51
|
Pădureanu V, Forțofoiu MC, Pîrșcoveanu M, Pădureanu R, Rădulescu D, Donoiu I, Pîrșcoveanu DFV. Cardiovascular Manifestations of Patients with Non-Alcoholic Fatty Liver Disease. Metabolites 2025; 15:149. [PMID: 40137114 PMCID: PMC11943630 DOI: 10.3390/metabo15030149] [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: 01/11/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD), more recently redefined as metabolic-associated fatty liver disease (MAFLD), is now recognized as the most prevalent cause of chronic liver disease. Its strong association with cardiovascular disease (CVD) underscores its emerging role in global morbidity and mortality. Objective: This review critically examines the pathophysiological mechanisms that link NAFLD/MAFLD with CVD. It focuses on shared metabolic disturbances, inflammatory pathways, and alterations in the gut microbiota that contribute to hepatic and cardiovascular pathology. Review and Gaps: Current evidence highlights insulin resistance, dyslipidemia, systemic inflammation, and gut dysbiosis as pivotal factors connecting NAFLD/MAFLD to CVD. Despite these insights, inconsistencies in diagnostic criteria and a lack of validated non-invasive biomarkers hinder a clear understanding of the causal relationship between liver and cardiovascular diseases. Conclusions: Addressing these knowledge gaps through standardized diagnostic protocols and large-scale longitudinal studies is essential. Improved biomarker validation and clearer delineation of the underlying mechanisms will improve cardiovascular risk stratification and enable more personalized therapeutic strategies for patients with NAFLD/MAFLD.
Collapse
Affiliation(s)
- Vlad Pădureanu
- Department of Internal Medicine, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania; (V.P.); (M.C.F.)
| | - Mircea Cătălin Forțofoiu
- Department of Internal Medicine, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania; (V.P.); (M.C.F.)
| | - Mircea Pîrșcoveanu
- Department of Surgery, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | - Rodica Pădureanu
- Department of Internal Medicine, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania; (V.P.); (M.C.F.)
| | - Dumitru Rădulescu
- Department of Surgery, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | - Ionuț Donoiu
- Department of Cardiology, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | | |
Collapse
|
52
|
Meade R, Ibrahim D, Engel C, Belaygorod L, Arif B, Hsu FF, Adak S, Catlett R, Zhou M, Ilagan MXG, Semenkovich CF, Zayed MA. Targeting fatty acid synthase reduces aortic atherosclerosis and inflammation. Commun Biol 2025; 8:262. [PMID: 39972116 PMCID: PMC11840040 DOI: 10.1038/s42003-025-07656-1] [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: 04/09/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
Fatty acid synthase (FAS) is predominantly expressed in the liver and adipose tissue. It plays vital roles in de novo synthesis of saturated fatty acids and regulates insulin sensitivity. We previously demonstrated that serum circulating FAS (cFAS) is a clinical biomarker for advanced atherosclerosis, and that it is conjugated to low-density lipoproteins (LDL). However, it remains unknown whether cFAS can directly impact atheroprogression. To investigate this, we evaluate whether cFAS impacts macrophage foam cell formation - an important cellular process leading to atheroprogression. Macrophages exposed to human serum containing high levels of cFAS show increased foam cell formation as compared to cells exposed to serum containing low levels of cFAS. This difference is not observed using serum containing either high or low LDL. Pharmacological inhibition of cFAS using Platensimycin (PTM) decreases foam cell formation in vitro. In Apoe-/- mice with normal FAS expression, administration of PTM over 16 weeks along with a high fat diet decreases cFAS activity and aortic atherosclerosis without affecting circulating total cholesterol. This effect is also observed in Apoe-/- mice with liver-specific knockout of hepatic Fasn. Reductions in aortic root plaque are associated with decreased macrophage infiltration. These findings demonstrate that cFAS plays an important role in arterial atheroprogression.
Collapse
Affiliation(s)
- Rodrigo Meade
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Dina Ibrahim
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Connor Engel
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Larisa Belaygorod
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Batool Arif
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Fong-Fu Hsu
- Metabolism & Lipid Research, Division of Endocrinology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sangeeta Adak
- Metabolism & Lipid Research, Division of Endocrinology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan Catlett
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Mingzhou Zhou
- Department Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ma Xenia G Ilagan
- Department Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Clay F Semenkovich
- Metabolism & Lipid Research, Division of Endocrinology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed A Zayed
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Veterans Affairs St. Louis Health Care System, St. Louis, MO, USA.
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
- Division of Molecular Cell Biology, Washington University School of Medicine, St. Louis, MO, USA.
- McKelvey School of Engineering, Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
- CardioVascular Research Innovation in Surgery & Engineering Center, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Division of Surgical Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
53
|
Nicholls SJ, Nelson AJ. Achieving More Optimal Lipid Control with Non-Statin Lipid Lowering Therapy. Curr Atheroscler Rep 2025; 27:32. [PMID: 39954169 PMCID: PMC11829850 DOI: 10.1007/s11883-025-01280-4] [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: 02/07/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE OF REVIEW The use of statins has transformed approaches to the prevention of cardiovascular disease. However, many patients remain at a major risk of experiencing cardiovascular events, due to a range of factors including suboptimal control of low-density lipoprotein cholesterol (LDL-C). Accordingly, there is an ongoing need to develop additional strategies, beyond the use of statins, to achieve more effective reductions in cardiovascular risk. RECENT FINDINGS Genomic studies have implicated the causal role of LDL in atherosclerosis and identified that polymorphisms influencing factors involved in lipid metabolism influence both the level of LDL-C and cardiovascular risk. These findings have highlighted the potential for cardiovascular benefit from development of therapies targeting these factors and incremental benefit when used in combination with statins. Clinical trials have demonstrated that these new agents have favourable effects on both atherosclerotic plaque and cardiovascular events. Additional work has sought to improve intensification of statin therapy and adherence with lipid lowering therapy, to achieve more effective cardiovascular prevention via lipid lowering. Emerging therapies, beyond statins, have the potential to optimise lipid levels and play an effective role in the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne, Australia.
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne, Australia
| |
Collapse
|
54
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 DOI: 10.1161/jaha.124.038857] [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: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-Communicable Disease Management Beijing Children's Hospital, Capital Medical University, National Center for Children's Health Beijing China
| | - Qin Liu
- Department of Ultrasound Children's Hospital of the Capital Institute of Pediatrics Beijing China
| | - Hong Cheng
- Department of Epidemiology Capital Institute of Pediatrics Beijing China
| | - Xiaoyuan Zhao
- Department of Epidemiology Capital Institute of Pediatrics Beijing China
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control Department Shenzhen Center for Chronic Disease Control Shenzhen China
| | - Jie Mi
- Center for Non-Communicable Disease Management Beijing Children's Hospital, Capital Medical University, National Center for Children's Health Beijing China
- Key Laboratory of Major Diseases in Children, Ministry of Education China
| |
Collapse
|
55
|
Yang B, Manifold B, Han W, DeSousa C, Zhu W, Streets A, Titov DV. SRS microscopy identifies inhibition of vitellogenesis as a mediator of lifespan extension by caloric restriction in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.636008. [PMID: 40034647 PMCID: PMC11875241 DOI: 10.1101/2025.01.31.636008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The molecular mechanisms of aging are not fully understood. Here, we used label-free Stimulated Raman scattering (SRS) microscopy to investigate changes in proteins and lipids throughout the lifespan of C. elegans. We observed a dramatic buildup of proteins within the body cavity or pseudocoelom of aged adults that was blunted by interventions that extend lifespan: caloric restriction (CR) and the reduced insulin/insulin-like growth factor signaling (IIS) pathway. Using a combination of microscopy, proteomic analysis, and validation with mutant strains, we identified vitellogenins as the key molecular components of the protein buildup in the pseudocoelom. Vitellogenins shuttle nutrients from intestine to embryos and are homologous to human apolipoprotein B, the causal driver of cardiovascular disease. We then showed that CR and knockdown of vitellogenins both extend lifespan by >60%, but their combination has no additional effect on lifespan, suggesting that CR extends the lifespan of C. elegans in part by inhibiting vitellogenesis. The extensive dataset of more than 12,000 images stitched into over 350 whole-animal SRS images of C. elegans at different ages and subjected to different longevity intervention will be a valuable resource for researchers interested in aging.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
- These authors contributed equally
| | - Bryce Manifold
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- These authors contributed equally
| | - Wuji Han
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- These authors contributed equally
| | - Catherin DeSousa
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Wanyi Zhu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Aaron Streets
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Denis V. Titov
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- These authors contributed equally
| |
Collapse
|
56
|
Yang Z, Han X, Liang P, Zhao X, Zhu Q, Ye H, Yang C, Jiang B. Study on the Effects of National Volume-Based Procurement of Chemical Drugs on Chinese Patent Medicines: Lipid-Lowering Drugs as an Example. HEALTH CARE SCIENCE 2025; 4:14-24. [PMID: 40026643 PMCID: PMC11869369 DOI: 10.1002/hcs2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 03/05/2025]
Abstract
Background Atherosclerotic cardiovascular disease remains the leading cause of death worldwide. This study aims to explore the impact of national volume-based procurement (NVBP) on Chinese patent medicines and provide evidence for improving policies and promoting rational drug use. Methods The study was based on data from the China National Health Insurance Agency that spanned January 2019 to December 2020. Descriptive analysis was conducted using volume and expenditure as variables. Interrupted time series analysis was applied to further analyze Chinese patent medicines. Results The unit prices of atorvastatin and rosuvastatin decreased by 25%-96%, whereas the prices of Zhibitai and Xuezhikang fluctuated slightly. The affordability is measured as the monthly expenditure on treatment divided by the daily wage. After policy implementation, the affordability of atorvastatin and rosuvastatin improved from 0.242 to 0.014 and from 0.247 to 0.019, respectively. The defined daily doses (DDDs) for atorvastatin and rosuvastatin also increased, whereas total expenditures decreased in hospitals of all levels. Both at the national level and at all levels of hospital, the policy had no significant impact on expenditures for Zhibitai and Xuezhikang and their defined daily doses. Conclusions The NVBP saved costs in the short term by incorporating high-quality, widely used lipid-lowering drugs. Notably, the policy impacted lipid-lowering chemical drugs, whereas Chinese patent medicines remained largely unaffected. Doctors' use of Chinese patent medicines did not decline, highlighting the clinical specificity of these medicines.
Collapse
Affiliation(s)
- Zhao Yang
- Center for Digital Health and Artificial IntelligencePeking University First HospitalBeijingChina
- Research Center of Public PolicyPeking UniversityBeijingChina
| | - Xiao Han
- School of Public HealthShanghai Jiaotong UniversityShanghaiChina
| | - Pei Liang
- Women and Children's Hospital of Chongqing Medical UniversityChongqing Health Center for Women and ChildrenChongqingChina
| | - Xiaoting Zhao
- School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Qiyun Zhu
- Revelle CollegeUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Hui Ye
- TCM‐Integrated TCM & Western Medicine DeptPeking University First HospitalBeijingChina
| | - Chao Yang
- Center for Digital Health and Artificial IntelligencePeking University First HospitalBeijingChina
- Renal Division, Department of MedicinePeking University First Hospital, Peking University Institute of NephrologyBeijingChina
| | - Bin Jiang
- Research Center of Public PolicyPeking UniversityBeijingChina
- School of Pharmaceutical SciencesPeking UniversityBeijingChina
| |
Collapse
|
57
|
Chandran M, Rameshkumar KB, Jaleel A, Ayyappan JP. Embelin Elevates Endoplasmic Reticulum Calcium Levels and Blocks the Sterol Regulatory Element-Binding Protein 2 Mediated Proprotein Convertase Subtilisin/Kexin Type 9 Expression and Improves the Low-Density Lipoprotein Receptor Mediated Lipid Clearance on Hepatocytes. Chem Biol Drug Des 2025; 105:e70055. [PMID: 39902651 DOI: 10.1111/cbdd.70055] [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: 03/11/2024] [Revised: 10/03/2024] [Accepted: 01/04/2025] [Indexed: 02/06/2025]
Abstract
Cardiovascular diseases (CVDs) continue to be one of the leading causes of morbidity and mortality worldwide, with a significant increase in recent years. Atherosclerosis, the pathological basis and prime reason for CVDs is primarily driven by dysregulated lipid metabolism and inflammation. Recently, proprotein convertase subtilisin kexin9 (PCSK9) has been evolved to be highly implicated in the circulatory low-density lipoprotein cholesterol levels by its modulatory effects on the low-density lipoprotein receptor (LDLR) mediated clearance. Even though not economical, the therapies targeting PCSK9 demonstrated appreciable levels of efficiency in managing hyperlipidaemic conditions. Embelin (2,5-dihydroxy-3-undecyl-1,4-benzoquinone) is a naturally occurring para-benzoquinone isolated from dried berries of Embelia ribes, which possess several effects in maintaining the cholesterol homeostasis. In this study, we have analysed the role of embelin in sterol regulatory element-binding protein 2 (SREBP2) mediated PCSK9 expression in cultured hepatocytes. The study showed that the embelin treatment attenuates the endoplasmic reticulum (ER) stress-induced reactive oxygen species levels and ER stress markers on cultured hepatocytes. The treatment of embelin modulates the mRNA and protein level expression of SREBP2 and its downstream targets like PCSK9, LDLR, and HMG-CoA reductase (HMGCR). Interestingly the Ca2+ levels and the calcium binding protein of ER were significantly increased with embelin treatment. The work revealed a putative mechanism of embelin in lowering PCSK9 levels by boosting ER Ca2+ levels, thereby blocking SREBP2 nuclear translocation. Further, this reduces LDLR degradation and increases receptor-mediated circulatory lipid clearance. The study summarized the potential clinical applications of embelin in addressing the cardio vascular diseases.
Collapse
Affiliation(s)
- Mahesh Chandran
- Translational Nanomedicine and Lifestyle Disease Research Laboratory, Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, India
- DBT-SAHAJ National Facility for Mass Spectrometry, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - K B Rameshkumar
- Jawaharlal Nehru Tropical Botanic Garden and Research Institute Palode, Thiruvananthapuram, Kerala, India
| | - Abdul Jaleel
- DBT-SAHAJ National Facility for Mass Spectrometry, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Janeesh Plakkal Ayyappan
- Translational Nanomedicine and Lifestyle Disease Research Laboratory, Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, India
- Centre for Advanced Cancer Research, Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
| |
Collapse
|
58
|
Abayomi J, Charnley M, Stone G, Lane K, Stevenson L, Davies I, Webb R. Editorial. Proc Nutr Soc 2025; 84:1-7. [PMID: 38240093 DOI: 10.1017/s0029665124000089] [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: 02/16/2024]
Abstract
Nutritional requirements of individuals vary across the lifecycle, according to activity, age and gender. To optimize human health, consideration of nutritional priorities at each stage is needed. This conference brought together multidisciplinary experts in maternal and child nutrition and health, cardiometabolic and plant-based nutrition and dietitians involved in the care of vulnerable populations, plus nutritional metabolism, health and ageing. The presentations highlighted the most important nutrition research in these areas, updating knowledge and suggesting how dietary advice and policy could be adapted to incorporate research findings. With the global increase in non-communicable disease (NCD) and nutrition being considered as a key modifiable risk factor for the prevention and management of NCD, this conference was much needed.
Collapse
Affiliation(s)
- Julie Abayomi
- School of Medicine & Nutrition, Faculty of Health, Social Care and Medicine, Edge Hill University, OrmskirkL39 4PQ, UK
| | - Margaret Charnley
- School of Health & Sport Sciences, Liverpool Hope University, Liverpool, UK
| | - Genevieve Stone
- School of Medicine & Nutrition, Faculty of Health, Social Care and Medicine, Edge Hill University, OrmskirkL39 4PQ, UK
| | - Katie Lane
- Research Institute for Sport and Exercise Sciences (RISES), Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Leo Stevenson
- School of Health & Sport Sciences, Liverpool Hope University, Liverpool, UK
| | - Ian Davies
- Research Institute for Sport and Exercise Sciences (RISES), Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Richard Webb
- School of Health & Sport Sciences, Liverpool Hope University, Liverpool, UK
| |
Collapse
|
59
|
Zhang T, Huang Y, Ji X, Wu T, Xiao P. CCL11 (Eotaxin) Promotes the Advancement of Aging-Related Cardiovascular Diseases. Rev Cardiovasc Med 2025; 26:26020. [PMID: 40026499 PMCID: PMC11868897 DOI: 10.31083/rcm26020] [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: 08/06/2024] [Revised: 10/13/2024] [Accepted: 10/31/2024] [Indexed: 03/05/2025] Open
Abstract
Aging-related diseases, such as cardiovascular diseases (CVDs), neurodegeneration, cancer, etc., have become important factors that threaten the lifespans of older individuals. A chronic inflammatory response is closely related to aging-related diseases. Establishing inflammatory aging clock (iAGE, deep-learning methods on blood immune biomarkers to construct a metric for age-related chronic inflammation) successfully predicted the positive correlation between several factors, including serum C-C-motif chemokine ligand 11 (CCL11) and aging-related diseases. Recently, the role and mechanism of CCL11, an eosinophilic chemokine, in neurodegenerative diseases have been widely reported. Additionally, many research studies have shown a positive correlation with CVDs, but the underlying mechanism remains unknown. This review focuses on the relationship between chronic inflammation and aging. The role of CCL11 will be discussed and summarized in relation to aging-related diseases, especially CVDs.
Collapse
Affiliation(s)
- Tanwei Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, 211166 Nanjing, Jiangsu, China
| | - Yanhong Huang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, 211166 Nanjing, Jiangsu, China
| | - Xinmeng Ji
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, 211166 Nanjing, Jiangsu, China
| | - Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, 211166 Nanjing, Jiangsu, China
| | - Pingxi Xiao
- Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, 210031 Nanjing, Jiangsu, China
| |
Collapse
|
60
|
Davies IG. Exploring high-protein diets in the context of cardiac rehabilitation. Proc Nutr Soc 2025; 84:75-86. [PMID: 37877360 DOI: 10.1017/s0029665123004779] [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: 10/26/2023]
Abstract
The review aims to explore the potential benefit and risk of high-protein diets (HPD) regarding the comorbidity of sarcopoenia and CVD in the setting of cardiac rehabilitation (CR). CR is standard care for individuals who have experienced a cardiac event, but the current practice of predominantly aerobic exercise, a lower-fat diet and weight loss poorly addresses the issue of sarcopoenia. HPD, especially when combined with resistance exercise (RE), may be valuable adjuncts to current CR practice and benefit both muscle and cardiovascular health. Meta-analyses and randomised controlled trials of HPD and CVD risk show beneficial but variable effects regarding weight loss, the lipid profile, insulin resistance and lean body mass in those living with or high risk of CVD. Meta-analyses of prospective cohort studies on hard CVD endpoints favour lower- and plant-protein diets over higher animal protein, but the evidence is inconsistent. HPD augment the strength and muscle gaining benefits of RE in older populations, but there are no published data in those living with CVD providing promising opportunities for CR research. HPD raise concern regarding renal and bone health, the microbiome, branched chain amino acids and environmental sustainability and findings suggest that plant-based HPD may confer ecological and overall health advantages compared to animal-based HPD. However, incorporating RE with HPD might alleviate certain health risks. In conclusion, a largely plant-based HPD is deemed favourable for CR when combined with RE, but further research regarding efficacy and safety in CR populations is needed.
Collapse
Affiliation(s)
- Ian G Davies
- Research Institute of Sports and Exercise Sciences, Student Life Building, Liverpool John Moores University, Copperas Hill, Liverpool L3 5LJ, UK
| |
Collapse
|
61
|
Ajoolabady A, Pratico D, Mazidi M, Davies IG, Lip GYH, Seidah N, Libby P, Kroemer G, Ren J. PCSK9 in metabolism and diseases. Metabolism 2025; 163:156064. [PMID: 39547595 DOI: 10.1016/j.metabol.2024.156064] [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: 08/10/2024] [Revised: 10/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
PCSK9 is a serine protease that regulates plasma levels of low-density lipoprotein (LDL) and cholesterol by mediating the endolysosomal degradation of LDL receptor (LDLR) in the liver. When PCSK9 functions unchecked, it leads to increased degradation of LDLR, resulting in elevated circulatory levels of LDL and cholesterol. This dysregulation contributes to lipid and cholesterol metabolism abnormalities, foam cell formation, and the development of various diseases, including cardiovascular disease (CVD), viral infections, cancer, and sepsis. Emerging clinical and experimental evidence highlights an imperative role for PCSK9 in metabolic anomalies such as hypercholesterolemia and hyperlipidemia, as well as inflammation, and disturbances in mitochondrial homeostasis. Moreover, metabolic hormones - including insulin, glucagon, adipokines, natriuretic peptides, and sex steroids - regulate the expression and circulatory levels of PCSK9, thus influencing cardiovascular and metabolic functions. In this comprehensive review, we aim to elucidate the regulatory role of PCSK9 in lipid and cholesterol metabolism, pathophysiology of diseases such as CVD, infections, cancer, and sepsis, as well as its pharmaceutical and non-pharmaceutical targeting for therapeutic management of these conditions.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK; King's College London, Department of Twin Research & Genetic Epidemiology, South Wing St Thomas', London, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ian G Davies
- School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Copperas Hill, Liverpool L3 5AJ, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Nabil Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC H2W 1R7, Canada.
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
62
|
Thomsen MN, Skytte MJ, Samkani A, Weber P, Fenger M, Frystyk J, Hansen E, Holst JJ, Madsbad S, Magkos F, Thomsen HS, Walzem RL, Haugaard SB, Krarup T. Replacing dietary carbohydrate with protein and fat improves lipoprotein subclass profile and liver fat in type 2 diabetes independent of body weight: evidence from 2 randomized controlled trials. Am J Clin Nutr 2025; 121:224-231. [PMID: 39617302 DOI: 10.1016/j.ajcnut.2024.11.030] [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: 08/24/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Dyslipidemia with elevated concentrations of triacylglycerol-rich lipoproteins (TRLs), small-dense LDL, and reduced HDL is linked to hepatic steatosis and promotes atherogenesis in type 2 diabetes (T2D). OBJECTIVES We aimed to analyze whether moderate carbohydrate restriction reduces liver fat in T2D independent of changes in body weight and whether this is accompanied by parallel improvements in plasma lipoprotein subclasses. METHODS We determined the density profile of circulating lipoproteins in patients with T2D from 2 previous randomized controlled trials. In the isoenergetic study, 30 participants were allocated in a crossover design to 6 + 6 wk of an isocaloric carbohydrate-reduced high-protein (CRHP, C/P/F = 30/30/40 E%) or conventional diabetes (CD, C/P/F = 50/17/33 E%) diet aimed at weight maintenance. In the hypoenergetic study, 72 participants were allocated in a parallel-group design to 6 wk of a hypocaloric CRHP or CD diet aimed at matched ∼6% weight loss. Both studies provided all meals from a metabolic kitchen to maximize adherence. RESULTS In the isoenergetic study, the CRHP diet reduced TRL (mean: -33%; 95% CI: -48%, -14%) and LDL5 (mean: -16%; 95% CI: -26%, -4%) and increased HDL2/HDL3 (mean: 10%; 95% CI: 0%, 22%) compared with the CD diet. In the hypoenergetic study, weight loss induced by CRHP diet tended to reduce TRL (mean: -16%; 95% CI: -30%, 1%), reduced LDL5 (mean: -13%; 95% CI: -22%, -3%), and increased HDL2/HDL3 (mean: 11%; 95% CI: 1%, 22%) compared with an equivalent weight loss induced by CD diet. The CRHP diet decreased intrahepatic triacylglycerol (IHTG) more than the CD diet (isoenergetic: -55%; 95% CI: -74%, -22%; hypoenergetic: -26%; 95% CI: -45%, 0%), and changes in IHTG correlated directly with changes in TRL and LDL5 (r = 0.36-0.55; P < 0.01 for all) in both studies. CONCLUSIONS Replacing dietary carbohydrate with protein and fat improves dyslipidemia in T2D independently of changes in body weight, by inducing an atheroprotective shift in the lipoprotein particle profile possibly facilitated by reduced IHTG accumulation. These trials were registered at clinicaltrials.gov as NCT02764021 (https://clinicaltrials.gov/study/NCT02764021?term=NCT02764021&rank=1) and NCT03814694 (https://clinicaltrials.gov/study/NCT03814694?term=NCT03814694&rank=1).
Collapse
Affiliation(s)
- Mads N Thomsen
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.
| | - Mads J Skytte
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Philip Weber
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jan Frystyk
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Elizaveta Hansen
- Department of Radiology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Henrik S Thomsen
- Department of Radiology, Copenhagen University Hospital Herlev, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Rosemary L Walzem
- Graduate Faculty of Nutrition, Texas A&M University, College Station, Texas, United States
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark; Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
63
|
Zhang M, Yang D, Wang J, Wang D, Xu J, Wang Y. Association of serum lipidomic profiles with risk of intracranial aneurysm: A Mendelian randomization study. J Neurochem 2025; 169:e16247. [PMID: 39449543 DOI: 10.1111/jnc.16247] [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: 07/05/2024] [Revised: 09/01/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024]
Abstract
A two-sample Mendelian randomization (MR) analysis was utilized to assess the causal relationship between lipidomic profiles and the risk of intracranial aneurysms (IAs). Genetic variants related to lipidomic profiles (227 components) and IA [IA, aneurysmal subarachnoid hemorrhage (aSAH) only, unruptured IA (uIA) only] were obtained from published genome-wide association studies (GWASs) or the IEU Open GWAS project and used as instrumental variables for MR analysis. The inverse-variance weighted method was used in the primary analyses to derive causality estimates and was expressed as odds ratio (OR) with 95% confidence interval (CI). Of these 227 lipidomic profiles, only genetically predicted high levels of cholesterol to total lipids ratio in very small very-low-density lipoproteins (VLDL) [OR = 0.629 (95% CI, 0.504-0.786)], cholesteryl esters to total lipids ratio in very small VLDL [OR = 0.637 (95% CI, 0.509-0.797)], ratio of docosahexaenoic acid to total fatty acids [OR = 0.691 (95% CI, 0.582-0.820)], and ratio of polyunsaturated fatty acids to monounsaturated fatty acids [OR = 0.630 (95% CI, 0.522-0.760)] reduced the risk of aSAH, whereas genetically predicted high ratio of monounsaturated fatty acids to total fatty acids [OR = 1.471 (95% CI, 1.215-1.781)] increased the risk of aSAH. Moreover, genetically predicted high levels of cholesterol to total lipids ratio in very small VLDL [OR = 0.657 (95% CI, 0.542-0.798)], cholesteryl esters to total lipids ratio in very small VLDL [OR = 0.663 (95% CI, 0.548-0.803)], free cholesterol to total lipids ratio in small VLDL [OR = 0.682 (95% CI, 0.560-0.832)], phospholipids to total lipids ratio in small VLDL [OR = 0.674 (95% CI, 0.548-0.830)], and ratio of polyunsaturated fatty acids to monounsaturated fatty acids [OR = 0.678 (95% CI, 0.569-0.808)] reduced the risk of IA. The results of multivariable MR demonstrated that these causal associations persisted after adjusting for systolic blood pressure and cigarettes smoked per day. The effect of serum lipids on IA and aSAH may be mainly caused by subclasses of lipids such as VLDL.
Collapse
Affiliation(s)
- Mingqin Zhang
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Dongyi Yang
- Interventional Department, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Jiabin Wang
- Interventional Department, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Dan Wang
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Jin Xu
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Yibo Wang
- Interventional Department, Henan Province Hospital of TCM, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
64
|
Griffin BA, Lovegrove JA. Saturated fat and CVD: importance of inter-individual variation in the response of serum low-density lipoprotein cholesterol. Proc Nutr Soc 2025; 84:87-97. [PMID: 38282001 DOI: 10.1017/s0029665124000107] [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: 01/30/2024]
Abstract
The aim of this review is to provide an overview of the history in support of the role of dietary saturated fatty acids (SFA) in the development of cardiovascular disease (CVD), and the controversy and consensus for the evidence in support of guidelines to remove and replace SFA with unsaturated fatty acids. The review will also examine the existence, origins, and implications for CVD risk of variability in serum LDL-cholesterol in response to these guidelines. While the quality of supporting evidence for the efficacy of restricting SFA on CVD risk has attracted controversy, this has helped to increase understanding of the inter-relationships between SFA, LDL-cholesterol and CVD, and reinforce confidence in this dietary recommendation. Nevertheless, there is significant inter-individual variation in serum LDL-C in response to this dietary change. The origins of this variation are multi-factorial and involve both dietary and metabolic traits. If serum biomarkers of more complex metabolic traits underlying LDL-responsiveness can be identified, this would have major implications for the targeting of these dietary guidelines to LDL-responders, to maximise the benefit to their cardiovascular health.
Collapse
Affiliation(s)
- Bruce A Griffin
- Department of Nutrition, Food & Exercise Sciences, University of Surrey, GuildfordGU2 7XH, UK
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular & Metabolic Research, Department of Food and Nutritional Sciences, University of Reading, ReadingRG6 6DZ, UK
| |
Collapse
|
65
|
Swirski FK, Binder CJ. Lower your cholesterol early, and stick with it! Nat Rev Cardiol 2025; 22:69-70. [PMID: 39424909 DOI: 10.1038/s41569-024-01095-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Affiliation(s)
- Filip K Swirski
- Cardiovascular Research Institute, Friedman Brain Institute, Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
66
|
Li X, Li ZF, Wu NQ. Remnant Cholesterol and Residual Risk of Atherosclerotic Cardiovascular Disease. Rev Cardiovasc Med 2025; 26:25985. [PMID: 40026498 PMCID: PMC11868899 DOI: 10.31083/rcm25985] [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: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 03/05/2025] Open
Abstract
Remnant cholesterol (RC) is increasingly recognized as a key target in the treatment of atherosclerotic cardiovascular disease (ASCVD), addressing much of the residual risk that persists despite standard therapies. However, integrating RC into clinical practice remains challenging. Key issues, such as the development of accessible RC measurement methods, the identification of safe and effective medications, the determination of optimal target levels, and the creation of RC-based risk stratification strategies, require further investigation. This article explores the complex role of RC in ASCVD development, including its definition, metabolic pathways, and its association with both the overall risk and residual risk of ASCVD in primary and secondary prevention. It also examines the effect of current lipid-lowering therapies on RC levels and their influence on cardiovascular outcomes. Recent research has highlighted promising advancements in therapies aimed at lowering RC, which show potential for reducing major adverse cardiovascular events (MACEs). Inhibitors such as angiopoietin-like protein 3 (ANGPTL3), apolipoprotein C-III (apoCIII), and proprotein convertase subtilisin/kexin type 9 (PCSK9) have demonstrated their ability to modulate RC and reduce MACEs by targeting specific proteins involved in RC synthesis and metabolism. There is a pressing need for larger randomized controlled trials to clarify the role of RC in relevant patient populations. The development of targeted RC-lowering therapies holds the promise of significantly reducing the high rates of morbidity and mortality associated with ASCVD.
Collapse
Affiliation(s)
- Xi Li
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| | - Zhi-Fan Li
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| | - Na-Qiong Wu
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| |
Collapse
|
67
|
Islam MM, Tamlander M, Hlushchenko I, Ripatti S, Pfisterer SG. Large-Scale Functional Characterization of Low-Density Lipoprotein Receptor Gene Variants Improves Risk Assessment in Cardiovascular Disease. JACC Basic Transl Sci 2025; 10:170-183. [PMID: 40131152 PMCID: PMC11897452 DOI: 10.1016/j.jacbts.2024.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 03/26/2025]
Abstract
Limited access to functional information of genetic variants reduces the applicability of genetic tools for precision medicine applications in cardiovascular disease. We established an automated analysis platform based on multiplexed high-content imaging and derived in-depth functional data for several hundred LDLR gene variants. Residual low-density lipoprotein receptor activity of genetic variants impacted the risk for cardiovascular disease and elevated low-density lipoprotein cholesterol as well as the utilization of lipid-lowering and combination therapy. This enables increased risk stratification for carriers of LDLR gene variants and opens up new opportunities for improved diagnosis, risk assessment, and treatment selection in familial hypercholesterolemia.
Collapse
Affiliation(s)
| | - Max Tamlander
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Iryna Hlushchenko
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Simon G Pfisterer
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
68
|
Zhai Q, Shang S, Zhang Z, Sun L, Huang Y, Feng S, Wu Q, Cui H, Shi X. Mechanism of salvianolic phenolic acids and hawthorn triterpenic acids combination in intervening atherosclerosis: network pharmacology, molecular docking, and experimental validation. Front Pharmacol 2025; 16:1501846. [PMID: 39950115 PMCID: PMC11821658 DOI: 10.3389/fphar.2025.1501846] [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: 09/25/2024] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
Background This study employs network pharmacology and molecular docking methods in conjunction with animal experimentation to elucidate the underlying mechanism by which the combination of salvianolic phenolic acids and hawthorn triterpenic acids (SHC) exerts its therapeutic effect on carotid atherosclerosis (AS) in ApoE-/- mice. Methods A network pharmacology research approach was used to predict potential core targets for SHC intervention in atherosclerosis. The predictions were subsequently validated through the implementation of animal in vivo experiments. ApoE-/- mice were randomly assigned to three experimental groups, namely, a model group, an atorvastatin group, and an SHC group. After the administration period, the plaque area in the carotid artery and aortic arch, blood lipid levels, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), and nitric oxide (NO) content were measured. Additionally, the expression of PI3K, Akt, NF-κB, JNK1, ERK1/2, and p38-MAPK in the aortic arteries was analyzed. Based on the protein expression results, molecular docking was used to predict the binding activity between the core compounds and core targets. Results A total of 23 core compounds were identified in SHC, and 55 core targets of SHC were screened as potential targets for intervention in AS. The results of the enrichment analysis indicated that the principal mechanisms through which SHC exerts its effects in AS are associated with lipid metabolism and the PI3K-Akt and MAPK pathways. The results from animal experiments demonstrated that atorvastatin and SHC markedly reduced the area of carotid plaque and downregulated the levels of TC and LDL-C in ApoE-/- mice. The administration of SHC was associated with an increase in SOD activity and a reduction in NO levels in the livers of mice. Furthermore, SHC was observed to downregulate the expression of NF-κB and p38-MAPK in the carotid region. The results of molecular docking demonstrated that the core compounds of SHC, including salvianolic acid A, B, and C, maslinic acid, ursolic acid, and oleic acid, were capable of stably binding to the core targets NF-κB and MAPK14. Conclusion It is hypothesized that SHC may reduce lipid deposition and plaque formation in AS by regulating blood lipids, a process that may be closely linked to the inhibition of inflammatory regulator expression, including NF-κB and p38-MAPK.
Collapse
Affiliation(s)
- Qu Zhai
- Institute of Executive Development, China National Medical Products Administration, Beijing, China
| | - Shixi Shang
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zihan Zhang
- Beijing University of Chinese Medical, Beijing, China
| | - Lihua Sun
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Huang
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuyi Feng
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Wu
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haifeng Cui
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Shi
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
69
|
Deng Y, Liu L, Li Y, Ma H, Li C, Yan K, Tian J, Li C. pH-sensitive nano-drug delivery systems dual-target endothelial cells and macrophages for enhanced treatment of atherosclerosis. Drug Deliv Transl Res 2025:10.1007/s13346-025-01791-2. [PMID: 39881105 DOI: 10.1007/s13346-025-01791-2] [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] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by vascular endothelial dysfunction. In the early stage of the disease, endothelial cell injury induces the infiltration of inflammatory macrophages, which secrete large amounts of inflammatory factors, further aggravating endothelial cell dysfunction and exacerbating the disease. Therefore, it is promising for co-targeting endothelial cells and macrophages further regulating the inflammatory microenvironment and endothelial cell function for effective treatment. The current nano-drug delivery system (NDDS) for AS treatment is mainly focused on anti-inflammatory therapy, while ignoring the potential value of suppressing inflammation and simultaneously improving vascular endothelial function. In this study, a pH-responsive dual-targeted NDDS based on plaque microenvironment, BC@CS/cRGD NPs, was prepared by combining baicalin (BC) with chondroitin sulfate (CS) through amidation reaction, and further modified with a targeting group cRGD peptide. In vitro release experiments illustrated a faster release of the nanoparticle at pH 5.0 than at pH 7.4. Meanwhile, in vitro cellular experiments demonstrated its ability to target activated endothelial cells and macrophages. In a mouse model of AS, BC@CS/cRGD NPs accumulated at plaque sites and effectively attenuated the plaque progression. In conclusion, this pH-sensitive BC@CS/cRGD NPs offered a very potential strategy for modulating endothelial dysfunction and inflammatory microenvironment for the treatment of AS.
Collapse
Affiliation(s)
- Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Li Liu
- Department of Anaesthesiology, The affiliated hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yao Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Science and Technology Department, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Huan Ma
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Kexin Yan
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
70
|
Garnås E. Saturated fat in an evolutionary context. Lipids Health Dis 2025; 24:28. [PMID: 39875911 PMCID: PMC11773866 DOI: 10.1186/s12944-024-02399-0] [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: 06/16/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025] Open
Abstract
Evolutionary perspectives have yielded profound insights in health and medical sciences. A fundamental recognition is that modern diet and lifestyle practices are mismatched with the human physiological constitution, shaped over eons in response to environmental selective pressures. This Darwinian angle can help illuminate and resolve issues in nutrition, including the contentious issue of fat consumption. In the present paper, the intake of saturated fat in ancestral and contemporary dietary settings is discussed. It is shown that while saturated fatty acids have been consumed by human ancestors across time and space, they do not feature dominantly in the diets of hunter-gatherers or projected nutritional inputs of genetic accommodation. A higher intake of high-fat dairy and meat products produces a divergent fatty acid profile that can increase the risk of cardiovascular and inflammatory disease and decrease the overall satiating-, antioxidant-, and nutrient capacity of the diet. By prioritizing fiber-rich and micronutrient-dense foods, as well as items with a higher proportion of unsaturated fatty acids, and in particular the long-chain polyunsaturated omega-3 fatty acids, a nutritional profile that is better aligned with that of wild and natural diets is achieved. This would help prevent the burdening diseases of civilization, including heart disease, cancer, and neurodegenerative conditions. Saturated fat is a natural part of a balanced diet; however, caution is warranted in a food environment that differs markedly from the one to which we are adapted.
Collapse
Affiliation(s)
- Eirik Garnås
- Institute of Health, Oslo New University College, Ullevålsveien 76, Oslo, 0454, Norway.
| |
Collapse
|
71
|
van Oortmerssen JAE, Mulder JWCM, van der Bijl MF, Mijnster RJM, Kavousi M, Roeters van Lennep JE. Lipid Lowering Therapy Utilization and Lipid Goal Attainment in Women. Curr Atheroscler Rep 2025; 27:29. [PMID: 39873822 PMCID: PMC11775078 DOI: 10.1007/s11883-025-01275-1] [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: 01/03/2025] [Indexed: 01/30/2025]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of the current status of lipid-lowering therapy utilization and lipid goal attainment in women. We focus on lipid-lowering therapy in individuals with and without established atherosclerotic cardiovascular disease, as well as familial hypercholesterolemia. Additionally, this review aims to explore the underlying mechanisms driving these sex differences and to identify existing knowledge gaps in this area. RECENT FINDINGS Despite the proven efficacy of lipid-lowering therapy in both sexes, real-world studies indicate that women with comparable risk profiles are less likely than men to receive these treatments. Furthermore, women who are prescribed statins typically receive lower-intensity regimens than men and are less likely to achieve guideline-recommended low-density lipoprotein cholesterol goals. Despite advancements in lipid-lowering therapies, women compared to men, are systematically undertreated. This difference is influenced by patient-related, physician-related, and societal factors.
Collapse
Affiliation(s)
| | - Janneke W C M Mulder
- Department of Internal Medicine, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marte F van der Bijl
- Department of Internal Medicine, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ruben J M Mijnster
- Department of Internal Medicine, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands.
| |
Collapse
|
72
|
Hua S, Zhang H, Li J, Zhou X, Zhang S, Zhu Y, Yan X, Gu P, Huang Z, Jiang W. Astragaloside IV ameliorates atherosclerosis by targeting TAK1 to suppress endothelial cell proinflammatory activation. Int Immunopharmacol 2025; 146:113842. [PMID: 39706043 DOI: 10.1016/j.intimp.2024.113842] [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: 08/28/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease mainly characterized by the activation of endothelial cells and recruitment of macrophages, leading to plaque formation. Astragaloside IV (AS-IV), a natural saponin derived from Astragalus mongholicus Bunge, has been shown to confer protective effects against cardiovascular diseases. PURPOSE The purpose of this study is to explore the role of AS-IV on atherosclerosis and the underlying mechanism. METHODS Mice with atherosclerosis were administered with AS-IV by oral gavage. Atherosclerotic plaques and blood lipid profiles of these mice were assessed. Endothelial cell activation and macrophage infiltration were examined by immunofluorescent or immunohistochemical staining. The effects of AS-IV on endothelial cell activation, macrophage migration and adhesion were determined by transwell experiments, RT-qPCR, and Western blot. RESULTS Mice treated with AS-IV exhibited a dose-dependent reduction in atherosclerotic plaque size, with no concomitant change in blood lipid levels. It significantly suppressed endothelial cell activation and macrophage infiltration in the vasculature. AS-IV inhibited TNF-α-induced endothelial cell activation and macrophage migration and adhesion in vitro. Furthermore, AS-IV reduced the phosphorylation of key kinases in the MAPK pathways and their upstream regulator TAK1 in endothelial cells. The inhibitory effects of AS-IV on MAPK pathways and endothelial cell activation were counteracted by TAK1 deficiency or overexpression of TAK1. Molecular docking analysis suggested AS-IV binds to TAK1 with high affinity. CONCLUSION AS-IV exhibits anti-atherosclerotic effects by targeting TAK1 in endothelial cells, thereby inhibiting endothelial cell activation, and the subsequent adhesion and migration of macrophages, providing a prospective therapeutic strategy for the management of atherosclerosis.
Collapse
Affiliation(s)
- Shuang Hua
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jixu Li
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaonian Zhou
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujie Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingqun Yan
- Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Gu
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China; Southeast University, School of Medicine, Nanjing, China.
| | - Zhe Huang
- Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China; Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China; Department of Cardiology, Shanghai Pudong New Area People's Hospital, Shanghai, China.
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
73
|
Tian Y, Kong Y, Liu X, Chen L, Wang L, Zhou L, Wang G, Zhang Q. Regenerated carboxymethyl cellulose beads for selective removal of low-density lipoprotein from whole blood. Carbohydr Polym 2025; 348:122848. [PMID: 39562119 DOI: 10.1016/j.carbpol.2024.122848] [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: 08/09/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 11/21/2024]
Abstract
Low-density lipoprotein (LDL) plays a crucial role in the development of cardiovascular disease. Lowering the level of LDL is an effective therapeutic strategy for treating cardiovascular disease. Here, we developed a facile and robust method to prepare carboxymethyl cellulose beads (CMCBs) for selectively adsorbing LDL. CMCBs had plentiful carboxyl groups and large numbers of nanopores on their surface. The in vitro assay reveals that CMCBs had a high LDL adsorbing capability of 7.67 ± 0.16 mg/g, owing to the carboxyl group-induced electrostatic adsorption and the nanopore-mediated trapping effect. CMCBs also possessed the excellent mechanical strength to resist the impact of rushing blood. Moreover, CMCBs were highly blood-compatible and had anticoagulant activity. The in vivo experiment reveals that LDL were significantly reduced from 15.02 ± 1.62 to 9.35 ± 1.71 mmol/L after blood perfusion using CMCBs, while minimal side effects were detected for the blood routine parameters. The study provides an easy-to-fabricate adsorbent for selective and efficient clearance of LDL in hemoperfusion.
Collapse
Affiliation(s)
- Yichen Tian
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Yuele Kong
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Xiaodi Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Lei Chen
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Lin Zhou
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China.
| | - Guodong Wang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China.
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, PR China.
| |
Collapse
|
74
|
Song Q, Zhang C, Wang W, Wang C, Yi C. Exploring the genetic landscape of the brain-heart axis: A comprehensive analysis of pleiotropic effects between heart disease and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111172. [PMID: 39423935 DOI: 10.1016/j.pnpbp.2024.111172] [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: 06/27/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The genetic links between heart disease and psychiatric disorders are complex and not well understood. This study uses genome-wide association studies (GWAS) and advanced multilevel analyses to explore these connections. METHODS We analyzed GWAS data from seven psychiatric disorders and five types of heart disease. Genetic correlations and overlaps were examined using linkage disequilibrium score regression (LDSC), high-definition likelihood (HDL), and Genetic analysis incorporating Pleiotropy and Annotation (GPA). Pleiotropic single-nucleotide variations (SNVs) were identified with pleiotropic analysis under the composite null hypothesis (PLACO) and annotated via Functional mapping and annotation of genetic associations (FUMA). Potential pleiotropic genes were identified using Multi-marker Analysis of GenoMic Annotation (MAGMA) and Summary data-based Mendelian Randomization (SMR). RESULTS Among 35 trait pairs, 32 showed significant genetic correlations or overlaps. PLACO identified 15,077 SNVs, with 287 recognized as pleiotropic loci and 20 colocalization sites. MAGMA and SMR revealed 75 potential pleiotropic genes involved in diverse pathways, including cancer, neurodevelopment, and cellular organization. Mouse Genome Informatics (MGI) queries provided evidence linking multiple genes to heart or psychiatric disorders. CONCLUSIONS This analysis reveals loci and genes with pleiotropic effects between heart disease and psychiatric disorders, highlighting shared biological pathways. These findings illuminate the genetic mechanisms underlying the brain-heart axis and suggest shared biological foundations for these conditions, offering potential targets for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Qifeng Song
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Cheng Zhang
- Nanjing Vocational Health College, Nanjing, Jiangsu 210000, China
| | - Wei Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Cihan Wang
- Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
75
|
Lin Z, Yi T, Hu F, Chen J, Chen L. U-shaped association between the non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and mortality risk in obese adults: evidence from NHANES 1999-2018. Front Cardiovasc Med 2025; 11:1524465. [PMID: 39866802 PMCID: PMC11759299 DOI: 10.3389/fcvm.2024.1524465] [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: 11/07/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Background Obesity, often accompanied by dyslipidemia and increased cardiovascular risk, poses a significant threat to overall mortality. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) has been identified as a valuable parameter for assessing dyslipidemia. The goal of the study was to elucidate the relationship between NHHR and mortality in obese populations. Methods Data for the study cohort were sourced from the National Health and Nutrition Examination Survey (1999-2018). The association between NHHR and mortality from all causes and cardiovascular disease was examined through multivariable Cox regression and restricted cubic splines (RCS). Segmented multivariable Cox regression and subgroup analyses were conducted when segmented effects were identified. The reliability of the results was confirmed through multiple sensitivity analyses. Results A total of 7,504 participants were included in the analysis. During a median follow-up of 119 months, 866 subjects died for all causes, of which 318 were related to cardiovascular diseases. A U-shaped association was found utilizing RCS analysis, with cardiovascular mortality and all-cause mortality exhibiting the lowest risk points at 3.409 and 3.369, respectively. The fully adjusted model revealed a negative relationship between the risk of cardiovascular mortality (HR = 0.68, 95% CI: 0.49-0.94) and all-cause mortality (HR = 0.82, 95% CI: 0.67-1.00) for per 1 mmol/L increase in NHHR levels below the cut-off value. On the other hand, above the cut-off point, NHHR was positively correlated with cardiovascular mortality (HR = 1.18, 95% CI: 1.02-1.36) and all-cause mortality (HR = 1.13, 95% CI: 1.01-1.28). The sensitivity results of this study were in accordance with earlier findings, and no significant interactions in NHHR levels were discovered across different subgroups. Conclusions In the obese adults, NHHR displayed a U-shaped relationship with cardiovascular and all-cause death. Monitoring and managing NHHR levels in obese population may help mitigate the risk of mortality.
Collapse
Affiliation(s)
- Zi Lin
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Cardiovascular Medical Center, Fuzhou, China
- Fujian Provincial Coronary Heart Disease Research Institute, Fuzhou, China
| | - Tao Yi
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Cardiovascular Medical Center, Fuzhou, China
- Fujian Provincial Coronary Heart Disease Research Institute, Fuzhou, China
| | - Feng Hu
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Cardiovascular Medical Center, Fuzhou, China
- Fujian Provincial Coronary Heart Disease Research Institute, Fuzhou, China
| | - Jinhua Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Cardiovascular Medical Center, Fuzhou, China
- Fujian Provincial Coronary Heart Disease Research Institute, Fuzhou, China
| |
Collapse
|
76
|
Di Giacomo-Barbagallo F, Andreychuk N, Scicali R, Gonzalez-Lleó A, Piro S, Masana L, Ibarretxe D. Inclisiran, Reasons for a Novel Agent in a Crowded Therapeutic Field. Curr Atheroscler Rep 2025; 27:25. [PMID: 39786678 PMCID: PMC11717820 DOI: 10.1007/s11883-024-01271-x] [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: 12/19/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE OF THE REVIEW A significant number of patients fail to achieve target LDL cholesterol (LDL-C) levels. This review aims to explore why inclisiran, a novel class of LLT, should be considered a valuable addition to the current treatment options. RECENT FINDINGS Inclisiran is a small interfering RNA (siRNA) that targets PCSK9 synthesis specifically in the hepatocytes. The drug remains in circulation for less than 48 h, but its effect lasts for over six months. Two subcutaneous injections per year consistently lowers LDL-C by approximately 55% with a favorable safety profile. In combination with other LLTs, it can achieve LDL-C reductions of over 80%, supporting its role in high-intensity LLT strategies. Inclisiran represents a novel class of LLT. Administered biannually, reduces baseline LDL-C levels by half. Additionally, it has a strong safety profile. Due to its pharmacokinetic properties, is likely to improve adherence to LLT and persistently maintain low LDL-C levels.
Collapse
Affiliation(s)
- Francesco Di Giacomo-Barbagallo
- Unitat de Medicina Vascular I Metabolismo, Hospital Universitario Sant Joan, Universitat Rovira I Virgili, IISPV, CIBERDEM, Reus, Spain
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122, Catania, Italy
| | - Natalia Andreychuk
- Unitat de Medicina Vascular I Metabolismo, Hospital Universitario Sant Joan, Universitat Rovira I Virgili, IISPV, CIBERDEM, Reus, Spain
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122, Catania, Italy
| | - Ana Gonzalez-Lleó
- Unitat de Medicina Vascular I Metabolismo, Hospital Universitario Sant Joan, Universitat Rovira I Virgili, IISPV, CIBERDEM, Reus, Spain
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122, Catania, Italy
| | - Lluis Masana
- Unitat de Medicina Vascular I Metabolismo, Hospital Universitario Sant Joan, Universitat Rovira I Virgili, IISPV, CIBERDEM, Reus, Spain.
- Faculty of Medicine, Universitat Rovira I Virgili, C/ Sant Llorenç, 21 43201, Reus, Spain.
| | - Daiana Ibarretxe
- Unitat de Medicina Vascular I Metabolismo, Hospital Universitario Sant Joan, Universitat Rovira I Virgili, IISPV, CIBERDEM, Reus, Spain
| |
Collapse
|
77
|
Lan NSR, Chen RT, Dwivedi G, Watts GF, Nicholls SJ, Nelson AJ. Learnings from Implementation Strategies to Improve Lipid Management. Curr Cardiol Rep 2025; 27:9. [PMID: 39775142 PMCID: PMC11711772 DOI: 10.1007/s11886-024-02174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW Lowering low-density lipoprotein (LDL)-cholesterol reduces cardiovascular risk. International lipid management guidelines recommend LDL-cholesterol goals or thresholds for initiating lipid-lowering therapy. However, contemporary real-world studies have shown that many high- and very high-risk patients are not attaining LDL-cholesterol goals and are not receiving intensive lipid-lowering therapies. In this review, recent examples of implementation strategies for optimising lipid management are discussed. RECENT FINDINGS Implementation studies are heterogenous in their strategies and design. At the clinician level, multidisciplinary team-based care (including multidisciplinary lipid clinics), pharmacist- or nurse-led interventions, decision-support algorithms or protocols, and educational initiatives have shown potential to improve lipid management. Various strategies to improve patient adherence to lipid-lowering therapies have demonstrated at least short-term efficacy, including education, shared decision-making, behavioural support and nudges. Electronic health records can be leveraged at low cost to identify patients requiring initiation or intensification of lipid-lowering therapies, but the optimal method of integrating automated alerts or nudges to influence decision-making requires further research. Moreover, telehealth and remote care delivery models can improve access to healthcare and facilitate lipid-lowering. Multifaceted strategies with a systematic approach to targeting clinician, patient and system related factors can be successful in improving lipid management. Future implementation research should evaluate longer-term outcomes and follow implementation science theories, models and/or frameworks at all stages. By doing so, ongoing implementation studies will help researchers better understand the impact, sustainability and scalability of strategies, and where barriers and facilitators to lipid management may exist in other contexts.
Collapse
Affiliation(s)
- Nick S R Lan
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA, Australia.
- Medical School, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, WA, 6009, Australia.
- Harry Perkins Institute of Medical Research, Perth, WA, Australia.
| | | | - Girish Dwivedi
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA, Australia
- Medical School, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, WA, 6009, Australia
- Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | - Gerald F Watts
- Medical School, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, WA, 6009, Australia
- Departments of Internal Medicine and Cardiology, Royal Perth Hospital, Perth, WA, Australia
| | | | - Adam J Nelson
- Victorian Heart Institute, Monash University, Clayton, VIC, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
78
|
Kirsch A, Gindlhuber J, Zabini D, Osto E. Bile acids and incretins as modulators of obesity-associated atherosclerosis. Front Cardiovasc Med 2025; 11:1510148. [PMID: 39834741 PMCID: PMC11743266 DOI: 10.3389/fcvm.2024.1510148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Obesity is one of the major global health concerns of the 21st century, associated with many comorbidities such as type 2 diabetes mellitus (T2DM), metabolic dysfunction-associated steatotic liver disease, and early and aggressive atherosclerotic cardiovascular disease, which is the leading cause of death worldwide. Bile acids (BAs) and incretins are gut hormones involved in digestion and absorption of fatty acids, and insulin secretion, respectively. In recent years BAs and incretins are increasingly recognized as key signaling molecules, which target multiple tissues and organs, beyond the gastro-intestinal system. Moreover, incretin-based therapy has revolutionized the treatment of T2DM and obesity. This mini review highlights the current knowledge about dysregulations in BA homeostasis in obesity with a special focus on atherosclerosis as well as athero-modulating roles of incretins and currently available incretin-based therapies.
Collapse
Affiliation(s)
- Andrijana Kirsch
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Juergen Gindlhuber
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Diana Zabini
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
- Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
79
|
Ko S, Anzai A, Liu X, Kinouchi K, Yamanoi K, Torimitsu T, Ichihara G, Kitakata H, Shirakawa K, Katsumata Y, Endo J, Hayashi K, Yoshida M, Nishimori K, Tanaka KF, Onaka T, Sano M, Ieda M. Social Bonds Retain Oxytocin-Mediated Brain-Liver Axis to Retard Atherosclerosis. Circ Res 2025; 136:78-90. [PMID: 39601150 DOI: 10.1161/circresaha.124.324638] [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: 03/25/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUNDS Social interaction with others is essential to life. Although social isolation and loneliness have been implicated as increased risks of cardiometabolic and cardiovascular diseases and all-cause mortality, the cellular and molecular mechanisms by which social connection maintains cardiometabolic and cardiovascular health remain largely unresolved. METHODS To investigate how social connection protects against cardiometabolic and cardiovascular diseases, atherosclerosis-prone, high-fat diet-fed Apoe-/- mouse siblings were randomly assigned to either individual or grouped housing for 12 weeks. Histological, flow cytometric, biochemical, gene, and protein analyses were performed to assess atherosclerotic lesions, systemic metabolism, inflammation, and stress response. The effects of oxytocin on hepatocytes and subsequent cardiometabolic and cardiovascular function were investigated by in vivo and in vitro approaches. RESULTS Apoe-/- mice housed individually developed larger vulnerable atherosclerotic lesions by disrupted lipid metabolism compared with those of mice in regular group housing, irrespective of body weight, eating behavior, feeding conditions, sympathetic nervous activity, glucocorticoid response, or systemic inflammation. Mechanistically, the chronic isolation reduced the hypothalamic production of oxytocin, which controls bile acid production and LPL (lipoprotein lipase) activity through the peripheral OXTR (oxytocin receptor) in hepatocytes, whose downstream targets include Cyp7a1, Angptl4, and Angptl8. While hepatocyte-specific OXTR-null mice and mice receiving adeno-associated virus targeting OXTR on hepatocytes led to severe dyslipidemia and aggravated atherosclerosis, oral oxytocin supplementation to socially isolated mice, but not to hepatocyte-specific OXTR conditional knockout mice, improved lipid profiles and retarded atherosclerosis development. CONCLUSIONS These results identify a novel brain-liver axis that links sociality to hepatic lipid metabolism, thus proposing a potential therapeutic strategy for loneliness-associated atherosclerosis progression.
Collapse
Affiliation(s)
- Seien Ko
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Anzai
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Xueyuan Liu
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Kenichiro Kinouchi
- Department of Endocrinology, Metabolism, and Nephrology (K.K., T.T., K.H.), Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Yamanoi
- Department of Pathology (K.Y.), Keio University School of Medicine, Tokyo, Japan
| | - Takuto Torimitsu
- Department of Endocrinology, Metabolism, and Nephrology (K.K., T.T., K.H.), Keio University School of Medicine, Tokyo, Japan
| | - Genki Ichihara
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Hiroki Kitakata
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Kohsuke Shirakawa
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Yoshinori Katsumata
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Jin Endo
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Kaori Hayashi
- Department of Endocrinology, Metabolism, and Nephrology (K.K., T.T., K.H.), Keio University School of Medicine, Tokyo, Japan
| | - Masahide Yoshida
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan (M.Y., T.O.)
| | - Katsuhiko Nishimori
- Department of Obesity and Internal Inflammation, Fukushima Medical University, Japan (K.N.)
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research (K.F.T.), Keio University School of Medicine, Tokyo, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan (M.Y., T.O.)
| | - Motoaki Sano
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Japan (M.S.)
| | - Masaki Ieda
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
80
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025:10.1038/s41569-024-01111-0. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
81
|
Ye Y, Markussen B, Engelsen SB, Khakimov B. The quality, uniqueness, and causality of NMR-based prediction models for low-density lipoprotein cholesterol subfractions in human blood plasma. Comput Biol Med 2025; 184:109379. [PMID: 39602980 DOI: 10.1016/j.compbiomed.2024.109379] [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: 06/26/2024] [Revised: 10/03/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Low-density lipoprotein (LDL) cholesterol (chol) subfractions are risk biomarkers for cardiovascular diseases (CVD). A reference analysis, ultracentrifugation (UC), is laborious and may be replaced with a rapid prediction using proton NMR spectra of human blood plasma. However, the quality and uniqueness of these prediction models of biologically related subfractions remains unknown. This study, using two independent cohorts (n = 277), investigates the inter-correlations between LDL cholesterol in the main fraction and five subfractions, as well as the independence of their NMR-based prediction models. The results reveal that the prediction models utilize both shared and unique spectral information from the NMR spectra to determine concentrations of LDL subfractions. Analysis of variance contributions for prediction and causality assessments demonstrate that the NMR spectra contain unique predictive information for the LDL1chol, LDL2chol, and LDL5chol subfractions. In contrast, the spectral signatures for LDL3chol and LDL4chol are either insufficient or confounded. Our findings indicate that these five CVD biomarkers represent two independent clusters, reflecting their biosynthetic pathways, and confirm the presence of causal relationships between certain LDL chol subfractions. This highlights the importance of employing caution when interpreting the concentrations of specific LDL subfractions as standalone biomarkers for CVD risk.
Collapse
Affiliation(s)
- Yongxin Ye
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958, Frederiksberg C, Denmark
| | - Bo Markussen
- Department of Mathematical Sciences, University of Copenhagen, Universitetsparken 5, DK-2100, Copenhagen OE, Denmark
| | - Søren Balling Engelsen
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958, Frederiksberg C, Denmark
| | - Bekzod Khakimov
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958, Frederiksberg C, Denmark.
| |
Collapse
|
82
|
Jomova K, Alomar SY, Nepovimova E, Kuca K, Valko M. Heavy metals: toxicity and human health effects. Arch Toxicol 2025; 99:153-209. [PMID: 39567405 PMCID: PMC11742009 DOI: 10.1007/s00204-024-03903-2] [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: 09/08/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Heavy metals are naturally occurring components of the Earth's crust and persistent environmental pollutants. Human exposure to heavy metals occurs via various pathways, including inhalation of air/dust particles, ingesting contaminated water or soil, or through the food chain. Their bioaccumulation may lead to diverse toxic effects affecting different body tissues and organ systems. The toxicity of heavy metals depends on the properties of the given metal, dose, route, duration of exposure (acute or chronic), and extent of bioaccumulation. The detrimental impacts of heavy metals on human health are largely linked to their capacity to interfere with antioxidant defense mechanisms, primarily through their interaction with intracellular glutathione (GSH) or sulfhydryl groups (R-SH) of antioxidant enzymes such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and other enzyme systems. Although arsenic (As) is believed to bind directly to critical thiols, alternative hydrogen peroxide production processes have also been postulated. Heavy metals are known to interfere with signaling pathways and affect a variety of cellular processes, including cell growth, proliferation, survival, metabolism, and apoptosis. For example, cadmium can affect the BLC-2 family of proteins involved in mitochondrial death via the overexpression of antiapoptotic Bcl-2 and the suppression of proapoptotic (BAX, BAK) mechanisms, thus increasing the resistance of various cells to undergo malignant transformation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of antioxidant enzymes, the level of oxidative stress, and cellular resistance to oxidants and has been shown to act as a double-edged sword in response to arsenic-induced oxidative stress. Another mechanism of significant health threats and heavy metal (e.g., Pb) toxicity involves the substitution of essential metals (e.g., calcium (Ca), copper (Cu), and iron (Fe)) with structurally similar heavy metals (e.g., cadmium (Cd) and lead (Pb)) in the metal-binding sites of proteins. Displaced essential redox metals (copper, iron, manganese) from their natural metal-binding sites can catalyze the decomposition of hydrogen peroxide via the Fenton reaction and generate damaging ROS such as hydroxyl radicals, causing damage to lipids, proteins, and DNA. Conversely, some heavy metals, such as cadmium, can suppress the synthesis of nitric oxide radical (NO·), manifested by altered vasorelaxation and, consequently, blood pressure regulation. Pb-induced oxidative stress has been shown to be indirectly responsible for the depletion of nitric oxide due to its interaction with superoxide radical (O2·-), resulting in the formation of a potent biological oxidant, peroxynitrite (ONOO-). This review comprehensively discusses the mechanisms of heavy metal toxicity and their health effects. Aluminum (Al), cadmium (Cd), arsenic (As), mercury (Hg), lead (Pb), and chromium (Cr) and their roles in the development of gastrointestinal, pulmonary, kidney, reproductive, neurodegenerative (Alzheimer's and Parkinson's diseases), cardiovascular, and cancer (e.g. renal, lung, skin, stomach) diseases are discussed. A short account is devoted to the detoxification of heavy metals by chelation via the use of ethylenediaminetetraacetic acid (EDTA), dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), 2,3-dimercapto-1-propane sulfonic acid (DMPS), and penicillamine chelators.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, 949 74, Nitra, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
83
|
Sritrakarn T, Lowhalidanon K, Khunkaewla P. CDR identification, epitope mapping and binding affinity determination of novel monoclonal antibodies generated against human apolipoprotein B-100. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141058. [PMID: 39454741 DOI: 10.1016/j.bbapap.2024.141058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/24/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
In-house generated mAbs to apolipoprotein B-100 (apoB-100) clones hLDL-E8, hLDL-2D8 and hLDL-F5 were extensively studied to determine their complementarity-determining regions (CDRs), binding epitopes and affinity. RT-PCR revealed that all mAbs consisted of kappa light chains and gamma heavy chains. DNA sequencing and bioinformatic analysis showed that the variable gene and protein sequences of their CDRs shared over 50 % identity with the existing databases. The 3D structures of the mAb variable fragments (Fv) with a QSQE score above 0.7 were constructed using the SWISS-MODEL platform. The structural accuracy was confirmed by Ramachandran plots, with 99 % of amino acid residues falling within acceptable regions. Thrombolytic cleavage of apoB-100 and Western blot analysis demonstrated that hLDL-E8 and hLDL-F5 specifically bind to the T3 fragment (aa 1297-3249), whereas hLDL-2D8 binds to the T4 fragment (aa 1-1297). These findings were supported with epitope-binding assays using inhibition ELISA, which indicated that hLDL-E8 binds at different epitopes from hLDL-2D8 and has some overlap with hLDL-F5. Lastly, the binding affinity of the mAbs was examined by indirect ELISA. The average affinity constants (Kaff) for mAbs hLDL-2D8, hLDL-E8 and hLDL-F5 are 1.51 ± 0.69 × 109 Mol-1, 7.25 ± 3.56 × 108 Mol-1 and 4.39 ± 2.63 × 106 Mol-1, respectively. Additionally, the behavior of the antibodies in the dose-response curve revealed that hLDL-F5 may recognize two epitopes of apoB-100 or have very low binding affinity. In contrast, hLDL-2D8 and hLDL-E8 each recognize a single epitope. These findings provide information that will be useful when selecting mAbs for both laboratory and clinical research purposes.
Collapse
Affiliation(s)
- Tariga Sritrakarn
- Biochemistry-Electrochemistry Research Unit, School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Kanokwan Lowhalidanon
- Biochemistry-Electrochemistry Research Unit, School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; Institute of Research and Development, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Panida Khunkaewla
- Biochemistry-Electrochemistry Research Unit, School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
84
|
Deconinck OG, Sharman JE, Bishop W, Lees CF, Dare L, Hardikar A, Fenton C, Pointon T, Watts GF, Black JA. Familial Hypercholesterolemia and Cardiovascular Outcomes Amongst Younger Patients Undergoing Coronary Bypass Surgery. Heart Lung Circ 2025; 34:77-83. [PMID: 39613585 DOI: 10.1016/j.hlc.2024.08.001] [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: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is an under-recognised but common genetic condition resulting in elevated levels of low-density lipoprotein cholesterol (LDL-C) and a high risk of premature coronary disease. The prevalence of FH among younger patients undergoing coronary bypass surgery is unknown, as is their post-surgical prognosis. METHOD This was a retrospective analysis of younger patients (aged <60 years) undergoing coronary bypass surgery at an Australian tertiary hospital between 2008 and 2022. A Dutch Lipid Clinical Network Score was calculated to determine the presence of underlying FH for each patient. Outcomes were FH prevalence, pre-surgical attainment of guideline-based secondary prevention LDL-C targets and post-surgical major adverse cardiovascular events. RESULTS Overall, 590 eligible patients (mean age 53.7 years, 85.6% male) were followed over a median of 7.9 years (interquartile range 4.7-12.1). Eighty (80; 13.6%) patients were categorised as 'FH', 249 (42.2%) 'possible FH' and 261 (44.2%) 'non-FH'. Compared to the non-FH group, patients with FH were less likely to achieve target LDL-C <1.8 mmol/L (15 [18.8%] vs 119 [45.6%]; p<0.001) and had higher rates of adverse cardiovascular events in the years following surgery (adjusted odds ratio 2.52; 95% confidence interval 1.0-6.4; p<0.001). CONCLUSIONS FH is highly prevalent among younger patients undergoing coronary bypass surgery. These patients are less likely to achieve adequate LDL reduction and are at higher risk of further adverse events. Detection and appropriate treatment of FH prior to bypass surgery should be a clinical priority.
Collapse
Affiliation(s)
| | - James E Sharman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia
| | | | | | - Luke Dare
- Royal Hobart Hospital, Hobart, Tas, Australia
| | - Ashutosh Hardikar
- Royal Hobart Hospital, Hobart, Tas, Australia; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia
| | | | | | | | - James A Black
- Royal Hobart Hospital, Hobart, Tas, Australia; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia.
| |
Collapse
|
85
|
Yin Z, Fu L, Wang Y, Tai S. Impact of gut microbiota on cardiac aging. Arch Gerontol Geriatr 2025; 128:105639. [PMID: 39312851 DOI: 10.1016/j.archger.2024.105639] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
Recent research has suggested imbalances in gut microbiota composition as contributors to cardiac aging. An individual's physical condition, along with lifestyle-associated factors, including diet and medication, are significant determinants of gut microbiota composition. This review discusses evidence of bidirectional associations between aging and gut microbiota, identifying gut microbiota-derived metabolites as potential regulators of cardiac aging. It summarizes the effects of gut microbiota on cardiac aging diseases, including cardiac hypertrophy and fibrosis, heart failure, and atrial fibrillation. Furthermore, this review discusses the potential anti-aging effects of modifying gut microbiota composition through dietary and pharmacological interventions. Lastly, it underscores critical knowledge gaps and outlines future research directions. Given the current limited understanding of the direct relationship between gut microbiota and cardiac aging, there is an urgent need for preclinical and clinical investigations into the mechanistic interactions between gut microbiota and cardiac aging. Such endeavors hold promise for shedding light on the pathophysiology of cardiac aging and uncovering new therapeutic targets for cardiac aging diseases.
Collapse
Affiliation(s)
- Zhiyi Yin
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Liyao Fu
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Yongjun Wang
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| | - Shi Tai
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| |
Collapse
|
86
|
Du Q, Chen B, Yang X, Zhu H, Shams Ul Hassan S, Liu Q. Bioactive Macromolecule-mediated Biogenic FeONPs Attenuate Inflammation in Atherosclerotic Rat by Activating PI3K/Akt/eNOS Pathway. Curr Pharm Des 2025; 31:843-854. [PMID: 39317998 DOI: 10.2174/0113816128298009240828062231] [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: 01/22/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Atherosclerosis refers to the thickening and hardening of artery walls. In our latest experiment, we utilized environmentally friendly techniques to produce multifunctional iron oxide nanoparticles (FeONPs) aimed at reducing inflammation in rats with atherosclerosis. METHODS The formulation was synthesized using curcumin (as the potent bioactive molecule) and was characterized. We assessed the in vitro antioxidant capability of the formulation against DPPH free radicals. Additionally, we quantified the mRNA levels of eNOS, PI3K, and Akt using Real Time-Polymerase Chain Reaction (RT-PCR). We tested the therapeutic impact of the bioactive formulation on a Triton X-100-induced atherosclerosis mouse model. RESULTS The crystallinity and magnetic behavior confirmed the magnetic properties of the FeONPs. The DPPH assay exhibited the dose-dependent radical scavenging characteristics of FeONPs. In the animal experiments, significant upregulation of the studied genes was noticed in treated groups 2 and 3 compared to treated group 1. Moreover, the expression of PI3K/eNOS/Akt was greater in treated group 3 than in treated group 2. These results indicate a dose-dependent elevation in target gene expression. CONCLUSION Nevertheless, the variation in gene expression between the negative control and the untreated control was not statistically significant (p > 0.05) across all genes.
Collapse
Affiliation(s)
- Qing Du
- Department of Emergency, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | - Bo Chen
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | - Xiaohan Yang
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | - Hecheng Zhu
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| | | | - Qiang Liu
- Department of Cardiology, The Fourth People's Hospital of Jinan, Shifan Road, No. 50, Jinan 250031, China
| |
Collapse
|
87
|
Shimizu Y, Kawashiri SY, Yamanashi H, Nakamichi S, Hayashida N, Nagata Y, Maeda T. Beneficial influence of low-density lipoprotein cholesterol on the endothelium in relation to endothelial repair. Environ Health Prev Med 2025; 30:24. [PMID: 40189259 PMCID: PMC11986262 DOI: 10.1265/ehpm.24-00332] [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: 10/10/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDLc) is regarded as a risk factor for endothelial dysfunction. However, LDLc stimulates the proliferation of hematopoietic stem cells (CD34-positive cells), which contribute to endothelial repair. Therefore, LDLc may have a beneficial influence on the endothelium of individuals with lower endothelial repair activity. METHODS This cross-sectional study included 245 men aged 60-69 years. Endothelial repair activity was categorized by the circulating levels of CD34-positive cells based on median values. The status of endothelium was evaluated using the cardio-ankle vascular index (CAVI). RESULTS Among individuals with low levels of circulating CD34-positive cells, LDL-c levels were significantly inversely correlated with CAVI and positively correlated with circulating CD34-positive cells. No significant correlations were observed among the participants with high levels of circulating CD34-positive cells. Among low levels of CD34-positive cells, the adjusted standardized parameter (β) and p value were -0.24 (p = 0.021) for CAVI and 0.41 (p < 0.001) for CD34-positive cells, whereas among high levels of CD34-positive cells, the corresponding values were 0.03 (p = 0.738) and -0.09 (p = 0.355). CONCLUSION LDLc has a beneficial influence on endothelial health among individuals with low endothelial repair activity, possibly by stimulating the proliferation of hematopoietic stem cells.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Seiko Nakamichi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naomi Hayashida
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Division of Strategic Collaborative Research, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Yasuhiro Nagata
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Islands and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
88
|
Modder M, het Panhuis WI, Li M, Afkir S, Dorn AL, Pronk ACM, Streefland TCM, Lalai RA, Pierrou S, Nilsson SK, Olivecrona G, Kooijman S, Rensen PCN, Schönke M. Liver-targeted Angptl4 silencing by antisense oligonucleotide treatment attenuates hyperlipidaemia and atherosclerosis development in APOE*3-Leiden.CETP mice. Cardiovasc Res 2024; 120:2179-2190. [PMID: 39259836 PMCID: PMC11687395 DOI: 10.1093/cvr/cvae195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 09/13/2024] Open
Abstract
AIMS Angiopoietin-like 3 (ANGPTL3) and 4 (ANGPTL4) inhibit lipoprotein lipase to regulate tissue fatty acid (FA) uptake from triglyceride (TG)-rich lipoproteins such as very low density lipoproteins (VLDL). While pharmacological inhibition of ANGPTL3 is being evaluated as a lipid-lowering strategy, systemic ANGPTL4 inhibition is not pursued due to adverse effects. This study aims to compare the therapeutic potential of liver-specific Angptl3 and Angptl4 silencing to attenuate hyperlipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice, a well-established humanized model for lipoprotein metabolism. METHODS AND RESULTS Mice were subcutaneously injected twice per week with saline or liver-targeted antisense oligonucleotides against Angptl3, Angptl4, both, or a scrambled oligonucleotide. Plasma lipid levels, VLDL clearance, and hepatic VLDL production were determined, and atherosclerosis development was assessed. For toxicological evaluation, cynomolgus monkeys were treated with three dosages of liver-targeted ANGPTL4-silencing oligonucleotides. Liver-targeted Angptl4 silencing reduced plasma TGs (-48%) and total cholesterol (-56%), explained by higher VLDL-derived FA uptake by brown adipose tissue and lower VLDL production by the liver. Accordingly, Angptl4 silencing reduced atherosclerotic lesion size (-86%) and improved lesion stability. Hepatic Angptl3 silencing similarly attenuated hyperlipidemia and atherosclerosis development. While Angptl3 and Angptl4 silencing lowered plasma TGs in the refed and fasted state, respectively, combined Angptl3/4 silencing lowered plasma TGs independent of the nutritional state. In cynomolgus monkeys, anti-ANGPTL4 ASO treatment was well tolerated without adverse effects. CONCLUSION Liver-targeted Angptl4 silencing potently attenuates hyperlipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice, and liver-targeted ANGPTL4 silencing is well tolerated in non-human primates. These data warrant further clinical development of liver-targeted ANGPTL4 silencing.
Collapse
Affiliation(s)
- Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Wietse In het Panhuis
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Mohan Li
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Salwa Afkir
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Alexandra L Dorn
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Trea C M Streefland
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Stefan Pierrou
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
| | - Stefan K Nilsson
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
| | - Gunilla Olivecrona
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
89
|
Bjørklund Holven K, Sonestedt E. Milk and dairy products - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2024; 68:10486. [PMID: 39781268 PMCID: PMC11708500 DOI: 10.29219/fnr.v68.10486] [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: 06/13/2022] [Revised: 01/12/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2025] Open
Abstract
Milk and dairy products are major sources of protein, calcium, and other micronutrients. Milk and dairy products contribute with approximately half of the total intake of saturated fat in the Nordic and Baltic diets. Saturated fat is an important determinant of plasma total and low density lipoprotein (LDL)-cholesterol concentrations, and a causal relationship between high LDL-cholesterol and atherosclerotic cardiovascular disease has consistently been documented. The aim of this scoping review is to describe the evidence for the role of milk and dairy products for health-related outcomes as a basis for setting and updating food-based dietary guidelines. Two qualified systematic reviews were included (World Cancer Research Fund and a systematic review for the US Dietary Guidelines Advisory Committee 2020). In addition, systematic reviews published between January 2011 and January 2022 were considered, screened (555 records) and evaluated (159 records) for this review. The systematic reviews suggest that milk or dairy consumption is not associated with increased risk of cardiovascular disease and dyslipidaemia. Current evidence suggests an inverse association with some cardiometabolic risk factors, such as total and LDL-cholesterol, especially regarding fermented dairy products (i.e. yogurt and cheese). There was evidence of an association between intake of dairy products and reduced risk of colorectal cancer. Some studies reported an inverse association between intake of dairy and type 2 diabetes or markers of impaired glucose homeostasis, especially for low-fat dairy, yoghurt, and cheese. Most studies suggest that intake of milk or dairy is not associated with increased risk of cardiovascular risk and some suggestions of inverse association, especially with low-fat products and fermented dairy products, were found with respect to cardiovascular disease, type 2 diabetes, and colorectal cancer. Milk or dairy products are important dietary sources of calcium and iodine, and are fully compatible with a healthy dietary pattern.
Collapse
Affiliation(s)
| | - Emily Sonestedt
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
90
|
Li G, Wu G, Huang Q, Dong S, Zhou Y, Lu M, Liang J, Zhou X, Zhou Z. Carbon-based light addressable potential aptasensor based on the synergy of C-MXene@rGO and OPD@NGQDs for low-density lipoprotein detection. Mikrochim Acta 2024; 192:35. [PMID: 39729216 DOI: 10.1007/s00604-024-06909-w] [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: 10/23/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
A novel carbon-based light-addressable potentiometric aptasensor (C-LAPS) was constructed for detection low-density lipoprotein (LDL) in serum. Carboxylated Ti3C2 MXene @reduced graphene oxide (C-MXene@rGO) was used as interface and o-phenylenediamine functionalized nitrogen-doped graphene quantum dots (OPD@NGQDs) as the photoelectric conversion element. The photosensitive layers composed of OPD@NGQDs/C-MXene@rGO exhibit superior photoelectric conversion efficiency and excellent biocompatibility, which contribute to an improved response signal. When LDL reacts with the LDL aptamer (LDLApt) immobilized on the photosensitive layers to form LDL-LDLApt complexes, the reaction process can induce the modification of the surface potential in the photosensitive layer, leading to potential shift observed through the I-V curves. The experimental conditions were successfully optimized with few planned tests by applying the Box-Behnken design and response surface methodology aspects of the Design-Expert software. Under the optimal condition, the potential shift had a linear relationship with concentrations of LDL from 0.02 to 0.30 μg/mL. The limit of detection (LOD) was 5.88 ng/mL (S/N = 3) and the sensitivity was 315.20 mV/μg·mL-1. In addition, the LDL C-LAPS demonstrated excellent specificity, reproducibility, and stability in detecting LDL. The sensor performed well in quantifying LDL in real samples. Therefore, the LDL C-LAPS has the potential for clinical applications.
Collapse
Affiliation(s)
- Guiyin Li
- College of Chemistry, Guangdong University of Petrochemical Technology, Guandu Road, Maoming, Guangdong, 525000, People's Republic of China
| | - Guangxiong Wu
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Qing Huang
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Shuaikang Dong
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Yu Zhou
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Mei Lu
- Department of Clinical Laboratory, The 924, Hospital of Chinese People's Liberation Army Joint Logistic Support Force, Guilin, Guangxi, 541002, People's Republic of China
| | - Jintao Liang
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China.
| | - Xueqing Zhou
- Department of Clinical Laboratory, The 924, Hospital of Chinese People's Liberation Army Joint Logistic Support Force, Guilin, Guangxi, 541002, People's Republic of China.
| | - Zhide Zhou
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China.
| |
Collapse
|
91
|
Zeng G, Zhang C, Song Y, Zhang Z, Xu J, Liu Z, Tang X, Wang X, Chen Y, Zhang Y, Zhu P, Guo X, Jiang L, Wang Z, Liu R, Wang Q, Yao Y, Feng Y, Han Y, Yuan J. The potential impact of inflammation on the lipid paradox in patients with acute myocardial infarction: a multicenter study. BMC Med 2024; 22:599. [PMID: 39710711 PMCID: PMC11664818 DOI: 10.1186/s12916-024-03823-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) is a well-recognized risk factor for cardiovascular diseases. However, several clinical studies demonstrated an inverse association between LDL-C and mortality risk in patients with acute myocardial infarction (AMI), known as the lipid paradox. This study aims to investigate the potential impact of inflammation on the association between LDL-C levels and mortality risks. METHODS A total of 5244 patients with AMI from a large nationwide prospective cohort were included in our analysis. Patients were stratified according to LDL-C quartiles. The primary outcome was all-cause mortality, and the secondary endpoint was cardiac mortality. High-sensitive C-reactive protein (hsCRP) > 3 mg/L was defined as high inflammatory risk. RESULTS During a median follow-up of 2.07 years, 297 mortality events (5.5%) and 227 cardiac mortality events (4.2%) occurred. Patients in the lowest LDL-C quartile had the highest incidence of all-cause mortality (7.3%) and cardiac mortality (5.8%). A U-shaped association between LDL-C levels and mortality risk was observed after multivariable adjustment, which persisted only in patients with high hsCRP levels. In contrast, a linear association between LDL-C and mortality risk was shown in patients with low hsCRP levels. CONCLUSIONS AMI patients with lower LDL-C levels had a higher risk of mortality. However, this association was only observed in those with high inflammatory risk. In contrast, the relationship between LDL-C and mortality risk was linear in patients with low inflammatory risk. This suggests the importance of considering inflammation when managing LDL-C levels in AMI patients.
Collapse
Affiliation(s)
- Guyu Zeng
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Ce Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Ying Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Zheng Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jingjing Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Xiaozeng Wang
- Department of Cardiology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110016, China
| | - Yan Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Yongzhen Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Jiang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Zhifang Wang
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, China
| | - Ru Liu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Qingsheng Wang
- Department of Cardiology, The First Hospital of QinHuangDao, Qinhuangdao, China
| | - Yi Yao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China
| | - Yingqing Feng
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenyang, 110016, China.
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Rd, Beijing, 100037, Xicheng District, China.
| |
Collapse
|
92
|
Shen L, Tian Q, Ran Q, Gan Q, Hu Y, Du D, Qin Z, Duan X, Zhu X, Huang W. Z-Ligustilide: A Potential Therapeutic Agent for Atherosclerosis Complicating Cerebrovascular Disease. Biomolecules 2024; 14:1623. [PMID: 39766330 PMCID: PMC11726876 DOI: 10.3390/biom14121623] [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: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis (AS) is one of the major catalysts of ischemic cerebrovascular disease, and the death and disease burden from AS and its cerebrovascular complications are increasing. Z-ligustilide (Z-LIG) is a key active ingredient in Angelica sinensis (Oliv.) Diels and Ligusticum chuanxiong Hort. In this paper, we first introduced LIG's physicochemical properties and pharmacokinetics. Then, we reviewed Z-LIG's intervention and therapeutic mechanisms on AS and its cerebrovascular complications. The mechanisms of Z-LIG intervention in AS include improving lipid metabolism, antioxidant and anti-inflammatory effects, protecting vascular endothelium, and inhibiting vascular endothelial fibrosis, pathological thickening, and plaque calcification. In ischemic cerebrovascular diseases complicated by AS, Z-LIG exerts practical neuroprotective effects in ischemic stroke (IS), transient ischemic attack (TIA), and vascular dementia (VaD) through anti-neuroinflammatory, anti-oxidation, anti-neuronal apoptosis, protection of the blood-brain barrier, promotion of mitochondrial division and angiogenesis, improvement of cholinergic activity, inhibition of astrocyte proliferation, and endoplasmic reticulum stress. This paper aims to provide a basis for subsequent studies of Z-LIG in the prevention and treatment of AS and its cerebrovascular complications and, thus, to promote the development of interventional drugs for AS.
Collapse
Affiliation(s)
- Longyu Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Qiqi Ran
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianrong Gan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Donglian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Zehua Qin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyi Duan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyun Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| |
Collapse
|
93
|
Gibson CM, Duffy D, Bahit MC, Chi G, White H, Korjian S, Alexander JH, Lincoff AM, Heise M, Kingwell BA, Nicolau JC, Lopes RD, Cornel JH, Lewis BS, Vinereanu D, Goodman SG, Bode C, Steg PG, Libby P, Sacks FM, Bainey KR, Ridker PM, Mahaffey KW, Aylward P, Nicholls SJ, Pocock SJ, Mehran R, Harrington RA. Apolipoprotein A-I infusions and cardiovascular outcomes in acute myocardial infarction according to baseline LDL-cholesterol levels: the AEGIS-II trial. Eur Heart J 2024; 45:5023-5038. [PMID: 39221651 DOI: 10.1093/eurheartj/ehae614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/08/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS In the AEGIS-II trial (NCT03473223), CSL112, a human apolipoprotein A1 derived from plasma that increases cholesterol efflux capacity, did not significantly reduce the risk of the primary endpoint through 90 days vs. placebo after acute myocardial infarction (MI). Nevertheless, given the well-established relationship between higher low-density lipoprotein cholesterol (LDL-C) and plaque burden, as well as greater risk reductions seen with PCSK9 inhibitors in patients with baseline LDL-C ≥ 100 mg/dL on statin therapy, the efficacy of CSL112 may be influenced by baseline LDL-C. METHODS Overall, 18 219 patients with acute MI, multivessel coronary artery disease, and additional risk factors were randomized to either four weekly infusions of 6 g CSL112 or placebo. This exploratory post-hoc analysis evaluated cardiovascular outcomes by baseline LDL-C in patients prescribed guideline-directed statin therapy at the time of randomization (n = 15 731). RESULTS As baseline LDL-C increased, the risk of the primary endpoint at 90 days lowered in those treated with CSL112 compared with placebo. In patients with LDL-C ≥ 100 mg/dL at randomization, there was a significant risk reduction of cardiovascular death, MI, or stroke in the CSL112 vs. placebo group at 90, 180, and 365 days [hazard ratio .69 (.53-.90), .71 (.57-.88), and .78 (.65-.93)]. In contrast, there was no difference between treatment groups among those with LDL-C < 100 mg/dL at baseline. CONCLUSIONS In this population, treatment with CSL112 compared to placebo was associated with a significantly lower risk of recurrent cardiovascular events among patients with a baseline LDL-C ≥ 100 mg/dL. Further studies need to confirm that CSL112 efficacy is influenced by baseline LDL-C.
Collapse
Affiliation(s)
- C Michael Gibson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - Danielle Duffy
- Clinical Development, CSL Behring, King of Prussia, PA, USA
| | | | - Gerald Chi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - Harvey White
- Health New Zealand - Te Whatu Ora, Te Toka Tumai, Green Lane Cardiovascular Service, Auckland City Hospital, Auckland 1142, New Zealand
| | - Serge Korjian
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - John H Alexander
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Duke Clinical Research Institute, Duke Health, Durham, NC, USA
| | - A Michael Lincoff
- The Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Mark Heise
- Clinical Development, CSL Behring, King of Prussia, PA, USA
- Deparment of Biostatistics, CSL Behring, King of Prussia, PA, USA
| | - Bronwyn A Kingwell
- Deparment of Research and Development, CSL Limited, Melbourne, Australia
| | - Jose C Nicolau
- Instituto do Coracao (InCor), Cardiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renato D Lopes
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Duke Clinical Research Institute, Duke Health, Durham, NC, USA
- Brazilian Clinical Research Institute, Sao Paulo, SP, Brazil
| | - Jan H Cornel
- Radboud University Medical Center, Nijmegen and Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | | | - Dragos Vinereanu
- University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
- Clinical Trials and Translation Unit, Peter Munk Cardiac Center, University Health Network, Toronto, Canada
| | - Christoph Bode
- Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ph Gabriel Steg
- Université Paris-Cité, INSERM U_1148, FACT and AP-HP, Hôpital Bichat, Paris, France
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Frank M Sacks
- Cardiovascular Disease Prevention, Department of Nutrition, Harvard T.H. Chan School of Public Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Kevin R Bainey
- Walter Mackenzie Health Sciences Centre, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Boston, MA, USA
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Philip Aylward
- Department of Cardiology, South Australian Health and Medical Research Institute/SAHMRI, Adelaide, Australia
| | - Stephen J Nicholls
- Department of Cardiology, Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Stuart J Pocock
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Roxana Mehran
- Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, Zena and Michael A. Wiener Cardiovascular Institute, New York, NY, USA
| | | |
Collapse
|
94
|
Liu J, Teng TQ, Li Z, Hu FW, Sha WW, Shen CX, Xia Y, Zhang YJ, Liang L. Influence of remnant lipoprotein particle cholesterol on non-target lesions progression in patients undergoing percutaneous coronary intervention. Front Cardiovasc Med 2024; 11:1471479. [PMID: 39720212 PMCID: PMC11666560 DOI: 10.3389/fcvm.2024.1471479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/28/2024] [Indexed: 12/26/2024] Open
Abstract
Background Low-Density Lipoprotein Cholesterol (LDL-C) is the primary lipid therapy target for coronary artery disease (CAD) patients after percutaneous coronary intervention (PCI). However, progression of coronary atherosclerosis occurs even LDL-C controlled well, some potentially important factors have been overlooked. Objective This study aims to elucidate the relationship between remnant lipoprotein particle cholesterol (RLP-C) and the progression of non-target lesions (NTLs) in patients with well-controlled lipid levels after PCI. Methods This retrospective study included 769 CAD patients who underwent PCI and followed up angiography within 6-24 months thereafter. Employing Multivariate Cox regression analysis, we assessed the correlation between RLP-C and NTLs progression. Based on the receiver operating characteristic curve analysis, we identified the optimal cutoff point for RLP-C, following which the patients were divided into two groups. Propensity score matching balanced confounding factors between groups, and Log-rank tests compared Kaplan-Meier curves for overall follow-up to assess NTLs progression. Results Multivariate Cox analysis revealed an independent association between RLP-C and NTLs progression when LDL-C was well-controlled. Additionally, the RLP-C level of 0.555 mmol/L was determined to be the best value for predicting NTLs progression. Following propensity score matching, Kaplan-Meier curves illustrated a significantly higher cumulative rate of NTLs progression in patients with RLP-C levels ≥0.555 mmol/L compared to the others (Log-rank P = 0.002). Elevated RLP-C levels were associated with high triglyceride concentrations, diabetes mellitus, and increased risk of revascularization. Conclusions This study illustrated the atherogenic impact of RLP-C in CAD patients. High RLP-C levels increased the risk of revascularization.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neurology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu, China
| | - Tian-Qi Teng
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zheng Li
- Department of Cardiology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu, China
| | - Feng-Wang Hu
- Department of Cardiology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu, China
| | - Wei-Wei Sha
- Department of Cardiology, Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, China
| | - Chang-Xian Shen
- Department of Cardiology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu, China
| | - Yong Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yao-Jun Zhang
- Department of Cardiology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu, China
- Department of Cardiology, Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, China
| | - Li Liang
- Department of Cardiology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu, China
- Department of Cardiology, Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
95
|
Jeeva P, Muthusamy A, Kesavan swaminathan J. Deciphering Structural Dynamics of Atherosclerosis Proteins: Insights from Crataegus oxyacantha Phytochemicals that Interceded Functional and Structural Changes in Targeted Atherosclerotic Proteins. ACS OMEGA 2024; 9:48159-48172. [PMID: 39676950 PMCID: PMC11635474 DOI: 10.1021/acsomega.4c04975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 12/17/2024]
Abstract
Atherosclerosis (ASC) is characterized by foam cell-mediated plaque formation, vascular endothelial inflammation, and lipidosis and is the rudimentary cause of cardiovascular diseases. This is the pre-eminent global factor of mortality. This etiological paradigm is significantly influenced by several proteins, where 23 pivotal proteins involved in ASC were meticulously gleaned on the basis of literature studies. The crux of the present study was aimed to probe the drugability of four active phytochemicals from Crataegus oxyacantha (COC): epicatechin, gallate, tyramine, and vitexin against the selected 23 proteins. The molecular docking analysis was judiciously administered via Glide, the binding free energy was calculated in detail utilizing the prime molecular mechanics-generalized Born surface area (MM-GBSA) module, and a deeper comprehensive investigation of protein-ligand dynamic associations was elucidated through Desmond. Drawing from the upper echelons of our docking results, the molecular dynamics simulation outcomes revealed that the macrophage migration inhibitory factor and prethrombin-1 showed persistent binding nature with gallate. The bioactive compound known as gallate sourced from COC shows the best molecular association with pivotal proteins involved in ASC and has a promising therapeutic potential for drug development endeavors.
Collapse
Affiliation(s)
- Praveen Jeeva
- Department
of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | - Anusuyadevi Muthusamy
- Department
of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | | |
Collapse
|
96
|
Ghasemzadeh Rahbardar M, Fazeli Kakhki H, Hosseinzadeh H. Ziziphus jujuba (Jujube) in Metabolic Syndrome: From Traditional Medicine to Scientific Validation. Curr Nutr Rep 2024; 13:845-866. [PMID: 39354208 DOI: 10.1007/s13668-024-00581-5] [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] [Accepted: 09/13/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE OF REVIEW This review evaluates the therapeutic potential of Ziziphus jujuba and its main components in managing complications of metabolic syndrome, including diabetes, dyslipidemia, obesity, and hypertension. RECENT FINDINGS The reviewed studies provide evidence supporting the use of Z. jujuba and its main components (lupeol and betulinic acid) as natural treatments for complications of metabolic syndrome. These substances enhance glucose uptake through the activation of signaling pathways such as phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), reduce hepatic glucose synthesis, and increase glucose uptake by adipocytes and skeletal muscle cells. They also improve insulin sensitivity by modulating AMP-activated protein kinase (AMPK) activity and regulating insulin signaling proteins and glucose transporters. In the field of dyslipidemia, they inhibit triglyceride synthesis, lipid accumulation, and adipogenic enzymes, while influencing key signaling pathways involved in adipogenesis. Z. jujuba and its constituents demonstrate anti-adipogenic effects, inhibiting lipid accumulation and modulating adipogenic enzymes and transcription factors. They also exhibit positive effects on endothelial function and vascular health by enhancing endothelial nitric oxide synthase (eNOS) expression, NO production, and antioxidant enzyme activity. Z. jujuba, lupeol, and betulinic acid hold promise as natural treatments for complications of metabolic syndrome. They improve glucose metabolism, insulin sensitivity, and lipid profiles while exerting anti-adipogenic effects and enhancing endothelial function. However, further research is needed to elucidate the mechanisms and confirm their efficacy in clinical trials. These natural compounds offer potential as alternative therapies for metabolic disorders and contribute to the growing body of evidence supporting the use of natural medicines in their management.
Collapse
Affiliation(s)
| | - Homa Fazeli Kakhki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
97
|
Masana L, Ibarretxe D. New drugs for treating dyslipidemias. From small molecules to small interfering RNAs. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36 Suppl 1:S15-S23. [PMID: 39645293 DOI: 10.1016/j.arteri.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 12/09/2024]
Abstract
Despite the various therapeutic tools available, many patients do not achieve therapeutic goals, and cardiovascular diseases remain a significant cause of death in our setting. Furthermore, even in patients who manage to reduce their LDL-C levels to the recommended targets, cardiovascular events continue to occur. The therapeutic challenge and the persistent risk have led to active research into new drugs targeting novel therapeutic pathways in the field of lipoprotein metabolism disorders. The therapeutic approach involves new pharmacological mechanisms, ranging from small molecules and monoclonal antibodies to RNA interference, with inclisiran being the first drug approved for clinical use in the cardiovascular domain. In this review, we aim to provide a comprehensive overview of the new therapeutic targets and pharmacological mechanisms under development, as well as their potential clinical impact.
Collapse
Affiliation(s)
- Lluís Masana
- Unitat de Recerca en Lípids i Arteriosclerosi, Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España.
| | - Daiana Ibarretxe
- Unitat de Recerca en Lípids i Arteriosclerosi, Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| |
Collapse
|
98
|
Wang Y, Wang C, Li J. Neutrophil extracellular traps: a catalyst for atherosclerosis. Mol Cell Biochem 2024; 479:3213-3227. [PMID: 38401035 DOI: 10.1007/s11010-024-04931-3] [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: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 02/26/2024]
Abstract
Neutrophil extracellular traps (NETs) are network-like structures released by activated neutrophils. They consist mainly of double-stranded DNA, histones, and neutrophil granule proteins. Continuous release of NETs in response to external stimuli leads to activation of surrounding platelets and monocytes/macrophages, resulting in damage to endothelial cells (EC) and vascular smooth muscle cells (VSMC). Some clinical trials have demonstrated the association between NETs and the severity and prognosis of atherosclerosis. Furthermore, experimental findings have shed light on the molecular mechanisms by which NETs contribute to atherogenesis. NETs play a significant role in the formation of atherosclerotic plaques. This review focuses on recent advancements in the understanding of the relationship between NETs and atherosclerosis. It explores various aspects, including the formation of NETs in atherosclerosis, clinical trials investigating NET-induced atherosclerosis, the mechanisms by which NETs promote atherogenesis, and the translational implications of NETs. Ultimately, we aim to propose new research directions for the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yinyu Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Cuiping Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Jiayan Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
99
|
Løvheim EB, Retterstøl K, Narverud I, Bogsrud MP, Halvorsen B, Ueland T, Aukrust P, Holven KB. Adherence to the Healthy Nordic Food Index is associated with reduced plasma levels of inflammatory markers in patients with heterozygous familial hypercholesterolemia. ATHEROSCLEROSIS PLUS 2024; 58:38-45. [PMID: 39525275 PMCID: PMC11550195 DOI: 10.1016/j.athplu.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/30/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Background and aims Familial hypercholesterolemia (FH) is an inherited disease associated with hypercholesterolemia, and dietary treatment is part of the treatment. We aimed to assess the dietary pattern in relation to the Healthy Nordic Food Index (HNFI) in adults with and without heterozygous FH (HeFH), and to examine the associations between dietary quality and biomarkers related to cardiovascular disease in adults with HeFH. Methods We included 205 adults (≥18 years) with HeFH who received follow-up at the Lipid Clinic in Oslo and compared them to controls (n = 228). Dietary intake was assessed using a food frequency questionnaire and dietary quality was assessed using the HNFI. Blood samples were analysed for levels of blood lipids, plasma fatty acids (FAs), and markers of inflammation and platelet activation. Results The HeFH patients (median 60 years; 50.2 % female; 25.9 % in secondary prevention) had lower intake of total and saturated fat compared to controls (32.6 energy percent (E%) vs. 34.9 E%, and 9.6 E% vs 12.0 E%, respectively; p < 0.001 for both). In the HeFH patients, increasing dietary quality was associated with increased plasma levels of the n-3 polyunsaturated FAs (PUFAs) eicosapentaenoic acid and docosahexaenoic acid, and the n-6 PUFA linoleic acid, and lower plasma levels of the inflammatory cytokines Tumor Necrosis Factor and interleukin-6, and of the platelet-derived inflammatory cytokines Platelet Factor 4 and Neutrophil-Activating Peptide-2. Conclusion Norwegian patients with HeFH followed up at a Lipid Clinic eat healthier than controls. Adherence to a healthy dietary pattern is associated with higher plasma levels of n-3 and n-6 PUFA, and lower levels of inflammatory markers, including platelet markers. This may suggest that adherence to an overall healthy dietary pattern might be beneficial for HeFH patients independent of the cholesterol-lowering effect of the diet.
Collapse
Affiliation(s)
- Eirin B. Løvheim
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O Box 1046 Blindern, 0317, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O Box 1046 Blindern, 0317, Oslo, Norway
- Lipid Clinic, Oslo University Hospital, Norway
| | - Ingunn Narverud
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Norway
| | - Martin P. Bogsrud
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Pål Aukrust
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Kirsten B. Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O Box 1046 Blindern, 0317, Oslo, Norway
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Norway
| |
Collapse
|
100
|
Tamehri Zadeh SS, Pang J, Chan DC, Watts GF. A contemporary snapshot of familial hypercholesterolemia registries. Curr Opin Lipidol 2024; 35:297-302. [PMID: 39508066 DOI: 10.1097/mol.0000000000000958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
PURPOSE OF REVIEW Familial hypercholesterolemia (FH) registries can capture unique data on FH concerning real-world practice, clinic epidemiology, natural history, cascade testing, cardiovascular consequences of late diagnosis, and use of healthcare resources. Such registries are also valuable for identifying and bridging the gaps between guidelines and clinical practice. We reviewed recent findings from the principal FH registries. RECENT FINDINGS Most adult patients with heterozygous FH (HeFH) are diagnosed late, undertreated, and do not reach guideline-recommended low density lipoprotein-cholesterol (LDL-C) goals. In children and adolescents with HeFH, detection relies principally on genetic testing and measurement of LDL-C levels. Similarly, the majority of patients with homozygous FH (HoFH) receive sub-optimal cholesterol-lowering treatments and do not attain recommended LDL-C goals, gaps being wider in lower income than higher income countries. In HeFH patients, men have a higher risk of atherosclerotic cardiovascular disease than women. SUMMARY The evolving data from FH registries provide real-world evidence for developing implementation strategies to address gaps across the continuum of care of FH worldwide.
Collapse
Affiliation(s)
| | - Jing Pang
- Medical School, University of Western Australia, Perth
| | - Dick C Chan
- Medical School, University of Western Australia, Perth
| | - Gerald F Watts
- Medical School, University of Western Australia, Perth
- Lipid Disorders Clinic, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|