1
|
Liu Y, Deng Y, Du Z, Zhang S, Chen L, Yan X, Pei Y. ADAMTS- 1 rs402007 Polymorphism Modulates Carotid Plaque Vulnerability and Atorvastatin Efficacy in Cerebral Infarction Patients. Transl Stroke Res 2025:10.1007/s12975-025-01350-4. [PMID: 40299202 DOI: 10.1007/s12975-025-01350-4] [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: 02/24/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
To investigate the association between rs402007 polymorphism in the ADAMTS-1 gene and carotid atherosclerotic plaque vulnerability, as well as the lipid-lowering efficacy of atorvastatin in cerebral infarction patients. Clinical data from 684 cerebral infarction patients admitted to The First Hospital of Hebei Medical University (2016-2019) were analyzed. Patients were stratified into stable plaque (n = 338) and vulnerable plaque (n = 346) groups based on carotid ultrasound. General information, biochemical markers, rs402007 (G/C) genotypes (dominant model), and allele frequencies were compared. Polymorphism genotyping was performed using TaqMan SNP assays (Applied Biosystems) on an ABI 7500 Fast Real-Time PCR system. Logistic regression evaluated plaque vulnerability risk factors and gene-risk factor interactions. Atorvastatin's lipid-lowering efficacy was compared across genotypes. Diabetes prevalence, LDL-C, TC, HCY, and FIB levels differed significantly between groups (P < 0.05). Genotypic distribution analysis revealed a higher frequency of the GG genotype in the stable plaque group (29.59% vs. 21.68%, χ2 = 5.618, P = 0.018). Diabetes, LDL-C, HCY, and FIB were independent risk factors for plaque vulnerability (P < 0.05). A significant interaction between rs402007 polymorphism and LDL-C was observed (P < 0.05). Atorvastatin efficacy rates were 82.29% (GG), 84.27% (GC), and 89.27% (CC), with significant post-treatment lipid improvements in all genotypes (P < 0.05). The CC genotype exhibited superior efficacy compared to GG (P < 0.05). The rs402007 polymorphism influences carotid plaque vulnerability and modulates atorvastatin efficacy, underscoring its potential role in genotype-guided therapeutic strategies.
Collapse
Affiliation(s)
- Yongjian Liu
- Health Management Center, The First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang, 050000, Hebei Province, China
| | - Yongmin Deng
- Pediatric Department, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Zhixing Du
- Health Management Center, The First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang, 050000, Hebei Province, China
| | - Shuowen Zhang
- Health Management Center, The First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang, 050000, Hebei Province, China
| | - Litao Chen
- Health Management Center, The First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang, 050000, Hebei Province, China
| | - Xiaojing Yan
- Health Management Center, The First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Yongbin Pei
- Health Management Center, The First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
2
|
Kawabe K, Izumi K, Fukasawa N, Takashina M, Taguchi M, Koike H, Sahashi Y, Ooba N. Association between statin use and cataract formation in a retrospective cohort study using Japanese health screening and claims data. Sci Rep 2025; 15:13594. [PMID: 40253569 PMCID: PMC12009338 DOI: 10.1038/s41598-025-97889-1] [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: 03/12/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
In this retrospective cohort study of data (recorded between 1 January 2005 and 31 December 2017) from a commercially available health screening and insurance claims database of the working-age Japanese population, we examined the association of statin use with cataract formation. Using the health screening data, we identified 1,178,560 patients who met the dyslipidaemia criteria; among them, 724,200 patients were enrolled. Based on person-years, the cohort was categorised by statin non-use and new use. Unadjusted, age-sex-adjusted, and multivariate-adjusted hazard ratios (hazard ratios [HRs]; with their 95% confidence intervals [CIs]) were estimated, and intergroup comparisons were undertaken using Cox proportional hazards regression. An increased risk of cataract incidence was associated with statin use (adjusted HR [95% CI]) compared with statin non-use (1.56 [1.43-1.70]). The adjusted HRs [95% CI] for cataract incidence for low- and high-potency statins were 1.48 [1.30-1.70] and 1.61 [1.44-1.79], respectively, whereas those for lipophilic and hydrophilic statins were 1.56 [1.39-1.75] and 1.56 [1.38-1.75], respectively. The adjusted HR for statin use with incidence of cataract was 1.35-1.73, except for fluvastatin and simvastatin. In the middle-aged Japanese working population, statin use was associated with a 1.5- to 1.6-fold higher risk of cataracts.
Collapse
Affiliation(s)
- Kazuhiro Kawabe
- Department of Pharmacoepidemiology, Nihon University School of Pharmacy, Chiba, Japan
- Department of Pharmacy, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | - Kanako Izumi
- Department of Pharmacoepidemiology, Nihon University School of Pharmacy, Chiba, Japan
| | - Naoko Fukasawa
- Department of Pharmacoepidemiology, Nihon University School of Pharmacy, Chiba, Japan
| | - Momoko Takashina
- Department of Pharmacoepidemiology, Nihon University School of Pharmacy, Chiba, Japan
| | - Minami Taguchi
- Department of Pharmacoepidemiology, Nihon University School of Pharmacy, Chiba, Japan
| | - Hirofumi Koike
- Department of Pharmacy, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | - Yukiko Sahashi
- Department of Pharmacy, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | - Nobuhiro Ooba
- Department of Pharmacoepidemiology, Nihon University School of Pharmacy, Chiba, Japan.
| |
Collapse
|
3
|
Jiang B, Wang Z, Wang M, Wang S, Li M, Meng Z, Yuan J, Ke Y. Safety Assessment of Two Human Fecal Bacteroides Strain Isolates in Immunodeficient Mice. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10529-y. [PMID: 40167961 DOI: 10.1007/s12602-025-10529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Bacteroides are potential candidates for next-generation probiotics (NGPs), which require preclinical safety and efficacy evaluations to ensure their rational use. This study aimed to verify the safety of two Bacteroides strains isolated from human fecal samples, Bacteroides dorei CK16 (B. dorei CK16) and Bacteroides vulgatus CK29 (B. vulgatus CK29), using genomic analysis and in vivo experiments. Whole-genome sequencing analysis of B. dorei CK16 revealed a predicted 4,898 protein-coding sequences (CDS), about 5.5 Mb of total genome length with a G + C content of 42.08%, and B. vulgatus CK29 revealed a predicted 4,610 CDS, about 5.3 Mb of total genome length with a G + C content of 42.56%. Moreover, the genome demonstrated the absence of virulence factors, and insertion sequences related to clinically relevant strains in either strain. A 42-day in vivo experiment was conducted on BALB/c and BALB/c nude mice, with each mouse receiving a daily dose of 1 × 108 colony forming units (CFU) /mL of B. dorei CK16 or B. vulgatus CK29. No significant in vivo pathogenic characteristics were observed based on body weight, organ index, hematological, serum biochemical, or histological analyses, particularly in nude mice. Therefore, the initial safety assessment of the two novel Bacteroides strains exhibited no notable adverse effects in both immunocompetent and immunodeficient mice models.
Collapse
Affiliation(s)
- Boyi Jiang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China
| | - Zhen Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Mingxuan Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, 050091, Hebei Province, China
| | - Shijie Wang
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, 050091, Hebei Province, China
| | - Mengmeng Li
- Department of Anesthesiology, Fourth Center of Chinese PLA General Hospital, Beijing, 100143, China.
| | - Zhaoting Meng
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China.
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China.
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China.
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China.
| |
Collapse
|
4
|
Wang T, Yao Y, Gao X, Luan H, Wang X, Liu L, Sun C. Genetic association of lipids and lipid-lowering drug target genes with breast cancer. Discov Oncol 2025; 16:331. [PMID: 40095250 PMCID: PMC11914663 DOI: 10.1007/s12672-025-02041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Although several preclinical and epidemiological studies have shown that blood lipids and lipid-lowering drugs can reduce the risk of breast cancer, this finding remains controversial. This study aimed to explore the causal relationship between dyslipidemia,lipid-lowering drugs, and breast cancer. We also aimed to evaluate the potential impact of lipid-lowering drug targets on breast cancer. METHOD Data of 431 lipid- and lipid-related phenotypes were obtained from genome-wide association study (GWAS), and mendelian randomization (MR) analyses were performed using two independent breast cancer datasets as endpoints. Genetic variants associated with genes encoding lipid-lowering drug targets were extracted from the Global Lipid Genetics Consortium. Expression quantitative trait loci data in relevant tissues were used to further validate lipid-lowering drug targets that reached significance and combined with bioinformatics approaches for molecular expression and prognostic exploration. Further mediation analyses were performed to explore potential mediators. RESULT In two independent datasets, phosphatidylcholine (18:1_0:0 levels) was associated with breast cancer risk (discovery: odds ratio (OR) = 1.255 [95% confidence interval (CI) 1.120-1.406]; p = 8.936 × 10-5, replication: OR = 1.016 [95% CI, 1.003-1.030]; p = 0.017), HMG- CoA reductase (HMGCR) inhibition was genetically modeled and associated with a reduced risk of breast cancer (discovery: OR = 0.833 [95% CI 0.752-0.923], p = 5.12 × 10-4; replication: OR = 0.975 [95% CI 0.960-0.990], p = 1.65 × 10-3). There was a significant MR correlation between HMGCR expression in whole blood and breast cancer (OR = 1.11 [95% 1.01-1.22] p = 0.04). Bioinformatics analysis revealed that HMGCR expression higher in breast cancer tissues than in normal tissues, along with poor overall survival and relapse-free survival, and was associated with multiple immune cell infiltration. Finally, the mediation analysis showed that HMGCR inhibitors affected breast cancer through different immune cell phenotypes and C-reactive protein levels. CONCLUSION In this study, we found for the first time that phosphatidylcholine (18:1_0:0) levels are associated with breast cancer risk. We found that HMGCR inhibitors are associated with a reduced risk of breast cancer, and part of their action may be through pathways other than lipid-lowering, including modulation of immune function and reduction of inflammation represented by C-reactive protein levels.
Collapse
Affiliation(s)
- Tianhua Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Xinhai Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Luan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - Lijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
5
|
Sun J, Liu HR, Zhu YX, Zhang W, Shi JS, Wu Q, Xu RX. Dendrobium nobile Lindl. alkaloids improve lipid metabolism by increasing LDL uptake through regulation of the LXRα/IDOL/LDLR pathway and inhibition of PCSK9 expression in HepG2 cells. Exp Ther Med 2025; 29:46. [PMID: 39885913 PMCID: PMC11775753 DOI: 10.3892/etm.2025.12796] [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: 10/16/2023] [Accepted: 10/11/2024] [Indexed: 02/01/2025] Open
Abstract
Dendrobium nobile Lindl. alkaloids (DNLA) are active ingredients that can be extracted from the traditional Chinese herb Dendrobium Nobile Lindl. DNLA exhibits hypoglycemic and antihyperlipidemia effects. However, to the best of our knowledge, the specific molecular mechanism by which DNLA can regulate lipid metabolism remains unclear. The aim of the present study was to investigate the effect of DNLA on lipopolysaccharide (LPS)-induced lipid metabolism in HepG2 cells and its potential mechanism. HepG2 cells were treated with LPS with or without different concentrations of DNLA (0, 0.035, 0.35 and 3.5 µg/ml) for 48 h. Cell viability was then detected using the Cell Counting Kit-8 assay. The 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanideperchlorate-low-density lipoprotein (LDL) uptake assay was used to examine LDL uptake. In addition, possible mechanisms were explored using western blot analysis. The effect of the combination of DNLA with rosuvastatin calcium on the expression levels of the LDL receptor (LDLR) and proprotein convertase subtilisin/Kexin type 9 (PCSK9) was examined. The results indicated that LPS stimulation reduced the uptake of LDL by HepG2 cells, decreased the intracellular LDLR content, and increased the expression levels of inducible degrader of the LDLR (IDOL) and liver X receptor (LXR)α. DNLA intervention reversed all of the aforementioned LPS-induced effects in HepG2 cells. Additional mechanistic experiments revealed that DNLA exerted its effects mainly by regulating the LXRα/IDOL/LDLR pathway. It was shown that DNLA also reduced the expression levels of PCSK9, sterol regulatory element binding protein 2 and hepatocyte nuclear factor 1α. In addition, DNLA decreased the expression levels of PCSK9 in rosuvastatin calcium-induced HepG2 cells. Notably, DNLA was able to decrease 3-hydroxy-3-methylglutaryl-coenzyme A reductase and increase cytochrome p450 7A1 expression at the protein level, which are rate-limiting enzymes in cholesterol synthesis and metabolism. Collectively, these data suggested that DNLA could enhance LDL uptake of HepG2 cells by increasing LDLR expression through the LXRα/IDOL/LDLR pathway to alleviate the effects induced by LPS, suggesting the potential benefit of DNLA in improving lipid metabolism disorders.
Collapse
Affiliation(s)
- Jian Sun
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, P.R. China
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, P.R. China
| | - Hao-Rui Liu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, P.R. China
| | - Ya-Xin Zhu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, P.R. China
| | - Wei Zhang
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, P.R. China
| | - Jing-Shan Shi
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, P.R. China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, P.R. China
| | - Rui-Xia Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, P.R. China
| |
Collapse
|
6
|
Han J, Kim Y, Kang HJ, Seo J, Choi H, Kim M, Kee G, Park S, Ko S, Jung H, Kim B, Jun TJ, Kim YH. Predicting low density lipoprotein cholesterol target attainment using machine learning in patients with coronary artery disease receiving moderate-dose statin therapy. Sci Rep 2025; 15:5346. [PMID: 39948422 PMCID: PMC11825908 DOI: 10.1038/s41598-025-88693-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) is an important factor in the development of cardiovascular disease, making its management a key aspect of cardiovascular health. While high-dose statin therapy is often recommended for LDL-C reduction, careful consideration is needed due to patient-specific factors and potential side effects. This study aimed to develop a machine learning (ML) model to estimate the likelihood of achieving target LDL-C levels in patients hospitalized for coronary artery disease and treated with moderate-dose statins. The predictive performance of three ML models, including Extreme Gradient Boosting (XGBoost), Random Forest, and Logistic Regression, was evaluated using electronic medical records from the Asan Medical Center in Seoul across six performance metrics. Additionally, all three models achieved an average AUROC of 0.695 despite reducing features by over 43%. SHAP analysis was conducted to identify key features influencing model predictions, aiming insights into patient characteristics associated with achieving LDL-C targets. This study suggests that ML-based approaches may help identify patients likely to benefit from moderate-dose statins, potentially supporting personalized treatment strategies and clinical decision-making for LDL-C management.
Collapse
Grants
- 1711195603 Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety)
- 1711195603 Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety)
- 1711195603 Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety)
- 1711195603 Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety)
- 1711195603 Korea government (the Ministry of Science and ICT, the Ministry of Trade, Industry and Energy, the Ministry of Health & Welfare, the Ministry of Food and Drug Safety)
- HR20C0026 Korea Health Industry Development Institute (KHIDI)
- HR20C0026 Korea Health Industry Development Institute (KHIDI)
- HR20C0026 Korea Health Industry Development Institute (KHIDI)
- HR20C0026 Korea Health Industry Development Institute (KHIDI)
- HR20C0026 Korea Health Industry Development Institute (KHIDI)
- HR20C0026 Korea Health Industry Development Institute (KHIDI)
Collapse
Affiliation(s)
- Jiye Han
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yunha Kim
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hee Jun Kang
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jiahn Seo
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Heejung Choi
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Minkyoung Kim
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Gaeun Kee
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Seohyun Park
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soyoung Ko
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - HyoJe Jung
- Department of Information Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Byeolhee Kim
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Tae Joon Jun
- Big Data Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic- ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Young-Hak Kim
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| |
Collapse
|
7
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 DOI: 10.1161/jaha.124.038857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-Communicable Disease Management Beijing Children's Hospital, Capital Medical University, National Center for Children's Health Beijing China
| | - Qin Liu
- Department of Ultrasound Children's Hospital of the Capital Institute of Pediatrics Beijing China
| | - Hong Cheng
- Department of Epidemiology Capital Institute of Pediatrics Beijing China
| | - Xiaoyuan Zhao
- Department of Epidemiology Capital Institute of Pediatrics Beijing China
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control Department Shenzhen Center for Chronic Disease Control Shenzhen China
| | - Jie Mi
- Center for Non-Communicable Disease Management Beijing Children's Hospital, Capital Medical University, National Center for Children's Health Beijing China
- Key Laboratory of Major Diseases in Children, Ministry of Education China
| |
Collapse
|
8
|
Shao H, Xu C, Zhang C, Li L, Wu P, Chen Z, Guan R. Genetic Insights Into Lipid Traits and Lipid-Modifying Drug Targets in Pregnancy Complications: A Two-Sample Mendelian Randomization Study. Int J Womens Health 2025; 17:221-234. [PMID: 39911358 PMCID: PMC11794394 DOI: 10.2147/ijwh.s496268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Background Dyslipidemia is linked to pregnancy complications, but its causal role remains uncertain. This two-sample Mendelian Randomization (MR) study investigated the causal relationship between lipid traits and pregnancy complications and evaluated the impact of lipid-modifying drug targets. Methods Genetic instruments for lipid traits and targets for lipid-modifying drugs were obtained from the Global Lipids Genetics Consortium. Three pregnancy complications' summary statistics came from the FinnGen R9 database. Significant drug targets underwent further analysis using Expression Quantitative Trait Loci data, and mediation analysis identified potential mediators. Results Increased high-density lipoprotein cholesterol (HDL-C) reduced the incidence of preeclampsia (OR: 0.755, 95% CI: 0.639-0.891, p=0.001, FDR=0.012) and gestational diabetes mellitus (GDM) (OR: 0.835, 95% CI: 0.741-0.942, p=0.003, FDR=0.018). Genetic proxies for cholesteryl ester transfer protein (CETP) inhibition correlated with a decreased risk of preeclampsia (OR: 0.863, 95% CI: 0.786-0.947, p=0.002, FDR=0.027), while genetic inhibition of HMG-CoA reductase (HMGCR) increased preeclampsia risk (OR: 1.700, 95% CI: 1.189-2.431, p=0.004, FDR=0.036). Genetically mimicking the enhancement of lipoprotein lipase (LPL) related to a reduced risk of GDM (OR: 0.681, 95% CI: 0.560-0.829, p=1.29×10-4, FDR=0.004). Higher LPL expression in subcutaneous adipose tissue also reduced GDM risk (OR: 0.642, 95% CI: 0.454-0.909, p=0.013). Waist circumference (4.2%) and waist-to-hip ratio adjusted by BMI (5.7%) partially mediated LPL's effect on GDM risk. Conclusion Elevated HDL-C levels help prevent preeclampsia and GDM. CETP and LPL could be therapeutic targets for preeclampsia and GDM, respectively. However, caution is advised with HMGCR-targeting drugs, as they may increase the preeclampsia risk.
Collapse
Affiliation(s)
- Huijing Shao
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, People’s Republic of China
| | - Chang Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Caihong Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Lirong Li
- Department of Traditional Chinese Gynecology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Pengfei Wu
- Department of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People’s Republic of China
| | - Zixi Chen
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Rui Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
9
|
Yaqoob MU, Qi Y, Hou J, Zhe L, Zhu X, Wu P, Li Z, Wang M, Li Y, Yue M. Coated cysteamine and choline chloride could be potential feed additives to mitigate the harmful effects of fatty liver hemorrhagic syndrome in laying hens caused by high-energy low-protein diet. Poult Sci 2024; 103:104296. [PMID: 39305615 PMCID: PMC11437759 DOI: 10.1016/j.psj.2024.104296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 10/01/2024] Open
Abstract
The research aimed to examine the impact of coated cysteamine (CS) and choline chloride (CC) on relieving the pathological effects of fatty liver hemorrhagic syndrome (FLHS) in laying hens. FLHS was induced by a high-energy low-protein (HELP) diet. Ninety laying hens were equally divided into 5 treatments with 6 replicates per treatment (3 hens/replicate). The control treatment (Cont) was fed a basal diet, while the remaining treatments were fed a HELP diet. Under the HELP dietary plan, 4 treatments were set by a 2 × 2 factorial design. Two levels of CS (CS-: 0.00 mg/kg CS; CS+: 100 mg/kg diet) and 2 levels of choline (CC-: 1,182 mg/kg; CC+: 4,124 mg/kg) were set and named CS-CC- (HELP), CS+CC-, CS-CC+ and CS+CC+. The liver of the CS-CC- (HELP) group became yellowish-brown and greasy, with hemorrhages and bleeding spots. Elevated (P < 0.05) plasma and hepatic ALT and AST and hepatic MDA levels, combined with reduced (P < 0.05) plasma and hepatic SOD and GSH-Px activities in the CS-CC- (HELP) group proved that FLHS was successfully induced. Dietary supplementation of CS, CC, or both (CS+CC+) in HELP diets relieved the pathological changes, significantly (P < 0.05) reduced the AST and ALT levels, and strengthened the antioxidant potential in laying hens under FLHS. The highest (P < 0.001) plasma adiponectin concentration was observed in the CS+CC- and lowest in the CS-CC- (HELP) group. In addition, CS and CC supplementation lowers the elevated levels of hepatic T-CHO and TG by increasing the HDL-C and reducing LDL-C levels (P < 0.05) than CS-CC- (HELP) group. CS supplementation, either alone or with CC, helps laying hens restore their egg production. It could be stated that CS and CC supplements could ameliorate the adverse effects of FLHS by regulating antioxidant enzymes activities, modulating the hepatic lipid metabolism, and restoring the production performance in laying hens. Hence, adding CS and CC could be an effective way to reduce FLHS in laying hens.
Collapse
Affiliation(s)
- Muhammad Umar Yaqoob
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Yingying Qi
- Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Jia Hou
- Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Li Zhe
- Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Xiangde Zhu
- Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Peng Wu
- Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Zhefeng Li
- Provincial Key Agricultural Enterprise Research Institute of King Techina, Hangzhou King Techina Feed Co., Ltd. Zhejiang Hangzhou 311107, China
| | - Minqi Wang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Yan Li
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Min Yue
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
10
|
Noordam R, Wang W, Nagarajan P, Wang H, Brown MR, Bentley AR, Hui Q, Kraja AT, Morrison JL, O'Connel JR, Lee S, Schwander K, Bartz TM, de las Fuentes L, Feitosa MF, Guo X, Hanfei X, Harris SE, Huang Z, Kals M, Lefevre C, Mangino M, Milaneschi Y, van der Most P, Pacheco NL, Palmer ND, Rao V, Rauramaa R, Sun Q, Tabara Y, Vojinovic D, Wang Y, Weiss S, Yang Q, Zhao W, Zhu W, Abu Yusuf Ansari M, Aschard H, Anugu P, Assimes TL, Attia J, Baker LD, Ballantyne C, Bazzano L, Boerwinkle E, Cade B, Chen HH, Chen W, Ida Chen YD, Chen Z, Cho K, De Anda-Duran I, Dimitrov L, Do A, Edwards T, Faquih T, Hingorani A, Fisher-Hoch SP, Gaziano JM, Gharib SA, Giri A, Ghanbari M, Grabe HJ, Graff M, Gu CC, He J, Heikkinen S, Hixson J, Ho YL, Hood MM, Houghton SC, Karvonen-Gutierrez CA, Kawaguchi T, Kilpeläinen TO, Komulainen P, Lin HJ, Linchangco GV, Luik AI, Ma J, Meigs JB, McCormick JB, Menni C, Nolte IM, Norris JM, Petty LE, Polikowsky HG, Raffield LM, Rich SS, Riha RL, Russ TC, Ruiz-Narvaez EA, Sitlani CM, Smith JA, Snieder H, Sofer T, Shen B, Tang J, Taylor KD, Teder-Laving M, Triatin R, et alNoordam R, Wang W, Nagarajan P, Wang H, Brown MR, Bentley AR, Hui Q, Kraja AT, Morrison JL, O'Connel JR, Lee S, Schwander K, Bartz TM, de las Fuentes L, Feitosa MF, Guo X, Hanfei X, Harris SE, Huang Z, Kals M, Lefevre C, Mangino M, Milaneschi Y, van der Most P, Pacheco NL, Palmer ND, Rao V, Rauramaa R, Sun Q, Tabara Y, Vojinovic D, Wang Y, Weiss S, Yang Q, Zhao W, Zhu W, Abu Yusuf Ansari M, Aschard H, Anugu P, Assimes TL, Attia J, Baker LD, Ballantyne C, Bazzano L, Boerwinkle E, Cade B, Chen HH, Chen W, Ida Chen YD, Chen Z, Cho K, De Anda-Duran I, Dimitrov L, Do A, Edwards T, Faquih T, Hingorani A, Fisher-Hoch SP, Gaziano JM, Gharib SA, Giri A, Ghanbari M, Grabe HJ, Graff M, Gu CC, He J, Heikkinen S, Hixson J, Ho YL, Hood MM, Houghton SC, Karvonen-Gutierrez CA, Kawaguchi T, Kilpeläinen TO, Komulainen P, Lin HJ, Linchangco GV, Luik AI, Ma J, Meigs JB, McCormick JB, Menni C, Nolte IM, Norris JM, Petty LE, Polikowsky HG, Raffield LM, Rich SS, Riha RL, Russ TC, Ruiz-Narvaez EA, Sitlani CM, Smith JA, Snieder H, Sofer T, Shen B, Tang J, Taylor KD, Teder-Laving M, Triatin R, Tsai MY, Völzke H, Westerman KE, Xia R, Yao J, Young KL, Zhang R, Zonderman AB, Zhu X, Below JE, Cox SR, Evans M, Fornage M, Fox ER, Franceschini N, Harlow SD, Holliday E, Ikram MA, Kelly T, Lakka TA, Lawlor DA, Li C, Liu CT, Mägi R, Manning AK, Matsuda F, Morrison AC, Nauck M, North KE, Penninx BW, Province MA, Psaty BM, Rotter JI, Spector TD, Wagenknecht LE, Willems van Dijk K, Study LC, Jaquish CE, Wilson PW, Peyser PA, Munroe PB, de Vries PS, Gauderman WJ, Sun YV, Chen H, Miller CL, Winkler TW, Rao DC, Redline S, van Heemst D. A Large-Scale Genome-Wide Gene-Sleep Interaction Study in 732,564 Participants Identifies Lipid Loci Explaining Sleep-Associated Lipid Disturbances. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.02.24312466. [PMID: 39281768 PMCID: PMC11398441 DOI: 10.1101/2024.09.02.24312466] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
We performed large-scale genome-wide gene-sleep interaction analyses of lipid levels to identify novel genetic variants underpinning the biomolecular pathways of sleep-associated lipid disturbances and to suggest possible druggable targets. We collected data from 55 cohorts with a combined sample size of 732,564 participants (87% European ancestry) with data on lipid traits (high-density lipoprotein [HDL-c] and low-density lipoprotein [LDL-c] cholesterol and triglycerides [TG]). Short (STST) and long (LTST) total sleep time were defined by the extreme 20% of the age- and sex-standardized values within each cohort. Based on cohort-level summary statistics data, we performed meta-analyses for the one-degree of freedom tests of interaction and two-degree of freedom joint tests of the main and interaction effect. In the cross-population meta-analyses, the one-degree of freedom variant-sleep interaction test identified 10 loci (P int <5.0e-9) not previously observed for lipids. Of interest, the ASPH locus (TG, LTST) is a target for aspartic and succinic acid metabolism previously shown to improve sleep and cardiovascular risk. The two-degree of freedom analyses identified an additional 7 loci that showed evidence for variant-sleep interaction (P joint <5.0e-9 in combination with P int <6.6e-6). Of these, the SLC8A1 locus (TG, STST) has been considered a potential treatment target for reduction of ischemic damage after acute myocardial infarction. Collectively, the 17 (9 with STST; 8 with LTST) loci identified in this large-scale initiative provides evidence into the biomolecular mechanisms underpinning sleep-duration-associated changes in lipid levels. The identified druggable targets may contribute to the development of novel therapies for dyslipidemia in people with sleep disturbances.
Collapse
|
11
|
Niu Q, Zhang T, Mao R, Zhao N, Deng S. Genetic association of lipid and lipid-lowering drug target genes with atopic dermatitis: a drug target Mendelian randomization study. Sci Rep 2024; 14:18097. [PMID: 39103489 PMCID: PMC11300444 DOI: 10.1038/s41598-024-69180-2] [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: 11/10/2023] [Accepted: 08/01/2024] [Indexed: 08/07/2024] Open
Abstract
Observational studies suggest dyslipidemia as an atopic dermatitis (AD) risk factor and posit that lipid-lowering drugs may influence AD risk, but the causal link remains elusive. Mendelian randomization was applied to elucidate the causal role of serum lipids in AD and assess the therapeutic potential of lipid-lowering drug targets. Genetic variants related to serum lipid traits and lipid-lowering drug targets were sourced from the Global Lipid Genetics Consortium GWAS data. Comprehensive AD data were collated from the UK Biobank, FinnGen, and Biobank Japan. Colocalization, Summary-data-based Mendelian Randomization (SMR), and mediation analyses were utilized to validate the results and pinpoint potential mediators. Among assessed targets, only Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) was significantly linked to a reduced AD risk, corroborated across three separate AD cohorts. No association between serum lipid concentrations or other lipid-lowering drug targets and diminished AD risk was observed. Mediation analysis revealed that beta nerve growth factor (b-NGF) might mediate approximately 12.8% of PCSK9's influence on AD susceptibility. Our findings refute dyslipidemia's role in AD pathogenesis. Among explored lipid-lowering drug targets, PCSK9 stands out as a promising therapeutic agent for AD.
Collapse
Affiliation(s)
- Qinwang Niu
- Sichuan Polytechnic University, Deyang, 618000, Sichuan, China
| | - Tongtong Zhang
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, 610031, Sichuan, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
| | - Nana Zhao
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, 610031, Sichuan, China
| | - Sui Deng
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde, China.
| |
Collapse
|
12
|
Cai G, Liu J, Cai M, Shao L. Exploring the causal effect between lipid-modifying drugs and idiopathic pulmonary fibrosis: a drug-target Mendelian randomization study. Lipids Health Dis 2024; 23:237. [PMID: 39090671 PMCID: PMC11293199 DOI: 10.1186/s12944-024-02218-6] [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: 05/08/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a respiratory disorder of obscure etiology and limited treatment options, possibly linked to dysregulation in lipid metabolism. While several observational studies suggest that lipid-lowering agents may decrease the risk of IPF, the evidence is inconsistent. The present Mendelian randomization (MR) study aims to determine the association between circulating lipid traits and IPF and to assess the potential influence of lipid-modifying medications for IPF. METHODS Summary statistics of 5 lipid traits (high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglyceride, apolipoprotein A, and apolipoprotein B) and IPF were sourced from the UK Biobank and FinnGen Project Round 10. The study's focus on lipid-regulatory genes encompassed PCSK9, NPC1L1, ABCG5, ABCG8, HMGCR, APOB, LDLR, CETP, ANGPTL3, APOC3, LPL, and PPARA. The primary effect estimates were determined using the inverse-variance-weighted method, with additional analyses employing the contamination mixture method, robust adjusted profile score, the weighted median, weighted mode methods, and MR-Egger. Summary-data-based Mendelian randomization (SMR) was used to confirm significant lipid-modifying drug targets, leveraging data on expressed quantitative trait loci in relevant tissues. Sensitivity analyses included assessments of heterogeneity, horizontal pleiotropy, and leave-one-out methods. RESULTS There was no significant effect of blood lipid traits on IPF risk (all P>0.05). Drug-target MR analysis indicated that genetic mimicry for inhibitor of NPC1L1, PCSK9, ABCG5, ABCG8, and APOC3 were associated with increased IPF risks, with odds ratios (ORs) and 95% confidence intervals (CIs) as follows: 2.74 (1.05-7.12, P = 0.039), 1.36 (1.02-1.82, P = 0.037), 1.66 (1.12-2.45, P = 0.011), 1.68 (1.14-2.48, P = 0.009), and 1.42 (1.20-1.67, P = 3.17×10-5), respectively. The SMR method identified a significant association between PCSK9 gene expression in whole blood and reduced IPF risk (OR = 0.71, 95% CI: 0.50-0.99, P = 0.043). Sensitivity analyses showed no evidence of bias. CONCLUSIONS Serum lipid traits did not significantly affect the risk of idiopathic pulmonary fibrosis. Drug targets MR studies examining 12 lipid-modifying drugs indicated that PCSK9 inhibitors could dramatically increase IPF risk, a mechanism that may differ from their lipid-lowering actions and thus warrants further investigation.
Collapse
Affiliation(s)
- Gexiang Cai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mengsi Cai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lianyou Shao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
13
|
Huang A, Wu X, Lin J, Wei C, Xu W. Genetic insights into repurposing statins for hyperthyroidism prevention: a drug-target Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1331031. [PMID: 38425755 PMCID: PMC10902122 DOI: 10.3389/fendo.2024.1331031] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Background Current therapeutic measures for thyroid dysfunction are limited and often accompanied by adverse effects. The use of lipid-lowering drugs like statins has recently been associated with lower thyroid eye diseases risk. Objective To investigate the implications of genetically proxied lipid-lowering drugs on thyroid dysfunction. Methods In this drug-target Mendelian randomization (MR) study, we utilized genetic variants within drug target genes associated with low-density lipoprotein (LDL) or triglyceride (TG), derived from a genome-wide association study (GWAS) meta-analysis (N ≤ 188,577), to simulate lifelong drug interventions. Genetic summary statistics for thyroid dysfunction outcomes were retrieved from GWAS datasets of Thyroid Omics Consortium (N ≤ 54,288) and UK Biobank (N = 484,598). Inverse-variance-weighted MR (IVW-MR) method was performed as primary analysis, followed by validation in colocalization analysis. A subsequent two-step MR analysis was conducted to identify biomarkers mediating the identified drug-outcome association. Results In IVW-MR analysis, genetic mimicry of 3-hydroxy-3-methylglutarylcoenzyme reductase (HMGCR) inhibitors (e.g. statins) was significantly associated with lower risk of hyperthyroidism in two independent datasets (OR1, 0.417 per 1-mmol/L lower in LDL-C; 95% CI 0.262 to 0.664; P1 = 2.262 × 10-4; OR2 0.996; 95% CI 0.993-0.998; P2 = 0.002). Two-step MR analysis revealed eighteen biomarkers linked to genetic mimicry of HMGCR inhibition, and identified insulin-like growth factor 1 (IGF-1) levels mediating 2.108% of the negative causal relationship between HMGCR inhibition and hyperthyroidism. Conclusion This study supports HMGCR inhibition as a promising therapeutic strategy for hyperthyroidism and suggests its underlying mechanisms may extend beyond lipid metabolism. Further investigations through laboratory studies and clinical trials are necessary to confirm and elucidate these findings.
Collapse
Affiliation(s)
- Anqi Huang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Xinyi Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Jiaqi Lin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Chiju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, China
| | - Wencan Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
14
|
Huang K, Huang S, Xiong M. Correlations between genetically predicted lipid-lowering drug targets and inflammatory bowel disease. Lipids Health Dis 2024; 23:31. [PMID: 38287401 PMCID: PMC10823737 DOI: 10.1186/s12944-024-02026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/21/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Millions of individuals globally suffer from Inflammatory bowel diseases (IBDs). There is a dearth of large population-based investigations on lipid metabolism and IBDs, and it is unclear whether lipid-lowering drugs target IBDs causally. Consequently, the aim of this study was to investigate the effects of lipid-lowering medication targets on the occurrence and progression of IBDs. METHODS Among the more than 400,000 participants in the UK Biobank cohort and the more than 170,000 participants in the Global Lipids Genetics Consortium, a total of nine genes linked to lipid-lowering drug targets were obtained (ABCG5/ABCG8, APOB, APOC3, LDLR, LPL, HMGCR, NPC1L1, PCSK9, and PPARA). IBD data were acquired from de Lange et al. (patients/sample size of IBDs: 25042/59957; ulcerative colitis (UC): 12366/45,975; Crohn's disease (CD): 12194/40,266) and the FinnGen cohort (patients/total sample size of IBDs: 4420/176,899; CD: 1520/171,906; UC: 3325/173,711). All four datasets were cross-combined for validation via Mendelian randomization analysis, and potential mediating factors were explored via mediation analysis. RESULTS Genetically proxied APOC3 inhibition was related to increased IBD risk (odds ratio (95% confidence interval): 0.87 (0.80-0.95); P < 0.01) and UC risk (0.83 (0.73-0.94); P < 0.01). IBD and CD risk were reduced by genetic mimicry of LDLR and LPL enhancements, respectively (odds ratioLDLR: 1.18 (1.03-1.36); P = 0.018; odds ratioCD: 1.26 (1.11-1.43); P = 2.60E-04). Genetically proxied HMGCR inhibition was associated with increased CD risk (0.68 (0.50-0.94); P = 0.018). These findings were confirmed through Mendelian analysis of the cross-combination of four separate datasets. APOC3-mediated triglyceride levels may contribute to IBDs partly through mediated triglycerides, Clostridium sensu stricto 1, Clostridiaceae 1, or the Lachnospiraceae FCS020 group. LDLR enhancement may contribute to IBDs partly through increasing Lactobacillaceae. CONCLUSION Vigilance is required to prevent adverse effects on IBDs (UC) for patients receiving volanesorsen (an antisense oligonucleotide targeting ApoC3 mRNA) and adverse effects on CD for statin users. LPL and LDLR show promise as candidate drug targets for CD and IBD, respectively, with mechanisms that are potentially independent of their lipid-lowering effects.
Collapse
Affiliation(s)
- Kuiyuan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, 330000, China
| | - Shenan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, 330000, China
| | - Ming Xiong
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, 330000, China.
| |
Collapse
|
15
|
Brandts J, Tittel SR, Bramlage P, Danne T, Brix JM, Zimny S, Heyer CHJ, Holl RW, Müller-Wieland D. Low-density lipoprotein cholesterol and non-high-density lipoprotein cholesterol in type 1 diabetes and type 2 diabetes: Lipid goal attainment in a large German-Austrian diabetes registry. Diabetes Obes Metab 2023; 25:3700-3708. [PMID: 37694759 DOI: 10.1111/dom.15264] [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: 06/12/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/12/2023]
Abstract
AIM To assess the implementation of the 2019 European Society of Cardiology (ESC)/European Atherosclerosis Society (EAS) guideline recommendations for lipid-lowering therapies among more than 30 000 patients with type 1 diabetes (T1D) and type 2 diabetes (T2D) in a German and Austrian registry from 2020 to 2022. MATERIALS AND METHODS Registry data from 2020 and 2021 of 32 170 adult patients (8314 patients with T1D and 23 856 with T2D) were stratified according to the 2019 ESC/EAS risk categories, and guideline-based low-density lipoprotein cholesterol (LDL-C) and non-high-density lipoprotein cholesterol (non-HDL-C) goal attainment was analysed. RESULTS In patients with T1D (median age 38.35 [20.51-57.13] years), overall statin use was 19.3%, ezetimibe use was 2.2% and the use of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors or fibrates was less than 1%. In patients with T2D (median age 68.76 [58.86-78.39] years), 45.7% received statins, 3.4% received ezetimibe, and fibrates and PCSK9 inhibitors were used by 1% and 0.1%, respectively. Among patients with T1D, 6.16% reached their risk-based recommended LDL-C goal of less than 55 mg/dL (very high risk), 10.97% of less than 70 mg/dL (high risk), and 69.50% of less than 100 mg/dL (moderate risk), respectively. In patients with T2D, 11.81% reached their risk-based goal of LDL-C less than 55 mg/dL, 16.25% of less than 70 mg/dL, and 51.33% of less than 100 mg/dL. Non-HDL-C goals were reached more often, with 15.3%, 25.52% and 91.61% in patients with T1D and 18.56%, 17.96% and 82.30% in patients with T2D for very high, high and moderate risk, respectively. CONCLUSION Approximately 2 years after publication of the guidelines, LDL-C and non-HDL-C goal attainment was rarely achieved in patients with T1D and T2D with a high or very high cardiovascular risk.
Collapse
Affiliation(s)
- Julia Brandts
- Department of Medicine I, University Hospital Aachen, Aachen, Germany
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Sascha R Tittel
- Institute for Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Thomas Danne
- Kinderkrankenhaus auf der Bult, Diabeteszentrum für Kinder und Jugendliche, Hannover, Germany
| | - Johanna M Brix
- Department of Medicine I, Vienna, Austria
- Karl Landsteiner Institute for obesity and metabolism, Vienna, Austria
| | - Stefan Zimny
- Department of General Internal Medicine, Endocrinology and Diabetes, HELIOS Clinic Schwerin, Schwerin, Germany
| | | | - Reinhard W Holl
- Institute for Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | | |
Collapse
|
16
|
Hu Y, Zhang RQ, Liu SL, Wang ZG. In-situ quantification of lipids in live cells through imaging approaches. Biosens Bioelectron 2023; 240:115649. [PMID: 37678059 DOI: 10.1016/j.bios.2023.115649] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/03/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023]
Abstract
Lipids are important molecules that are widely distributed within the cell, and they play a crucial role in several biological processes such as cell membrane formation, signaling, cell motility and division. Monitoring the spatiotemporal dynamics of cellular lipids in real-time and quantifying their concentrations in situ is crucial since the local concentration of lipids initiates various signaling pathways that regulate cellular processes. In this review, we first introduced the historical background of lipid quantification methods. We then delve into the current state of the art of in situ lipid quantification, including the establishment and utility of fluorescence imaging techniques based on sensors of lipid-binding domains labeled with organic dyes or fluorescent proteins, and Raman and magnetic resonance imaging (MRI) techniques that do not require lipid labeling. Next, we highlighted the biological applications of live-cell lipid quantification techniques in the study of in situ lipid distribution, lipid transformation, and lipid-mediated signaling pathways. Finally, we discussed the technical challenges and prospects for the development of lipid quantification in live cells, with the aim of promoting the development of in situ lipid quantification in live cells, which may have a profound impact on the biological and medical fields.
Collapse
Affiliation(s)
- Yusi Hu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry and School of Medicine, Nankai University, Tianjin, 300071, China
| | - Rui-Qiao Zhang
- Qingdao Academy of Agricultural Sciences, Qingdao, 266100, China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry and School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry and School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
17
|
Laubach K, Turan T, Mathew R, Wilsbacher J, Engelhardt J, Samayoa J. Tumor-intrinsic metabolic reprogramming and how it drives resistance to anti-PD-1/PD-L1 treatment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:611-641. [PMID: 37842241 PMCID: PMC10571065 DOI: 10.20517/cdr.2023.60] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/15/2023] [Accepted: 08/29/2023] [Indexed: 10/17/2023]
Abstract
The development of immune checkpoint blockade (ICB) therapies has been instrumental in advancing the field of immunotherapy. Despite the prominence of these treatments, many patients exhibit primary or acquired resistance, rendering them ineffective. For example, anti-programmed cell death protein 1 (anti-PD-1)/anti-programmed cell death ligand 1 (anti-PD-L1) treatments are widely utilized across a range of cancer indications, but the response rate is only 10%-30%. As such, it is necessary for researchers to identify targets and develop drugs that can be used in combination with existing ICB therapies to overcome resistance. The intersection of cancer, metabolism, and the immune system has gained considerable traction in recent years as a way to comprehensively study the mechanisms that drive oncogenesis, immune evasion, and immunotherapy resistance. As a result, new research is continuously emerging in support of targeting metabolic pathways as an adjuvant to ICB to boost patient response and overcome resistance. Due to the plethora of studies in recent years highlighting this notion, this review will integrate the relevant articles that demonstrate how tumor-derived alterations in energy, amino acid, and lipid metabolism dysregulate anti-tumor immune responses and drive resistance to anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kyra Laubach
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
- Immuno-Oncology, AbbVie, South San Francisco, CA 94080, USA
| | - Tolga Turan
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
| | - Rebecca Mathew
- Immuno-Oncology, AbbVie, South San Francisco, CA 94080, USA
| | | | | | - Josue Samayoa
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
| |
Collapse
|
18
|
Fang Y, Su J, Zhao C, Meng Y, Wei B, Zhang B, Huang Y, Dai L, Ouyang S. Association between nontraditional lipid profiles and the severity of obstructive sleep apnea: A retrospective study. J Clin Lab Anal 2023; 37:e24499. [PMID: 35576500 PMCID: PMC10623523 DOI: 10.1002/jcla.24499] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Due to the significant role of dyslipidemia, cardiovascular diseases (CVDs) are very common in obstructive sleep apnea (OSA). Nontraditional lipid indices are considered to be a better predictive index for cardiovascular risk. Nevertheless, the association between nontraditional lipid profiles and the severity of OSA is not clear. METHODS A retrospective study was proceeded on 635 patients. Subjects were diagnosed with OSA through polysomnography (PSG). The association between severe OSA and nontraditional lipid profiles [triglyceride (TG)/high-density lipoprotein cholesterol (HDL-C) ratio, total cholesterol (TC)/HDL-C ratio, low-density lipoprotein cholesterol (LDL-C)/HDL-C ratio, non-high-density lipoprotein cholesterol (non-HDL-C), atherogenic index (AI), and lipoprotein combine index (LCI)] was examined by utilizing the restricted cubic spline and multivariate logistic regression analysis. RESULTS All nontraditional lipid indices had positive relationships with the severity of OSA. By multivariable adjustment, the per SD increment of the TG/HDL-C, TC/ HDL-C, LDL-C/HDL-C, non-HDL-C, AI, and LCI were significantly associated with 88%, 50%, 42%, 40%, 50%, and 125% higher risk for severe OSA respectively. Compared with the lowest tertiles, the adjusted ORs (95% CI) were 2.42 (1.57-3.75), 2.39 (1.53-3.73), 2.35 (1.52-3.64), 1.86 (1.21-2.86), 2.39 (1.53-3.73), and 2.23 (1.43-3.48) for the top tertiles of TG/HDL-C, TC/ HDL-C, LDL-C/HDL-C, non-HDL-C, AI, and LCI respectively. CONCLUSION All nontraditional lipid indices had positive relationship with the severity of OSA. In addition, TG/HDL-C, TC/HDL-C, and AI had better performance than the other nontraditional lipid indices for predicting severe OSA. These findings could help to determine the risk of cardiovascular diseases and improve the dyslipidemia management of OSA patients.
Collapse
Affiliation(s)
- Yifei Fang
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jiao Su
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chunling Zhao
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yang Meng
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Beilei Wei
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Binglu Zhang
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyang Huang
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Songyun Ouyang
- Department of Respiratory and Sleep MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
19
|
Cao J, Wang Z, Zhu M, Huang Y, Jin Z, Xiong Z. Low-density lipoprotein cholesterol and risk of hepatocellular carcinoma: a Mendelian randomization and mediation analysis. Lipids Health Dis 2023; 22:110. [PMID: 37525197 PMCID: PMC10388495 DOI: 10.1186/s12944-023-01877-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/21/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND A previous study demonstrated that low-density lipoprotein cholesterol (LDL-C) is associated with hepatocellular carcinoma (HCC); however, the causality between them has not been proven due to conflicting research results and the interference of confounders. This study utilized Mendelian randomization (MR) to investigate the causal relationship between LDL-C and HCC and identify the mediating factors. METHODS LDL-C, HCC, and coronary artery disease (CAD) genome-wide association study (GWAS) data were obtained from a public database. To investigate causality, inverse variance weighting (IVW) was the main analysis approach. MR‒Egger, simple mode, weighted median (WM), and weighted mode were employed as supplementary analytic methods. In addition, horizontal pleiotropy and heterogeneity were tested. To evaluate the stability of the MR results, a "leave-one-out" approach was used. Multivariate MR (MVMR) was utilized to correct the confounders that might affect causality, and mediation analysis was used to investigate the potential mediating effects. Finally, we used HCC risk to infer the reverse causality with LDL-C level. RESULTS Random effects IVW results were (LDL-C-HCC: odds ratio (OR) = 0.703, 95% confidence interval (CI) = [0.508, 0.973], P = 0.034; CAD-HCC: OR = 0.722, 95% CI = [0.645, 0.808], P = 1.50 × 10-8; LDL-C-CAD: OR = 2.103, 95% CI = [1.862, 2.376], P = 5.65 × 10-33), demonstrating a causal link between LDL-C levels and a lower risk of HCC. Through MVMR, after mutual correction, the causal effect of LDL-C and CAD on HCC remained significant (P < 0.05). Through mediation analysis, it was proven that CAD mediated the causative connection between LDL-C and HCC, and the proportion of mediating effect on HCC was 58.52%. Reverse MR showed that HCC could affect LDL-C levels with a negative correlation (ORIVW = 0.979, 95% CI = [0.961, 0.997], P = 0.025). CONCLUSION This MR study confirmed the causal effect between LDL-C levels and HCC risk, with CAD playing a mediating role. It may provide a new view on HCC occurrence and development mechanisms, as well as new metabolic intervention targets for treatment.
Collapse
Affiliation(s)
- Jiali Cao
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Ziwen Wang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Mengpei Zhu
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Yumei Huang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Ze Jin
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Zhifan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, China.
| |
Collapse
|
20
|
Wang D, Zhang P, Li J. Crossover point and maximal fat oxidation training effects on blood lipid metabolism in young overweight women: a pilot study. Front Physiol 2023; 14:1190109. [PMID: 37398909 PMCID: PMC10311904 DOI: 10.3389/fphys.2023.1190109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
Purpose: To determine the effects of weight reduction schemes using the exercise intensities corresponding to maximal fat oxidation (FATmax) and crossover point (COP). The effects of different intervention protocols on blood lipid metabolism were compared to explore how fat can be consumed and used more efficiently and provide a theoretical basis for weight loss through exercise. Methods: This study included 30 young overweight women randomly divided into the COP, FATmax, and control groups. Participants in the COP and FATmax groups exercised for 45 min four times a week for 8 weeks after the individual treadmill exercise test. The control group did not perform any exercise. Results: After 8 weeks of training, participants in the COP group significantly decreased weight (2.6 ± 3.3 kg), body mass index (0.91 ± 1.26 kg/m2), body fat percentage (1.21% ± 1.50%), and fat mass (1.90 ± 2.30 kg) (p < 0.05). They also had significantly decreased hip circumference (4.8 ± 3.3 cm), serum apolipoprotein B (ApoB) levels (15.48 ± 14.19 mg/dL), and ApoB/apolipoprotein AI (ApoAI) ratios (0.47 ± 0.37) (p < 0.01). However, their serum ApoAI levels were significantly increased (14.18 ± 10.24 mg/dL; p < 0.01). Participants in the FATmax group had significantly decreased hip circumference (2.4 ± 2.0 cm), serum ApoB levels (14.49 ± 11.00 mg/dL), and ApoB/ApoAI ratios (0.59 ± 0.30) (p < 0.01) but significantly increased serum ApoAI levels (29.53 ± 13.29 mg/dL; p < 0.01). No significant changes in physiological indexes were observed in participants in the control group. Conclusion: Personalised exercise intervention positively affected central obesity, effectively improving blood lipid metabolism and fat oxidation, reducing cardiovascular disease risk in young overweight women. COP training improved weight and body composition better than the FATmax exercise, while the latter provided greater improvements in serum ApoAI levels.
Collapse
Affiliation(s)
- Dizhi Wang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Peizhen Zhang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Jin Li
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| |
Collapse
|
21
|
Elis A. Current and future options in cholesterol lowering treatments. Eur J Intern Med 2023; 112:1-5. [PMID: 36813611 DOI: 10.1016/j.ejim.2023.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023]
Abstract
The relative risk reduction of cardiovascular events is proportional to the absolute reduction in LDL-C levels, the primary target of therapy, no matter the way of reduction. During the last decades, the therapeutic regimens for reducing the LDL-C levels have been immerged and improved, with favorable effects on the atherosclerotic process and clinical benefits of various cardiovascular outcomes. From a practical view of point, this review is focusing only on the current available lipid lowering agents: statins, ezetimibe, anti PCSK9 monoclonal antibodies, the small interfering RNA (siRNA) agent, Inclisiran, and Bempedoic acid. The recent changes in lipid lowering regimens, including the early combination of lipid lowering agents and "Low LDL-C" levels <30 mg/dL for high/very high cardiovascular risk patients will also be discussed.
Collapse
Affiliation(s)
- Avishay Elis
- Department of Internal Medicine "C", Beilinson Hospital, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
22
|
Li Z, Zhang B, Liu Q, Tao Z, Ding L, Guo B, Zhang E, Zhang H, Meng Z, Guo S, Chen Y, Peng J, Li J, Wang C, Huang Y, Xu H, Wu Y. Genetic association of lipids and lipid-lowering drug target genes with non-alcoholic fatty liver disease. EBioMedicine 2023; 90:104543. [PMID: 37002989 PMCID: PMC10070091 DOI: 10.1016/j.ebiom.2023.104543] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Some observational studies found that dyslipidaemia is a risk factor for non-alcoholic fatty liver disease (NAFLD), and lipid-lowering drugs may lower NAFLD risk. However, it remains unclear whether dyslipidaemia is causative for NAFLD. This Mendelian randomisation (MR) study aimed to explore the causal role of lipid traits in NAFLD and evaluate the potential effect of lipid-lowering drug targets on NAFLD. METHODS Genetic variants associated with lipid traits and variants of genes encoding lipid-lowering drug targets were extracted from the Global Lipids Genetics Consortium genome-wide association study (GWAS). Summary statistics for NAFLD were obtained from two independent GWAS datasets. Lipid-lowering drug targets that reached significance were further tested using expression quantitative trait loci data in relevant tissues. Colocalisation and mediation analyses were performed to validate the robustness of the results and explore potential mediators. FINDINGS No significant effect of lipid traits and eight lipid-lowering drug targets on NAFLD risk was found. Genetic mimicry of lipoprotein lipase (LPL) enhancement was associated with lower NAFLD risks in two independent datasets (OR1 = 0.60 [95% CI 0.50-0.72], p1 = 2.07 × 10-8; OR2 = 0.57 [95% CI 0.39-0.82], p2 = 3.00 × 10-3). A significant MR association (OR = 0.71 [95% CI, 0.58-0.87], p = 1.20 × 10-3) and strong colocalisation association (PP.H4 = 0.85) with NAFLD were observed for LPL expression in subcutaneous adipose tissue. Fasting insulin and type 2 diabetes mediated 7.40% and 9.15%, respectively, of the total effect of LPL on NAFLD risk. INTERPRETATION Our findings do not support dyslipidaemia as a causal factor for NAFLD. Among nine lipid-lowering drug targets, LPL is a promising candidate drug target in NAFLD. The mechanism of action of LPL in NAFLD may be independent of its lipid-lowering effects. FUNDING Capital's Funds for Health Improvement and Research (2022-4-4037). CAMS Innovation Fund for Medical Sciences (CIFMS, grant number: 2021-I2M-C&T-A-010).
Collapse
Affiliation(s)
- Ziang Li
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Bin Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Qingrong Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhihang Tao
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Ding
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Bo Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Erli Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haitong Zhang
- Department of Cardiology, the Third-Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhen Meng
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Shuai Guo
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yang Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jia Peng
- Department of Cardiology, the First-Affiliated Hospital, Xiangya Hospital Central South University, Changsha, China
| | - Jinyue Li
- Key Laboratory of Cardiovascular Epidemiology & Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Can Wang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yingbo Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyan Xu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Yongjian Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
23
|
Ennezat PV, Guerbaai RA, Maréchaux S, Le Jemtel TH, François P. Extent of Low-density Lipoprotein Cholesterol Reduction and All-cause and Cardiovascular Mortality Benefit: A Systematic Review and Meta-analysis. J Cardiovasc Pharmacol 2023; 81:35-44. [PMID: 36027598 PMCID: PMC9812424 DOI: 10.1097/fjc.0000000000001345] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023]
Abstract
ABSTRACT Lipid-modifying agents steadily lower low-density lipoprotein cholesterol (LDL-C) levels with the aim of reducing mortality. A systematic review and meta-analysis were conducted to determine whether all-cause or cardiovascular (CV) mortality effect size for lipid-lowering therapy varied according to the magnitude of LDL-C reduction. Electronic databases were searched, including PubMed and ClinicalTrials.gov , from inception to December 31, 2019. Eligible studies included randomized controlled trials that compared lipid-modifying agents (statins, ezetimibe, and PCSK-9 inhibitors) versus placebo, standard or usual care or intensive versus less-intensive LDL-C-lowering therapy in adults, with or without known history of CV disease with a follow-up of at least 52 weeks. All-cause and CV mortality as primary end points, myocardial infarction, stroke, and non-CV death as secondary end points. Absolute risk differences [ARD (ARDs) expressed as incident events per 1000 person-years], number needed to treat (NNT), and rate ratios (RR) were assessed. Sixty randomized controlled trials totaling 323,950 participants were included. Compared with placebo, usual care or less-intensive therapy, active or more potent lipid-lowering therapy reduced the risk of all-cause death [ARD -1.33 (-1.89 to -0.76); NNT 754 (529-1309); RR 0.92 (0.89-0.96)]. Intensive LDL-C percent lowering was not associated with further reductions in all-cause mortality [ARD -0.27 (-1.24 to 0.71); RR 1.00 (0.94-1.06)]. Intensive LDL-C percent lowering did not further reduce CV mortality [ARD -0.28 (-0.83 to 0.38); RR 1.02 (0.94-1.09)]. Our findings indicate that risk reduction varies across subgroups and that overall NNTs are high. Identifying patient subgroups who benefit the most from LDL-C levels reduction is clinically relevant and necessary.
Collapse
Affiliation(s)
| | | | - Sylvestre Maréchaux
- Department of cardiology, Groupement des Hôpitaux de l’Institut Catholique de Lille, Lomme, France
| | - Thierry H. Le Jemtel
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine; Tulane University Heart and Vascular Institute, New Orleans, LA; and
| | - Patrice François
- Department of Epidemiology, University of Grenoble Alpes, TIMC UMR 5525 CNRS and Centre Hospitalier Universitaire de Grenoble-Alpes, La Tronche, France
| |
Collapse
|
24
|
Zhang F, Li J, Chang C, Gu L, Xiong W, Su Y, Yang Y. The Association of Dietary Cholesterol from Egg Consumption on Cardiovascular Diseases Risk Varies from Person to Person. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14977-14988. [PMID: 36416372 DOI: 10.1021/acs.jafc.2c04634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The public and scientists remain skeptical about egg consumption, given that cardiovascular diseases (CVDs) are the leading causes of death in worldwide. This review mainly explained the recurrence of contradictory conclusions about relationships between egg consumption and CVD risk and discussed effects of egg cholesterol intake on cholesterol homeostasis. Factors including individual health status and cholesterol sensitivity, dietary pattern, region, and race should be distinguished when understanding generalized conclusions. Identified compensatory mechanisms in response to dietary cholesterol and the resulting balance in cholesterol biosynthesis, absorption, and efflux supported the view that moderate egg consumption had no substantial overall impacts on cholesterol homeostasis in healthy people. Excessive cholesterol intake is not recommended in individuals with distempered metabolism. More than cholesterol metabolism, impacts of egg consumption as a part of overall diet on CVD risk should be considered from aspects of nutrient intake, lipid metabolism, and energy supply in the future.
Collapse
Affiliation(s)
- Fan Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Junhua Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Cuihua Chang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Luping Gu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Wen Xiong
- Hunan Engineering and Technology Research Center for Food Flavors and Flavorings, Jinshi, Hunan 415400, PR China
| | - Yujie Su
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yanjun Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| |
Collapse
|
25
|
Xiao W, Li J, Huang X, Zhu Q, Liu T, Xie H, Deng Z, Tang Y. Mediation roles of neutrophils and high-density lipoprotein (HDL) on the relationship between HLA-DQB1 and rosacea. Ann Med 2022; 54:1530-1537. [PMID: 35622385 PMCID: PMC9891224 DOI: 10.1080/07853890.2022.2077427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/18/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Though the previous genome-wide association studies found the association between HLA alleles and rosacea in the European populations, the data is lacking among the Asians. Moreover, neutrophils are important in the immune-related mechanism of rosacea, and dyslipidemia is closely related to rosacea. We aimed to explore the association between HLA genes and rosacea in Chinese rosacea patients, as well as the mediation effect of neutrophils, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) on the relationship between HLA genes and rosacea. METHODS A total of 249 rosacea and 150 controls were ranked by the international investigator global rosacea severity scores. HLA genes, neutrophils, HDL, and LDL were detected. And their mediation effects on the relationship between HLA and rosacea risk or severity were analysed. RESULTS HLA-DQB1*03:03 allele (OR = 41.89, 95% CI: 9.80 ∼ 179.09, p = 4.7*10-7), HLA-DQB1*04:02 allele (OR = 0.16, 95% CI: 0.03 ∼ 0.81, p = 0.026) and HLA-DQB1*03:03/05:02 genotype (OR = 5.57, 95% CI: 1.13 ∼ 27.52, p = 0.0351) were significantly associated with rosacea. Moreover, HLA-DQB1*03:03 allele (b = 1.434, SE = 0.217, p = 2.0*10-10), HLA-DQB1*05:01 allele (b = 0.894, SE = 0.33520, p = 0.008) and HLA-DQB1*03:03/06:01 genotype (b = 0.998, SE = 0.472, p = 0.040) were positively associated with rosacea severity. Furthermore, we found both neutrophils and HDL, instead of LDL, have mediation effects on the relationship between HLA-DQB1*03:03 and risk or severity of rosacea. CONCLUSIONS We discovered novel susceptible HLA alleles for rosacea in the Chinese population, and disclosed the mediation effect of neutrophils and HDL on the relationship between HLA-DQB1 and rosacea, implying a possible correlation between rosacea and inflammatory or metabolic factors, providing hints for future studies in the mechanism of rosacea. Key messagesHLA-DQB1*03:03 allele, HLA-DQB1*04:02 allele and HLA-DQB1*03:03/05:02 genotype were significantly associated with rosacea.HLA-DQB1*03:03 allele, HLA-DQB1*05:01 allele and HLA-DQB1*03:03/06:01 genotype were positively associated with rosacea severity.Neutrophils and HDL have mediation effects on the relationship between HLA-DQB1*03:03 and risk or severity of rosacea.
Collapse
Affiliation(s)
- Wenqin Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South Univerisity, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South Univerisity, Changsha, China
| | - Xin Huang
- Department of Epidemiology and Biostatistics, School of Medicine, Hunan Normal University, Changsha, China
| | - Quan Zhu
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Tangxiele Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South Univerisity, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South Univerisity, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South Univerisity, Changsha, China
| | - Yan Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South Univerisity, Changsha, China
| |
Collapse
|
26
|
A novel, orally bioavailable, small-molecule inhibitor of PCSK9 with significant cholesterol-lowering properties in vivo. J Lipid Res 2022; 63:100293. [DOI: 10.1016/j.jlr.2022.100293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022] Open
|
27
|
Elevated triglycerides and reduced high-density lipoprotein cholesterol are independently associated with the onset of advanced chronic kidney disease: a cohort study of 911,360 individuals from the United Kingdom. BMC Nephrol 2022; 23:312. [PMID: 36109725 PMCID: PMC9479392 DOI: 10.1186/s12882-022-02932-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Increased total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and decreased high-density lipoprotein cholesterol (HDL-C) concentrations, are established risk factors for cardiovascular morbidity and mortality; but their impact on the risk of advanced chronic kidney disease (CKD) is unclear. This study evaluates the association between the different lipid profiles and the onset of advanced CKD using a general population sample.
Methods
This observational study used records of 911,360 individuals from the English Clinical Practice Research Datalink (from 2000 to 2014), linked to coded hospital discharges and mortality registrations. Cox models were used to examine the independent association between the equal quarters of TC, TG, LDL-C, and HDL-C and the risk of advanced CKD, after adjustment for sex and age, and potential effect mediators.
Results
During a median follow-up of 7.5 years, 11,825 individuals developed CKD stages 4–5. After adjustment for sex and age, the hazard ratios (HRs) and confidence intervals (CIs) for CKD stages 4–5 comparing the 4th vs. 1st quarters of TG and 1st vs. 4th quarters of HDL-C were 2.69 (95% CI, 2.49–2.90) and 2.61 (95% CI, 2.42–2.80), respectively. Additional adjustment for potential effect mediators reduced the HRs to 1.28 (95% CI, 1.15–1.43), and 1.27 (95% CI, 1.14–1.41), respectively. There was no evidence of fully adjusted associations with CKD stages 4–5 for levels of either TC or LDL-C.
Conclusions
Elevated TG and reduced HDL-C levels are independently associated with the onset of advanced CKD. Future studies, such as in basic science and randomized trials, are needed to understand whether associations between TG and HDL-C and the development of CKD are causal.
Collapse
|
28
|
Brandts J, Verket M, Müller-Wieland D. [Lipid lowering: new agents and new concepts]. Herz 2022; 47:419-425. [PMID: 36018378 DOI: 10.1007/s00059-022-05133-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 12/01/2022]
Abstract
Low-density lipoprotein (LDL) cholesterol (LDL-C) is a causal risk factor for cardiovascular complications. A target value is set according to risk, guideline-based and individual basis. We now have the means to lower LDL‑C levels to ranges that are even associated with plaque volume regression. Moreover, lipid treatment is an example of how pharmacotherapy has evolved from classical selective inhibition of enzymes by drugs (e.g. statins) to targeted neutralization of proteins by antibodies. The reduction of atherogenic lipoproteins by specific inhibition or reduction of mRNA of target proteins, e.g. PCSK‑9, ANGPLT3, ApoC-III or Apo (a), and possibly one day by vaccination or even CRISP-based gene therapy will in the long term lead to new concepts in the treatment and prevention of dyslipidemia and cardiovascular complications. The cumulative exposure of atherogenic lipoproteins to the vessel wall is determined by the time-averaged LDL‑C level. This essentially depends on patient adherence and prescribed treatment intensity by physicians. Therefore, it is likely that treatment adherence influences the cumulative benefit of treatment. Accordingly, the new therapeutic strategies mentioned above with presumably higher adherence rates could help to optimize cardiovascular prevention. Early and effective LDL‑C lowering could drastically reduce the incidence of cardiovascular complications in the long term and help to maintain the health of our patients.
Collapse
Affiliation(s)
- Julia Brandts
- Medizinische Klinik I, Universitätsklinikum RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland
| | - Marlo Verket
- Medizinische Klinik I, Universitätsklinikum RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland
| | - Dirk Müller-Wieland
- Medizinische Klinik I, Universitätsklinikum RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland.
| |
Collapse
|
29
|
Watanabe K, Kakeda S, Nemoto K, Onoda K, Yamaguchi S, Kobayashi S, Yamakawa Y. Effects of Obesity, Blood Pressure, and Blood Metabolic Biomarkers on Grey Matter Brain Healthcare Quotient: A Large Cohort Study of a Magnetic Resonance Imaging Brain Screening System in Japan. J Clin Med 2022; 11:jcm11112973. [PMID: 35683364 PMCID: PMC9181611 DOI: 10.3390/jcm11112973] [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: 04/03/2022] [Revised: 05/12/2022] [Accepted: 05/18/2022] [Indexed: 02/05/2023] Open
Abstract
This study investigated the relationship between grey matter (GM) volume and blood biomarkers, blood pressure, and obesity. We aimed to elucidate lifestyle factors that promote GM volume loss. A total of 1799 participants underwent the brain dock as a medical checkup. Data regarding blood pressure, obesity measurements, and standard blood biomarkers were obtained. Further, brain magnetic resonance imaging (MRI), including high-resolution T1-weighted imaging, was performed. We calculated the grey matter brain healthcare quotient (GM-BHQ), which represents GM volume as a deviation value. After adjusting for confounding variables, multiple regression analysis revealed that body mass index (BMI) (b = −0.28, p < 0.001), gamma-glutamyltransferase (γ-GTP) (b = −0.01, p = 0.16), and fasting blood glucose (b = −0.02, p = 0.049) were significantly correlated with GM-BHQ. Although the current cross-sectional study cannot determine a cause-and-effect relationship, elevated BMI, γ-GTP, and fasting blood glucose could promote GM volume loss.
Collapse
Affiliation(s)
- Keita Watanabe
- Institution of Open Innovation, Kyoto University, Kyoto 606-8501, Japan;
- Correspondence: ; Tel.: +81-075-753-5534
| | - Shingo Kakeda
- Department of Diagnostic Radiology, Hirosaki University Graduate School of Medicine Radiology, Aomori 036-8562, Japan;
| | - Kiyotaka Nemoto
- Division of Clinical Medicine, Department of Neuropsychiatry, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan;
| | - Keiichi Onoda
- Department of Psychology, Otemon Gakuin University, Osaka 567-8502, Japan;
| | - Shuhei Yamaguchi
- Department of Neurology, Shimane University, Izumo 690-0823, Japan; (S.Y.); (S.K.)
- Department of Neurology, Shimane Prefectural Central Hospital, Izumo 693-0068, Japan
| | - Shotai Kobayashi
- Department of Neurology, Shimane University, Izumo 690-0823, Japan; (S.Y.); (S.K.)
| | - Yoshinori Yamakawa
- Institution of Open Innovation, Kyoto University, Kyoto 606-8501, Japan;
- Institute of Innovative Research, Tokyo Institute of Technology, Tokyo 152-8550, Japan
- Academic and Industrial Innovation, Kobe University, Kobe 657-8501, Japan
- ImPACT Program of Council for Science, Technology, and Innovation, Cabinet Office, Tokyo 100-8914, Japan
- Brain Impact General Incorporated Association, Kyoto 606-8501, Japan
| |
Collapse
|
30
|
Yuefeng Y, Zhiqi L, Yi C, Keyu Z, Heng W, Yuying W, Ningjian W, Yuetian Y, Xinjie G, Yihao Z, Yingli L, Fangzhen X. Testosterone Deficiency Promotes Hypercholesteremia and Attenuates Cholesterol Liver Uptake via AR/PCSK9/LDLR Pathways. Int J Endocrinol 2022; 2022:7989751. [PMID: 35599686 PMCID: PMC9122719 DOI: 10.1155/2022/7989751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/05/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Testosterone deficiency is reportedly correlated with an elevation of cholesterol in plasma, but the mechanism remains unclear. Our objective was to investigate the effects of testosterone deficiency on cholesterol metabolism and the corresponding molecular changes in vivo and in vitro. METHODS SD rats were randomized into three groups: sham-operated (SHAM), subtotal orchiectomized (SO), and orchiectomized (ORX) and fed for 8 weeks. HepG2 cells were cultured with medium containing testosterone with the final concentrations of 0, 10, 30, and 300 nM. Method of isotope tracing and fluorescence labelling was adopted to investigate cholesterol metabolism. Several key molecules of cholesterol metabolism were also analyzed. RESULTS SO and ORX rats displayed dysfunctional liver uptake of cholesterol. HepG2 cells incubated with testosterone of lower and excessive level exhibited reduced capacity of cholesterol uptake. Further investigation revealed that lack of testosterone induced increased proprotein convertase subtilisin/kexin type 9 (PCSK9) and decreased low-density lipoprotein receptor (LDLR) both in vivo and in vitro. Moreover, the androgen receptor (AR) antagonist flutamide mimicked the effects of testosterone deficiency on PCSK9 and LDLR indicating the role of AR as a mediator in triggering attenuating liver cholesterol uptake in which testosterone instead of dihydrotestosterone (DHT) is the major functional form of androgen. CONCLUSION Testosterone deficiency attenuated cholesterol liver uptake mediated by the PCSK9-LDLR pathway, in which AR and testosterone without transforming to DHT play important roles.
Collapse
Affiliation(s)
- Yu Yuefeng
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Lin Zhiqi
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Chen Yi
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Zhu Keyu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Wan Heng
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Wang Yuying
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Wang Ningjian
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yu Yuetian
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Gu Xinjie
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Zhang Yihao
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Lu Yingli
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Xia Fangzhen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
31
|
Lange KW. Tea in cardiovascular health and disease: a critical appraisal of the evidence. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.12.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
MiR-199a-3p Restrains Foaming and Inflammation by Regulating RUNX1 in Macrophages. Mol Biotechnol 2022; 64:1130-1142. [DOI: 10.1007/s12033-022-00484-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/20/2022] [Indexed: 10/18/2022]
|
33
|
Clinical Phenotypes of Cardiovascular and Heart Failure Diseases Can Be Reversed? The Holistic Principle of Systems Biology in Multifaceted Heart Diseases. CARDIOGENETICS 2022. [DOI: 10.3390/cardiogenetics12020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Recent advances in cardiology and biological sciences have improved quality of life in patients with complex cardiovascular diseases (CVDs) or heart failure (HF). Regardless of medical progress, complex cardiac diseases continue to have a prolonged clinical course with high morbidity and mortality. Interventional coronary techniques together with drug therapy improve quality and future prospects of life, but do not reverse the course of the atherosclerotic process that remains relentlessly progressive. The probability of CVDs and HF phenotypes to reverse can be supported by the advances made on the medical holistic principle of systems biology (SB) and on artificial intelligence (AI). Studies on clinical phenotypes reversal should be based on the research performed in large populations of patients following gathering and analyzing large amounts of relative data that embrace the concept of complexity. To decipher the complexity conundrum, a multiomics approach is needed with network analysis of the biological data. Only by understanding the complexity of chronic heart diseases and explaining the interrelationship between different interconnected biological networks can the probability for clinical phenotypes reversal be increased.
Collapse
|
34
|
Matsuo M. ABCA1 and ABCG1 as potential therapeutic targets for the prevention of atherosclerosis. J Pharmacol Sci 2022; 148:197-203. [DOI: 10.1016/j.jphs.2021.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022] Open
|
35
|
Hu C, Luo W, Xu J, Han X. RECOGNITION AND AVOIDANCE OF ION SOURCE-GENERATED ARTIFACTS IN LIPIDOMICS ANALYSIS. MASS SPECTROMETRY REVIEWS 2022; 41:15-31. [PMID: 32997818 PMCID: PMC8287896 DOI: 10.1002/mas.21659] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 05/04/2023]
Abstract
Lipid research is attracting more and more attention as various key roles and novel biological functions of lipids have been demonstrated and discovered in the organism. Mass spectrometry (MS)-based lipidomics approaches are the most powerful and effective tools for analysis of cellular lipidomes with very high sensitivity and specificity. However, the artifacts generated from in-source fragmentation are always present in all kinds of ion sources, even soft ionization techniques (i.e., electrospray ionization and matrix-assisted laser desorption/ionization [MALDI]). These artifacts can cause many problems for lipidomics, especially when the fragment ions correspond to/are isomeric species of other endogenous lipid species in complex biological samples. These commonly observed artifacts could lead to misannotation, false identification, and consequently, incorrect attribution of phenotypes, and will have negative impact on any MS-based lipidomics research including but not limited to biomarker discovery, drug development, etc. Liquid chromatography-MS, shotgun lipidomics, and MALDI-MS imaging are three representative lipidomics approaches in which ion source-generated artifacts are all manifested and are comprehensively summarized in this article. The strategies on how to avoid/reduce the artifacts of in-source fragmentation on lipidomics analysis are also discussed in detail. We believe that with the recognition and avoidance of ion source-generated artifacts, MS-based lipidomics approaches will provide better accuracy on comprehensive analysis of biological samples and will make greater contribution to the research on metabolism and translational/precision medicine (collectively termed functional lipidomics). © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- Changfeng Hu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
| | - Wenqing Luo
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003 China
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229 USA
- Department of Medicine – Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229 USA
| |
Collapse
|
36
|
Müller-Wieland D, Merkel M, Verket M, März W, von Eckardstein A. [Pathophysiological principles of dyslipoproteinaemia]. Dtsch Med Wochenschr 2021; 146:e103-e111. [PMID: 34731902 DOI: 10.1055/a-1516-2441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The effective reduction of atherogenic lipoproteins has contributed to the rate of atherosclerosis-related cardiovascular complications being approximately halved over the last 50 years. Nevertheless, cardiovascular disease will be the leading cause of death worldwide in the coming years. The focus of this review is on the clinical significance of the pathophysiology of changes in lipid and lipoprotein metabolism. Elevated levels of atherogenic lipoproteins are a causal risk factor for atherosclerotic cardiovascular disease. Primary forms of hypercholesterolaemia have a significantly higher ASCVD risk because of the already lifelong LDL elevation (higher cumulative LDL exposure for the vessel wall). Secondary changes in lipid and lipoprotein metabolism (e. g. in diabetes or hypothyroidism) must be excluded or treated. Regulatory key steps in the pathophysiology of lipid metabolism and atherosclerotic plaque are "drug targets" for existing and new lipid and lipoprotein modifying therapies.
Collapse
|
37
|
Wang M, Zhang R, Dehaen W, Fang Y, Qian S, Ren Y, Cheng F, Guo Y, Guo C, Li Y, Deng Y, Cao Z, Peng C. Specific recognition, intracellular assay and detoxification of fluorescent curcumin derivative for copper ions. JOURNAL OF HAZARDOUS MATERIALS 2021; 420:126490. [PMID: 34252661 DOI: 10.1016/j.jhazmat.2021.126490] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 06/13/2023]
Abstract
Recognition and excretion of metal ions play an important role in the diagnosis and treatment of various diseases and poisoning. Although copper (Cu) is a cofactor of many key enzymes in the human body, its accumulation caused by genetic ATP7B mutation or environmental pollution can lead to hepatotoxicity, renal failure, Wilson's disease, inflammation, and even Parkinson's disease (PD) and Alzheimer's disease (AD). Therefore, in this work, a difluoroboron curcumin derivative (DF-Cur) was used for the specific recognition of copper ions (Cu2+). DF-Cur could be further used to as a rapid diagnostic agent for the copper detection in cells and zebrafish at the nanomolar level. DF-Cur could significantly reduce the toxic damage caused by high Cu2+ dose. Inductively coupled plasma-mass spectrometry (ICP-MS) analysis indicated that DF-Cur could promote the excretion of copper ions in the urine and bile and reduce the accumulation of copper ions in vivo. In addition, DF-Cur could selectively detect cholesterol in the blood and adipose tissue in vivo by fluorescent staining. These results demonstrated that this molecule might represent a new and promising diagnostic and therapeutic agent to combat diseases related to copper ions accumulation.
Collapse
Affiliation(s)
- Miao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wim Dehaen
- Department of Chemistry, KU Leuven, Celestijnenlaan 200f-bus 02404, 3001 Leuven, Belgium
| | - Yuyu Fang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Chemistry, KU Leuven, Celestijnenlaan 200f-bus 02404, 3001 Leuven, Belgium.
| | - Shan Qian
- Department of Pharmaceutical Engineering, School of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Yali Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuying Guo
- Department of Pharmaceutical Engineering, School of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Chuanjie Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yun Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
38
|
Apolipoprotein B and Cardiovascular Disease: Biomarker and Potential Therapeutic Target. Metabolites 2021; 11:metabo11100690. [PMID: 34677405 PMCID: PMC8540246 DOI: 10.3390/metabo11100690] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein (apo) B, the critical structural protein of the atherogenic lipoproteins, has two major isoforms: apoB48 and apoB100. ApoB48 is found in chylomicrons and chylomicron remnants with one apoB48 molecule per chylomicron particle. Similarly, a single apoB100 molecule is contained per particle of very-low-density lipoprotein (VLDL), intermediate density lipoprotein, LDL and lipoprotein(a). This unique one apoB per particle ratio makes plasma apoB concentration a direct measure of the number of circulating atherogenic lipoproteins. ApoB levels indicate the atherogenic particle concentration independent of the particle cholesterol content, which is variable. While LDL, the major cholesterol-carrying serum lipoprotein, is the primary therapeutic target for management and prevention of atherosclerotic cardiovascular disease, there is strong evidence that apoB is a more accurate indicator of cardiovascular risk than either total cholesterol or LDL cholesterol. This review examines multiple aspects of apoB structure and function, with a focus on the controversy over use of apoB as a therapeutic target in clinical practice. Ongoing coronary artery disease residual risk, despite lipid-lowering treatment, has left patients and clinicians with unsatisfactory options for monitoring cardiovascular health. At the present time, the substitution of apoB for LDL-C in cardiovascular disease prevention guidelines has been deemed unjustified, but discussions continue.
Collapse
|
39
|
Xu X, Dong Y, Ma N, Kong W, Yu C, Gong L, Chen J, Ren J. MiR-337-3p lowers serum LDL-C level through targeting PCSK9 in hyperlipidemic mice. Metabolism 2021; 119:154768. [PMID: 33775647 DOI: 10.1016/j.metabol.2021.154768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 03/04/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Reducing serum low-density lipoprotein cholesterol (LDL-C) in hyperlipemia is recognized as an effective strategy to minimize the risk of atherosclerotic cardiovascular disease (ASCVD). MiR-337-3p has already been discovered to play regulatory roles in tumor proliferation and metastasis, adipocyte browning and ischemic brain injury, etc. However, the association between miR-337-3p and LDL-C is unknown. METHODS Gene Expression Omnibus (GEO) dataset and two hyperlipidemic murine models were used to analyze the potential relationship between miR-337-3p and LDL-C. AAV-mediated liver-directed miRNA overexpression in high fat diet (HFD)-fed mouse model was used to examine the effect of miR-337-3p on LDL-C and WB/RT-PCR/ELISA/luciferase assays were used to investigate the underlying mechanism. RESULTS The expressions of miR-337-3p were obviously lower in multiple hyperlipidemic mouse models and had a negative correlation with serum LDL-C levels. After confirming the effect of miR-337-3p on the improvement of serum LDL-C in vivo, we discovered PCSK9 might be a possible target of miR-337-3p, which was further proved by in vitro experiments. MiR-337-3p could directly interact with both the PCSK9 3'UTR and promoter to inhibit PCSK9 translation and transcription. Furthermore, the result from DiI-LDL uptake assay under the knockdown of PCSK9 demonstrated that miR-337-3p promoting the absorption of LDL-C in HepG2 cells was dependent on PCSK9, and the result from LDLR-/- mouse model indicated that miR-337-3p regulating LDL-C was dependent on PCSK9/LDLR pathway. CONCLUSION We discovered a new function of miR-337-3p in regulating PCSK9 expression and LDL-C absorption, suggesting miR-337-3p might be a new therapeutic target for the development of antihyperlipidemic drug.
Collapse
Affiliation(s)
- Xiaoding Xu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Yunxia Dong
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Ningning Ma
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| | - Weiwen Kong
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Chuwei Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Likun Gong
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jing Chen
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China.
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
40
|
Garcia-Arguinzonis M, Diaz-Riera E, Peña E, Escate R, Juan-Babot O, Mata P, Badimon L, Padro T. Alternative C3 Complement System: Lipids and Atherosclerosis. Int J Mol Sci 2021; 22:ijms22105122. [PMID: 34066088 PMCID: PMC8151937 DOI: 10.3390/ijms22105122] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Familial hypercholesterolemia (FH) is increasingly associated with inflammation, a phenotype that persists despite treatment with lipid lowering therapies. The alternative C3 complement system (C3), as a key inflammatory mediator, seems to be involved in the atherosclerotic process; however, the relationship between C3 and lipids during plaque progression remains unknown. The aim of the study was to investigate by a systems biology approach the role of C3 in relation to lipoprotein levels during atherosclerosis (AT) progression and to gain a better understanding on the effects of C3 products on the phenotype and function of human lipid-loaded vascular smooth muscle cells (VSMCs). By mass spectrometry and differential proteomics, we found the extracellular matrix (ECM) of human aortas to be enriched in active components of the C3 complement system, with a significantly different proteomic signature in AT segments. Thus, C3 products were more abundant in AT-ECM than in macroscopically normal segments. Furthermore, circulating C3 levels were significantly elevated in FH patients with subclinical coronary AT, evidenced by computed tomographic angiography. However, no correlation was identified between circulating C3 levels and the increase in plaque burden, indicating a local regulation of the C3 in AT arteries. In cell culture studies of human VSMCs, we evidenced the expression of C3, C3aR (anaphylatoxin receptor) and the integrin αMβ2 receptor for C3b/iC3b (RT-PCR and Western blot). C3mRNA was up-regulated in lipid-loaded human VSMCs, and C3 protein significantly increased in cell culture supernatants, indicating that the C3 products in the AT-ECM have a local vessel-wall niche. Interestingly, C3a and iC3b (C3 active fragments) have functional effects on VSMCs, significantly reversing the inhibition of VSMC migration induced by aggregated LDL and stimulating cell spreading, organization of F-actin stress fibers and attachment during the adhesion of lipid-loaded human VSMCs. This study, by using a systems biology approach, identified molecular processes involving the C3 complement system in vascular remodeling and in the progression of advanced human atherosclerotic lesions.
Collapse
MESH Headings
- Adult
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Case-Control Studies
- Cell Adhesion
- Cells, Cultured
- Complement C3/metabolism
- Female
- Humans
- Hyperlipoproteinemia Type II/immunology
- Hyperlipoproteinemia Type II/metabolism
- Hyperlipoproteinemia Type II/pathology
- Male
- Middle Aged
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proteome/analysis
- Proteome/metabolism
- Vascular Remodeling
- Wound Healing
- Young Adult
Collapse
Affiliation(s)
- Maisa Garcia-Arguinzonis
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
| | - Elisa Diaz-Riera
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
| | - Esther Peña
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael Escate
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oriol Juan-Babot
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, 28010 Madrid, Spain;
| | - Lina Badimon
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program-ICCC, Research Institute-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain; (M.G.-A.); (E.D.-R.); (E.P.); (R.E.); (O.J.-B.); (L.B.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-935-565-886; Fax: +34-935-565-559
| |
Collapse
|
41
|
Giglio RV, Pantea Stoian A, Al-Rasadi K, Banach M, Patti AM, Ciaccio M, Rizvi AA, Rizzo M. Novel Therapeutical Approaches to Managing Atherosclerotic Risk. Int J Mol Sci 2021; 22:4633. [PMID: 33924893 PMCID: PMC8125277 DOI: 10.3390/ijms22094633] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a multifactorial vascular disease that leads to inflammation and stiffening of the arteries and decreases their elasticity due to the accumulation of calcium, small dense Low Density Lipoproteins (sdLDL), inflammatory cells, and fibrotic material. A review of studies pertaining to cardiometabolic risk factors, lipids alterations, hypolipidemic agents, nutraceuticals, hypoglycaemic drugs, atherosclerosis, endothelial dysfunction, and inflammation was performed. There are several therapeutic strategies including Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) inhibitors, inclisiran, bempedoic acid, Glucagon-Like Peptide-1 Receptor agonists (GLP-1 RAs), and nutraceuticals that promise improvement in the atheromatous plaque from a molecular point of view, because have actions on the exposure of the LDL-Receptor (LDL-R), on endothelial dysfunction, activation of macrophages, on lipid oxidation, formations on foam cells, and deposition extracellular lipids. Atheroma plaque reduction both as a result of LDL-Cholesterol (LDL-C) intensive lowering and reducing inflammation and other residual risk factors is an integral part of the management of atherosclerotic disease, and the use of valid therapeutic alternatives appear to be appealing avenues to solving the problem.
Collapse
Affiliation(s)
- Rosaria Vincenza Giglio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (R.V.G.); (M.C.)
| | - Anca Pantea Stoian
- Diabetes, Nutrition and Metabolic Diseases Department, Faculty of General Medicine, Carol Davila University, 050474 Bucharest, Romania;
| | - Khalid Al-Rasadi
- Medical Research Centre, Sultan Qaboos University, Muscat 123, Oman;
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, 90-419 Lodz, Poland;
- Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Angelo Maria Patti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90133 Palermo, Italy;
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (R.V.G.); (M.C.)
- Department of Laboratory Medicine, University-Hospital, 90127 Palermo, Italy
| | - Ali A. Rizvi
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University, Atlanta, GA 30322, USA;
- Division of Endocrinology, Diabetes and Metabolism, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Manfredi Rizzo
- Diabetes, Nutrition and Metabolic Diseases Department, Faculty of General Medicine, Carol Davila University, 050474 Bucharest, Romania;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90133 Palermo, Italy;
- Division of Endocrinology, Diabetes and Metabolism, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
42
|
Ma B, Wang X, Zhang R, Niu S, Rong Z, Ni L, Di X, Han Q, Liu C. Cigarette smoke extract stimulates PCSK9 production in HepG2 cells via ROS/NF‑κB signaling. Mol Med Rep 2021; 23:331. [PMID: 33760160 PMCID: PMC7974406 DOI: 10.3892/mmr.2021.11970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/19/2021] [Indexed: 12/14/2022] Open
Abstract
Cigarette smoke (CS) exposure is a risk factor for dyslipidemia and atherosclerosis. Reduced expression of low-density lipoprotein receptor (LDLR) in hepatocytes may be one of the underlying mechanisms for these disorders. The aim of the present study was to investigate the molecular mechanism underlying the regulatory effect of CS extract (CSE) on proprotein convertase subtilisin/kexin type 9 (PCSK9) and low LDLR expression in HepG2 cells. PCSK9 and LDLR mRNA and protein expression levels in HepG2 cells were evaluated after CSE treatment via reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. In addition, total intracellular reactive oxygen species (ROS) production was determined via 2,7-dichlorofluorescein diacetate fluorescence. CSE significantly increased PCSK9 expression and inhibited LDLR expression in a time- and concentration-dependent manner. Furthermore, CSE significantly induced ROS production and nuclear factor κB (NF-κB) activation. However, pretreatment with a ROS scavenger or an NF-κB inhibitor significantly attenuated the CSE-induced changes in PCSK9 and LDLR expression. In addition, pretreatment with melatonin markedly reduced ROS production, NF-κB activation and PCSK9 expression, and increased LDLR expression in the CSE-treated cells. These data suggest that melatonin inhibits CSE-regulated PCSK9 and LDLR production in HepG2 cells via ROS/NF-κB signaling.
Collapse
Affiliation(s)
- Baitao Ma
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xuebin Wang
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Rui Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Niu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Zhihua Rong
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Leng Ni
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xiao Di
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Qin Han
- Center of Excellence in Tissue Engineering, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing 100005, P.R. China
| | - Changwei Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
43
|
Sá ACMGND, Machado ÍE, Bernal RTI, Malta DC. Factors associated with high LDL-Cholesterol in the Brazilian adult population: National Health Survey. CIENCIA & SAUDE COLETIVA 2021; 26:541-553. [PMID: 33605332 DOI: 10.1590/1413-81232021262.37102020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 01/21/2023] Open
Abstract
The study analyzed factors associated with high LDL-Cholesterol in Brazilian population. This is a cross-sectional study with laboratory data from 8,534 individuals collected in National Health Survey were analyzed. The prevalence levels of LDL-Cholesterol <130 and ≥ 130 mg/dL were calculated. The outcome variable was high LDL-Cholesterol (≥ 130 mg/dL) and explanatory variables were sociodemographic, anthropometric, lifestyle, chronic diseases and self-rated health. To Poisson regression was used and estimated prevalence ratios (PR) with 95% confidence levels (CI) to verify associations. The prevalence of high LDL-Cholesterol was 18.58%. In the final multivariate model were associated with the outcome: 30 to 44 years (PR 1.99; CI 1.58-2.54), 45 to 59 years (PR 2.89; CI 2.29-3.64), 60 years or more (PR 2.90; CI 2.29-3.68), living in the Northeast Region (PR 1.16; CI 1.02 - 1.32), overweight (PR 1.32; CI 1.15-1.51), obesity (PR 1.41; CI 1.19-1.65) or anemia (PR 0.66; CI 0.54-0.80). The LDL-Cholesterol was associated with aging, overweight, obesity, live in the Northeast and anemia. The monitoring of LDL levels is relevant, due to the increased risk with age, and can guide the adopting healthy lifestyles and diagnosis in places with lower access.
Collapse
Affiliation(s)
- Ana Carolina Micheletti Gomide Nogueira de Sá
- Programa de Pós-Graduação em Enfermagem, Escola de Enfermagem, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena 190, Santa Efigênia. 30130-100 Belo Horizonte MG Brasil.
| | - Ísis Eloah Machado
- Departamento de Medicina de Família, Saúde Mental e Coletiva, Escola de Medicina, Universidade Federal de Ouro Preto. Ouro Preto MG Brasil
| | - Regina Tomie Ivata Bernal
- Programa de Pós-Graduação em Enfermagem, Escola de Enfermagem, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena 190, Santa Efigênia. 30130-100 Belo Horizonte MG Brasil.
| | - Deborah Carvalho Malta
- Departamento de Enfermagem Materno-Infantil e Saúde Pública, Escola de Enfermagem, UFMG. Belo Horizonte MG Brasil
| |
Collapse
|
44
|
Buscemi S, Corleo D, Buscemi C, Randazzo C, Borzì AM, Barile AM, Rosafio G, Ciaccio M, Caldarella R, Meli F, Maestri S, Currenti W, Cincione RI, Murabito P, Galvano F. Influence of Habitual Dairy Food Intake on LDL Cholesterol in a Population-Based Cohort. Nutrients 2021; 13:593. [PMID: 33670170 PMCID: PMC7916907 DOI: 10.3390/nu13020593] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/23/2021] [Accepted: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cholesterol has a pivotal role in human physiology, exerting both structural and functional activity. However, higher blood cholesterol levels, especially low-density lipoprotein cholesterol (LDL-C), are a major cardiovascular risk factor. Therefore, special attention has been given to the effect of dietary factors in influencing LDL-C blood levels. In particular, much research has focused on dairy products, since they are a main component of different dietary patterns worldwide. A large body of evidence did not support the hypothesis that dairy products significantly increase circulating LDL-C, but no definitive data are available. Hence, we aimed to assess the relationships among LDL-C, habitual dairy food intake and anthropometric variables in a cohort representative of the general population in a Mediterranean area. METHODS We evaluated 802 healthy adults included in the ABCD_2 (Alimentazione, Benessere Cardiovascolare e Diabete) study (ISRCTN15840340), a longitudinal observational single-center study of a cohort representative of the general population of Palermo, Sicily. The habitual intake of dairy products was assessed with a validated food frequency questionnaire, and LDL-C serum levels and several anthropometric parameters were measured. RESULTS The group with high LDL-C serum concentrations (≥130 vs. <130 mg/dL) exhibited higher age, body mass index (BMI), waist-to-hip ratio (WHR), body fat percentage, systolic and diastolic blood pressure, carotid intima-media thickness and glycated hemoglobin. The habitual diet was not different between the groups in terms of macronutrient, cholesterol, egg and dairy food intake, with the exception of the weekly number of portions of milk (higher in the low LDL-C group vs. the high LDL-C group) and ricotta cheese (higher in the high LDL-C group vs. the LDL-C group). No significant correlation was found between LDL-C blood levels and the habitual intake of dairy products or the dietary intake of cholesterol and fats. The multivariate regression analyses (R2 = 0.94) showed that LDL-C blood levels were significantly associated with the habitual intake of milk (p < 0.005) and ricotta cheese (p < 0.001) and with BMI (p < 0.001). CONCLUSION Our study reported that total dairy food consumption was not correlated with LDL-C blood levels. However, multivariate analyses showed an inverse association between serum LDL-C and milk intake as well as a positive association between ricotta cheese intake and LDL-C concentrations. More studies are needed to better characterize the relationship between dairy products and circulating LDL-C.
Collapse
Affiliation(s)
- Silvio Buscemi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Davide Corleo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Carola Buscemi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Cristiana Randazzo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Antonio Maria Borzì
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Anna Maria Barile
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Giuseppe Rosafio
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Clinical Nutrition, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Marcello Ciaccio
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata (BIND), University of Palermo, I-90127 Palermo, Italy;
- Unit of Laboratory Medicine, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Rosalia Caldarella
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Laboratory Medicine, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Francesco Meli
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Laboratory Medicine, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Salvatore Maestri
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, I-90127 Palermo, Italy; (D.C.); (C.B.); (C.R.); (A.M.B.); (A.M.B.); (G.R.); (R.C.); (F.M.); (S.M.)
- Unit of Laboratory Medicine, AOU Policlinico “P. Giaccone”, I-90127 Palermo, Italy
| | - Walter Currenti
- Dipartimento di Scienze Biomediche e Biotecnologiche, University of Catania, I-95100 Catania, Italy; (W.C.); (F.G.)
| | - Raffaele Ivan Cincione
- Dipartimento di Medicina Clinica e Sperimentale, University of Foggia, I-71100 Foggia, Italy;
| | - Paolo Murabito
- Dipartimento di Chirurgia Generale e Specialità Medico-Chirurgiche, University of Catania, I-95100 Catania, Italy;
| | - Fabio Galvano
- Dipartimento di Scienze Biomediche e Biotecnologiche, University of Catania, I-95100 Catania, Italy; (W.C.); (F.G.)
| |
Collapse
|
45
|
Miao YF, Gao XN, Xu DN, Li MC, Gao ZS, Tang ZH, Mhlambi NH, Wang WJ, Fan WT, Shi XZ, Liu GL, Song SQ. Protective effect of the new prepared Atractylodes macrocephala Koidz polysaccharide on fatty liver hemorrhagic syndrome in laying hens. Poult Sci 2021; 100:938-948. [PMID: 33518147 PMCID: PMC7858188 DOI: 10.1016/j.psj.2020.11.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/10/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023] Open
Abstract
Fatty liver hemorrhage syndrome (FLHS) is the most common noninfectious cause of death in backyard chickens worldwide, which can cause a sudden drop in egg production in the affected flocks and cause huge losses to the laying hens breeding industry. In this study, we prepared polysaccharide from Atractylodes macrocephala Koidz (PAMK) by one-step alcohol precipitation. The structural analysis found that PAMK with a molecular weight of 2.816 × 103 Da was composed of glucose and mannose, in a molar ratio of 0.582 to 0.418. Furthermore, we investigated the hepatoprotective effects of PAMK on high-energy and low-protein (HELP) diet-induced FLHS in laying hens. The results showed that the hens' livers of the HELP diet showed yellowish-brown, greasy, and soft, whereas the supplement of PAMK (200 mg/kg or 400 mg/kg) could alleviate such pathological changes. The liver index, the abdominal fat percentage, and liver injury induced by the HELP diet were reduced in PAMK (200 mg/kg or 400 mg/kg). Supplementing 200 mg/kg or 400 mg/kg PAMK showed improvements of the antioxidant capacity in laying hens. Furthermore, we found that the HELP diet increased the expression of hepatic lipogenesis genes and decreased the expression of fatty acid β-oxidation genes, which could be reversed by 200 mg/kg or 400 mg/kg PAMK supplementation. Nevertheless, there is no difference between the addition of 40 mg/kg PAMK and the HELP group. Collectively, these results showed that PAMK supplements could ameliorate HELP diet-induced liver injury through regulating activities of antioxidant enzymes and hepatic lipid metabolism. Therefore, PAMK could be a potential feedstuff additive to alleviate FLHS in laying hens.
Collapse
Affiliation(s)
- Y F Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - X N Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - D N Xu
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - M C Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Z S Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Z H Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - N H Mhlambi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - W J Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - W T Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - X Z Shi
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - G L Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - S Q Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
46
|
Abstract
Atherosclerotic cardiovascular disease (ASCVD) proceeds through a series of stages: initiation, progression (or regression), and complications. By integrating known biology regarding molecular signatures of each stage with recent advances in high-dimensional molecular data acquisition platforms (to assay the genome, epigenome, transcriptome, proteome, metabolome, and gut microbiome), snapshots of each phase of atherosclerotic cardiovascular disease development can be captured. In this review, we will summarize emerging approaches for assessment of atherosclerotic cardiovascular disease risk in humans using peripheral blood molecular signatures and molecular imaging approaches. We will then discuss the potential (and challenges) for these snapshots to be integrated into a personalized movie providing dynamic readouts of an individual's atherosclerotic cardiovascular disease risk status throughout the life course.
Collapse
Affiliation(s)
- Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Kemar J. Brown
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ramachandran S. Vasan
- Sections of Preventive Medicine & Epidemiology, and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA; Department of Epidemiology, Boston University School of Public Health; Boston University Center for Computing and Data Sciences
| |
Collapse
|
47
|
Lowhalidanon K, Khunkaewla P. Discrimination between minimally modified LDL and fully oxidized LDL using monoclonal antibodies. Anal Biochem 2021; 619:114103. [PMID: 33453163 DOI: 10.1016/j.ab.2021.114103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 11/26/2022]
Abstract
Low density lipoprotein (LDL) can be oxidized in a stepwise process that leads to the production of minimally modified low density lipoprotein (mm-LDL), in which only the lipid component is oxidized, and then of fully oxidized LDL (oxLDL), in which both the lipids and the protein are oxidized. The thiobarbituric acid-reactive substances (TBARS) assay is a recognized method for determination of oxidized LDL, however this method is unable to distinguish between mm-LDL and oxLDL. In this study, seven specific monoclonal antibodies (mAbs) against human LDL were generated and selectively bound to the apolipoprotein B-100 (apoB-100) component of LDL. Oxidized LDL was produced by incubation of human LDL with 10 μM CuSO4 for various times. The TBARS assay revealed that the optimal incubation time to achieve maximal lipid oxidation was 9 h. Indirect ELISA using the newly generated mAbs was implemented to differentiate between mm-LDL and oxLDL and it was found that binding of the mAbs to oxLDL was significantly decreased after 48 h of incubation, reflecting the oxidative modification of apoB-100. Our results suggest that the optimal times for incubation of LDL with CuSO4 for generation of mm-LDL and oxLDL were 9 h and 48 h, respectively.
Collapse
Affiliation(s)
- Kanokwan Lowhalidanon
- Biochemistry-Electrochemistry Research Unit, School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Panida Khunkaewla
- Biochemistry-Electrochemistry Research Unit, School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand.
| |
Collapse
|
48
|
Xue NY, Ge DY, Dong RJ, Kim HH, Ren XJ, Tu Y. Effect of electroacupuncture on glial fibrillary acidic protein and nerve growth factor in the hippocampus of rats with hyperlipidemia and middle cerebral artery thrombus. Neural Regen Res 2021; 16:137-142. [PMID: 32788468 PMCID: PMC7818884 DOI: 10.4103/1673-5374.286973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Electroacupuncture (EA) has been shown to reduce blood lipid level and improve cerebral ischemia in rats with hyperlipemia complicated by cerebral ischemia. However, there are few studies on the results and mechanism of the effect of EA in reducing blood lipid level or promoting neural repair after stroke in hyperlipidemic subjects. In this study, EA was applied to a rat model of hyperlipidemia and middle cerebral artery thrombosis and the condition of neurons and astrocytes after hippocampal injury was assessed. Except for the normal group, rats in other groups were fed a high-fat diet throughout the whole experiment. Hyperlipidemia models were established in rats fed a high-fat diet for 6 weeks. Middle cerebral artery thrombus models were induced by pasting 50% FeCl3 filter paper on the left middle cerebral artery for 20 minutes on day 50 as the model group. EA1 group rats received EA at bilateral ST40 (Fenglong) for 7 days before the thrombosis. Rats in the EA1 and EA2 groups received EA at GV20 (Baihui) and bilateral ST40 for 14 days after model establishment. Neuronal health was assessed by hematoxylin-eosin staining in the brain. Hyperlipidemia was assessed by biochemical methods that measured total cholesterol, triglyceride, low-density lipoprotein and high-density lipoprotein in blood sera. Behavioral analysis was used to confirm the establishment of the model. Immunohistochemical methods were used to detect the expression of glial fibrillary acidic protein and nerve growth factor in the hippocampal CA1 region. The results demonstrated that, compared with the model group, blood lipid levels significantly decreased, glial fibrillary acidic protein immunoreactivity was significantly weakened and nerve growth factor immunoreactivity was significantly enhanced in the EA1 and EA2 groups. The repair effect was superior in the EA1 group than in the EA2 group. These findings confirm that EA can reduce blood lipid, inhibit glial fibrillary acidic protein expression and promote nerve growth factor expression in the hippocampal CA1 region after hyperlipidemia and middle cerebral artery thrombosis. All experimental procedures and protocols were approved by the Animal Use and Management Committee of Beijing University of Chinese Medicine, China (approval No. BUCM-3-2018022802-1002) on April 12, 2018.
Collapse
Affiliation(s)
- Na-Ying Xue
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Dong-Yu Ge
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Rui-Juan Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hyung-Hwan Kim
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Xiu-Jun Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ya Tu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
49
|
de Leeuw FA, Karamujić-Čomić H, Tijms BM, Peeters CFW, Kester MI, Scheltens P, Ahmad S, Vojinovic D, Adams HHH, Hankemeier T, Bos D, van der Lugt A, Vernooij MW, Ikram MA, Amin N, Barkhof F, Teunissen CE, van Duijn CM, van der Flier WM. Circulating metabolites are associated with brain atrophy and white matter hyperintensities. Alzheimers Dement 2020; 17:205-214. [PMID: 32886448 PMCID: PMC7984157 DOI: 10.1002/alz.12180] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 07/25/2020] [Accepted: 08/08/2020] [Indexed: 01/01/2023]
Abstract
Introduction Our aim was to study whether systemic metabolites are associated with magnetic resonance imaging (MRI) measures of brain and hippocampal atrophy and white matter hyperintensities (WMH). Methods We studied associations of 143 plasma‐based metabolites with MRI measures of brain and hippocampal atrophy and WMH in three independent cohorts (n = 3962). We meta‐analyzed the results of linear regression analyses to determine the association of metabolites with MRI measures. Results Higher glucose levels and lower levels of three small high density lipoprotein (HDL) particles were associated with brain atrophy. Higher glucose levels were associated with WMH. Discussion Glucose levels were associated with brain atrophy and WMH, and small HDL particle levels were associated with brain atrophy. Circulating metabolites may aid in developing future intervention trials.
Collapse
Affiliation(s)
- Francisca A de Leeuw
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hata Karamujić-Čomić
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Carel F W Peeters
- Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maartje I Kester
- Department of Neurology, Flevoziekenhuis, Almere, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hieab H H Adams
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Thomas Hankemeier
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, Leiden, The Netherlands.,Translational Epidemiology, Faculty Science, Leiden University, Leiden, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Institutes of Neurology & Healthcare Engineering, UCL London, London, UK
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Wang M, Liu M, Li F, Guo C, Liu Z, Pan Y, Liu Y, Liu F, Cai H, Wu Y, He Z, Ke Y. Gender heterogeneity in dyslipidemia prevalence, trends with age and associated factors in middle age rural Chinese. Lipids Health Dis 2020; 19:135. [PMID: 32532299 PMCID: PMC7291723 DOI: 10.1186/s12944-020-01313-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 06/04/2020] [Indexed: 11/22/2022] Open
Abstract
Background Heterogeneity should be carefully addressed to facilitate establishment of effective population-level blood lipid management. The primary aim of the study was to investigate gender heterogeneity in prevalence of dyslipidemia, including trends with age and associated factors in middle age rural Chinese. Methods This is a cross-sectional study based on a baseline investigation of a population-based randomized controlled trial in rural China, involving 26,378 permanent residents of age 45–69. The age-specific prevalence of dyslipidemia was estimated for men and women, and the trends of prevalence with age were compared. Logistic regression was used to explore the factors associated with prevalent risk of dyslipidemia. Results The overall prevalence of dyslipidemia was significantly higher in females than in males for borderline high and above (BHA) total cholesterol (TC ≥ 200 mg/dL), BHA triglycerides (TG ≥ 150 mg/dL) and BHA low-density lipoprotein cholesterol (LDL-C ≥ 130 mg/dL), but was lower for low high-density lipoprotein cholesterol (HDL-C < 40 mg/dL) in females than the corresponding prevalence in males. The prevalence of borderline high and above TC, TG and LDL-C all rose with age in females, but was stable or even decreased with age in males. In contrast, graphic representation of the prevalence of low HDL-C showed no striking age related trend in both genders. Risk of dyslipidemia was associated predominantly with obesity in males, but was more predominantly associated with hypertension in females. Conclusion Heterogeneity was found in comparing the prevalence of dyslipidemia in men and women, and gender heterogeneity was found in its trend with age and associated factors in middle aged rural Chinese. The effectiveness of population-level blood lipid management and CVD primary prevention programs in China is expected to be improved if gender heterogeneity is considered.
Collapse
Affiliation(s)
- Minmin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fenglei Li
- Hua County People's Hospital, Anyang, China
| | - Chuanhai Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yangfeng Wu
- Peking University Clinical Research Institute, Beijing, China
| | - Zhonghu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|