1
|
Wulff AB, Nordestgaard BG. Genetics of remnant cholesterol. Curr Opin Lipidol 2025:00041433-990000000-00119. [PMID: 40277396 DOI: 10.1097/mol.0000000000000991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
PURPOSE OF REVIEW Remnant cholesterol is receiving increasing attention as a target to reduce residual atherosclerotic cardiovascular disease (ASCVD) risk in individuals already treated with statins. New therapeutic options as antisense oligonucleotides, small interfering RNA, and monoclonal antibodies allow specific targeting of genes and proteins to counter pathological pathways promoted by these genes. Identifying genetic determinants of remnant cholesterol and relating these to risk of ASCVD is thus an appealing path to identifying and evaluating new and existing drug targets. RECENT FINDINGS Human genetic epidemiology has identified several genetic variants in genes involved in lipoprotein metabolism with effect on plasma concentrations of remnant cholesterol. Lipoprotein lipase (LPL) is central to the metabolism of remnant lipoproteins and plasma concentrations of remnant cholesterol, and several genes, including APOC3, ANGPTL3 and ANGPTL4, whose gene products regulate activity of LPL, are important determinants of remnant cholesterol. SUMMARY Current opinion is that remnant cholesterol is a likely causal factor in the development of ASCVD. Human genetic studies have identified several genes, many involved in LPL function, affecting remnant cholesterol concentrations, some of which are already used as therapeutic targets, and others which are subject to investigation of their remnant cholesterol and triglyceride-lowering effect in clinical trials.
Collapse
Affiliation(s)
- Anders B Wulff
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Sällström T, Bärebring L, Hulander E, Gjertsson I, Winkvist A, Lindqvist HM. Inflammatory and lipemic response to red meat intake in women with and without Rheumatoid Arthritis: a single meal study within a randomized controlled trial. BMC Nutr 2025; 11:74. [PMID: 40217385 PMCID: PMC11987391 DOI: 10.1186/s40795-025-01055-9] [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: 01/29/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The risk of atherosclerotic cardiovascular disease (ASCVD) is increased in Rheumatoid Arthritis (RA). Previous research has suggested that lipid metabolism is altered in RA, but research under postprandial conditions is scarce. The aim of this study was to investigate whether women with RA have a different lipemic and inflammatory response to a mixed meal containing red meat compared to women without RA. METHODS Twenty-two women with RA, with modest disease activity, and 22 women without RA matched for age and body mass index (BMI) at the group level consumed a hamburger meal containing ca. 700 kcal (53 E% from fat, 27 E% from carbohydrate). Venous blood was sampled in the fasted state and after 30 min, 1, 2, 3 and 5 h and analysed for lipid species using nuclear magnetic resonance spectroscopy. Postprandial inflammation was measured by interleukin- 6 (IL- 6). The postprandial lipid response was calculated as the incremental area under the curve minimal value, and serial measurements were analysed by repeated measures analysis of variance. Lipid and inflammatory responses were compared by linear regression analysis, adjusted for age, BMI, physical activity, and baseline plasma concentration. RESULTS Plasma concentrations of IL- 6, triglycerides (TGs) and very low-density lipoprotein (VLDL) particles increased significantly after the meal compared to baseline within both groups, but no differences were observed between groups. However, the women with RA had a less pronounced response in cholesterol carried in VLDL particles (p = 0.03) and in TGs in the subfraction of VLDL particles with highest density (p = 0.03). No association was found between the response in TGs and IL- 6. CONCLUSION This study does not provide compelling evidence for any difference in the lipemic or inflammatory response in women with RA compared with age- and BMI-matched women without RA following ingestion of a mixed, high-fat meal containing red meat. The modest disease activity in women with RA should be considered when interpreting these findings. Subtle group differences found in the lipids carried by VLDL particles warrant further investigation. TRIAL REGISTRATION The PIRA (Postprandial Inflammation in Rheumatoid Arthritis) trial was registered 2020-01 - 28 at Clinicaltrials.gov (NCT04247009).
Collapse
Affiliation(s)
- Torsten Sällström
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, PO Box 459, Gothenburg, 405 30, Sweden
| | - Linnea Bärebring
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, PO Box 459, Gothenburg, 405 30, Sweden
| | - Erik Hulander
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, PO Box 459, Gothenburg, 405 30, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Winkvist
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, PO Box 459, Gothenburg, 405 30, Sweden
| | - Helen M Lindqvist
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, PO Box 459, Gothenburg, 405 30, Sweden.
| |
Collapse
|
3
|
Lin C, Xia M, Dai Y, Huang Q, Sun Z, Zhang G, Luo R, Peng Q, Li J, Wang X, Lin H, Gao X, Tang H, Shen X, Wang S, Jin L, Hao X, Zheng Y. Cross-ancestry analyses of Chinese and European populations reveal insights into the genetic architecture and disease implication of metabolites. CELL GENOMICS 2025; 5:100810. [PMID: 40118068 PMCID: PMC12008806 DOI: 10.1016/j.xgen.2025.100810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025]
Abstract
Differential susceptibilities to various diseases and corresponding metabolite variations have been documented across diverse ethnic populations, but the genetic determinants of these disparities remain unclear. Here, we performed large-scale genome-wide association studies of 171 directly quantifiable metabolites from a nuclear magnetic resonance-based metabolomics platform in 10,792 Han Chinese individuals. We identified 15 variant-metabolite associations, eight of which were successfully replicated in an independent Chinese population (n = 4,480). By cross-ancestry meta-analysis integrating 213,397 European individuals from the UK Biobank, we identified 228 additional variant-metabolite associations and improved fine-mapping precision. Moreover, two-sample Mendelian randomization analyses revealed evidence that genetically predicted levels of triglycerides in high-density lipoprotein were associated with a higher risk of coronary artery disease and that of glycine with a lower risk of heart failure in both ancestries. These findings enhance our understanding of the shared and specific genetic architecture of metabolites as well as their roles in complex diseases across populations.
Collapse
Affiliation(s)
- Chenhao Lin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Guoqing Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; National Genomics Data Center& Bio-Med Big Data Center, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai 200031, China
| | - Ruijin Luo
- Shanghai Southgene Technology Co., Ltd., Shanghai 201203, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinxi Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xiaofeng Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan University-the People's Hospital of Rugao Joint Research Institute of Longevity and Aging, Rugao, Jiangsu 226500, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xia Shen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, Guangdong 511400, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Li J, Hou D, Li J, Li R, Sun M. Association between the atherogenic index of plasma and the systemic immuno-inflammatory index using NHANES data from 2005 to 2018. Sci Rep 2025; 15:11245. [PMID: 40175471 PMCID: PMC11965486 DOI: 10.1038/s41598-025-96090-8] [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: 12/23/2024] [Accepted: 03/26/2025] [Indexed: 04/04/2025] Open
Abstract
The atherogenic index of plasma (AIP) is used to evaluate the risk of atherosclerosis, while the systemic immune-inflammation index (SII) measures inflammation. The AIP and SII are indicators used to predict diseases in various areas. This study aims to explore the relationship between AIP and SII. A cross-sectional study design was used to recruit 70,190 participants from the National Health and Nutrition Examination Survey (NHANES) conducted between 2005 and 2018, excluding AIP missing data, SII missing data, participants under 20 years of age, and participants with missing covariates to eventually include 8163 participants. We used weighted multiple linear regression analysis, trend test, smooth curve fitting and threshold effect analysis to examine the relationship between AIP and SII. Among the 8163 participants included in the study, the mean (± SD) age was 48.412 ± 16.842 years. The mean SII (± SD) for all participants was 519.910 ± 316.974. In a model adjusted for all covariates (Model 3), AIP showed a significant positive correlation with SII [β (95% CI) 32.497 (5.425, 59.569), P = 0.021]. The smooth curve fitting results of AIP and SII are an "inverted U-shape" non-linear relationship, and the inflection point is at AIP = 0.82. This positive association between AIP and SII was found only in females and participants under 50. Specifically, for females, the positive correlation between AIP and SII was linear [β (95% CI) 80.791 (44.625, 116.958); P < 0.001]. In participants under 50, the positive correlation between AIP and SII was [β (95% CI) 34.198 (3.087, 65.310); P = 0.034], and there was also an "inverted U-shape" non-linear relationship with an inflection point of AIP = 0.549. For participants aged 20-50 years and males, the smooth curve showed a "down-flat-down" non-linear relationship. There is a significant positive correlation between AIP and SII. A positive association between AIP and SII was observed exclusively in females and among participants under 50. Furthermore, AIP and SII demonstrated a nonlinear relationship that resembles an "inverted U-shape". These findings offer new insights into the prevention, treatment, and management of cardiovascular disease. However, further comprehensive cohort studies are necessary to validate the relationship between AIP and SII.
Collapse
Affiliation(s)
- Jiayu Li
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Dan Hou
- PLA Northern Theater Command General Hospital, Shenyang, Liaoning, China.
| | - Jiarong Li
- Shaoguan University, Shaoguan, Guangdong, China
| | - Rongcai Li
- Guangzhou Institute of Technology, Guangzhou, Guangdong, China
| | - Ming Sun
- PLA Northern Theater Command General Hospital, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Tramontano D, D'Erasmo L, Larouche M, Brisson D, Lauzière A, Di Costanzo A, Bini S, Minicocci I, Covino S, Baratta F, Pasquali M, Cerbelli B, Gaudet D, Arca M. The vicious circle of chronic kidney disease and hypertriglyceridemia: What is first, the hen or the egg? Atherosclerosis 2025; 403:119146. [PMID: 40056689 DOI: 10.1016/j.atherosclerosis.2025.119146] [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: 05/21/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
Chronic kidney disease (CKD) is documented to cause alterations in lipid metabolism, and this was considered a potent driver of increased cardiovascular risk. Among the diverse alteration of lipid traits in CKD, research endeavours have predominantly concentrated on low-density lipoproteins (LDL) in view of the potent pro-atherogenic role of these lipoprotein particles and the demonstration of protective cardiovascular effect of reducing LDL. However, few studies have focused on the metabolism of triglyceride-rich lipoproteins and even fewer on their role in causing kidney damage. Therefore, the comprehensive description of the impact of hypertriglyceridemia (HTG) in CKD pathophysiology remains largely undetermined. This reflects the difficulty of disentangling the independent role of triglycerides (TG) in the complex, bidirectional relationship between TG and kidney disease. Abnormal neutral lipid accumulation in the intrarenal vasculature and renal cells eventually due to HTG may also promote glomerular injury, throughout mechanisms including oxidative stress, mitochondrial dysfunction and proinflammatory responses. While epidemiological and experimental evidence suggests a potential role of TG in kidney damage, the causal mechanisms and their clinical relevance remain unclear, representing a significant area for future investigation. This review aims to highlight the intricate interplay between TG metabolism and kidney disease, shedding light on the mechanisms through which HTG may influence kidney functionality.
Collapse
Affiliation(s)
- Daniele Tramontano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy.
| | - Miriam Larouche
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical Research Center, Chicoutimi, QC, Canada
| | - Diane Brisson
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical Research Center, Chicoutimi, QC, Canada
| | - Alex Lauzière
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical Research Center, Chicoutimi, QC, Canada
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy
| | - Simone Bini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy
| | - Ilenia Minicocci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy
| | - Stella Covino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy
| | - Francesco Baratta
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Marzia Pasquali
- Department of Internal Medicine and Medical Specialities, Nephrology Unit, University Policlinico Umberto I Hospital, Rome, Italy
| | - Bruna Cerbelli
- Department of Medical-Surgical Sciences and Biotechnologies Sapienza University of Rome, Rome, Italy
| | - Daniel Gaudet
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical Research Center, Chicoutimi, QC, Canada
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell' Università 37, 00161, Rome, Italy
| |
Collapse
|
6
|
Do H, Kwon OC, Ha JW, Chung J, Park YB, Huh JH, Lee SW. Remnant Cholesterol Levels at Diagnosis May Predict Acute Coronary Syndrome Occurrence During Follow-Up in Patients with Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. J Clin Med 2025; 14:2260. [PMID: 40217710 PMCID: PMC11989813 DOI: 10.3390/jcm14072260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Previous studies have revealed the predictive potential of remnant cholesterol (RC) for acute coronary syndrome (ACS) occurrence in the general population. However, whether this association applies to patients with antineutrophil cytoplasmic antibody-associated vasculitis (AAV), in which a lipid paradox exists, remains unclear. We investigated whether RC levels at diagnosis could predict ACS occurrence during follow-up in patients with AAV. Methods: This study included 139 patients with AAV. ACS was defined as ST-elevation myocardial infarction (STEMI), non-STEMI, or unstable angina occurring after AAV diagnosis. RC levels were calculated as (total cholesterol)-(low-density lipoprotein cholesterol)-(high-density lipoprotein cholesterol). Patients were categorised into three groups by RC tertiles: highest (≥26.2 mg/dL), middle (19.1-26.1 mg/dL), and lowest (≤19.0 mg/dL) tertile groups. Results: The median age of the 139 patients (male, 31.7%) was 58.0 years. During follow-up, six, two, and one patients were diagnosed with ACS in the highest, middle, and lowest tertile groups, respectively. Patients in the highest tertile group exhibited a significantly lower ACS-free survival rate than those in the lowest tertile (p = 0.030). In the multivariable Cox hazards model, male sex (hazard ratio [HR] 9.054, 95% confidence interval [CI] 1.786-45.910), Birmingham vasculitis activity score (HR 1.147, 95% CI 1.033-1.274), and the highest tertile of RC levels (HR 10.818, 95% CI 1.867-62.689) were significantly and independently associated with ACS occurrence during follow-up in patients with AAV. Conclusions: Our findings indicate that RC levels at diagnosis may predict ACS occurrence during follow-up in patients with AAV, regardless of the traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Hyunsue Do
- Division of Rheumatology, Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon-si 24341, Republic of Korea;
| | - Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea;
| | - Jang Woo Ha
- Division of Rheumatology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Republic of Korea;
| | - Jihye Chung
- Division of Rheumatology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.C.); (Y.-B.P.)
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.C.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji Hye Huh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.C.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
7
|
Kumari A, Larsen SWR, Bondesen S, Qian Y, Tian HD, Walker SG, Davies BSJ, Remaley AT, Young SG, Konrad RJ, Jørgensen TJD, Ploug M. ANGPTL3/8 is an atypical unfoldase that regulates intravascular lipolysis by catalyzing unfolding of lipoprotein lipase. Proc Natl Acad Sci U S A 2025; 122:e2420721122. [PMID: 40112106 PMCID: PMC11962473 DOI: 10.1073/pnas.2420721122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
Lipoprotein lipase (LPL) carries out the lipolytic processing of triglyceride-rich lipoproteins (TRL) along the luminal surface of capillaries. LPL activity is regulated by the angiopoietin-like proteins (ANGPTL3, ANGPTL4, ANGPTL8), which control the delivery of TRL-derived lipid nutrients to tissues in a temporal and spatial fashion. This regulation of LPL mediates the partitioning of lipid delivery to adipose tissue and striated muscle according to nutritional status. A complex between ANGPTL3 and ANGPTL8 (ANGPTL3/8) inhibits LPL activity in oxidative tissues, but its mode of action has remained unknown. Here, we used biophysical techniques to define how ANGPTL3/8 and ANGPTL3 interact with LPL and how they drive LPL inactivation. We demonstrate, by mass photometry, that ANGPTL3/8 is a heterotrimer with a 2:1 ANGPTL3:ANGPTL8 stoichiometry and that ANGPTL3 is a homotrimer. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) studies revealed that ANGPTL3/8 and ANGPTL3 use the proximal portion of their N-terminal α-helices to interact with sequences surrounding the catalytic pocket in LPL. That binding event triggers unfolding of LPL's α/β-hydrolase domain and irreversible loss of LPL catalytic activity. The binding of LPL to its endothelial transporter protein (GPIHBP1) or to heparan-sulfate proteoglycans protects LPL from unfolding and inactivation, particularly against the unfolding triggered by ANGPTL3. Pulse-labeling HDX-MS studies revealed that ANGPTL3/8 and ANGPTL3 catalyze LPL unfolding in an ATP-independent fashion, which categorizes these LPL inhibitors as atypical unfoldases. The catalytic nature of LPL unfolding by ANGPTL3/8 explains why low plasma concentrations of ANGPTL3/8 are effective in inhibiting a molar excess of LPL in capillaries.
Collapse
Affiliation(s)
- Anni Kumari
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, Copenhagen NDK–2200, Denmark
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
| | - Sanne W. R. Larsen
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense MDK–5320, Denmark
| | - Signe Bondesen
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense MDK–5320, Denmark
| | - Yuewei Qian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN462585
| | - Hao D. Tian
- Laboratory of Lipoprotein Metabolism, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20814
| | - Sydney G. Walker
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa, IA52242
| | - Brandon S. J. Davies
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa, IA52242
| | - Alan T. Remaley
- Laboratory of Lipoprotein Metabolism, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20814
| | - Stephen G. Young
- Department of Medicine, University of California, Los Angeles, CA90095
- Department of Human Genetics, University of California, Los Angeles, CA90095
| | - Robert J. Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN462585
| | - Thomas J. D. Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense MDK–5320, Denmark
| | - Michael Ploug
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, Copenhagen NDK–2200, Denmark
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
| |
Collapse
|
8
|
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
|
9
|
Long H, Huang R, Zhu S, Wang Z, Liu X, Zhu Z. Polysaccharide from Caulerpa lentillifera alleviates hyperlipidaemia through altering bile acid metabolism mediated by gut microbiota. Int J Biol Macromol 2025; 306:141663. [PMID: 40044008 DOI: 10.1016/j.ijbiomac.2025.141663] [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: 10/07/2024] [Revised: 01/07/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Polysaccharide from Caulerpa lentillifera (CLP) offers preventative health benefits, but its efficacy against hyperlipidaemia and underlying mechanisms still elusive. This investigation assessed CLP's potential to mitigate high-fat diet (HFD)-induced hyperlipidaemia via the gut microbiota-bile acid (BA) axis. In hyperlipidaemic mice, 8 weeks of CLP treatment improved body weight, lipid profiles, and hepatic function, correlating with shifts in BA concentrations. Additionally, CLP not only repaired HFD-induced gut dysbiosis by increasing SCFA-producing bacteria but also positively modulated gut metabolites, including acetic and butyric acids. Spearman's correlation analysis illustrated strong associations between the altered microbes, metabolites, and the expression of genes involved in BA metabolism. Remarkably, CLP significantly influenced BA levels related to hyperlipidaemia, partly by augmenting the population of Parabacteroides and associated butyric acid level. These results indicate that CLP may serve as a functional food to guard against dyslipidaemia through impacting specific gut microbes and metabolites such as Parabacteroides and butyrate, and thus presenting promising therapeutic prospects for diseases associated with BA metabolism.
Collapse
Affiliation(s)
- Hairong Long
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239001, Anhui, PR China; College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, Guangxi, PR China
| | - Rui Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong, PR China
| | - Shuangjie Zhu
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239001, Anhui, PR China
| | - Zuhan Wang
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239001, Anhui, PR China
| | - Xiaoling Liu
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, Guangxi, PR China.
| | - Zhenjun Zhu
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong, PR China.
| |
Collapse
|
10
|
Qian Q, Wu J, Wang C, Yang Z, Cheng Y, Zheng Y, Wang X, Wang H. 6-PPD triggered lipid metabolism disorder and inflammatory response in larval zebrafish (Danio rerio) by regulating PPARγ/NF-κB pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125785. [PMID: 39900129 DOI: 10.1016/j.envpol.2025.125785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/05/2025]
Abstract
As a synthetic rubber antioxidant, the environmental monitoring concentrations of N-(1,3-Dimethylbutyl)-N'-phenyl-p-phenylenediamine (6-PPD) have exceeded the risk threshold, attracting widespread attention. Although investigations into the harmful effects on zebrafish have commenced, a comprehensive exploration of its toxicological impacts and underlying molecular mechanisms remains to be conducted. By using zebrafish as a model, this study systematically evaluated 6-PPD-induced lipid metabolism disorders and inflammation response following environmental exposure. Bioinformatics analysis revealed that 6-PPD target genes enriched in the hepatitis B pathway, indicating potential hepatic toxicity via inflammatory pathways. Therefore, we hypothesize that 6-PPD could trigger hepatotoxicity through the crosstalk between lipid metabolism and inflammation. Further experiments substantiated this hypothesis by showing lipid accumulation in the liver following 6-PPD exposure, along with elevated triglyceride (TG) and total cholesterol (TC) levels, and imbalanced expression of lipid metabolism-related marker genes. Additionally, 6-PPD exposure induced the accumulation of reactive oxygen species (ROS) and inhibited the differentiation and maturation of immune cells, resulting in immune evasion. Most of these abnormalities were exacerbated in a dose-dependent manner with increasing concentrations of 6-PPD. The addition of the PPARγ pathway agonist puerarin (PUE) or NF-κB pathway inhibitor quinazoline (QNZ) to 6-PPD exposure group mitigated these toxic effects, validating our conjecture that lipid metabolism disorder and inflammatory responses may result from the regulation of the PPARγ/NF-κB pathway.
Collapse
Affiliation(s)
- Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ji Wu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Cuizhen Wang
- Sanquan College of Xinxiang Medical University, Xinxiang, 453513, China
| | - Zheng Yang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Ying Cheng
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
11
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2025; 36:262-277. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [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: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
12
|
Dakal TC, Xiao F, Bhusal CK, Sabapathy PC, Segal R, Chen J, Bai X. Lipids dysregulation in diseases: core concepts, targets and treatment strategies. Lipids Health Dis 2025; 24:61. [PMID: 39984909 PMCID: PMC11843775 DOI: 10.1186/s12944-024-02425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/30/2024] [Indexed: 02/23/2025] Open
Abstract
Lipid metabolism is a well-regulated process essential for maintaining cellular functions and energy homeostasis. Dysregulation of lipid metabolism is associated with various conditions, including cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. This review explores the mechanisms underlying lipid metabolism, emphasizing the roles of key lipid species such as triglycerides, phospholipids, sphingolipids, and sterols in cellular physiology and pathophysiology. It also examines the genetic and environmental factors contributing to lipid dysregulation and the challenges of diagnosing and managing lipid-related disorders. Recent advancements in lipid-lowering therapies, including PCSK9 inhibitors, ezetimibe, bempedoic acid, and olpasiran, provide promising treatment options. However, these advancements are accompanied by challenges related to cost, accessibility, and patient adherence. The review highlights the need for personalized medicine approaches to address the interplay between genetics and environmental factors in lipid metabolism. As lipidomics and advanced diagnostic tools continue to progress, a deeper understanding of lipid-related disorders could pave the way for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, 313001, India
| | - Feng Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Chandra Kanta Bhusal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | | | - Rakesh Segal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Juan Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Xiaodong Bai
- Department of Plastic Surgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, China.
| |
Collapse
|
13
|
Paoli A. The Influence of Physical Exercise, Ketogenic Diet, and Time-Restricted Eating on De Novo Lipogenesis: A Narrative Review. Nutrients 2025; 17:663. [PMID: 40004991 PMCID: PMC11858292 DOI: 10.3390/nu17040663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
De novo lipogenesis (DNL) is a metabolic pathway that converts carbohydrates into fatty acids, primarily occurring in the liver and, to a lesser extent, in adipose tissue. While hepatic DNL is highly responsive to dietary carbohydrate intake and regulated by insulin via transcription factors like SREBP-1c, adipose DNL is more modest and less sensitive to dietary overfeeding. Dysregulated DNL contributes to metabolic disorders, including metabolic dysfunction-associated steatotic liver disease (MASLD). Lifestyle interventions, such as physical exercise, ketogenic diets, and time-restricted eating (TRE) offer promising strategies to regulate DNL and improve metabolic health. Physical exercise enhances glucose uptake in muscles, reduces insulin levels, and promotes lipid oxidation, thereby suppressing hepatic DNL. Endurance and resistance training also improve mitochondrial function, further mitigating hepatic triglyceride accumulation. Ketogenic diets shift energy metabolism toward fatty acid oxidation and ketogenesis, lower insulin, and directly downregulate lipogenic enzyme activity in the liver. TRE aligns feeding with circadian rhythms by optimizing AMP-activated protein kinase (AMPK) activation during fasting periods, which suppresses DNL and enhances lipid metabolism. The combined effects of these interventions demonstrate significant potential for improving lipid profiles, reducing hepatic triglycerides, and preventing lipotoxicity. By addressing the distinct roles of the liver and adipose DNL, these strategies target systemic and localized lipid metabolism dysregulation. Although further research is needed to fully understand their long-term impact, these findings highlight the transformative potential of integrating these approaches into clinical practice to manage metabolic disorders and their associated complications.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35100 Padua, Italy;
- Research Center for High Performance Sport, UCAM Catholic University of Murcia, 30107 Murcia, Spain
| |
Collapse
|
14
|
Sun Z, Torphy RJ, Miller EN, Darehshouri A, Vigil I, Terai T, Eck E, Sun Y, Guo Y, Yee EJ, Hu J, Kedl RM, Lasda EL, Hesselberth JR, MacLean P, Bruce KD, Randolph GJ, Schulick RD, Zhu Y. GPR182 is a lipoprotein receptor for dietary fat absorption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.634329. [PMID: 39975353 PMCID: PMC11838411 DOI: 10.1101/2025.02.03.634329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The lymphatic system plays a central role in lipid absorption, which transports chylomicrons from the small intestine to the circulation. However, the molecular mechanism by which chylomicrons get into the intestinal lymphatics is unknown. Here we demonstrated that GPR182, a receptor in lymphatic endothelial cells (LECs), mediates dietary fat absorption. GPR182 knockout mice are resistant to dietary-induced obesity. GPR182 ablation in mice leads to poor lipid absorption and thereby a delay in growth during development. GPR182 binds and endocytoses lipoproteins broadly. Mechanistically, loss of GPR182 prevents chylomicrons from entering the lacteal lumen of the small intestine. GPR182 blockage with a monoclonal antibody (mAb) protects mice from dietary induced obesity. Together, our study identifies GPR182 as a lipoprotein receptor that mediates dietary fat absorption.
Collapse
|
15
|
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
|
16
|
Biswas A, Arshid S, Kristensen KK, Jørgensen TJD, Ploug M. Competitive displacement of lipoprotein lipase from heparan sulfate is orchestrated by a disordered acidic cluster in GPIHBP1. J Lipid Res 2025; 66:100745. [PMID: 39814316 PMCID: PMC11869522 DOI: 10.1016/j.jlr.2025.100745] [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: 11/25/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Movement of lipoprotein lipase (LPL) from myocytes or adipocytes to the capillary lumen is essential for intravascular lipolysis and plasma triglyceride homeostasis-low LPL activity in the capillary lumen causes hypertriglyceridemia. The trans-endothelial transport of LPL depends on ionic interactions with GPIHBP1's intrinsically disordered N-terminal tail, which harbors two acidic clusters at positions 5-12 and 19-30. This polyanionic tail provides a molecular switch that controls LPL detachment from heparan sulfate proteoglycans (HSPGs) by competitive displacement. When the acidic tail was neutralized in gene-edited mice, LPL remained trapped in the sub-endothelial spaces triggering hypertriglyceridemia. Due to its disordered state, the crystal structure of LPL•GPIHBP1 provided no information on these electrostatic interactions between LPL and GPIHBP1 acidic tail. In the current study, we positioned the acidic tail on LPL using zero-length crosslinking. Acidic residues at positions 19-30 in GPIHBP1 mapped to Lys445, Lys441, Lys414, and Lys407 close to the interface between the C- and N-terminal domains in LPL. Modeling this interface revealed widespread polyelectrolyte interactions spanning both LPL domains, which explains why the acidic tail stabilizes LPL activity and protein conformation. In functional assays, we showed that the acidic cluster at 19-30 also had the greatest impact on preserving LPL activity, mitigating ANGPTL4-catalyzed LPL inactivation, preventing PSCK3-mediated LPL cleavage, and, importantly, displacing LPL from HSPGs. Our current study provides key insights into the biophysical mechanism(s) orchestrating intravascular compartmentalization of LPL activity-an intriguing pathway entailing competitive displacement of HSPG-bound LPL by a disordered acidic tail in GPIHBP1.
Collapse
Affiliation(s)
- Anamika Biswas
- Finsen Laboratory, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Samina Arshid
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Kristian Kølby Kristensen
- Finsen Laboratory, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Thomas J D Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Michael Ploug
- Finsen Laboratory, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
17
|
Arai K, Ono Y, Hirai N, Sugiura Y, Kaneko K, Matsuda S, Iio K, Kajino K, Saitoh T, Wei FY, Katagiri H, Inoue A. Chemogenetic activation of hepatic G 12 signaling ameliorates hepatic steatosis and obesity. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167566. [PMID: 39542224 DOI: 10.1016/j.bbadis.2024.167566] [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: 05/03/2024] [Revised: 09/20/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Hepatic steatosis, the early stage of nonalcoholic fatty liver disease (NAFLD), currently lacks targeted pharmacological treatments. G protein-coupled receptors (GPCRs) in hepatocytes differentially regulate lipid metabolism depending on their coupling profile of G protein subtypes. Unlike Gs, Gi, and Gq signaling, the role of G12 signaling in hepatic steatosis remains elusive. The objective of this study was to investigate the effect of G12 signaling on hepatic steatosis and obesity and its mechanisms. METHODS We generated mice expressing a G12-coupled designer GPCR in a liver-specific manner. We performed phenotypic analysis in the mice under the condition of fasting (acute hepatic steatosis model) or high-fat diet feeding (chronic hepatic steatosis model). RESULTS In acute and chronic hepatic steatosis models, chemogenetic activation of hepatic G12 signaling suppressed the progression of hepatic steatosis. The treatment led to an increased triglyceride secretion with little effect on mitochondrial respiratory activity, fatty acid oxidation, de novo lipogenesis, and fatty acid uptake. Furthermore, in a high-fat-diet-induced obesity model, activation of the G12-coupled designer GPCR exerted anti-obesity effects with increased whole-body energy expenditure and fat oxidation. Anti-FGF21 antibody treatment showed that the anti-obesity effects of the hepatic G12D activation relied in part on the hepatokine FGF21. CONCLUSIONS Our findings indicate that the activation of G12 signaling in the liver has the potential to prevent hepatic steatosis and obesity. This discovery provides a strong rationale for the development of drugs targeting G12-coupled GPCRs expressed in the liver.
Collapse
Affiliation(s)
- Kaito Arai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Yuki Ono
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Natsumi Hirai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology (CCII), Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan; Human Biology Microbiome Quantum Research Center (WPI-Bio2Q), Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Shigeru Matsuda
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Keita Iio
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8571, Japan
| | - Keita Kajino
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8571, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Fan-Yan Wei
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
18
|
Li X, Li ZF, Wu NQ. Remnant Cholesterol and Residual Risk of Atherosclerotic Cardiovascular Disease. Rev Cardiovasc Med 2025; 26:25985. [PMID: 40026498 PMCID: PMC11868899 DOI: 10.31083/rcm25985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 03/05/2025] Open
Abstract
Remnant cholesterol (RC) is increasingly recognized as a key target in the treatment of atherosclerotic cardiovascular disease (ASCVD), addressing much of the residual risk that persists despite standard therapies. However, integrating RC into clinical practice remains challenging. Key issues, such as the development of accessible RC measurement methods, the identification of safe and effective medications, the determination of optimal target levels, and the creation of RC-based risk stratification strategies, require further investigation. This article explores the complex role of RC in ASCVD development, including its definition, metabolic pathways, and its association with both the overall risk and residual risk of ASCVD in primary and secondary prevention. It also examines the effect of current lipid-lowering therapies on RC levels and their influence on cardiovascular outcomes. Recent research has highlighted promising advancements in therapies aimed at lowering RC, which show potential for reducing major adverse cardiovascular events (MACEs). Inhibitors such as angiopoietin-like protein 3 (ANGPTL3), apolipoprotein C-III (apoCIII), and proprotein convertase subtilisin/kexin type 9 (PCSK9) have demonstrated their ability to modulate RC and reduce MACEs by targeting specific proteins involved in RC synthesis and metabolism. There is a pressing need for larger randomized controlled trials to clarify the role of RC in relevant patient populations. The development of targeted RC-lowering therapies holds the promise of significantly reducing the high rates of morbidity and mortality associated with ASCVD.
Collapse
Affiliation(s)
- Xi Li
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| | - Zhi-Fan Li
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| | - Na-Qiong Wu
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| |
Collapse
|
19
|
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
|
20
|
Su B, Fan Z, Wu J, Zhan H. Genetic association of lipid-lowering drug target genes with pancreatic cancer: a Mendelian randomization study. Sci Rep 2025; 15:3282. [PMID: 39863728 PMCID: PMC11762976 DOI: 10.1038/s41598-025-87490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Previous studies have found that dyslipidemia is a risk factor for pancreatic cancer (PC), and that lipid-lowering drugs may reduce the risk of PC. However, it is not clear whether dyslipidemia causes PC. The Mendelian randomization (MR) study aimed to investigate the causal role of lipid traits in pancreatic cancer and to assess the potential impact of lipid-lowering drug targets on pancreatic cancer. 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 PC were obtained from an independent GWAS datasets. Colocalization analyses were performed to validate the robustness of the results. No significant effect of lipid-lowering drug targets on PC risk was found. Genetic mimicry of lipoprotein lipase (LPL) was potentially associated with PC risks. Significant MR associations were observed in the discovery dataset (OR 1.64 [95% CI 1.24-2.16], p = 4.48*10-4) with PC in one dataset. However, the finding was not verified in the replication dataset. Our findings do not support dyslipidemia as a causal factor for PC. Among lipid-lowering drug targets, LPL is the potential drug target in PC.
Collapse
Affiliation(s)
- Bohan Su
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jiexi Wu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
21
|
Smith K, Dennis KMJH, Hodson L. The ins and outs of liver fat metabolism: The effect of phenotype and diet on risk of intrahepatic triglyceride accumulation. Exp Physiol 2025. [PMID: 39861959 DOI: 10.1113/ep092001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/22/2024] [Indexed: 01/27/2025]
Abstract
In health, the liver is a metabolically flexible organ that plays a key role in regulating systemic lipid and glucose concentrations. There is a constant flux of fatty acids (FAs) to the liver from multiple sources, including adipose tissue, dietary, endogenously synthesized from non-lipid precursors, intrahepatic lipid droplets and recycling of triglyceride-rich remnants. Within the liver, FAs are used for triglyceride synthesis, which can be oxidized, stored or secreted in very low-density lipoproteins into the systemic circulation. The processes of FA uptake, FA synthesis and the intracellular partitioning of FAs into storage, oxidation or secretory pathways are tightly regulated. An imbalance in these processes causes intrahepatic triglyceride to accumulate and is associated with the development of metabolic dysfunction-associated steatotic liver disease. It is well appreciated that many factors can influence intrahepatic FA partitioning, and although there is good evidence that both phenotype (e.g., sex, ethnicity and adiposity) and dietary macronutrient composition can play a role in intrahepatic triglyceride accumulation, their interaction remains poorly understood. The aim of this review is to explore how the respective pathways of FA delivery, synthesis and disposal are altered by phenotype and understand how dietary macronutrient composition might influence the partitioning of FAs in the liver in vivo, in humans.
Collapse
Affiliation(s)
- Kieran Smith
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Kaitlyn M J H Dennis
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
22
|
Li H, Ke X, Feng B, Tian H, Cai Z, Zhang A, Man Q. Research progress on the mechanism and markers of metabolic disorders in the occurrence and development of cognitive dysfunction after ischemic stroke. Front Endocrinol (Lausanne) 2025; 16:1500650. [PMID: 39911922 PMCID: PMC11794095 DOI: 10.3389/fendo.2025.1500650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025] Open
Abstract
Post-stroke cognitive impairment (PSCI) is a common complication following a stroke that significantly affects patients' quality of life and rehabilitation outcomes. It also imposes a heavy economic burden. There is an urgent need to better understand the pathophysiology and pathogenesis of PSCI, as well as to identify markers that can predict PSCI early in the clinical stage, facilitating early prevention, monitoring, and treatment. Although the mechanisms underlying PSCI are complex and multifaceted, involving factors such as atherosclerosis and neuroinflammation, metabolic disorders also play a critical role. This article primarily reviews the relationship between metabolic disorders of the three major nutrients-sugar, fat, and protein-and the development of cognitive dysfunction following ischemic stroke (IS). It aims to elucidate how these metabolic disturbances contribute to cognitive dysfunction post-stroke and to explore potential metabolic biomarkers for PSCI. We believe that this review will offer new insights into the early identification, treatment, and prognostic assessment of PSCI.
Collapse
Affiliation(s)
- Huaqiang Li
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bianying Feng
- Department of Clinical Laboratory, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huan Tian
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenzhen Cai
- Department of Clinical Laboratory, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
23
|
Li X, Xie E, Sun S, Shen J, Ding Y, Wang J, Peng X, Zheng R, Farag MA, Xiao J. Flavonoids for gastrointestinal tract local and associated systemic effects: A review of clinical trials and future perspectives. J Adv Res 2025:S2090-1232(25)00033-5. [PMID: 39798849 DOI: 10.1016/j.jare.2025.01.014] [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: 10/09/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Flavonoids are naturally occurring dietary phytochemicals with significant antioxidant effects aside from several health benefits. People often consume them in combination with other food components. Compiling data establishes a link between bioactive flavonoids and prevention of several diseases in animal models, including cardiovascular diseases, diabetes, gut dysbiosis, and metabolic dysfunction-associated steatotic liver disease (MASLD). However, numerous clinical studies have demonstrated the ineffectiveness of flavonoids contradicting rodent models, thereby challenging the validity of using flavonoids as dietary supplements. AIM OF REVIEW This review provides a clinical perspective to emphasize the effective roles of dietary flavonoids as well as to summarize their specific mechanisms in animals briefly. KEY SCIENTIFIC CONCEPTS OF REVIEW First, this review offers an in-depth elucidation of the metabolic processes of flavonoids within human, encompassing the small, large intestine, and the liver. Furthermore, the review provides a comprehensive overview of the various functions of flavonoids in the gastrointestinal tract, including hindering the breakdown and assimilation of macronutrients, such as polysaccharides and lipids, regulating gut hormone secretion as well as inhibition of mineral iron absorption. In the large intestine, an unabsorbed major portion of flavonoids interact with the gut flora leading to their biotransformation. Once absorbed and circulated in the bloodstream, bioactive flavonoids or their metabolites exert numerous beneficial systemic effects. Lastly, we examine the protective effects of flavonoids in several metabolic disorders, including endothelial dysfunction, MASLD, cardiovascular disease, obesity, hyperlipidemia, and insulin resistance. In conclusion, this review outlines the safety and future prospects of flavonoids in the field of health, especially in the prevention of metabolic syndrome (MetS).
Collapse
Affiliation(s)
- Xiaopeng Li
- Center of Nutrition and Food Sciences Hunan Agricultural Products Processing Institute Hunan Academy of Agricultural Sciences Changsha China.
| | - Enjun Xie
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Shumin Sun
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Jie Shen
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Yujin Ding
- National Clinical Research Center for Metabolic Diseases Metabolic Syndrome Research Center Department of Metabolism and Endocrinology The Second Xiangya Hospital of Central South University Changsha China
| | - Jiaqi Wang
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Xiaoyu Peng
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Ruting Zheng
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Mohamed A Farag
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562 Egypt
| | - Jianbo Xiao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI 36310 Vigo, Spain; Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21 39011 Santander, Spain.
| |
Collapse
|
24
|
Li Z, Yu C, Zhang H, Chen R, Zhao Y, Zheng Z. Impact of remnant cholesterol on short-term and long-term prognosis in patients with prediabetes or diabetes undergoing coronary artery bypass grafting: a large-scale cohort study. Cardiovasc Diabetol 2025; 24:8. [PMID: 39780174 PMCID: PMC11708299 DOI: 10.1186/s12933-024-02537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Remnant cholesterol (remnant-C) contributes to atherosclerotic cardiovascular disease (ASCVD), particularly in individuals with impaired glucose metabolism. Patients with impaired glucose metabolism and ASCVD remain at significant residual risk after coronary artery bypass grafting (CABG). However, the role of remnant-C in this population has not yet been investigated. METHODS Adult patients with prediabetes or diabetes undergoing isolated CABG were consecutively enrolled in a longitudinal cohort between 2013 and 2018. The impact of remnant-C on short-term and long-term outcomes after CABG was evaluated. The short-term outcomes included major perioperative complications. The long-term outcomes were major adverse cardiovascular and cerebrovascular events (MACCEs). Remnant-C was analyzed as both a categorical and continuous variable. Logistic regression, Cox regression, and restricted cubic spline analyses were performed with multivariate adjustments. RESULTS In terms of perioperative outcomes, patients with elevated remnant-C had a higher incidence of acute kidney injury (AKI) stage 2/3 (high vs. low remnant-C: 3.2% vs. 2.4%; OR: 1.404, 95% CI 1.080-1.824). Each 1-standard deviation (SD) increase in remnant-C was associated with a 16.6% higher risk of AKI stage 2/3 (OR: 1.160, 95% CI 1.067-1.260). Long-term outcomes were assessed after a median follow-up of 3.2 years, during which 1,251 patients (9.3%) experienced MACCEs. Each 1-SD increase in remnant-C was associated with a 6.6% higher risk of MACCEs (HR: 1.066, 95% CI 1.012-1.124), a 7.1% higher risk of all-cause death (HR: 1.071, 95% CI 1.008-1.209), and an 11.2% higher risk of myocardial infarction (HR: 1.112, 95% CI 1.011-1.222). These associations remained consistent when remnant-C was treated as a categorical variable. Importantly, the association between remnant-C and MACCEs was independent of LDL-C levels; higher remnant-C levels were associated with increased MACCE risk regardless of whether LDL-C was ≤ 2.6 mmol/L or > 2.6 mmol/L. Subgroup analysis indicated that this risk was more pronounced in insulin-treated patients. CONCLUSIONS Remnant-C is associated with AKI and MACCEs in patients with diabetes or prediabetes undergoing CABG. The MACCE risk associated with remnant-C is independent of LDL-C and is more pronounced in insulin-treated patients.
Collapse
Affiliation(s)
- Zhongchen Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Rd, Xicheng District, Beijing, 100037, People's Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Chunyu Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Rd, Xicheng District, Beijing, 100037, People's Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Heng Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Rd, Xicheng District, Beijing, 100037, People's Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Runze Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Rd, Xicheng District, Beijing, 100037, People's Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yan Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Rd, Xicheng District, Beijing, 100037, People's Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Zhe Zheng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Rd, Xicheng District, Beijing, 100037, People's Republic of China.
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
- Key Laboratory of Coronary Heart Disease Risk Prediction and Precision Therapy, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
25
|
Corral P, Nardelli N, Elbert A, Aranguren F, Schreier L. Impact of SGLT2 Inhibitors on Lipoproteins in Type 2 Diabetes. Curr Diab Rep 2025; 25:16. [PMID: 39762665 DOI: 10.1007/s11892-024-01572-0] [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] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW This article explores the cardiovascular effects of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes mellitus (T2DM), with a particular focus on their impact on lipid profiles. As evidence grows of the cardiovascular benefits of SGLT2i beyond glucose control, it is essential to better understand their effects on lipoproteins and their impact on cardiovascular disease. RECENT FINDINGS SGLT2i have shown significant cardiovascular benefits in patients with type 2 diabetes mellitus, beyond their role in lowering blood glucose. Studies indicate that SGLT2i reduce major adverse cardiovascular events by impacting factors such as blood pressure, body weight, and arterial stiffness. However, their effects on lipid profile remain complex and somewhat inconsistent. Some research points to modest increases in LDL cholesterol, while others report shifts toward less atherogenic lipid profile, including reductions in triglycerides and small, dense LDL particles, and increases in HDL-C. SGLT2i represent a significant advancement in managing diabetes and associated cardiovascular risks, with benefits such as triglyceride reduction and HDL-C increase. While their impact on LDL-C remains controversial and varies across studies, the reduction of small, dense LDL particles may mitigate negative effects. This article highlights the need for future research to better understand the specific mechanisms behind lipid modulation.
Collapse
Affiliation(s)
- Pablo Corral
- Facultad de Medicina, Departamento de Farmacología e Investigación, Universidad FASTA, Instituto de Investigaciones Clínicas (IIC), Mar del Plata, Argentina.
| | - Natalia Nardelli
- Centro de Nutrición y Diabetes (CENUDIAB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Alicia Elbert
- Centro de Enfermedades Renales e Hipertensión Arterial (CEREHA S.A.), Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Laura Schreier
- Facultad de Farmacia y Bioquímica, Laboratorio de Lípidos y Aterosclerosis, Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC-UBA), Buenos Aires, Argentina
| |
Collapse
|
26
|
Lam SM, Wang Z, Song JW, Shi Y, Liu WY, Wan LY, Duan K, Chua GH, Zhou Y, Wang G, Huang X, Wang Y, Wang FS, Zheng MH, Shui G. Non-invasive lipid panel of MASLD fibrosis transition underscores the role of lipoprotein sulfatides in hepatic immunomodulation. Cell Metab 2025; 37:69-86.e7. [PMID: 39500328 DOI: 10.1016/j.cmet.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/18/2024] [Accepted: 09/13/2024] [Indexed: 01/11/2025]
Abstract
There exists a pressing need for a non-invasive panel that differentiates mild fibrosis from non-fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD). In this work, we applied quantitative lipidomics and sterolomics on sera from the PERSONS cohort with biopsy-based histological assessment of liver pathology. We trained a lasso regression model using quantitative omics data and clinical variables, deriving a combinatorial panel of lipids and clinical indices that differentiates mild fibrosis (>F1, n = 324) from non-fibrosis (F0, n = 195), with an area under receiver operating characteristic curve (AUROC) at 0.775 (95% confidence interval [CI]: 0.735-0.816). Circulating sulfatides (SLs) emerged as central lipids distinctly associated with fibrosis pathogenesis in MASLD. Lipidomics analysis of lipoprotein fractions revealed a redistribution of circulating SLs from high-density lipoproteins (HDLs) onto low-density lipoproteins (LDLs) in MASLD fibrosis. We further verified that patient LDLs with reduced SL content triggered a smaller activation of type II natural killer T lymphocytes, compared with control LDLs. Our results suggest that hepatic crosstalk with systemic immunity mediated by lipoprotein metabolism underlies fibrosis progression at early-stage MASLD.
Collapse
Affiliation(s)
- Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; LipidALL Technologies Company Limited, Changzhou 213022, Jiangsu, China
| | - Zehua Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jin-Wen Song
- Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Yue Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin-Yu Wan
- Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Kaibo Duan
- Centre for Biomedical Informatics, Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Gek Huey Chua
- LipidALL Technologies Company Limited, Changzhou 213022, Jiangsu, China
| | - Yingjuan Zhou
- LipidALL Technologies Company Limited, Changzhou 213022, Jiangsu, China
| | - Guibin Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Fu-Sheng Wang
- Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China.
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
27
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025:10.1038/s41569-024-01111-0. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
29
|
Zhao S, Giles C, Huynh K, Kettunen J, Järvelin MR, Kähönen M, Viikari J, Lehtimäki T, Raitakari OT, Meikle PJ, Mäkinen VP, Ala-Korpela M. Personalized Profiling of Lipoprotein and Lipid Metabolism Based on 1018 Measures from Combined Quantitative NMR and LC-MS/MS Platforms. Anal Chem 2024; 96:20362-20370. [PMID: 39680883 PMCID: PMC11696825 DOI: 10.1021/acs.analchem.4c03229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Applications of advanced omics methodologies are increasingly popular in biomedicine. However, large-scale studies aiming at clinical translation are typically siloed to single technologies. Here, we present the first comprehensive large-scale population data combining 209 lipoprotein measures from a quantitative NMR spectroscopy platform and 809 lipid classes and species from a quantitative LC-MS/MS platform. These data with 1018 molecular measures were analyzed in two population cohorts totaling 7830 participants. The association and cluster analyses revealed excellent coherence between the methodologically independent data domains and confirmed their quantitative compatibility and suitability for large-scale studies. The analyses elucidated the detailed molecular characteristics of the heterogeneous circulatory macromolecular lipid transport system and the underlying structural and compositional relationships. Unsupervised neural network analysis─the so-called self-organizing maps (SOMs)─revealed that these deep molecular and metabolic data are inherently related to key physiological and clinical population characteristics. The data-driven population subgroups uncovered marked differences in the population distribution of multiple cardiometabolic risk factors. These include, e.g., multiple lipoprotein lipids, apolipoprotein B, ceramides, and oxidized lipids. All 79 structurally unique triglyceride species showed similar associations over the entire lipoprotein cascade and indicated systematically increased risk for carotid intima media thickening and other atherosclerosis risk factors, including obesity and inflammation. The metabolic attributes for 27 individual cholesteryl ester species, which formed six distinct clusters, were more intricate with associations both with higher─e.g., CE(16:1)─and lower─e.g., CE(20:4)─cardiometabolic risk. The molecular details provided by these combined data are unprecedented for molecular epidemiology and demonstrate a new potential avenue for population studies.
Collapse
Affiliation(s)
- Siyu Zhao
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
| | - Corey Giles
- Baker
Heart and Diabetes Institute, Melbourne 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Melbourne 3004, Australia
| | - Kevin Huynh
- Baker
Heart and Diabetes Institute, Melbourne 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Melbourne 3004, Australia
| | - Johannes Kettunen
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
- Department
of Public Health and Welfare, Finnish Institute
for Health and Welfare, 00271 Helsinki, Finland
| | - Marjo-Riitta Järvelin
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Department
of Epidemiology and Biostatistics, MRC Centre for Environment and
Health, School of Public Health, Imperial
College London, London W12 0BZ, U.K.
- Department
of Life Sciences, College of Health and Life Sciences, Brunel University London, London UB8 3PH, U.K.
| | - Mika Kähönen
- Department
of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular
Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, 33270 Tampere, Finland
| | - Jorma Viikari
- Department
of Medicine, University of Turku, 20014 Turku, Finland
- Division
of Medicine, Turku University Hospital, 20014 Turku, Finland
| | - Terho Lehtimäki
- Department
of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular
Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, 33270 Tampere, Finland
| | - Olli T. Raitakari
- Research
Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20014 Turku, Finland
- Centre
for Population Health Research, University
of Turku and Turku University Hospital, 20014 Turku, Finland
- Department
of Clinical Physiology and Nuclear Medicine, Turku University Hospital, 20014 Turku, Finland
- InFLAMES
Research Flagship, University of Turku, 20014 Turku, Finland
| | - Peter J. Meikle
- Baker
Heart and Diabetes Institute, Melbourne 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Melbourne 3004, Australia
- Monash
University, Melbourne 3004, Australia
| | - Ville-Petteri Mäkinen
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
| | - Mika Ala-Korpela
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
- Monash
University, Melbourne 3004, Australia
- NMR Metabolomics
Laboratory, School of Pharmacy, University
of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
30
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
31
|
Corbalan JJ, Jagadeesan P, Frietze KK, Taylor R, Gao GL, Gallagher G, Nickels JT. Humanized monoacylglycerol acyltransferase 2 mice develop metabolic dysfunction-associated steatohepatitis. J Lipid Res 2024; 65:100695. [PMID: 39505262 DOI: 10.1016/j.jlr.2024.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Mice lacking monoacylglycerol acyltransferase 2 (mMGAT21) are resistant to diet-induced fatty liver, suggesting hMOGAT2 inhibition is a viable option for treating metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). We generated humanized hMOGAT2 mice (HuMgat2) for use in pre-clinical studies testing the efficacy of hMOGAT2 inhibitors for treating MASLD/MASH. HuMgat2 mice developed MASH when fed a steatotic diet. Computer-aided histology revealed the presence of hepatocyte cell ballooning, immune cell infiltration, and fibrosis. Hepatocytes accumulated Mallory-Denk bodies containing phosphorylated p62/sequestosome-1-ubiquitinated protein aggregates likely caused by defects in autophagy. Metainflammation and apoptotic cell death were seen in the livers of HuMgat2 mice. Treating HuMgat2 mice with elafibranor reduced several MASH phenotypes. RNASeq analysis predicted changes in bile acid transporter expression that correlated with altered bile acid metabolism indicative of cholestasis. Our results suggest that HuMgat2 mice will serve as a pre-clinical model for testing hMOGAT2 inhibitor efficacy and toxicity and allow for the study of hMOGAT2 in the context of MASH.
Collapse
Affiliation(s)
- J Jose Corbalan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Pranavi Jagadeesan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Karla K Frietze
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Grace L Gao
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA
| | - Grant Gallagher
- Oncoveda, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Joseph T Nickels
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
32
|
Xu M, Chen ZY, Li Y, Li Y, Guo G, Dai RZ, Ni N, Tao J, Wang HY, Chen QL, Wang H, Zhou H, Yang YN, Chen S, Chen L. Rab2A-mediated Golgi-lipid droplet interactions support very-low-density lipoprotein secretion in hepatocytes. EMBO J 2024; 43:6383-6409. [PMID: 39496977 PMCID: PMC11649929 DOI: 10.1038/s44318-024-00288-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Lipid droplets (LDs) serve as crucial hubs for lipid trafficking and metabolic regulation through their numerous interactions with various organelles. While the interplay between LDs and the Golgi apparatus has been recognized, their roles and underlying mechanisms remain poorly understood. Here, we reveal the role of Ras-related protein Rab-2A (Rab2A) in mediating LD-Golgi interactions, thereby contributing to very-low-density lipoprotein (VLDL) lipidation and secretion in hepatocytes. Mechanistically, our findings identify a selective interaction between Golgi-localized Rab2A and 17-beta-hydroxysteroid dehydrogenase 13 (HSD17B13) protein residing on LDs. This complex facilitates dynamic organelle communication between the Golgi apparatus and LDs, thus contributing to lipid transfer from LDs to the Golgi apparatus for VLDL2 lipidation and secretion. Attenuation of Rab2A activity via AMP-activated protein kinase (AMPK) suppresses the Rab2A-HSD17B13 complex formation, impairing LD-Golgi interactions and subsequent VLDL secretion. Furthermore, genetic inhibition of Rab2A and HSD17B13 in the liver reduces the serum triglyceride and cholesterol levels. Collectively, this study provides a new perspective on the interactions between the Golgi apparatus and LDs.
Collapse
Affiliation(s)
- Min Xu
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Zi-Yue Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China
| | - Yang Li
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, 830000, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, 830000, Urumqi, China
| | - Yue Li
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Ge Guo
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Rong-Zheng Dai
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Na Ni
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Jing Tao
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, 830000, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, 830000, Urumqi, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China
| | - Qiao-Li Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Hong Zhou
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China.
| | - Yi-Ning Yang
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, 830000, Urumqi, China.
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, 830000, Urumqi, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 830000, Urumqi, China.
- Key Laboratory of Cardiovascular Disease Research, First Affiliated Hospital of Xinjiang Medical University, 830000, Urumqi, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China.
| | - Liang Chen
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China.
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, 230001, Hefei, China.
| |
Collapse
|
33
|
Xiao X, Wu Y, Jie Z, Lin L, Li Y, Hu W, Li Y, Zhong S. Akkermansia Muciniphila supplementation improves hyperlipidemia, cardiac function, and gut microbiota in high fat fed apolipoprotein E-deficient mice. Prostaglandins Other Lipid Mediat 2024; 175:106906. [PMID: 39265779 DOI: 10.1016/j.prostaglandins.2024.106906] [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: 05/07/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Hyperlipidemia, obesity and gut dysbiosis are pivotal risk factors for atherosclerotic cardiovascular disease (ACVD). Supplementation of Akkermansia muciniphila (AKK) has also been proven to be effective in the prevention and treatment of obesity and other metabolic disorders. Here we found that AKK was more abundant in healthy control than ACVD patients via metagenomic sequencing on fecal samples. Subsequently, we investigated the role and underlying mechanism of AKK on obesity-associated atherosclerosis. AKK intervention partially reversed the exacerbation of atherosclerotic lesion formation in ApoE-/- mice by improving dyslipidemia. Interestingly, replenishment with AKK significantly enhanced cardiac function and reduced the body weight. It also reduced pro-inflammatory cytokine IL-6 and increased anti-inflammatory IL-10 in the circulation. Additionally, AKK colonization dramatically regulated gut microbiota and increased the abundance of Lactobacillaceae. Our findings have provided novel insights into the therapeutic potential of AKK as a beneficial microbe for treating atherosclerotic-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yuanyuan Wu
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, PR China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Zhuye Jie
- BGI-Shenzhen, Shenzhen 518083, PR China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, PR China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Lu Lin
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yangchen Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Weixian Hu
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, PR China
| | - Yong Li
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, PR China.
| | - Shilong Zhong
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, PR China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
34
|
Elías-López D, Wadström BN, Vedel-Krogh S, Kobylecki CJ, Nordestgaard BG. Impact of Remnant Cholesterol on Cardiovascular Risk in Diabetes. Curr Diab Rep 2024; 24:290-300. [PMID: 39356419 DOI: 10.1007/s11892-024-01555-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE OF REVIEW Individuals with diabetes face increased risk of atherosclerotic cardiovascular disease (ASCVD), in part due to hyperlipidemia. Even after LDL cholesterol-lowering, residual ASCVD risk persists, part of which may be attributed to elevated remnant cholesterol. We describe the impact of elevated remnant cholesterol on ASCVD risk in diabetes. RECENT FINDINGS Preclinical, observational, and Mendelian randomization studies robustly suggest that elevated remnant cholesterol causally increases risk of ASCVD, suggesting remnant cholesterol could be a treatment target. However, the results of recent clinical trials of omega-3 fatty acids and fibrates, which lower levels of remnant cholesterol in individuals with diabetes, are conflicting in terms of ASCVD prevention. This is likely partly due to neutral effects of these drugs on the total level of apolipoprotein B(apoB)-containing lipoproteins. Elevated remnant cholesterol remains a likely cause of ASCVD in diabetes. Remnant cholesterol-lowering therapies should also lower apoB levels to reduce risk of ASCVD.
Collapse
Affiliation(s)
- Daniel Elías-López
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- Department of Endocrinology and Metabolism and Research Center of Metabolic Diseases, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Secc. 16, Tlalpan, 14080, México City, México
| | - Benjamin Nilsson Wadström
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Signe Vedel-Krogh
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Camilla Jannie Kobylecki
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark.
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| |
Collapse
|
35
|
Yao B, Ma J, Ran Q, Chen H, He X. Mechanism of Valeriana officinalis L. extract improving atherosclerosis by regulating PGC-1α/Sirt3/Epac1 pathway. Front Pharmacol 2024; 15:1483518. [PMID: 39629078 PMCID: PMC11611558 DOI: 10.3389/fphar.2024.1483518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/07/2024] [Indexed: 12/06/2024] Open
Abstract
Objective To investigate the protective effect of the of Valeriana officinalis L. extract on mitochondrial injury in AS mice and the underlying mechanism. Methods Firstly, Ultra-High performance liquid chromatography-quadrupole time-of-flight mass spectrometer (UPLC / Q-TOF-MS) was proposed to explore the chemical composition of Valeriana officinalis L. extract. ApoE-/- mice were employed for in vivo experiments. The efficacy of Valeriana officinalis L. extract was detected by B-ultrasound, Biochemical, Oil Red O staining, HE staining and Masson staining analysis. The molecular mechanism of Valeriana officinalis L. extract in regulating mitochondrial energy metabolism for the treatment of atherosclerosis was elucidated after Monitoring System of Vascular Microcirculation in Vivo and transmission electron microscopy. Use the corresponding reagent kit to detect ACTH level, CHRNα1 level and ATP level, and measure the expression levels of PGC-1α, Sirt3, Epac1, Caspase-3, and Caspase-9 through real-time qPCR, and Western blot. Results A total of 29 metabolites were newly discovered from KYXC using UPLC-MS. The drug had a significant positive effect on the growth of atherosclerotic plaque in mice. It also improved the microcirculation of the heart and mesentery, reduced the levels of CHOL, TG, and VLDL in the serum, and increased the levels of HDL-C to maintain normal lipid metabolism in the body. Additionally, it increased the levels of ATP, improved the ultrastructure of mitochondria to maintain mitochondrial energy metabolism, and increased the levels of T-SOD to combat oxidative stress of the organism. Furthermore, the drug significantly increased the mRNA and protein expression of PGC-1α and Sirt3 in aortic tissue, while decreasing the mRNA and protein expression of Epac1, Caspase-3, and Caspase-9. Conclusion This study has verified that the extract of Valeriana officinalis L. is highly effective in enhancing atherosclerosis disease. The mechanism is suggested through the PGC-1α/Sirt3/Epac1 signaling pathway, which improves mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Bo Yao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Jingzhuo Ma
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingzhi Ran
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hengwen Chen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuanhui He
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
36
|
Sidorkiewicz M. The Cardioprotective Effects of Polyunsaturated Fatty Acids Depends on the Balance Between Their Anti- and Pro-Oxidative Properties. Nutrients 2024; 16:3937. [PMID: 39599723 PMCID: PMC11597422 DOI: 10.3390/nu16223937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are not only structural components of membrane phospholipids and energy storage molecules in cells. PUFAs are important factors that regulate various biological functions, including inflammation, oxidation, and immunity. Both n-3 and n-6 PUFAs from cell membranes can be metabolized into pro-inflammatory and anti-inflammatory metabolites that, in turn, influence cardiovascular health in humans. The role that PUFAs play in organisms depends primarily on their structure, quantity, and the availability of enzymes responsible for their metabolism. n-3 PUFAs, such as eicosapentaenoic (EPA) and docosahexaenoic (DHA), are generally known for anti-inflammatory and atheroprotective properties. On the other hand, n-6 FAs, such as arachidonic acid (AA), are precursors of lipid mediators that display mostly pro-inflammatory properties and may attenuate the efficacy of n-3 by competition for the same enzymes. However, a completely different light on the role of PUFAs was shed due to studies on the influence of PUFAs on new-onset atrial fibrillation. This review analyzes the role of PUFAs and PUFA derivatives in health-related effects, considering both confirmed benefits and newly arising controversies.
Collapse
Affiliation(s)
- Malgorzata Sidorkiewicz
- Department of Medical Biochemistry, Faculty of Health Sciences, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
37
|
Wen Y, Li Y, Liu T, Huang L, Yao L, Deng D, Luo W, Cai W, Zhong S, Jin T, Yang X, Wang Q, Wang W, Xue J, Mukherjee R, Hong J, Phillips AR, Windsor JA, Sutton R, Li F, Sun X, Huang W, Xia Q. Chaiqin chengqi decoction treatment mitigates hypertriglyceridemia-associated acute pancreatitis by modulating liver-mediated glycerophospholipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155968. [PMID: 39217651 DOI: 10.1016/j.phymed.2024.155968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The incidence of hypertriglyceridemia-associated acute pancreatitis (HTG-AP) is increasing globally and more so in China. The characteristics of liver-mediated metabolites and related key enzymes are rarely reported in HTG-AP. Chaiqin chengqi decoction (CQCQD) has been shown to protect against AP including HTG-AP in both patients and rodent models, but the underlying mechanisms in HTG-AP remain unexplored. PURPOSE To assess the characteristics of liver-mediated metabolism and the therapeutic mechanisms of CQCQD in HTG-AP. METHODS Male human apolipoprotein C3 transgenic (hApoC3-Tg; leading to HTG) mice or wild-type littermates received 7 intraperitoneal injections of cerulein (100 μg/kg) to establish HTG-AP and CER-AP, respectively. In HTG-AP, some mice received CQCQD (5.5 g/kg) gavage at 1, 5 or 9 h after disease induction. AP severity and related liver injury were determined by serological and histological parameters; and underlying mechanisms were identified by lipidomics and molecular biology. Molecular docking was used to identify key interactions between CQCQD compounds and metabolic enzymes, and subsequently validated in vitro in hepatocytes. RESULTS HTG-AP was associated with increased disease severity indices including augmented liver injury compared to CER-AP. CQCQD treatment reduced severity and liver injury of HTG-AP. Glycerophospholipid (GPL) metabolism was the most disturbed pathway in HTG-AP in comparison to HTG alone. In HTG-AP, the mRNA level of GPL enzymes involved in phosphocholine (PC) and phosphatidylethanolamine (PE) synthesis (Pcyt1a, Pcyt2, Pemt, and Lpcat) were markedly upregulated in the liver. Of the GPL metabolites, lysophosphatidylethanolamine LPE(16:0) in serum of HTG-AP was significantly elevated and positively correlated with the pancreas histopathology score (r = 0.65). In vitro, supernatant from Pcyt2-overexpressing hepatocytes co-incubated with LPE(16:0) or phospholipase A2 (a PC- and PE-hydrolyzing enzyme) alone induced pancreatic acinar cell death. CQCQD treatment downregulated PCYT1a and PCYT2 enzyme levels in the liver. Hesperidin and narirutin were identified top two CQCQD compounds with highest affinity docking to PCYT1a and PCYT2. Both hesperidin and narirutin reduced the level of some GPL metabolites in hepatocytes. CONCLUSION Liver-mediated GPL metabolism is excessively activated in HTG-AP with serum LPE(16:0) level correlating with disease severity. CQCQD reduces HTG-AP severity partially via modulating key enzymes in GPL metabolism pathway.
Collapse
Affiliation(s)
- Yongjian Wen
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuying Li
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijia Huang
- West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linbo Yao
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Deng
- West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjuan Luo
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenhao Cai
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shaoqi Zhong
- West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Jin
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinmin Yang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiqi Wang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wen Wang
- Chinese Evidence-based Medicine Centre, and National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Xue
- Laboratory of Oncogenes and Related Genes, Stem Cell Research Centre, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Rajarshi Mukherjee
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool and Liverpool University Hospitals NHS Foundation Trust, Liverpoo,l L69 3GE, UK
| | - Jiwon Hong
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Anthony R Phillips
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - John A Windsor
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool and Liverpool University Hospitals NHS Foundation Trust, Liverpoo,l L69 3GE, UK
| | - Fei Li
- Department of Pharmacy, Laboratory of Metabolomics and Drug-Induced Liver Injury, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Sun
- Chinese Evidence-based Medicine Centre, and National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China; West China Biobank, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
38
|
Wang J, Liu Y, Xiu C, Wang X, Liu Y, Hu Y, Yang J, Lei Y. Network Pharmacology-Based Strategy to Explore the Effect and Mechanism of Zhizhu Granule Improving Glucose-Lipid Metabolism in Rats with Metabolic Syndrome. Diabetes Metab Syndr Obes 2024; 17:3833-3846. [PMID: 39440025 PMCID: PMC11495215 DOI: 10.2147/dmso.s477410] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Objective To explore the mechanism of the traditional Chinese medicine (TCM), Zhizhu granule (ZZG), in treating metabolic syndrome (MS) based on network pharmacology and pharmacodynamic experiment. Materials and Methods Network pharmacology combined with a pharmacodynamic experiment was used to elucidate the therapeutic mechanism of ZZG in MS. Serum samples were collected from rats with MS, induced by a high-sugar-fat-salt diet (HSFSD) combined with streptozotocin (STZ), to measure the levels of biochemical markers. The glucose (GLU), total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), systolic blood pressure (SBP), and diastolic blood pressure (DBP) were detected. The liver tissue of rats was used for histological examination and Western blot analysis. Results Network pharmacology analysis generated 69 drug-disease common targets and 10 hub genes closely related to ZZG against MS. KEGG pathway analysis revealed that the PI3K/AKT signaling pathway was the most potential pathway, which took part in the therapeutic mechanisms. In the animal experiments section, the therapeutic effect of ZZG on MS and the therapeutic pathway of ZZG on MS were verified. ZZG could significantly decrease the body weight, TC, TG, LDL-C and GLU levels in MS rats (all p<0.01), alleviate hepatocyte steatosis and decrease liver lipid droplet deposition. Western blot analysis indicated that compared with the model group, the expression levels of PI3K, AKT, and IRS-1 protein were significantly increased (all p<0.05), and the FOXO-1 was significantly decreased (all p<0.05) in the ZZG group. Conclusion ZZG can improve glucose-lipid metabolism disorder in rats with metabolic syndrome. The reported results provide experimental evidence for ZZG in the treatment of MS.
Collapse
Affiliation(s)
- Jiali Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People’s Republic of China
| | - Yiqing Liu
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Chengkui Xiu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xue Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yinan Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yanhong Hu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Jing Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yan Lei
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
39
|
Shagdarova B, Melnikova V, Kostenko V, Konovalova M, Zhuikov V, Varlamov V, Svirshchevskaya E. Effects of Chitosan and N-Succinyl Chitosan on Metabolic Disorders Caused by Oral Administration of Olanzapine in Mice. Biomedicines 2024; 12:2358. [PMID: 39457671 PMCID: PMC11504887 DOI: 10.3390/biomedicines12102358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The issue of human mental health is gaining more and more attention nowadays. However, most mental disorders are treated with antipsychotic drugs that cause weight gain and metabolic disorders, which include olanzapine (OLZ). The search for and development of natural compounds for the prevention of obesity when taking antipsychotic drugs is an urgent task. The biopolymer chitosan (Chi) and its derivatives have lipid-lowering and anti-diabetic properties, which makes them potential therapeutic substances for use in the treatment of metabolic disorders. The purpose of this work was to analyze the effect of the natural biopolymer Chi, its derivative N-succinyl chitosan (SuChi), and Orlistat (ORL) as a control on the effects caused by the intake of OLZ in a mouse model. METHODS Mice were fed with pearl barley porridge mixed with OLZ or combinations OLZ + Chi, OLZ + SuChi, or OLZ + ORL for 2 months. The weight, lipid profile, blood chemokines, expression of genes associated with appetite regulation, and behavior of the mice were analyzed in dynamics. RESULTS For the first time, data were obtained on the effects of Chi and SuChi on metabolic changes during the co-administration of antipsychotics. Oral OLZ increased body weight, food and water intake, and glucose, triglyceride, and cholesterol levels in blood. ORL and SuChi better normalized lipid metabolism than Chi, decreasing triglyceride and cholesterol levels. OLZ decreased the production of all chemokines tested at the 4th week of treatment and increased CXCL1, CXCL13, and CCL22 chemokine levels at the 7th week. All of the supplements corrected the level of CXCL1, CXCL13, and CCL22 chemokines but did not recover suppressed chemokines. SuChi and ORL stimulated the expression of satiety associated proopiomelanocortin (POMC) and suppressed the appetite-stimulating Agouti-related protein (AgRP) genes. All supplements improved the locomotion of mice. CONCLUSIONS Taken collectively, we found that SuChi more than Chi possessed an activity close to that of ORL, preventing metabolic disorders in mice fed with OLZ. As OLZ carries positive charge and SuChi is negatively charged, we hypothesized that SuChi's protective effect can be explained by electrostatic interaction between OLZ byproducts and SuChi in the gastrointestinal tract.
Collapse
Affiliation(s)
- Balzhima Shagdarova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia; (V.Z.); (V.V.)
| | - Viktoria Melnikova
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Valentina Kostenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (V.K.); (M.K.); (E.S.)
| | - Mariya Konovalova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (V.K.); (M.K.); (E.S.)
| | - Vsevolod Zhuikov
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia; (V.Z.); (V.V.)
| | - Valery Varlamov
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia; (V.Z.); (V.V.)
| | - Elena Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (V.K.); (M.K.); (E.S.)
| |
Collapse
|
40
|
Gonçalves M, Costa M, Paiva-Martins F, Silva P. Olive Oil Industry By-Products as a Novel Source of Biophenols with a Promising Role in Alzheimer Disease Prevention. Molecules 2024; 29:4841. [PMID: 39459209 PMCID: PMC11510978 DOI: 10.3390/molecules29204841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
This review explores the potential health benefits and applications of phenolic secoiridoids derived from olive oil by-products in the prevention of Alzheimer's disease (AD). As reviewed herein, polyphenols, such as epigallocatechin-3-gallate, epicatechin, and resveratrol, show in vitro and in vivo antioxidant, anti-inflammatory, and neuroprotective properties, and are particularly relevant in the context of AD, a leading cause of dementia globally. The olive oil industry, particularly in the Mediterranean region, produces significant amounts of waste, including leaves, pomace, and wastewater, which pose environmental challenges but also offer an untapped source of bioactive compounds. Despite promising in vitro and in vivo studies indicating that olive-derived polyphenols, such as oleuropein and hydroxytyrosol, may mitigate AD pathology, human clinical trials remain limited. The variability in extraction methods and the complex nature of AD further complicate research. Future studies should focus on standardizing the protocols and conducting robust clinical trials to fully assess the therapeutic potential of these compounds. This approach not only supports the development of new treatments for AD but also promotes environmental sustainability by valorizing olive oil industry waste.
Collapse
Affiliation(s)
- Marta Gonçalves
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| | - Marlene Costa
- REQUIMTE/LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal; (M.C.); (F.P.-M.)
| | - Fátima Paiva-Martins
- REQUIMTE/LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal; (M.C.); (F.P.-M.)
| | - Paula Silva
- Laboratory of Histology and Embryology, Department of Microscopy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- iNOVA Media Lab, ICNOVA-NOVA Institute of Communication, NOVA School of Social Sciences and Humanities, Universidade NOVA de Lisboa, 1069-061 Lisbon, Portugal
| |
Collapse
|
41
|
Xiao Q, Wang L, Wang J, Wang M, Wang DW, Ding H. A novel lncRNA GM47544 modulates triglyceride metabolism by inducing ubiquitination-dependent protein degradation of APOC3. Mol Metab 2024; 88:102011. [PMID: 39173944 PMCID: PMC11399561 DOI: 10.1016/j.molmet.2024.102011] [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: 04/04/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
OBJECTIVE Emerging evidence highlights the pivotal roles of long non-coding RNAs (lncRNAs) in lipid metabolism. Apoprotein C3 (ApoC3) is a well-established therapeutic target for hypertriglyceridemia and exhibits a strong association with cardiovascular disease. However, the exact mechanisms via which the lncRNAs control ApoC3 expression remain unclear. METHODS We identified a novel long noncoding RNA (lncRNA), GM47544, within the ApoA1/C3/A4/A5 gene cluster. Subsequently, the effect of GM47544 on intracellular triglyceride metabolism was analyzed. The diet-induced mouse models of hyperlipidemia and atherosclerosis were established to explore the effect of GM47544 on dyslipidemia and plaque formation in vivo. The molecular mechanism was explored through RNA sequencing, immunoprecipitation, RNA pull-down assay, and RNA immunoprecipitation. RESULTS GM47544 was overexpressed under high-fat stimulation. GM47544 effectively improved hepatic steatosis, reduced blood lipid levels, and alleviated atherosclerosis in vitro and in vivo. Mechanistically, GM47544 directly bound to ApoC3 and facilitated the ubiquitination at lysine 79 in ApoC3, thereby facilitating ApoC3 degradation via the ubiquitin-proteasome pathway. Moreover, we identified AP006216.5 as the human GM47544 transcript, which fulfills a comparable function in human hepatocytes. CONCLUSIONS The identification of GM47544 as a lncRNA modulator of ApoC3 reveals a novel mechanism of post-translational modification, with significant clinical implications for the treatment of hypertriglyceridemia and atherosclerosis.
Collapse
Affiliation(s)
- Qianqian Xiao
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Luyun Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China; Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Jing Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China; Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Man Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Dao Wen Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China; Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China; Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
42
|
Lichtenstein L, Cheng CW, Bajarwan M, Evans EL, Gaunt HJ, Bartoli F, Chuntharpursat-Bon E, Patel S, Konstantinou C, Futers TS, Reay M, Parsonage G, Moore JB, Bertrand-Michel J, Sukumar P, Roberts LD, Beech DJ. Endothelial force sensing signals to parenchymal cells to regulate bile and plasma lipids. SCIENCE ADVANCES 2024; 10:eadq3075. [PMID: 39331703 PMCID: PMC11430402 DOI: 10.1126/sciadv.adq3075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024]
Abstract
How cardiovascular activity interacts with lipid homeostasis is incompletely understood. We postulated a role for blood flow acting at endothelium in lipid regulatory organs. Transcriptome analysis was performed on livers from mice engineered for deletion of the flow-sensing PIEZO1 channel in endothelium. This revealed unique up-regulation of Cyp7a1, which encodes the rate-limiting enzyme for bile synthesis from cholesterol in hepatocytes. Consistent with this effect were increased gallbladder and plasma bile acids and lowered hepatic and plasma cholesterol. Elevated portal fluid flow acting via endothelial PIEZO1 and genetically enhanced PIEZO1 conversely suppressed Cyp7a1. Activation of hepatic endothelial PIEZO1 channels promoted phosphorylation of nitric oxide synthase 3, and portal flow-mediated suppression of Cyp7a1 depended on nitric oxide synthesis, suggesting endothelium-to-hepatocyte coupling via nitric oxide. PIEZO1 variants in people were associated with hepatobiliary disease and dyslipidemia. The data suggest an endothelial force sensing mechanism that controls lipid regulation in parenchymal cells to modulate whole-body lipid homeostasis.
Collapse
Affiliation(s)
- Laeticia Lichtenstein
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Chew W. Cheng
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Muath Bajarwan
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | | | - Fiona Bartoli
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | - Shaili Patel
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- Department of Hepatobiliary and Transplant Surgery, St James's University Hospital, Leeds LS9 7TF, UK
| | - Charalampos Konstantinou
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
- Department of Hepatobiliary and Transplant Surgery, St James's University Hospital, Leeds LS9 7TF, UK
| | | | - Melanie Reay
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | | | - J. Bernadette Moore
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Justine Bertrand-Michel
- MetaToul-Lipidomics Facility, INSERM UMR1048, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, INSERM, Toulouse, France
| | | | - Lee D. Roberts
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - David J. Beech
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
43
|
Wu Y, Foollee A, Chan AY, Hille S, Hauke J, Challis MP, Johnson JL, Yaron TM, Mynard V, Aung OH, Cleofe MAS, Huang C, Lim Kam Sian TCC, Rahbari M, Gallage S, Heikenwalder M, Cantley LC, Schittenhelm RB, Formosa LE, Smith GC, Okun JG, Müller OJ, Rusu PM, Rose AJ. Phosphoproteomics-directed manipulation reveals SEC22B as a hepatocellular signaling node governing metabolic actions of glucagon. Nat Commun 2024; 15:8390. [PMID: 39333498 PMCID: PMC11436942 DOI: 10.1038/s41467-024-52703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
The peptide hormone glucagon is a fundamental metabolic regulator that is also being considered as a pharmacotherapeutic option for obesity and type 2 diabetes. Despite this, we know very little regarding how glucagon exerts its pleiotropic metabolic actions. Given that the liver is a chief site of action, we performed in situ time-resolved liver phosphoproteomics to reveal glucagon signaling nodes. Through pathway analysis of the thousands of phosphopeptides identified, we reveal "membrane trafficking" as a dominant signature with the vesicle trafficking protein SEC22 Homolog B (SEC22B) S137 phosphorylation being a top hit. Hepatocyte-specific loss- and gain-of-function experiments reveal that SEC22B was a key regulator of glycogen, lipid and amino acid metabolism, with SEC22B-S137 phosphorylation playing a major role in glucagon action. Mechanistically, we identify several protein binding partners of SEC22B affected by glucagon, some of which were differentially enriched with SEC22B-S137 phosphorylation. In summary, we demonstrate that phosphorylation of SEC22B is a hepatocellular signaling node mediating the metabolic actions of glucagon and provide a rich resource for future investigations on the biology of glucagon action.
Collapse
Affiliation(s)
- Yuqin Wu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Ashish Foollee
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Andrea Y Chan
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Susanne Hille
- Department of Internal Medicine V, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Jana Hauke
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Matthew P Challis
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Department of Cell Biology, Harvard Medical School, Boston, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Tomer M Yaron
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Victoria Mynard
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Okka H Aung
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Maria Almira S Cleofe
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Cheng Huang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, Victoria, Australia
| | | | - Mohammad Rahbari
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Department of Surgery, Theodor-Kutzer-Ufer 1-3, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
| | - Suchira Gallage
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard-Karls University, Tübingen, Germany
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Department of Cell Biology, Harvard Medical School, Boston, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Ralf B Schittenhelm
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Greg C Smith
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine V, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia.
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia.
| |
Collapse
|
44
|
Gugliucci A. Angiopoietin-like Proteins and Lipoprotein Lipase: The Waltz Partners That Govern Triglyceride-Rich Lipoprotein Metabolism? Impact on Atherogenesis, Dietary Interventions, and Emerging Therapies. J Clin Med 2024; 13:5229. [PMID: 39274442 PMCID: PMC11396212 DOI: 10.3390/jcm13175229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Over 50% of patients who take statins are still at risk of developing atherosclerotic cardiovascular disease (ASCVD) and do not achieve their goal LDL-C levels. This residual risk is largely dependent on triglyceride-rich lipoproteins (TRL) and their remnants. In essence, remnant cholesterol-rich chylomicron (CM) and very-low-density lipoprotein (VLDL) particles play a role in atherogenesis. These remnants increase when lipoprotein lipase (LPL) activity is inhibited. ApoCIII has been thoroughly studied as a chief inhibitor and therapeutic options to curb its effect are available. On top of apoCIII regulation of LPL activity, there is a more precise control of LPL in various tissues, which makes it easier to physiologically divide the TRL burden according to the body's requirements. In general, oxidative tissues such as skeletal and cardiac muscle preferentially take up lipids during fasting. Conversely, LPL activity in adipocytes increases significantly after feeding, while its activity in oxidative tissues decreases concurrently. This perspective addresses the recent improvements in our understanding of circadian LPL regulations and their therapeutic implications. Three major tissue-specific lipolysis regulators have been identified: ANGPTL3, ANGPTL4, and ANGPTL8. Briefly, during the postprandial phase, liver ANGPTL8 acts on ANGPTL3 (which is released continuously from the liver) to inhibit LPL in the heart and muscle through an endocrine mechanism. On the other hand, when fasting, ANGPTL4, which is released by adipocytes, inhibits lipoprotein lipase in adipose tissue in a paracrine manner. ANGPTL3 inhibitors may play a therapeutic role in the treatment of hypertriglyceridemia. Several approaches are under development. We look forward to future studies to clarify (a) the nature of hormonal and nutritional factors that determine ANGPTL3, 4, and 8 activities, along with what long-term impacts may be expected if their regulation is impaired pharmacologically; (b) the understanding of the quantitative hierarchy and interaction of the regulatory actions of apoCIII, apoAV, and ANGPTL on LPL activity; (c) strategies for the safe and proper treatment of postprandial lipemia; and (d) the effect of fructose restriction on ANGPTL3, ANGPTL4, and ANGPTL8.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
45
|
Gómez-Álvarez N, Boppre G, Hermosilla-Palma F, Reyes-Amigo T, Oliveira J, Fonseca H. Effects of Small-Sided Soccer Games on Physical Fitness and Cardiometabolic Health Biomarkers in Untrained Children and Adolescents: A Systematic Review and Meta-Analysis. J Clin Med 2024; 13:5221. [PMID: 39274434 PMCID: PMC11396522 DOI: 10.3390/jcm13175221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Objective: This systematic review and meta-analysis aimed to determine the benefits of an exercise intervention based on small-sided soccer games (SSSGs) on health-related physical fitness and cardiometabolic health in previously untrained children and adolescents. Methods: A systematic search on PubMed/MEDLINE, Web of Science, Scopus, Cochrane, and EBSCO databases was performed. Randomized or non-randomized controlled trials conducted in previously untrained children or adolescents (age < 18 years) that assessed the effect of SSSG-based intervention on health-related physical fitness and cardiometabolic risk biomarkers were included. Primary outcomes were cardiorespiratory fitness and waist circumference. Evidence was synthesized as the mean difference or standardized mean difference using a random-effects meta-analysis. The quality of evidence was assessed using ROB2 and ROBINS-I tools. Results: Sixteen studies (n = 2872 participants) were included in this meta-analysis. SSSGs significantly improved cardiorespiratory fitness (SMD, 0.12 [0.01; 0.23]) and showed a non-significant trend in decreased waist circumference (-7.49 cm [-15.03; 0.06]). Additionally, SSSGs significantly decreased systolic (MD, -3.85 mmHg [-5.75; -1.94]) and diastolic blood pressure (MD, -1.26 mmHg [-2.44; -0.08]) and triglycerides (-30.34 mg·dL-1 [-45.99; -14.69]). No effects on body composition or other cardiometabolic risk biomarkers were observed. After a sensitivity analysis, waist circumference and muscle strength were also shown to improve significantly following SSSGs. Comparisons between SSSG and other types of exercise interventions showed no differences in improved physical fitness or cardiometabolic risk. Conclusions: SSSG-based interventions effectively improve cardiorespiratory fitness, blood pressure, triglycerides, muscle strength, and waist circumference. There is less evidence of the effects of SSSGs on other health markers. Particular attention should be given to improving SSSG protocol reporting in future studies.
Collapse
Affiliation(s)
- Nicolás Gómez-Álvarez
- Centre for Research, Education, Innovation and Intervention in Sport, Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Nucleus of Research in Human Motricity Sciences, Universidad Adventista de Chile, Chillán 3780000, Chile
| | - Giorjines Boppre
- Research Centre in Physical Activity, Health, and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600 Porto, Portugal
| | - Felipe Hermosilla-Palma
- Pedagogía en Educación Física, Facultad de Educación, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Tomás Reyes-Amigo
- Physical Activity Sciences Observatory (OCAF), Department of Physical Activity Sciences, Universidad de Playa Ancha, Valparaíso 2360072, Chile
| | - José Oliveira
- Research Centre in Physical Activity, Health, and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600 Porto, Portugal
| | - Hélder Fonseca
- Research Centre in Physical Activity, Health, and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600 Porto, Portugal
| |
Collapse
|
46
|
Arrobas-Velilla T, Ariza MJ, Rico-Corral MÁ, Valdivielso P. Early detection of severe hypertriglyceridemia using teleconsultation in a clinical laboratory setting. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:299-302. [PMID: 38702205 DOI: 10.1016/j.arteri.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/26/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Teleconsultation in the context of clinical laboratories is a valuable tool for the early detection of dyslipidemia and prevention of cardiovascular risk. Here, we describe a patient who was referred to the Lipid Unit of the Virgen Macarena Hospital due to an alert for severe hypertriglyceridemia through its teleconsultation program. CASE PRESENTATION A comprehensive clinical and biochemical study of the patient was carried out, and genetic testing was performed on the patient and his family. The proband and his family showed mild to severe hypertriglyceridemia and various secondary factors, together with a genetic background associated with a triglyceride-raising effect. CONCLUSION This extensive study has identified a family at high risk of cardiovascular disease and acute pancreatitis. These findings can help maximize lifestyle changes and improve the clinical management of their dyslipidemia.
Collapse
Affiliation(s)
| | - María José Ariza
- Lipids and Atherosclerosis Laboratory, Department of Medicine and Dermatology, Centro de Investigaciones Médico Sanitarias (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, Málaga, Spain.
| | | | - Pedro Valdivielso
- Lipids and Atherosclerosis Laboratory, Department of Medicine and Dermatology, Centro de Investigaciones Médico Sanitarias (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, Málaga, Spain; Internal Medicine and Lipid Units, University Hospital Virgen de la Victoria, Málaga, Spain
| |
Collapse
|
47
|
Kang T, Zhou Y, Fan C, Zhang Y, Yang Y, Jiang J. Genetic association of lipid traits and lipid-related drug targets with normal tension glaucoma: a Mendelian randomization study for predictive preventive and personalized medicine. EPMA J 2024; 15:511-524. [PMID: 39239107 PMCID: PMC11371969 DOI: 10.1007/s13167-024-00373-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/05/2024] [Indexed: 09/07/2024]
Abstract
Background Glaucoma is the leading cause of irreversible blindness worldwide. Normal tension glaucoma (NTG) is a distinct subtype characterized by intraocular pressures (IOP) within the normal range (< 21 mm Hg). Due to its insidious onset and optic nerve damage, patients often present with advanced conditions upon diagnosis. NTG poses an additional challenge as it is difficult to identify with normal IOP, complicating its prediction, prevention, and treatment. Observational studies suggest a potential association between NTG and abnormal lipid metabolism, yet conclusive evidence establishing a direct causal relationship is lacking. This study aims to explore the causal link between serum lipids and NTG, while identifying lipid-related therapeutic targets. From the perspective of predictive, preventive, and personalized medicine (PPPM), clarifying the role of dyslipidemia in the development of NTG could provide a new strategy for primary prediction, targeted prevention, and personalized treatment of the disease. Working hypothesis and methods In our study, we hypothesized that individuals with dyslipidemia may be more susceptible to NTG due to a dysregulation of microvasculature in optic nerve head. To verify the working hypothesis, univariable Mendelian randomization (UVMR) and multivariable Mendelian randomization (MVMR) were utilized to estimate the causal effects of lipid traits on NTG. Drug target MR was used to explore possible target genes for NTG treatment. Genetic variants associated with lipid traits and variants of genes encoding seven lipid-related drug targets were extracted from the Global Lipids Genetics Consortium genome-wide association study (GWAS). GWAS data for NTG, primary open angle glaucoma (POAG), and suspected glaucoma (GLAUSUSP) were obtained from FinnGen Consortium. For apolipoproteins, we used summary statistics from a GWAS study by Kettunen et al. in 2016. For metabolic syndrome, summary statistics were extracted from UK Biobank participants. In the end, these findings could help identify individuals at risk of NTG by screening for lipid dyslipidemia, potentially leading to new targeted prevention and personalized treatment approaches. Results Genetically assessed high-density cholesterol (HDL) was negatively associated with NTG risk (inverse-variance weighted [IVW] model: OR per SD change of HDL level = 0.64; 95% CI, 0.49-0.85; P = 1.84 × 10-3), and the causal effect was independent of apolipoproteins and metabolic syndrome (IVW model: OR = 0.29; 95% CI, 0.14-0.60; P = 0.001 adjusted by ApoB and ApoA1; OR = 0.70; 95% CI, 0.52-0.95; P = 0.023 adjusted by BMI, HTN, and T2DM). Triglyceride (TG) was positively associated with NTG risk (IVW model: OR = 1.62; 95% CI, 1.15-2.29; P = 6.31 × 10-3), and the causal effect was independent of metabolic syndrome (IVW model: OR = 1.66; 95% CI, 1.18-2.34; P = 0.003 adjusted by BMI, HTN, and T2DM), but not apolipoproteins (IVW model: OR = 1.71; 95% CI, 0.99-2.95; P = 0.050 adjusted by ApoB and ApoA1). Genetic mimicry of apolipoprotein B (APOB) enhancement was associated with lower NTG risks (IVW model: OR = 0.09; 95% CI, 0.03-0.26; P = 9.32 × 10-6). Conclusions Our findings supported dyslipidemia as a predictive causal factor for NTG, independent of other factors such as metabolic comorbidities. Among seven lipid-related drug targets, APOB is a potential candidate drug target for preventing NTG. Personalized health profiles can be developed by integrating lipid metabolism with life styles, visual quality of life such as reading, driving, and walking. This comprehensive approach will aid in shifting from reactive medical services to PPPM in the management of NTG. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00373-5.
Collapse
Affiliation(s)
- Tianyi Kang
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Yi Zhou
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Cong Fan
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Yue Zhang
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Yu Yang
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Jian Jiang
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| |
Collapse
|
48
|
Taylor R. Understanding the cause of type 2 diabetes. Lancet Diabetes Endocrinol 2024; 12:664-673. [PMID: 39038473 DOI: 10.1016/s2213-8587(24)00157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024]
Abstract
Type 2 diabetes has long been thought to have heterogenous causes, even though epidemiological studies uniformly show a tight relationship with overnutrition. The twin cycle hypothesis postulated that interaction of self-reinforcing cycles of fat accumulation inside the liver and pancreas, driven by modest but chronic positive calorie balance, could explain the development of type 2 diabetes. This hypothesis predicted that substantial weight loss would bring about a return to the non-diabetic state, permitting observation of the pathophysiology determining the transition. These changes were postulated to reflect the basic mechanisms of causation in reverse. A series of studies over the past 15 years has elucidated these underlying mechanisms. Together with other research, the interaction of environmental and genetic factors has been clarified. This knowledge has led to successful implementation of a national programme for remission of type 2 diabetes. This Review discusses the paucity of evidence for heterogeneity in causes of type 2 diabetes and summarises the in vivo pathophysiological changes, which cause this disease of overnutrition. Type 2 diabetes has a homogenous cause expressed in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Newcastle Magnetic Resonance Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
49
|
Lin Y, Wang S, Li Z, Zhou Y, Wang R, Wang Y, Chen Y. Short-Term Statin Therapy Induces Hepatic Insulin Resistance Through HNF4α/PAQR9/PPM1α Axis Regulated AKT Phosphorylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403451. [PMID: 38970167 PMCID: PMC11425881 DOI: 10.1002/advs.202403451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Statins, the first-line medication for dyslipidemia, are linked to an increased risk of type 2 diabetes. But exactly how statins cause diabetes is yet unknown. In this study, a developed short-term statin therapy on hyperlipidemia mice show that hepatic insulin resistance is a cause of statin-induced diabetes. Statin medication raises the expression of progesterone and adiponectin receptor 9 (PAQR9) in liver, which inhibits insulin signaling through degradation of protein phosphatase, Mg2+/Mn2+ dependent 1 (PPM1α) to activate ERK pathway. STIP1 homology and U-box containing protein 1 (STUB1) is found to mediate ubiquitination of PPM1α promoted by PAQR9. On the other hand, decreased activity of hepatocyte nuclear factor 4 alpha (HNF4α) seems to be the cause of PAQR9 expression under statin therapy. The interventions on PAQR9, including deletion of PAQR9, caloric restriction and HNF4α activation, are all effective treatments for statin-induced diabetes, while liver specific over-expression of PPM1α is another possible tactic. The results reveal the importance of HNF4α-PAQR9-STUB1-PPM1α axis in controlling the statin-induced hepatic insulin resistance, offering a fresh insight into the molecular mechanisms underlying statin therapy.
Collapse
Affiliation(s)
- Yijun Lin
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Shuying Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zixuan Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuling Zhou
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Ruiying Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
50
|
Jin Z, Wang X. Traditional Chinese medicine and plant-derived natural products in regulating triglyceride metabolism: Mechanisms and therapeutic potential. Pharmacol Res 2024; 208:107387. [PMID: 39216839 DOI: 10.1016/j.phrs.2024.107387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
The incidence of cardiometabolic disease is increasing globally, with a trend toward younger age of onset. Among these, atherosclerotic cardiovascular disease is a leading cause of mortality worldwide. Despite the efficacy of traditional lipid-lowering drugs, such as statins, in reducing low-density lipoprotein cholesterol levels, a significant residual risk of cardiovascular events remains, which is closely related to unmet triglyceride (TG) targets. The clinical application of current TG-lowering Western medicines has certain limitations, necessitating alternative or complementary therapeutic strategies. Traditional Chinese medicine (TCM) and plant-derived natural products, known for their safety owing to their natural origins and diverse biological activities, offer promising avenues for TG regulation with potentially fewer side effects. This review systematically summarises the mechanisms of TG metabolism and subsequently reviews the regulatory effects of TCM and plant-derived natural products on TG metabolism, including the inhibition of TG synthesis (via endogenous and exogenous pathways), promotion of TG catabolism, regulation of fatty acid absorption and transport, enhancement of lipophagy, modulation of the gut microbiota, and other mechanisms. In conclusion, through a comprehensive analysis of recent studies, this review consolidates the multifaceted regulatory roles of TCM and plant-derived natural products in TG metabolism and elucidates their potential as safer, multi-target therapeutic agents in managing hypertriglyceridemia and mitigating cardiovascular risk, thereby providing a basis for new drug development.
Collapse
Affiliation(s)
- Zhou Jin
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaolong Wang
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|