the AUC is 0.9958 (0.9947-0.9969) and 0.9928 (0.9899-0.9951), respectively. Among the nine shared key features of the two models are indicators of glucose metabolism, lipid metabolism, and liver metabolism. In the models for predicting CI and TIA among patients with hypertension, hypercholesterolemia, diabetes, and those with all three metabolic disorders combined, the AUCs ranged from 0.6990 to 0.8591. We found that the indicators K significantly contributed to predict CI and TIA from those with dysmetabolism. Additionally, metabolic-related indicators, such as glucose (GLU) and high-density lipoprotein cholesterol (HDL-C), are ranked highly among the top ten contributing features. XGBoost performed the best in all models. It can effectively differentiate CI and TIA from healthy and dysmetabolic patients by combining blood routine and blood biochemical indicators. Moreover, it can also differentiate CI from TIA. Although any suspicious findings from this model would still require confirmatory imaging, the simplicity and low cost of blood-based testing may offer a practical adjunct for clinicians-particularly in areas lacking advanced imaging or extensive stroke expertise-and could facilitate earlier diagnostic decision-making.
Collapse
Affiliation(s)
- Yunyun Yang
- Department of Laboratory Medicine, Xiamen Key Laboratory of Genetic Testing, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Lindan Huang
- Department of Laboratory Medicine, Xiamen Key Laboratory of Genetic Testing, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- School of Public Health, Xiamen University, Xiamen, 361003, Fujian, China
| | - Ying Gu
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Zhicheng Wang
- Department of Laboratory Medicine, Xiamen Key Laboratory of Genetic Testing, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Department of Otolaryngology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Shuai Liu
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Qun Chen
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Wanshan Ning
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
| | - Guolin Hong
- Department of Laboratory Medicine, Xiamen Key Laboratory of Genetic Testing, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
- School of Public Health, Xiamen University, Xiamen, 361003, Fujian, China.
| |
Collapse
|
58
|
Zare P, Bideshki MV, Sohrabi Z, Behzadi M, Sartang MM. Effect of Dietary Approaches to Stop Hypertension (DASH) diet on lipid profile in individuals with overweight/ obesity: A GRADE-assessed systematic review and meta-analysis of clinical trials. Nutr Metab Cardiovasc Dis 2025:104057. [PMID: 40268568 DOI: 10.1016/j.numecd.2025.104057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND AND AIM Obesity is a major nutritional disease that increases the risk of developing serious health conditions like dyslipidemia. Plant-based diets, like DASH (dietary approaches to stop hypertension), can help lower the risk of dyslipidemia. However, evidence on the effect of DASH diet on lipid profile in populations with overweight/obesity is inconsistent. This meta-analysis of controlled trials investigated the effects of the DASH diet on lipid profile in individuals with overweight/obesity. METHODS AND RESULTS A search for relevant studies was conducted in databases like PubMed, Web of Science, and Scopus until January 2024. The calculation of weighted-mean differences (WMDs) and 95 % confidence intervals (CIs) was performed based on the random-effects model. Sensitivity, meta-regression and publication bias analyses were also conducted. 22 eligible studies with 26 arms and 3562 participants were included. DASH diet significantly reduced total cholesterol (TC) (WMD: 5.05 mg/dl, 95 % CI: 8.78, -1.31, p = 0.008), low-density lipoprotein cholesterol (LDL-C) (WMD: 5.33 mg/dl, 95 % CI: 8.54, -2.11, p = 0.001) and very low-density lipoprotein cholesterol (VLDL-C) (WMD: 3.26, 95 % CI: 6.19, -0.34, p = 0.029) levels. Greater reductions were observed in studies with durations ≤8 weeks. All of the included studies were classified as high quality except two, which were classified as moderate quality. LDL-C and VLDL-C were categorized as high-grade evidence, while others were categorized as moderate. CONCLUSIONS DASH diet could improve the lipid profile of individuals with overweight/obesity by decreasing TC, LDL-C and VLDL-C levels. However, it doesn't have significant effects on high-density lipoprotein cholesterol and triglyceride levels.
Collapse
Affiliation(s)
- Paniz Zare
- Student Research Committee, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Vesal Bideshki
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Diet Therapy, School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Sohrabi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Behzadi
- Student Research Committee, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Mohammadi Sartang
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
59
|
Zhang Z, Liu C, Zhao L, Tan X, Yu X, Wang J, Yao J. Non-High-Density Lipoprotein-to-High-Density Lipoprotein Cholesterol Ratio as a Predictive Biomarker for Diabetic Retinopathy Risk: A Population-Based Analysis of US Adults. Ophthalmic Res 2025; 68:301-309. [PMID: 40228483 DOI: 10.1159/000545816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
INTRODUCTION Diabetic retinopathy (DR) is a severe complication of diabetes, and lipid imbalances play a key role in its progression. The non-high-density lipoprotein cholesterol-to-high-density lipoprotein cholesterol ratio (NHHR) has been identified as a predictor of cardiovascular diseases, but its link to DR remains unclear. This study aimed to assess the association between NHHR and DR risk in diabetic patients. METHODS Data from the 2005-2018 National Health and Nutrition Examination Survey (NHANES) were analyzed. Multivariate logistic regression models were used to evaluate the relationship between NHHR and DR. Nonlinear associations were assessed using restricted cubic spline analysis. RESULTS Of the 4,935 participants, 1,193 had DR. Higher NHHR was strongly associated with increased DR risk. Each unit rise in NHHR increased the risk by 19% (OR = 1.19, 95% CI: 1.07-1.31, p < 0.05). In quartile analysis, participants in the highest NHHR quartile had nearly double the risk of DR compared to those in the lowest quartile (OR = 1.84, 95% CI: 1.62-2.06, p < 0.001). Subgroup analysis showed this association was consistent across different demographic groups, including age, gender, BMI, and smoking status. CONCLUSION NHHR is significantly linked to DR risk in diabetic patients and may be a valuable biomarker for early detection and prevention strategies in clinical settings.
Collapse
Affiliation(s)
- Zhirui Zhang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China,
| | - Lingying Zhao
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xufang Tan
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ximing Yu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiadi Wang
- The First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Yao
- The First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
60
|
Zaman S, Wasfy JH, Kapil V, Ziaeian B, Parsonage WA, Sriswasdi S, Chico TJA, Capodanno D, Colleran R, Sutton NR, Song L, Karam N, Sofat R, Fraccaro C, Chamié D, Alasnag M, Warisawa T, Gonzalo N, Jomaa W, Mehta SR, Cook EES, Sundström J, Nicholls SJ, Shaw LJ, Patel MR, Al-Lamee RK. The Lancet Commission on rethinking coronary artery disease: moving from ischaemia to atheroma. Lancet 2025; 405:1264-1312. [PMID: 40179933 DOI: 10.1016/s0140-6736(25)00055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/01/2025] [Accepted: 01/09/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Sarah Zaman
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Jason H Wasfy
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikas Kapil
- William Harvey Research Institute, Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| | - Boback Ziaeian
- Division of Cardiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | - William A Parsonage
- Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, QLD, Australia; Department of Cardiology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sira Sriswasdi
- Center of Excellence in Computational Molecular Biology, Chulalongkorn University, Pathum Wan, Bangkok, Thailand; Faculty of Medicine, Chulalongkorn University, Pathum Wan, Bangkok, Thailand
| | - Timothy J A Chico
- School of Medicine and Population Health, University of Sheffield, Sheffield, UK; British Heart Foundation Data Science Centre, Health Data Research UK, London, UK
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico, University of Catania, Catania, Italy
| | - Róisín Colleran
- Department of Cardiology and Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Nadia R Sutton
- Department of Internal Medicine, and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lei Song
- Department of Cardiology, National Clinical Research Centre for Cardiovascular Diseases, Fuwai Hospital, Beijing, China; Peking Union Medical College (Chinese Academy of Medical Sciences), Beijing, China
| | - Nicole Karam
- Cardiology Department, European Hospital Georges Pompidou, Paris City University, Paris, France
| | - Reecha Sofat
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Chiara Fraccaro
- Division of Cardiology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Daniel Chamié
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mirvat Alasnag
- Cardiac Center, King Fahd Armed Forces Hospital, Jeddah, Saudi Arabia
| | | | - Nieves Gonzalo
- Cardiology Department, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Walid Jomaa
- Cardiology B Department, Fattouma Bourguiba University Hospital, University of Monastir, Monastir, Tunisia
| | - Shamir R Mehta
- Population Health Research Institute, Hamilton Health Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Elizabeth E S Cook
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Johan Sundström
- Uppsala University, Uppsala, Sweden; The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | | | - Leslee J Shaw
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manesh R Patel
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Rasha K Al-Lamee
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
61
|
Jiang R, Ruan H, Zhang W, Chen J, Yang Y, Tang S, Wang Y, Ruan X, Cao J, Zhu H, Xie N, Liu Y, Tan N. Association between neutrophil-lymphocyte ratio levels and mortality related to cardiovascular cause and all causes in coronary artery disease patients with low-density lipoprotein cholesterol below 1.4 mmol/L: A multicenter cohort study. Nutr Metab Cardiovasc Dis 2025:104058. [PMID: 40268567 DOI: 10.1016/j.numecd.2025.104058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND AND AIMS Neutrophil-lymphocyte ratio (NLR) can reflect the residual risk of patients with coronary atherosclerotic disease (CAD). However, in CAD patients with low density lipoprotein cholesterol (LDL-C) less than 1.4 mmol/L, the relationship between NLR and poor prognosis remains unclear. The purpose of this research is to examine the relationship between NLR and mortality from cardiovascular causes as well as all causes in the study group. METHODS AND RESULTS From the beginning of 2007 to the end of 2020, 2749 individuals with LDL-C under 1.4 mmol/L at baseline and CAD diagnosed via coronary angiography were part of this research. The selected population was divided into four groups according to the level of NLR, and Cox regression model was used mainly to evaluate the association between NLR and cardiovascular and all-cause mortality. After a follow-up of median 6 years, a total of 182 cardiovascular deaths and 460 all-cause deaths had occurred. The Cox regression analysis of fully adjusted model showed that an elevated NLR is connected to a higher risk of mortality from cardiovascular cause (HR: 2.18, 95 %CI: 1.38-3.43) and all causes (HR: 1.33, 95 %CI: 1.02-1.74) in the fourth quartile group. Subgroup analysis indicated that younger and middle-aged patients, as well as those with multivessel coronary artery disease, may have a heightened residual risk of inflammation. CONCLUSION NLR can be a useful risk factor indicator for cardiovascular and all-cause mortality in CAD patients with LDL-C levels below 1.4 mmol/L, and it also points to residual risk.
Collapse
Affiliation(s)
- Rengui Jiang
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), Ganzhou, 341000, China
| | - Huangtao Ruan
- Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Weipeng Zhang
- School of Medicine South China University of Technology, Guangzhou, 510100, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China
| | - Jinming Chen
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yupeng Yang
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Shangyi Tang
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yueting Wang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), Ganzhou, 341000, China
| | - Xianlin Ruan
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jun Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), Ganzhou, 341000, China
| | - Hengqing Zhu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), Ganzhou, 341000, China
| | - Nianjin Xie
- Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), Ganzhou, 341000, China.
| | - Yong Liu
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Ning Tan
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510515, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| |
Collapse
|
62
|
Worledge EA, Billups SJ, Titus OJ, Saseen JJ. Achievement of low-density lipoprotein cholesterol thresholds in very high-risk atherosclerotic cardiovascular disease. J Clin Lipidol 2025:S1933-2874(25)00263-6. [PMID: 40360376 DOI: 10.1016/j.jacl.2025.04.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND The 2022 American College of Cardiology Expert Consensus Decision Pathway on the Role of Nonstatin Therapies recommended more aggressive management of low-density lipoprotein cholesterol (LDL-C) compared to the 2018 American Heart Association/American College of Cardiology/Multisociety Guideline. In congruence with European guidelines, a target LDL-C threshold of 55 mg/dL is recommended for patients with very high-risk atherosclerotic cardiovascular disease (ASCVD). Real-world status of the early implementation of this recommendation remains uncertain. OBJECTIVE To determine the proportion of patients with very high-risk ASCVD achieving an LDL-C <55 mg/dL and characterize differences between patients who did and did not achieve this LDL-C value. METHODS This retrospective cohort study evaluated patients between January 1, 2023, and December 31, 2023, who were ≥ 18 years old with very high-risk ASCVD from the University of Colorado Health system primary care practices. Very high-risk ASCVD was defined as a history of a major ASCVD event with at least 2 high-risk conditions. RESULTS A total of 8974 patients met inclusion criteria for very high-risk ASCVD. Of these, only 21.0% achieved an LDL-C <55 mg/dL, while 39.5% of patients achieved the previously recommended LDL-C threshold of <70 mg/dL. More than half of patients were prescribed a high-intensity statin-based regimen. CONCLUSION Within a large, academic health-system, achievement of LDL-C threshold among patients with very high-risk ASCVD was low. Recommendations from the 2022 ACC Expert Consensus Decision Pathway that endorse more aggressive LDL-C lowering and emphasize nonstatin therapies as an adjunct to statins are not fully implemented in real-world clinical practice.
Collapse
Affiliation(s)
- Elisa A Worledge
- Department of Clinical Pharmacy, University of Colorado Anschutz Medical Campus Skaggs School of Pharmacy, Aurora, CO (Drs Worledge, Billups and Saseen, and Mr. Titus)
| | - Sarah J Billups
- Department of Clinical Pharmacy, University of Colorado Anschutz Medical Campus Skaggs School of Pharmacy, Aurora, CO (Drs Worledge, Billups and Saseen, and Mr. Titus)
| | - Oliver J Titus
- Department of Clinical Pharmacy, University of Colorado Anschutz Medical Campus Skaggs School of Pharmacy, Aurora, CO (Drs Worledge, Billups and Saseen, and Mr. Titus)
| | - Joseph J Saseen
- Department of Clinical Pharmacy, University of Colorado Anschutz Medical Campus Skaggs School of Pharmacy, Aurora, CO (Drs Worledge, Billups and Saseen, and Mr. Titus); Department of Family Medicine, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO (Dr Saseen).
| |
Collapse
|
63
|
Akiyama Y, Katsuki S, Koga Y, Yamamoto M, Hironaga K, Suematsu N, Miyata K, Mukai Y, Inoue S, Nishi JI, Tashiro H, Nakano Y, Funakoshi K, Tagawa K, Ichi I, Tsutsui H, Abe K, Matoba T. Effect of PCSK9 inhibitor usage on coronary endothelial dysfunction in patients with hypercholesterolemia after coronary stenting: The CuVIC-2 trial. J Cardiol 2025:S0914-5087(25)00101-7. [PMID: 40221085 DOI: 10.1016/j.jjcc.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND The effects of evolocumab on coronary endothelial dysfunction (CED), a hallmark of atherogenesis, are unknown. The aim of this study was to investigate whether evolocumab, in combination with high-dose statins, could ameliorate CED in patients who underwent coronary stenting. METHODS The CuVIC-2 trial was a multicenter randomized controlled trial. CED was defined as intracoronary acetylcholine (ACh)-induced contractile responses with signs of myocardial ischemia. We originally intended to enroll 160 participants but altered the study design due to the COVID-19 pandemic and then recruited 41 participants. The revised primary endpoint was the coronary contraction rate in response to ACh assessed in a core laboratory, ensuring a statistical power of over 80 % using the mixed model for repeated measures. RESULTS The evolocumab in combination with high-dose statins with or without ezetimibe (EV + S) group included 19 males and 4 females aged 62 ± 13 years. The high-dose statins with or without ezetimibe (S) group included 13 males and 5 females aged 64 ± 11 years. Compared with the S group, the EV + S group presented a significantly greater decrease in low-density lipoprotein cholesterol at 28 weeks; 83 ± 17 to 20 ± 16 mg/dL (-76 % from the baseline) in the EV + S group and 88 ± 16 to 81 ± 20 mg/dL (-7 % from the baseline) in the S group (p < 0.0001). At 28 weeks, there was no difference between the two groups in terms of the coronary artery constriction rate across all doses [mean difference: 4.8 % (95 % CI: -13.6 to 23.2); p = 0.6]. CONCLUSIONS Amelioration of CED by evolocumab was not observed in this trial with several limitations.
Collapse
Affiliation(s)
- Yusuke Akiyama
- Department of Cardiovascular Medicine, Oita Prefectural Hospital, Oita, Japan; Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yasuaki Koga
- Department of Cardiovascular Medicine, Oita Prefectural Hospital, Oita, Japan
| | - Mitsutaka Yamamoto
- Department of Cardiovascular Medicine, Harasanshin Hospital, Fukuoka, Japan
| | - Kiyoshi Hironaga
- Department of Cardiovascular Medicine, Fukuoka City Hospital, Fukuoka, Japan
| | - Nobuhiro Suematsu
- Department of Cardiovascular Medicine, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - Kenji Miyata
- Department of Cardiovascular Medicine, Japan Community Health Care Organization, Kyushu Hospital, Fukuoka, Japan
| | - Yasushi Mukai
- Department of Cardiovascular Medicine, Japanese Red Cross Fukuoka Hospital, Fukuoka, Japan
| | - Shujiro Inoue
- Department of Cardiovascular Medicine, National Hospital Organization Kyushu Medical Centre, Fukuoka, Japan
| | - Jun-Ichiro Nishi
- Department of Cardiovascular Medicine, Iizuka Hospital, Fukuoka, Japan
| | - Hideki Tashiro
- Department of Cardiovascular Medicine, St. Mary's Hospital, Fukuoka, Japan
| | - Yasuhiro Nakano
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kouta Funakoshi
- Center for Clinical and Translational Research of Kyushu University Hospital, Fukuoka, Japan
| | - Koshiro Tagawa
- Center for Clinical and Translational Research of Kyushu University Hospital, Fukuoka, Japan
| | - Ikuyo Ichi
- Graduate School of Humanities and Science, Ochanomizu University, Tokyo, Japan
| | - Hiroyuki Tsutsui
- School of Medicine and Graduate School, International University of Health and Welfare, Fukuoka, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.
| |
Collapse
|
64
|
Li Y, Liu X, Li Q, Zhou P, Chen Q, Jiang B, Zhu T. Association of helicobacter pylori infection with lipid metabolism and 10-year cardiovascular risk in diabetes mellitus: A cross-sectional study. PLoS One 2025; 20:e0319688. [PMID: 40203057 PMCID: PMC11981135 DOI: 10.1371/journal.pone.0319688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/05/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Previous studies have shown that Helicobacter pylori infection is not only a risk factor for gastrointestinal diseases but also associated with various non-digestive conditions. This study aimed to investigate the effect of Helicobacter pylori infection on the risk of lipid metabolism disorders and cardiovascular disease in individuals with diabetes mellitus. METHODS This cross-sectional study was conducted at a health examination center. Data from life questionnaires, laboratory tests, the carbon-13 urea breath test, and the Framingham Risk Score were collected from 266 patients with diabetes. All participants were categorized into Helicobacter pylori-uninfected and Helicobacter pylori-infected groups based on the carbon-13 urea breath test results. Differences in lipid levels, Framingham Risk Score, and cardiovascular disease risk were compared between the two groups. A logistic regression model was applied to analyze whether Helicobacter pylori infection is an independent risk factor for dyslipidemia in patients with diabetes. RESULTS Total cholesterol and low-density lipoprotein cholesterol levels were higher in the Helicobacter pylori-infected group than in the uninfected group, and high-density lipoprotein cholesterol levels were lower in the infected group (both P < 0.05). There was no statistically significant difference in triglyceride levels between the two groups. Regression analysis showed that Helicobacter pylori infection was an independent risk factor for dyslipidemia in patients with diabetes (P < 0.05). The Framingham Risk Score and 10-year cardiovascular disease risk were higher in the Helicobacter pylori-infected group compared with the uninfected group (P < 0.001). CONCLUSION Helicobacter pylori infection is associated with dyslipidemia and may contribute to an increased risk of cardiovascular disease in individuals with diabetes.
Collapse
Affiliation(s)
- Yuexi Li
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Xiaoqin Liu
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Qing Li
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Peng Zhou
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Qian Chen
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Bolan Jiang
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Taiju Zhu
- Health Management Center, Deyang People’s Hospital, Deyang, Sichuan, China
| |
Collapse
|
65
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
66
|
Di Giovanni G, Fujino M, Kataoka Y, Butters J, Hucko T, Puri R, Nissen SE, Nelson AJ, Psaltis PJ, Nicholls SJ. Impact of evolocumab on plaque phenotypic changes in patients with acute coronary syndrome and elevated lipoprotein(a) levels: a HUYGENS secondary analysis. Eur J Prev Cardiol 2025:zwaf211. [PMID: 40424182 DOI: 10.1093/eurjpc/zwaf211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 05/29/2025]
Abstract
AIMS The proprotein convertase subtilisin/kexin Type 9 inhibitor, evolocumab, promoted plaque stabilization on serial imaging in patients following an acute coronary syndrome. The impact of evolocumab in patients with varying lipoprotein(a) [Lp(a)] levels is unknown. METHODS AND RESULTS Serial optical coherence tomography imaging was performed to evaluate changes in plaque composition in response to treatment with evolocumab 420 mg or placebo for 50 weeks. The current post hoc analysis compared demographics, biochemistry, and plaque imaging changes in those with baseline Lp(a) levels <125 (n = 71) and ≥125 nmol/L (n = 46). Among those with high Lp(a) levels, evolocumab treatment produced lower levels of LDL cholesterol (LDL-C) (21.7 ± 10.3 vs. 94.5 ± 22.9 mg/dL; P < 0.001) and Lp(a) [156.0 (136.0, 187.0) vs. 204.0 (170.5, 290.5) nmol/L; P = 0.007], compared with placebo. Changes in minimum fibrous cap thickness (FCT) (+51.6 ± 40.9 vs. +12.4 ± 23.9 μm; P < 0.001) and lipid arc (-60.9 ± 56.5° vs. -9.1 ± 70.8°; P = 0.008) were greater in the high Lp(a) group with evolocumab compared with placebo. Among patients with low Lp(a) levels, evolocumab produced lower levels of LDL-C (23.3 ± 34.9 vs. 82.9 ± 46.5 mg/dL; P < 0.001) and Lp(a) [11.5 (5.8, 23.8) vs. 25.0 (13.5, 41.0) nmol/L; P = 0.01] compared with placebo, but no differences were observed between groups in changes in minimum FCT (+45.9 ± 37.8 vs. +34.7 ± 36.0 μm; P = 0.21) and lipid arc (-59.9 ± 50.1° vs. -44.5 ± 46.1°; P = 0.18). Baseline Lp(a) levels significantly interacted with the impact of evolocumab on changes in minimum FCT (interaction P = 0.04). CONCLUSION The ability of evolocumab to more effectively promote plaque stabilization, compared with statin monotherapy, appears more pronounced in patients with higher Lp(a) levels, suggesting that Lp(a) may help identify those who benefit most from intensive lipid-lowering therapy. REGISTRATION ClinicalTrials.gov: NCT03570697.
Collapse
Affiliation(s)
- Giuseppe Di Giovanni
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne 3168, Australia
| | - Masashi Fujino
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne 3168, Australia
| | - Yu Kataoka
- Department of Cardiology, National Cerebral and Cardiovascular Centre, Suita, Japan
| | - Julie Butters
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne 3168, Australia
| | | | - Rishi Puri
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland Clinic, Cleveland, OH, USA
| | - Steven E Nissen
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland Clinic, Cleveland, OH, USA
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne 3168, Australia
| | - Peter J Psaltis
- Lifelong Health Theme, South Australian Health and Medical Research Institute, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne 3168, Australia
| |
Collapse
|
67
|
Wang L, Xu Y, Chen L, Zhang H. Estimated glucose disposal rate mediates the association between Life's Crucial 9 and congestive heart failure: a population-based study. Front Endocrinol (Lausanne) 2025; 16:1540794. [PMID: 40248150 PMCID: PMC12004698 DOI: 10.3389/fendo.2025.1540794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/04/2025] [Indexed: 04/19/2025] Open
Abstract
Background Life's Crucial 9 (LC9) is the latest indicator of cardiovascular health (CVH), and the estimated glucose disposal rate (eGDR) is a non-invasive indicator of insulin resistance (IR). However, the relationships between LC9 and eGDR and congestive heart failure (CHF) remain unknown. Methods In this cross-sectional study, participants aged ≥20 years in the NHANES database from 2005 to 2018 were analyzed. Weighted linear regression, logistic regression, subgroup analysis, and restricted cubic spline (RCS) analysis were employed to analyze the associations among LC9, eGDR, and CHF. Mediation analysis was used to explore the mediating role of eGDR in the association between LC9 and CHF. Results A total of 22,699 adult participants were included, among whom 661 suffered from CHF. The mean age of the participants was 47.52 (0.26) years old, with 11186 (48.68%) males and 11513 (51.32%) females. The average value of LC9 was 71.16 (0.22), and that of eGDR was 7.91 (0.04). After adjusting for confounding factors, linear regression showed that LC9 was independently and positively associated with eGDR (β: 1.11, 95%CI: 1.07 - 1.14, P < 0.0001). Logistic regression indicated that both LC9 (OR: 0.76, 95%CI: 0.65 - 0.88, P < 0.001) and eGDR (OR: 0.81, 95%CI: 0.76 - 0.86, P < 0.0001) were independently and negatively associated with the prevalence of CHF. Mediation analysis revealed that the association between LC9 and CHF was mainly mediated by eGDR, with a proportion of 66%. Conclusion This study suggests that higher LC9 scores and eGDR values imply a lower prevalence of CHF. Meanwhile, eGDR is the main intermediate factor in the association between LC9 and CHF, indicating that good CVH may reduce the prevalence of CHF by improving IR.
Collapse
Affiliation(s)
- Liping Wang
- Department of Cardiovascular, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yaying Xu
- Department of Endocrinology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Lele Chen
- Henan Provincial People's Hospital, Zhumadian, China
| | - Huifeng Zhang
- Department of Cardiovascular, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
68
|
Wang LW. Non-invasive screening for coronary artery disease: current perspectives, patient, public health and ethical considerations in evaluating symptomatic and asymptomatic individuals. Intern Med J 2025; 55:555-563. [PMID: 40047329 PMCID: PMC11981024 DOI: 10.1111/imj.16585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/30/2024] [Indexed: 04/10/2025]
Abstract
Coronary artery disease (CAD) is a leading cause of morbidity worldwide. Although non-invasive testing for CAD aims at reducing future disease burden, testing can often be associated with significant economic and other health-related costs, at both an individual and societal level. Although there is an established role for screening symptomatic patients for CAD, there is still considerable debate as to the best approach for individuals who are asymptomatic. In this review, various non-invasive tests commonly used in clinical practice will be discussed, including their potential utility, known limitations, and other considerations regarding their use. The use of such testing requires careful consideration of their diagnostic accuracy, availability, cost and patient-specific factors that may limit their utility and safety. Future recommendations for CAD screening, especially for lower-risk or asymptomatic individuals, should offer clinicians and patients some degree of flexibility and take into account the nuanced clinical approach that is often required to address the variability of each individual patient's biopsychosocial context and other factors relating to the suitability and accessibility of screening (e.g. financial cost and geographic location). Recommendations that are well suited to certain geographic locations or societal groups may be less appropriate for other populations, especially those that are marginalised, less well resourced or experiencing significant socioeconomic disadvantage. Screening for CAD should therefore endeavour to ensure equity and aim to improve outcomes in all patient groups, including those who are disadvantaged and most at risk.
Collapse
Affiliation(s)
- Louis W. Wang
- Department of MedicineSydney HospitalSydneyNew South WalesAustralia
- Department of Vascular MedicineSt Vincent's HospitalSydneyNew South WalesAustralia
- St Vincent's & Mater Clinical School, University of Notre Dame AustraliaSydneyNew South WalesAustralia
- St Vincent’s Healthcare Clinical CampusSchool of Clinical Medicine, University of New South WalesSydneyAustralia
| |
Collapse
|
69
|
Bookmeyer CHM, Correig FX, Masana L, Magni P, Yanes Ó, Vinaixa M. Advancing atherosclerosis research: The Power of lipid imaging with MALDI-MSI. Atherosclerosis 2025; 403:119130. [PMID: 40059002 DOI: 10.1016/j.atherosclerosis.2025.119130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/20/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease that is one of the leading causes of mortality globally. It is characterized by the formation of atheromatous plaques in the intima layer of larger arteries. The (fibro-)fatty plaques usually develop asymptomatically within the vessel until a serious event such as myocardial infarction or stroke occurs. Lipids play a pivotal role in disease progression, but while the causal role of cholesterol is beyond doubt, the distribution of numerous other lipids within the heterogeneous layers of atherosclerotic plaques, and their biological function remain unclear. A deeper understanding of the pathophysiological progression of the disease for prognostics, diagnostics, treatment, and prevention is of great need. Mass spectrometry imaging (MSI), in particular with matrix-assisted laser desorption/ionization (MALDI) offers an unprecedented untargeted characterization of the physiological microenvironment, unraveling the spatial distribution of numerous biochemical compounds. MALDI-MSI offers an advantageous balance of sample preparation, chemical sensitivity, and spatial resolution, and thus has been established as a key technology in modern biomedical analysis. This review focuses on the analysis of lipids in atherosclerotic lesions with MALDI-MSI, for which the past years showed major developments in the spatial characterization of lipids and their interaction within atherosclerotic plaques. We will cover main contributions with a focus on the recent decade, elaborate possibilities, limitations, main findings, and recent developments from sample handling to instrumentation, and estimate current challenges and potentials of MALDI-MSI with respect to a clinical application.
Collapse
Affiliation(s)
- Christoph H M Bookmeyer
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain.
| | - F Xavier Correig
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Luis Masana
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; Universitat Rovira i Virgili, Research Unit on Lipids and Atherosclerosis, Reus, Spain
| | - Paolo Magni
- Dept. of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Italy
| | - Óscar Yanes
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Maria Vinaixa
- Universitat Rovira i Virgili, Department of Electronic Engineering, Metabolomics Interdisciplinary Laboratory, Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
| |
Collapse
|
70
|
Yuan X, Zhang L, Li X, Chen K, Chu Q, Chen L, Hu S. Differential lipid impact on mortality in patients undergoing CABG versus PCI. Atherosclerosis 2025; 403:119141. [PMID: 40022948 DOI: 10.1016/j.atherosclerosis.2025.119141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND AND AIMS The impact of revascularization type, including coronary artery bypass grafting (CABG) and percutaneous coronary intervention (PCI), on lipid control efficacy remains unclear. We aim to assess the associations of lipid levels with all-cause and cardiovascular mortality in patients undergoing PCI or CABG. METHODS We used data from the ChinaHEART cohort and enrolled patients with a self-reported history of PCI or CABG. We employed Cox proportional hazards regression models to evaluate the associations between lipid levels and mortality. RESULTS Our analysis included 29 003 participants, of whom 23 959 (82.6 %) undergoing PCI and 5044 (17.4 %) undergoing CABG. Over a median follow-up of 3.22 years, 1007 deaths were recorded, with 579 attributed to cardiovascular causes. Each 1 mmol/L increase in TC, LDL-C, and non-HDL-C was associated with multivariable-adjusted HRs of 1.16 (95 % CI 1.10,1.22), 1.23 (1.15,1.32) and 1.16 (1.10,1.23) for all-cause mortality, respectively. Similar results were observed in patients undergoing PCI, while no significant associations were found in patients undergoing CABG. CONCLUSIONS Elevated lipid levels are associated with all-cause and cardiovascular mortality in revascularization patients. Suboptimal lipid control appears to have a more pronounced effect on mortality in patients undergoing PCI. The effects of elevated lipid levels on mortality in CABG patients may need a longer follow-up to manifest due to the more complex nature of the grafts and the long-term adaptation process.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihua Zhang
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi Li
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Chen
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Chu
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Chen
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengshou Hu
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
71
|
Abstract
PURPOSE Peripheral arterial disease (PAD) is characterized by atherosclerotic arterial occlusive disease of the lower extremities and is associated with an increased risk of major adverse cardiovascular events (MACE) in addition to disabling clinical sequelae, including intermittent claudication and chronic limb-threatening ischemia (CLTI). Given the growing burden of disease, knowledge of modern practices to prevent MACE and major adverse limb events (MALE) is essential. This review article examines evidence for medical management of PAD and its associated risk factors, as well as wound prevention and care. METHODS A thorough review of the literature was performed, with attention to evidence for the management of modifiable atherosclerotic risk factors, claudication symptoms, wound prevention, and wound care. RESULTS Contemporary management of PAD requires a multi-faceted approach to care, with medical optimization of smoking, hypertension, hyperlipidemia, and diabetes mellitus. The use of supervised exercise therapy for intermittent claudication is highlighted. The anatomic disease patterns of smoking and diabetes mellitus are discussed further, and best practices for diabetic foot ulcer prevention, including offloading footwear, are described. Quality wound care is essential in this patient population and involves strategic use of debridement, wound-healing adjuncts, and skin substitutes, when appropriate. CONCLUSION The objective of medical management of PAD is to reduce the risk of MACE and MALE. Atherosclerotic risk factor optimization, appropriate wound care, and management of diabetic foot ulcers, foot infections, gangrene, and chronic, non-healing wounds are critical components of PAD care. Interdisciplinary care is essential to coordinate care, leverage expertise, and improve outcomes.
Collapse
Affiliation(s)
- Ian O Cook
- Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, BCM 390, Houston, TX, 77030, USA
| | - Jayer Chung
- Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, BCM 390, Houston, TX, 77030, USA.
| |
Collapse
|
72
|
Gallo Ruelas M, Queiroz I, Pimentel T, Tavares AH, Defante MLR, Barbosa LM, Eckert I. Effects of seal oil supplementation on lipid profile biomarkers: A systematic review and meta-analysis of randomized controlled trials. Prostaglandins Leukot Essent Fatty Acids 2025; 204:102666. [PMID: 39914123 DOI: 10.1016/j.plefa.2025.102666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Seal oil (SO) supplementation has been purported to have cardiovascular health benefits due to its content of omega-3 fatty acids; however, the clinical evidence base for this intervention has yet to be comprehensively assessed. OBJECTIVE We aimed to evaluate the effects of oral SO supplementation on lipid profile biomarkers. METHODS A systematic search was performed on Pubmed, Embase, Web of Science and Cochrane Library, from inception to August 2024. Only randomized controlled trials (RCTs) assessing the effect of SO on lipid profile biomarkers were included. A random-effects meta-analysis was applied to determine the overall effect estimate. The certainty of evidence (CoE) was evaluated using the GRADE approach. RESULTS Nine RCTs were included in the review after the screening of 242 studies, comprising a total of 626 patients. Supplementation of SO resulted in no statistically significant effects on LDL-C (MD -0.07 mmol/L; 95 % CI [-0.19, 0.05]; CoE: Low) and total cholesterol (MD -0.12 mmol/L; 95 % CI [-0.30, 0.06]; CoE: Very low). There were statistically significant results of modest-to-trivial clinical importance on triglycerides (MD -0.19 mmol/L, 95 % CI [-0.30, -0.08]; CoE: Low) and trivial importance on HDL-C (MD 0.07 mmol/L, 95 % CI [0.003, 0.13]; CoE: Very low). CONCLUSION There is no sufficiently certain evidence to determine the effects of SO on cardiovascular lipid biomarkers. Our analyses may suggest a modest-to-trivial, clinically uncertain beneficial effect on triglyceride levels; and little to no effect on LDL-C. Effect estimates for HDL-C and total cholesterol levels were highly uncertain. Further evidence is required to conclusively determine the effects of oral SO on lipid biomarkers. PROTOCOL REGISTRATION NUMBER CRD42024583739.
Collapse
Affiliation(s)
| | - Ivo Queiroz
- Catholic University of Pernambuco, Medicine Department, Brazil
| | - Túlio Pimentel
- Federal University of Pernambuco, Medicine Department, Brazil
| | | | - Maria L R Defante
- Redentor University Center, Medicine Department, Itaperuna, Rio de Janeiro, Brazil
| | | | - Igor Eckert
- Independent Researcher, Porto Alegre, Brazil
| |
Collapse
|
73
|
Zueva AS, Shevchenko AI, Medvedev SP, Elisaphenko EA, Sleptcov AA, Nazarenko MS, Tmoyan NA, Zakian SM, Zakharova IS. Isogenic induced pluripotent stem cell line ICGi036-A-1 from a patient with familial hypercholesterolaemia, derived by correcting a pathogenic variant of the gene LDLR c.530C>T. Vavilovskii Zhurnal Genet Selektsii 2025; 29:189-199. [PMID: 40264801 PMCID: PMC12011625 DOI: 10.18699/vjgb-25-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 04/24/2025] Open
Abstract
Familial hypercholesterolaemia is a common monogenic disorder characterized by high plasma cholesterol levels leading to chronic cardiovascular disease with high risk and often early manifestation due to atherosclerotic lesions of the blood vessels. The atherosclerotic lesions in familial hypercholesterolaemia are mainly caused by pathogenic variants of the low-density lipoprotein receptor (LDLR) gene, which plays an important role in cholesterol metabolism. Normally, cholesterol-laden low-density lipoproteins bind to the LDLR receptor on the surface of liver cells to be removed from the bloodstream by internalisation with hepatocytes. In familial hypercholesterolaemia, the function of the receptor is impaired and the uptake of low-density lipoproteins is significantly reduced. As a result, cholesterol accumulates in the subendothelial space on the inner wall of blood vessels, triggering atherogenesis, the formation of atherosclerotic plaques. At present, there are no effective and universal approaches to the diagnosis and treatment of familial hypercholesterolaemia. A relevant approach to study the molecular genetic mechanisms of the disease and to obtain systems for screening chemical compounds as potential drugs is the generation of cellular models based on patient-specific induced pluripotent stem cells. The aim of our work was to derive an isogenic genetically modified induced pluripotent stem cell line by correcting the pathogenic allelic variant c.530C of the LDLR gene in the original iPSC previously obtained from a compound heterozygote patient with familial hypercholesterolaemia. The resulting isogenic iPSC line differs from the original by only one corrected nucleotide substitution, allowing us to study the direct effect of this pathogenic genetic variant on physiological changes in relevant differentiated cells. CRISPR/Cas-mediated base editing was used to correct the single nucleotide substitution. The resulting genetically modified iPSC line has pluripotency traits, a normal karyotype, a set of short tandem repeats identical to that in the original line and can be used to obtain differentiated derivatives necessary for the elaboration of relevant cell models.
Collapse
Affiliation(s)
- A S Zueva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - A I Shevchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S P Medvedev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E A Elisaphenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Sleptcov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - M S Nazarenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - N A Tmoyan
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia National Medical Research Center of Cardiology named after academician E.I. Chazov, Moscow, Russia
| | - S M Zakian
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - I S Zakharova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
74
|
Corsini A, Ginsberg HN, Chapman MJ. Therapeutic PCSK9 targeting: Inside versus outside the hepatocyte? Pharmacol Ther 2025; 268:108812. [PMID: 39947256 DOI: 10.1016/j.pharmthera.2025.108812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
As a major regulator of LDL receptor (LDLR) activity and thus of LDL-cholesterol (LDL-C) levels, proprotein convertase subtilisin/kexin type 9 (PCSK9) represents an obvious therapeutic target for lipid lowering. The PCSK9 inhibitors, alirocumab and evolocumab, are human monoclonal antibodies (mAbs) that act outside the cell by complexing circulating PCSK9 and thus preventing its binding to the LDLR. In contrast, inclisiran, a small interfering RNA (siRNA), inhibits hepatic synthesis of PCSK9, thereby resulting in reduced amounts of the protein inside and outside the cell. Both approaches result in decreased plasma LDL-C concentrations and improved cardiovascular outcomes. Marginally superior LDL-C reduction (≈ 60 %) is achieved with mAbs as compared to the siRNA (≈ 50 %); head-to-head comparisons are required to confirm between-class differences in efficacy. Both drug classes have shown variability in LDL-C lowering response between individuals in waterfall analyses. Whereas mAb-mediated inhibition leads to a compensatory increase in plasma PCSK9 levels, siRNA treatment reduces them. These agents differ in their pharmacokinetic and pharmacodynamic features, which may translate into distinct clinical opportunities under acute (e.g. acute coronary syndromes) as compared to chronic conditions. Both drug classes provide additional reduction in LDL-C levels (up to 50 %) beyond those achieved with statin therapy, facilitating attainment of guideline-recommended LDL-C goals in high and very high-risk patients. Additional PCSK9 inhibitors, including an oral macrocyclic peptide, a small PCSK9 binding protein and a novel small molecule, plus hepatic gene editing of PCSK9, are under development. This review critically appraises pharmacological strategies to target PCSK9 either inside or outside the cell.
Collapse
Affiliation(s)
- Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Henry N Ginsberg
- Irving Institute for Clinical and Translational Research, Columbia University, New York, USA
| | - M John Chapman
- Sorbonne University Medical Faculty, Lipidology and Cardiovascular Prevention Unit, Pitie-Salpetriere University Hospital, Paris, France.
| |
Collapse
|
75
|
Epigenetic editing of PCSK9 for a durable reduction in cholesterol. Nat Med 2025; 31:1083-1084. [PMID: 40000801 DOI: 10.1038/s41591-025-03583-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
|
76
|
Li D, Xu M, Wang D, Wang S, Li C. Polyguluronate sulfate has potential to mitigate hyperlipidemia by inhibiting the PCSK9-mediated degradation of LDLR. Int J Biol Macromol 2025; 302:140585. [PMID: 39904449 DOI: 10.1016/j.ijbiomac.2025.140585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/09/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
Hyperlipidemia has become a major global health challenge and one of the leading causes of mortality. Proprotein convertase subtilisin/kexin type 9 (PCSK9), a circulating plasma protein, promotes the lysosomal degradation of hepatic low-density lipoprotein receptors (LDLR), thereby reducing the clearance of low-density lipoprotein (LDL) from blood plasma. PCSK9 has increasingly become a prominent therapeutic target for the development of lipid-lowering agents. In this study, we firstly identified polyguluronate sulfate (PGS) as a novel PCSK9 inhibitor. PGS could bind to the positively charged domain of PCSK9 with a KD value of 3.198 μM, effectively blocking its mediated LDLR degradation. This interaction leads to increase in LDLR levels on hepatocyte surface, enhancing LDL clearance. Furthermore, we demonstrated that PGS more effectively activates the AMP-activated protein kinase (AMPK) pathway compared to polymannuronate sulfate (PMS) at 200 μg/mL, resulting in about 2-fold greater lipid-lowering effect. In summary, our findings highlight PGS as a promising candidate for the development of novel lipid-lowering drugs, offering new insights into the therapeutic potential of sulfate polysaccharides targeting PCSK9.
Collapse
Affiliation(s)
- Dan Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
| | - Meijie Xu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
| | - Dingfu Wang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
| | - Shixin Wang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory of Marine Glycodrug Research and Development, Marine Biomedical Research Institute of Qingdao, Qingdao, China
| | - Chunxia Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China; Laboratory of Marine Glycodrug Research and Development, Marine Biomedical Research Institute of Qingdao, Qingdao, China.
| |
Collapse
|
77
|
Wolska M, Peruzzi M, Kaziród-Wolski K, Wróbel P, Oleś I, Sielski J, Jankowski P. Risk factors for cardiovascular diseases: the focus on primary prevention. Minerva Cardiol Angiol 2025; 73:245-253. [PMID: 37971709 DOI: 10.23736/s2724-5683.23.06360-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Views on the etiopathogenesis of atherosclerosis are subject to evolution. In addition to the classic well-known risk factors, new ones related to mental state, social life and environment are being discovered. Both acute and chronic stress stimulate inflammatory processes. Due to the change in lifestyle and eating habits, the accumulation of risk factors in childhood is an increasing problem. Knowledge of risk factors allows for effective primary prevention of cardiovascular diseases. The effectiveness of prevention increases when the activities cover the largest possible part of the society, and access to a doctor is easy. Therefore, government programs are being implemented offering patients easier access to diagnostics of cardiovascular diseases at the level of primary health care, which enables faster identification of people at the greatest cardiovascular risk. Easier access to primary care and a good doctor-patient relationship improve patient compliance. In this situation, the importance of the family doctor as a key link in the diagnosis, prevention and treatment of cardiovascular diseases is increasing.
Collapse
Affiliation(s)
| | - Mariangela Peruzzi
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University, Rome, Italy -
- Mediterranea Cardiocentro, Naples, Italy
| | - Karol Kaziród-Wolski
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Paweł Wróbel
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Izabela Oleś
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Janusz Sielski
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Piotr Jankowski
- Department of Epidemiology and Health Promotion, School of Public Health, Center of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
78
|
Choi J, Wen W, Jia G, Tao R, Long J, Shu XO, Zheng W. Associations of Blood Lipid-Related Polygenic Scores, Lifestyle Factors and Their Combined Effects with Risk of Coronary Artery Disease in the UK Biobank Cohort. J Cardiovasc Transl Res 2025; 18:331-340. [PMID: 39680354 DOI: 10.1007/s12265-024-10578-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
Circulating lipids play a crucial role in the development of coronary artery disease (CAD). However, it is unclear whether the genetic susceptibility to hyperlipidemia may interact with lifestyle factors in CAD risk. Using UK Biobank data from 328,606 participants, we evaluated combined effects of genetic susceptibility to hyperlipidemia and lifestyle factors with risk of CAD. We found that both blood lipid-related polygenic score (PGS) and healthy lifestyle score (HLS) are independently associated with CAD risk, and individuals with the highest-risk lipid-related PGS and the least healthy HLS had the highest CAD risk. This association was stronger in younger (< 60 years, hazard ratio: 4.46, 95% confidence interval: 3.44-5.78) than older adults (2.54, 2.13-3.03). Our study suggests that individuals, particularly younger adults, with higher-risk PGSs of blood lipid traits would benefit more substantially by adherence to a healthy lifestyle than those with lower PGSs.
Collapse
Affiliation(s)
- Jungyoon Choi
- Division of Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-Do, Korea
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guochong Jia
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
79
|
Mortezaei A, Ghorbani M, Hajikarimloo B, Sameer O, Kazemi T, Salavati E, Hamidpour M, Gheydari ME. Is L-Arginine an Appropriate Alternative for Conventional Anti-Atherosclerotic Therapy?: A Comprehensive Review. Health Sci Rep 2025; 8:e70544. [PMID: 40161001 PMCID: PMC11949766 DOI: 10.1002/hsr2.70544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 04/02/2025] Open
Abstract
Background Atherosclerosis is the leading cause of cardiovascular disease (CVD). Historically, the management of atherosclerosis was focused on decreasing lipid profile levels; however, recent evidence demonstrated that platelets and leukocytes play an important role in forming and exacerbating atherosclerosis. L-arginine (L-Arg), a precursor to nitric oxide (NO), plays a critical role in modulating oxidative stress and influencing platelet-leukocyte recruitment and has been extensively addressed in the context of CVD. Objective We aimed to perform a comprehensive literature review on l-Arg metabolism in the causative pathway of atherosclerosis compared to conventional treatment and it as a putative therapeutic approach. Results L-Arg supplementation has shown promising effects on NO production, improving endothelial function and reducing oxidative stress in preclinical models. Clinical studies have indicated moderate improvements in vascular health markers, including reductions in inflammation and oxidative stress, although results have varied across studies. The potential of l-Arg to modify platelet-leukocyte recruitment and slow the progression of atherosclerotic plaque development has been observed in certain studies. However, these benefits remain inconsistent, and more robust clinical trials are needed to confirm its effectiveness. Additionally, while l-Arg appears to be relatively safe, some studies reported mild gastrointestinal discomfort as a common side effect. Conclusion l-Arg holds potential as a complementary or alternative treatment for atherosclerosis, particularly in improving endothelial function and reducing inflammation and oxidative stress. However, the variability in clinical outcomes and the lack of long-term data required further investigation into assessing therapeutic benefits. Future studies should focus on determining optimal dosing regimens, evaluating their long-term safety, and assessing their potential in combination with other therapies to enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- Ali Mortezaei
- Student Research CommitteeGonabad University of Medical SciencesGonabadIran
| | - Mohammad Ghorbani
- Faculty of Allied Medicine, Department of Medical Laboratory SciencesGonabad University of Medical SciencesGonabadIran
- Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | | | - Omar Sameer
- College of MedicineUniversity of SharjahSharjahUAE
| | - Toba Kazemi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Ebrahim Salavati
- Allameh Bohlool HospitalGonabad University of Medical SciencesGonabadIran
| | - Mohsen Hamidpour
- HSC Research Center, Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Esmail Gheydari
- Department of Cardiology, School of Medicine, Taleghani HospitalShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
80
|
Michikura M, Ogura M, Hori M, Matsuki K, Makino H, Fujioka S, Shishikura D, Hoshiga M, Harada-Shiba M. Association of Achilles tendon thickness with lipid profile and carotid IMT in patients with familial hypercholesterolemia. Atherosclerosis 2025; 403:119173. [PMID: 40158303 DOI: 10.1016/j.atherosclerosis.2025.119173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/10/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND AND AIMS Familial hypercholesterolemia (FH) is characterized by high levels of low-density lipoprotein cholesterol (LDL-C) and Achilles tendon (AT) thickening. AT thickness (ATT) is useful for diagnosing FH and assessing the risk of coronary artery disease (CAD). Nevertheless, the relationship between AT thickening and lipid profile is not clear. We investigated the association of ATT with lipid Profile and carotid IMT. METHODS We included 450 patients with clinically diagnosed heterozygous FH. ATT was measured by ultrasonography. RESULTS The rate of thickening increased for both AT and carotid-IMT according to age (p < 0.001). In the teens, there was no carotid-IMT thickening, but 39 % of the subjects had a thickened AT. The thresholds of cumulative LDL-C values for AT and carotid-IMT thickening based on ROC curves were 9210 mg/dL∗years (AUC: 0.66, 95 % CI: 0.61-0.72) for ATT and 11,255 mg/dL∗years (AUC: 0.79, 95 % CI: 0.75-0.84) for carotid-IMT. For cumulative LDL-C levels ≥ median, untreated HDL-C level was lower in the AT thickened group than in the non-thickened group (AT thickened: 52 (43-63) mg/dL, non-thickened: 63 (52-72) mg/dL). There was a significant correlation between ATT and cumulative LDL-C (Male: R = 0.48 p < 0.001, Female: R = 0.33 p < 0.001). CONCLUSIONS We clarified that both cumulative exposure to LDL-C and untreated HDL-C levels were closely related to AT thickening. Our results show that in ultrasonographic assessment, ATT is more useful than carotid-IMT for predicting the degree of lipid deposition in tissues, especially in young adults.
Collapse
Affiliation(s)
- Masahito Michikura
- Department of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center Hospital, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan; Department of Cardiology, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan.
| | - Masatsune Ogura
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, 6-8-1 Hinode, Urayasu, Chiba, 279-0013, Japan
| | - Mika Hori
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Kota Matsuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho Hirosaki City, Aomori, 036-8562. Japan
| | - Hisashi Makino
- Department of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center Hospital, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Shimpei Fujioka
- Department of Cardiology, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Daisuke Shishikura
- Department of Cardiology, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Masaaki Hoshiga
- Department of Cardiology, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| |
Collapse
|
81
|
Ahmed A, Naeem N, Jain A, Arora S, Elgendy IY. Acute Limb Ischemia Interventions. Interv Cardiol Clin 2025; 14:273-282. [PMID: 40049853 DOI: 10.1016/j.iccl.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Acute limb ischemia (ALI) is characterized by a sudden decrease in limb perfusion that threatens the viability of the limb. ALI can result from various causes including arterial embolism, thrombosis, or trauma. The diagnosis is predominantly clinical, guided by the "6 Ps" mnemonic-pain, pallor, pulse deficit, paralysis, paresthesia, and poikilothermia-and confirmed through imaging modalities if needed. Treatment decisions are informed by the Rutherford classification, ranging from viable to irreversibly damaged limbs. Early diagnosis and timely intervention are critical for improving limb salvage and reducing amputation rates.
Collapse
Affiliation(s)
- Asmaa Ahmed
- Department of Internal Medicine, Rochester General Hospital, 1425 Portland Avenue, Rochester, NY 14621, USA
| | - Nauman Naeem
- Department of Internal Medicine, Rochester General Hospital, 1425 Portland Avenue, Rochester, NY 14621, USA
| | - Aakriti Jain
- Department of Internal Medicine, Rochester General Hospital, 1425 Portland Avenue, Rochester, NY 14621, USA
| | - Sahej Arora
- Department of Internal Medicine, Rochester General Hospital, 1425 Portland Avenue, Rochester, NY 14621, USA
| | - Islam Y Elgendy
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, 900 South Limestone Street, Suite CTW 320, Lexington, KY 40536, USA.
| |
Collapse
|
82
|
Moya Mateo E, García Alonso R, Sánchez Sánchez C, Tung-Chen Y, Rodilla E, Beltrán Romero L, García-Donaire JA, Bonilla-Hernández MV, Muñoz-Rivas N, Castilla-Guerra L. Positioning for the use of multivessel clinicalultrasound in vascular risk evaluation: VASUS+protocol. 2024 Recommendations of the Vascular Risk WorkingGroup of The Spanish Society of Internal Medicine (SEMI), Clinical Ultrasound Working Group of the Spanish Society of Internal Medicine (SEMI) and the Spanish Society of Hypertension-Spanish League for the Fight against Arterial Hypertension (SHE-LELHA). Rev Clin Esp 2025; 225:223-230. [PMID: 40180493 DOI: 10.1016/j.rceng.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/30/2024] [Indexed: 04/05/2025]
Abstract
Atherosclerosis is the underlying disease in the entire spectrum of atherosclerotic vascular disease. Point of care ultrasound is a useful tool for its detection. Current guidelines recommend the use of scales such as SCORE 2 and SCORE 2OP in apparently healthy individuals and in those at intermediate-low risk, they recognize the role of the arterial plaque by ultrasound to refine risk stratification and the need for more aggressive preventive strategies. However, the way to evaluate the vascular territories in which there is presence of plaque, the amount or load of plaque is not homogeneous nor is it well protocolized. In this document, 2 protocols are proposed for the evaluation of vascular risk, VASUS and VASUS+, including the presence of ventricular hypertrophy with the objective of homogenizing clinical ultrasound in the assessment of vascular risk in clinical practice.
Collapse
Affiliation(s)
- E Moya Mateo
- Medicina Interna, Unidad de Riesgo Vascular, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - R García Alonso
- Medicina Interna, Complejo Asistencial de Ávila, Ávila, Spain
| | | | - Y Tung-Chen
- Medicina Interna, Hospital Universitario La Paz, Madrid, Spain
| | - E Rodilla
- Medicina Interna, Hospital Universitario de Sagunto, Universidad Cardenal Herrera-CEU, Sagunto, Spain
| | | | - J A García-Donaire
- Medicina Interna, Hospital Universitario Clínico San Carlos, Madrid, Spain; Universidad Complutense Madrid, Madrid, Spain
| | | | | | - L Castilla-Guerra
- Medicina Interna, Hospital Universitario Virgen Macarena, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
83
|
Gomes DA, Presume J, de Araújo Gonçalves P, Almeida MS, Mendes M, Ferreira J. Association Between the Magnitude of Glycemic Control and Body Weight Loss With GLP-1 Receptor Agonists and Risk of Atherosclerotic Cardiovascular Disease: A Systematic Review and Meta-analyses of Randomized Diabetes Cardiovascular Outcomes Trials. Cardiovasc Drugs Ther 2025; 39:337-345. [PMID: 38214869 DOI: 10.1007/s10557-024-07547-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
PURPOSE Reduction of major atherosclerotic cardiovascular events (MACE) has not been consistent among different glucagon-like peptide-1 receptor agonists (GLP-1 RAs) in patients with type 2 diabetes mellitus (T2DM). The aim of this study was to assess the association between the magnitude of glycemic control, body weight loss, and reductions in systolic blood pressure (SBP) and low-density lipoprotein cholesterol (LDL-C) achieved through GLP-1 RA therapy and MACE. METHODS Electronic databases (MEDLINE, CENTRAL, SCOPUS) were searched through March 2023. Studies were eligible if they were cardiovascular outcome trials (CVOTs) comparing GLP-1 RAs versus placebo in T2DM patients. The outcome of interest was 3-point MACE - cardiovascular death, myocardial infarction, or stroke. Random-effects meta-regression analyses evaluated the associations between reductions of HbA1c, body weight, SBP and LDL-C and reduction of MACE. RESULTS Overall, 8 CVOTs were included (60079 patients, 30693 with GLP-1 RAs). Reductions of HbA1C were associated with the reduction of 3P-MACE (Log RR -0.290 [95% CI -0.515;-0.064], p = 0.012), with an estimated RR reduction of 25% for each 1% absolute reduction in HbA1C levels. Body weight loss was associated with the reduction of 3P-MACE (Log RR -0.068 [95% CI -0.135;-0.001], p = 0.047), with an estimated RR reduction of 7% for each 1 kg reduction in body weight. Reductions of SBP (Log RR -0.058 [95% CI -0.192;0.076], p = 0.396) and LDL-C (Log RR -0.602 [95% CI -4.157;2.953], p = 0.740) were not associated with the reduction of 3P-MACE. CONCLUSIONS In T2DM patients, more potent GLP-1 RAs in reducing HbA1c and body weight were associated with greater reductions of MACE.
Collapse
Affiliation(s)
- Daniel A Gomes
- Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo Dos Santos, Carnaxide, 2790-134, Lisbon, Portugal.
| | - João Presume
- Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo Dos Santos, Carnaxide, 2790-134, Lisbon, Portugal
- Comprehensive Health Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Pedro de Araújo Gonçalves
- Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo Dos Santos, Carnaxide, 2790-134, Lisbon, Portugal
- Comprehensive Health Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Manuel Sousa Almeida
- Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo Dos Santos, Carnaxide, 2790-134, Lisbon, Portugal
- Comprehensive Health Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Miguel Mendes
- Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo Dos Santos, Carnaxide, 2790-134, Lisbon, Portugal
| | - Jorge Ferreira
- Cardiology Department, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo Dos Santos, Carnaxide, 2790-134, Lisbon, Portugal
| |
Collapse
|
84
|
Lee M, Lee KJ, Kim J, Lee DY, Park RW, Rhee SY, Cha JM, Yang HJ, Jang JW, Jung S, Lee J, Lee SH, Kim C, Bae JS, Kim YJ, Lee JH, Bae H, Kim Y. Low-density lipoprotein cholesterol levels and risk of incident dementia: a distributed network analysis using common data models. J Neurol Neurosurg Psychiatry 2025:jnnp-2024-334708. [PMID: 40169354 DOI: 10.1136/jnnp-2024-334708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/03/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND The link between low-density lipoprotein cholesterol (LDL-C) levels and dementia risk is poorly understood, with conflicting evidence on the role of LDL-C and the impact of statin therapy on cognitive outcomes. Thus, we aimed to examine the association between low-density LDL-C levels and the risk of dementia and assess the influence of statin therapy. METHODS We retrospectively analysed data from 11 university hospitals participating in the Observational Medical Outcomes Partnership (OMOP) Common Data Model (CDM). Participants with a prior diagnosis of dementia or those with <180 days of observation before cohort inclusion, and those included in both cohorts were excluded. The primary outcome was all-cause dementia, with the secondary outcome being Alzheimer's disease-related dementia (ADRD). The study utilised 1:1 propensity score matching to compare individuals with LDL-C levels below 70 mg/dL (1.8 mmol/L) against those with levels above 130 mg/dL (3.4 mmol/L), resulting in a primary analysis cohort of 108 980 matched patients. Secondary analyses further examined LDL-C thresholds below 55 mg/dL (1.4 mmol/L) and the influence of statin use. RESULTS The LDL-C levels below 70 mg/dL (1.8 mmol/L) were associated with a 26% reduction in the risk of all-cause dementia and a 28% reduction in the risk of ADRD, compared with levels above 130 mg/dL (3.4 mmol/L). For LDL-C levels below 55 mg/dL (1.4 mmol/L), there was an 18% risk reduction for both outcomes. Among those with LDL-C <70 mg/dL (<1.8 mmol/L), statin use was associated with a 13% reduction in all-cause dementia risk and a 12% decrease in ADRD risk compared with non-users. CONCLUSION Low LDL-C levels (<70 mg/dL (<1.8 mmol/L)) are significantly associated with a reduced risk of dementia, including ADRD, with statin therapy providing additional protective effects. These findings support the necessity of targeted lipid management as a preventive strategy against dementia, indicating the importance of personalised treatment approaches.
Collapse
Affiliation(s)
- Minwoo Lee
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea (the Republic of)
| | - Kyung Joo Lee
- Department of Medical Informatics & Statistics, Kangdong Sacred Heart Hospital, Seoul, Korea (the Republic of)
| | - Jinseob Kim
- Zarathu Co Ltd, Seoul, Korea (the Republic of)
| | - Dong Yun Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Korea (the Republic of)
- Bongdam forest mental health clinic, Hwaseong, Korea (the Republic of)
| | - Rae Woong Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Korea (the Republic of)
| | - Sang Youl Rhee
- Department of Digital Health and Department of Endocrinology and Metabolism, Kyung Hee University College of Medicine, Seoul, Korea (the Republic of)
| | - Jae Myung Cha
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, Korea (the Republic of)
| | - Hyeon-Jong Yang
- Department of Pediatrics, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea (the Republic of)
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University Hospital, Kangwon National University College of Medicine, Chuncheon, Korea (the Republic of)
| | - Seunguk Jung
- Department of Neurology, Gyeongsang National University Changwon Hospital, Changwon, Korea (the Republic of)
| | - Jeeun Lee
- Department of Neurology, Myongji Hospital, Hanyang University College of Medicine, Goyang, Korea (the Republic of)
| | - Sang-Hwa Lee
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Korea (the Republic of)
| | - Chulho Kim
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Korea (the Republic of)
| | - Jong-Seok Bae
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea (the Republic of)
| | - Yeo Jin Kim
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea (the Republic of)
| | - Ju-Hun Lee
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea (the Republic of)
| | - Hyoeun Bae
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea (the Republic of)
| | - Yerim Kim
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea (the Republic of)
| |
Collapse
|
85
|
Ao L, van Heemst D, Jukema JW, Rensen PCN, Willems van Dijk K, Noordam R. Potential causal evidence for an ApoB-independent and HDL-related risk profile associated with coronary artery disease. J Lipid Res 2025; 66:100778. [PMID: 40089107 PMCID: PMC12000745 DOI: 10.1016/j.jlr.2025.100778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025] Open
Abstract
Plasma 1H-NMR metabolomic measures have yielded significant insight into the pathophysiology of cardiometabolic disease, but their inter-related nature complicates causal inference and clinical interpretation. This study aimed to investigate the associations of unrelated 1H-NMR metabolomic profiles with coronary artery disease (CAD) and ischemic stroke (IS). Principal component (PC) analysis was performed on 168 1H-NMR metabolomic measures in 56,712 unrelated European participants from UK Biobank to retrieve uncorrelated PCs, which were used in Cox-proportional hazard models. For each outcome, two-sample Mendelian randomization analyses were then conducted based on three nonoverlapping databases, followed by a meta-analysis. The first six PCs collectively explaining 88% of the total variance were identified. For CAD, results from Cox and Mendelian randomization analyses were generally directionally consistent. The pooled odds ratios (95% CI) for CAD per one-SD increase in genetically influenced PC1 and PC3 (both characterized by distinct apolipoprotein B [ApoB]-associated lipoprotein profiles) were 1.04 (1.03, 1.05) and 0.94 (0.93, 0.96), respectively. Besides, the pooled odds ratio (95% CI) for CAD per one-SD increase in genetically influenced PC4, characterized by simultaneously decreased small HDL and increased large HDL, and independent of ApoB, was 1.05 (1.03, 1.07). For IS, increases of PC3 and PC5 (characterized by increased amino acids) were associated with a lower risk and a higher risk, respectively. This study confirms associations of ApoB-associated lipoprotein profiles with CAD and IS, and highlights the possible existence of an ApoB-independent lipoprotein profile, characterized by a distinctive HDL subparticle distribution, driving CAD.
Collapse
Affiliation(s)
- Linjun Ao
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
86
|
Avogaro A, Buzzetti R, Candido R, Cosmo SD, Notarianni L, Consolo E, Luciano M. Exploring the benefits of alirocumab as lipid-lowering therapy in people with diabetes and very high cardiovascular risk. Diabetes Res Clin Pract 2025; 222:112055. [PMID: 40020784 DOI: 10.1016/j.diabres.2025.112055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
People with diabetes mellitus (DM) are at a higher risk (2-4 times) for cardiovascular (CV) death and atherosclerotic CV disease (ASCVD) than the general population. A multifactorial approach is recommended to reduce CV risk. Since low-density lipoprotein cholesterol (LDL-C) is a major causal and cumulative risk factor for ASCVD, the management of lipids is a fundamental element in global risk reduction. Intensive lipid lowering therapy (LLT), such as the addition of a proprotein convertase subtilisin/kexin type 9 inhibitor (PCSK9i), to achieve LDL-C goals and reduce the risk of first or recurrent CV events in people with DM at very high CV risk (VHCVR) of ASCVD (i.e. acute coronary syndrome, coronary artery disease, peripheral artery disease) is often required. Alirocumab, a monoclonal antibody against PCSK9, as lipid-lowering therapy offers significant CV benefits and a favourable safety profile in people with DM and a VHCVR, with or without previous CV events. This review highlights the role of LDL-C in the complex pathogenesis of atherosclerosis, summarises the guidelines for CV risk reduction related to LDL-C in patients with DM and a VHCVR, and focuses on the role of alirocumab in managing LDL-C and consequent CV risk reduction in these patients.
Collapse
Affiliation(s)
- Angelo Avogaro
- Department of Medicine, Section of Diabetes and Metabolic Diseases, University of Padua Metabolic Diseases Division, University Hospital of Padova, Padua, Italy.
| | - Raffaella Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Riccardo Candido
- Diabetes Unit, Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Salvatore De Cosmo
- Department of Medical Sciences, Unit of Internal Medicine, IRCCS "Casa Sollievo della Sofferenza" San Giovanni Rotondo (FG), Italy
| | | | | | - Myriam Luciano
- Medical and Scientific Department, Sanofi S.r.l., Milan, Italy
| |
Collapse
|
87
|
Wienbergen H, Hanses U, Kerniss H, Hambrecht R. [Update on cardiovascular prevention 2025]. Herz 2025:10.1007/s00059-025-05305-1. [PMID: 40153005 DOI: 10.1007/s00059-025-05305-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/30/2025]
Abstract
Despite an expensive healthcare system Germany performs poorly with respect to life-expectancy compared to other countries, for which cardiovascular diseases and deficits in cardiovascular prevention in particular are responsible. The basis of cardiovascular prevention is a healthy lifestyle with regular physical exercise, a predominantly plant-based diet, nonsmoking, good sleep and mental health. In many cases additional lipid-lowering, antidiabetic and antihypertensive medications are necessary. Recent studies have proven the prognostic effects of different groups of medications, such as proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, sodium-glucose transporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP-1) receptor agonists, in the appropriate indication areas. To improve the cardiovascular prevention in Germany, intensified public efforts are crucial. In addition, individual support of patients is effective for long-term preventive measures. To achieve this healthcare professionals must be trained (physicians, cardiovascular prevention assistants), who can sustainably support patients in lifestyle modifications and medicinal prevention.
Collapse
Affiliation(s)
- Harm Wienbergen
- Bremer Institut für Herz- und Kreislaufforschung (BIHKF), Stiftung Bremer Herzen, Senator-Weßling-Str. 2, 28277, Bremen, Deutschland.
- Klinik für Innere Medizin II, Gesundheit Nord - Klinikverbund Bremen gGmbH, Bremen, Deutschland.
- Universitäres Herzzentrum Lübeck, Medizinische Klinik II, Universität zu Lübeck, Lübeck, Deutschland.
| | - Ulrich Hanses
- Bremer Institut für Herz- und Kreislaufforschung (BIHKF), Stiftung Bremer Herzen, Senator-Weßling-Str. 2, 28277, Bremen, Deutschland
- Klinik für Innere Medizin II, Gesundheit Nord - Klinikverbund Bremen gGmbH, Bremen, Deutschland
| | - Hatim Kerniss
- Bremer Institut für Herz- und Kreislaufforschung (BIHKF), Stiftung Bremer Herzen, Senator-Weßling-Str. 2, 28277, Bremen, Deutschland
- Klinik für Innere Medizin II, Gesundheit Nord - Klinikverbund Bremen gGmbH, Bremen, Deutschland
| | - Rainer Hambrecht
- Bremer Institut für Herz- und Kreislaufforschung (BIHKF), Stiftung Bremer Herzen, Senator-Weßling-Str. 2, 28277, Bremen, Deutschland
- Klinik für Innere Medizin II, Gesundheit Nord - Klinikverbund Bremen gGmbH, Bremen, Deutschland
| |
Collapse
|
88
|
Kavanagh ME, Chiavaroli L, Quibrantar SM, Viscardi G, Ramboanga K, Amlin N, Paquette M, Sahye-Pudaruth S, Patel D, Grant SM, Glenn AJ, Ayoub-Charette S, Zurbau A, Josse RG, Malik VS, Kendall CWC, Jenkins DJA, Sievenpiper JL. Acceptability of a Web-Based Health App (PortfolioDiet.app) to Translate a Nutrition Therapy for Cardiovascular Disease in High-Risk Adults: Mixed Methods Randomized Ancillary Pilot Study. JMIR Cardio 2025; 9:e58124. [PMID: 40152922 PMCID: PMC11992491 DOI: 10.2196/58124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 01/09/2025] [Accepted: 02/03/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The Portfolio Diet is a dietary pattern for cardiovascular disease (CVD) risk reduction with 5 key categories including nuts and seeds; plant protein from specific food sources; viscous fiber sources; plant sterols; and plant-derived monounsaturated fatty acid sources. To enhance implementation of the Portfolio Diet, we developed the PortfolioDiet.app, an automated, web-based, multicomponent, patient-facing health app that was developed with psychological theory. OBJECTIVE We aimed to evaluate the effect of the PortfolioDiet.app on dietary adherence and its acceptability among adults with a high risk of CVD over 12 weeks. METHODS Potential participants with evidence of atherosclerosis and a minimum of one additional CVD risk factor in an ongoing trial were invited to participate in a remote web-based ancillary study by email. Eligible participants were randomized in a 1:1 ratio using a concealed computer-generated allocation sequence to the PortfolioDiet.app group or a control group for 12 weeks. Adherence to the Portfolio Diet was assessed by weighed 7-day diet records at baseline and 12 weeks using the clinical Portfolio Diet Score, ranging from 0 to 25. Acceptability of the app was evaluated using a multifaceted approach, including usability through the System Usability Scale ranging from 0 to 100, with a score >70 being considered acceptable, and a qualitative analysis of open-ended questions using NVivo 12. RESULTS In total, 41 participants were invited from the main trial to join the ancillary study by email, of which 15 agreed, and 14 were randomized (8 in the intervention group and 6 in the control group) and completed the ancillary study. At baseline, adherence to the Portfolio Diet was high in both groups with a mean clinical Portfolio Diet Score of 13.2 (SD 3.7; 13.2/25, 53%) and 13.7 (SD 5.8; 13.7/25, 55%) in the app and control groups, respectively. After the 12 weeks, there was a tendency for a mean increase in adherence to the Portfolio Diet by 1.25 (SD 2.8; 1.25/25, 5%) and 0.19 (SD 4.4; 0.19/25, 0.8%) points in the app and control group, respectively, with no difference between groups (P=.62). Participants used the app on average for 18 (SD 14) days per month and rated the app as usable (System Usability Scale of mean 80.9, SD 17.3). Qualitative analyses identified 4 main themes (user engagement, usability, external factors, and added components), which complemented the quantitative data obtained. CONCLUSIONS Although adherence was higher for the PortfolioDiet.app group, no difference in adherence was found between the groups in this small ancillary study. However, this study demonstrates that the PortfolioDiet.app is considered usable by high-risk adults and may reinforce dietitian advice to follow the Portfolio Diet when it is a part of a trial for CVD management. TRIAL REGISTRATION ClinicalTrials.gov NCT02481466; https://clinicaltrials.gov/study/NCT02481466.
Collapse
Affiliation(s)
- Meaghan E Kavanagh
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Laura Chiavaroli
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Selina M Quibrantar
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gabrielle Viscardi
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Kimberly Ramboanga
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Natalie Amlin
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Melanie Paquette
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Sandhya Sahye-Pudaruth
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Darshna Patel
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Shannan M Grant
- Departments of Pediatrics and Obstetrics and Gynaecology, IWK Health, Halifax, NS, Canada
- Department of Obstetrics and Gynaecology, Faulty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Andrea J Glenn
- Department of Nutrition and Food Studies, New York University, New York, NY, United States
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Sabrina Ayoub-Charette
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Andreea Zurbau
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Robert G Josse
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Vasanti S Malik
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Cyril W C Kendall
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - David J A Jenkins
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - John L Sievenpiper
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
89
|
Campuzano R, Mostaza JM, Barrios V, Egocheaga-Cabello I, Gómez-Cerezo JF, Pallarés-Carratalá V, Martínez-López I, Castellanos M, Hernández-Subirá I, Morant-Talamante N, Parrondo J, Arigita-Lastra L, Gámez JM. Assessing LDL-C Levels and Lipid-Modifying Therapies in a Real-World Cohort of Patients with Atherosclerotic Cardiovascular Disease: The REALITY Study. J Clin Med 2025; 14:2340. [PMID: 40217791 PMCID: PMC11989470 DOI: 10.3390/jcm14072340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Background and Objectives: Patients with atherosclerotic cardiovascular disease (ASCVD) remain at high risk of recurrent events. REALITY aims to analyse the actual levels of low-density lipoprotein cholesterol (LDL-C), the modalities of lipid-lowering therapies (LLT) used, and ASCVD patient outcomes in a real-world setting in Spain. Methods: REALITY is a retrospective observational study conducted using a healthcare database of 1.8 million patients representative of the Spanish population. The study included 26,976 patients with new or recurrent ASCVD events recruited from 2017 to 2019 and followed up for two years. Results: Management of ASCVD involved the utilisation of high-intensity (53%) or very high-intensity (36%) LLT. After two years, a decrease in total cholesterol (from 228.7 to 176.7 mg/dL), LDL-C (from 126.4 to 99.0 mg/dL), and triglycerides (from 216.7 to 163.7 mg/dL), accompanied by a moderate increase in HDL-C levels was observed. However, LDL-C goals of ESC/EAS guidelines were only reached by a minority of patients. Less than 15% of ASCVD patients achieved their LDL-C goals of <70, as stated in contemporary guidelines (3% if the present <55 mg/dL threshold in the ESC/EAS guidelines is considered). During the follow-up period, 9% of ASCVD patients died and 25% experienced a new ASCVD event. Conclusions: In the REALITY study, most patients did not achieve their target LDL-C goals despite receiving high- or very high-intensity LLT. Increasing the utilisation of extreme LLT is crucial to reducing recurrent ASCVD events and mitigating these patients' high morbidity and mortality risk.
Collapse
Affiliation(s)
- Raquel Campuzano
- Department of Cardiology, Alcorcón Foundation University Hospital, 28922 Madrid, Spain;
| | - José M. Mostaza
- Department of Internal Medicine, La Paz University Hospital, 28046 Madrid, Spain
| | - Vivencio Barrios
- Department of Cardiology, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | | | | | - Vicente Pallarés-Carratalá
- Health Surveillance Unit, Unión de Mutuas, 12004 Castellón, Spain;
- Department of Medicine, Jaume I University, 12006 Castellón, Spain
| | - Iciar Martínez-López
- Pharmacy Department and Molecular Diagnostic and Clinical Genetics Unit, Son Espases University Hospital, 07120 Palma de Mallorca, Spain
| | - Mar Castellanos
- Department of Neurology, A Coruña University Hospital and Biomedical Research Institute, 15006 A Coruña, Spain;
| | | | | | - Javier Parrondo
- Early Products and Health Economics Department, Novartis Pharmaceuticals, 08013 Barcelona, Spain
| | | | - José M. Gámez
- Department of Cardiology, Son Llátzer University Hospital, 07198 Palma de Mallorca, Spain
- Departamento de Medicina, Universidad de las Islas Baleares, 07120 Palma de Mallorca, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN CB 12/03/30038), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
90
|
Cinza-Sanjurjo S, Pallarés-Carratalá V, Díaz Rodríguez A, Fierro-González D, Turégano-Yedro M, Polo-García J. [Practical approach to the patient with hypercholesterolemia in Spain. SEMERGEN position statement]. Semergen 2025; 51:102460. [PMID: 40139108 DOI: 10.1016/j.semerg.2025.102460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025]
Abstract
Hypercholesterolemia, specifically the increase in the set of lipoproteins containing apolipoproteinB and, in particular, low-density lipoprotein cholesterol (LDL-C), together with the decrease in high-density lipoprotein cholesterol (HDL-C) constitute the etiopathogenic basis of atherosclerotic vascular disease. Multiple clinical trials have shown that lowering LDL-C by lipid-lowering therapy is associated with a significant decrease in the risk of vascular complications. Thus, LDL-C is the main therapeutic target in patients with dyslipidemia. Unfortunately, current LDL-C control figures are still very low, partly due to insufficient intensification of lipid-lowering therapy, but also due to the need for new tools to achieve these goals. This paper reviews the different lipid-lowering treatment options, including the latest available therapies, and provides a practical approach to achieving LDL-C control goals in patients with hypercholesterolemia, as well as in different patient subgroups.
Collapse
Affiliation(s)
- S Cinza-Sanjurjo
- Centro de Salud Milladoiro, Área Sanitaria de Santiago de Compostela, Santiago de Compostela, A Coruña, España; Instituto de Investigación de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, España; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, España; Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Agencia de Investigación de SEMERGEN; Junta directiva de SEMERGEN.
| | - V Pallarés-Carratalá
- Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Junta directiva de SEMERGEN; Departamento de Medicina, Universitat Jaume I, Castellón, España
| | - A Díaz Rodríguez
- Centro de Salud Bembibre, Bembibre, León, España; Grupo de Trabajo de Lípidos de SEMERGEN
| | - D Fierro-González
- Grupo de Trabajo de Lípidos de SEMERGEN; Centro de Salud de Armunia, León, España
| | - M Turégano-Yedro
- Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Centro de Salud Casar de Cáceres, Casar de Cáceres, Cáceres, España
| | - J Polo-García
- Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Junta directiva de SEMERGEN; Centro de Salud Casar de Cáceres, Casar de Cáceres, Cáceres, España
| |
Collapse
|
91
|
Ren BX, Zeng ZL, Deng L, Hu JM, Chen MZ, Jiang HW, Zang CZ, Fang ST, Weiss SJ, Liu J, Fu R, Wu ZQ. Genetic and pharmacological targeting of Snail inhibits atherosclerosis by relieving intraplaque endothelium dysfunction and associated inflammation. Acta Pharmacol Sin 2025:10.1038/s41401-025-01519-5. [PMID: 40133628 DOI: 10.1038/s41401-025-01519-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
The intraplaque endothelium dysfunction and associated inflammation contribute to the progression of atherosclerosis. We previously show that zinc-finger transcription factor Snail is predominantly expressed in embryonic vascular endothelial cells (ECs), and deletion of Snail in ECs induces severe defects in vascular development and thus causes embryonic lethality. Snail is essentially absent at postnatal stage, and inducible deletion of Snail in ECs has no impact on physiological angiogenesis in postnatally developing or adult mice. In this study we investigated whether Snail was reactivated in vascular ECs during pathologically angiogenic process (e.g. the formation of atherosclerotic plaque) or could play a functional role in atherosclerosis progression. We showed that the expression levels of Snail were significantly elevated in ECs of human and mouse atherosclerotic plaques, and associated with the disease severity. In the accelerated and canonical mouse models of atherosclerosis, tamoxifen-inducible, EC-specific Snail deletion significantly reduced intraplaque endothelial dysfunction, inflammation and lipid uptake accompanied by enhanced plaque stability. By conducting scRNA-sequencing in ECs of ApoE-/-SnailiΔEC versus ApoE-/-Snailfl/fl arterial vessels, we demonstrated that Snail deletion significantly decreased histone acetylation on Ccl5 and Cxcl10 promoters, thereby decreased CCL5/CXCL10-driven vascular damage and inflammation. Administration with recombinant CXCL10 protein (2 μg/kg, i.v., once per week for three weeks) efficiently restored atherosclerosis in EC-specific Snail-deleted mice. Finally, we developed an orally bioavailable small-molecule Snail inhibitor LFW273 that displayed potent anti-atherosclerotic effects in mice. These results reveal Snail as a promising therapeutic target in atherosclerotic disease.
Collapse
Affiliation(s)
- Bo-Xue Ren
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhao-Lan Zeng
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Department of Organic Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Li Deng
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jia-Meng Hu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ming-Zhen Chen
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hao-Wei Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chen-Zi Zang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Shen-Tong Fang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Stephen J Weiss
- The Life Sciences Institute, Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jie Liu
- Department of Organic Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, China.
| | - Rong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Zhao-Qiu Wu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
92
|
Mahmoud A, Abdelsayed K, Mohamed AA, Najah Q, Abdulkader A, Ali K, Tabassum S, Abouzid MR, Abdelazeem B, Mills JD. Safety and efficacy of antisense oligonucleotides on triglyceride, apolipoprotein C-III, and other lipid parameters levels in hypertriglyceridemia; a network meta-analysis of randomized controlled trials. Lipids Health Dis 2025; 24:109. [PMID: 40119340 PMCID: PMC11929262 DOI: 10.1186/s12944-024-02389-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/27/2024] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Hypertriglyceridemia is an independent risk factor for cardiovascular diseases. In previous trials, apolipoprotein C-III (APOC3) inhibition through the antisense oligonucleotides volanesorsen, olezarsen, and plozasiran reduced triglyceride levels. However, the three medications' safety and efficacy have yet to be compared. METHODS A network meta-analysis was performed to compare multiple doses of the three medications to each other through the placebo. Randomized controlled trials (RCTs) were retrieved by searching PubMed, EMBASE, Web of Science, SCOPUS, and Cochrane until November 22nd, 2024. The mean difference (MD) and 95% confidence interval (CI) were used for continuous outcomes. The risk ratio (RR) and 95% CI were used for dichotomous outcomes. RESULTS Ten RCTs with a total of 1,129 patients were included. volanesorsen 300 mg once weekly showed the most significant percent reduction in triglyceride levels (MD = -91.0%, 95% CI: (-109.2%; -72.8%); P < 0.01). Only plozasiran once monthly, regardless of the dose, showed a non-significant percent reduction in triglycerides. This finding should be taken cautiously as the data were derived from a phase 1 trial with a small sample size. All the regimens significantly reduced APOC3 levels compared to placebo, with plozasiran 100 mg monthly and volanesorsen 300 mg once weekly showing the most significant reduction (MD range: -92.8% to -88.5%; P < 0.01). None of the treatments showed a statistically significant difference in overall adverse events rate compared to the placebo. CONCLUSION APOC3 antisense oligonucleotide inhibitors effectively reduced triglyceride and APOC3 levels in hypertriglyceridemia with an acceptable safety profile. However, the results should be interpreted cautiously due to the small sample size. Further research is needed to confirm the beneficial effects of APOC3 inhibitors and show strong evidence of the impact of each regimen.
Collapse
Affiliation(s)
| | - Kerollos Abdelsayed
- Clinical Research Department, Aswan Heart Centre, Magdi Yacoub Foundation, Aswan, Egypt.
| | - Ahmed Almahdy Mohamed
- Department of Medicine, Division of Cardiology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Qasi Najah
- Faculty of Medicine, Elmergib University, Alkhums, Libya
| | - Anas Abdulkader
- College of Medicine, AlMaarefa University, Riyadh, KSA, Saudi Arabia
| | - Karim Ali
- Department of Internal Medicine, Hennepin County Medical Center, Minneapolis, MN, USA
| | - Shehroze Tabassum
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Mohamed Riad Abouzid
- Department of Internal Medicine, Baptist Hospitals of Southeast Texas, Beaumont, TX, USA
| | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | - James D Mills
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
93
|
Thethwayo ST, Madoroba E, Masikane S, Opoku AR, Cele ND. The Effect of Bauhinia bowkeri Extracts on Hypercholesterolemia: Insights from In Vitro and In Silico Investigations. PLANTS (BASEL, SWITZERLAND) 2025; 14:979. [PMID: 40265934 PMCID: PMC11945628 DOI: 10.3390/plants14060979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025]
Abstract
Despite the many current cholesterol-lowering drugs on the market, the persistent surge of hypercholesterolemic-related complications ignites a fascinating search for the discovery of novel therapeutics. This study aimed at investigating the anti-hypercholesterolemic effect of Bauhinia bowkeri extracts. The plant material was sequentially extracted with n-hexane, dichloromethane (DCM), and 70% ethanol. The phytochemical constituents of the extracts were analyzed through GC-MS and the antioxidant activity of the extracts was screened against a wide range of free radicals (ABTS, DPPH, hydroxyl radical, and nitric oxide). The extracts were also screened for the metal iron chelating and reducing power potential. The enzyme inhibitory activity of the extracts on pancreatic lipase, cholesterol esterase, and HMG-CoA reductase as well as the bile acid binding capacity were evaluated. Among the total of 122 compounds detected in the three extracts, only 7 compounds (E-15-Heptadecenal, Diethyl Phthalate, 9,12,15-Octadecatrienoic acid ethyl ester, (Z,Z) Tetradecane 5-methyl, and Octadecane 5-methyl) were found to be common in all the extracts. The extract displayed a varying degree of efficiency on free radicals with IC50 values ranging from 0.07 mg/mL to 0.41 mg/mL. A concentration-dependent inhibition of pancreatic lipase and cholesterol esterase activities, along with a reduction in the bile-binding capacity exhibited by the extracts, was noted. In silico investigations of some of the phytoconstituent revealed significant inhibition of HMG-CoA reductase, cyclooxygenase, and hormone-sensitive lipase with a binding affinity that ranged between -5.1 and -7.0 kcal/mol. These findings suggest that Bauhinia bowkeri extracts possess potential antioxidant and anti-hypercholesterolemic properties.
Collapse
Affiliation(s)
- Siphelele T. Thethwayo
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture, and Engineering, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa; (S.T.T.); (E.M.); (A.R.O.)
| | - Evelyn Madoroba
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture, and Engineering, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa; (S.T.T.); (E.M.); (A.R.O.)
| | - Sphamandla Masikane
- Department of Chemistry, Faculty of Science, Agriculture and Engineering, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa;
| | - Andrew R. Opoku
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture, and Engineering, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa; (S.T.T.); (E.M.); (A.R.O.)
| | - Nkosinathi D. Cele
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture, and Engineering, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa; (S.T.T.); (E.M.); (A.R.O.)
| |
Collapse
|
94
|
Wang Y, Courcelles E, Peyronnet E, Porte S, Diatchenko A, Jacob E, Angoulvant D, Amarenco P, Boccara F, Cariou B, Mahé G, Steg PG, Bastien A, Portal L, Boissel JP, Granjeon-Noriot S, Bechet E. Credibility assessment of a mechanistic model of atherosclerosis to predict cardiovascular outcomes under lipid-lowering therapy. NPJ Digit Med 2025; 8:171. [PMID: 40108310 PMCID: PMC11923190 DOI: 10.1038/s41746-025-01557-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
Demonstrating cardiovascular (CV) benefits with lipid-lowering therapy (LLT) requires long-term randomized clinical trials (RCTs) with thousands of patients. Innovative approaches such as in silico trials applying a disease computational model to virtual patients receiving multiple treatments offer a complementary approach to rapidly generate comparative effectiveness data. A mechanistic computational model of atherosclerotic cardiovascular disease (ASCVD) was built from knowledge, describing lipoprotein homeostasis, LLT effects, and the progression of atherosclerotic plaques leading to myocardial infarction, ischemic stroke, major acute limb event and CV death. The ASCVD model was successfully calibrated and validated, and reproduced LLT effects observed in selected RCTs (ORION-10 and FOURIER for calibration; ORION-11, ODYSSEY-OUTCOMES and FOURIER-OLE for validation) on lipoproteins and ASCVD event incidence at both population and subgroup levels. This enables the future use of the model to conduct the SIRIUS programme, which intends to predict CV event reduction with inclisiran, an siRNA targeting hepatic PCSK9 mRNA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Denis Angoulvant
- Cardiology department, Hôpital Trousseau, CHRU de Tours & UMR Inserm 1327 ISCHEMIA "Membrane Signaling and Inflammation in Reperfusion Injuries" Université de Tours, F37000, Tours, France
| | - Pierre Amarenco
- Department of Neurology and Stroke center, APHP, Bichat Hospital, Université Paris-Cité, Paris, France and McMaster University, Population Health Research Institute, Hamilton, ON, Canada
| | - Franck Boccara
- Sorbonne Université, GRC n°22, C2MV-Complications Cardiovasculaires et Métaboliques chez les patients vivant avec le Virus de l'immunodéficience humaine, Inserm UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), Cardiologie, Hôpital Saint Antoine AP-HP, Paris, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Guillaume Mahé
- Vascular Medicine Unit, CHU Rennes, Univ Rennes, CIC1414, M2S-EA 7470, Rennes, France
| | | | | | | | | | | | | |
Collapse
|
95
|
Li CY, Wang WT, Ma SH, Lo LW, Wu CY, Chang WC, Chen YJ, Chen TL. Association of proprotein convertase subtilisin/kexin type-9 inhibitors with risk of nonmelanoma skin cancer: a retrospective cohort study. Br J Dermatol 2025; 192:697-705. [PMID: 39585798 DOI: 10.1093/bjd/ljae438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/03/2024] [Accepted: 11/03/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Growing evidence has shown that cholesterol metabolism abnormalities involve carcinogenesis. Proprotein convertase subtilisin/kexin type-9 (PCSK9) inhibitors have been reported to inhibit tumour progression and prevent ultraviolet-related skin damage. OBJECTIVES To investigate the association of PCSK9 inhibitors with the risk of nonmelanoma skin cancer (NMSC). METHODS This retrospective cohort study analysed data from the US Collaborative Network in the TriNetX database. Adults aged ≥ 40 years with atherosclerotic cardiovascular disease (ASCVD) under statin therapy between 2016 and 2022 were identified. A target trial design was used to compare the risk of NMSC, including basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC), in patients also treated with PCSK9 inhibitors or continuing statin treatment (the control group). Each head-to-head comparison involved propensity score matching. Hazard ratios (HRs) were estimated using Cox proportional hazard models. Stratified analyses based on age, sex, Fitzpatrick skin type and immune status were also performed. RESULTS A total of 73 636 patients with ASCVD were analysed. Compared with the control group, patients with ASCVD initiating PCSK9 inhibitors had lower risks of developing NMSC [HR 0.78, 95% confidence interval (CI) 0.71-0.87], BCC (HR 0.78, 95% CI 0.69-0.89) and cSCC (HR 0.79, 95% CI 0.67-0.93). Subanalyses revealed a reduced risk of NMSC with each PCSK9 inhibitor, namely evolocumab and alirocumab. Stratified analyses showed similar results in patients aged 65-79 years, those older than 80 years and in men. CONCLUSIONS Our study indicated that patients with ASCVD taking PCSK9 inhibitors have a lower risk of incident NMSC than those not taking PCSK9 inhibitors.
Collapse
Affiliation(s)
- Cheng-Yuan Li
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Ting Wang
- School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taiwan
| | - Sheng-Hsiang Ma
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
- Department of Public Health, Institute of Public Health, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Li-Wei Lo
- Cardiovascular Center, Department of Internal Medicine, Taipei Veterans General Hospital, Taiwan
- Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chen-Yi Wu
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
- Department of Public Health, Institute of Public Health, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Wei-Chuan Chang
- Epidemiology and Biostatistics Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tai-Li Chen
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
| |
Collapse
|
96
|
Rodriguez CP, Burka S, Michos ED. Impact of Sex Differences on Lipids and Statin Utilization. Curr Atheroscler Rep 2025; 27:38. [PMID: 40100457 DOI: 10.1007/s11883-025-01286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE OF THE REVIEW Hypercholesterolemia is a significant and modifiable risk factor for cardiovascular disease (CVD), with Low-Density Lipoprotein Cholesterol (LDL-C) being the primary target for lipid-lowering therapies in both primary and secondary prevention. This review aims to explore the efficacy of statin therapy in women, its safety and application during pregnancy, and treatment disparities that contribute to undertreatment of dyslipidemia in women. RECENT FINDINGS Statins has demonstrated efficacy in reducing LDL-C and CVD risk in women. However, women are less likely to achieve LDL-C targets compared to men, largely due to undertreatment and delays in initiating therapy, often influenced by sex-specific factors. The unique considerations of lipid management during pregnancy, including suspension of statin therapy, present additional challenges in achieving optimal lipid control in high-risk women. Evidence also points to systemic disparities in healthcare delivery and treatment allocation, further exacerbating undertreatment of dyslipidemia among women. While LDL-C lowering remains a cornerstone of CVD prevention, women face distinct challenges in achieving lipid goals due to biological, clinical, and healthcare disparities. Addressing these barriers, including improving timely initiation of statins and addressing gaps in care during pregnancy, is essential to enhance the dyslipidemia management and reduce CVD risk in women.
Collapse
Affiliation(s)
- Carla P Rodriguez
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Semenawit Burka
- Division of Cardiology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD, USA.
| |
Collapse
|
97
|
Jiao P, Yang N, Jia Q, Fan B, Feng K, Yu J, Zhao S. A dual-reporter LDLR system integrating fluorescence and luminescence for understanding LDLR regulation and facilitating drug discovery. Front Mol Biosci 2025; 12:1552085. [PMID: 40182619 PMCID: PMC11966430 DOI: 10.3389/fmolb.2025.1552085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The low-density lipoprotein receptor (LDLR) is integral to cholesterol metabolism and cardiovascular health. Enhancing LDLR expression is a promising strategy for treating hyperlipidemia and reducing the risk of atherosclerosis. However, current LDLR reporter systems have limitations in detecting both transcriptional and translational regulation. To address this, we developed a novel dual-reporter LDLR system incorporating Enhanced Green Fluorescent Protein (EGFP) and Gaussia luciferase (Gluc) to enable precise monitoring of LDLR expression and function. Methods A CRISPR/Cas9-mediated knock-in strategy was used to integrate EGFP and Gluc upstream of the stop codon located in exon 18 of the LDLR gene in HEK293 cells. The dual-reporter system allows real-time visualization of LDLR expression via EGFP fluorescence and quantitative assessment through secreted Gluc activity. The system was validated using western blotting, immunofluorescence, and functional assays, including DiI-LDL uptake and drug response analyses with statins and PCSK9 inhibitors. Results The established LDLR-EGFP-Gluc knock-in cell line faithfully recapitulates endogenous LDLR expression and function. EGFP fluorescence accurately reflects LDLR expression dynamics, while Gluc activity provides a highly sensitive and quantitative readout. Functional assays confirmed that LDLR expression responds appropriately to statins and PCSK9 inhibitors. Additionally, screening for transcriptional regulators identified FOXP3 and CREB as novel modulators of LDLR expression, with CREB-mediated regulation involving the sterol regulatory element-binding protein 2 (SREBP2) pathway. Discussion This dual-reporter system enables complementary monitoring of LDLR dynamics, providing enhanced sensitivity, accuracy, and versatility for studying LDLR regulation and function, as well as facilitating drug discovery targeting hyperlipidemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Peng Jiao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Na Yang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Qianfeng Jia
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Baozhen Fan
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Ke Feng
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Jian Yu
- Department of Basic Medical Education, Yantai Nursing School, Yantai, Shandong, China
| | - Shengtian Zhao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
98
|
Mateo EM, Alonso RG, Sánchez CS, Tung-Chen Y, Rodilla E, Romero LB, García-Donaire JA, Bonilla-Hernández MV, Muñoz-Rivas N, Castilla-Guerra L. Positioning for the use of multivessel clinical ultrasound in vascular risk evaluation: VASUS+ protocol. 2024 recommendations of the Vascular Risk Group, clinical ultrasound of the Spanish Society of Internal Medicine and Spanish Society of Hypertension and Vascular Risk. HIPERTENSION Y RIESGO VASCULAR 2025:S1889-1837(24)00115-6. [PMID: 40118714 DOI: 10.1016/j.hipert.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 03/23/2025]
Abstract
Atherosclerosis is the underlying disease in the entire spectrum of atherosclerotic vascular disease. Point of care ultrasound is a useful tool for its detection. Current guidelines recommend the use of scales such as SCORE 2 and SCORE 2OP in apparently healthy individuals and in those at intermediate-low risk, they recognize the role of the arterial plaque by ultrasound to refine risk stratification and the need for more aggressive preventive strategies. However, the way to evaluate the vascular territories in which there is presence of plaque, the amount or load of plaque is not homogeneous nor is it well protocolized. In this document, 2 protocols are proposed for the evaluation of vascular risk, VASUS and VASUS+, including the presence of ventricular hypertrophy with the objective of homogenizing clinical ultrasound in the assessment of vascular risk in clinical practice.
Collapse
Affiliation(s)
- E Moya Mateo
- Medicina Interna, Unidad de Riesgo Vascular. Hospital Universitario Infanta Leonor, Madrid, Spain
| | - R García Alonso
- Medicina Interna, Complejo Asistencial de Ávila, Ávila, Spain
| | | | - Y Tung-Chen
- Medicina Interna, Hospital Universitario La Paz, Madrid, Spain
| | - E Rodilla
- Medicina Interna, Hospital Universitario de Sagunto, Universidad Cardenal Herrera-CEU, Sagunto, Spain
| | | | - J A García-Donaire
- Medicina Interna, Hospital Universitario Clínico San Carlos, Madrid, Spain; Universidad Complutense Madrid, Madrid, Spain
| | | | | | - L Castilla-Guerra
- Medicina Interna, Hospital Universitario Virgen Macarena, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
99
|
Lee Y, Shelke S, Lee C. Cardiac Repair and Regeneration via Advanced Technology: Narrative Literature Review. JMIR BIOMEDICAL ENGINEERING 2025; 10:e65366. [PMID: 40056468 PMCID: PMC11956377 DOI: 10.2196/65366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are the leading cause of death globally, and almost one-half of all adults in the United States have at least one form of heart disease. This review focused on advanced technologies, genetic variables in CVD, and biomaterials used for organ-independent cardiovascular repair systems. OBJECTIVE A variety of implantable and wearable devices, including biosensor-equipped cardiovascular stents and biocompatible cardiac patches, have been developed and evaluated. The incorporation of those strategies will hold a bright future in the management of CVD in advanced clinical practice. METHODS This study employed widely used academic search systems, such as Google Scholar, PubMed, and Web of Science. Recent progress in diagnostic and treatment methods against CVD, as described in the content, are extensively examined. The innovative bioengineering, gene delivery, cell biology, and artificial intelligence-based technologies that will continuously revolutionize biomedical devices for cardiovascular repair and regeneration are also discussed. The novel, balanced, contemporary, query-based method adapted in this manuscript defined the extent to which an updated literature review could efficiently provide research on the evidence-based, comprehensive applicability of cardiovascular devices for clinical treatment against CVD. RESULTS Advanced technologies along with artificial intelligence-based telehealth will be essential to create efficient implantable biomedical devices, including cardiovascular stents. The proper statistical approaches along with results from clinical studies including model-based risk probability prediction from genetic and physiological variables are integral for monitoring and treatment of CVD risk. CONCLUSIONS To overcome the current obstacles in cardiac repair and regeneration and achieve successful therapeutic applications, future interdisciplinary collaborative work is essential. Novel cardiovascular devices and their targeted treatments will accomplish enhanced health care delivery and improved therapeutic efficacy against CVD. As the review articles contain comprehensive sources for state-of-the-art evidence for clinicians, these high-quality reviews will serve as a first outline of the updated progress on cardiovascular devices before undertaking clinical studies.
Collapse
Affiliation(s)
- Yugyung Lee
- Division of Pharmacology and Pharmaceutics Sciences, School of Pharmacy, University of Missouri Kansas City, Kansas City, MO, United States
| | - Sushil Shelke
- Division of Pharmacology and Pharmaceutics Sciences, School of Pharmacy, University of Missouri Kansas City, Kansas City, MO, United States
| | - Chi Lee
- Division of Pharmacology and Pharmaceutics Sciences, School of Pharmacy, University of Missouri Kansas City, Kansas City, MO, United States
| |
Collapse
|
100
|
Jiang Z, Chen X, Lai Y, Liu J, Ye X, Chen P, Zhang Z. Early prediction of postpartum dyslipidemia in gestational diabetes using machine learning models. Sci Rep 2025; 15:8028. [PMID: 40055456 PMCID: PMC11889255 DOI: 10.1038/s41598-025-92299-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
This study addresses a gap in research on predictive models for postpartum dyslipidemia in women with gestational diabetes mellitus (GDM). The goal was to develop a machine learning-based model to predict postpartum dyslipidemia using early pregnancy clinical data, and the model's robustness was evaluated through both internal and temporal validation. Clinical data from 15,946 pregnant women were utilized. After cleaning, the data were divided into two sets: Dataset A (n = 1,116), used for training and evaluating the model, and Dataset B (n = 707), used for temporal validation. Several machine learning algorithms were applied, and the performance of the model was assessed with Dataset A, while Dataset B was used to validate the model across a different time period. Feature significance was evaluated through Information Value (IV), model importance analysis, and SHAP (SHapley Additive exPlanations) analysis. The results showed that among the five machine learning algorithms tested, tree-based ensemble models, such as XGBoost, LightGBM, and Random Forest, outperformed others in predicting postpartum dyslipidemia. In Dataset A, these models achieved accuracies of 70.54%, 70.54%, and 69.64%, respectively, with AUC-ROC values of 73.10%, 71.94%, and 76.14%. Temporal validation with Dataset B indicated that XGBoost performed best, achieving an accuracy of 81.05% and an AUC-ROC of 87.92%. The predictive power of the model was strengthened by key variables such as total cholesterol, fasting glucose, triglycerides, and BMI, with total cholesterol being identified as the most important feature. Further IV and SHAP analyses confirmed the pivotal role of these variables in predicting dyslipidemia. The study concluded that the XGBoost-based predictive model for postpartum dyslipidemia in GDM showed strong and consistent performance in both internal and temporal validations. By introducing new variables, the model can identify high-risk groups during early pregnancy, supporting early intervention and potentially improving pregnancy outcomes and reducing complications.
Collapse
Affiliation(s)
- Zhifa Jiang
- Obstetrics and Gynaecology, Huizhou First Maternal and Child Health Care Hospital, Huizhou, 516000, Guangdong, China
| | - Xiekun Chen
- School of Computer Science and Engineering, Huizhou University, Huizhou, 516000, Guangdong, China
| | - Yuhang Lai
- School of Computer Science and Engineering, Huizhou University, Huizhou, 516000, Guangdong, China
| | - Jingwen Liu
- Obstetrics and Gynaecology, Huizhou First Maternal and Child Health Care Hospital, Huizhou, 516000, Guangdong, China
| | - Xiangyun Ye
- Obstetrics and Gynaecology, Huizhou First Maternal and Child Health Care Hospital, Huizhou, 516000, Guangdong, China
| | - Ping Chen
- Obstetrics and Gynaecology, Huizhou First Maternal and Child Health Care Hospital, Huizhou, 516000, Guangdong, China
| | - Zhen Zhang
- School of Computer Science and Engineering, Huizhou University, Huizhou, 516000, Guangdong, China.
| |
Collapse
|