1
|
Zhao S, Song Y, Nakashima Y, Zou X, Koga T, Ishida T, Li R, Hirota Y, Tanaka Y, Ishii Y. Ablation of Mouse Selenium-Binding Protein 1 and 2 Elevates LDL by Disruption of Cholesterol Efflux and Lipid Metabolism. Int J Mol Sci 2025; 26:3363. [PMID: 40244197 PMCID: PMC11989624 DOI: 10.3390/ijms26073363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Selenium-binding protein 1 (SeBP1) is an anticancer factor that affects lipid metabolism in mouse kidneys via the peroxisome proliferator-activated receptor-alpha (PPARA) pathway. However, its physiological role in the liver is difficult to explain because of the presence of the highly homologous selenium-binding protein 2 (SeBP2). To investigate the role of these proteins in the liver, we generated SeBP1 and SeBP2 double-knockout mice (SeBP1/2-DK). SeBP1/2 deletion did not significantly alter the mice phenotypic compared to that of the wild-type strain. Then, we identified the genes involved in hepatic lipid metabolism. The double knockout did not affect fatty acid and cholesterol synthesis, but inhibited fatty acid oxidation and cholesterol efflux. Furthermore, transfection of HepG2 cells with human selenium-binding protein 1 (hSeBP1) positively regulated PPARA and the genes controlled by it. Overexpression of hSeBP1 reduced the levels of non-esterified fatty acids in the culture medium. The serum levels of low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, and triglycerides were significantly different among the three groups. In summary, we elucidated the potential signaling pathways of SeBP1 and SeBP2 in fatty acid oxidation and hepatic cholesterol efflux. Our findings provide insights relevant for developing new strategies to prevent and treat lipid metabolism disorders.
Collapse
Grants
- Scientific Research (A) JSPS KAKENHI JP17H00788, Recipient YI Japan Society for the Promotion of Science
- Scientific Research (A) JSPS KAKENHI JP21H04928, Recipient YI Japan Society for the Promotion of Science
- JSPS Fellows JSPS KAKENHI 24KJ1773, Recipient SZ Japan Society for the Promotion of Science
- Research on Food Safety (H30-Designated Research-005, Recipient YI) the Ministry of Health, Labor and Welfare, Japan
- the Ministry of Health, Labor and Welfare, Japan [Research on Food Safety (R3-Designated Research JP21KA2003, Recipient YI) the Ministry of Health, Labor and Welfare, Japan
- Research on Food Safety ( R6-Designated Research JP24KA2001, Recipient YI) the Ministry of Health, Labor and Welfare, Japan
Collapse
Affiliation(s)
- Shuangli Zhao
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| | - Yingxia Song
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| | - Yuko Nakashima
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| | - Xing Zou
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| | - Takayuki Koga
- Laboratory of Hygienic Chemistry, Daiichi University of Pharmacy, Fukuoka 815-8511, Japan;
| | - Takumi Ishida
- School of Pharmacy, International University of Health and Welfare Fukuoka, Ohkawa 831-8501, Japan;
| | - Renshi Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Yuko Hirota
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| | - Yoshitaka Tanaka
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| | - Yuji Ishii
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (S.Z.); (Y.S.); (X.Z.); (Y.H.); (Y.T.)
| |
Collapse
|
2
|
Yang Z, Ren J, Lu S, Feng Y, Fan Y, Liu TX, Jing X. In vivo functional analysis of the cotton bollworm Helicoverpa armigera 24-dehydrocholesterol reductase (HaDHCR24) in phytosterol metabolism. INSECT SCIENCE 2025; 32:398-408. [PMID: 38973264 DOI: 10.1111/1744-7917.13405] [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: 03/15/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 07/09/2024]
Abstract
Insects have to obtain sterols from food due to the inability to synthesize this essential nutrient de novo. For lepidopteran insects, they can convert a variety of phytosterols into cholesterol to meet their growth needs. The final step of the cholesterol biosynthesis is the metabolism of desmosterol catalyzed by 24-dehydrocholesterol reductase (DHCR24). In this study, we identified a DHCR24 homolog in the cotton bollworm Helicoverpa armigera, designated as H. armigera 24-dehydrocholesterol reductase (HaDHCR24)-1. The quantitative expression analyses indicated that HaDHCR24-1 was highly enriched in the midgut where dietary sterol uptake occurs. Compared to the control, the DHCR24-1 mutant larvae generated by clustered regularly interspaced palindromic repeats (CRISPR) / CRISPR-associated nuclease 9 technology accumulated more desmosterol in the gut, while the content of cholesterol was significantly reduced. A similar phenomenon was observed when the DHCR24 inhibitor, amiodarone, was applied to the insects. Moreover, DHCR24-1 played an important role for the usage of β-sitosterol, a major sterol in plants, in H. armigera, and loss of function of DHCR24-1 resulted in higher mortality on β-sitosterol. However, the DHCR24 homolog does not necessarily exist in the genomes of all insects. The loss of this gene occurred more frequently in the insects feeding on animals, which further support the role of DHCR24-1 in using phytosterols. This gene may have important potential in developing new strategies to control herbivory pests in Lepidoptera and other insect orders.
Collapse
Affiliation(s)
- Zhen Yang
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Jinchan Ren
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuning Lu
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuanze Feng
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yongliang Fan
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Tong-Xian Liu
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiangfeng Jing
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Key Laboratory of Integrated Pest Management on Crops in Northwestern Loess Plateau of Ministry of Agriculture and Rural Affairs, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
3
|
Bodard Q, Fredon G, Riche A, Tessier X, Liberatore J. [Refractory immune thrombocytopenia revealing MYH9 related disease in a 64-year-old man]. Rev Med Interne 2025; 46:236-239. [PMID: 39939231 DOI: 10.1016/j.revmed.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/31/2024] [Accepted: 12/12/2024] [Indexed: 02/14/2025]
Abstract
INTRODUCTION MYH9 related disease is a rare autosomal dominant inherited disorder characterized in its full expression by thrombocytopenia, platelets macrocytosis, deafness, chronic kidney disease and cataract. CASE REPORT We present an original and didactic observation of refractory immune thrombocytopenia revealing a late diagnosis of MYH9 related disease. A 64-year-old man with longstanding thrombocytopenia less than 30 G/L without any history of bleeding, was treated with corticosteroids, intravenous immune globulin and thrombopoietin receptor agonists for a coronary angioplasty. The presence of platelets macrocytosis, increased mean platelet volume and deafness led to genetic testing of MYH9 related disease. CONCLUSION MYH9 related disease is the most common inherited thrombocytopenia. If no other symptoms are associated, it is an important differential diagnosis for immune thrombocytopenia that should be known.
Collapse
Affiliation(s)
- Quentin Bodard
- Service de médecine interne, maladies infectieuses et rhumatologie, centre hospitalier d'Angoulême, Angoulême, France.
| | - Guillaume Fredon
- Service de médecine interne, maladies infectieuses et rhumatologie, centre hospitalier d'Angoulême, Angoulême, France
| | - Agnès Riche
- Service de médecine interne, maladies infectieuses et rhumatologie, centre hospitalier d'Angoulême, Angoulême, France
| | - Xavier Tessier
- Laboratoire de biologie médicale, centre hospitalier d'Angoulême, Angoulême, France
| | - Johanne Liberatore
- Service de médecine interne, maladies infectieuses et rhumatologie, centre hospitalier d'Angoulême, Angoulême, France
| |
Collapse
|
4
|
Ruscica M, Loh WJ, Sirtori CR, Watts GF. Phytosterols and phytostanols in context: From physiology and pathophysiology to food supplementation and clinical practice. Pharmacol Res 2025; 214:107681. [PMID: 40049428 DOI: 10.1016/j.phrs.2025.107681] [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: 01/14/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Phytosterols and phytostanols are two classes of sterol derivatives naturally synthesised in plants, but not in humans. Structurally, phytosterols and phytostanols have a sterane ring in common, but phytostanols do not have a double bond between carbons 5 and 6. The therapeutic potential of phytosterols and phytostanols supplementation in cholesterol reduction is the main reason for its wide usage in an expansive food matrix, including milk, yoghurt, margarine, mayonnaise, chocolate, tartare, chips, esterification with omega-3, and recently, as a successful nutraceutical among athletes is its fortification with whey protein. The heterogeneous effect of phytosterols and phytostanols in cholesterol lowering appears to be related to whether the individuals' inherent physiologic tendencies to "hyper-synthesise" cholesterol in the liver or "hyperabsorb" cholesterol via the small intestine. Individuals who are 'hypersynthesizers" of cholesterol tend to have a good reduction in plasma low-density lipoprotein cholesterol (LDLc) in response to statin therapy. Conversely, "hyper-absorbers" of cholesterol show a greater LDLc lowering in response to phytosterols or phytostanols. The ratios of cholestanol to cholesterol and lathosterol to cholesterol are good biomarkers of intestinal absorption of cholesterol and hepatic cholesterol synthesis. Animal data and human observational data suggest that phytosterols and phytostanols may have anti-atherosclerotic activities, e.g. reduction of the formation of nitric oxide, antagonism to the formation of LDL aggregates and plaque formation. The absence of cardiovascular outcome trials using phytosterol or phytostanol supplementation, makes it difficult to confirm a wider use in clinical practice, especially with the rapidly expanding list of effective and safe lipid-lowering medications.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy; Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Wann Jia Loh
- School of Medicine, University of Western Australia, Australia; Department of Endocrinology, Changi General Hospital, Changi, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Australia; Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
5
|
Henry A, Mo X, Finan C, Chaffin MD, Speed D, Issa H, Denaxas S, Ware JS, Zheng SL, Malarstig A, Gratton J, Bond I, Roselli C, Miller D, Chopade S, Schmidt AF, Abner E, Adams L, Andersson C, Aragam KG, Ärnlöv J, Asselin G, Raja AA, Backman JD, Bartz TM, Biddinger KJ, Biggs ML, Bloom HL, Boersma E, Brandimarto J, Brown MR, Brunak S, Bruun MT, Buckbinder L, Bundgaard H, Carey DJ, Chasman DI, Chen X, Cook JP, Czuba T, de Denus S, Dehghan A, Delgado GE, Doney AS, Dörr M, Dowsett J, Dudley SC, Engström G, Erikstrup C, Esko T, Farber-Eger EH, Felix SB, Finer S, Ford I, Ghanbari M, Ghasemi S, Ghouse J, Giedraitis V, Giulianini F, Gottdiener JS, Gross S, Guðbjartsson DF, Gui H, Gutmann R, Hägg S, Haggerty CM, Hedman ÅK, Helgadottir A, Hemingway H, Hillege H, Hyde CL, Aagaard Jensen B, Jukema JW, Kardys I, Karra R, Kavousi M, Kizer JR, Kleber ME, Køber L, Koekemoer A, Kuchenbaecker K, Lai YP, Lanfear D, Langenberg C, Lin H, Lind L, Lindgren CM, Liu PP, London B, Lowery BD, Luan J, Lubitz SA, Magnusson P, Margulies KB, Marston NA, Martin H, März W, Melander O, Mordi IR, Morley MP, et alHenry A, Mo X, Finan C, Chaffin MD, Speed D, Issa H, Denaxas S, Ware JS, Zheng SL, Malarstig A, Gratton J, Bond I, Roselli C, Miller D, Chopade S, Schmidt AF, Abner E, Adams L, Andersson C, Aragam KG, Ärnlöv J, Asselin G, Raja AA, Backman JD, Bartz TM, Biddinger KJ, Biggs ML, Bloom HL, Boersma E, Brandimarto J, Brown MR, Brunak S, Bruun MT, Buckbinder L, Bundgaard H, Carey DJ, Chasman DI, Chen X, Cook JP, Czuba T, de Denus S, Dehghan A, Delgado GE, Doney AS, Dörr M, Dowsett J, Dudley SC, Engström G, Erikstrup C, Esko T, Farber-Eger EH, Felix SB, Finer S, Ford I, Ghanbari M, Ghasemi S, Ghouse J, Giedraitis V, Giulianini F, Gottdiener JS, Gross S, Guðbjartsson DF, Gui H, Gutmann R, Hägg S, Haggerty CM, Hedman ÅK, Helgadottir A, Hemingway H, Hillege H, Hyde CL, Aagaard Jensen B, Jukema JW, Kardys I, Karra R, Kavousi M, Kizer JR, Kleber ME, Køber L, Koekemoer A, Kuchenbaecker K, Lai YP, Lanfear D, Langenberg C, Lin H, Lind L, Lindgren CM, Liu PP, London B, Lowery BD, Luan J, Lubitz SA, Magnusson P, Margulies KB, Marston NA, Martin H, März W, Melander O, Mordi IR, Morley MP, Morris AP, Morrison AC, Morton L, Nagle MW, Nelson CP, Niessner A, Niiranen T, Noordam R, Nowak C, O'Donoghue ML, Ostrowski SR, Owens AT, Palmer CNA, Paré G, Pedersen OB, Perola M, Pigeyre M, Psaty BM, Rice KM, Ridker PM, Romaine SPR, Rotter JI, Ruff CT, Sabatine MS, Sallah N, Salomaa V, Sattar N, Shalaby AA, Shekhar A, Smelser DT, Smith NL, Sørensen E, Srinivasan S, Stefansson K, Sveinbjörnsson G, Svensson P, Tammesoo ML, Tardif JC, Teder-Laving M, Teumer A, Thorgeirsson G, Thorsteinsdottir U, Torp-Pedersen C, Tragante V, Trompet S, Uitterlinden AG, Ullum H, van der Harst P, van Heel D, van Setten J, van Vugt M, Veluchamy A, Verschuuren M, Verweij N, Vissing CR, Völker U, Voors AA, Wallentin L, Wang Y, Weeke PE, Wiggins KL, Williams LK, Yang Y, Yu B, Zannad F, Zheng C, Asselbergs FW, Cappola TP, Dubé MP, Dunn ME, Lang CC, Samani NJ, Shah S, Vasan RS, Smith JG, Holm H, Shah S, Ellinor PT, Hingorani AD, Wells Q, Lumbers RT. Genome-wide association study meta-analysis provides insights into the etiology of heart failure and its subtypes. Nat Genet 2025; 57:815-828. [PMID: 40038546 PMCID: PMC11985341 DOI: 10.1038/s41588-024-02064-3] [Show More Authors] [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: 10/09/2023] [Accepted: 12/17/2024] [Indexed: 03/06/2025]
Abstract
Heart failure (HF) is a major contributor to global morbidity and mortality. While distinct clinical subtypes, defined by etiology and left ventricular ejection fraction, are well recognized, their genetic determinants remain inadequately understood. In this study, we report a genome-wide association study of HF and its subtypes in a sample of 1.9 million individuals. A total of 153,174 individuals had HF, of whom 44,012 had a nonischemic etiology (ni-HF). A subset of patients with ni-HF were stratified based on left ventricular systolic function, where data were available, identifying 5,406 individuals with reduced ejection fraction and 3,841 with preserved ejection fraction. We identify 66 genetic loci associated with HF and its subtypes, 37 of which have not previously been reported. Using functionally informed gene prioritization methods, we predict effector genes for each identified locus, and map these to etiologic disease clusters through phenome-wide association analysis, network analysis and colocalization. Through heritability enrichment analysis, we highlight the role of extracardiac tissues in disease etiology. We then examine the differential associations of upstream risk factors with HF subtypes using Mendelian randomization. These findings extend our understanding of the mechanisms underlying HF etiology and may inform future approaches to prevention and treatment.
Collapse
Affiliation(s)
- Albert Henry
- Institute of Cardiovascular Science, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Xiaodong Mo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, London, UK
| | - Mark D Chaffin
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Doug Speed
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Hanane Issa
- Institute of Health Informatics, University College London, London, UK
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
- British Heart Foundation Data Science Centre, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - James S Ware
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Hammersmith Hospital, Imperial College Hospitals NHS Trust, London, UK
| | - Sean L Zheng
- National Heart & Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Anders Malarstig
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - Jasmine Gratton
- Institute of Cardiovascular Science, University College London, London, UK
| | - Isabelle Bond
- Institute of Cardiovascular Science, University College London, London, UK
| | - Carolina Roselli
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - David Miller
- Division of Biosciences, University College London, London, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, University College London, London, UK
| | - A Floriaan Schmidt
- Institute of Cardiovascular Science, University College London, London, UK
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Charlotte Andersson
- Department of Cardiology, Herlev Gentofte Hospital, Herlev, Denmark
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
| | - Krishna G Aragam
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society/Section of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
- School of Health and Social Sciences, Dalarna University, Falun, Sweden
| | | | - Anna Axelsson Raja
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Joshua D Backman
- Analytical Genetics, Regeneron Genetics Center, Tarrytown, NY, USA
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kiran J Biddinger
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mary L Biggs
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Heather L Bloom
- Department of Medicine, Division of Cardiology, Emory University Medical Center, Atlanta, GA, USA
| | - Eric Boersma
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeffrey Brandimarto
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael R Brown
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mie Topholm Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | | | - Henning Bundgaard
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - David J Carey
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Xing Chen
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Tomasz Czuba
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Simon de Denus
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Abbas Dehghan
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Alexander S Doney
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Samuel C Dudley
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Deparment of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Tõnu Esko
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Eric H Farber-Eger
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Sarah Finer
- Centre for Primary Care and Public Health, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Ian Ford
- Robertson Center for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sahar Ghasemi
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Jonas Ghouse
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John S Gottdiener
- Department of Medicine, Division of Cardiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stefan Gross
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Daníel F Guðbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Hongsheng Gui
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Rebecca Gutmann
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Åsa K Hedman
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | | | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
| | - Hans Hillege
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Craig L Hyde
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | | | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, the Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ravi Karra
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health System, and Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Lars Køber
- Department of Cardiology, Nordsjaellands Hospital, Copenhagen, Denmark
| | - Andrea Koekemoer
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Karoline Kuchenbaecker
- Division of Psychiatry, University College London, London, UK
- UCL Genetics Institute, University College London, London, UK
| | - Yi-Pin Lai
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - David Lanfear
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
- Heart and Vascular Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Honghuang Lin
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Peter P Liu
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barry London
- Division of Cardiovascular Medicine and Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| | - Brandon D Lowery
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Steven A Lubitz
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrik Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kenneth B Margulies
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas A Marston
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Hilary Martin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Olle Melander
- Department of Internal Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Malmö, Sweden
| | - Ify R Mordi
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Michael P Morley
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Lori Morton
- Cardiovascular Research, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alexander Niessner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Teemu Niiranen
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society/Section of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
| | - Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anjali T Owens
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colin N A Palmer
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, Finland
| | - Marie Pigeyre
- Population Health Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Simon P R Romaine
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, CA, USA
- Departments of Pediatrics and Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Christian T Ruff
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Neneh Sallah
- Institute of Health Informatics, University College London, London, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Naveed Sattar
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Alaa A Shalaby
- Department of Medicine, Division of Cardiology, University of Pittsburgh Medical Center and VA Pittsburgh HCS, Pittsburgh, PA, USA
| | - Akshay Shekhar
- Cardiovascular Research, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Diane T Smelser
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Nicholas L Smith
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Veterans Affairs Office of Research & Development, Seattle Epidemiologic Research and Information Center, Seattle, WA, USA
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sundararajan Srinivasan
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Per Svensson
- Department of Cardiology, Söderjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education-Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Mari-Liis Tammesoo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Alexander Teumer
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Guðmundur Thorgeirsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Internal Medicine, Division of Cardiology, National University Hospital of Iceland, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - David van Heel
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - Jessica van Setten
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marion van Vugt
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Abirami Veluchamy
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Monique Verschuuren
- Department Life Course and Health, Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Christoffer Rasmus Vissing
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Yunzhang Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Peter E Weeke
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kerri L Wiggins
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - L Keoki Williams
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Yifan Yang
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Faiez Zannad
- Université de Lorraine, CHU de Nancy, Inserm and INI-CRCT (F-CRIN), Institut Lorrain du Coeur et des Vaisseaux, Vandoeuvre Lès Nancy, France
| | - Chaoqun Zheng
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Folkert W Asselbergs
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Thomas P Cappola
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie-Pierre Dubé
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michael E Dunn
- Cardiovascular Research, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Chim C Lang
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Svati Shah
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Ramachandran S Vasan
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Sections of Cardiology, Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - J Gustav Smith
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Sonia Shah
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Patrick T Ellinor
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, University College London, London, UK
| | - Quinn Wells
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - R Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK.
- Health Data Research UK, London, UK.
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK.
| |
Collapse
|
6
|
Yuan X, Hou Y, Qin N, Xiang L, Jiang Z, Bao X. Flaxseed-derived peptide, Ile-Pro-Pro-Phe (IPPF), ameliorates hepatic cholesterol metabolism to treat metabolic dysfunction-associated steatotic liver disease by promoting cholesterol conversion and excretion. Food Funct 2025; 16:2808-2823. [PMID: 40094418 DOI: 10.1039/d4fo04478a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Flaxseed-derived peptide IPPF has been reported to effectively inhibit cholesterol micellization and reduce cholesterol accumulation in vitro. However, its effects on hepatic cholesterol accumulation and related dysfunction-associated steatotic liver disease (MASLD) in vivo, along with the underlying mechanisms and specific molecular targets, remain unclear. This study investigated the impact of IPPF on hepatic cholesterol accumulation to ameliorate MASLD and its potential mechanisms in vivo. Six-week-old male C57BL/6J mice were fed a high-cholesterol, high-fat diet and treated with different doses of IPPF via oral gavage for six weeks. IPPF intervention significantly reduced hepatic cholesterol levels and oxidative stress damage while increasing fecal cholesterol and bile acid excretion. Non-targeted metabolomics analysis revealed that IPPF primarily affected pathways related to ABC transporters and bile acid metabolism. IPPF intake upregulated the mRNA expression of Abcg5/8 and Cyp7a1 in the liver. Molecular docking, dynamics and Surface plasmon resonance (SPR) simulations demonstrated that IPPF binds strongly to ABCG5/8 and CYP7A1, forming stable complexes. Furthermore, cholesterol accumulation and MASLD in HepG2 cells induced by palmitic acid (PA) was alleviated by IPPF, but this effect was partly stopped when CYP7A1 or ABCG5/8 was inhibited. In conclusion, flaxseed-derived peptide IPPF targets CYP7A1 and ABCG5/8, promoting cholesterol conversion and excretion, thereby reducing hepatic cholesterol accumulation and offering a potential nutritional treatment for MASLD. IPPF can be used as a novel dietary cholesterol-lowering functional ingredient. This study provides a scientific basis and new perspective for the development of cholesterol-lowering functional foods and dietary supplements.
Collapse
MESH Headings
- Animals
- Male
- Mice, Inbred C57BL
- Mice
- Flax/chemistry
- Liver/metabolism
- Liver/drug effects
- Cholesterol/metabolism
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Cholesterol 7-alpha-Hydroxylase/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- Humans
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics
- Diet, High-Fat/adverse effects
- Bile Acids and Salts/metabolism
- Hep G2 Cells
- Molecular Docking Simulation
- Peptides/pharmacology
- Peptides/chemistry
- Oxidative Stress/drug effects
- Lipoproteins
Collapse
Affiliation(s)
- Xingyu Yuan
- Department of life science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China
| | - Yifeng Hou
- Department of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongoli, P. R. China.
| | - Narisu Qin
- Department of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongoli, P. R. China.
| | - Lu Xiang
- Department of life science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China
| | - Zhe Jiang
- Department of life science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China
| | - Xiaolan Bao
- Department of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongoli, P. R. China.
| |
Collapse
|
7
|
Schaefer EJ, Tedder B, Asztalos BF, Callu R, Geller AS, Diffenderfer MR, Roth M. Low-density lipoprotein cholesterol elevation, ketogenic diets, body mass index, and heterozygous ABCG5 genetic variation: Review, case report, and large population analysis. J Clin Lipidol 2025:S1933-2874(25)00060-1. [PMID: 40240243 DOI: 10.1016/j.jacl.2025.03.009] [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: 06/06/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Low-body mass index (BMI) has been associated with marked low-density-lipoprotein-cholesterol (LDL-C) elevations in response to very-low-carbohydrate diets (VLCD). METHODS We report a case (51-year-old woman, BMI 18.5 kg/m2) whose LDL-C was >500 mg/dL on a VLCD diet. We characterized her plasma lipoproteins and noncholesterol-sterols (gas chromatography/mass spectrometry) and the DNA sequences of her genes affecting lipid metabolism. We also carried out a large population analysis (224,126 subjects, 54% female, mean age 54 years) examining interrelationships between BMI and serum lathosterol/total cholesterol and β-sitosterol/total cholesterol ratios. RESULTS In this case, her LDL-C concentration increased from 142 mg/dL to 555 mg/dL on a VLCD, and her plasma β-sitosterol level was very high at 12.8 mg/L. DNA analysis revealed a heterozygous pathogenic ABCG5 exon 9 variant (c.1323_1324+2del at position g.44051049 TACAC>T). With dietary cholesterol restriction and ezetimibe therapy, her LDL-C and β-sitosterol levels decreased by 75% and 46% to 139 mg/dL and 7.1 mg/L, respectively. In the population analysis, we noted a significant inverse correlation between BMI and the plasma β-sitosterol/total cholesterol ratio (r = -0.573, P < .00001). Those with a BMI <20 kg/m2 had mean β-sitosterol/total cholesterol values that were significantly higher (+63%, P < .00001) than values in obese women. The converse was true for the plasma lathosterol/total cholesterol ratio. Similar findings were noted in men. CONCLUSIONS Our data are consistent with the concepts that low BMI predisposes to increased plasma β-sitosterol/total cholesterol ratios and an increased serum LDL-C when on high cholesterol VLCD diets, and that this response may be markedly enhanced in subjects with pathogenic heterozygous ABCG5 variants.
Collapse
Affiliation(s)
- Ernst J Schaefer
- Boston Heart Diagnostics/Eurofins Scientific Network, Framingham, MA, USA (Schaefer, Diffenderfer); Tufts University School of Medicine and Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA (Schaefer, Asztalos).
| | - Barry Tedder
- Department of Internal Medicine, St. Bernard's Regional Medical Center, Jonesboro, AR, USA (Tedder, Callu)
| | - Bela F Asztalos
- Tufts University School of Medicine and Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA (Schaefer, Asztalos)
| | - Ritu Callu
- Department of Internal Medicine, St. Bernard's Regional Medical Center, Jonesboro, AR, USA (Tedder, Callu)
| | - Andrew S Geller
- Clinical Enterprise/Eurofins Scientific Network, Framingham, MA, USA (Geller)
| | - Margaret R Diffenderfer
- Boston Heart Diagnostics/Eurofins Scientific Network, Framingham, MA, USA (Schaefer, Diffenderfer)
| | | |
Collapse
|
8
|
Anspach GB, Chauhan I, Savage EL, Poole B, Noffsinger VP, Fu X, Wang Z, Temel RE, Gordon SR, Helsley RN, Graf GA. ABCG5 ABCG8-independent mechanisms fail to maintain sterol balance in mice fed a high cholesterol diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637687. [PMID: 39990413 PMCID: PMC11844483 DOI: 10.1101/2025.02.11.637687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The ABCG5 ABCG8 (G5G8) sterol transporter opposes the accumulation of dietary xenosterols, but is also the primary mediator of biliary cholesterol secretion in the cholesterol elimination pathway. In humans and in mouse models of disrupted biliary cholesterol secretion, fecal neutral sterols remain constant, indicating the presence of a G5G8-independent mechanism for cholesterol excretion. Transintestinal cholesterol elimination (TICE) is thought to compensate for biliary G5G8 insufficiency. We sought to measure the compensatory increase in intestinal cholesterol secretion and provide mechanistic insight for how TICE maintains sterol balance in the absence of G5G8. Differences were not observed in fecal neutral sterols between control, acute, and chronic liver-specific G5G8 deficient mice (G5G8 LKO ). Cholesterol content did not differ at any point along the intestinal tract between genotypes. We also observed no change in the expression of apical or basolateral sterol transporting enzymes in the proximal small intestine. We then measured biliary and intestinal cholesterol secretion rates using cholesterol free and cholesterol enriched bile acid micelles as acceptors. While biliary cholesterol secretion was reduced, the intrinsic rate of intestinal cholesterol secretion did not differ between genotypes. G5G8 LKO and whole-body G5G8-deficient mice were challenged with a cholesterol-containing diet. While control mice upregulate fecal neutral sterol excretion, G5G8 LKO and G5G8 -/- mice fail to do so and accumulate sterol in the liver and plasma. These studies indicate that while G5G8-independent mechanisms can mediate cholesterol excretion, TICE is not upregulated in response to a loss of hepatic G5G8 and is unable to compensate for hepatic or whole-body G5G8-deficiency in response to dietary cholesterol in mice.
Collapse
|
9
|
Stellaard F, Lütjohann D. Phytosterol-Enriched Dietary Supplements for Lowering Plasma LDL-Cholesterol: Yes or No? Nutrients 2025; 17:654. [PMID: 40004982 PMCID: PMC11858453 DOI: 10.3390/nu17040654] [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: 12/08/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Elevated plasma low-density lipoprotein cholesterol (LDL-C) is associated with an increased risk for atherosclerosis and development of cardiovascular disease. An elevated plasma LDL-C concentration is the result of enhanced C synthesis, C absorption, and/or altered C homeostasis. Plasma LDL-C lowering can be achieved using pharmaceutical means. Statin therapy inhibits endogenous C synthesis and leads to a mean 40% LDL-C reduction. Ezetimibe inhibits C absorption and achieves an average 20% LDL-C reduction with a 10 mg daily intake. Phytosterol therapy is established by dietary supplements enriched in phytosterols and/or phytostanols. A dosage of 2 to 3 g a day reduces C absorption and leads to an average 10% LDL-C reduction. This dosage expresses a 10-fold increased daily intake for phytosterols or a 100-fold increased intake of phytostanols. Phytosterol- and -stanol-enriched dietary supplements are freely available in the supermarket. The majority of consumers may be healthy subjects with a plasma LDL-C in the normal range. Scientific evidence reveals that increased phytosterol intake may be associated with the development of atherosclerosis. The degree of increased risk is dependent on the patient's genetic polymorphisms in NPC1L1 and ABCG5/G8 transport proteins as well as on the established risk reduction due to LDL-C lowering. Subjects with a normal or only slightly elevated LDL-C have only minimal LDL-C lowering and lack the compensation for the potential increased risk for atherosclerosis by phytosterols.
Collapse
Affiliation(s)
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany;
| |
Collapse
|
10
|
Qiao W, Feng H, Zhang YF, Zhang Z, Yang J, Wu M, Xie J, Huang J, Zhou T, Zhang Y. Protective association between dietary phytosterol intake and cardiovascular health: an analysis of the UK Biobank cohort. Food Funct 2025; 16:1157-1168. [PMID: 39846866 DOI: 10.1039/d4fo05439c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Background: Cardiovascular diseases (CVDs) remain a leading cause of morbidity and mortality worldwide, with dietary interventions showing promise in reducing CVD risk factors. Phytosterols (PSs) in plant-based foods may reduce CVD risk by lowering low-density lipoprotein cholesterol. However, the relationship between dietary PS intake and CVD outcomes remains inconclusive. Methods: This study investigated the association between dietary PS intake and CVD outcomes, including coronary heart disease (CHD) and cardiovascular mortality, using a large cohort of 167 209 UK Biobank participants. PS intake was assessed through repeated 24 hour dietary recall data, with participants stratified into quintiles. The Cox proportional-hazards model was used to estimate hazard ratios (HRs) for CVD risk across quintiles of PS intake, adjusting for potential confounders. Restricted cubic splines were used to examine the nonlinear relationship between phytosterol intake and cardiovascular disease risk. Sensitivity and subgroup analyses explored interactions with demographic and lifestyle factors. Results: Higher dietary PS intake was significantly associated with a reduced risk of CVD events, including CHD and cardiovascular mortality. Each 100 mg increase in PS intake was linked to an 8% reduction in CVD risk (HR = 0.92, 95% CI: 0.87, 0.97). Multivariable-adjusted analyses revealed that participants in the highest quintile of PS intake had significantly lower CVD hazard ratios (HR = 0.81, 95% CI: 0.77, 0.84) compared to those in the lowest quintile. Significant inverse associations were also observed for cardiovascular mortality (HR: 0.86, 95% CI: 0.80, 0.94) and CHD (HR: 0.91, 95% CI: 0.84, 0.98). Subgroup analysis highlighted stronger inverse associations in current smokers, individuals with lower body mass index (BMI), and those with moderate to high physical activity levels, with variations observed based on dyslipidemia status. Sensitivity analyses, excluding early events and adjusting for energy intake, confirmed the robustness of the findings. Conclusions: This large cohort study provides evidence supporting the cardioprotective effects of dietary PS intake, particularly for CHD and cardiovascular mortality. Dietary PS may be considered an integral component of heart-healthy diets.
Collapse
Affiliation(s)
- Wanning Qiao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Hanxiao Feng
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Yi-Feng Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Zhilan Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Jinzhao Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Manni Wu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Jiyu Xie
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Juan Huang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, Guangdong, China
| | - Tao Zhou
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
- Guangdong Engineering Technology Research Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| |
Collapse
|
11
|
Huang X, Hao X, Wang T, Zhang X, Wu P, Shen L, Yang Y, Zhang W, Zhang K. Sex-related association between smoke exposure and gallstones in a US population: a cross-sectional study. BMC Public Health 2025; 25:344. [PMID: 39871261 PMCID: PMC11773891 DOI: 10.1186/s12889-024-21173-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/20/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Cholelithiasis are a condition that creates an economic and medical burden on society, and women are more susceptible to gallbladder stones. However, the effect of smoking on the development of gallstones remains controversial. No studies, to our knowledge, have discussed the association between smoke exposure and gallstones separately in men and women. We evaluated the association between smoking and gallstones in both sexes. METHODS We conducted a cross-sectional analysis using data obtained from the National Health and Nutrition Examination Survey from 2017 to March 2020. The analysis was limited to individuals aged ≥ 20 years with complete information available. Self-reported smoking status, serum cotinine concentration, and self-reported gallstones conditions were combined to analyze the association of gallstones with smoking and cotinine concentration. RESULTS Of the 6,982 participants, a total of 6.2% (212) men and 14.5% (512) women reported having gallstones. Logistic regression analysis showed smoking and high serum cotinine level were risk factors associated with gallstones among women, both in the model 1 (current smoker: odds ratio [OR] = 1.563, 95% confidence interval [CI] = 1.044-2.339, p = 0.032; former smoker: OR = 1.434, 95% CI = 1.116-1.842, p = 0.007; cotinine ≥ 3 ng/mL: OR = 1.800, 95% CI = 1.247-2.596, p = 0.005; and cotinine 0.05-2.99 ng/mL: OR = 1.640, 95% CI = 1.188-2.263, p = 0.005) and model 2 (current smoker: OR = 1.588, 95% CI = 1.015-2.483, p = 0.044; cotinine ≥ 3 ng/mL: OR = 1.825; 95% CI = 1.181-2.821, p = 0.011; and cotinine 0.05-2.99 ng/mL: OR = 1.509, 95% CI = 1.075-2.126, p = 0.022). However, the association was statistically insignificant in men. The subgroup analysis showed the robustness of the association. CONCLUSIONS This study indicates smoking and elevated serum cotinine levels may be risk factors associated with the development of gallstones. Notably, the associations were specifically observed among women. The findings suggest the significance of smoking in the incidence of gallstones, which may potentially provide insights for future research on strategies to prevent gallstones, particularly among women. The validation of these findings necessitates the conduction of large-scale, high-quality prospective studies.
Collapse
Affiliation(s)
- Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Xuanyu Hao
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Xiaoyue Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Wanchuan Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China.
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive, Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
12
|
Lv G, Wang D, Huang Y, Shi R, Qin C, Chen X, Zeng X, Luo H, Yang P, Chen S, Wang J. High serum uric acid levels are associated with increased prevalence of gallstones in adult women: a cross-sectional study based on NHANES. Front Med (Lausanne) 2025; 12:1487974. [PMID: 39897593 PMCID: PMC11782260 DOI: 10.3389/fmed.2025.1487974] [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: 08/29/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Objective We investigated the association between serum uric acid (SUA) levels and gallstone (GS) prevalence in adult women. Methods Participants' information were taken from the United States National Health and Nutrition Examination Survey (2017-2020). Logistic regression analysis and dose-response curve were used to assess the association between SUA levels and the prevalence of GS in adult women. Subgroup analyses were performed to investigate associations between SUA levels and age, ethnicity, body mass index, hypertension, and diabetes. Results A total of 600 participants self-reported a history of GS. After adjusting for confounding, the prevalence of GS in adult women increased by 14% for every 1 mg/dL increase in SUA (odds ratio [OR]: 1.14, 95% confidence interval [CI]: 1.06, 1.22). Testing SUA as a categorical variable for sensitivity analyses indicated a 1.6-fold increase in the prevalence of GS in tertile 3 (OR=1.60, 95% CI: 1.25, 2.04) compared to tertile 1. Dose-response curves showed a nonlinear correlation between SUA levels and the prevalence of GS. Subgroup analyses indicated that SUA level was associated with an increased prevalence of GS in most subgroups, although subtle differences existed. Conclusion SUA was positively and non-linearly associated with the prevalence of GS in adult females. Despite the inability to clarify the causal relationship between them, our results remain interesting.
Collapse
Affiliation(s)
- Guozheng Lv
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Decai Wang
- Department of Urology, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Yu Huang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Ruizi Shi
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Chuan Qin
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Xi Chen
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Xintao Zeng
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Hua Luo
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Pei Yang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Sirui Chen
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Jianjun Wang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
13
|
Do TTM, Vu CD, Dien TM, Can TBN, Nguyen TTN, Nguyen HH, Tran VK, Nguyen NL, Tran HT, Mai TTC, Nguyen KN. Phenotypes, Genotypes, Treatment, and Outcomes of 14 Children with Sitosterolemia at Vietnam National Children's Hospital. J Clin Med 2025; 14:325. [PMID: 39860331 PMCID: PMC11765834 DOI: 10.3390/jcm14020325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Sitosterolemia is a rare autosomal recessive disorder characterized by diverse clinical manifestations ranging from asymptomatic cases to the development of xanthomas, hypercholesterolemia, premature atherosclerosis, or even sudden death during childhood. It results from homozygous or compound heterozygous pathogenic variants in the ABCG5 or ABCG8 genes. Prompt detection and intervention are essential to managing this condition and preventing severe outcomes. Methods: This study aims to retrospectively analyze the phenotype, genotype, treatment, and outcomes of 14 children-seven boys and seven girls-all of Vietnamese origin, diagnosed with sitosterolemia at the Vietnam National Children's Hospital between March 2015 and July 2024. Results: The median ages at disease onset and diagnosis were 5.7 years (range: 1.5-17.9) and 7.2 years (range: 1.7-17.9), respectively. Xanthomas were observed in 85.7% of patients (12/14), arthralgia in 14.3% (2/14), and anemia in 7.1% (1/14), with no cases of thrombocytopenia. At diagnosis, all patients exhibited elevated total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C), with considerably higher levels in patients with xanthomas compared to those without. Mutations in the ABCG5 gene were identified in 71.4% (10/14) of the patients, while 28.6% (4/14) had mutations in the ABCG8 gene. Fourteen variants were detected, nine in ABCG5 and five in ABCG8, with five variants reported for the first time in sitosterolemia patients. Initial management for all patients involved dietary modifications. After three months, 10 patients with persistently elevated TC and LDL-C received ezetimibe or cholestyramine treatment. Among the eight patients who continued treatment for over three months, the median TC and LDL-C concentrations decreased by 54.9% and 67.3%, respectively. Conclusions: Among Vietnamese patients with sitosterolemia, variants in the ABCG5 gene were more prevalent than those in the ABCG8 gene. Patients showed a positive response to ezetimibe or cholestyramine treatment. Genetic testing is essential for establishing a diagnosis of sitosterolemia and guiding accurate management strategies.
Collapse
Affiliation(s)
- Thi Thanh Mai Do
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Chi Dung Vu
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Tran Minh Dien
- Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Thi Bich Ngoc Can
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Thi Thanh Ngan Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 100000, Vietnam; (T.T.N.N.); (H.H.N.)
| | - Huy Hoang Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 100000, Vietnam; (T.T.N.N.); (H.H.N.)
| | - Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (V.K.T.); (N.L.N.)
| | - Ngoc Lan Nguyen
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (V.K.T.); (N.L.N.)
| | - Huy Thinh Tran
- Biochemistry Department, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam;
| | - Tran Thi Chi Mai
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
| | - Khanh Ngoc Nguyen
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| |
Collapse
|
14
|
Gu Q, Wang Y, Yi P, Cheng C. Theoretical framework and emerging challenges of lipid metabolism in cancer. Semin Cancer Biol 2025; 108:48-70. [PMID: 39674303 DOI: 10.1016/j.semcancer.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Elevated lipid metabolism is one of hallmarks of malignant tumors. Lipids not only serve as essential structural components of biological membranes but also provide energy and substrates for the proliferation of cancer cells and tumor growth. Cancer cells meet their lipid needs by coordinating the processes of lipid absorption, synthesis, transport, storage, and catabolism. As research in this area continues to deepen, numerous new discoveries have emerged, making it crucial for scientists to stay informed about the developments of cancer lipid metabolism. In this review, we first discuss relevant concepts and theories or assumptions that help us understand the lipid metabolism and -based cancer therapies. We then systematically summarize the latest advancements in lipid metabolism including new mechanisms, novel targets, and up-to-date pre-clinical and clinical investigations of anti-cancer treatment with lipid metabolism targeted drugs. Finally, we emphasize emerging research directions and therapeutic strategies, and discuss future prospective and emerging challenges. This review aims to provide the latest insights and guidance for research in the field of cancer lipid metabolism.
Collapse
Affiliation(s)
- Qiuying Gu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Chunming Cheng
- Department of Oncology Science, OU Health Stephenson Cancer Center at University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
15
|
Hazra S, Chakraborthy G. Effects of Diabetes and Hyperlipidemia in Physiological Conditions - A Review. Curr Diabetes Rev 2025; 21:24-34. [PMID: 38409688 DOI: 10.2174/0115733998289406240214093815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Diabetes mellitus (DM) is an autoimmune manifestation defined by persistent hyperglycemia and alterations in protein, fatty substances, and carbohydrate metabolism as an effect of problems with the secretion of insulin action or both. Manifestations include thirst, blurred eyesight, weight loss, and ketoacidosis, which can majorly lead to coma. There are different types of diabetes according to class or by cellular level. They are interrelated with hyperlipidemia as they are involved in the metabolism and regulation of physiological factors. Most parameters are seen at cellular or humoral levels, yet the underlying concern remains the same. OBJECTIVE To create a systematic correlation between the disease and locate the exact mechanism and receptors responsible for it. So, this article covers a proper way to resolve the conditions and their manifestation through literacy and diagrammatic. CONCLUSION Hence, this will be an insight for many scholars to understand the exact mechanism involved in the process.
Collapse
Affiliation(s)
- Sayan Hazra
- Department of Pharmacology, Parul Institute of Pharmacy and Research, Parul University, Vadodara, Gujarat, 391760, India
| | - Gunosindhu Chakraborthy
- Parul Institute of Pharmacy and Research, Parul University, Vadodara, Gujarat, 391760, India
| |
Collapse
|
16
|
Wei M, LYu P, Li P, Hu J, Wu R, Ouyang Q, Guo K. Baolier Capsule's Secret Weapon: Piperine Boosts Cholesterol Excretion to Combat Atherosclerosis. Drug Des Devel Ther 2024; 18:6427-6446. [PMID: 39749187 PMCID: PMC11693944 DOI: 10.2147/dddt.s499598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Purpose The Baolier capsule (BLEC) is a proprietary Mongolian medicine administered for treating hypercholesterolemia and atherosclerosis (AS). However, the therapeutic effects, primary bioactive ingredients, and potential mechanisms underlying hypercholesterolemia and AS remain unclear. This study aimed to investigate the pharmacological effects, principal active ingredients, and mechanisms of BLEC against hypercholesterolemia and AS. Methods Adeno-associated virus tail vein injection was utilized to construct liver-specific LXRα knockout ApoE-/- mice. A high-fat diet was utilized to feed ApoE-/- mice to build hyperlipidemia and AS mouse models. The aorta or liver stained with Oil Red O was used to assess the effect of the drugs on AS or fatty liver formation after the oral administration of BLEC, piperine, statins, or ezetimibe to the mice following the experimental protocol. Biochemical assays were utilized to evaluate the effect of the drugs on serum lipid levels and cholesterol efflux indicators. Transcriptomics was employed to investigate the effect of BLEC on liver gene expression levels. HPLC-MS/MS was used to determine BLEC and its major components in the liver. Western blotting or quantitative reverse transcription polymerase chain reaction was conducted to detect LXRα, ABCA1, ABCG5, ABCG8, and CYP7A1 expression. Results Here, we revealed that BLEC decreases lipid levels in the serum and liver, as well as decelerates AS by promoting cholesterol excretion. BLEC and piperine, which are the main components exposed in the target liver tissue, activate LXRα to upregulate ABCA1, ABCG5, ABCG8, and CYP7A1, which promotes cholesterol transport to high-density lipoprotein and excretion to bile and feces. Notably, piperines demonstrated synergistic beneficial effects with atorvastatin or ezetimibe, which are two widely used hypocholesterolemic and anti-atherosclerotic drugs. Conclusion BLEC and its main active ingredient, piperine, promote cholesterol excretion, reduce serum cholesterol levels, inhibit AS, and exhibit good clinical application value and prospects.
Collapse
Affiliation(s)
- Mengqiu Wei
- Intensive Care Unit, Zhongshan City People’s Hospital, Zhongshan, 528400, People’s Republic of China
| | - Ping LYu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Peng Li
- Department of Geriatrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, People’s Republic of China
| | - Jing Hu
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, People’s Republic of China
| | - Ruozhuo Wu
- School of Biological and Pharmaceutical Engineering, Wuhan Huaxia Institute of Technology, Wuhan, 430223, People’s Republic of China
| | - Qingqing Ouyang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Kai Guo
- Department of Cardiology, The Seventh Affiliated Hospital of Southern Medical University, Southern Medical University, Foshan, 528244, People’s Republic of China
| |
Collapse
|
17
|
Du X, Zeng Y, Li Y, Peng Q, Miao J, Liu X. A Protein with Unknown Function, Ps495620, Is Critical for the Sporulation and Oospore Production of Phytophthora sojae. J Fungi (Basel) 2024; 11:12. [PMID: 39852431 PMCID: PMC11766772 DOI: 10.3390/jof11010012] [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: 11/25/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025] Open
Abstract
While the rapid rise in bioinformatics has facilitated the identification of the domains and functions of many proteins, some still have no domain annotation or largely uncharacterized functions. However, the biological roles of unknown proteins were not clear in oomycetes. An analysis of the Phytophthora sojae genome database identified the protein Ps495620, which has no domain annotations and functional predictions in Phytophthora. This study used a CRISPR/Cas9-mediated gene replacement system to knock out Ps495620 to elucidate its function. The Ps495620-knockout mutants exhibited significantly increased oospore production and decreased sporangium formation compared to the wild-type strain P6497. Transcriptomics showed that it is a key regulator of nitrogen, pyruvate, ascorbate, and adorate metabolism in P. sojae. Our findings indicate that Ps495620 is critical in regulating sporangium formation and oospore production in P. sojae.
Collapse
Affiliation(s)
- Xiaoran Du
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Xianyang 712100, China; (X.D.); (Y.Z.); (Y.L.); (Q.P.)
| | - Yan Zeng
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Xianyang 712100, China; (X.D.); (Y.Z.); (Y.L.); (Q.P.)
| | - Yiying Li
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Xianyang 712100, China; (X.D.); (Y.Z.); (Y.L.); (Q.P.)
| | - Qin Peng
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Xianyang 712100, China; (X.D.); (Y.Z.); (Y.L.); (Q.P.)
| | - Jianqiang Miao
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Xianyang 712100, China; (X.D.); (Y.Z.); (Y.L.); (Q.P.)
| | - Xili Liu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Xianyang 712100, China; (X.D.); (Y.Z.); (Y.L.); (Q.P.)
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, 2 Yuanmingyuanxi Road, Beijing 100193, China
| |
Collapse
|
18
|
Wang J, Shen Z, Liang Y, Qin C, Chen S, Shi R, Huang Y, Chen X, Luo H, Yang P, Gong J, Zeng X, Wang D. Association of neutrophil percentage to albumin ratio with gallstones: a cross-sectional study from the United States NHANES. BMC Public Health 2024; 24:3503. [PMID: 39696262 PMCID: PMC11657001 DOI: 10.1186/s12889-024-21071-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: 08/08/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVE The neutrophil percentage to albumin ratio (NPAR) is an emerging, costimulatory indicator of inflammation that is associated with a variety of diseases, such as non-alcoholic fatty liver disease, liver fibrosis, stroke, and cardiovascular disease. However, the relationship between NPAR and gallstones (GS) has not yet been explored. Therefore, this study aimed to evaluate the association of the NPAR with the odds of GS and the age of patients at the time of their first GS surgery. METHODS Participants were selected from the National Health and Nutrition Examination Survey (NHANES) in the United States, a nationally representative survey. Logistic regression analysis and dose-response curve were performed to analyze the relationship between NPAR and the prevalence of GS. Multiple linear regression analysis and dose-response curve were used to analyze the association between NPAR and the age of patients at the time of their first GS surgery. Subgroup analyses further explored the relationships between NPAR and age, sex, race, body mass index, hypertension, and diabetes. RESULTS In total, 7805 adults aged > 20 years were included in this study, of whom 838 had a history of GS. After adjusting for all potential confounders, each 1-unit increase in NPAR was found to be associated with a 4% increase in the prevalence of GS (odds ratio (OR): 1.04, 95% confidence interval (CI): 1.02, 1.07) and an advancement in the age of the patient at the time of the first GS surgery by 0.35 years (β = - 0.35, 95% CI: - 0.68, - 0.02). Dose-response curves further confirmed that NPAR was positively associated with the prevalence of GS and negatively associated with the age of patients at the time of their first GS surgery. The results of the subgroup analyses suggested that after adjusting for all potential confounders, the positive association of NPAR with the prevalence of GS was more pronounced in the 40-59-year-old (OR: 1.07, 95% CI: 1.02, 1.12), male (OR: 1.07, 95% CI: 1.02, 1.12), non-Hispanic Black (OR: 1.06, 95% CI: 1.01, 1.12), non-hypertensive (OR: 1.06, 95% CI: 1.02, 1.10), and non-diabetic populations (OR=: 1.05, 95% CI: 1.02, 1.08). CONCLUSIONS The higher the NPAR, the higher the prevalence of GS, and the earlier the age of the patient at the time of the first GS surgery. Due to the nature of cross-sectional study, it is not possible to determine a causal relationship between them.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhiwen Shen
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, 317000, China
| | - Yuwen Liang
- Department of Oncology, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Chuan Qin
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Sirui Chen
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Ruizi Shi
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Yu Huang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Xi Chen
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Hua Luo
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Pei Yang
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Jianping Gong
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Xintao Zeng
- Department of Hepatobiliary Surgery, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China.
| | - Decai Wang
- NHC Key Laboratory of Nuclear Technology Medical Transformation, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China.
- Department of Urology, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, China.
| |
Collapse
|
19
|
Elbahnsi A, Dudas B, Callebaut I, Hinzpeter A, Miteva MA. ATP-Binding Cassette and Solute Carrier Transporters: Understanding Their Mechanisms and Drug Modulation Through Structural and Modeling Approaches. Pharmaceuticals (Basel) 2024; 17:1602. [PMID: 39770445 PMCID: PMC11676857 DOI: 10.3390/ph17121602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The ATP-binding cassette (ABC) and solute carrier (SLC) transporters play pivotal roles in cellular transport mechanisms, influencing a wide range of physiological processes and impacting various medical conditions. Recent advancements in structural biology and computational modeling have provided significant insights into their function and regulation. This review provides an overview of the current knowledge of human ABC and SLC transporters, emphasizing their structural and functional relationships, transport mechanisms, and the contribution of computational approaches to their understanding. Current challenges and promising future research and methodological directions are also discussed.
Collapse
Affiliation(s)
- Ahmad Elbahnsi
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| | - Balint Dudas
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| | - Isabelle Callebaut
- Muséum National d’Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie—IMPMC, Sorbonne Université, 75005 Paris, France
| | - Alexandre Hinzpeter
- CNRS, INSERM, Institut Necker Enfants Malades—INEM, Université Paris Cité, 75015 Paris, France
| | - Maria A. Miteva
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| |
Collapse
|
20
|
Yamanashi Y, Komine T, Hirota Y, Suzuki H, Osuga Y, Takada T. Dietary phytosterols induce infertility in female mice via epigenomic modulations. Commun Biol 2024; 7:1535. [PMID: 39562830 DOI: 10.1038/s42003-024-07233-y] [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: 05/20/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Dietary modifications to overcome infertility have attracted attention; however, scientifically substantiated information on specific dietary components affecting fertility and their mechanisms is limited. Herein, we investigated diet-induced, reversible infertility in female mice lacking the heterodimer of ATP-binding cassette transporters G5 and G8 (ABCG5/G8), which functions as a lipid exporter in the intestine. We found that dietary phytosterols, especially β-sitosterol and brassicasterol, which are substrates of ABCG5/G8, have potent but reversible reproductive toxicities in mice. Mechanistically, these phytosterols inhibited ovarian folliculogenesis and reduced egg quality by enhancing polycomb repressive complex 2-mediated histone H3 trimethylation at lysine 27 in the ovary. Clinical analyses showed that serum phytosterol levels were significantly and negatively correlated with the blastocyst development rate of fertilized eggs in women undergoing in vitro fertilization, suggesting that phytosterols affect egg quality in both humans and mice. Thus, avoiding excessive intake of certain phytosterols would be beneficial for female reproductive health.
Collapse
Affiliation(s)
- Yoshihide Yamanashi
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.
| | - Toko Komine
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
21
|
Chen Z, Ding H, Zhu H, Huang S, Yan C, Chen ZY. Additional mechanism for selective absorption of cholesterol and phytosterols. Food Chem 2024; 458:140300. [PMID: 38964108 DOI: 10.1016/j.foodchem.2024.140300] [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: 05/15/2024] [Revised: 06/16/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Phytosterols are structurally similar to cholesterol but they are much less absorbed (<2%) than cholesterol (>50%) in the intestine. We hypothesize that phytosterols are poor substrates of intestinal acyl-CoA: cholesterol acyltransferase 2 (ACAT2), and thus minimal phytosterol esters are formed and packed into chylomicrons, leading to their low absorption. Two isotope tracing models, including a radioactive hamster microsomal ACAT2 reaction model and a differentiated Caco-2 cell model, were established to examine the specificity of ACAT2 to various sterols, including cholesterol, sitosterol, stigmasterol, and campesterol. Both models consistently demonstrated that only cholesterol but not phytosterols could be efficiently esterified by ACAT2 in a time- and dose-dependent manner. Molecular docking further suggested that unfavorable interactions existed between ACAT2 and phytosterols. In conclusion, phytosterols are poor substrates of ACAT2 and thus minimally absorbed. This work provides a theoretical basis for the use of phytosterol-based supplements in treating dyslipidemia and preventing heart diseases.
Collapse
Affiliation(s)
- Zixing Chen
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Huafang Ding
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Hanyue Zhu
- School of Food Science and Engineering / Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Shouhe Huang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| |
Collapse
|
22
|
Li T, Chiang JYL. Bile Acid Signaling in Metabolic and Inflammatory Diseases and Drug Development. Pharmacol Rev 2024; 76:1221-1253. [PMID: 38977324 PMCID: PMC11549937 DOI: 10.1124/pharmrev.124.000978] [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: 04/16/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
Bile acids are the end products of cholesterol catabolism. Hepatic bile acid synthesis accounts for a major fraction of daily cholesterol turnover in humans. Biliary secretion of bile acids generates bile flow and facilitates biliary secretion of lipids, endogenous metabolites, and xenobiotics. In intestine, bile acids facilitate the digestion and absorption of dietary lipids and fat-soluble vitamins. Through activation of nuclear receptors and G protein-coupled receptors and interaction with gut microbiome, bile acids critically regulate host metabolism and innate and adaptive immunity and are involved in the pathogenesis of cholestasis, metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, type-2 diabetes, and inflammatory bowel diseases. Bile acids and their derivatives have been developed as potential therapeutic agents for treating chronic metabolic and inflammatory liver diseases and gastrointestinal disorders. SIGNIFICANCE STATEMENT: Bile acids facilitate biliary cholesterol solubilization and dietary lipid absorption, regulate host metabolism and immunity, and modulate gut microbiome. Targeting bile acid metabolism and signaling holds promise for treating metabolic and inflammatory diseases.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| | - John Y L Chiang
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| |
Collapse
|
23
|
Dong H, Chen R, Wang J, Chai N, Linghu E. Can NPC1L1 inhibitors reduce the risk of biliary tract cancer? Evidence from a mendelian randomization study. Dig Liver Dis 2024; 56:1599-1604. [PMID: 38342741 DOI: 10.1016/j.dld.2024.01.211] [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: 08/30/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND & AIMS Oxysterols have been implicated in biliary tract cancer (BTC), and Niemann-Pick C1-like 1 (NPC1L1) has been associated with oxysterol uptake in biliary and intestinal cells. Thus, our study aims to investigate the potential causal link between genetically proxied NPC1L1 inhibitors and the risk of BTC. METHODS In this study, we employed two genetic instruments as proxies for NPC1L1 inhibitors, which included LDL cholesterol-associated genetic variants located within or in close proximity to the NPC1L1 gene, as well as expression quantitative trait loci (eQTLs) of the NPC1L1 gene. Effect estimates were calculated using the Inverse-variance-weighted MR (IVW-MR) and summary-data-based MR (SMR) methods. RESULTS In MR analysis using the IVW method, both proxy instruments from the UK Biobank and the GLGC demonstrated a positive association between NPC1L1-mediated LDL cholesterol and BTC risk, with odds ratios (OR) of 10.30 (95% CI = 1.51-70.09; P = 0.017) and 5.61 (95% CI = 1.43-21.91; P = 0.013), respectively. Moreover, SMR analysis revealed a significant association between elevated NPC1L1 expression and increased BTC risk (OR = 1.19, 95% CI = 1.04-1.37; P = 0.014). CONCLUSIONS This MR study suggests a causal link between NPC1L1 inhibition and reduced BTC risk. NPC1L1 inhibitors, like ezetimibe, show potential for chemoprevention in precancerous BTC patients, requiring further clinical investigation.
Collapse
Affiliation(s)
- Hao Dong
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Rong Chen
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Jiafeng Wang
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Ningli Chai
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, PR China.
| | - Enqiang Linghu
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, PR China.
| |
Collapse
|
24
|
Kwon GE, Son HH, Moon JY, Lee A, Jung MK, Rhie S, Park MJ, Garg A, Yoo EG, Choi MH. Dried blood spot-based free sterol signatures in sitosterolemia diagnostics. Clin Chim Acta 2024; 562:119886. [PMID: 39053727 DOI: 10.1016/j.cca.2024.119886] [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: 08/25/2023] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Sitosterolemia is a rare inherited lipid metabolic disorder characterized by increased levels of plant sterols and accelerated atherosclerosis. Although early detection is beneficial for the prevention of disease progression, it is largely underdiagnosed by routine screening based on conventional lipid profiles. MATERIALS AND METHODS A gas chromatography-mass spectrometry (GC-MS)-based profiling has been developed and validated to measure the levels of biologically active free sterols, including five endogenous sterols and three plant sterols (sitosterol, campesterol, and stigmasterol) in dried blood spot (DBS). RESULTS Within- and between-run precisions were 1.4-11.1 % and 2.2-14.1 %, respectively, while the accuracies were all 86.3 ∼ 121.9 % with the correlation coefficients (r2) > 0.988 for all the sterols. In the patients (four girls and two boys, 6.5 ± 2.8 years), sitosterol levels were significantly increased, with an optimal cut-off value of 2.5 µg/mL distinguishing them from ninety-three age-matched healthy children. A cut-off value of 31.9 µg/mL differentiated the patients from six ABCG5/ABCG8 heterozygous carriers. In addition, the molecular ratios of sitosterol to cholesterol, desmosterol, and 7-dehydrocholesterol provided excellent cut-off values of 26.3, 67.6, and 21.6, respectively, to distinguish patients from both healthy controls and heterozygous carriers. CONCLUSIONS The novel DBS-based GC-MS profiling of free sterols accurately identified patients with sitosterolemia, with a performance comparable to that of a serum assay. The DBS profiling could be more feasible method in clinical practice as well as population screening programs, and it can provide diagnostic cut-off values for individual plant sterols.
Collapse
MESH Headings
- Humans
- Lipid Metabolism, Inborn Errors/blood
- Lipid Metabolism, Inborn Errors/diagnosis
- Female
- Male
- Intestinal Diseases/blood
- Intestinal Diseases/diagnosis
- Gas Chromatography-Mass Spectrometry
- Child
- Phytosterols/blood
- Phytosterols/adverse effects
- Dried Blood Spot Testing/methods
- Hypercholesterolemia/blood
- Hypercholesterolemia/diagnosis
- Child, Preschool
- ATP Binding Cassette Transporter, Subfamily G, Member 5/blood
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- Sterols/blood
- ATP Binding Cassette Transporter, Subfamily G, Member 8/blood
- ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics
- Lipoproteins/blood
Collapse
Affiliation(s)
- Go Eun Kwon
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Hyun-Hwa Son
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju-Yeon Moon
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Ayoung Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Mo Kyung Jung
- Department of Pediatrics, CHA Bundang Medical Center, Gyeonggi-do 13496, Republic of Korea
| | - Seonkyeong Rhie
- Department of Pediatrics, CHA Bundang Medical Center, Gyeonggi-do 13496, Republic of Korea
| | - Mi Jung Park
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul 01757, Republic of Korea
| | - Abhimanyu Garg
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eun-Gyong Yoo
- Department of Pediatrics, CHA Bundang Medical Center, Gyeonggi-do 13496, Republic of Korea.
| | - Man Ho Choi
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
25
|
Alenbawi J, Al-Sarraj YA, Umlai UKI, Kadhi A, Hendi NN, Nemer G, Albagha OME. Genome-wide association study and meta-analysis of phytosterols identifies a novel locus for serum levels of campesterol. Hum Genomics 2024; 18:85. [PMID: 39090729 PMCID: PMC11295598 DOI: 10.1186/s40246-024-00649-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Sitosterolemia is a rare inherited disorder caused by mutations in the ABCG5/ABCG8 genes. These genes encode proteins involved in the transport of plant sterols. Mutations in these genes lead to decreased excretion of phytosterols, which can accumulate in the body and lead to a variety of health problems, including premature coronary artery disease. We conducted the first genome-wide association study (GWAS) in the Middle East/North Africa population to identify genetic determinants of plant sterol levels in Qatari people. GWAS was performed on serum levels of β-sitosterol and campesterol using the Metabolon platform from Qatar Biobank (QBB) and genome sequence data provided by Qatar Genome Program. A trans-ancestry meta-analysis of data from our Qatari cohort with summary statistics from a previously published large cohort (9758 subjects) of European ancestry was conducted. Using conditional analysis, we identified two independent single nucleotide polymorphisms associated with β-sitosterol (rs145164937 and rs4299376), and two others with campesterol (rs7598542 and rs75901165) in the Qatari population in addition to previously reported variants. All of them map to the ABCG5/8 locus except rs75901165 which is located within the Intraflagellar Transport 43 (IFT43) gene. The meta-analysis replicated most of the reported variants, and our study provided significant support for the association of variants in SCARB1 and ABO with sitosterolemia. Evaluation of a polygenic risk score devised from European GWAS data showed moderate performance when applied to QBB (adjusted-R2 = 0.082). These findings provide new insights into the genetic architecture of phytosterol metabolism while showing the importance including under-represented populations in future GWAS studies.
Collapse
Affiliation(s)
- Jamil Alenbawi
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| | - Yasser A Al-Sarraj
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
- Qatar Genome Program (QGP), Qatar Foundation Research, Development and Innovation, Qatar Foundation (QF), P.O. Box 5825, Doha, Qatar
| | - Umm-Kulthum I Umlai
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| | - Ayat Kadhi
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
- College of Health and Sciences, University of Doha for Science and Technology, P.O. Box 24449, Doha, Qatar
| | - Nagham N Hendi
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| | - Georges Nemer
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar.
| | - Omar M E Albagha
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar.
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
26
|
Onuma K, Watanabe K, Isayama K, Ogi S, Tokunaga Y, Mizukami Y. Bardoxolone methyl prevents metabolic dysfunction-associated steatohepatitis by inhibiting macrophage infiltration. Br J Pharmacol 2024; 181:2545-2565. [PMID: 38599607 DOI: 10.1111/bph.16374] [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: 10/20/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Bardoxolone methyl (2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid methyl ester, CDDO-Me) is a potent activator of nuclear factor erythroid 2-related factor 2 (Nrf2), which induces the expression of antioxidative-associated genes. CDDO-Me exerts protective effects against chronic inflammatory diseases in the kidneys and lungs. However, its pharmacological effects on metabolic dysfunction-associated steatohepatitis (MASH) caused by fat accumulation remain unknown. In this study, we examined the hepatoprotective effects of CDDO-Me in a diet-induced MASH mouse model and elucidated its pharmacological mechanisms using RNA-seq analysis. EXPERIMENTAL APPROACH CDDO-Me was orally administered to mice fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD), and histological, biochemical, and transcriptomic analyses were performed on livers of mice that developed MASH. KEY RESULTS CDDO-Me administration induced the expression of antioxidant genes and cholesterol transporters downstream of Nrf2 and significantly prevented the symptoms of MASH. Whole-transcriptome analysis revealed that CDDO-Me inhibited the inflammatory pathway that led to phagocyte recruitment, in addition to activating the Nrf2-dependent pathway. Among inflammatory pathways, CC chemokine ligands (CCL)3 and CCL4, which are downstream of NF-κB and are associated with the recruitment of macrophages expressing CC chemokine receptors (CCR)1 and CCR5, were released into the blood in MASH mice. However, CDDO-Me directly inhibited the expression of CCL3-CCR1 and CCL4-CCR5 in macrophages. CONCLUSIONS AND IMPLICATIONS Overall, we revealed the potent hepatoprotective effect of CDDO-Me in a MASH mouse model and demonstrated that its pharmacological effects were closely associated with a reduction of macrophage infiltration, through CCL3-CCR1 and CCL4-CCR5 inhibition, in addition to Nrf2-mediated hepatoprotective effects.
Collapse
Affiliation(s)
- Kazuhiro Onuma
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
- Pharmaceutical Research Laboratory, Pharmaceutical Division, UBE Corporation, Yamaguchi, Japan
| | - Kenji Watanabe
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| | - Keishiro Isayama
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| | - Sayaka Ogi
- Pharmaceutical Research Laboratory, Pharmaceutical Division, UBE Corporation, Yamaguchi, Japan
| | - Yasunori Tokunaga
- Pharmaceutical Research Laboratory, Pharmaceutical Division, UBE Corporation, Yamaguchi, Japan
| | - Yoichi Mizukami
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| |
Collapse
|
27
|
Li F, Xie X, Xu S, Zhou F, Yu Y, Fang X, Zhou M, Zhu M, Hong D. Cerebral involvement in sitosterolemia. Lipids Health Dis 2024; 23:222. [PMID: 39039599 PMCID: PMC11265170 DOI: 10.1186/s12944-024-02216-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Sitosterolemia, an autosomal recessive condition, is characterized by impaired metabolism of plant sterols. Clinical symptoms include skin xanthoma, premature atherosclerotic disease, arthritis, and unexplained hematological abnormalities. However, there is a dearth of studies on sitosterolemia-related brain damage. METHODS This study focused on the family of two sitosterolemia patients who presented with severe hypercholesterolemia and xanthoma. Radiological examinations, biopsies, whole-exome sequencing (WES), and plant sterol tests were conducted. RESULTS The index patient, a 66-year-old female, initially exhibited weakness in both lower limbs and later developed urinary and fecal incontinence. Neuroimaging showed that the falx of the brain had irregular fusiform thickening. Significant tissue edema was observed around the lesions in the bilateral frontal-parietal lobes. Pathological analysis of the biopsied brain lesion revealed extensive cholesterol crystal deposition and lymphocyte infiltration in the matrix. The index patient who experienced cerebral impairment and her sister both carried two compound heterozygous variants in ATP binding cassette transporter G5 (ABCG5). These included the nonsense variants NM_022436: c.751 C > T (p.Q251X) in exon 6 and NM_022436: c.1336 C > T (p.R446X) in exon 10. A notable increase in plant sterol levels was observed in the younger sister of the index patient. CONCLUSION This study highlights a previously unreported neurological aspect of sitosterolemia. Imaging and pathology findings suggest that cholesterol crystals may be deposited in connective tissues such as the cerebral falx and pia mater through blood circulation.
Collapse
Affiliation(s)
- Fangjun Li
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China
- Departerment of Neurology, Gaoxin Branch of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xufang Xie
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China
| | - Shan Xu
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fuqing Zhou
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yaqing Yu
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China
| | - Xin Fang
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China
| | - Meihong Zhou
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China
| | - Min Zhu
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, Jiangxi, China
- Rare Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health Commission, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, Jiangxi, P.R. China.
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, Jiangxi, China.
- Rare Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health Commission, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
28
|
Wang J, Li H, Hu J, Shi R, Qin C, Chen X, Chen S, Zeng X, Luo H, Luo H, Zhou Y, Yang P, Wang D. Relationship of triglyceride-glucose index to gallstone prevalence and age at first gallstone surgery in American adults. Sci Rep 2024; 14:16749. [PMID: 39033195 PMCID: PMC11271289 DOI: 10.1038/s41598-024-67883-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
The triglyceride-glucose (TyG) index is a novel marker of insulin resistance that has been strongly associated with many diseases related to metabolic disorders, such as diabetes, coronary heart disease, myocardial infarction, obesity, nonalcoholic fatty liver disease, and stroke. However, whether the TyG index is associated with the prevalence of gallstones has not been determined. Therefore, the purpose of this study was to evaluate the relationship between the TyG index and the prevalence of gallstones in American adults, as well as the age at which adults in America undergo their first gallstone surgery. We selected individuals from the National Health and Nutrition Examination Survey (NHANES) database from 2017 to March 2020. Based on the goal of our study, comprehensive inclusion and exclusion criteria were created. A logistic regression analysis, dose-response curve, and subgroup analysis were computed to assess the relationship between the TyG index and gallstone prevalence and age at first surgery for gallstone. A total of 3905 participants aged > 20 years were included in our study, of whom 421 had a self-reported history of gallstones. A total of 1884 (48.2%) males and 2021 (51.8%) females were included. After confounders adjustment, it was found single-unit increases in the TyG index were linked with a 25.0% increase in gallstone prevalence (odds ratio [OR] = 1.25, 95% confidence interval [95%CI]: 1.04, 1.51). After conversion of the TyG index values from continuous to categorical variables with tertiles, a marked 48% increase in gallstone incidence was found in tertile 3 relative to tertile 1 (OR = 1.48, 95% CI: 1.09, 1.99). The dose-response curve results indicated positive associations between gallstone prevalence and the TyG index, while the latter was negatively associated with age at first gallstone surgery. Based on subgroup analysis, the positive association between TyG index and high-incidence of gallstones was more significant in females (OR = 1.39, 95% CI: 1.09, 1.77), age < 40 years (OR = 2.02, 95% CI: 1.23, 3.29), and other race (OR = 1.46, 95% CI: 1.06, 2.02). A higher TyG index is associated with a higher incidence of gallstones and may lead to an earlier age of first gallstone surgery. However, a causal relationship between TyG and gallstones cannot be established.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Han Li
- Department of Cardiology, The Fifth Hospital of Wuhan, Wuhan, 430050, China
| | - Junchao Hu
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Ruizi Shi
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Chuan Qin
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Xi Chen
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Sirui Chen
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Xintao Zeng
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Hua Luo
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Huiwen Luo
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Yulong Zhou
- Department of General Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Pei Yang
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China.
| | - Decai Wang
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China.
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China.
| |
Collapse
|
29
|
Castellanos AA, Castillo MDC, Montoya L, Ruiz ME, Zapateiro JL, Nogueira JP. Family sitosterolemia: report of two cases in Colombia. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:229-233. [PMID: 38443216 DOI: 10.1016/j.arteri.2024.02.003] [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: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
Sitosterolemia is an autosomal recessive and very rare disease. Its main characteristic is that there is a greater absorption and a decrease in the excretion of sterols, which leads to them being deposited in tissues. It is given by mutations in the ABCG5 or ABCG8 genes found on chromosome 2p21. In this clinical note, we describe the first two patients with familial sitosterolemia described in Colombia, brothers, one of them with xanthomas in extremities as the only symptom, and the other, completely asymptomatic. Genetic studies were performed as a diagnostic test in both patients, where a pathogenic homozygous variant could be identified in the ABCG8 gene in the first case (symptomatic), and a heterozygous variant in the ABCG8 gene in the second case (asymptomatic); the first patient has responded to treatment with ezetimibe. In conclusion, xanthomas should be studied in depth in pediatric age as they may be the only visible sign of such complex and hereditary diseases as familial sitosterolemia, which can be controlled and prevent cardiovascular complications of the disease.
Collapse
Affiliation(s)
| | | | - Laura Montoya
- Hospital San Jerónimo de Montería, Montería, Colombia
| | | | | | - Juan Patricio Nogueira
- Centro de Investigación en Endocrinología, Nutrición y Metabolismo (CIENM), Facultad de Ciencias de La Salud, Universidad Nacional de Formosa, Formosa, Argentina; Universidad Internacional de las Américas, San José, Costa Rica.
| |
Collapse
|
30
|
Li C, Tian J, Liu N, Song D, Steer CJ, Han Q, Song G. MicroRNA-206 as a potential cholesterol-lowering drug is superior to statins in mice. J Lipid Res 2024; 65:100576. [PMID: 38866328 PMCID: PMC11292365 DOI: 10.1016/j.jlr.2024.100576] [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: 05/16/2023] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Hypercholesterolemia is frequently intertwined with hepatosteatosis, hypertriglyceridemia, and hyperglycemia. This study is designed to assess the therapeutic efficacy of miR-206 in contrast to statins in the context of managing hypercholesterolemia in mice. We previously showed that miR-206 is a potent inhibitor of de novo lipogenesis (DNL), cholesterol synthesis, and gluconeogenesis in mice. Given that these processes occur within hepatocytes, we employed a mini-circle (MC) system to deliver miR-206 specifically to hepatocytes (designated as MC-miR-206). A single intravenous injection of MC-miR-206 maintained high levels of miR-206 in the liver for at least two weeks, thereby maintaining suppression of hepatic DNL, cholesterol synthesis, and gluconeogenesis. MC-miR-206 significantly reduced DNA damage, endoplasmic reticulum and oxidative stress, and hepatic toxicity. Therapeutically, both MC-miR-206 and statins significantly reduced total serum cholesterol and triglycerides as well as LDL cholesterol and VLDL cholesterol in mice maintained on the normal chow and high-fat high-cholesterol diet. MC-miR-206 reduced liver weight, hepatic triglycerides and cholesterol, and blood glucose, while statins slightly increased hepatic cholesterol and blood glucose and failed to affect levels of liver weight and hepatic triglycerides. Mechanistically, miR-206 alleviated hypercholesterolemia by inhibiting hepatic cholesterol synthesis, while statins increased HMGCR activity, hepatic cholesterol synthesis, and fecal-neutral steroid excretion. MiR-206 facilitates the regression of hypercholesterolemia, hypertriglyceridemia, hyperglycemia, and hepatosteatosis. MiR-206 outperforms statins by reducing hyperglycemia, hepatic cholesterol levels, and hepatic toxicity.
Collapse
Affiliation(s)
- Chao Li
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan City, China; The First College of Clinical Medicine, Shanxi Medical University, Taiyuan City, China
| | - Jing Tian
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan City, China
| | - Ningning Liu
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - David Song
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Clifford J Steer
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Qinghua Han
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan City, China.
| | - Guisheng Song
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
31
|
Le A, Peng H, Golinsky D, Di Scipio M, Lali R, Paré G. What Causes Premature Coronary Artery Disease? Curr Atheroscler Rep 2024; 26:189-203. [PMID: 38573470 DOI: 10.1007/s11883-024-01200-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW This review provides an overview of genetic and non-genetic causes of premature coronary artery disease (pCAD). RECENT FINDINGS pCAD refers to coronary artery disease (CAD) occurring before the age of 65 years in women and 55 years in men. Both genetic and non-genetic risk factors may contribute to the onset of pCAD. Recent advances in the genetic epidemiology of pCAD have revealed the importance of both monogenic and polygenic contributions to pCAD. Familial hypercholesterolemia (FH) is the most common monogenic disorder associated with atherosclerotic pCAD. However, clinical overreliance on monogenic genes can result in overlooked genetic causes of pCAD, especially polygenic contributions. Non-genetic factors, notably smoking and drug use, are also important contributors to pCAD. Cigarette smoking has been observed in 25.5% of pCAD patients relative to 12.2% of non-pCAD patients. Finally, myocardial infarction (MI) associated with spontaneous coronary artery dissection (SCAD) may result in similar clinical presentations as atherosclerotic pCAD. Recognizing the genetic and non-genetic causes underlying pCAD is important for appropriate prevention and treatment. Despite recent progress, pCAD remains incompletely understood, highlighting the need for both awareness and research.
Collapse
Affiliation(s)
- Ann Le
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Helen Peng
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Danielle Golinsky
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- School of Nursing, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada.
| |
Collapse
|
32
|
Del Castillo J, Tool ATJ, van Leeuwen K, van Alphen FPJ, Brands MM, Suijker MH, Meijer AB, Hoogendijk AJ, Kuijpers TW. Platelet proteomic profiling in sitosterolemia suggests thrombocytopenia is driven by lipid disorder and not platelet aberrations. Blood Adv 2024; 8:2466-2477. [PMID: 38513134 PMCID: PMC11112606 DOI: 10.1182/bloodadvances.2023012018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
ABSTRACT Sitosterolemia is a rare autosomal recessive genetic disorder in which patients develop hypercholesterolemia and may exhibit abnormal hematologic and/or liver test results. In this disease, dysfunction of either ABCG5 or ABCG8 results in the intestinal hyperabsorption of all sterols, including cholesterol and, more specifically, plant sterols or xenosterols, as well as in the impaired ability to excrete xenosterols into the bile. It remains unknown how and why some patients develop hematologic abnormalities. Only a few unrelated patients with hematologic abnormalities at the time of diagnosis have been reported. Here, we report on 2 unrelated pedigrees who were believed to have chronic immune thrombocytopenia as their most prominent feature. Both consanguineous families showed recessive gene variants in ABCG5, which were associated with the disease by in silico protein structure analysis and clinical segregation. Hepatosplenomegaly was absent. Thrombopoietin levels and megakaryocyte numbers in the bone marrow were normal. Metabolic analysis confirmed the presence of strongly elevated plasma levels of xenosterols. Potential platelet proteomic aberrations were longitudinally assessed following dietary restrictions combined with administration of the sterol absorption inhibitor ezetimibe. No significant effects on platelet protein content before and after the onset of treatment were demonstrated. Although we cannot exclude that lipotoxicity has a direct and platelet-specific impact in patients with sitosterolemia, our data suggest that thrombocytopenia is neither caused by a lack of megakaryocytes nor driven by proteomic aberrations in the platelets themselves.
Collapse
Affiliation(s)
- Jessica Del Castillo
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Anton T. J. Tool
- Department of Blood Cell Research, Sanquin Research, Amsterdam, The Netherlands
| | - Karin van Leeuwen
- Department of Research Facilities, Sanquin Research, Amsterdam, The Netherlands
| | | | - Marion M. Brands
- Department of Pediatric Metabolic Diseases, Emma Children’s Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Monique H. Suijker
- Department of Pediatric Hematology, Emma Children’s Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Alexander B. Meijer
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Arie J. Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Taco W. Kuijpers
- Department of Blood Cell Research, Sanquin Research, Amsterdam, The Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Ghosh S, Devereaux MW, Liu C, Sokol RJ. LRH-1 agonist DLPC through STAT6 promotes macrophage polarization and prevents parenteral nutrition-associated cholestasis in mice. Hepatology 2024; 79:986-1004. [PMID: 37976384 PMCID: PMC11023811 DOI: 10.1097/hep.0000000000000690] [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: 08/30/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND AIMS Parenteral nutrition-associated cholestasis (PNAC) is an important complication in patients with intestinal failure with reduced LRH-1 expression. Here, we hypothesized that LRH-1 activation by its agonist, dilauroylphosphatidylcholine (DLPC), would trigger signal transducer and activator of transcription 6 (STAT6) signaling and hepatic macrophage polarization that would mediate hepatic protection in PNAC. APPROACH AND RESULTS PNAC mouse model (oral DSSx4d followed by PNx14d; DSS-PN) was treated with LRH-1 agonist DLPC (30 mg/kg/day) intravenously. DLPC treatment prevented liver injury and cholestasis while inducing hepatic mRNA expression of Nr5a2 (nuclear receptor subfamily 5 group A member 2), Abcb11 (ATP binding cassette subfamily B member 11), Abcg5 (ATP-binding cassette [ABC] transporters subfamily G member 5), Abcg8 (ATP-binding cassette [ABC] transporters subfamily G member 8), nuclear receptor subfamily 0, and ATP-binding cassette subfamily C member 2 ( Abcc2) mRNA, all of which were reduced in PNAC mice. To determine the mechanism of the DLPC effect, we performed RNA-sequencing analysis of the liver from Chow, DSS-PN, and DSS-PN/DLPC mice, which revealed DLPC upregulation of the anti-inflammatory STAT6 pathway. In intrahepatic mononuclear cells or bone-marrow derived macrophages (BMDM) from PNAC mice, DLPC treatment prevented upregulation of pro-inflammatory (M1) genes, suppressed activation of NFκB and induced phosphorylation of STAT6 and its target genes, indicating M2 macrophage polarization. In vitro, incubation of DLPC with cultured macrophages showed that the increased Il-1b and Tnf induced by exposure to lipopolysaccharides or phytosterols was reduced significantly, which was associated with increased STAT6 binding to promoters of its target genes. Suppression of STAT6 expression by siRNA in THP-1 cells exposed to lipopolysaccharides, phytosterols, or both resulted in enhanced elevation of IL-1B mRNA expression. Furthermore, the protective effect of DLPC in THP-1 cells was abrogated by STAT6 siRNA. CONCLUSIONS These results indicate that activation of LRH-1 by DLPC may protect from PNAC liver injury through STAT6-mediated macrophage polarization.
Collapse
Affiliation(s)
- Swati Ghosh
- Department of Pediatrics, University of Colorado School of Medicine, Section of Gastroenterology, Hepatology and Nutrition
| | - Michael W. Devereaux
- Department of Pediatrics, University of Colorado School of Medicine, Section of Gastroenterology, Hepatology and Nutrition
| | - Cuining Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado‐Denver Anschutz Medical Campus, Aurora, 80045, Colorado, USA
| | - Ronald J. Sokol
- Department of Pediatrics, University of Colorado School of Medicine, Section of Gastroenterology, Hepatology and Nutrition
- Digestive Health Institute, Children’s Hospital Colorado
| |
Collapse
|
34
|
Miszczuk E, Bajguz A, Kiraga Ł, Crowley K, Chłopecka M. Phytosterols and the Digestive System: A Review Study from Insights into Their Potential Health Benefits and Safety. Pharmaceuticals (Basel) 2024; 17:557. [PMID: 38794127 PMCID: PMC11124171 DOI: 10.3390/ph17050557] [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: 04/04/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Phytosterols are a large group of substances belonging to sterols-compounds naturally occurring in the tissues of plants, animals, and humans. The most well-known animal sterol is cholesterol. Among phytosterols, the most significant compounds are β-sitosterol, stigmasterol, and campesterol. At present, they are mainly employed in functional food products designed to counteract cardiovascular disorders by lowering levels of 'bad' cholesterol, which stands as their most extensively studied purpose. It is currently understood that phytosterols may also alleviate conditions associated with the gastrointestinal system. Their beneficial pharmacological properties in relation to gastrointestinal tract include anti-inflammatory and hepatoprotective activity. Also, the anti-cancer properties as well as the impact on the gut microbiome could be a very interesting area of research, which might potentially lead to the discovery of their new application. This article provides consolidated knowledge on a new potential use of phytosterols, namely the treatment or prevention of gastrointestinal diseases. The cited studies indicate high therapeutic efficacy in conditions such as peptic ulcer disease, IBD or liver failure caused by hepatotoxic xenobiotics, however, these are mainly in vitro or in vivo studies. Nevertheless, studies to date indicate their therapeutic potential as adjunctive treatments to conventional therapies, which often exhibit unsatisfactory efficacy or serious side effects. Unfortunately, at this point there is a lack of significant clinical study data to use phytosterols in clinical practice in this area.
Collapse
Affiliation(s)
- Edyta Miszczuk
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (E.M.); (K.C.)
| | - Andrzej Bajguz
- Department of Biology and Plant Ecology, Faculty of Biology, University of Bialystok, Ciołkowskiego 1J, 15-245 Bialystok, Poland;
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (E.M.); (K.C.)
| | - Kijan Crowley
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (E.M.); (K.C.)
| | - Magdalena Chłopecka
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (E.M.); (K.C.)
| |
Collapse
|
35
|
Garrido-Sanchez L, Leiva-Badosa E, Llop-Talaveron J, Pintó-Sala X, Lozano-Andreu T, Corbella-Inglés E, Alia-Ramos P, Arias-Barquet L, Ramon-Torrel JM, Badía-Tahull MB. Blood Phytosterol Concentration and Genetic Variant Associations in a Sample Population. Nutrients 2024; 16:1067. [PMID: 38613098 PMCID: PMC11013666 DOI: 10.3390/nu16071067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
The main objective of this study was to determine plasma levels of PS and to study SNVs rs41360247, rs4245791, rs4148217, and rs11887534 of ABCG8 and the r657152 SNV at the ABO blood group locus in a sample of a population treated at our hospital, and to determine whether these SNVs are related to plasma PS concentrations. The secondary objective was to establish the variables associated with plasma PS concentrations in adults. Participants completed a dietary habit questionnaire and a blood sample was collected to obtain the following variables: campesterol, sitosterol, sitostanol, lanosterol, stigmasterol, biochemical parameters, and the SNVs. In addition, biometric and demographic variables were also recorded. In the generalized linear model, cholesterol and age were positively associated with total PS levels, while BMI was negatively related. For rs4245791, homozygous T allele individuals showed a significantly lower campesterol concentration compared with C homozygotes, and the GG alleles of rs657152 had the lowest levels of campesterol compared with the other alleles of the SNV. Conclusions: The screening of certain SNVs could help prevent the increase in plasma PS and maybe PNALD in some patients. However, further studies on the determinants of plasma phytosterol concentrations are needed.
Collapse
Affiliation(s)
- Leticia Garrido-Sanchez
- Pharmacy Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Elisabet Leiva-Badosa
- Pharmacy Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Josep Llop-Talaveron
- Pharmacy Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Xavier Pintó-Sala
- Cardiovascular Risk Unit, Internal Medicine, Hospital Universitari de Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain; (X.P.-S.)
| | - Toni Lozano-Andreu
- Pharmacy Department, Institut Català d’Oncologia, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Emili Corbella-Inglés
- Cardiovascular Risk Unit, Internal Medicine, Hospital Universitari de Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain; (X.P.-S.)
| | - Pedro Alia-Ramos
- Clinical Laboratory Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Lluis Arias-Barquet
- Ophthalmology Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Josep Maria Ramon-Torrel
- Preventive Medicine Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| | - Maria B. Badía-Tahull
- Pharmacy Department, Hospital Universitari Bellvitge, IDIBELL, Universitat Barcelona, 08907 L’Hospitalet de Llobregat, Spain
| |
Collapse
|
36
|
Gu R, Wang H, Wang CL, Lu M, Miao M, Huang MN, Chen Y, Dai YL, Zhu MQ, Zhou Q, Zou CC. Gene variants and clinical characteristics of children with sitosterolemia. Lipids Health Dis 2024; 23:83. [PMID: 38509578 PMCID: PMC10953262 DOI: 10.1186/s12944-024-02077-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
OBJECTIVE To enhance the detection, management and monitoring of Chinese children afflicted with sitosterolemia by examining the physical characteristics and genetic makeup of pediatric patients. METHODS In this group, 26 children were diagnosed with sitosterolemia, 24 of whom underwent genetic analysis. Patient family medical history, physical symptoms, tests for liver function, lipid levels, standard blood tests, phytosterol levels, cardiac/carotid artery ultrasounds, fundus examinations, and treatment were collected. RESULTS The majority (19, 73.1%) of the 26 patients exhibited xanthomas as the most prevalent manifestation. The second most common symptoms were joint pain (7, 26.9%) and stunted growth (4, 15.4%). Among the 24 (92.3%) patients whose genetics were analyzed, 16 (66.7%) harbored ABCG5 variants (type 2 sitosterolemia), and nearly one-third (8, 33.3%) harbored ABCG8 variants (type 1 sitosterolemia). Additionally, the most common pathogenic ABCG5 variant was c.1166G > A (p.Arg389His), which was found in 10 patients (66.7%). Further analysis did not indicate any significant differences in pathological traits among those carrying ABCG5 and ABCG8 variations (P > 0.05). Interestingly, there was a greater abundance of nonsense variations in ABCG5 than in ABCG8 (P = 0.09), and a greater frequency of splicing variations in ABCG8 than ABCG5 (P = 0.01). Following a change in diet or a combination of ezetimibe, the levels of cholesterol and low-density lipoprotein were markedly decreased compared to the levels reported before treatment. CONCLUSION Sitosterolemia should be considered for individuals presenting with xanthomas and increased cholesterol levels. Phytosterol testing and genetic analysis are important for early detection. Managing one's diet and taking ezetimibe can well control blood lipids.
Collapse
Affiliation(s)
- Rui Gu
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
- Department of NICU, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Hui Wang
- Department of Rehabilitation, Children's Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Chun-Lin Wang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Mei Lu
- Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Miao Miao
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Meng-Na Huang
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Yi Chen
- Department of Pulmonology, Children's Hospital of Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Yang-Li Dai
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Ming-Qiang Zhu
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Qiong Zhou
- Department of Pediatrics, Hangzhou Children's Hospital, Hangzhou, 310005, China
| | - Chao-Chun Zou
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China.
| |
Collapse
|
37
|
Peng J, Li H, Tong F, Hu J, Li M, Chen G, Liu D, Liu J, Wang R, Xu H, Li X, Zhong X, Yao J, Cao B. Methylation changes of liver DNA during the formation of gallstones. Epigenomics 2024; 16:529-547. [PMID: 38444389 PMCID: PMC11160444 DOI: 10.2217/epi-2023-0391] [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: 11/06/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Aim: To explore the overall methylation changes in liver tissues during the formation of gallstones, as well as the key pathways and genes involved in the process. Methods: Reduced-representation bisulfite sequencing and RNA sequencing were conducted on the liver tissues of mice with gallstones and control normal mice. Results: A total of 8705 differentially methylated regions in CpG and 1410 differentially expressed genes were identified. The joint analysis indicated that aberrant DNA methylation may be associated with dysregulated gene expression in key pathways such as cholesterol metabolism and bile secretion. Conclusion: We propose for the first time that methylation changes in some key pathway genes in liver tissue may be involved in the formation of gallstones.
Collapse
Affiliation(s)
- Junbin Peng
- Medical School of Anhui University Of Science & Technology, Huainan, 232001, Anhui, China
| | - Haojie Li
- Medical School of Anhui University Of Science & Technology, Huainan, 232001, Anhui, China
| | - Fang Tong
- Medical School of Anhui University Of Science & Technology, Huainan, 232001, Anhui, China
| | - Jinlong Hu
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, China
| | - Min Li
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
| | - Gan Chen
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
| | - Dongquan Liu
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
| | - Jinshan Liu
- Anhui Medical University, Hefei, 230032, Anhui, China
| | - Rui Wang
- Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Hongyu Xu
- Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xuanxuan Li
- Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Xinguo Zhong
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
| | - Jiaming Yao
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
| | - Baoqiang Cao
- Medical School of Anhui University Of Science & Technology, Huainan, 232001, Anhui, China
- Department of General Surgery, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, Anhui, China
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, 230041, China
| |
Collapse
|
38
|
Niknafs S, Meijer MMY, Khaskheli AA, Roura E. In ovo delivery of oregano essential oil activated xenobiotic detoxification and lipid metabolism at hatch in broiler chickens. Poult Sci 2024; 103:103321. [PMID: 38100943 PMCID: PMC10762474 DOI: 10.1016/j.psj.2023.103321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
In ovo interventions are used to improve embryonic development and robustness of chicks. The objective of this study was to identify the optimal dose for in ovo delivery of oregano essential oil (OEO), and to investigate metabolic impacts. Broiler chickens Ross 308 fertile eggs were injected with 7 levels of OEO (0, 5, 10, 20, 30, 40, and 50 µL) into the amniotic fluid at embryonic d 17.5 (E17.5) (n = 48). Chick quality was measured by navel score (P < 0.05) and/or hatchability rates (P < 0.01) were significantly decreased at doses at or above 10 or 20 µL/egg, respectively, indicating potential toxicity. However, no effects were observed at the 5 µL/egg, suggesting that compensatory mechanisms were effective to maintain homeostasis in the developing embryo. To pursue a better understanding of these mechanisms, transcriptomic analyses of the jejunum were performed comparing the control injected with saline and the group injected with 5 µL of OEO. The transcriptomic analyses identified that 167 genes were upregulated and 90 were downregulated in the 5 µL OEO compared to the control group injected with saline (P < 0.01). Functional analyses of the differentially expressed genes (DEG) showed that metabolic pathways related to the epoxygenase cytochrome P450 pathway associated with xenobiotic catabolic processes were significantly upregulated (P < 0.05). In addition, long-chain fatty acid metabolism associated with ATP binding transporters was also upregulated in the OEO treated group (P < 0.05). The results indicated that low doses of OEO in ovo have the potential to increase lipid metabolism in late stages (E17.5) of embryonic development. In conclusion, in ovo delivery of 5 µL OEO did not show any negative impact on hatchability and chick quality. OEO elevated expression of key enzymes and receptors involved in detoxification pathways and lipid metabolism in the jejunum of hatchling broiler chicks.
Collapse
Affiliation(s)
- Shahram Niknafs
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia
| | - Mila M Y Meijer
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia
| | - Asad A Khaskheli
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia
| | - Eugeni Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia.
| |
Collapse
|
39
|
Terasaki M, Izumi M, Yamagishi SI. A Clinical Case of Probable Sitosterolemia. Int J Mol Sci 2024; 25:1535. [PMID: 38338819 PMCID: PMC10855567 DOI: 10.3390/ijms25031535] [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: 12/18/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Sitosterolemia is a rare genetic lipid disorder characterized by elevated plant sterols in the serum. A 24-year-old Japanese woman was referred to our hospital due to a high serum low-density lipoprotein cholesterol (LDL-C) level of 332 mg/dL. At first, she was suspected to suffer from familial hypercholesterolemia, and thus received lipid-lowering agents. Although her LDL-C level remained high (220 mg/dL) with diet therapy plus 10 mg/day rosuvastatin, it was drastically decreased to 46 mg/dL with the addition of 10 mg/day ezetimibe. Finally, her LDL-C level was well-controlled at about 70 mg/dL with 10 mg/day ezetimibe alone. Furthermore, while her serum sitosterol level was elevated at 10.5 μg/mL during the first visit to our hospital, it decreased to 3.6 μg/mL with the 10 mg/day ezetimibe treatment alone. These observations suggest that she might probably suffer from sitosterolemia. Therefore, targeted gene sequencing analysis was performed using custom panels focusing on the exome regions of 21 lipid-associated genes, including ABCG5, ABCG8, and familial hypercholesterolemia-causing genes (LDL receptor, LDLRAP1, PCSK9, and apolipoprotein B). We finally identified a heterozygous ABCG8 variant (NM_022437.2:c.1285A>G or NP_071882.1:p.Met429Val) in our patient. The same gene mutation was detected in her mother. We report here a rare case exhibiting probable sitosterolemia caused by a heterozygous Met429Val variant in the ABCG8 gene and additional unknown variants.
Collapse
Affiliation(s)
- Michishige Terasaki
- Division of Diabetes, Metabolism and Endocrinology, Showa University Graduate School of Medicine, 1-5-8 Shinagawa, Tokyo 142-8666, Japan;
| | - Mikiko Izumi
- Center for Clinical Genetics, Showa University Hospital, 1-5-8 Shinagawa, Tokyo 142-8666, Japan;
| | - Sho-ichi Yamagishi
- Division of Diabetes, Metabolism and Endocrinology, Showa University Graduate School of Medicine, 1-5-8 Shinagawa, Tokyo 142-8666, Japan;
| |
Collapse
|
40
|
Pan E, Tao F, Smorodina E, Zhang S. Structural bioinformatics studies of six human ABC transporters and their AlphaFold2-predicted water-soluble QTY variants. QRB DISCOVERY 2024; 5:e1. [PMID: 38577032 PMCID: PMC10988169 DOI: 10.1017/qrd.2024.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 04/06/2024] Open
Abstract
Human ATP-binding cassette (ABC) transporters are one of the largest families of membrane proteins and perform diverse functions. Many of them are associated with multidrug resistance that often results in cancer treatment with poor outcomes. Here, we present the structural bioinformatics study of six human ABC membrane transporters with experimentally determined cryo-electron microscopy (CryoEM) structures including ABCB7, ABCC8, ABCD1, ABCD4, ABCG1, ABCG5, and their AlphaFold2-predicted water-soluble QTY variants. In the native structures, there are hydrophobic amino acids such as leucine (L), isoleucine (I), valine (V), and phenylalanine (F) in the transmembrane alpha helices. These hydrophobic amino acids are systematically replaced by hydrophilic amino acids glutamine (Q), threonine (T), and tyrosine (Y). Therefore, these QTY variants become water soluble. We also present the superposed structures of native ABC transporters and their water-soluble QTY variants. The superposed structures show remarkable similarity with root mean square deviations between 1.064 and 3.413 Å despite significant (41.90-54.33%) changes to the protein sequence of the transmembrane domains. We also show the differences in hydrophobicity patches between the native ABC transporters and their QTY variants. We explain the rationale behind why the QTY membrane protein variants become water soluble. Our structural bioinformatics studies provide insight into the differences between the hydrophobic helices and hydrophilic helices and will likely further stimulate designs of water-soluble multispan transmembrane proteins and other aggregated proteins. The water-soluble ABC transporters may be useful as soluble antigens to generate therapeutic monoclonal antibodies for combating multidrug resistance in clinics.
Collapse
Affiliation(s)
- Emily Pan
- The Lawrenceville School, Lawrenceville, NJ, USA
| | - Fei Tao
- Laboratory of Food Microbial Technology, State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Eva Smorodina
- Laboratory for Computational and Systems Immunology, Department of Immunology, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
41
|
Bhattacharya T, Nandi A, Das A, El-Shazly M. Role of liver in gallstone formation. GALLSTONE FORMATION, DIAGNOSIS, TREATMENT AND PREVENTION 2024:51-70. [DOI: 10.1016/b978-0-443-16098-1.00014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
42
|
Olkkonen VM, Gylling H. Oxy- and Phytosterols as Biomarkers: Current Status and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:353-375. [PMID: 38036889 DOI: 10.1007/978-3-031-43883-7_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Oxysterols and phytosterols are sterol compounds present at markedly low levels in tissues and serum of healthy individuals. A wealth of evidence suggests that they could be employed as biomarkers for human diseases or for cholesterol absorption.An increasing number of reports suggest circulating or tissue oxysterols as putative biomarkers for cardiovascular and neurodegenerative diseases or cancers. Thus far most of the studies have been carried out on small study populations. To achieve routine biomarker use, large prospective cohort studies are absolutely required. This, again, would necessitate thorough standardization of the oxysterol analytical methodology across the different laboratories, which now employ different technologies resulting in inconsistencies in the measured oxysterol levels. Routine use of oxysterol biomarkers would also necessitate the development of a new targeted analytical methodology suitable for high-throughput platforms.The most important use of phytosterols as biomarkers involves their use as markers for cholesterol absorption. For this to be achieved, (1) their quantitative analyses should be available in routine lipid laboratories, (2) it should be generally acknowledgment that the profile of cholesterol metabolism can reveal the risk of the development of atherosclerotic cardiovascular diseases (ASCVD), and (3) screening of the profile of cholesterol metabolism should be included in the ASCVD risk surveys. This should be done e.g. in families with a history of early onset or frequent ASCVD and in young adults aged 18-20 years, to exclude the presence of high cholesterol absorption. Individuals in high cholesterol absorption families need preventive measures from young adulthood to inhibit the possible development and progression of atherosclerosis.
Collapse
Affiliation(s)
- Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Helena Gylling
- Heart and Lung Center, Cardiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
43
|
Bydlowski SP, Levy D. Association of ABCG5 and ABCG8 Transporters with Sitosterolemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:31-42. [PMID: 38036873 DOI: 10.1007/978-3-031-43883-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Sitosterolemia is a rare genetic lipid disorder, mainly characterized by the accumulation of dietary xenosterols in plasma and tissues. It is caused by inactivating mutations in either ABCG5 or ABCG8 subunits, a subfamily-G ATP-binding cassette (ABCG) transporters. ABCG5/G8 encodes a pair of ABC half transporters that form a heterodimer (G5G8). This heterodimeric ATP-binding cassette (ABC) sterol transporter, ABCG5/G8, is responsible for the hepatobiliary and transintestinal secretion of cholesterol and dietary plant sterols to the surface of hepatocytes and enterocytes, promoting the secretion of cholesterol and xenosterols into the bile and the intestinal lumen. In this way, ABCG5/G8 function in the reverse cholesterol transport pathway and mediate the efflux of cholesterol and xenosterols to high-density lipoprotein and bile salt micelles, respectively. Here, we review the biological characteristics and function of ABCG5/G8, and how the mutations of ABCG5/G8 can cause sitosterolemia, a loss-of-function disorder characterized by plant sterol accumulation and premature atherosclerosis, among other features.
Collapse
Affiliation(s)
- Sergio Paulo Bydlowski
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil.
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera) CNPq, Rio de Janeiro, Brazil.
| | - Debora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
| |
Collapse
|
44
|
Al-Dejeli AAB, Al-Mudhafar MA, Al-Sabri IKA. Adenosine triphosphate binding cassette transporters G5 and G8 early diagnostic tools for cardiovascular disease in human. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:262-267. [PMID: 38592987 DOI: 10.36740/wlek202402111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
OBJECTIVE Aim: The current study was designed to investigate the role of ABCG5 and ABCG5 in serum with normal and expected cardiac complaints with CVDs as individual early diagnostic tools. PATIENTS AND METHODS Materials and Methods: Data was collected in paper form and recorded from 100 healthy personals and 100 personals suspected with CVS after take the case history and clinical signs in private clinical hospital and the serum was collected for measurements the activity of ABCG5 and ABCG5 by used ELISA reader and the results illustrated that activity of ABCG5 and ABCG5 in all aged groups. RESULTS Results: Activity of ABCG5 and ABCG5 in all aged groups periods in patient person male and female significant decrease as compared with same age in same period of live, so that the researched depicted that can used the serum activity of ABCG5 and ABCG5 as a diagnostics tools for atherosclerotic cardiovascular disease. CONCLUSION Conclusions: We identified areas of further exploration on cholesterol transport related with CVD risk and concluded that changes in the Adenosine Triphosphate Binding Cassette transporters mainly G5 and G8 early diagnostic tools for cardiovascular disease in Human. We correlated areas of farther disquisition on nutrient cholesterol and CVD threat, in the included trials, healthy grown-ups consumed high doses of dietary cholesterol.
Collapse
|
45
|
Wu L, Wang X, Jiang J, Chen Y, Peng B, Jin W. Mechanism of rhubarb in the treatment of hyperlipidemia: A recent review. Open Med (Wars) 2023; 18:20230812. [PMID: 37808167 PMCID: PMC10552914 DOI: 10.1515/med-2023-0812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Hyperlipidemia is a metabolic disorder, which is a major risk factor for atherosclerosis, stroke, and coronary heart disease. Although lipid-lowering treatments have been extensively studied, safer treatments with fewer adverse effects are needed. Rhubarb is a traditional Chinese medicine that has lipid-lowering, anti-inflammatory, and antioxidant properties. Disturbance in lipid metabolism is the basis of tissue damage caused by hyperlipidemia and plays a key role in the development of hyperlipidemia; however, the molecular mechanisms by which rhubarb regulates lipid metabolism to lower lipid levels are yet to be elucidated. We conducted this study to summarize the phytochemical constituents of Rheum officinale and provide a comprehensive review of the molecular mechanisms underlying the regulation of lipid metabolism during hyperlipidemia treatment. It was found that rhubarb extracts, including emodin, rhubarb acid, and rhubarb phenol, regulate total cholesterol, triglyceride, TNF-α, and IL-1β levels through signaling pathways such as C/EBP α, 3T3-L1, PPAR α, and AMPK, thereby improving the hyperlipidemic state. This suggests that rhubarb is a natural drug with lipid-lowering potential, and an in-depth exploration of its lipid-lowering mechanism can provide new ideas for the prevention and treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Lijiao Wu
- Chengdu University of Traditional Chinese Medicine School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangjin Wang
- College of Sports Medicine and Health, Chengdu Sports University, Chengdu, China
| | - Jihang Jiang
- Chengdu University of Traditional Chinese Medicine School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Chen
- Emergency Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Peng
- Respiratory Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Jin
- Emergency Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
46
|
Xu H, Xin Y, Wang J, Liu Z, Cao Y, Li W, Zhou Y, Wang Y, Liu P. The TICE Pathway: Mechanisms and Potential Clinical Applications. Curr Atheroscler Rep 2023; 25:653-662. [PMID: 37736845 DOI: 10.1007/s11883-023-01147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE OF REVIEW Transintestinal cholesterol excretion (TICE) is a non-biliary pathway that excretes excess cholesterol from the body through feces. This article focuses on the research progress of the TICE pathway in the last few years, including the discovery process of the TICE pathway, its molecular mechanism, and potential clinical applications. RECENT FINDINGS Cholesterol homeostasis is vital for cardiovascular diseases, stroke, and neurodegenerative diseases. Beyond the cholesterol excretion via hepatobiliary pathway, TICE contributes significantly to reverse cholesterol transport ex vivo and in vivo. Nuclear receptors are ligand-activated transcription factors that regulate cholesterol metabolism. The farnesoid X receptor (FXR) and liver X receptor (LXR) activated, respectively, by oxysterols and bile acids promote intestinal cholesterol secretion through ABCG5/G8. Nutrient regulators and intestinal flora also modulate cholesterol secretion through the TICE pathway. TICE allows direct elimination of plasma cholesterol, which may provide an attractive therapeutic targets. TICE pathway may provide a potential target to stimulate cholesterol elimination and reduce the risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Huimin Xu
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Henan, China
| | - Yiyang Xin
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Henan, China
| | - Jiaxin Wang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Henan, China
| | - Zixin Liu
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Henan, China
| | - Yutong Cao
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Henan, China
| | - Weiguo Li
- People's Hospital of Hebi, Henan University, Henan, China
| | - Yun Zhou
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Henan, China.
| | - Yandong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Peng Liu
- People's Hospital of Hebi, Henan University, Henan, China.
| |
Collapse
|
47
|
Jiang W, Xu Y, Fu Z, Hu M, Wu Q, Ji Y, Li JZ, Gong Y, Zhou H. Genetic analysis and functional study of a novel ABCG5 mutation in sitosterolemia with hematologic disease. Gene 2023; 879:147596. [PMID: 37390873 DOI: 10.1016/j.gene.2023.147596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/24/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Sitosterolemia is a rare autosomal recessive hereditary disease caused by loss-of-function genetic mutations in either ATP-binding cassette subfamily G member 5 or member 8 (ABCG5 or ABCG8). Here, we investigate novel variants in ABCG5 and ABCG8 that are associated with the sitosterolemia phenotype. We describe a 32-year-old woman with hypercholesterolemia, tendon and hip xanthomas, autoimmune hemolytic anemia and macrothrombocytopenia from early life, which make us highly suspicious of the possibility of sitosterolemia. A novel homozygous variant in ABCG5 (c.1769C>A, p.S590X) was identified by genomic sequencing. We also examined the lipid profile, especially plant sterols levels, using gas chromatography-mass spectrometry. Functional studies, including western blotting and immunofluorescence staining, showed that the nonsense mutation ABCG5 1769C>A hinders the formation of ABCG5 and ABCG8 heterodimers and the function of transporting sterols. Our study expands the knowledge of variants in sitosterolemia and provides diagnosis and treatment recommendations.
Collapse
Affiliation(s)
- Wanzi Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China
| | - Yiwen Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China
| | - Zhenzhen Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China
| | - Moran Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China
| | - Qinyi Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Suzhou, China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yingyun Gong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China.
| | - Hongwen Zhou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China.
| |
Collapse
|
48
|
Risinger M, Kim PS, Rodriguez RX, Rivas MN, Setchell KDR, Zhang W, Kalfa TA. Hemolytic anemia and macrothrombocytopenia: A lipid problem? Am J Hematol 2023; 98:1335-1340. [PMID: 36974979 PMCID: PMC10523966 DOI: 10.1002/ajh.26916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/09/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Affiliation(s)
- Mary Risinger
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Phyllis S. Kim
- Hematology and Medical Oncology Department, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA
| | - Roberto X. Rodriguez
- Hematology and Medical Oncology Department, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA
| | - Monica Narvaez Rivas
- Division of Pathology & Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kenneth D. R. Setchell
- Division of Pathology & Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Wenying Zhang
- Genetics and Genomics Diagnostic Laboratory, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Theodosia A. Kalfa
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
49
|
Ontsouka E, Schroeder M, Albrecht C. Revisited role of the placenta in bile acid homeostasis. Front Physiol 2023; 14:1213757. [PMID: 37546542 PMCID: PMC10402276 DOI: 10.3389/fphys.2023.1213757] [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: 04/28/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
To date, the discussion concerning bile acids (BAs) during gestation is almost exclusively linked to pregnancy complications such as intrahepatic cholestasis of pregnancy (ICP) when maternal serum BA levels reach very high concentrations (>100 μM). Generally, the placenta is believed to serve as a protective barrier avoiding exposure of the growing fetus to excessive amounts of maternal BAs that might cause detrimental effects (e.g., intrauterine growth restriction and/or increased vulnerability to metabolic diseases). However, little is known about the precise role of the placenta in BA biosynthesis, transport, and metabolism in healthy pregnancies when serum BAs are at physiological levels (i.e., low maternal and high fetal BA concentrations). It is well known that primary BAs are synthesized from cholesterol in the liver and are later modified to secondary BA species by colonic bacteria. Besides the liver, BA synthesis in extrahepatic sites such as the brain elicits neuroprotective actions through inhibition of apoptosis as well as oxidative and endoplasmic reticulum stress. Even though historically BAs were thought to be only "detergent molecules" required for intestinal absorption of dietary fats, they are nowadays acknowledged as full signaling molecules. They modulate a myriad of signaling pathways with functional consequences on essential processes such as gluconeogenesis -one of the principal energy sources of the fetus- and cellular proliferation. The current manuscript discusses the potential multipotent roles of physiologically circulating BAs on developmental processes during gestation and provides a novel perspective in terms of the importance of the placenta as a previously unknown source of BAs. Since the principle "not too much, not too little" applicable to other signaling molecules may be also true for BAs, the risks associated with fetal exposure to excessive levels of BAs are discussed.
Collapse
|
50
|
Simonen P, Öörni K, Sinisalo J, Strandberg TE, Wester I, Gylling H. High cholesterol absorption: A risk factor of atherosclerotic cardiovascular diseases? Atherosclerosis 2023; 376:53-62. [PMID: 37290267 DOI: 10.1016/j.atherosclerosis.2023.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
Lowering elevated low-density lipoprotein cholesterol (LDL-C) concentrations reduces the risk of atherosclerotic cardiovascular diseases (ASCVDs). However, increasing evidence suggests that cholesterol metabolism may also be involved in the risk reduction of ASCVD events. In this review, we discuss if the different profiles of cholesterol metabolism, with a focus on high cholesterol absorption, are atherogenic, and what could be the possible mechanisms. The potential associations of cholesterol metabolism and the risk of ASCVDs are evaluated from genetic, metabolic, and population-based studies and lipid-lowering interventions. According to these studies, loss-of-function genetic variations in the small intestinal sterol transporters ABCG5 and ABCG8 result in high cholesterol absorption associated with low cholesterol synthesis, low cholesterol elimination from the body, and a high risk of ASCVDs. In contrast, loss-of-function genetic variations in another intestinal sterol transporter, NPC1L1 result in low cholesterol absorption associated with high cholesterol synthesis, elevated cholesterol elimination from the body, and low risk of ASCVDs. Statin monotherapy is not sufficient to reduce the ASCVD risk in cases of high cholesterol absorption, and these individuals need combination therapy of statin with cholesterol absorption inhibition. High cholesterol absorption, i.e., >60%, is estimated to occur in approximately one third of a population, so taking it into consideration is important to optimise lipid-lowering therapy to prevent atherosclerosis and reduce the risk of ASCVD events.
Collapse
Affiliation(s)
- Piia Simonen
- Heart and Lung Center, Cardiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Katariina Öörni
- Wihuri Research Institute, Helsinki, Finland; Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Cardiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Timo E Strandberg
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Center for Life-Course Health Research, University of Oulu, Oulu, Finland
| | | | - Helena Gylling
- Heart and Lung Center, Cardiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| |
Collapse
|