1
|
Ryu HE, Jung DH, Heo SJ, Park B, Lee YJ. Extremely high HDL cholesterol paradoxically increases the risk of all-cause mortality in non-diabetic males from the Korean population: Korean genome and epidemiology study-health examinees (KoGES-HEXA) cohorts. Front Med (Lausanne) 2025; 12:1534524. [PMID: 40443511 PMCID: PMC12119502 DOI: 10.3389/fmed.2025.1534524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 05/01/2025] [Indexed: 06/02/2025] Open
Abstract
Background High-density lipoprotein cholesterol (HDL-C) is associated with lower risk of mortality and cardiovascular disease. However, the relationship between extremely high HDL cholesterol level and all-cause mortality has not been thoroughly investigated. In this study, we examined the longitudinal effects of very high HDL cholesterol on all-cause mortality in a large cohort of Korean adults without type 2 diabetes mellitus. Methods Data from 173,195 Korean participants over 40 years of age enrolled in the Korean Genome and Epidemiology Study-Health Examinees (KoGES-HEXA) cohort, linked with the death certificate database of the National Statistical Office, were assessed. Participants were grouped into four according to HDL-C levels. We used multivariate Cox proportional-hazard regression models to prospectively assess hazard ratios (HRs) for all-cause mortality with 95% confidence intervals (CIs) over an 11-year baseline period. Results During a mean follow-up of 11.7 years, there were a total of 3,906 deaths from all causes, including 2,258 in men and 1,648 in women. The relationship between HDL-C and all-cause mortality showed a U-shaped pattern, especially in men. Compared to the reference group, the HR (95% CI) for mortality in males in the highest HDL cholesterol group was 1.31 (95% CI, 1.01-1.71) after adjusting for potential confounding variables. Moreover, low HDL cholesterol showed a statistically significant association with increased mortality in both men and women. Conclusion Extremely high HDL-C levels could paradoxically increase the risk of all-cause mortality, particularly among males, in the general population without type 2 diabetes mellitus. Non-protective effects of very high HDL-C level should be noted when predicting incident metabolic syndrome, particularly in men, in clinical settings.
Collapse
Affiliation(s)
- Ha-Eun Ryu
- Department of Family Medicine, Yongin Severance Hospital, Yongin, Gyeonggi-do, Republic of Korea
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong Hyuk Jung
- Department of Family Medicine, Yongin Severance Hospital, Yongin, Gyeonggi-do, Republic of Korea
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Jae Heo
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byoungjin Park
- Department of Family Medicine, Yongin Severance Hospital, Yongin, Gyeonggi-do, Republic of Korea
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Jae Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Family Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
2
|
Kjeldsen EW, Frikke-Schmidt R. Causal cardiovascular risk factors for dementia: insights from observational and genetic studies. Cardiovasc Res 2025; 121:537-549. [PMID: 39498825 PMCID: PMC12054631 DOI: 10.1093/cvr/cvae235] [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: 07/18/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024] Open
Abstract
The escalating prevalence of dementia worldwide necessitates preventive strategies to mitigate its extensive health, psychological, and social impacts. As the prevalence of dementia continues to rise, gaining insights into its risk factors and causes becomes paramount, given the absence of a definitive cure. Cardiovascular disease has emerged as a prominent player in the complex landscape of dementia. Preventing dyslipidaemia, unhealthy western-type diets, hypertension, diabetes, being overweight, physical inactivity, smoking, and high alcohol intake have the potential to diminish not only cardiovascular disease but also dementia. The purpose of this review is to present our current understanding of cardiovascular risk factors for Alzheimer's disease and vascular dementia (VaD) by using clinical human data from observational, genetic studies and clinical trials, while elaborating on potential mechanisms. Hypertension and Type 2 diabetes surface as significant causal risk factors for both Alzheimer's disease and VaD, as consistently illustrated in observational and Mendelian randomization studies. Anti-hypertensive drugs and physical activity have been shown to improve cognitive function in clinical trials. Important to note is that robust genome-wide association studies are lacking for VaD, and indeed more and prolonged clinical trials are needed to establish these findings and investigate other risk factors. Trials should strategically target individuals at the highest dementia risk, identified using risk charts incorporating genetic markers, biomarkers, and cardiovascular risk factors. Understanding causal risk factors for dementia will optimize preventive measures, and the implementation of well-known therapeutics can halt or alleviate dementia symptoms if started early. Needless to mention is that future health policies should prioritize primordial prevention from early childhood to prevent risk factors from even occurring in the first place. Together, understanding the role of cardiovascular risk factors in dementia, improving genome-wide association studies for VaD, and advancing clinical trials are crucial steps in addressing this significant public health challenge.
Collapse
Affiliation(s)
- Emilie Westerlin Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
3
|
Pérez-Medina C, Fisher EA, Fayad ZA, Mulder WJM, Teunissen AJP. Radiolabeling lipoproteins to study and manage disease. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07281-4. [PMID: 40293448 DOI: 10.1007/s00259-025-07281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Lipoproteins are endogenous nanoparticles with essential roles in lipid transport and inflammation. Lipoproteins are also valuable in diagnosing and treating disease. For instance, certain lipoproteins are overexpressed in patients with atherosclerotic cardiovascular disease, and reconstituted lipoproteins have been extensively used for drug delivery. Radiolabeling has proven an especially powerful approach for studying and therapeutically exploiting lipoproteins. This review details how radiochemistry and nuclear imaging can facilitate the study of lipoproteins in health and disease. Among other topics, we discuss approaches for radiolabeling lipoproteins and detail how these have helped advance our understanding of lipoprotein biology and the diagnosis and treatment of diseases, including atherosclerosis, cancer, and hypercholesteremia. METHODS We performed an extensive literature search on all peer-reviewed studies involving radiolabeled lipoproteins and selected representative examples to provide a high-level overview of the most important discoveries and technological advancements. RESULTS More than 200 peer-reviewed papers involved radiolabeled lipoproteins, spanning mechanistic, diagnostic, and therapeutic studies across a wide range of diseases. CONCLUSION Radiolabeling has been critical in advancing our understanding of lipoprotein biology and leveraging these nanomaterials for diagnosing and treating disease.
Collapse
Affiliation(s)
| | - Edward A Fisher
- Department of Medicine (Cardiology), New York University Grossman School of Medicine, New York, NY, USA
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Willem J M Mulder
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Hung RKY, Costeira R, Chen J, Schlosser P, Grundner-Culemann F, Booth JW, Sharpe CC, Bramham K, Sun YV, Marconi VC, Teumer A, Winkler CA, Post FA, Bell JT. Epigenetic associations with kidney disease in individuals of African ancestry with APOL1 high-risk genotypes and HIV. Nephrol Dial Transplant 2025; 40:997-1006. [PMID: 39448372 PMCID: PMC12035534 DOI: 10.1093/ndt/gfae237] [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: 03/25/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Apolipoprotein L1 (APOL1) high-risk variants are major determinants of chronic kidney disease (CKD) in people of African ancestry. Previous studies have identified epigenetic changes in relation to kidney function and CKD, but not in individuals with APOL1 high-risk genotypes. We conducted an epigenome-wide analysis of CKD and estimated glomerular filtration rate (eGFR) in in people of African ancestry and APOL1 high-risk genotypes with HIV. METHODS DNA methylation profiles from peripheral blood mononuclear cells of 119 individuals with APOL1 high-risk genotypes (mean age 48 years, 49% female, median CD4 count 515 cells/mm3, 90% HIV-1 RNA <200 copies/mL, 23% with CKD) were obtained by Illumina MethylationEPIC BeadChip. Differential methylation analysis of CKD considered technical and biological covariates. We also assessed associations with eGFR. Replication was pursued in three independent multi-ancestry cohorts with and without HIV. RESULTS DNA methylation levels at 14 regions were associated with CKD. The strongest signals were located in SCARB1, DNAJC5B and C4orf50. Seven of the 14 signals also associated with eGFR, and most showed evidence for a genetic basis. Four signals (in SCARB1, FRMD4A, CSRNP1 and RAB38) replicated in other cohorts, and 11 previously reported epigenetic signals for kidney function or CKD replicated in our cohort. We found no significant DNA methylation signals in, or near, the APOL1 promoter region. CONCLUSIONS We report several novel as well as previously reported epigenetic associations with CKD and eGFR in individuals with HIV having APOL1 high-risk genotypes. Further investigation of pathways linking DNA methylation to APOL1 nephropathies is warranted.
Collapse
Affiliation(s)
- Rachel K Y Hung
- Department of HIV and Sexual Health, King's College Hospital, London, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Ricardo Costeira
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Franziska Grundner-Culemann
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - John W Booth
- Department of Renal Medicine, Bart's Health NHS Foundation Trust, London, UK
| | - Claire C Sharpe
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Kate Bramham
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Vincent C Marconi
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- School of Medicine, Emory University, Atlanta, GA, USA
| | - Alexander Teumer
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Cheryl A Winkler
- Basic Reseach Program, Frederick National Laboratory for Cancer Research and the Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Frank A Post
- Department of HIV and Sexual Health, King's College Hospital, London, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
5
|
Ma Y, Wu Y, Hu L, Chen W, Zhang X, Zheng D, Congdon N, Jin G, Liu Z. Associations between serum lipids and glaucoma: a cohort study of 400 229 UK Biobank participants. Br J Ophthalmol 2025:bjo-2024-326062. [PMID: 39904580 DOI: 10.1136/bjo-2024-326062] [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: 06/25/2024] [Accepted: 11/25/2024] [Indexed: 02/06/2025]
Abstract
PURPOSE To examine the associations of commonly-used serum lipid measures (high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC) and triglycerides (TG)) with glaucoma. METHODS This prospective cohort study included 400 229 participants from the UK Biobank. Cox regression and restricted cubic spline models and polygenic risk scores were employed to investigate the associations between serum lipids and glaucoma. RESULTS Over a mean follow-up of 14.44 years, 6868 (1.72%) participants developed glaucoma. Multivariate Cox regression revealed that higher levels of HDL-C were associated with an increased risk of glaucoma (HR for 1-SD increase in HDL-C 1.05, 95% CI 1.02 to 1.08, p=0.001), while elevated levels of LDL-C (HR 0.96, 95% CI 0.94 to 0.99, p=0.005), TC (HR 0.97, 95% CI 0.94 to 1.00, p=0.037) and TG (HR 0.96, 95% CI 0.94 to 0.99, p=0.008) were all associated with reduced risk. The analysis examining the associations between polygenic risk score of serum lipids and glaucoma showed per 1-SD increment of HDL-C genetic risk was associated with a 5% greater hazard of glaucoma (HR 1.05, 95% CI 1.00 to 1.11, p=0.031). However, the polygenic risk score of LDL-C, TC, and TG did not show a significant association with glaucoma. CONCLUSIONS Elevated HDL-C is associated with an increased risk of glaucoma, while elevated LDL-C, TC, and TG levels are associated with a lower risk of glaucoma. This study enhances our understanding of the association between lipid profile and glaucoma and warrants further investigation of lipid-focused treatments in glaucoma management.
Collapse
Affiliation(s)
- Yiyuan Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Yue Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Leyi Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Wen Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Xinyu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Danying Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Nathan Congdon
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
- Centre for Public Health, Queen's University Belfast, Belfast, UK
- Orbis International, New York, New York, USA
| | - Guangming Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Zhenzhen Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Bolanle IO, de Liedekerke Beaufort GC, Weinberg PD. Transcytosis of LDL Across Arterial Endothelium: Mechanisms and Therapeutic Targets. Arterioscler Thromb Vasc Biol 2025; 45:468-480. [PMID: 40013359 PMCID: PMC11936472 DOI: 10.1161/atvbaha.124.321549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Transport of LDL (low-density lipoprotein) from plasma to arterial intima is thought to be rate limiting in the development of atherosclerosis. Its variation likely determines where lesions develop within arteries and might account for some of the currently unexplained difference in disease susceptibility between individuals. It may also be critical in the development of lipid-rich, unstable plaques. Mechanisms have been controversial but recent evidence suggests that caveolar transcytosis across endothelial cells is the dominant pathway. Receptors involved are LDLR (LDL receptor), SR-B1 (scavenger receptor class B type 1), and ALK1 (activin receptor-like kinase 1). The role of LDLR is influenced by IL-1β (interleukin-1β); the role of SR-B1 by HDL (high-density lipoprotein), DOCK4 (dedicator of cytokinesis 4), GPER (G-protein-coupled estrogen receptor), and HMGB1 (high mobility group box 1); and the role of ALK1 by BMP (bone morphogenetic protein) 9. Additionally, BMP4 stimulates transcytosis and FSTL1 (follistatin-like 1 protein) inhibits it. Fundamental transcytotic mechanisms include caveola formation, undocking, trafficking, and docking; they are influenced by cholesterol-lowering agents, MYDGF (myeloid-derived growth factor), MFSD2a (major facilitator superfamily domain containing 2a) in the blood-brain barrier, and inhibitors of dynamin-2 and tubulin polymerization. The relative merits of different therapeutic approaches are discussed, with statins, colchicine, benzimidazoles, and metformin being existing drugs that might be repurposed and salidroside and glycyrrhizic acid being nutraceuticals worth investigating. Finally, we discuss evidence against the ferry-boat model of transcytosis, the contributions of receptor-mediated, fluid-phase, and active transcytosis, and where inhibition of transcytosis might be most beneficial.
Collapse
Affiliation(s)
- Israel O. Bolanle
- Department of Bioengineering, Imperial College London, United Kingdom
| | | | - Peter D. Weinberg
- Department of Bioengineering, Imperial College London, United Kingdom
| |
Collapse
|
7
|
Ao L, van Heemst D, Jukema JW, Rensen PCN, Willems van Dijk K, Noordam R. Potential causal evidence for an ApoB-independent and HDL-related risk profile associated with coronary artery disease. J Lipid Res 2025; 66:100778. [PMID: 40089107 PMCID: PMC12000745 DOI: 10.1016/j.jlr.2025.100778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025] Open
Abstract
Plasma 1H-NMR metabolomic measures have yielded significant insight into the pathophysiology of cardiometabolic disease, but their inter-related nature complicates causal inference and clinical interpretation. This study aimed to investigate the associations of unrelated 1H-NMR metabolomic profiles with coronary artery disease (CAD) and ischemic stroke (IS). Principal component (PC) analysis was performed on 168 1H-NMR metabolomic measures in 56,712 unrelated European participants from UK Biobank to retrieve uncorrelated PCs, which were used in Cox-proportional hazard models. For each outcome, two-sample Mendelian randomization analyses were then conducted based on three nonoverlapping databases, followed by a meta-analysis. The first six PCs collectively explaining 88% of the total variance were identified. For CAD, results from Cox and Mendelian randomization analyses were generally directionally consistent. The pooled odds ratios (95% CI) for CAD per one-SD increase in genetically influenced PC1 and PC3 (both characterized by distinct apolipoprotein B [ApoB]-associated lipoprotein profiles) were 1.04 (1.03, 1.05) and 0.94 (0.93, 0.96), respectively. Besides, the pooled odds ratio (95% CI) for CAD per one-SD increase in genetically influenced PC4, characterized by simultaneously decreased small HDL and increased large HDL, and independent of ApoB, was 1.05 (1.03, 1.07). For IS, increases of PC3 and PC5 (characterized by increased amino acids) were associated with a lower risk and a higher risk, respectively. This study confirms associations of ApoB-associated lipoprotein profiles with CAD and IS, and highlights the possible existence of an ApoB-independent lipoprotein profile, characterized by a distinctive HDL subparticle distribution, driving CAD.
Collapse
Affiliation(s)
- Linjun Ao
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
8
|
Hashem C, Altin SE, Guyton JR, Boden WE. Nonlinearity of the inverse relationship between high-density lipoprotein (HDL) cholesterol and incident cardiovascular risk: Is it time to revisit the "HDL hypothesis"? J Clin Lipidol 2025; 19:238-246. [PMID: 39934033 DOI: 10.1016/j.jacl.2024.12.009] [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: 08/12/2024] [Revised: 10/16/2024] [Accepted: 12/03/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Low levels of high-density lipoprotein cholesterol (HDL-C) are clearly associated with atherosclerotic cardiovascular disease (ASCVD), but the risk curve is not well defined, especially at very high and low HDL-C levels. Current proportional hazards prediction models assume inverse linearity of effect, which may not accurately represent risk at these levels. SOURCES OF MATERIAL Clinical inattention to risk associated with low HDL-C may derive from randomized controlled trials (RCTs) aimed at raising HDL-C, though most failed to reduce ASCVD events when combined with statin-based therapy. However, these prior trials enrolled patients with HDL-C levels largely in the 35-45 mg/dL range. ABSTRACT OF FINDINGS Mounting post hoc evidence from RCTs as well as new genetic and observational data suggest that very low HDL-C (less than 30 or 35 mg/dL) may signal a further increase in incident cardiovascular events. Moreover, when HDL-C exceeds 90 mg/dL, monotonic reduction of ASCVD risk appears to reverse. Because a pervasively agnostic view of the importance of both very low and high levels of HDL-C now exists, consideration should be given to incorporating nonlinear effects of HDL-C into future risk prediction models such that very low HDL-C and/or very high HDL-C levels could be considered as new risk-enhancing factors to promote more optimal risk stratification. CONCLUSION When revision of the U.S. Cholesterol Guideline recommences, consideration should be directed to whether HDL-associated risk matches the assumptions of current statistical models. Thus, it may be both timely and opportune to revisit the "HDL hypothesis" based on evolving scientific evidence.
Collapse
Affiliation(s)
- Carl Hashem
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States (Hashem)
| | - S Elissa Altin
- Division of Cardiology, Department of Medicine, Yale New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut, United States (Altin); Division of Cardiology, Department of Medicine, West Haven VA Medical Center, West Haven, Connecticut, United States (Altin)
| | - John R Guyton
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States (Guyton)
| | - William E Boden
- VA Boston Healthcare System, Boston, Massachusetts, United States (Boden); Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States (Boden).
| |
Collapse
|
9
|
Kalwick M, Roth M. A Comprehensive Review of the Genetics of Dyslipidemias and Risk of Atherosclerotic Cardiovascular Disease. Nutrients 2025; 17:659. [PMID: 40004987 PMCID: PMC11858766 DOI: 10.3390/nu17040659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Dyslipidemias are often diagnosed based on an individual's lipid panel that may or may not include Lp(a) or apoB. But these values alone omit key information that can underestimate risk and misdiagnose disease, which leads to imprecise medical therapies that reduce efficacy with unnecessary adverse events. For example, knowing whether an individual's dyslipidemia is monogenic can granularly inform risk and create opportunities for precision therapeutics. This review explores the canonical and non-canonical causes of dyslipidemias and how they impact atherosclerotic cardiovascular disease (ASCVD) risk. This review emphasizes the multitude of genetic causes that cause primary hypercholesterolemia, hypertriglyceridemia, and low or elevated high-density lipoprotein (HDL)-cholesterol levels. Within each of these sections, this review will explore the evidence linking these genetic conditions with ASCVD risk. Where applicable, this review will summarize approved therapies for a particular genetic condition.
Collapse
Affiliation(s)
| | - Mendel Roth
- GBinsight, GB Healthwatch, San Diego, CA 92122, USA;
| |
Collapse
|
10
|
Zhang J. Associations of neutrophil/high-density lipoprotein cholesterol ratio with frailty and its mortality. Front Endocrinol (Lausanne) 2025; 15:1495139. [PMID: 39835260 PMCID: PMC11743577 DOI: 10.3389/fendo.2024.1495139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Background Frailty is an increasingly important determinant in the field of health, and its identification has important clinical significance in the field of critical care medicine. However, there are still a large number of challenges in quick and accurate identification of frailty. This study aims to evaluate the value of the neutrophil/high-density lipoprotein cholesterol ratio (NHR) in frailty and its long-term survival. Methods Adult participants from seven study cycles of the National Health and Nutrition Examination Survey (NHANES) database were included. Frailty was assessed with a 49-item Frailty Index (FI). Weighted logistic regression, restricted cubic spline (RCS), and Cox regression were used to analyze the association of NHR with frailty and its long-term survival. In addition, subgroup and interaction analyses were also performed. Results A total of 34,382 adult participants aged 47.6 on average were included, and 16,950 (48.8%) of them were males. After the adjustment of potential confounding variables, an increase of one standard deviation (SD) in NHR resulted in the increase of the incidence of frailty by 11% (OR: 1.11, 95% CI: 1.04-1.18, P = 0.002). RCS showed a J-shaped association between NHR and frailty, which was robust in all subgroups according to the subgroup analysis. In addition, the survival analysis revealed that NHR was significantly positively associated with all-cause (HR: 1.12, 95% CI: 1.07-1.17, P < 0.0001), cardiocerebrovascular disease (CCD)-specific (HR: 1.21, 95% CI: 1.11-1.33, P < 0.0001), and cancer-specific mortality risks (HR: 1.13, 95% CI: 1.07-1.19, P < 0.0001) in frail individuals. Conclusion In the American adult population, NHR maintains a J-shaped relationship with frailty. In addition, NHR can help predict long-term mortality in frail individuals. This study demonstrates that NHR may become an effective predictor of frailty and its mortality.
Collapse
Affiliation(s)
- Jianqiang Zhang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
11
|
Romero-Jiménez MJ. HDL and cardiovascular risk. Atherosclerosis 2025; 400:119050. [PMID: 39551681 DOI: 10.1016/j.atherosclerosis.2024.119050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024]
|
12
|
Wang Y, Wang L, Zhao Z, Yin S, Tang X, Zhang K. The predictive role of the hs-CRP/HDL-C ratio for long-term mortality in the general population: evidence from a cohort study. BMC Cardiovasc Disord 2024; 24:758. [PMID: 39736563 PMCID: PMC11684128 DOI: 10.1186/s12872-024-04446-1] [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: 07/24/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND The high-sensitivity C-reactive protein (hs-CRP) to high-density lipoprotein cholesterol (HDL-C) ratio, a composite marker of low-grade inflammation and lipid metabolism, is reportedly associated with the occurrence of new cardiovascular diseases (CVDs) in certain people. However, the predictive value of the hs-CRP/HDL-C ratio for long-term mortality in the general population remains unclear. METHODS This retrospective cohort study included data from 9,492 adults obtained from the National Health and Nutrition Examination Survey (NHANES) (2015-2018) in the United States. Multivariate Cox regression, two-piecewise linear regression, restricted cubic spline (RCS) models and subgroup analysis by age, sex, smoking status and drinking status were applied to evaluate the associations of the hs-CRP/HDL-C ratio with long-term all-cause and cardiovascular mortality. RESULTS The overall median age of the cohort was 47.0 years (interquartile range (IQR) 32.0-62.0), and 4,585 (48.30%) patients were male. During a median follow-up period of 37.0 months, 239 (2.52%) all-cause deaths occurred, 59 (0.62%) of which were attributed to cardiovascular events. Participants with all-cause and cardiovascular mortality presented a higher hs-CRP/HDL-C ratio than did those without events [0.56 (0.24-1.38) vs. 0.37 (0.14-0.94) and 0.60 (0.23-1.60) vs. 0.37 (0.14-0.95), P < 0.001 and P = 0.002]. According to multivariate Cox regression models, the hs-CRP/HDL-C ratio was found to be an independent risk factor for both long-term all-cause mortality [hazard ratio (HR) = 1.09, 95% confidence interval (CI): 1.05-1.13] and cardiovascular mortality (HR = 1.11, 95% CI: 1.05-1.19). A two-piecewise linear regression model indicated that the risk of all-cause mortality increased more prominently when the hs-CRP/HDL-C ratio was less than 1.21. In addition, a significant interaction effect with smoking status was discovered (P = 0.006), indicating that the association of the hs-CRP/HDL-C ratio with all-cause mortality was stronger in nonsmokers. The RCS curve revealed a positive linear association of the hs-CRP/HDL-C ratio with long-term mortality after adjustment for potential confounders. CONCLUSIONS The hs-CRP/HDL-C ratio is a crucial predictor of long-term mortality in the general population, independent of potential confounding factors.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Li Wang
- Community Health Center, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, Jiangsu, 215000, China
| | - Zongquan Zhao
- Community Health Center, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, Jiangsu, 215000, China
| | - Song Yin
- Community Health Center, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, Jiangsu, 215000, China
| | - Xuejun Tang
- Community Health Center, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, Jiangsu, 215000, China
| | - Kerui Zhang
- Center for Cardiovascular Disease, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, Jiangsu, 215000, China.
| |
Collapse
|
13
|
Lui DTW, Li L, Liu X, Xiong X, Tang EHM, Lee CH, Woo YC, Lang BHH, Wong CKH, Tan KCB. The association of HDL-cholesterol levels with incident major adverse cardiovascular events and mortality in 0.6 million individuals with type 2 diabetes: a population-based retrospective cohort study. BMC Med 2024; 22:586. [PMID: 39696353 DOI: 10.1186/s12916-024-03810-4] [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: 06/19/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND High levels of high-density lipoprotein cholesterol (HDL-C) are previously considered protective against cardiovascular diseases (CVD), but recent studies suggest an increased risk of adverse events at very high HDL-C levels in the general population. It remains to be elucidated such a relationship in diabetes, a condition with high cardiovascular risks. We examined the association of HDL-C levels with the risk of major adverse cardiovascular events (MACE) and mortality in type 2 diabetes. METHODS This retrospective cohort study identified individuals with type 2 diabetes who had HDL-C records (2008-2020) from the electronic health record database of the Hong Kong Hospital Authority. They were classified into three groups based on their first-recorded HDL-C levels following diabetes diagnosis: low (≤ 40 mg/dL), medium (> 40 and ≤ 80 mg/dL) and high HDL-C (> 80 mg/dL) groups. The primary outcome was incident MACE (composite of myocardial infarction, stroke, heart failure, and cardiovascular mortality). Cox regression model and restricted cubic spline analysis were employed to assess the relationship between HDL-C and adverse outcomes. RESULTS Among 596,943 individuals with type 2 diabetes included, 168,931 (28.30%), 412,863 (69.16%), and 15,149 (2.54%) were classified as low HDL-C, medium HDL-C, and high HDL-C groups, respectively. Over a median follow-up of 79.5 months, both low and high HDL-C groups had higher risk of incident MACE compared to the medium HDL-C group (HR 1.24, 95% CI 1.23-1.26, P < 0.001; HR 1.09, 95% CI 1.04-1.13, P < 0.001). The spline curves revealed a U-shaped association between HDL-C levels and incident MACE (non-linear p < 0.001). Similar U-shaped relationship was observed for all-cause and non-cardiovascular mortality. CONCLUSIONS Our study demonstrated a U-shaped association between HDL-C levels and incident MACEs and all-cause and non-cardiovascular mortality in individuals with type 2 diabetes, highlighting the need for mechanistic studies on the adverse outcomes seen at high HDL-C levels in type 2 diabetes.
Collapse
Affiliation(s)
- David Tak Wai Lui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Lanlan Li
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaodong Liu
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
| | - Xi Xiong
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Research Department of Practice and Policy, School of Pharmacy, University College London, London, UK
| | - Eric Ho Man Tang
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chi Ho Lee
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Cho Woo
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Brian Hung Hin Lang
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Carlos King Ho Wong
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China.
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Department of Infectious Disease Epidemiology & Dynamics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| | - Kathryn Choon Beng Tan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
14
|
Hao D, Xue JY, Wang Q, Guo L, Li XA. The Role of Scavenger Receptor BI in Sepsis. Int J Mol Sci 2024; 25:13441. [PMID: 39769206 PMCID: PMC11677381 DOI: 10.3390/ijms252413441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Sepsis is a life-threatening condition resulting from a dysregulated host response to infection. Currently, there is no effective therapy for sepsis due to an incomplete understanding of its pathogenesis. Scavenger receptor BI (SR-BI) is a high-density lipoprotein (HDL) receptor that plays a key role in HDL metabolism by modulating the selective uptake of cholesteryl ester from HDL. Recent studies, including those from our laboratory, indicate that SR-BI protects against sepsis through multiple mechanisms: (1) preventing nitric oxide-induced cytotoxicity; (2) promoting hepatic lipopolysaccharide (LPS) clearance and regulating cholesterol metabolism in the liver; (3) inhibiting LPS-induced inflammatory signaling in macrophages; and (4) mediating the uptake of cholesterol from HDL for inducible glucocorticoid (iGC) synthesis in the adrenal gland, which controls systemic inflammatory response. In this article, we review the roles of SR-BI in sepsis.
Collapse
Affiliation(s)
- Dan Hao
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jian-Yao Xue
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Qian Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Ling Guo
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Xiang-An Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
- Lexington VA Healthcare System, Lexington, KY 40502, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
15
|
Tillison EA, Sahoo D. Sticky Business: Correlating Oligomeric Features of Class B Scavenger Receptors to Lipid Transport. Curr Atheroscler Rep 2024; 27:15. [PMID: 39630384 DOI: 10.1007/s11883-024-01260-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE OF THE REVIEW Atherosclerotic plaques result from imbalanced lipid metabolism and maladaptive chronic immune responses. Class B scavenger receptors are lipid transporters and regulators of their metabolism. The purpose of this review is to explore recent structural findings of these membrane-associated receptors, with particular focus on their higher-order oligomeric organization and impact on lipid transport. RECENT FINDINGS Class B scavenger receptors have evidence for oligomerization, with recent efforts placed on identifying residues and motifs responsible for mediating this process. The first studies correlating scavenger receptor oligomerization to function are described. This review highlights two emerging hypotheses regarding the function of scavenger receptor oligomerization. The first is a hydrophobic channel created by self-association of receptors to promote transport. The second hypothesis suggests that homo-oligomerization stabilizes receptors, prevents internalization and thereby promotes transport indirectly. Novel computational and in vitro experimental techniques with purified receptors are also described.
Collapse
Affiliation(s)
- Emma A Tillison
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Division of Endocrinology & Molecular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- H4930 Health Research Center, Medical College of Wisconsin, 8701 W. Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
16
|
Potapenko A, Frey K, Schlumpf E, Robert J, Wollscheid B, von Eckardstein A, Rohrer L. The two major splice variants of scavenger receptor BI differ by their interactions with lipoproteins and cellular localization in endothelial cells. J Lipid Res 2024; 65:100665. [PMID: 39393447 PMCID: PMC11585690 DOI: 10.1016/j.jlr.2024.100665] [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: 10/23/2023] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/13/2024] Open
Abstract
The scavenger receptor BI (SR-BI) facilitates the transport of both HDL and LDL through endothelial cells. Its two splice variants, SR-BIvar1 and SR-BIvar2, differ in their carboxy terminal domains. Only SR-BIvar1 contains the putative binding sites for the adapter proteins PDZ domain containing protein 1 (PDZK1) and dedicator of cytokinesis 4 (DOCK4), which limit the cell surface abundance and internalization of the receptor. To investigate the cellular localization of the SR-BI variants and their interaction with lipoproteins in endothelial cells, EA.hy926 cells were stably transfected with vectors encoding untagged, GFP- or mCherry-tagged constructs of the two SR-BI variants. Additionally, the cells were transfected with shRNAs against PDZK1 or DOCK4. Microscopy investigation showed that SR-BIvar1 was predominantly localized on the cell surface together with clathrin whereas SR-BIvar2 was absent from the cell surface but retrieved in endosomes and lysosomes. Accordingly, only SR-BIvar1 increased lipoprotein binding to endothelial while HDL and LDL uptake were enhanced by both variants. Silencing of PDZK1 or DOCK4 only reduced HDL association in SR-BIvar2 overexpressing cells while LDL association was reduced both in WT and SR-BIvar2 overexpressing cells. In conclusion, either SR-BI variant facilitates the uptake of HDL and LDL into endothelial cells, however by different mechanisms and trafficking routes. This dual role may explain why the loss of DOCK4 or PDZK1 differently affects the uptake of HDL and LDL in different endothelial cells.
Collapse
Affiliation(s)
- Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Kathrin Frey
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland; Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Jérôme Robert
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland.
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Al Zein M, Khazzeka A, El Khoury A, Al Zein J, Zoghaib D, Eid AH. Revisiting high-density lipoprotein cholesterol in cardiovascular disease: Is too much of a good thing always a good thing? Prog Cardiovasc Dis 2024; 87:50-59. [PMID: 39442601 DOI: 10.1016/j.pcad.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular disease (CVD) continues to be a leading cause of global mortality and morbidity. Various established risk factors are linked to CVD, and modifying these risk factors is fundamental in CVD management. Clinical studies underscore the association between dyslipidemia and CVD, and therapeutic interventions that target low-density lipoprotein cholesterol elicit clear benefits. Despite the correlation between low high-density lipoprotein cholesterol (HDLC) and heightened CVD risk, HDL-raising therapies have yet to showcase significant clinical benefits. Furthermore, evidence from epidemiological and genetic studies reveals that not only low HDL-C levels, but also very high levels of HDL-C are linked to increased risk of CVD. In this review, we focus on HDL metabolism and delve into the relationship between HDL and CVD, exploring HDL functions and the observed alterations in its roles in disease. Altogether, the results discussed herein support the conventional wisdom that "too much of a good thing is not always a good thing". Thus, our recommendation is that a careful reconsideration of the impact of high HDL-C levels is warranted, and shall be revisited in future research.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Alicia Khazzeka
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Jana Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Dima Zoghaib
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
18
|
Jakubowski H. The Molecular Bases of Anti-Oxidative and Anti-Inflammatory Properties of Paraoxonase 1. Antioxidants (Basel) 2024; 13:1292. [PMID: 39594433 PMCID: PMC11591180 DOI: 10.3390/antiox13111292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
The anti-oxidative and anti-inflammatory properties of high-density lipoprotein (HDL) are thought to be mediated by paraoxonase 1 (PON1), a calcium-dependent hydrolytic enzyme carried on a subfraction of HDL that also carries other anti-oxidative and anti-inflammatory proteins. In humans and mice, low PON1 activity is associated with elevated oxidized lipids and homocysteine (Hcy)-thiolactone, as well as proteins that are modified by these metabolites, which can cause oxidative stress and inflammation. PON1-dependent metabolic changes can lead to atherothrombotic cardiovascular disease, Alzheimer's disease, and cancer. The molecular bases underlying these associations are not fully understood. Biochemical, proteomic, and metabolic studies have significantly expanded our understanding of the mechanisms by which low PON1 leads to disease and high PON1 is protective. The studies discussed in this review highlight the changes in gene expression affecting proteostasis as a cause of the pro-oxidative and pro-inflammatory phenotypes associated with attenuated PON1 activity. Accumulating evidence supports the conclusion that PON1 regulates the expression of anti-oxidative and anti-inflammatory proteins, and that the disruption of these processes leads to disease.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, University of Life Sciences, 60-637 Poznań, Poland; ; Tel.: +1-973-972-8733; Fax: 973-972-8981
- Department of Microbiology, Biochemistry and Molecular Genetics, International Center for Public Health, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
19
|
Madaudo C, Bono G, Ortello A, Astuti G, Mingoia G, Galassi AR, Sucato V. Dysfunctional High-Density Lipoprotein Cholesterol and Coronary Artery Disease: A Narrative Review. J Pers Med 2024; 14:996. [PMID: 39338250 PMCID: PMC11432852 DOI: 10.3390/jpm14090996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
High-density lipoprotein (HDL) cholesterol is traditionally viewed as protective against cardiovascular disease (CVD). However, emerging evidence reveals that dysfunctional HDL, characterized by impaired reverse cholesterol transport (RCT), reduced anti-inflammatory and antioxidant activities and increased endothelial dysfunction, which can contribute to coronary artery disease (CAD). Dysfunctional HDL, resulting from oxidative modifications of Apolipoprotein A-1 (Apo A-1) and enzyme inactivation, fails to effectively remove cholesterol from peripheral tissues and may promote inflammation and atherosclerosis. Genetic mutations affecting HDL metabolism further complicate its role in cardiovascular health. Studies have shown that conventional therapies aimed at raising HDL-C levels do not necessarily reduce cardiovascular events, highlighting the need for new approaches that improve HDL functionality. Therapeutic strategies such as Apo A-1 mimetic peptides, reconstituted HDL infusions, and drugs targeting specific HDL metabolic pathways are being explored. Additionally, weight loss, statin therapy, and niacin have shown potential in enhancing HDL function. The pathophysiology of dysfunctional HDL involves complex mechanisms, including oxidative stress, inflammation, and genetic mutations, which alter its structure and function, diminishing its cardioprotective effects. New functional assays, such as the cholesterol efflux capacity (CEC) and HDL inflammatory index, provide more accurate predictions of cardiovascular risk by assessing HDL quality rather than quantity. As research progresses, the focus is shifting towards therapeutic strategies that enhance HDL function and address the root causes of its dysfunction, offering a more effective approach to reducing cardiovascular risk and preventing CAD.
Collapse
Affiliation(s)
- Cristina Madaudo
- Division of Cardiology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital Paolo Giaccone, University of Palermo, 90127 Palermo, Italy
| | | | | | | | | | | | | |
Collapse
|
20
|
Xiang N, Su S, Yang Y, Luo Y, Fu T, Wang L, Lin Y, Huang J. Genetic support of causal association between lipid and glucose metabolism and stress urinary incontinence in women: a bidirectional Mendelian randomization and multivariable-adjusted study. Front Endocrinol (Lausanne) 2024; 15:1394252. [PMID: 39351534 PMCID: PMC11439682 DOI: 10.3389/fendo.2024.1394252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/23/2024] [Indexed: 10/04/2024] Open
Abstract
Background Stress urinary incontinence (SUI) is a common condition characterized by urethral sphincter failure and urine leakage. Its prevalence in women is higher than in men, and estimates of crude prevalence rates vary widely due to factors such as research methodologies, study populations, and underreporting by patients. This variability hinders research and impacts patient diagnosis, treatment, and quality of life. The complex etiology of SUI is not fully understood, and previous studies have primarily focused on non-invasive indicators. While emerging observational research suggests a correlation between SUI in women and abnormalities in lipid and blood metabolism, the underlying biological mechanisms and causal relationships require further investigation. This study aims to explore the causalities between SUI in women and lipid and blood metabolism. Methods Using bidirectional univariate Mendelian randomization (MR), we investigated the causal association between SUI liability in women (case/control = 5,924/399,509) from UK Biobank and lipid and glucose metabolism, indicated by total cholesterol (TC, N = 61,166), low-density lipoproteins (LDL, N = 58,381), high-density lipoproteins (HDL, N = 60,812), triglycerides (TG, N = 60,027), fasting glucose (FG, N = 19,745), and fasting insulin (FI, N = 38,238) from ENGAGE consortium. To account for potential confounding effects, multivariable MR (MVMR) analyses were performed, adjusting for body mass index (BMI) and separately among lipid and glucose metabolism. Results We found that increased genetically proxied TC, LDL, and HDL levels were associated with an elevated risk of SUI in women (OR: 1.090-1.117, all P < 0.05), These associations were further supported by MVMR analyses with adjustment for BMI (OR: 1.087-1.114, all P < 0.05). Conversely, increased FG and FI were associated with reduced SUI reliability in women (OR: 0.731-0.815, all P < 0.05). When adjusting among lipid and glucose metabolism, only HDL and FI demonstrated causal effects. Reverse MR analyses provided no genetic evidence supporting the causal effect of SUI in women on lipid and blood metabolism (all P > 0.05). Conclusions Our results reported that increased TC, LDL, and HDL are linked to higher SUI susceptibility in women, while higher FG and FI levels have a protective effect. In overweight/obese women with metabolic abnormalities, the positive associations between TC, LDL, and HDL levels and SUI indicate a higher risk.
Collapse
Affiliation(s)
- Nanyan Xiang
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
| | - Shiqi Su
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
| | - Yong Yang
- Health Management Center, General Practice Medical Center, Innovation Institute for Integration of Medicine and Engineering, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yurui Luo
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
| | - Tingting Fu
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Le Wang
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yifei Lin
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Jin Huang
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Kumar M, Ali W, Yadav K, Kaumri S, Mishra S, Nardi P, Iellamo F, Bernardini S, Pradhan A, Perrone MA. High-Density Lipoprotein-Associated Paraoxonase-1 (PON-1) and Scavenger Receptor Class B Type 1 (SRB-1) in Coronary Artery Disease: Correlation with Disease Severity. J Clin Med 2024; 13:5480. [PMID: 39336967 PMCID: PMC11432482 DOI: 10.3390/jcm13185480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Coronary artery disease (CAD) is the leading cause of death worldwide. High-Density lipoprotein (HDL) is a well-established marker associated with CAD. The current research goes beyond the conventional HDL-C measurement in previous studies and dives into the functional intricacies of HDL. By understanding how HDL works, rather than just how much of it exists, we can better tailor diagnostic and therapeutic strategies for CAD and related conditions. Hence, the current study quantifies the serum levels of two novel HDL-associated markers, Paraoxonase-1 (PON-1) and Scavenger Receptor Class B Type 1 (SRB-1), in CAD cases vs. controls. Methods: A total of 92 subjects, including 69 CAD and 23 healthy controls, were included, based on the prevalence of the disease. Further, based on the severity of the disease, CAD cases were subcategorized as CAD-I, -II, and -III. Serum PON-1 and SRB-1 levels were measured and compared between patient and control groups. Results: The levels of PON-1 and SRB-1 (32.6 ng/mL and 12.49 ng/mL) were significantly lower in CAD patients vs. the healthy control, at 60.36 ng/mL and 15.85 ng/mL, respectively (p < 0.000). A further intergroup comparison showed a statistically significant difference between the CAT-I and -III for PON-1 (p < 0.025), the CAT-I and -III, and CAT-II and -III for SRB-1 (p < 0.000). The receiver operating characteristics (ROC) curve showed cutoff values of 48.20 ng/mL and 14.90 ng/mL for PON-1 and SRB-1. Conclusions: The current study found that serum levels of HDL-associated PON-1 and SRB-1 are significantly lower in CAD cases, and were also inversely related to the increasing severity of coronary artery disease. This inference implies that serum PON-1 and SRB-1 could be used as non-invasive tools for the identification of coronary atherosclerosis and risk assessment in CAD cases.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Wahid Ali
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Kusum Yadav
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226003, Uttar Pradesh, India; (K.Y.); (S.M.)
| | - Swati Kaumri
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226003, Uttar Pradesh, India; (K.Y.); (S.M.)
| | - Paolo Nardi
- Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Ferdinando Iellamo
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.I.); (M.A.P.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Akshyaya Pradhan
- Lari Cardiology Center, King George Medical University, Lucknow 226003, Uttar Pradesh, India;
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.I.); (M.A.P.)
| |
Collapse
|
22
|
Mamede I, Braga MAP, Martins OC, Franchini AEO, Silveira Filho RB, Santos MCF. Association between very high HDL-C levels and mortality: A systematic review and meta-analysis. J Clin Lipidol 2024; 18:e701-e709. [PMID: 39278774 DOI: 10.1016/j.jacl.2024.06.002] [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: 02/03/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Recent research has raised questions about the assumed cardiovascular (CV) benefits of high-density lipoprotein cholesterol (HDL-C) and the potential for adverse outcomes with extremely high levels. OBJECTIVE We conducted a meta-analysis to investigate the association between very high HDL-C levels (≥80 mg/dL) and mortality outcomes in individuals without coronary artery disease (CAD). METHODS We systematically searched PubMed, Embase, and Cochrane databases for studies comparing very high HDL-C levels to normal levels (40-60 mg/dL) in CAD-free individuals. We assessed heterogeneity using I2 statistics with a random-effects model. RESULTS Our analysis included 1,004,584 individuals from 8 studies, of whom 133,646 (13.3%) had very high HDL-C levels. All-cause mortality did not significantly differ between groups (p = 0.55), nor did cancer mortality (p = 0.45). Cardiovascular mortality showed no change in those with very high HDL-C (hazard ratio [HR] 1.05; 95% confidence interval [CI] 0.94-1.17; p = 0.37). Fatal and non-fatal coronary heart disease events were less frequent in the very high HDL-C group (HR 0.79; 95% CI 0.73-0.86; p < 0.00001). Subgroup dose-response analysis revealed that very high HDL-C levels increased cardiovascular death in women above 116 mg/dL (HR 1.47; 95% CI 1.01-2.15) and in men above 94 mg/dL (HR 1.29; 95% CI 1.01-1.65) (p_nonlinearity <0.01). CONCLUSIONS These findings suggest that very high HDL-C levels are not protective against CV mortality and may, in fact, increase CV mortality risk especially in men.
Collapse
Affiliation(s)
- Isadora Mamede
- Faculty of Medicine, Federal University of Sao Joao del-Rei, Centro Oeste Campus, Divinopolis, Brazil (Dr Mamede).
| | | | - Otavio C Martins
- Faculty of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Brazil (Dr Martins)
| | - Anne E O Franchini
- Faculty of Medicine, President Tancredo de Almeida Neves University Center, Sao Joao del Rei, Brazil (Dr Franchini)
| | | | - Marcel C F Santos
- Department of Internal Medicine, University of Sao Paulo, Sao Paulo, Brazil (Dr Santos)
| |
Collapse
|
23
|
Wang Z, Barinas-Mitchell E, Brooks MM, Crawford SL, Leis AM, Derby CA, Thurston RC, Hedderson MM, Janssen I, Jackson EA, McConnell DS, El Khoudary SR. HDL-C criterion of the metabolic syndrome and future diabetes and atherosclerosis in midlife women: The SWAN Study. Am J Prev Cardiol 2024; 19:100687. [PMID: 39070021 PMCID: PMC11279330 DOI: 10.1016/j.ajpc.2024.100687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/21/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Objective High-density lipoprotein cholesterol (HDL-C) is one of 5 components [high blood pressure, glucose, triglycerides, waist circumference, low HDL-C], 3 of which, needed to diagnose metabolic syndrome (MetS). Evolving research shows that higher HDL-C is not necessarily cardioprotective in midlife women, supporting a need to re-evaluate HDL-C's contribution to risks related to MetS. We tested whether risk of future diabetes and higher carotid intima-media thickness (cIMT) differ by HDL-C status in midlife women diagnosed with MetS based on the other 4 components. Methods Midlife women were classified into 3 groups 1) no MetS, 2) MetS with HDL-C ≥ 50 mg/dL (MetS hiHDL), and 3) MetS with HDL-C < 50 mg/dL (MetS loHDL). cIMT was measured 13.8 ± 0.6 years post baseline. Incident diabetes was assessed yearly. Results Among 2773 women (1350 (48 %) of them had cIMT), 2383 (86 %) had no MetS, 117 (4 %) had MetS hiHDL, 273 (10 %) had MetS loHDL. Compared with no MetS, both MetS- hiHDL and loHDL groups had higher cIMT and diabetes risk. Risk of having high cIMT did not differ between MetS loHDL vs. hiHDL groups. Adjusting for levels of MetS criteria other than HDL-C at baseline explained the associations of each of the two MetS groups with cIMT. Conversely, after adjustment, associations of MetS hiHDL and MetS loHDL with incident diabetes persisted. Conclusions In midlife women, HDL-C status matters for predicting risk of incident diabetes but not higher cIMT beyond other MetS components.
Collapse
Affiliation(s)
- Ziyuan Wang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh PA, USA
| | - Emma Barinas-Mitchell
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh PA, USA
| | - Maria M. Brooks
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh PA, USA
| | - Sybil L. Crawford
- Tan Chingfen Graduate School of Nursing, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Aleda M. Leis
- Department of Epidemiology, The University of Michigan, Ann Arbor, MI, USA
| | - Carol A. Derby
- Departments of Neurology, and of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rebecca C. Thurston
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Monique M. Hedderson
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA, USA
| | - Imke Janssen
- Department of Family and Preventive Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Elizabeth A. Jackson
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Samar R. El Khoudary
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh PA, USA
| |
Collapse
|
24
|
Shamsuzzaman S, Deaton RA, Salamon A, Doviak H, Serbulea V, Milosek VM, Evans MA, Karnewar S, Saibaba S, Alencar GF, Shankman LS, Walsh K, Bekiranov S, Kocher O, Krieger M, Kull B, Persson M, Michaëlsson E, Bergenhem N, Heydarkhan-Hagvall S, Owens GK. Novel Mouse Model of Myocardial Infarction, Plaque Rupture, and Stroke Shows Improved Survival With Myeloperoxidase Inhibition. Circulation 2024; 150:687-705. [PMID: 38881440 PMCID: PMC11347105 DOI: 10.1161/circulationaha.123.067931] [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: 11/10/2023] [Accepted: 05/22/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Thromboembolic events, including myocardial infarction (MI) or stroke, caused by the rupture or erosion of unstable atherosclerotic plaques are the leading cause of death worldwide. Although most mouse models of atherosclerosis develop lesions in the aorta and carotid arteries, they do not develop advanced coronary artery lesions. Moreover, they do not undergo spontaneous plaque rupture with MI and stroke or do so at such a low frequency that they are not viable experimental models to study late-stage thrombotic events or to identify novel therapeutic approaches for treating atherosclerotic disease. This has stymied the development of more effective therapeutic approaches for reducing these events beyond what has been achieved with aggressive lipid lowering. Here, we describe a diet-inducible mouse model that develops widespread advanced atherosclerosis in coronary, brachiocephalic, and carotid arteries with plaque rupture, MI, and stroke. METHODS We characterized a novel mouse model with a C-terminal mutation in the scavenger receptor class B, type 1 (SR-BI), combined with Ldlr knockout (designated SR-BI∆CT/∆CT/Ldlr-/-). Mice were fed Western diet (WD) for 26 weeks and analyzed for MI and stroke. Coronary, brachiocephalic, and carotid arteries were analyzed for atherosclerotic lesions and indices of plaque stability. To validate the utility of this model, SR-BI∆CT/∆CT/Ldlr-/- mice were treated with the drug candidate AZM198, which inhibits myeloperoxidase, an enzyme produced by activated neutrophils that predicts rupture of human atherosclerotic lesions. RESULTS SR-BI∆CT/∆CT/Ldlr-/- mice show high (>80%) mortality rates after 26 weeks of WD feeding because of major adverse cardiovascular events, including spontaneous plaque rupture with MI and stroke. Moreover, WD-fed SR-BI∆CT/∆CT/Ldlr-/- mice displayed elevated circulating high-sensitivity cardiac troponin I and increased neutrophil extracellular trap formation within lesions compared with control mice. Treatment of WD-fed SR-BI∆CT/∆CT/Ldlr-/- mice with AZM198 showed remarkable benefits, including >90% improvement in survival and >60% decrease in the incidence of plaque rupture, MI, and stroke, in conjunction with decreased circulating high-sensitivity cardiac troponin I and reduced neutrophil extracellular trap formation within lesions. CONCLUSIONS WD-fed SR-BI∆CT/∆CT/Ldlr-/- mice more closely replicate late-stage clinical events of advanced human atherosclerotic disease than previous models and can be used to identify and test potential new therapeutic agents to prevent major adverse cardiac events.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Diet, Western/adverse effects
- Disease Models, Animal
- Enzyme Inhibitors/therapeutic use
- Enzyme Inhibitors/pharmacology
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Infarction/pathology
- Myocardial Infarction/drug therapy
- Peroxidase/metabolism
- Plaque, Atherosclerotic/drug therapy
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Rupture, Spontaneous
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Stroke/drug therapy
- Stroke/prevention & control
Collapse
Affiliation(s)
- Sohel Shamsuzzaman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Rebecca A. Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Anita Salamon
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Heather Doviak
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Victoria M. Milosek
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA, USA
| | - Megan A. Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Subhi Saibaba
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Gabriel F. Alencar
- Beirne B Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kenneth Walsh
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Stefan Bekiranov
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Medical Deaconess Medical Center Harvard Medical School, Boston, MA, USA
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bengt Kull
- BioPharmaceuticals R&D Early Cardiovascular Renal and Metabolism (CVRM) Bioscience Cardiovascular, AstraZeneca, Mölndal 43183, Sweden
| | - Marie Persson
- BioPharmaceuticals R&D Early Cardiovascular Renal and Metabolism (CVRM) DMPK, AstraZeneca, Mölndal 43183, Sweden
| | - Erik Michaëlsson
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal, and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nils Bergenhem
- Alliance Management, Business Development & Licensing, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - Sepideh Heydarkhan-Hagvall
- AstraZeneca R&D, Chief Medical Office, Global Patient Safety, Pepparedsleden 1, Mölndal, SE43183, Sweden
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
25
|
Dong X, Xiong YT, He T, Zheng C, Li J, Zhuang Y, Xu Y, Xiu Y, Wu Z, Zhao X, Xiao X, Bai Z, Gao L. Protective effects of Nogo-B deficiency in NAFLD mice and its multiomics analysis of gut microbiology and metabolism. GENES & NUTRITION 2024; 19:17. [PMID: 39182019 PMCID: PMC11344411 DOI: 10.1186/s12263-024-00754-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver ailment that can lead to serious conditions such as cirrhosis and hepatocellular carcinoma. Hepatic Nogo-B regulates glucose and lipid metabolism, and its inhibition has been shown to be protective against metabolic syndrome. Increasing evidence suggests that imbalances in the gut microbiota (GM) and lipid metabolism disorders are significant contributors to NAFLD progression. Nevertheless, it is not yet known whether Nogo-B can affect NAFLD by influencing the gut microbiota and metabolites. Hence, the aim of the present study was to characterize this process and explore its possible underlying mechanisms. METHODS A NAFLD model was constructed by administering a high-fat diet (HFD) to Nogo-B-/- and WT mice from the same litter, and body weight was measured weekly in each group. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed to assess blood glucose levels. At the end of the 12-week period, samples of serum, liver, and intestinal contents were collected and used for serum biochemical marker and inflammatory factor detection; pathology evaluation; and gut microbiome and metabolomics analysis. Spearman's correlation analysis was performed to determine possible correlations between differential gut microbiota and differential serum metabolites between groups. RESULTS Nogo-B deficiency attenuated the effects of the HFD, including weight gain, liver weight gain, impaired glucose tolerance, hepatic steatosis, elevated serum lipid biochemicals levels, and liver function. Nogo-B deficiency suppressed M1 polarization and promoted M2 polarization, thus inhibiting inflammatory responses. Furthermore, Nogo-B-/--HFD-fed mice presented increased gut microbiota richness and diversity, decreased Firmicutes/Bacteroidota (F/B) ratios, and altered serum metabolites compared with those of WT-HFD-fed mice. During analysis, several differential gut microbiota, including Lachnoclostridium, Harryflintia, Odoribacter, UCG-009, and unclassified_f_Butyricoccaceae, were screened between groups. These microbiota were found to be positively correlated with upregulated purine metabolism and bile acid metabolites in Nogo-B deficiency, while they were negatively correlated with downregulated corticosterone and tricarboxylic acid cyclic metabolites in Nogo-B deficiency. CONCLUSION Nogo-B deficiency delayed NAFLD progression, as demonstrated by reduced hepatocellular lipid accumulation, attenuated inflammation and liver injury, and ameliorated gut microbiota dysbiosis and metabolic disorders. Importantly, Odoribacter was strongly positively correlated with ALB and taurodeoxycholic acid, suggesting that it played a considerable role in the influence of Nogo-B on the progression of NAFLD, a specific feature of NAFLD in Nogo-B-/- mice. The regulation of bile acid metabolism by the gut microbiota may be a potential target for Nogo-B deficiency to ameliorate NAFLD.
Collapse
Affiliation(s)
- Xu Dong
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yu-Ting Xiong
- 307 Clinical Medical College of PLA, Anhui Medical University, Beijing, China
| | - Tingting He
- Department of Hepatology Medicine of Traditional Chinese Medicine, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Congyang Zheng
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junjie Li
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Chengde Medical University, Chengdeshi, China
| | - Yingjie Zhuang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingjie Xu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ye Xiu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhixin Wu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaomei Zhao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Lili Gao
- Medical School of Chinese PLA, Beijing, China.
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
26
|
Huang H, Yang B, Yu R, Ouyang W, Tong J, Le Y. Very high high-density lipoprotein cholesterol may be associated with higher risk of cognitive impairment in older adults. Nutr J 2024; 23:79. [PMID: 39020341 PMCID: PMC11253464 DOI: 10.1186/s12937-024-00983-9] [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: 11/26/2023] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Previous studies have shown that high-density lipoprotein cholesterol (HDL-C) levels are positively associated with cognitive function across a range of concentrations. However, recent studies have suggested that very high HDL-C levels may lead to poorer outcomes. Therefore, we aimed to investigate the relationship between different concentrations of HDL-C and cognitive impairment risk. METHODS We collected data from 3632 participants aged over 60 years from the U.S. National Health and Nutrition Examination Survey (NHANES) between 2011 and 2014 to assess the relationship between HDL-C and cognitive function. Cognitive function was evaluated with the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) test, the animal fluency test (AFT), and the digit symbol substitution test (DSST). We used restricted cubic spline models and logistic regression to examine the association between HDL-C and cognitive function. RESULTS A U-shaped was observed between HDL-C and cognitive outcomes, individuals with higher risk in those with both low and very high HDL-C levels compared with those with midrange values. Very high HDL-C levels (≥ 2.50 mmol/L) were associated with increased risk of cognitive impairment (OR = 2.19; 95% CI, 1.12-4.28) compared with those with HDL-C levels in the range of 1.50 to 1.99 mmol/L in older adults after adjustment for confounding factors. Interaction test demonstrated that relationship between very high HDL-C and the risk of cognitive impairment was not changed in different sex and race group (P for interaction > 0.05). CONCLUSIONS Very high HDL-C levels were associated with an increased risk of cognitive impairment. HDL-C may not be a protective factor for maintaining brain health in older adults at very high levels.
Collapse
Affiliation(s)
- Huifan Huang
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Anesthesiology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Bin Yang
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Renhe Yu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Wen Ouyang
- Department of Anesthesiology, the Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Province Key Laboratory of Brain Homeostasis, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Jianbin Tong
- Department of Anesthesiology, the Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Province Key Laboratory of Brain Homeostasis, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Le
- Department of Anesthesiology, the Third Xiangya Hospital, Central South University, Changsha, China.
- Hunan Province Key Laboratory of Brain Homeostasis, the Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
27
|
Guo S, Tian Y, Li J, Zeng X. A Glimpse into Humoral Response and Related Therapeutic Approaches of Takayasu's Arteritis. Int J Mol Sci 2024; 25:6528. [PMID: 38928233 PMCID: PMC11203527 DOI: 10.3390/ijms25126528] [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: 05/04/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Takayasu's arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing body of evidence bears testimony to the widespread effects of B cells on disease pathogenesis and progression. Distinct alterations in peripheral B cell subsets have been described in individuals with TAK. Advancements in technology have facilitated the identification of novel autoantibodies in TAK. Moreover, emerging data suggest that dysregulated signaling cascades downstream of B cell receptor families, including interactions with innate pattern recognition receptors such as toll-like receptors, as well as co-stimulatory molecules like CD40, CD80 and CD86, may result in the selection and proliferation of autoreactive B cell clones in TAK. Additionally, ectopic lymphoid neogenesis within the aortic wall of TAK patients exhibits functional characteristics. In recent decades, therapeutic interventions targeting B cells, notably utilizing the anti-CD20 monoclonal antibody rituximab, have demonstrated efficacy in TAK. Despite the importance of the humoral immune response, a systematic understanding of how autoreactive B cells contribute to the pathogenic process is still lacking. This review provides a comprehensive overview of the biological significance of B cell-mediated autoimmunity in TAK pathogenesis, as well as insights into therapeutic strategies targeting the humoral response. Furthermore, it examines the roles of T-helper and T follicular helper cells in humoral immunity and their potential contributions to disease mechanisms. We believe that further identification of the pathogenic role of autoimmune B cells and the underlying regulation system will lead to deeper personalized management of TAK patients. We believe that further elucidation of the pathogenic role of autoimmune B cells and the underlying regulatory mechanisms holds promise for the development of personalized approaches to managing TAK patients.
Collapse
Affiliation(s)
- Shuning Guo
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Yixiao Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| |
Collapse
|
28
|
Tricorache DF, Dascalu AM, Alexandrescu C, Bobirca A, Grigorescu C, Tudor C, Cristea BM. Correlations Between the Neutrophil-Lymphocyte Ratio, Platelet-Lymphocyte Ratio, and Serum Lipid Fractions With Neovascular Age-Related Macular Degeneration. Cureus 2024; 16:e62503. [PMID: 39022525 PMCID: PMC11252630 DOI: 10.7759/cureus.62503] [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] [Accepted: 06/16/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Age-related macular degeneration, a chronic and progressive disease, is one of the leading causes of vision loss globally among the elderly population. Multiple hypotheses have been proposed regarding its pathogenesis, including the presence of lipid metabolism alteration. Dysfunctional lipid handling within retinal pigment epithelial cells has been implicated in the accumulation of lipofuscin and subsequent induction of oxidative stress and inflammation, all contributing to retinal degeneration. The present study aims to comparatively analyze the serum lipid fraction distributions in patients with neovascular age-related macular degeneration (AMD) and controls. Materials and methods A retrospective study was carried out between January 2021 and December 2023 on 91 naïve patients with neovascular AMD and 90 controls admitted for routine cataract surgery. All subjects underwent a comprehensive ophthalmological exam, including ophthalmoscopy and optical coherence tomography (OCT) with central macular thickness (CMT) measurement. A complete blood count with differential and lipid fractions values was analyzed. The neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG) were comparatively analyzed between the control group and the test group. Results The groups were comparable in terms of age (73.84 ±7.52 years for the neovascular AMD group vs 72.1±10.92 years in controls; p=0.8) and gender distribution (p=0.243). The mean NLR and PLR values were slightly higher in the AMD group but not statistically significant (p=0.51, p>0.99, respectively). Comparative analysis of lipid profile fractions showed significantly higher HDL-C values in the exudative AMD group compared to normal subjects (61.27±19.4 mg/dL vs 50.99±7.86 mg/dL, p=0.006). Also, the proportion of subjects with HDL-C>60 mg/dL was higher in the exudative AMD group (p=0.014). There were no significant differences in total cholesterol (189.77±53.39 mg/dL vs 190.43±37.84 mg/dL, p=0.681), LDL-C, and TG. Logistic regression analysis showed that serum HDL-C and HDL-C values >60 mg/dL are significantly associated factors with neovascular AMD. However, there is no statistical correlation between the values of these biochemical parameters and visual acuity or CMT in the neovascular AMD patient group. Conclusions There were no correlations between NLR and PLR with neovascular AMD in the study group. Higher HDL-C values exceeding 60 mg/dL were associated with neovascular age-related macular degeneration and could represent a possible therapeutic target in neovascular AMD.
Collapse
Affiliation(s)
- Diana F Tricorache
- Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Ana M Dascalu
- Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | | | - Anca Bobirca
- Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | | | - Corneliu Tudor
- Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Bogdan M Cristea
- Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
29
|
Zhang L, Zhang T, Chen X, Wang F, Liu L, Yang Y, Zeng Y, Si Y, Yang N. Overexpression of CuZn superoxide dismutase improves high-density lipoprotein function in swine. Prostaglandins Other Lipid Mediat 2024; 172:106817. [PMID: 38331090 DOI: 10.1016/j.prostaglandins.2024.106817] [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: 10/12/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Cardiovascular disease (CVD) has been the leading cause of death worldwide. As a chronic inflammatory disease, atherosclerosis (AS) acts as the initiating factor for CVD and reactive oxygen species (ROS) play a vital role in its development. Superoxide dismutases (SOD) can alleviate the detrimental effects of ROS and serve as the first line of defense through detoxifying the products derived from oxidative stress in vivo. Considering the potential preventive effects of high-density lipoprotein (HDL) on AS and the close relationship between CuZn superoxide dismutase (CuZnSOD) and HDL, the present work investigated whether CuZnSOD overexpression in swine could improve the function of HDL. Seven CuZnSOD transgenic swine, constructed by sperm and magnetic nanoparticles, demonstrated overexpressed CuZnSOD in the liver (P < 0.01) but comparable level to control in plasma (P > 0.05). CuZnSOD overexpression significantly down-regulated the levels of triglyceride (TG), apolipoprotein A-I (apoA-I) (P < 0.05), and high-density lipoprotein cholesterol (HDL-C) (P < 0.01) in plasma. In the presence of CuZnSOD overexpression, HDL3 significantly inhibited levels of IL-6 and TNF-α induced by oxidized low-density lipoprotein (oxLDL) (P < 0.05), indicating enhanced anti-inflammatory activity of HDL. At the same time, HDL-mediated cholesterol efflux did not decrease (P > 0.05). CuZnSOD overexpression improves the anti-inflammatory function of HDL despite decreased levels of HDL-C. In Conclusion, CuZnSOD overexpression improves HDL function in swine.
Collapse
Affiliation(s)
- Lichun Zhang
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Tianliang Zhang
- Experimental Center for Medical Research, Weifang Medical University, Weifang, Shandong, China
| | - Xiaofeng Chen
- Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Fengjiao Wang
- Medical Laboratory Animal Center, School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Li Liu
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Yanmei Yang
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Yongqing Zeng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China.
| | - Yanhong Si
- Basic Medical College, Shandong First Medical University, Taian, Shandong, China.
| | - Nana Yang
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China; Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular Diseases, Weifang, Shandong, China.
| |
Collapse
|
30
|
Chen XF, Xiang YF, Cai XL, Chen JH, Lin XQ, Lin BT, Liang WJ, Zhou GY, Guo YS, Lin KY. A V-shaped association between high-density lipoprotein cholesterol levels and poor outcomes in patients after percutaneous coronary intervention. Int J Cardiol 2024; 400:131773. [PMID: 38211670 DOI: 10.1016/j.ijcard.2024.131773] [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: 09/13/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 01/13/2024]
Abstract
BACKGROUND High density lipoprotein cholesterol (HDL-C) is considered as "good cholesterol". Recent evidence suggests that a high HDL-C level may increase the risk of poor outcomes in some populations. PURPOSE To investigate the association between HDL-C levels and poor outcomes in patients after percutaneous coronary intervention (PCI). METHODS Patients undergoing PCI during January 2012 and December 2018 were consecutively recruited and divided into three groups with different HDL-C levels: HDL-C ≤ 25 mg/dL, 25 < HDL-C ≤ 60 mg/dL, HDL-C > 60 mg/dL by the restricted cubic spline (RCS) analysis and assessed for all-cause mortality (ACM). The association between HDL-C levels and poor outcomes was assessed by multivariable cox regression analysis. RESULTS The patients were followed with a median duration of 4 years. Of the 7284 participants, 727 all-cause deaths and 334 cardiovascular deaths occurred. A V-shaped association of HDL-C with the prognosis was observed, patients with either excessively low or high HDL-C levels reporting a higher risk than those with midrange values. After adjustment for confounding factors, the former exhibited a higher cumulative rate of ACM and cardiovascular mortality (CM) than the latter [low HDL-C: for ACM, hazard ratio (HR), 1.96; 95%CI, 1.41, 2.73, P < 0.001; for CM, HR, 1.66; 95%CI, 1.03, 2.67; P = 0.037; high HDL-C: for ACM, HR, 1.73; 95%CI, 1.29, 2.32, P < 0.001; for CM, HR, 1.73; 95%CI, 1.16, 2.58; P = 0.007]. CONCLUSION HDL-C levels display a V-shaped association with poor outcomes in patients after PCI, with excessively high or low HDL-C suggesting a higher mortality risk. An optimal HDL-C level may fall in the range of 25-60 mg/dL.
Collapse
Affiliation(s)
- Xiao-Fang Chen
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Yi-Fei Xiang
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Xiao-Ling Cai
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Jun-Han Chen
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Xue-Qin Lin
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Bi-Ting Lin
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Wen-Jia Liang
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Geng-Yu Zhou
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China
| | - Yan-Song Guo
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China; Fujian Heart Failure Center Alliance, Fuzhou, China.
| | - Kai-Yang Lin
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China; Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, Fuzhou, China; Fujian Heart Failure Center Alliance, Fuzhou, China.
| |
Collapse
|
31
|
Teng D, Wang W, Jia W, Song J, Gong L, Zhong L, Yang J. The effects of glycosylation modifications on monocyte recruitment and foam cell formation in atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167027. [PMID: 38237743 DOI: 10.1016/j.bbadis.2024.167027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
The monocyte recruitment and foam cell formation have been intensively investigated in atherosclerosis. Nevertheless, as the study progressed, it was obvious that crucial molecules participated in the monocyte recruitment and the membrane proteins in macrophages exhibited substantial glycosylation modifications. These modifications can exert a significant influence on protein functions and may even impact the overall progression of diseases. This article provides a review of the effects of glycosylation modifications on monocyte recruitment and foam cell formation. By elaborating on these effects, we aim to understand the underlying mechanisms of atherogenesis further and to provide new insights into the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Da Teng
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wenlong Wang
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wenjuan Jia
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jikai Song
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Lei Gong
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lin Zhong
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China.
| | - Jun Yang
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
32
|
Liu R, Cheng W. Association between HDL-C and intensive blood pressure control in patients with hypertension: A post-hoc analysis of SPRINT. J Clin Hypertens (Greenwich) 2024; 26:225-234. [PMID: 38318688 PMCID: PMC10918727 DOI: 10.1111/jch.14754] [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: 09/13/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 02/07/2024]
Abstract
Previous studies in patients with hypertension have demonstrated that there is a U-shaped association between HDL-C (high-density lipoprotein cholesterol) and the risk of cardiovascular events in male patients with hypertension. However, to the best of our knowledge, the relationship between HDL-C and intensive blood pressure control in specific cardiovascular events has never been investigated. To fill this knowledge gap, the authors analyzed the relationship between HDL-C levels and cardiovascular events in hypertensive patients within the Systolic Blood Pressure Intervention Trial (SPRINT). The SPRINT evaluated the impact of intensive blood pressure control (systolic blood pressure < 120 mm Hg) versus standard blood pressure control (systolic blood pressure < 140 mm Hg). The Cox proportional risk regression was used to investigate the association between different HDL-C status and clinical outcomes. Additional stratified analyzes were performed to evaluate the robustness of sex difference. A total of 9323 participants (6016 [64.53%] males and 3307 [35.47%] females) with hypertension from the SPRINT research were included in the analysis. The median follow-up period was 3.26 years. Our population was divided into five groups based on the HDL-C plasma levels: HDL-C < 30 mg/dL, HDL-C between 30 and 40 mg/dL, HDL-C between 40 and 60 mg/dL, HDL-C between 60 and 80 mg/dL and HDL-C > 80 mg/dL. Sensitivity analyzes showed that in the SPRINT, women in the HDL-C high population had a higher risk of mortality from all causes than men. In this cohort study, results suggest that patients with HDL-C levels higher than 80 mg/dL had lower risk of SPRINT primary outcome, cardiovascular death, and stroke, but this study tested association, not causation. HDL-C levels were associated with composite cardiovascular outcomes in male but not female patients. Our results demonstrated that in patients with hypertension, the association between HDL-C and risk of cardiovascular events is L-shaped.
Collapse
Affiliation(s)
- Rufei Liu
- Hypertension CenterBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Wenli Cheng
- Hypertension CenterBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
33
|
Borja MS, Hammerson B, Tang C, Juarez-Serrano L, Savinova OV, Harris WS, Oda MN, Shearer GC. Effects of niacin and omega-3 fatty acids on HDL-apolipoprotein A-I exchange in subjects with metabolic syndrome. PLoS One 2024; 19:e0296052. [PMID: 38408107 PMCID: PMC10896500 DOI: 10.1371/journal.pone.0296052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/29/2023] [Indexed: 02/28/2024] Open
Abstract
HDL-apolipoprotein A-I exchange (HAE) measures a functional property associated with HDL's ability to mediate reverse cholesterol transport. HAE has been used to examine HDL function in case-control studies but not in studies of therapeutics that alter HDL particle composition. This study investigates whether niacin and omega-3 fatty acids induce measurable changes in HAE using a cohort of fifty-six subjects with metabolic syndrome (MetS) who were previously recruited to a double-blind trial where they were randomized to 16 weeks of treatment with dual placebo, extended-release niacin (ERN, 2g/day), prescription omega-3 ethyl esters (P-OM3, 4g/day), or the combination. HAE was assessed at the beginning and end of the study. Compared to placebo, ERN and P-OM3 alone significantly increased HAE by 15.1% [8.2, 22.0] (P<0.0001) and 11.1% [4.5, 17.7] (P<0.0005), respectively, while in combination they increased HAE by 10.0% [2.5, 15.8] (P = 0.005). When HAE was evaluated per unit mass of apoA-I ERN increased apoA-I specific exchange activity by 20% (2, 41 CI, P = 0.02) and P-OM3 by 28% (9.6, 48 CI, P<0.0006). However the combination had no statistically significant effect, 10% (-9, 31 CI, P = 0.39). With regard to P-OM3 therapy in particular, the HAE assay detected an increase in this property in the absence of a concomitant rise in HDL-C and apoA-I levels, suggesting that the assay can detect functional changes in HDL that occur in the absence of traditional biomarkers.
Collapse
Affiliation(s)
- Mark S. Borja
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- Department of Chemistry and Biochemistry, California State University East Bay, Hayward, California, United States of America
| | - Bradley Hammerson
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Chongren Tang
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Litzy Juarez-Serrano
- Department of Chemistry and Biochemistry, California State University East Bay, Hayward, California, United States of America
| | - Olga V. Savinova
- Cardiovascular Research Center, Sanford Research, University of South Dakota, Sioux Falls, South Dakota, United States of America
| | - William S. Harris
- Cardiovascular Research Center, Sanford Research, University of South Dakota, Sioux Falls, South Dakota, United States of America
- OmegaQuant, Sioux Falls, South Dakota, United States of America
| | - Michael N. Oda
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Gregory C. Shearer
- Cardiovascular Research Center, Sanford Research, University of South Dakota, Sioux Falls, South Dakota, United States of America
| |
Collapse
|
34
|
Li W, Wang Y, Huang R, Lian F, Xu G, Wang W, Xue S. Rare and common coding variants in lipid metabolism-related genes and their association with coronary artery disease. BMC Cardiovasc Disord 2024; 24:97. [PMID: 38336686 PMCID: PMC10858582 DOI: 10.1186/s12872-024-03759-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is a complex disease that is influenced by environmental and genetic factors. In this study, we aimed to investigate the relationship between coding variants in lipid metabolism-related genes and CAD in a Chinese Han population. METHODS A total of 252 individuals were recruited for this study, including 120 CAD patients and 132 healthy control individuals. Rare and common coding variants in 12 lipid metabolism-related genes (ANGPTL3, ANGPTL4, APOA1, APOA5, APOC1, APOC3, CETP, LDLR, LIPC, LPL, PCSK9 and SCARB1) were detected via next-generation sequencing (NGS)-based targeted sequencing. Associations between common variants and CAD were evaluated by Fisher's exact test. A gene-based association test of rare variants was performed by the sequence kernel association test-optimal (SKAT-O test). RESULTS We found 51 rare variants and 17 common variants in this study. One common missense variant, LIPC rs6083, was significantly associated with CAD after Bonferroni correction (OR = 0.47, 95% CI = 0.29-0.76, p = 1.9 × 10- 3). Thirty-three nonsynonymous rare variants were identified, including two novel variants located in the ANGPTL4 (p.Gly47Glu) and SCARB1 (p.Leu233Phe) genes. We did not find a significant association between rare variants and CAD via gene-based analysis via the SKAT-O test. CONCLUSIONS Targeted sequencing is a powerful tool for identifying rare and common variants in CAD. The common missense variant LIPC rs6083 confers protection against CAD. The clinical relevance of rare variants in CAD aetiology needs to be investigated in larger sample sizes in the future.
Collapse
Affiliation(s)
- Wei Li
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Yongyi Wang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Ritai Huang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Feng Lian
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Genxing Xu
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Weijun Wang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
35
|
Walker ME, De Matteis R, Perretti M, Dalli J. Resolvin T4 enhances macrophage cholesterol efflux to reduce vascular disease. Nat Commun 2024; 15:975. [PMID: 38316794 PMCID: PMC10844649 DOI: 10.1038/s41467-024-44868-1] [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: 06/28/2020] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
While cardiovascular disease (CVD) is one of the major co-morbidities in patients with rheumatoid arthritis (RA), the mechanism(s) that contribute to CVD in patients with RA remain to be fully elucidated. Herein, we observe that plasma concentrations of 13-series resolvin (RvT)4 negatively correlate with vascular lipid load in mouse inflammatory arthritis. Administration of RvT4 to male arthritic mice fed an atherogenic diet significantly reduces atherosclerosis. Assessment of the mechanisms elicited by this mediator demonstrates that RvT4 activates cholesterol efflux in lipid laden macrophages via a Scavenger Receptor class B type 1 (SR-BI)-Neutral Cholesterol Ester Hydrolase-dependent pathway. This leads to the reprogramming of lipid laden macrophages yielding tissue protection. Pharmacological inhibition or knockdown of macrophage SR-BI reverses the vasculo-protective activities of RvT4 in vitro and in male mice in vivo. Together these findings elucidate a RvT4-SR-BI centered mechanism that orchestrates macrophage responses to limit atherosclerosis during inflammatory arthritis.
Collapse
Affiliation(s)
- Mary E Walker
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Roberta De Matteis
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Mauro Perretti
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Jesmond Dalli
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
36
|
Li Y, Xiong Z, Jiang WL, Tian D, Zhou H, Hou Q, Xiao L, Zhang M, Huang L, Zhong L, Zhou L, Zeng GG. An innovative viewpoint on the existing and prospectiveness of SR-B1. Curr Probl Cardiol 2024; 49:102226. [PMID: 38040207 DOI: 10.1016/j.cpcardiol.2023.102226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Scavenger Receptor Class B Type 1 (SR-B1), a receptor protein expressed on the cell membrane, plays a crucial role in the metabolism and transport of cholesterol and other lipids, contributing significantly to the homeostasis of lipid levels within the body. Bibliometric analysis involves the application of mathematical and statistical methods to quantitatively analyze different types of documents. It involves the analysis of structural and temporal trends in scholarly articles, coupled with the identification of subject emphasis and variations. Through a bibliometric analysis, this study examines the historical background, current research trends, and future directions in the exploration of SR-B1. By offering insights into the research status and development of SR-B1, this paper aims to assist researchers in identifying novel pathways and areas of investigation in this field of study. Following the screening process, it can be concluded that research on SR-B1 has consistently remained a topic of significant interest over the past 17 years. Interestingly, SR-B1 has recently garnered attention in areas beyond its traditional research focus, including the field of cancer. The primary objective of this review is to provide a concise and accessible overview of the development process of SR-B1 that can help readers who are not well-versed in SR-B1 research quickly grasp its key aspects. Furthermore, this review aims to offer insights and suggestions to researchers regarding potential future research directions and areas of emphasis relating to SR-B1.
Collapse
Affiliation(s)
- Yonggui Li
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhijie Xiong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wan-Li Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Dandan Tian
- School of Nursing, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Haiyou Zhou
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Qin Hou
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liang Xiao
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Mengjie Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liubin Huang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Lianping Zhong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Zhou
- Department of Pathology, Chongqing Public Health Medical Center, Southwest University Public Health Hospital, Chongqing, China
| | - Guang-Gui Zeng
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
37
|
Perswani P, Ismail SM, Mumtaz H, Uddin N, Asfand M, Khalil ABB, Ijlal A, Khan SE, Usman M, Younas H, Rai A. Rethinking HDL-C: An In-Depth Narrative Review of Its Role in Cardiovascular Health. Curr Probl Cardiol 2024; 49:102152. [PMID: 37852560 DOI: 10.1016/j.cpcardiol.2023.102152] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
The interplay between HDL-C and LDL levels are closely intertwined with the cardiovascular system. High-Density Lipoprotein Cholesterol (HDL-C) is a well-known biomarker traditionally being interpreted as higher the HDL-C levels, minimal the risk of adverse cardiovascular disease (CVD) outcomes. However, recent research has unveiled a more complex relationship between HDL-C levels and cardiovascular outcomes, including genetic influences and potential risks associated with extremely high HDL-C levels. Intriguingly, extremely high HDL-C levels have been linked to unexpected cardiovascular risks. Up To date research suggests that individuals with genetically linked ultra-high HDL-C levels may depict an increased susceptibility to CVD, challenging the conventional realm that higher HDL-C is always beneficial. The mechanisms underlying this mystery are not fully understood but may involve HDL particle functionality and composition. In a nutshell, the relationship between HDL-C levels and cardiovascular outcomes is multifactorial. While low HDL-C remains a recognized risk factor for CVD, the genetic determinants of HDL-C levels add complexity to this association. Furthermore, extremely high HDL-C levels may not exhibit the expected protective benefits and may even pose unprecedented cardiovascular risks. A comprehensive understanding of these dynamics is essential for advancing our knowledge of CVD risk assessment and developing targeted therapeutic interventions. Further studies are needed to unravel the intricacies of HDL-C's role in cardiovascular health and disease.
Collapse
Affiliation(s)
| | | | - Hassan Mumtaz
- Care Coordinator: Association for Social Development, Islamabad, Pakistan; International Practitioner: Faculty of Public Health UK.
| | - Naseer Uddin
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan.
| | | | | | - Aisha Ijlal
- South City Institute of physical therapy and rehabilitation, Karachi.
| | - Shaheer Ellahi Khan
- Associate Professor of Public Health: Health services Academy, Islamabad, Pakistan; Adjunct Professor: Dala Lana School Of Public Health, University of Toronto, Canada.
| | | | - Hadia Younas
- Services institute of medical Sciences, Lahore, Pakistan.
| | - Anushree Rai
- Govt. Chhattisgarh institute of Medical sciences, Bilaspur, Chhattisgarh, India.
| |
Collapse
|
38
|
Yamakawa M, Rexach JE. Cell States and Interactions of CD8 T Cells and Disease-Enriched Microglia in Human Brains with Alzheimer's Disease. Biomedicines 2024; 12:308. [PMID: 38397909 PMCID: PMC10886701 DOI: 10.3390/biomedicines12020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a multi-stage neurodegenerative disorder characterized by beta-amyloid accumulation, hyperphosphorylated Tau deposits, neurodegeneration, neuroinflammation, and cognitive impairment. Recent studies implicate CD8 T cells as neuroimmune responders to the accumulation of AD pathology in the brain and potential contributors to toxic neuroinflammation. However, more evidence is needed to understand lymphocytes in disease, including their functional states, molecular mediators, and interacting cell types in diseased brain tissue. The scarcity of lymphocytes in brain tissue samples has limited the unbiased profiling of disease-associated cell types, cell states, drug targets, and relationships to common AD genetic risk variants based on transcriptomic analyses. However, using recent large-scale, high-quality single-nuclear sequencing datasets from over 84 Alzheimer's disease and control cases, we leverage single-nuclear RNAseq data from 800 lymphocytes collected from 70 individuals to complete unbiased molecular profiling. We demonstrate that effector memory CD8 T cells are the major lymphocyte subclass enriched in the brain tissues of individuals with AD dementia. We define disease-enriched interactions involving CD8 T cells and multiple brain cell subclasses including two distinct microglial disease states that correlate, respectively, to beta-amyloid and tau pathology. We find that beta-amyloid-associated microglia are a major hub of multicellular cross-talk gained in disease, including interactions involving both vulnerable neuronal subtypes and CD8 T cells. We reproduce prior reports that amyloid-response microglia are depleted in APOE4 carriers. Overall, these human-based studies provide additional support for the potential relevance of effector memory CD8 T cells as a lymphocyte population of interest in AD dementia and provide new candidate interacting partners and drug targets for further functional study.
Collapse
Affiliation(s)
| | - Jessica E. Rexach
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| |
Collapse
|
39
|
Wu H, Yang Z, Wang J, Bu Y, Wang Y, Xu K, Li J, Yan C, Liu D, Han Y. Exploring shared therapeutic targets in diabetic cardiomyopathy and diabetic foot ulcers through bioinformatics analysis. Sci Rep 2024; 14:230. [PMID: 38168477 PMCID: PMC10761883 DOI: 10.1038/s41598-023-50954-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Advanced diabetic cardiomyopathy (DCM) patients are often accompanied by severe peripheral artery disease. For patients with DCM combined with diabetic foot ulcer (DFU), there are currently no good therapeutic targets and drugs. Here, we investigated the underlying network of molecular actions associated with the occurrence of these two complications. The datasets were downloaded from the Gene Expression Omnibus (GEO) database. We performed enrichment and protein-protein interaction analyses, and screened for hub genes. Construct transcription factors (TFs) and microRNAs regulatory networks for validated hub genes. Finally, drug prediction and molecular docking verification were performed. We identified 299 common differentially expressed genes (DEGs), many of which were involved in inflammation and lipid metabolism. 6 DEGs were identified as hub genes (PPARG, JUN, SLC2A1, CD4, SCARB1 and SERPINE1). These 6 hub genes were associated with inflammation and immune response. We identified 31 common TFs and 2 key miRNAs closely related to hub genes. Interestingly, our study suggested that fenofibrate, a lipid-lowering medication, holds promise as a potential treatment for DCM combined with DFU due to its stable binding to the identified hub genes. Here, we revealed a network involves a common target for DCM and DFU. Understanding these networks and hub genes is pivotal for advancing our comprehension of the multifaceted complications of diabetes and facilitating the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Hanlin Wu
- Dalian Medical University, Dalian, 116044, Liaoning Province, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Zheming Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Jing Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Yuxin Bu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Yani Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Kai Xu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Jing Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China
| | - Dan Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China.
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang, 110016, Liaoning Province, China.
| |
Collapse
|
40
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
41
|
Hoekstra M, Zhang Z, Lindenburg PW, Van Eck M. Scavenger Receptor BI Deficiency in Mice Is Associated With Plasma Ceramide and Sphingomyelin Accumulation and a Reduced Cholesteryl Ester Fatty Acid Length and Unsaturation Degree. J Lipid Atheroscler 2024; 13:69-79. [PMID: 38299166 PMCID: PMC10825577 DOI: 10.12997/jla.2024.13.1.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 02/02/2024] Open
Abstract
Objective Scavenger receptor class B type I (SR-BI) is primarily known for its role in the selective uptake of cholesteryl esters (CEs) from high-density lipoproteins (HDLs). Here we investigated whether SR-BI deficiency is associated with other potentially relevant changes in the plasma lipidome than the established effect of HDL-cholesterol elevation. Methods Targeted ultra-high-performance liquid chromatography-tandem mass spectrometry was utilized to measure lipid species in plasma from female wild-type and SR-BI knockout mice. Results SR-BI deficiency was associated with a reduction in the average CE fatty acid length (-2%; p<0.001) and degree of CE fatty acid unsaturation (-18%; p<0.001) due to a relative shift from longer, polyunsaturated CE species CE (20:4), CE (20:5), and CE (22:6) towards the mono-unsaturated CE (18:1) species. Sphingomyelin (SM) levels were 64% higher (p<0.001) in SR-BI knockout mice without a parallel change in (lyso)phosphatidylcholine (LPC) concentrations, resulting in an increase in the SM/LPC ratio from 0.102±0.005 to 0.163±0.003 (p<0.001). In addition, lower LPC lengths (-5%; p<0.05) and fatty acid unsaturation degrees (-20%; p<0.01) were detected in SR-BI knockout mice. Furthermore, SR-BI deficiency was associated with a 4.7-fold increase (p<0.001) in total plasma ceramide (Cer) levels, with a marked >9-fold rise (p<0.001) in Cer (d18:1/24:1) concentrations. Conclusion We have shown that SR-BI deficiency in mice not only impacts the CE concentrations, length, and saturation index within the plasma compartment, but is also associated with plasma accumulation of several Cer and SM species that may contribute to the development of specific hematological and metabolic (disease) phenotypes previously detected in SR-BI knockout mice.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| | - Zhengzheng Zhang
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Peter W. Lindenburg
- Research Group Metabolomics, Faculty Science & Technology, University of Applied Sciences Leiden, Hogeschool Leiden, Leiden, The Netherlands
| | - Miranda Van Eck
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| |
Collapse
|
42
|
Price TR, Emfinger CH, Schueler KL, King S, Nicholson R, Beck T, Yandell BS, Summers SA, Holland WL, Krauss RM, Keller MP, Attie AD. Identification of genetic drivers of plasma lipoprotein size in the Diversity Outbred mouse population. J Lipid Res 2023; 64:100471. [PMID: 37944753 PMCID: PMC10750189 DOI: 10.1016/j.jlr.2023.100471] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Despite great progress in understanding lipoprotein physiology, there is still much to be learned about the genetic drivers of lipoprotein abundance, composition, and function. We used ion mobility spectrometry to survey 16 plasma lipoprotein subfractions in 500 Diversity Outbred mice maintained on a Western-style diet. We identified 21 quantitative trait loci (QTL) affecting lipoprotein abundance. To refine the QTL and link them to disease risk in humans, we asked if the human homologs of genes located at each QTL were associated with lipid traits in human genome-wide association studies. Integration of mouse QTL with human genome-wide association studies yielded candidate gene drivers for 18 of the 21 QTL. This approach enabled us to nominate the gene encoding the neutral ceramidase, Asah2, as a novel candidate driver at a QTL on chromosome 19 for large HDL particles (HDL-2b). To experimentally validate Asah2, we surveyed lipoproteins in Asah2-/- mice. Compared to wild-type mice, female Asah2-/- mice showed an increase in several lipoproteins, including HDL. Our results provide insights into the genetic regulation of circulating lipoproteins, as well as mechanisms by which lipoprotein subfractions may affect cardiovascular disease risk in humans.
Collapse
Affiliation(s)
- Tara R Price
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Kathryn L Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Sarah King
- School of Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Rebekah Nicholson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Tim Beck
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Brian S Yandell
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Ronald M Krauss
- School of Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
43
|
Hu S, Zhu Y, Zhao X, Li R, Shao G, Gong D, Hu C, Liu H, Xu K, Liu C, Xu M, Zhao Z, Li T, Hu Z, Shao M, Liu J, Li X, Wu H, Li J, Xu Y. Hepatocytic lipocalin-2 controls HDL metabolism and atherosclerosis via Nedd4-1-SR-BI axis in mice. Dev Cell 2023; 58:2326-2337.e5. [PMID: 37863040 DOI: 10.1016/j.devcel.2023.09.007] [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: 02/28/2023] [Revised: 07/03/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023]
Abstract
High-density lipoprotein (HDL) metabolism is regulated by complex interplay between the scavenger receptor group B type 1 (SR-BI) and multiple signaling molecules in the liver. Here, we show that lipocalin-2 (Lcn2) is a key regulator of hepatic SR-BI, HDL metabolism, and atherosclerosis. Overexpression of human Lcn2 in hepatocytes attenuates the development of atherosclerosis via SR-BI in western-diet-fed Ldlr-/- mice, whereas hepatocyte-specific ablation of Lcn2 has the opposite effect. Mechanistically, hepatocyte Lcn2 improves HDL metabolism and alleviates atherogenesis by blocking Nedd4-1-mediated SR-BI ubiquitination at K500 and K508. The Lcn2-improved HDL metabolism is abolished in mice with hepatocyte-specific Nedd4-1 or SR-BI deletion and in SR-BI (K500A/K508A) mutation mice. This study identifies a regulatory axis from Lcn2 to HDL via blocking Nedd4-1-mediated SR-BI ubiquitination and demonstrates that hepatocyte Lcn2 may be a promising target to improve HDL metabolism to treat atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Shuwei Hu
- School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China; Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yingdong Zhu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Xiaojie Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Rui Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Guangze Shao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Dongxu Gong
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Chencheng Hu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Hongjun Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Kexin Xu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Chenxi Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Minghuan Xu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Tao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital Xizhimen South Street, West District, Beijing 100044, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Mengle Shao
- Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun- Liu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yanyong Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Pathology of School of Basic Medical Sciences, Frontier Innovation Center, Fudan University Shanghai, Shanghai 200032, China; Diabetes, Obesity and Metabolism Research Center, Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| |
Collapse
|
44
|
Sheikhy A, Fallahzadeh A, Sadeghian S, Pashang M, Karimi AA, Bagheri J, Ahmadi‐Tafti H, Hosseini K. A realistic approach to evaluating the effect of baseline lipid profile in postcoronary artery bypass grafting surgery. Clin Cardiol 2023; 46:1408-1417. [PMID: 37594287 PMCID: PMC10642323 DOI: 10.1002/clc.24132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND There are still many uncertainties in the association between lipid profile and postcoronary artery bypass grafting (CABG) outcomes. Although simplifying the association to linear equations makes it understandable but cannot explain many findings. HYPOTHESIS There is a nonlinear associatin between lipid profile indices and adverse outcomes after CABG. METHODS A total of 17 555 patients who underwent isolated CABG between 2005 and 2016 were evaluated. During the median follow-up of 75.24 months, the Restricted Cubic Splines (RCS) estimated from the Cox regression model adjusted for all possible confounders was applied to show a nonlinear relationship of lipid profile contents with the "ln hazard ratio" of mortality and major cerebro-cardiac events (MACCE). RESULTS The relationship between LDL-C and HDL-C with all-cause mortality was nonlinear (nonlinear p were .004 and <.001, respectively). The relationship between remnant cholesterol and all-cause mortality was linear (linearity p = .023). Among men, those in the highest LDL-C level (Q4, LDL-C > 114) and those in the lowest HDL-C level (Q1, HDL-C < 30) showed a significantly higher risk of all-cause mortality compared to other groups (compared with Q3, LDL-C Q4, HR = 1.16, 95% confidence interval [CI]:1.02-1.26, p = .014; HDL-C Q1, HR = 1.14, 95% CI: 1.01-1.31, p = .041). Female patients in the lowest HDL-C level (Q1, HDL-C < 30) showed a significantly higher (compared with Q3, HR = 1.14, 95% CI:1.01-1.31, p = .028) and those in the highest HDL-C level (Q4, HDL-C > 43) showed a significantly lower (compared with Q3, HR = 0.74, 95% CI:0.58-0.98, p = .019) risk of all-cause mortality. CONCLUSION Determining a universal cut off for components of lipid profile may be misleading and should better be revised. Extreme values (very low or very high) for HDL-C and LDL-C have different effects on cardiovascular outcomes.
Collapse
Affiliation(s)
- Ali Sheikhy
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Non‐Communicable Disease Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Aida Fallahzadeh
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Non‐Communicable Disease Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Saeed Sadeghian
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Mina Pashang
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Abbas Ali Karimi
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Jamshid Bagheri
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Hossein Ahmadi‐Tafti
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Kaveh Hosseini
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
45
|
Wang Z, Yelamanchili D, Liu J, Gotto AM, Rosales C, Gillard BK, Pownall HJ. Serum opacity factor normalizes erythrocyte morphology in Scarb1 -/- mice in an HDL-free cholesterol-dependent way. J Lipid Res 2023; 64:100456. [PMID: 37821077 PMCID: PMC10641538 DOI: 10.1016/j.jlr.2023.100456] [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: 08/09/2023] [Revised: 09/16/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023] Open
Abstract
Compared with WT mice, HDL receptor-deficient (Scarb1-/-) mice have higher plasma levels of free cholesterol (FC)-rich HDL and exhibit multiple pathologies associated with a high mol% FC in ovaries, platelets, and erythrocytes, which are reversed by lowering HDL. Bacterial serum opacity factor (SOF) catalyzes the opacification of plasma by targeting and quantitatively converting HDL to neo HDL (HDL remnant), a cholesterol ester-rich microemulsion, and lipid-free APOA1. SOF delivery with an adeno-associated virus (AAVSOF) constitutively lowers plasma HDL-FC and reverses female infertility in Scarb1-/- mice in an HDL-dependent way. We tested whether AAVSOF delivery to Scarb1-/- mice will normalize erythrocyte morphology in an HDL-FC-dependent way. We determined erythrocyte morphology and FC content (mol%) in three groups-WT, untreated Scarb1-/- (control), and Scarb1-/- mice receiving AAVSOF-and correlated these with their respective HDL-mol% FC. Plasma-, HDL-, and tissue-lipid compositions were also determined. Plasma- and HDL-mol% FC positively correlated across all groups. Among Scarb1-/- mice, AAVSOF treatment normalized reticulocyte number, erythrocyte morphology, and erythrocyte-mol% FC. Erythrocyte-mol% FC positively correlated with HDL-mol% FC and with both the number of reticulocytes and abnormal erythrocytes. AAVSOF treatment also reduced FC of extravascular tissues to a lesser extent. HDL-FC spontaneously transfers from plasma HDL to cell membranes. AAVSOF treatment lowers erythrocyte-FC and normalizes erythrocyte morphology and lipid composition by reducing HDL-mol% FC.
Collapse
Affiliation(s)
- Ziyi Wang
- Center for Bioenergetics, Houston Methodist, Houston, TX, USA; Departments of Endocrinology and Xiangya Hospital, Central South University, Changsha, China
| | | | - Jing Liu
- Center for Bioenergetics, Houston Methodist, Houston, TX, USA; Departments of Endocrinology and Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Antonio M Gotto
- Center for Bioenergetics, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Corina Rosales
- Center for Bioenergetics, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Baiba K Gillard
- Center for Bioenergetics, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
46
|
Preechasuk L, Luksameejaroenchai C, Tangjittipokin W, Kunavisarut T. Short-term effects of allulose consumption on glucose homeostasis, metabolic parameters, incretin levels, and inflammatory markers in patients with type 2 diabetes: a double-blind, randomized, controlled crossover clinical trial. Eur J Nutr 2023; 62:2939-2948. [PMID: 37432472 DOI: 10.1007/s00394-023-03205-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
PURPOSE Allulose is a rare monosaccharide with almost zero calories. There is no study of short-term allulose consumption in patients with type 2 diabetes (T2D). Thus, we aimed to study the effect of allulose consumption for 12 weeks on glucose homeostasis, lipid profile, body composition, incretin levels, and inflammatory markers in patients with T2D. METHODS A double-blind, randomized, controlled crossover study was conducted on sixteen patients with T2D. Patients were randomly assigned to allulose 7 g twice daily or aspartame 0.03 g twice daily for 12 weeks. After a 2-week washout, patients were crossed over to the other sweetener for an additional 12 weeks. Oral glucose tolerance tests, laboratory measurements, and dual-energy X-ray absorptiometry were conducted before and after each phase. RESULTS This study revealed that short-term allulose consumption exerted no significant effect on glucose homeostasis, incretin levels, or body composition but significantly increased MCP-1 levels (259 ± 101 pg/ml at baseline vs. 297 ± 108 pg/mL after 12 weeks of allulose, p = 0.002). High-density lipoprotein cholesterol (HDL-C) significantly decreased from 51 ± 13 mg/dl at baseline to 41 ± 12 mg/dL after 12 weeks of allulose, p < 0.001. CONCLUSION Twelve weeks of allulose consumption had a neutral effect on glucose homeostasis, body composition, and incretin levels. Additionally, it decreased HDL-C levels and increased MCP-1 levels. TRIAL REGISTRATION This trial was retrospectively registered on the Thai Clinical Trials Registry (TCTR20220516006) on December 5, 2022.
Collapse
Affiliation(s)
- Lukana Preechasuk
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanoknan Luksameejaroenchai
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Watip Tangjittipokin
- Siriraj Center of Research Excellence for Diabetes, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tada Kunavisarut
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand.
| |
Collapse
|
47
|
Song B, Wang K, Lu W, Zhao X, Yao T, Liu T, Gao G, Fan H, Liu C. A U-shaped association between the triglyceride to high-density lipoprotein cholesterol ratio and the risk of incident type 2 diabetes mellitus in Japanese men with normal glycemic levels: a population-based longitudinal cohort study. Front Endocrinol (Lausanne) 2023; 14:1180910. [PMID: 37810876 PMCID: PMC10552638 DOI: 10.3389/fendo.2023.1180910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Background Several studies have verified that a high baseline TG/HDL-C ratio is a risk factor for incident type 2 diabetes mellitus (T2DM). However, for low baseline TG/HDL-C levels, the findings were inconsistent with ours. In addition, the association between baseline TG/HDL-C ratio and the risk of incident T2DM in Japanese men with normal glycemic levels is unclear. As a result, our study further investigated the relationship between baseline TG/HDL-C and the risk of incident T2DM in Japanese men with normal glycemic levels. Methods This was a secondary longitudinal cohort study. We selected 7,684 male participants between 2004 and 2015 from the NAGALA database. A standardized Cox regression model and two piecewise Cox regression models were used to explore the relationship between the baseline high-density lipoprotein cholesterol ratio (TG/HDL-C) and incident T2DM. Results During a median follow-up of 2,282 days, 162 men developed incident T2DM. In the adjusted model, the baseline TG/HDL-C ratio was strongly associated with the risk of incident T2DM, and no dose-dependent positive association was observed between the baseline TG/HDL-C ratio and incidence of T2DM throughout the baseline TG/HDL-C quartiles. Two-piecewise linear regression analysis showed a U-shaped association between baseline TG/HDL-C ratio and incidence of incident T2DM. A baseline TG/HDL-C ratio below 1.188 was negatively associated with incident T2DM (H.R. = 0.105, 95% CI = 0.025, 0.451; P = 0.002). In contrast, a baseline TG/HDL-C ratio >1.188 was positively associated with incident T2DM (H.R. = 1.248, 95% CI = 1.113, 1.399; P<0.001). The best TG/HDL-C threshold for predicting incident T2DM was 1.8115 (area under the curve, 0.6837). Conclusion A U-shaped relationship between baseline TG/HDL-C ratio and incident T2DM in Japanese men with normal glycemic levels was found.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chengyun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
48
|
Graham A. Modulation of the Cellular microRNA Landscape: Contribution to the Protective Effects of High-Density Lipoproteins (HDL). BIOLOGY 2023; 12:1232. [PMID: 37759631 PMCID: PMC10526091 DOI: 10.3390/biology12091232] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
High-density lipoproteins (HDL) play an established role in protecting against cellular dysfunction in a variety of different disease contexts; however, harnessing this therapeutic potential has proved challenging due to the heterogeneous and relative instability of this lipoprotein and its variable cargo molecules. The purpose of this study is to examine the contribution of microRNA (miRNA; miR) sequences, either delivered directly or modulated endogenously, to these protective functions. This narrative review introduces the complex cargo carried by HDL, the protective functions associated with this lipoprotein, and the factors governing biogenesis, export and the uptake of microRNA. The possible mechanisms by which HDL can modulate the cellular miRNA landscape are considered, and the impact of key sequences modified by HDL is explored in diseases such as inflammation and immunity, wound healing, angiogenesis, dyslipidaemia, atherosclerosis and coronary heart disease, potentially offering new routes for therapeutic intervention.
Collapse
Affiliation(s)
- Annette Graham
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Cowcaddens Road, Glasgow G4 0BA, UK
| |
Collapse
|
49
|
Hoekstra M, Van Eck M. High-density lipoproteins and non-alcoholic fatty liver disease. ATHEROSCLEROSIS PLUS 2023; 53:33-41. [PMID: 37663008 PMCID: PMC10469384 DOI: 10.1016/j.athplu.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023]
Abstract
Background and aims Non-alcoholic fatty liver disease (NAFLD), a high incidence liver pathology, is associated with a ∼1.5-fold higher cardiovascular disease risk. This phenomenon is generally attributed to the NAFLD-associated increase in circulating levels of pro-atherogenic apolipoprotein B100-containing small dense low-density lipoprotein and plasma hypertriglyceridemia. However, also a significant reduction in cholesterol transported by anti-atherogenic high-density lipoproteins (HDL) is frequently observed in subjects suffering from NAFLD as compared to unaffected people. In this review, we summarize data regarding the relationship between NAFLD and plasma HDL-cholesterol levels, with a special focus on highlighting potential causality between the NAFLD pathology and changes in HDL metabolism. Methods and results Publications in PUBMED describing the relationship between HDL levels and NAFLD susceptibility and/or disease severity, either in human clinical settings or genetically-modified mouse models, were critically reviewed for subsequent inclusion in this manuscript. Furthermore, relevant literature describing effects on lipid loading in cultured hepatocytes of models with genetic alterations related to HDL metabolism have been summarized. Conclusions Although in vitro observations suggest causality between HDL formation by hepatocytes and protection against NAFLD-like lipid accumulation, current literature remains inconclusive on whether relative HDL deficiency is actually driving the development of fatty liver disease in humans. In light of the current obesity pandemic and the associated marked rise in NAFLD incidence, it is of clear scientific and societal interest to gain further insight into the relationship between HDL-cholesterol levels and fatty liver development to potentially uncover the therapeutic potential of pharmacological HDL level and/or function modulation.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
- Pharmacy Leiden, Leiden, the Netherlands
| | - Miranda Van Eck
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
- Pharmacy Leiden, Leiden, the Netherlands
| |
Collapse
|
50
|
Chen W, Bao L, Ren Q, Zhang Z, Yi L, Lei W, Yang Z, Lu Y, You B, You Y, Gu M. SCARB1 in extracellular vesicles promotes NPC metastasis by co-regulating M1 and M2 macrophage function. Cell Death Discov 2023; 9:323. [PMID: 37644041 PMCID: PMC10465564 DOI: 10.1038/s41420-023-01621-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 07/30/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Distant metastasis is currently the main factor affecting the prognosis of nasopharyngeal carcinoma (NPC), and understanding the mechanisms of metastasis and identifying reliable therapeutic targets are critical for improving prognosis and achieving clinical translation. Macrophages, as important immune cells in the tumor microenvironment (TME), have been shown to regulate metastasis. And extracellular vesicles (EVs) secreted by stromal cells and tumor cells play the important role in intercellular communication in the tumor microenvironment. However, the role of NPC-EVs on macrophages and their function in regulating macrophages to affect metastasis has not been fully clarified. In this study, we report that NPC-EVs can be uptake by macrophages and alter macrophage polarization, for the first time, we identified the genes implicated in these regulatory functions: SCARB1, HAAO, and CYP1B1. Moreover, we found that SCARB1 was positively associated with metastasis and poor prognosis of NPC. Interestingly, we found that SCARB1-rich EVs promoted M1 macrophages ferroptosis to decrease M1 macrophages infiltration by upregulating the HAAO level while decreasing phagocytosis of M2 macrophages by upregulating the CYP1B1 level. Finally, we identified the SCARB1-binding gene KLF9, which is involved in the transcription of HAAO and CYP1B1. Our findings showed that SCARB1-EVs promoted metastasis by co-regulating M1 and M2 macrophage function. The related mechanism will provide a new therapeutic strategy to help patients with NPC improve their prognosis.
Collapse
Affiliation(s)
- Wenhui Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Lili Bao
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Qianqian Ren
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zixiang Zhang
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Lu Yi
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wei Lei
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhiyuan Yang
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yingna Lu
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Bo You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| | - Miao Gu
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|