1
|
Lu F, Li E, Yang X. Proprotein convertase subtilisin/kexin type 9 deficiency in extrahepatic tissues: emerging considerations. Front Pharmacol 2024; 15:1413123. [PMID: 39139638 PMCID: PMC11319175 DOI: 10.3389/fphar.2024.1413123] [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: 04/06/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily secreted by hepatocytes. PCSK9 is critical in liver low-density lipoprotein receptors (LDLRs) metabolism. In addition to its hepatocellular presence, PCSK9 has also been detected in cardiac, cerebral, islet, renal, adipose, and other tissues. Once perceived primarily as a "harmful factor," PCSK9 has been a focal point for the targeted inhibition of both systemic circulation and localized tissues to treat diseases. However, PCSK9 also contributes to the maintenance of normal physiological functions in numerous extrahepatic tissues, encompassing both LDLR-dependent and -independent pathways. Consequently, PCSK9 deficiency may harm extrahepatic tissues in close association with several pathophysiological processes, such as lipid accumulation, mitochondrial impairment, insulin resistance, and abnormal neural differentiation. This review encapsulates the beneficial effects of PCSK9 on the physiological processes and potential disorders arising from PCSK9 deficiency in extrahepatic tissues. This review also provides a comprehensive analysis of the disparities between experimental and clinical research findings regarding the potential harm associated with PCSK9 deficiency. The aim is to improve the current understanding of the diverse effects of PCSK9 inhibition.
Collapse
Affiliation(s)
- Fengyuan Lu
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - En Li
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Yang
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Zimodro JM, Mucha M, Berthold HK, Gouni-Berthold I. Lipoprotein Metabolism, Dyslipidemia, and Lipid-Lowering Therapy in Women: A Comprehensive Review. Pharmaceuticals (Basel) 2024; 17:913. [PMID: 39065763 PMCID: PMC11279947 DOI: 10.3390/ph17070913] [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: 06/20/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Lipid-lowering therapy (LLT) is a cornerstone of atherosclerotic cardiovascular disease prevention. Although LLT might lead to different reductions in low-density lipoprotein cholesterol (LDL-C) levels in women and men, LLT diminishes cardiovascular risk equally effectively in both sexes. Despite similar LLT efficacy, the use of high-intensity statins, ezetimibe, and proprotein convertase subtilisin/kexin type 9 inhibitors is lower in women compared to men. Women achieve the guideline-recommended LDL-C levels less often than men. Greater cholesterol burden is particularly prominent in women with familial hypercholesterolemia. In clinical practice, women and men with dyslipidemia present with different cardiovascular risk profiles and disease manifestations. The concentrations of LDL-C, lipoprotein(a), and other blood lipids differ between women and men over a lifetime. Dissimilar levels of LLT target molecules partially result from sex-specific hormonal and genetic determinants of lipoprotein metabolism. Hence, to evaluate a potential need for sex-specific LLT, this comprehensive review (i) describes the impact of sex on lipoprotein metabolism and lipid profile, (ii) highlights sex differences in cardiovascular risk among patients with dyslipidemia, (iii) presents recent, up-to-date clinical trial and real-world data on LLT efficacy and safety in women, and (iv) discusses the diverse medical needs of women and men with dyslipidemia and increased cardiovascular risk.
Collapse
Affiliation(s)
- Jakub Michal Zimodro
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Magda Mucha
- Faculty of Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Heiner K. Berthold
- Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), 33611 Bielefeld, Germany
| | - Ioanna Gouni-Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
3
|
Sachan V, Le Dévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Resiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQH, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, Seidah NG. PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease. Metabolism 2024; 150:155736. [PMID: 37967646 DOI: 10.1016/j.metabol.2023.155736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Epidemiological evidence links the proprotein convertase subtilisin/kexin 7 (PCSK7) to triglyceride (TG) metabolism. We associated the known PCSK7 gain-of-function non-coding SNP rs236918 with higher levels of plasma apolipoprotein B (apoB) and the loss-of-function coding variant p.Pro777Leu (SNP rs201598301) with lower apoB and TG. Herein, we aimed to unravel the in vivo role of liver PCSK7. METHODS We biochemically defined the functional role of PCSK7 in lipid metabolism using hepatic cell lines and Pcsk7-/- mice. Our findings were validated following subcutaneous administration of hepatocyte-targeted N-acetylgalactosamine (GalNAc)-antisense oligonucleotides (ASOs) against Pcsk7. RESULTS Independent of its proteolytic activity, membrane-bound PCSK7 binds apoB100 in the endoplasmic reticulum and enhances its secretion. Mechanistically, the loss of PCSK7/Pcsk7 leads to apoB100 degradation, triggering an unfolded protein response, autophagy, and β-oxidation, eventually reducing lipid accumulation in hepatocytes. Non-alcoholic fatty liver disease (NAFLD) was induced by a 12-week high fat/fructose/cholesterol diet in wild type (WT) and Pcsk7-/- mice that were then allowed to recover on a 4-week control diet. Pcsk7-/- mice recovered more effectively than WT mice from all NAFLD-related liver phenotypes. Finally, subcutaneous administration of GalNAc-ASOs targeting hepatic Pcsk7 to WT mice validated the above results. CONCLUSIONS Our data reveal hepatic PCSK7 as one of the major regulators of apoB, and its absence reduces apoB secretion from hepatocytes favoring its ubiquitination and degradation by the proteasome. This results in a cascade of events, eventually reducing hepatic lipid accumulation, thus supporting the notion of silencing PCSK7 mRNA in hepatocytes for targeting NAFLD.
Collapse
Affiliation(s)
- Vatsal Sachan
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Maïlys Le Dévéhat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Anna Roubtsova
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Rachid Essalmani
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Jean-Francois Laurendeau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Damien Garçon
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Delia Susan-Resiga
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Stéphanie Duval
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Sahar Mikaeeli
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Josée Hamelin
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Alexandra Evagelidis
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Michael Chong
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | | | - Zu-Hua Gao
- Department of Pathology, McGill University Health Centre, Montréal, QC, Canada
| | - Isabelle Robillard
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Matthieu Ruiz
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Vincent Quoc-Huy Trinh
- Departement of Pathology and Cellular Biology, Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, QC, Canada
| | - Jennifer L Estall
- Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - May Faraj
- Nutrition Department, Université de Montréal, Research Unit on Nutrition, Lipoproteins and Cardiometabolic Diseases, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, Canada
| | - Martin Sauvageau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Annik Prat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Robert S Kiss
- McGill University Health Centre Research Institute, Montréal, QC, Canada
| | - Nabil G Seidah
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
4
|
Luo Q, Tang Z, Wu P, Chen Z, Fang Z, Luo F. A bibliometric analysis of PCSK9 inhibitors from 2007 to 2022. Front Endocrinol (Lausanne) 2023; 14:1218968. [PMID: 38093957 PMCID: PMC10716461 DOI: 10.3389/fendo.2023.1218968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Background Since the approval of the proprotein convertase subtilisin/kexin type 9 (PCSK9) monoclonal antibodies for marketing in 2015, PCSK9 inhibitors have attracted significant interest in the field of cardiovascular endocrinology. A large number of clinical trials have confirmed the efficacy and safety of PCSK9 inhibitors in reducing cholesterol and the risk of cardiovascular events. No bibliometric analysis of PCSK9 inhibitors has been performed as of yet. This study aims to analyze the research trends and hotspots of PCSK9 inhibitors through bibliometric analysis. Methods We searched the Web of Science Core Collection (WoSCC) database for PCSK9 inhibitor-related publications from 2007 to 2022. Data visualization analysis was performed using CiteSpace software. Microsoft Excel and Graphpad software were used for the drawing of some tables and figures. Results A total of 1072 pieces of literature were retrieved between 2007 and 2022. The number of publications concerning PCSK9 inhibitors is growing annually. The top five countries with the most articles published were the United States, England, Canada, Italy, and France. Harvard University, Amgen, Brigham & Women's Hospital, Harvard Medical School, and Imperial College London are the five institutions with the highest output. The Journal of Clinical Lipidology is the most popular journal in this field. The most frequently cited journal is the New England Journal of Medicine. As for authors, Sabatine MS and Giugliano RP from Brigham & Women's Hospital have the highest number of published articles. Amgen is the funding agency for most of the research. According to keyword analysis, "low density lipoprotein", "familial hypercholesterolemia", "PCSK9 inhibitor", "PCSK9", and "efficacy" are the five keywords with the highest frequency of co-occurrence. Conclusion The past 15 years have witnessed a rapid and fruitful development of PCSK9 inhibitors. The research trend and focus for PCSK9 inhibitors are from the mechanism of reducing low-density lipoprotein cholesterol to related clinical trials. Developed countries such as the United States have contributed prominently in this area. Coronary artery and inflammation are currently at the forefront of research in the field and are in an explosion period.
Collapse
Affiliation(s)
- Qin Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenchu Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Panyun Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhangling Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fei Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Parente M, Tonini C, Segatto M, Pallottini V. Regulation of cholesterol metabolism: New players for an old physiological process. J Cell Biochem 2023; 124:1449-1465. [PMID: 37796135 DOI: 10.1002/jcb.30477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Identified more than two centuries ago, cholesterol plays a pivotal role in human physiology. Since cholesterol metabolism is a physiologically significant process, it is not surprising that its alterations are associated with several pathologies. The discovery of new molecular targets or compounds able to modulate this sophisticated metabolism has been capturing the attention of research groups worldwide since many years. Endogenous and exogenous compounds are known to regulate cellular cholesterol synthesis and uptake, or reduce cholesterol absorption at the intestinal level, thereby regulating cholesterol homeostasis. However, there is a great need of new modulators and diverse new pathways have been uncovered. Here, after illustrating cholesterol metabolism and its well-known regulators, some new players of this important physiological process are also described.
Collapse
Affiliation(s)
| | | | - Marco Segatto
- Department of Bioscience and Territory, University of Molise, Pesche, Italy
| | - Valentina Pallottini
- Department of Science, University Roma Tre, Rome, Italy
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Via del Fosso Fiorano, Rome, Italy
| |
Collapse
|
6
|
Roubtsova A, Scipione CA, Garçon D, Boffa MB, Seidah NG, Koschinsky ML, Prat A. Surface LDLR is a major receptor for lipoprotein(a) clearance in male mice lacking PCSK9. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159288. [PMID: 36708961 DOI: 10.1016/j.bbalip.2023.159288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Affiliation(s)
- Anna Roubtsova
- Institut de Recherches Cliniques de Montréal (IRCM), Université de Montréal, Montreal, QC, Canada
| | - Corey A Scipione
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Damien Garçon
- Institut de Recherches Cliniques de Montréal (IRCM), Université de Montréal, Montreal, QC, Canada
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Nabil G Seidah
- Institut de Recherches Cliniques de Montréal (IRCM), Université de Montréal, Montreal, QC, Canada
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Annik Prat
- Institut de Recherches Cliniques de Montréal (IRCM), Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
7
|
Sex differences in LDL-C response to PCSK9 inhibitors: A real world experience. J Clin Lipidol 2023; 17:142-149. [PMID: 36641299 DOI: 10.1016/j.jacl.2022.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies have shown the efficacy of PCSK9 inhibitors (PCSK9i) in lowering LDL-C. One clinical trial with alirocumab suggested that the LDL-C reduction effect is larger in men than women. In contrast, none of the studies with evolocumab have observed a difference in the treatment effect between men and women. However, sex differences data from real life experience is lacking. In addition, the difference in LDL-C response to PCSK9i between pre- and post-menopausal women has not been investigated so far. OBJECTIVES To compare the relative change in LDL-C following the introduction of a PCSK9i in a real-life clinical setting according to sex and menopausal status. METHODS All patients were recruited at the IRCM lipid clinic. Lipid profiles before and after the introduction of PCSK9i were available in the medical file for 259 patients (160 men and 99 women (72 post-menopausal, 20 pre-menopausal and 7 unknown menopausal status). RESULTS We observed a significant difference in relative LDL-C change between men (-70%) and women (-59%), p<0.0001. However, no difference was observed between pre-menopausal (-58%) and post-menopausal (-58%) women. In a linear regression model, sex remains a significant predictor of the response to PCSK9i after correction for confounding factors such as statin intensity (beta coefficient=-0.245, p<0.0001). CONCLUSION We observed a greater relative LDL-C response to PCSK9i in men than in women in a real-life clinical context. However, it is still unknown whether this difference in LDL-C change between men and women translates into a meaningful difference on long-term cardiovascular risk.
Collapse
|
8
|
Pakalniškytė D, Schönberger T, Strobel B, Stierstorfer B, Lamla T, Schuler M, Lenter M. Rosa26-LSL-dCas9-VPR: a versatile mouse model for tissue specific and simultaneous activation of multiple genes for drug discovery. Sci Rep 2022; 12:19268. [PMID: 36357523 PMCID: PMC9649745 DOI: 10.1038/s41598-022-23127-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
Transgenic animals with increased or abrogated target gene expression are powerful tools for drug discovery research. Here, we developed a CRISPR-based Rosa26-LSL-dCas9-VPR mouse model for targeted induction of endogenous gene expression using different Adeno-associated virus (AAV) capsid variants for tissue-specific gRNAs delivery. To show applicability of the model, we targeted low-density lipoprotein receptor (LDLR) and proprotein convertase subtilisin/kexin type 9 (PCSK9), either individually or together. We induced up to ninefold higher expression of hepatocellular proteins. In consequence of LDLR upregulation, plasma LDL levels almost abolished, whereas upregulation of PCSK9 led to increased plasma LDL and cholesterol levels. Strikingly, simultaneous upregulation of both LDLR and PCSK9 resulted in almost unaltered LDL levels. Additionally, we used our model to achieve expression of all α1-Antitrypsin (AAT) gene paralogues simultaneously. These results show the potential of our model as a versatile tool for optimized targeted gene expression, alone or in combination.
Collapse
Affiliation(s)
- Dalia Pakalniškytė
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88400 Biberach an der Riß, Germany
| | - Tanja Schönberger
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88400 Biberach an der Riß, Germany
| | - Benjamin Strobel
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88400 Biberach an der Riß, Germany
| | - Birgit Stierstorfer
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Nonclinical Drug Safety Germany, 88400 Biberach an der Riß, Germany
| | - Thorsten Lamla
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Discovery Research Coordination, 88400 Biberach an der Riß, Germany
| | - Michael Schuler
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88400 Biberach an der Riß, Germany
| | - Martin Lenter
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88400 Biberach an der Riß, Germany
| |
Collapse
|
9
|
Jia F, Fei SF, Tong DB, Xue C, Li JJ. Sex difference in circulating PCSK9 and its clinical implications. Front Pharmacol 2022; 13:953845. [PMID: 36160427 PMCID: PMC9490038 DOI: 10.3389/fphar.2022.953845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Proprotein convertase subtilisin kexin type 9 (PCSK9) is a proprotein convertase that increases plasma low-density lipoprotein cholesterol (LDL-C) levels by triggering the degradation of LDL receptors (LDLRs). Beyond the regulation of circulating LDL-C, PCSK9 also has direct atherosclerotic effects on the vascular wall and is associated with coronary plaque inflammation. Interestingly, emerging data show that women have higher circulating PCSK9 concentrations than men, suggesting that the potential roles of PCSK9 may have different impacts according to sex. In this review, we summarize the studies concerning sex difference in circulating levels of PCSK9. In addition, we report on the sex differences in the relations of elevated circulating PCSK9 levels to the severity and prognosis of coronary artery disease, the incidence of type 2 diabetes mellitus, and neurological damage after cardiac arrest and liver injury, as well as inflammatory biomarkers and high-density lipoprotein cholesterol (HDL-C). Moreover, sex difference in the clinical efficacy of PCSK9 inhibitors application are reviewed. Finally, the underlying mechanisms of sex difference in circulating PCSK9 concentrations and the clinical implications are also discussed.
Collapse
Affiliation(s)
- Fang Jia
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Si-Fan Fei
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - De-Bing Tong
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Cong Xue
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- *Correspondence: Cong Xue, ; Jian-Jun Li,
| | - Jian-Jun Li
- Cardio-Metabolic Center, Fu Wai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- *Correspondence: Cong Xue, ; Jian-Jun Li,
| |
Collapse
|
10
|
PCSK9 deficiency results in a specific shedding of excess LDLR in female mice only: Role of hepatic cholesterol. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159217. [PMID: 35985474 DOI: 10.1016/j.bbalip.2022.159217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022]
Abstract
PCSK9 promotes the lysosomal degradation of cell surface LDL receptor (LDLR). We analyzed how excess LDLR generated by PCSK9 deficiency is differently handled in male and female mice to possibly unveil the mechanism leading to the lower efficacy of PCSK9 mAb on LDL-cholesterol levels in women. Analysis of intact or ovariectomized PCSK9 knockout (KO) mice supplemented with placebo or 17β-estradiol (E2) demonstrated that female, but not male mice massively shed the soluble ectodomain of the LDLR in the plasma. Liver-specific PCSK9 KO or alirocumab-treated WT mice exhibit the same pattern. This shedding is distinct from the basal one and is inhibited by ZLDI-8, a metalloprotease inhibitor pointing at ADAM10/ADAM17. In PCSK9 KO female mice, ZLDI-8 raises by 80 % the LDLR liver content in a few hours. This specific shedding is likely cholesterol-dependent: it is prevented in PCSK9 KO male mice that exhibit low intra-hepatic cholesterol levels without activating SREBP-2, and enhanced by mevalonate or high cholesterol feeding, or by E2 known to stimulate cholesterol synthesis via the estrogen receptor-α. Liver transcriptomics demonstrates that critically low liver cholesterol in ovariectomized female or knockout male mice also hampers the cholesterol-dependent G2/M transition of the cell cycle. Finally, higher levels of shed LDLR were measured in the plasma of women treated with PCSK9 mAb. PCSK9 knockout female mice hormonally sustain cholesterol synthesis and shed excess LDLR, seemingly like women. In contrast, male mice rely on high surface LDLR to replenish their stocks, despite 80 % lower circulating LDL.
Collapse
|
11
|
Benito-Vicente A, Uribe KB, Larrea-Sebal A, Palacios L, Cenarro A, Calle X, Galicia-Garcia U, Jebari-Benslaiman S, Sánchez-Hernández RM, Stef M, Lambert G, Civeira F, Martín C. Leu22_Leu23 Duplication at the Signal Peptide of PCSK9 Promotes Intracellular Degradation of LDLr and Autosomal Dominant Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2022; 42:e203-e216. [PMID: 35510551 DOI: 10.1161/atvbaha.122.315499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND PCSK9 (Proprotein convertase subtilisin/kexin type 9) regulates LDL-C (low-density lipoprotein cholesterol) metabolism by targeting LDLr (LDL receptor) for lysosomal degradation. PCSK9 gain-of-function variants cause autosomal dominant hypercholesterolemia by reducing LDLr levels, the D374Y variant being the most severe, while loss-of-function variants are associated with low LDL-C levels. Gain-of-function and loss-of-function activities have also been attributed to variants occurring in the PCSK9 signal peptide. Among them, L11 is a very rare PCSK9 variant that seems to increase LDL-C values in a moderate way causing mild hypercholesterolemia. METHODS Using molecular biology and biophysics methodologies, activities of L8 and L11 variants, both located in the leucine repetition stretch of the signal peptide, have been extensively characterized in vitro. RESULTS L8 variant is not associated with increased LDLr activity, whereas L11 activity is increased by ≈20% compared with wt PCSK9. The results suggest that the L11 variant reduces LDLr levels intracellularly by a process resulting from impaired cleavage of the signal peptide. This would lead to less efficient LDLr transport to the cell membrane and promote LDLr intracellular degradation. CONCLUSIONS Deletion of a leucine in the signal peptide in L8 variant does not affect PCSK9 activity, whereas the leucine duplication in the L11 variant enhances LDLr intracellular degradation. These findings highlight the importance of deep in vitro characterization of PCSK9 genetic variants to determine pathogenicity and improve clinical diagnosis and therapy of inherited familial hypercholesterolemia disease.
Collapse
Affiliation(s)
- Asier Benito-Vicente
- Biofisika Institute (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain (A.B.-V., A.L.-S., U.G.-G., S.J.-B., C.M.).,Department of Biochemistry and Molecular Biology, UPV/EHU, University of the Basque Country, Bilbao, Spain (A.B.-V., S.J.-B., C.M.)
| | - Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain (K.B.U.)
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain (A.B.-V., A.L.-S., U.G.-G., S.J.-B., C.M.).,Fundación Biofísica Bizkaia, Leioa, Spain (A.L.-S., U.G.-G.)
| | - Lourdes Palacios
- Progenika Biopharma, a Grifols Company, Derio, Spain (L.P., M.S.)
| | - Ana Cenarro
- Lipid Unit, Hospital Universitario Miguel Servet, IIS Aragon, CIBERCV, Universidad de Zaragoza, Spain (A.C., F.C.)
| | - Xabier Calle
- Institute of Biological Phychiatry, Mental Health Services, University Hospital, Copenhagen, Denmark (X.C.)
| | - Unai Galicia-Garcia
- Biofisika Institute (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain (A.B.-V., A.L.-S., U.G.-G., S.J.-B., C.M.).,Fundación Biofísica Bizkaia, Leioa, Spain (A.L.-S., U.G.-G.)
| | - Shifa Jebari-Benslaiman
- Biofisika Institute (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain (A.B.-V., A.L.-S., U.G.-G., S.J.-B., C.M.).,Department of Biochemistry and Molecular Biology, UPV/EHU, University of the Basque Country, Bilbao, Spain (A.B.-V., S.J.-B., C.M.)
| | - Rosa M Sánchez-Hernández
- Endocrinology Department, Complejo Hospitalario Universitario Insular Materno Infantil de Gran Canaria and Instituto Universitario de Investigación Biomédica y Sanitaria (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Spain (R.M.S.-H.)
| | - Marianne Stef
- Progenika Biopharma, a Grifols Company, Derio, Spain (L.P., M.S.)
| | - Gilles Lambert
- Inserm, Laboratoire UMR1188 DéTROI, Sainte Clotilde, France (G.L.).,Université de La Réunion, Faculté de Médecine, Saint Denis de La Réunion, France (G.L.)
| | - Fernando Civeira
- Lipid Unit, Hospital Universitario Miguel Servet, IIS Aragon, CIBERCV, Universidad de Zaragoza, Spain (A.C., F.C.)
| | - Cesar Martín
- Biofisika Institute (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain (A.B.-V., A.L.-S., U.G.-G., S.J.-B., C.M.).,Department of Biochemistry and Molecular Biology, UPV/EHU, University of the Basque Country, Bilbao, Spain (A.B.-V., S.J.-B., C.M.)
| |
Collapse
|
12
|
Schreckenberg R, Wolf A, Szabados T, Gömöri K, Szabó IA, Ágoston G, Brenner G, Bencsik P, Ferdinandy P, Schulz R, Schlüter KD. Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) Deletion but Not Inhibition of Extracellular PCSK9 Reduces Infarct Sizes Ex Vivo but Not In Vivo. Int J Mol Sci 2022; 23:ijms23126512. [PMID: 35742954 PMCID: PMC9223354 DOI: 10.3390/ijms23126512] [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: 05/02/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxia upregulates PCSK9 expression in the heart, and PCSK9 affects the function of myocytes. This study aimed to investigate the impact of PCSK9 on reperfusion injury in rats and mice fed normal or high-fat diets. Either the genetic knockout of PCSK9 (mice) or the antagonism of circulating PCSK9 via Pep2-8 (mice and rats) was used. Isolated perfused hearts were exposed to 45 min of ischemia followed by 120 min of reperfusion. In vivo, mice were fed normal or high-fat diets (2% cholesterol) for eight weeks prior to coronary artery occlusion (45 min of ischemia) and reperfusion (120 min). Ischemia/reperfusion upregulates PCSK9 expression (rats and mice) and releases it into the perfusate. The inhibition of extracellular PCSK9 does not affect infarct sizes or functional recovery. However, genetic deletion largely reduces infarct size and improves post-ischemic recovery in mice ex vivo but not in vivo. A high-fat diet reduced the survival rate during ischemia and reperfusion, but in a PCSK9-independent manner that was associated with increased plasma matrix metalloproteinase (MMP)9 activity. PCSK9 deletion, but not the inhibition of extracellular PCSK9, reduces infarct sizes in ex vivo hearts, but this effect is overridden in vivo by factors such as MMP9.
Collapse
Affiliation(s)
- Rolf Schreckenberg
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35390 Gießen, Germany; (R.S.); (A.W.); (R.S.)
| | - Annemarie Wolf
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35390 Gießen, Germany; (R.S.); (A.W.); (R.S.)
| | - Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (T.S.); (K.G.); (I.A.S.); (G.Á.); (P.B.)
- Pharmahungary Group, 6722 Szeged, Hungary; (G.B.); (P.F.)
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (T.S.); (K.G.); (I.A.S.); (G.Á.); (P.B.)
- Pharmahungary Group, 6722 Szeged, Hungary; (G.B.); (P.F.)
| | - István Adorján Szabó
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (T.S.); (K.G.); (I.A.S.); (G.Á.); (P.B.)
| | - Gergely Ágoston
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (T.S.); (K.G.); (I.A.S.); (G.Á.); (P.B.)
| | - Gábor Brenner
- Pharmahungary Group, 6722 Szeged, Hungary; (G.B.); (P.F.)
- Department of Pharmacology and Phamacotherapy, Faculty of Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (T.S.); (K.G.); (I.A.S.); (G.Á.); (P.B.)
- Pharmahungary Group, 6722 Szeged, Hungary; (G.B.); (P.F.)
| | - Péter Ferdinandy
- Pharmahungary Group, 6722 Szeged, Hungary; (G.B.); (P.F.)
- Department of Pharmacology and Phamacotherapy, Faculty of Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35390 Gießen, Germany; (R.S.); (A.W.); (R.S.)
| | - Klaus-Dieter Schlüter
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35390 Gießen, Germany; (R.S.); (A.W.); (R.S.)
- Correspondence:
| |
Collapse
|
13
|
Abstract
This article reviews the discovery of PCSK9, its structure-function characteristics, and its presently known and proposed novel biological functions. The major critical function of PCSK9 deduced from human and mouse studies, as well as cellular and structural analyses, is its role in increasing the levels of circulating low-density lipoprotein (LDL)-cholesterol (LDLc), via its ability to enhance the sorting and escort of the cell surface LDL receptor (LDLR) to lysosomes. This implicates the binding of the catalytic domain of PCSK9 to the EGF-A domain of the LDLR. This also requires the presence of the C-terminal Cys/His-rich domain, its binding to the secreted cytosolic cyclase associated protein 1, and possibly another membrane-bound "protein X". Curiously, in PCSK9-deficient mice, an alternative to the downregulation of the surface levels of the LDLR by PCSK9 is taking place in the liver of female mice in a 17β-estradiol-dependent manner by still an unknown mechanism. Recent studies have extended our understanding of the biological functions of PCSK9, namely its implication in septic shock, vascular inflammation, viral infections (Dengue; SARS-CoV-2) or immune checkpoint modulation in cancer via the regulation of the cell surface levels of the T-cell receptor and MHC-I, which govern the antitumoral activity of CD8+ T cells. Because PCSK9 inhibition may be advantageous in these processes, the availability of injectable safe PCSK9 inhibitors that reduces by 50% to 60% LDLc above the effect of statins is highly valuable. Indeed, injectable PCSK9 monoclonal antibody or small interfering RNA could be added to current immunotherapies in cancer/metastasis.
Collapse
Affiliation(s)
- Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC, Canada
| |
Collapse
|
14
|
Vachon L, Smaani A, Tessier N, Jean G, Demers A, Milasan A, Ardo N, Jarry S, Villeneuve L, Alikashani A, Finherty V, Ruiz M, Sorci-Thomas MG, Mayer G, Martel C. Downregulation of low-density lipoprotein receptor mRNA in lymphatic endothelial cells impairs lymphatic function through changes in intracellular lipids. Theranostics 2022; 12:1440-1458. [PMID: 35154499 PMCID: PMC8771568 DOI: 10.7150/thno.58780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 12/20/2021] [Indexed: 11/18/2022] Open
Abstract
Rationale: Impairment in lymphatic transport is associated with the onset and progression of atherosclerosis in animal models. The downregulation of low-density-lipoprotein receptor (LDLR) expression, rather than increased circulating cholesterol level per se, is involved in early atherosclerosis-related lymphatic dysfunction. Enhancing lymphatic function in Ldlr-/- mice with a mutant form of VEGF-C (VEGF-C 152s), a selective VEGFR-3 agonist, successfully delayed atherosclerotic plaque onset when mice were subsequently fed a high-fat diet. However, the specific mechanisms by which LDLR protects against lymphatic function impairment is unknown. Methods and results: We have thus injected wild-type and Pcsk9-/- mice with an adeno-associated virus type 1 expressing a shRNA for silencing Ldlr in vivo. We herein report that lymphatic contractility is reduced upon Ldlr dowregulation in wild-type mice only. Our in vitro experiments reveal that a decrease in LDLR expression at the mRNA level reduces the chromosome duplication phase and the protein expression of VEGFR-3, a membrane-bound key lymphatic marker. Furthermore, it also significantly reduced the levels of 18 lipid subclasses, including key constituents of lipid rafts as well as the transcription of several genes involved in cholesterol biosynthesis and cellular and metabolic processes. Exogenous PCSK9 only reduces lymphatic endothelial-LDLR at the protein level and does not affect lymphatic endothelial cell integrity. This puts forward that PCSK9 may act upon lymphatic muscle cells to mediate its effect on lymphatic contraction capacity in vivo. Conclusion: Our results suggest that treatments that specifically palliate the down regulation of LDLR mRNA in lymphatic endothelial cells preserve the integrity of the lymphatic endothelium and sustain lymphatic function, a prerequisite player in atherosclerosis.
Collapse
Affiliation(s)
- Laurent Vachon
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Ali Smaani
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Gabriel Jean
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Annie Demers
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Nadine Ardo
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Stéphanie Jarry
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | | | - Vincent Finherty
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Matthieu Ruiz
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Metabolomics platform, Montreal, Quebec, Canada
| | | | - Gaétan Mayer
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Susan-Resiga D, Girard E, Essalmani R, Roubtsova A, Marcinkiewicz J, Derbali RM, Evagelidis A, Byun JH, Lebeau PF, Austin RC, Seidah NG. Asialoglycoprotein receptor 1 is a novel PCSK9-independent ligand of liver LDLR cleaved by furin. J Biol Chem 2021; 297:101177. [PMID: 34508778 PMCID: PMC8479480 DOI: 10.1016/j.jbc.2021.101177] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 01/06/2023] Open
Abstract
The hepatic carbohydrate-recognizing asialoglycoprotein receptor (ASGR1) mediates the endocytosis/lysosomal degradation of desialylated glycoproteins following binding to terminal galactose/N-acetylgalactosamine. Human heterozygote carriers of ASGR1 deletions exhibit ∼34% lower risk of coronary artery disease and ∼10% to 14% reduction of non-HDL cholesterol. Since the proprotein convertase PCSK9 is a major degrader of the low-density lipoprotein receptor (LDLR), we investigated the degradation and functionality of LDLR and/or PCSK9 by endogenous/overexpressed ASGR1 using Western blot and immunofluorescence in HepG2-naïve and HepG2-PCSK9-knockout cells. ASGR1, like PCSK9, targets LDLR, and both independently interact with/enhance the degradation of the receptor. This lack of cooperativity between PCSK9 and ASGR1 was confirmed in livers of wildtype (WT) and Pcsk9−/− mice. ASGR1 knockdown in HepG2-naïve cells significantly increased total (∼1.2-fold) and cell-surface (∼4-fold) LDLR protein. In HepG2-PCSK9-knockout cells, ASGR1 silencing led to ∼2-fold higher levels of LDLR protein and DiI (1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate)-LDL uptake associated with ∼9-fold increased cell-surface LDLR. Overexpression of WT-ASGR1/2 primarily reduced levels of immature non-O-glycosylated LDLR (∼110 kDa), whereas the triple Ala-mutant of Gln240/Trp244/Glu253 (characterized by loss of carbohydrate binding) reduced expression of the mature form of LDLR (∼150 kDa), suggesting that ASGR1 binds the LDLR in both a sugar-dependent and -independent fashion. The protease furin cleaves ASGR1 at the RKMK103↓ motif into a secreted form, likely resulting in a loss of function on LDLR. Altogether, we demonstrate that LDLR is the first example of a liver-receptor ligand of ASGR1. We conclude that silencing of ASGR1 and PCSK9 may lead to higher LDL uptake by hepatocytes, thereby providing a novel approach to further reduce LDL cholesterol levels.
Collapse
Affiliation(s)
- Delia Susan-Resiga
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Emmanuelle Girard
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Rachid Essalmani
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Anna Roubtsova
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Jadwiga Marcinkiewicz
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Rabeb M Derbali
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Alexandra Evagelidis
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Jae H Byun
- Division of Nephrology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Paul F Lebeau
- Division of Nephrology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Peyot ML, Roubtsova A, Lussier R, Chamberland A, Essalmani R, Murthy Madiraju SR, Seidah NG, Prentki M, Prat A. Substantial PCSK9 inactivation in β-cells does not modify glucose homeostasis or insulin secretion in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158968. [PMID: 33992809 DOI: 10.1016/j.bbalip.2021.158968] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 01/06/2023]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in cholesterol homeostasis by promoting the degradation of the LDL receptor (LDLR). PCSK9 loss-of-function mutations are associated with increased fasting plasma glucose levels and slightly elevated risk of type 2-diabetes. Considering the known detrimental effects of cholesterol accumulation in β-cell, and the widespread use of PCSK9 inhibitors to treat hypercholesterolemia, it is important to gain insight into the role of pancreatic PCSK9 in glucose homeostasis and β-cell function. We generated the first β-cell-specific KO of PCSK9 (βKO). PCSK9 mRNA and protein expression were reduced by 48% and 78% in βKO islets, respectively, indicating that β-cells constitute a major site of PCSK9 expression. In islets, loss of β-cell PCSK9 resulted in unchanged LDLR protein levels, but reduced LDLR mRNA, indicating that cholesterol internalization is enhanced and that β-cell PCSK9 promotes LDLR degradation. In contrast, whole body PCSK9 KO mice exhibited 2-fold higher LDLR protein levels in islets and a stable expression of cholesterogenic genes. Whole body KO and βKO mice presented normal glucose tolerance, insulin release in response to glucose load and insulin sensitivity. Ex vivo glucose-stimulated insulin secretion in presence or absence of fatty acids was similar in WT and KO islets. Like KO mice, individuals carrying loss-of-function PCSK9 variants may be protected from cholesterol-induced toxicity due to reduced circulating cholesterol levels. Using both whole body KO or βKO models, our data demonstrate that PCSK9 deletion in mouse does not have any toxic effect on β-cell function and glucose homeostasis.
Collapse
Affiliation(s)
- Marie-Line Peyot
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Anna Roubtsova
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - Roxane Lussier
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Ann Chamberland
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - Rachid Essalmani
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - S R Murthy Madiraju
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - Marc Prentki
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada.
| |
Collapse
|
17
|
Affiliation(s)
- Kevin Chemello
- Université de La Réunion, Laboratoire Inserm UMR1188 DéTROI, Sainte-Clotilde, FRANCE
| | - Ali K Jaafar
- Université de La Réunion, Laboratoire Inserm UMR1188 DéTROI, Sainte-Clotilde, FRANCE
| | - Gilles Lambert
- Université de La Réunion, Laboratoire Inserm UMR1188 DéTROI, Sainte-Clotilde, FRANCE
| |
Collapse
|
18
|
Abstract
The kexin-like proprotein convertases perform the initial proteolytic cleavages that ultimately generate a variety of different mature peptide and proteins, ranging from brain neuropeptides to endocrine peptide hormones, to structural proteins, among others. In this review, we present a general introduction to proprotein convertase structure and biochemistry, followed by a comprehensive discussion of each member of the kexin-like subfamily of proprotein convertases. We summarize current knowledge of human proprotein convertase insufficiency syndromes, including genome-wide analyses of convertase polymorphisms, and compare these to convertase null and mutant mouse models. These mouse models have illuminated our understanding of the roles specific convertases play in human disease and have led to the identification of convertase-specific substrates; for example, the identification of procorin as a specific PACE4 substrate in the heart. We also discuss the limitations of mouse null models in interpreting human disease, such as differential precursor cleavage due to species-specific sequence differences, and the challenges presented by functional redundancy among convertases in attempting to assign specific cleavages and/or physiological roles. However, in most cases, knockout mouse models have added substantively both to our knowledge of diseases caused by human proprotein convertase insufficiency and to our appreciation of their normal physiological roles, as clearly seen in the case of the furin, proprotein convertase 1/3, and proprotein convertase 5/6 mouse models. The creation of more sophisticated mouse models with tissue- or temporally-restricted expression of specific convertases will improve our understanding of human proprotein convertase insufficiency and potentially provide support for the emerging concept of therapeutic inhibition of convertases.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Kucsera D, Tóth VE, Gergő D, Vörös I, Onódi Z, Görbe A, Ferdinandy P, Varga ZV. Characterization of the CDAA Diet-Induced Non-alcoholic Steatohepatitis Model: Sex-Specific Differences in Inflammation, Fibrosis, and Cholesterol Metabolism in Middle-Aged Mice. Front Physiol 2021; 12:609465. [PMID: 33692700 PMCID: PMC7937716 DOI: 10.3389/fphys.2021.609465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/11/2021] [Indexed: 01/14/2023] Open
Abstract
Background The prevalence of non-alcoholic steatohepatitis (NASH) rapidly increases with associated metabolic disorders such as dyslipidemia; therefore, NASH is now considered an independent risk factor of cardiovascular diseases. NASH displays sex-linked epidemiological, phenotypical, and molecular differences; however, little is known about the background of these sex-specific differences on the molecular level. Objectives We aimed to assess sex-specific differences in the expression of inflammatory and fibrotic genes, as well as in cholesterol metabolism, focusing on the expression of Pcsk9 in several tissues in a mouse model of NASH that shows the typical features of the human condition. Methods and Results We fed 10-months-old male and female C57Bl/6J mice with a NASH-inducing CDAA or corresponding control diet for 8 weeks. We found that, compared to the control male mice baseline, hepatic Pcsk9 expression as well as serum PCSK9 level was significantly higher in females, and both circulating PCSK9 level and the hepatic Pcsk9 gene were markedly decreased in female mice during NASH development. Histological analysis revealed that male and female mice develop a similar degree of steatosis; however, fibrosis was more pronounced in males upon CDAA diet feeding. Strikingly, female mice have higher hepatic expression of the pro-inflammatory cytokines (Il1b, Ifng), and increased IL-1β cleavage by the NLRP3 inflammasome, and a decrease in Clec4f+ resident Kupffer cell population in comparison to males in the CDAA-fed groups. Conclusion This is the first demonstration that there are critical sex-specific differences during NASH development in middle-aged mice regarding inflammation, fibrosis, and cholesterol metabolism and that changes in PCSK9 and IL-1β are likely important contributors to sex-specific changes during the transition to NASH.
Collapse
Affiliation(s)
- Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Viktória E Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Dorottya Gergő
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Imre Vörös
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Cardiometabolic Immunology Research Group, Budapest, Hungary
| |
Collapse
|
20
|
Simeone PG, Vadini F, Tripaldi R, Liani R, Ciotti S, Di Castelnuovo A, Cipollone F, Santilli F. Sex-Specific Association of Endogenous PCSK9 With Memory Function in Elderly Subjects at High Cardiovascular Risk. Front Aging Neurosci 2021; 13:632655. [PMID: 33776743 PMCID: PMC7990768 DOI: 10.3389/fnagi.2021.632655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Growing evidence indicates that cognitive decline and cardiovascular diseases (CVDs) share common vascular risk factors. Protease proprotein convertase subtilisin/kexin type 9 (PCSK9) is associated with CV disease risk and has been also involved in neuronal differentiation. Aim: Evaluate whether in patients at high CV risk cognitive function is related to PCSK9 levels. Methods. One hundred sixty-six patients (67 female) were enrolled. A detailed neuropsychological (NP) assessment was performed. PCSK9 levels were measured with ELISA. Results: Men had significantly higher short-term memory, executive function, and praxic and mental representation skills, as reflected by Forward Digit Span (FDS) (p = 0.005), Trail Making Test-A (TMT-A) (p = 0.047), Clock Drawing Test (CDT) (0.016). Endogenous PCSK9 levels were higher in female (p = 0.005). On linear regression analysis PCSK9 predicts short term memory only in females (Beta = 0.408, p = 0.001), with an interaction between PCSK9 and gender (p = 0.004 for interaction PCSK9 by sex). The association of PCSK9 with FDS in female was partially mediated by waist circumference (mediation effect 8.5%). Conclusions: In patients at high CV risk short term memory was directly related to PCSK9 levels only in women, revealing the relevance of sex in this relationship. The association of PCSK9 with memory function may be mediated, at least in part, by waist circumference.
Collapse
Affiliation(s)
- Paola G Simeone
- Department of Medicine and Aging and Center for Advanced Studies and Technology, Chieti, Italy
| | - Francesco Vadini
- Psychoinfectivology Service, Pescara General Hospital, Pescara, Italy
| | - Romina Tripaldi
- Department of Medicine and Aging and Center for Advanced Studies and Technology, Chieti, Italy
| | - Rossella Liani
- Department of Medicine and Aging and Center for Advanced Studies and Technology, Chieti, Italy
| | - Sonia Ciotti
- Department of Medicine and Aging and Center for Advanced Studies and Technology, Chieti, Italy
| | | | - Francesco Cipollone
- Department of Medicine and Aging and Center for Advanced Studies and Technology, Chieti, Italy
| | - Francesca Santilli
- Department of Medicine and Aging and Center for Advanced Studies and Technology, Chieti, Italy
| |
Collapse
|
21
|
Nuclear magnetic resonance reveals postprandial low-density lipoprotein cholesterol determined by enzymatic method could be a misleading indicator. Clin Chim Acta 2020; 514:59-65. [PMID: 33333042 DOI: 10.1016/j.cca.2020.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Serum concentration of low-density lipoprotein cholesterol (LDL-C) is markedly reduced after a meal. Does postprandial cholesterol in LDL truly decline via clearance of LDL particles or is there simply a redistribution of cholesterol in LDL subclasses? Thus, we sought to evaluate whether postprandial decline of LDL-C reflects a reduction of LDL particle and to assess the correlation between proprotein convertase subtilisin/kexin type 9 (PCSK9) concentration and postprandial atherogenic lipoproteins profile. METHODS Eighty-seven persons were enrolled in this study. We measured lipid profiles by enzymatic and nuclear magnetic resonance (NMR)-based methods and serum PCSK9 concentration by enzyme-linked immunosorbent assays before and after a meal. Plasma samples were collected after a 10-h fasting and 2 and 4 h post-meal. RESULTS Compared to the fasting status, there was significant postprandial decline of LDL-C measured enzymatically (LDL-Ce) at 2nd and 4th h [99.38 (80.43, 120.65) vs 95.51 (74.25, 117.17) vs 87.01 (69.99, 108.28) mg/dl, p < 0.000]. But there was no significant reduction in LDL particle and its cholesterol content (LDL-Cn) determined by NMR. Just the postprandial large LDL particle [186.45 (151.36, 229.42) vs 176.92 (147.43, 220.91) vs 181.77 (149.05, 224.17), p < 0.000] and its cholesterol content [19.10 (15.09, 22.37) vs 18.28 (14.59, 21.84) vs 17.79 (14.62, 22.14), p < 0.000] were greatly decreased at 2nd and 4th h compared to the fasting one. Interestingly, postprandial serum PCSK9 was decreased at 2nd and 4th h compared with fasting concentration [298.75 (233.25, 396.92) vs 257.34 (207.52, 342.36) vs 250.57 (215.02, 339.66) ng/ml, p < 0.000]. The postprandial percent decrease in serum PCSK9 at 4th h was positively correlated to the percent decline in postprandial LDL-Ce (r = 0.252, p = 0.019) but was independently associated with the percent increase in remnant cholesterol (r = 0.262, p = 0.016). CONCLUSIONS Postprandial decline of LDL-C determined enzymatically was not confirmed by NMR-based methods. Indeed, there exists cholesterol redistribution in LDL subclasses following a meal. The decrease of postprandial PCSK9 may be secondary to the increase in intrahepatic lipids following food intake.
Collapse
|
22
|
Tang Y, Li SL, Hu JH, Sun KJ, Liu LL, Xu DY. Research progress on alternative non-classical mechanisms of PCSK9 in atherosclerosis in patients with and without diabetes. Cardiovasc Diabetol 2020; 19:33. [PMID: 32169071 PMCID: PMC7071562 DOI: 10.1186/s12933-020-01009-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022] Open
Abstract
The proprotein convertase subtilisin/kexin type 9 (PCSK9) acts via a canonical pathway to regulate circulating low-density lipoprotein-cholesterol (LDL-C) via degradation of the LDL receptor (LDLR) on the liver cell surface. Published research has shown that PCSK9 is involved in atherosclerosis via a variety of non-classical mechanisms that involve lysosomal, inflammatory, apoptotic, mitochondrial, and immune pathways. In this review paper, we summarized these additional mechanisms and described how anti-PCSK9 therapy exerts effects through these mechanisms. These additional pathways further illustrate the regulatory role of PCSK9 in atherosclerosis and offer an in-depth interpretation of how the PCSK9 inhibitor exerts effects on the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Ying Tang
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Sheng-Lan Li
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jia-Hui Hu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Kai-Jun Sun
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Lei-Ling Liu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Dan-Yan Xu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
23
|
Development of a Predictive Model to Induce Atherogenesis and Hepato-Renal Impairment in Female Rats. Biomolecules 2019; 9:biom9110664. [PMID: 31671756 PMCID: PMC6921007 DOI: 10.3390/biom9110664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Therapeutic approaches for the treatment of dyslipidemia and atherosclerosis have radically changed in recent decades. Part of this advance undeniably stems from basic biomedical research that has provided a better understanding and identification of new therapeutic targets. The aim of this work was to develop a model to induce atherogenesis and hepato-renal impairment in female Wistar rats. The following groups received the respective treatments for 60 days: control animals, non-ovariectomized rats that received an atherogenic diet (NEAD), ovariectomized rats that received an atherogenic diet (NOAD), non-ovariectomized rats that received an atherogenic diet and oral Nω-nitro-l-arginine methyl ester hydrochloride (l-NAME; LEAD), and ovariectomized rats that received an atherogenic diet and oral l-NAME (LOAD). Animals in the NEAD, NOAD, LEAD, and LOAD groups also received methimazole and cholecalciferol daily. Urinary, biochemical, hemodynamic, and electrocardiographic parameters and renal function were assessed. Samples of the liver, heart, kidney, and arteries were collected to investigate redox status and perform histopathological analyses. All of the groups developed dyslipidemia and hepatic steatosis. Only the NEAD group developed arterial lesions that were compatible with fatty streaks. Renal function was significantly impaired in the LEAD and NOAD groups. These results indicate a viable alternative to induce atherogenesis and hepato-renal impairment in female rats.
Collapse
|
24
|
Ben Djoudi Ouadda A, Gauthier MS, Susan-Resiga D, Girard E, Essalmani R, Black M, Marcinkiewicz J, Forget D, Hamelin J, Evagelidis A, Ly K, Day R, Galarneau L, Corbin F, Coulombe B, Çaku A, Tagliabracci VS, Seidah NG. Ser-Phosphorylation of PCSK9 (Proprotein Convertase Subtilisin-Kexin 9) by Fam20C (Family With Sequence Similarity 20, Member C) Kinase Enhances Its Ability to Degrade the LDLR (Low-Density Lipoprotein Receptor). Arterioscler Thromb Vasc Biol 2019; 39:1996-2013. [PMID: 31553664 DOI: 10.1161/atvbaha.119.313247] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE PCSK9 (proprotein convertase subtilisin-kexin 9) enhances the degradation of the LDLR (low-density lipoprotein receptor) in endosomes/lysosomes. This study aimed to determine the sites of PCSK9 phosphorylation at Ser-residues and the consequences of such posttranslational modification on the secretion and activity of PCSK9 on the LDLR. Approach and Results: Fam20C (family with sequence similarity 20, member C) phosphorylates serines in secretory proteins containing the motif S-X-E/phospho-Ser, including the cholesterol-regulating PCSK9. In situ hybridization of Fam20C mRNA during development and in adult mice revealed a wide tissue distribution, including liver, but not small intestine. Here, we show that Fam20C phosphorylates PCSK9 at Serines 47, 666, 668, and 688. In hepatocytes, phosphorylation enhances PCSK9 secretion and maximizes its induced degradation of the LDLR via the extracellular and intracellular pathways. Replacing any of the 4 Ser by the phosphomimetic Glu or Asp enhanced PCSK9 activity only when the other sites are phosphorylated, whereas Ala substitutions reduced it, as evidenced by Western blotting, Elisa, and LDLR-immunolabeling. This newly uncovered PCSK9/LDLR regulation mechanism refines our understanding of the implication of global PCSK9 phosphorylation in the modulation of LDL-cholesterol and rationalizes the consequence of natural mutations, for example, S668R and E670G. Finally, the relationship of Ser-phosphorylation to the implication of PCSK9 in regulating LDL-cholesterol in the neurological Fragile X-syndrome disorder was investigated. CONCLUSIONS Ser-phosphorylation of PCSK9 maximizes both its secretion and activity on the LDLR. Mass spectrometric approaches to measure such modifications were developed and applied to quantify the levels of bioactive PCSK9 in human plasma under normal and pathological conditions.
Collapse
Affiliation(s)
- Ali Ben Djoudi Ouadda
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Marie-Soleil Gauthier
- Translational Proteomics Research Unit, Clinical Research Institute of Montreal (IRCM, affiliated to the Université de Montréal), QC, Canada (M.-S.G., D.F., B.C.)
| | - Delia Susan-Resiga
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Emmanuelle Girard
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Rachid Essalmani
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Miles Black
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (M.B., V.S.T.)
| | - Jadwiga Marcinkiewicz
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Diane Forget
- Translational Proteomics Research Unit, Clinical Research Institute of Montreal (IRCM, affiliated to the Université de Montréal), QC, Canada (M.-S.G., D.F., B.C.)
| | - Josée Hamelin
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Alexandra Evagelidis
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| | - Kevin Ly
- Institut de Pharmacologie, Université de Sherbrooke, QC, Canada (K.L., R.D.)
| | - Robert Day
- Institut de Pharmacologie, Université de Sherbrooke, QC, Canada (K.L., R.D.)
| | - Luc Galarneau
- Department of Biochemistry, Université de Sherbrooke, and Centre de Recherche du CHUS, QC, Canada (L.G., F.C., A.Ç.)
| | - Francois Corbin
- Department of Biochemistry, Université de Sherbrooke, and Centre de Recherche du CHUS, QC, Canada (L.G., F.C., A.Ç.)
| | - Benoit Coulombe
- Translational Proteomics Research Unit, Clinical Research Institute of Montreal (IRCM, affiliated to the Université de Montréal), QC, Canada (M.-S.G., D.F., B.C.)
| | - Artuela Çaku
- Department of Biochemistry, Université de Sherbrooke, and Centre de Recherche du CHUS, QC, Canada (L.G., F.C., A.Ç.)
| | - Vincent S Tagliabracci
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (M.B., V.S.T.)
| | - Nabil G Seidah
- From the Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM; affiliated to the Université de Montréal), QC, Canada (A.B.D.O., D.S.-R., E.G., R.E., J.M., J.H., A.E., N.G.S.)
| |
Collapse
|
25
|
Nakajima K, Tokita Y, Tanaka A, Takahashi S. The VLDL receptor plays a key role in the metabolism of postprandial remnant lipoproteins. Clin Chim Acta 2019; 495:382-393. [PMID: 31078566 DOI: 10.1016/j.cca.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022]
Abstract
A new concept to account for the process of postprandial remnant lipoprotein metabolism is proposed based on the characteristics of lipoprotein particles and their receptors. The characteristics of remnant lipoprotein (RLP) were investigated using an immuno-separation method. The majority of the postprandial lipoproteins increased after fat intake was shown to be VLDL remnants, not chylomicron (CM) remnants, based on the significantly high ratio of apoB100/apoB48 in the RLP and the high degree of similarity in the particle size of the apoB48 and apoB100 carrying lipoproteins, which fluctuate in parallel during a 6 h period after fat intake. The VLDL receptor was discovered as a receptor for TG-rich lipoprotein metabolism and is located in peripheral tissues such as skeletal muscle, adipose tissue, etc., but not in the liver. Postprandial VLDL particles are strongly bound and internalized into cells expressing the VLDL receptor. Ligands that bind to VLDL receptor, such as LPL and Lp(a), present in RLP. The presence of various specific ligands in VLDL remnants may enhance the capacity for binding to the VLDL receptor, which play the role primarily for energy delivery to the peripheral tissues, but is also a causal factor in atherogenic diseases when excessively and/or continuously remained in plasma.
Collapse
Affiliation(s)
- Katsuyuki Nakajima
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan.
| | - Yoshiharu Tokita
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Akira Tanaka
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan
| | - Sadao Takahashi
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Division of Diabetes, Ageo Central General Hospital, Saitama, Japan
| |
Collapse
|
26
|
Bordicchia M, Spannella F, Ferretti G, Bacchetti T, Vignini A, Di Pentima C, Mazzanti L, Sarzani R. PCSK9 is Expressed in Human Visceral Adipose Tissue and Regulated by Insulin and Cardiac Natriuretic Peptides. Int J Mol Sci 2019; 20:ijms20020245. [PMID: 30634533 PMCID: PMC6358804 DOI: 10.3390/ijms20020245] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/05/2018] [Accepted: 01/04/2019] [Indexed: 01/14/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to and degrades the low-density lipoprotein receptor (LDLR), contributing to hypercholesterolemia. Adipose tissue plays a role in lipoprotein metabolism, but there are almost no data about PCSK9 and LDLR regulation in human adipocytes. We studied PCSK9 and LDLR regulation by insulin, atrial natriuretic peptide (ANP, a potent lipolytic agonist that antagonizes insulin), and LDL in visceral adipose tissue (VAT) and in human cultured adipocytes. PCSK9 was expressed in VAT and its expression was positively correlated with body mass index (BMI). Both intracellular mature and secreted PCSK9 were abundant in cultured human adipocytes. Insulin induced PCSK9, LDLR, and sterol-regulatory element-binding protein-1c (SREBP-1c) and -2 expression (SREBP-2). ANP reduced insulin-induced PCSK9, especially in the context of a medium simulating hyperglycemia. Human LDL induced both mature and secreted PCSK9 and reduced LDLR. ANP indirectly blocked the LDLR degradation, reducing the positive effect of LDL on PCSK9. In conclusion, PCSK9 is expressed in human adipocytes. When the expression of PCSK9 is induced, LDLR is reduced through the PCSK9-mediated degradation. On the contrary, when the induction of PCSK9 by insulin and LDL is partially blocked by ANP, the LDLR degradation is reduced. This suggests that NPs could be able to control LDLR levels, preventing PCSK9 overexpression.
Collapse
Affiliation(s)
- Marica Bordicchia
- Internal Medicine and Geriatrics, Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", 60126 Ancona, Italy.
| | - Francesco Spannella
- Internal Medicine and Geriatrics, Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", 60126 Ancona, Italy.
- Internal Medicine and Geriatrics, "Hypertension Excellence Centre" of the European Society of Hypertension, IRCCS-INRCA, 60127 Ancona, Italy.
| | - Gianna Ferretti
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, School of Nutrition, University "Politecnica delle Marche", 60126 Ancona, Italy.
| | - Tiziana Bacchetti
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, School of Nutrition, University "Politecnica delle Marche", 60126 Ancona, Italy.
| | - Arianna Vignini
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, School of Nutrition, University "Politecnica delle Marche", 60126 Ancona, Italy.
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", 60126 Ancona, Italy.
- Internal Medicine and Geriatrics, "Hypertension Excellence Centre" of the European Society of Hypertension, IRCCS-INRCA, 60127 Ancona, Italy.
| | - Laura Mazzanti
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, School of Nutrition, University "Politecnica delle Marche", 60126 Ancona, Italy.
| | - Riccardo Sarzani
- Internal Medicine and Geriatrics, Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", 60126 Ancona, Italy.
- Internal Medicine and Geriatrics, "Hypertension Excellence Centre" of the European Society of Hypertension, IRCCS-INRCA, 60127 Ancona, Italy.
| |
Collapse
|
27
|
Dufurrena Q, Bäck N, Mains R, Hodgson L, Tanowitz H, Mandela P, Eipper B, Kuliawat R. Kalirin/Trio Rho GDP/GTP exchange factors regulate proinsulin and insulin secretion. J Mol Endocrinol 2018; 62:JME-18-0048.R2. [PMID: 30407917 PMCID: PMC6494717 DOI: 10.1530/jme-18-0048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022]
Abstract
Key features for progression to pancreatic β-cell failure and disease are loss of glucose responsiveness and an increased ratio of secreted proinsulin to insulin. Proinsulin and insulin are stored in secretory granules (SGs) and the fine-tuning of hormone output requires signal mediated recruitment of select SG populations according to intracellular location and age. The GTPase Rac1 coordinates multiple signaling pathways that specify SG release and Rac1 activity is controlled in part by GDP/GTP exchange factors (GEFs). To explore the function of two large multidomain GEFs, Kalirin and Trio in β-cells, we manipulated their Rac1-specific GEF1 domain activity by using small molecule inhibitors and by genetically ablating Kalirin. We examined age related secretory granule behavior employing radiolabeling protocols. Loss of Kalirin/Trio function attenuated radioactive proinsulin release by reducing constitutive-like secretion and exocytosis of 2-hour old granules. At later chase times or at steady state, Kalirin/Trio manipulations decreased glucose stimulated insulin output. Finally, use of a Rac1 FRET biosensor with cultured β-cell lines, demonstrated that Kalirin/Trio GEF1 activity was required for normal rearrangement of Rac1 to the plasma membrane in response to glucose. Rac1 activation can be evoked by both glucose metabolism and signaling through the incretin glucagon-like peptide 1 (GLP-1) receptor. GLP-1 addition restored Rac1 localization/activity and insulin secretion in the absence of Kalirin, thereby assigning Kalirin's participation to stimulatory glucose signaling.
Collapse
Affiliation(s)
- Quinn Dufurrena
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY
| | - Nils Bäck
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Richard Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Herbert Tanowitz
- Departments of Pathology, Medicine, Albert Einstein College of Medicine, Bronx, NY
| | | | - Betty Eipper
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT
| | - Regina Kuliawat
- Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
28
|
Palmisano BT, Zhu L, Eckel RH, Stafford JM. Sex differences in lipid and lipoprotein metabolism. Mol Metab 2018; 15:45-55. [PMID: 29858147 PMCID: PMC6066747 DOI: 10.1016/j.molmet.2018.05.008] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Endogenous sex hormones are important for metabolic health in men and women. Before menopause, women are protected from atherosclerotic cardiovascular disease (ASCVD) relative to men. Women have fewer cardiovascular complications of obesity compared to men with obesity. Endogenous estrogens have been proposed as a mechanism that lessens ASCVD risk, as risk of glucose and lipid abnormalities increases when endogenous estrogens decline with menopause. While baseline risk is higher in males than females, endogenously produced androgens are also protective against fatty liver, diabetes and ASCVD, as risk goes up with androgen deprivation and with the decline in androgens with age. SCOPE OF REVIEW In this review, we discuss evidence of how endogenous sex hormones and hormone treatment approaches impact fatty acid, triglyceride, and cholesterol metabolism to influence metabolic and cardiovascular risk. We also discuss potential reasons for why treatment strategies with estrogens and androgens in older individuals fail to fully recapitulate the effects of endogenous sex hormones. MAJOR CONCLUSIONS The pathways that confer ASCVD protection for women are of potential therapeutic relevance. Despite protection relative to men, ASCVD is still the major cause of mortality in women. Additionally, diabetic women have similar ASCVD risk as diabetic men, suggesting that the presence of diabetes may offset the protective cardiovascular effects of being female through unknown mechanisms.
Collapse
Affiliation(s)
- Brian T Palmisano
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, USA
| | - Lin Zhu
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, USA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, USA
| | - John M Stafford
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, USA; Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, USA.
| |
Collapse
|
29
|
Kawakami R, Nozato Y, Nakagami H, Ikeda Y, Shimamura M, Yoshida S, Sun J, Kawano T, Takami Y, Noma T, Rakugi H, Minamino T, Morishita R. Development of vaccine for dyslipidemia targeted to a proprotein convertase subtilisin/kexin type 9 (PCSK9) epitope in mice. PLoS One 2018; 13:e0191895. [PMID: 29438441 PMCID: PMC5811007 DOI: 10.1371/journal.pone.0191895] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates expression of low-density lipoprotein (LDL) receptors via receptor internalization and subsequent lysosomal degradation. Thus, an anti-PCSK9 antibody is well known as an anti-hyperlipidemia drug. Here, we aimed to develop vaccine for a long-term treatment of dyslipidemia targeted to PCSK9. In This study, we designed a peptide vaccine for mouse PCSK-9, which consisted of short peptides conjugated to keyhole limpet hemocyanin (KLH) as a carrier protein. Vaccines were administered to male apolipoprotein E (ApoE) deficient mice with adjuvants and significantly elicited an antibody response against PCSK9. The PCSK9 vaccines were administered to mice three times in 2-week intervals, and antibody titers and lipoprotein levels were evaluated up to 24 weeks after the first immunization to determine the therapeutic effect. Anti-PCSK9 antibody titers reached peak levels 6 weeks after the first immunization, and theses titers were maintained for up to 24 weeks. Decreased plasma levels of total cholesterol, very low-density lipoprotein (VLDL), and chylomicron (CM) were maintained for up to 24 weeks. Immunized mice exhibited a significant increase in cell-surface LDL receptor expression. Stimulation with KLH, but not PCSK9, induced the production of INF-gamma and interleukin-4 (IL-4), as determined with ELISPOT assays, thus indicating that PCSK9 vaccine did not elicit T-cell activation in our vaccine system. The present anti-PCSK9 vaccine induced long-lasting anti-PCSK9 antibody production and improved lipoprotein profiles. Thus, anti-PCSK9 vaccine could become a new option for the treatment of dyslipidemia as a long-acting therapy in future.
Collapse
Affiliation(s)
- Ryo Kawakami
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan.,Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuka Ikeda
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Munehisa Shimamura
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shota Yoshida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jiao Sun
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomohiro Kawano
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takahisa Noma
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
30
|
Ce O, Rs P, Ab W, S D, Cj W, Qm M, D L. Potential Link Between Proprotein Convertase Subtilisin/Kexin Type 9 and Alzheimer's Disease. ACTA ACUST UNITED AC 2018; 1. [PMID: 32352077 DOI: 10.31531/2581-4745.1000106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease [AD] is not only the most common neurodegenerative disease but is also currently incurable. Proprotein convertase subtilisin/kexin-9 [PCSK9] is an indirect regulator of plasma low density lipoprotein [LDL] levels controlling LDL receptor expression at the plasma membrane. PCSK9 also appears to regulate the development of glucose intolerance, insulin resistance, abdominal obesity, inflammation, and hypertension, conditions that have been identified as risk factors for AD. PCSK9 levels also depend on age, sex, and ethnic background, factors associated with AD. Herein, we will review indirect evidence that suggests a link between PCSK9 levels and AD.
Collapse
Affiliation(s)
- Oldham Ce
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Powell Rs
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Williams Ab
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Dixon S
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Wooten Cj
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Melendez Qm
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Lopez D
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| |
Collapse
|
31
|
Zhang N, Zhang Y, Lin J, Qiu X, Chen L, Pan X, Lu Y, Zhang J, Wang Y, Li D, Wang L. Low-density lipoprotein receptor deficiency impaired mice osteoblastogenesis in vitro. Biosci Trends 2017; 11:658-666. [PMID: 29269714 DOI: 10.5582/bst.2017.01267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Postmenopausal osteoporosis affected most elderly women with co-existence of lipid and bone metabolism disorders. However, the cellular and molecular mechanisms underlying the parallel progression and cross-talk of these systems remained unclear. In the present study, low-density lipoprotein receptor knockout (LDLR-/-) mice were chosen to elucidate the effect of LDLR in regulating the differentiation of osteoblasts, which were responsible for bone formation and modulation of osteoclastogenesis. Primary osteoblasts were isolated from the calvarium of newborn LDLR-/- or wild-type mice followed by osteoblastic differentiation culture in vitro. Alkaline phosphatase activity was significantly decreased in LDLR-/- osteoblasts compared to wild-type controls, combined with calcium deposit formation delay, implying impaired osteoblastogenesis in vitro. Consistent with these findings, the expression of runt-related transcription factor 2 (Runx2) was decreased 3 days after differentiation in LDLR-/- osteoblasts compared to wild-type controls. Moreover, the expression of Osterix was decreased 7 days after differentiation in LDLR-/- osteoblasts compared to wild-type controls, later than Runx2.However, the osteoclastogenesis modulation role of osteoblasts was unaffected by the LDLR deficiency, evidenced by the same level of osteoprotegerin (OPG)/receptor activator of nuclear factor-κ B ligand (RANKL) axis between LDLR-/- and wild-type control osteoblasts. Our results provide a novel insight into the role of LDLR during osteoblastic differentiation and improve understanding of cross-talk between bone and lipid metabolisms.
Collapse
Affiliation(s)
- Na Zhang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Yang Zhang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine
| | - Jing Lin
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Xuemin Qiu
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Lanting Chen
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Xinyao Pan
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Youhui Lu
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Jiali Zhang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Yan Wang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Dajin Li
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| | - Ling Wang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University.,The Academy of Integrative Medicine of Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases
| |
Collapse
|
32
|
Levenson AE, Shah AS, Khoury PR, Kimball TR, Urbina EM, de Ferranti SD, Maahs DM, Dolan LM, Wadwa RP, Biddinger SB. Obesity and type 2 diabetes are associated with elevated PCSK9 levels in young women. Pediatr Diabetes 2017; 18:755-760. [PMID: 28093849 PMCID: PMC5513789 DOI: 10.1111/pedi.12490] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 11/02/2016] [Accepted: 11/22/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key regulator of low-density lipoprotein cholesterol and cardiovascular disease risk, and is an emerging therapeutic target. OBJECTIVE We compared serum PCSK9 levels in young adults, with and without type 2 diabetes. SUBJECTS AND METHODS Cross-sectional analysis was conducted in a cohort, aged 15 to 26 years, in Cincinnati, OH, from 2005 to 2010. Serum PCSK9 levels were measured in 94 youth with type 2 diabetes, 93 obese control subjects, and 99 lean control subjects. Correlative analyses were conducted to determine significant covariates of PCSK9 by group and sex, and multivariate linear regression models were used to study the independent determinants of PCSK9. RESULTS In females, PCSK9 levels were significantly increased in the obese and type 2 diabetes subjects relative to the lean controls (P < .01). Moreover, PCSK9 was positively correlated with multiple metabolic parameters in females: body mass index, systolic blood pressure, fasting glucose, fasting insulin, and C-reactive protein levels (P ≤ .02). In males, PCSK9 levels were decreased overall compared with females (P = .03), and did not differ between the lean, obese, or type 2 diabetes groups. CONCLUSIONS Obesity and type 2 diabetes were associated with significantly higher levels of PCSK9 in young women, but not in young men. These data suggest that sex could modify the effects of obesity and diabetes on PCSK9 in young adults.
Collapse
Affiliation(s)
- Amy E. Levenson
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Amy S. Shah
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229, United States
| | - Philip R. Khoury
- Division of Cardiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229, United States
| | - Thomas R. Kimball
- Division of Cardiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229, United States
| | - Elaine M. Urbina
- Division of Cardiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229, United States
| | - Sarah D. de Ferranti
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - David M. Maahs
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Lawrence M. Dolan
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229, United States
| | - R. Paul Wadwa
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Sudha B. Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
33
|
Hypercholesterolemia: The role of PCSK9. Arch Biochem Biophys 2017; 625-626:39-53. [DOI: 10.1016/j.abb.2017.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 01/06/2023]
|
34
|
Takahashi S. Triglyceride Rich Lipoprotein -LPL-VLDL Receptor and Lp(a)-VLDL Receptor Pathways for Macrophage Foam Cell Formation. J Atheroscler Thromb 2017; 24:552-559. [PMID: 28428482 PMCID: PMC5453679 DOI: 10.5551/jat.rv17004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Very low-density lipoprotein (VLDL) receptor is a member of the low-density lipoprotein (LDL) receptor family. It binds triglyceride rich lipoprotein (TGRL) but not LDL, because it recognizes apolipoprotein (apo)E only but not apoB. The VLDL receptor functions as a peripheral lipoprotein receptor in concert with lipoprotein lipase (LPL) in heart, muscle, adipose tissue and macrophages. In contrast to the LDL receptor, VLDL receptor binds apo E2/2 VLDL and apoE3/3 VLDL particles, and its expression is not down-regulated by intracellular lipoproteins. It has been reported that both LDL-cholesterol (LDL-C) and postprandial triglyceride (chyromicron and VLDL remnants) are risk factors for human atherosclerotic cardiovascular disease (ASCVD). True ligands such as lipoprotein particles of the VLDL receptor are chyromicron remnant (CMR) and VLDL remnant (postprandial hyperlipidemia). Although the oxidized LDL (oxLDL)-scavenger receptors pathway is considered to be the main mechanism for macrophage foam cell formation, it seems that the TGRL-LPL-VLDL receptor pathway is also involved. Since Lp(a) is one of the ligands for the VLDL receptor, the Lp(a)- VLDL receptor pathway is another potential alternative. The expression of VLDL receptor protein in mouse macrophages is modest compared to that in rabbit and human macrophages, both in vitro and in vivo. Therefore, we need to elucidate the mechanism of human ASCVD not by using the mouse model and scavenger receptors pathway but instead using the rabbit model and VLDL receptor pathway, respectively.
Collapse
|
35
|
Scott Kiss R, Sniderman A. Shunts, channels and lipoprotein endosomal traffic: a new model of cholesterol homeostasis in the hepatocyte. J Biomed Res 2017; 31:95-107. [PMID: 28808191 PMCID: PMC5445212 DOI: 10.7555/jbr.31.20160139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The liver directs cholesterol metabolism in the organism. All the major fluxes of cholesterol within the body involve the liver: dietary cholesterol is directed to the liver; cholesterol from peripheral cells goes to the liver; the liver is a major site of cholesterol synthesis for the organism; cholesterol is secreted from the liver within the bile, within apoB lipoproteins and translocated to nascent HDL. The conventional model of cholesterol homeostasis posits that cholesterol from any source enters a common, rapidly exchangeable pool within the cell, which is in equilibrium with a regulatory pool. Increased influx of cholesterol leads rapidly to decreased synthesis of cholesterol. This model was developed based on in vitro studies in the fibroblast and validated only for LDL particles. The challenges the liver must meet in vivo to achieve cholesterol homeostasis are far more complex. Our model posits that the cholesterol derived from three different lipoproteins endosomes has three different fates: LDL-derived cholesterol is largely recycled within VLDL with most of the cholesterol shunted through the hepatocyte without entering the exchangeable pool of cholesterol; high density lipoprotein-derived CE is transcytosed into bile; and chylomicron remnant-derived cholesterol primarily enters the regulatory pool within the hepatocyte. These endosomal channels represent distinct physiological pathways and hepatic homeostasis represents the net result of the outcomes of these distinct channels. Our model takes into account the distinct physiological challenges the hepatocyte must meet, underlie the pathophysiology of many of the apoB dyslipoproteinemias and account for the sustained effectiveness of therapeutic agents such as statins.
Collapse
Affiliation(s)
- Robert Scott Kiss
- Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Allan Sniderman
- Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
36
|
Besse-Patin A, Léveillé M, Oropeza D, Nguyen BN, Prat A, Estall JL. Estrogen Signals Through Peroxisome Proliferator-Activated Receptor-γ Coactivator 1α to Reduce Oxidative Damage Associated With Diet-Induced Fatty Liver Disease. Gastroenterology 2017; 152:243-256. [PMID: 27658772 DOI: 10.1053/j.gastro.2016.09.017] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Inefficient fatty acid oxidation in mitochondria and increased oxidative damage are features of non-alcoholic fatty liver disease (NAFLD). In rodent models and patients with NAFLD, hepatic expression of peroxisome proliferator-activated receptor-γ (PPARG) coactivator 1α (PPARGC1A or PGC1A) is inversely correlated with liver fat and disease severity. A common polymorphism in this gene (rs8192678, encoding Gly482Ser) has been associated with NAFLD. We investigated whether reduced expression of PGC1A contributes to development of NAFLD using mouse models, primary hepatocytes, and human cell lines. METHODS HepG2 cells were transfected with variants of PPARGC1A and protein and messenger RNA levels were measured. Mice with liver-specific hemizygous or homozygous disruption of Ppargc1a (Ppargc1af/+Alb-cre+/0 and Ppargc1af/f Alb-cre+/0 mice, respectively) were fed regular chow (control) or a high-fat diet supplemented with 30% d-fructose in drinking water (obesogenic diet) for 25-33 weeks. Liver tissues were analyzed by histology and by immunoblotting. Primary hepatocytes were analyzed for insulin signaling, reactive oxygen species, and estrogen response. Luciferase reporter expression was measured in transfected H2.35 cells expressing an estrogen receptor reporter gene, estrogen receptor 1, and/or PGC1A/B. RESULTS The serine 482 variant of the human PGC1A protein had a shorter half-life than the glycine 482 variant when expressed in HepG2 cells. Liver tissues from mice with liver-specific hemizygous disruption of Ppargc1a placed on an obesogenic diet expressed increased markers of inflammation and fibrosis and decreased levels of antioxidant enzymes compared with the Ppargc1a+/+ on the same diet. Oxidative damage was observed in livers from Ppargc1af/+Alb-cre+/0 mice of each sex, in a cell-autonomous manner, but was greater in livers from the female mice. Expression of PGC1A in H2.35 cells coactivated estrogen receptor 1 and was required for estrogen-dependent expression of genes that encode antioxidant proteins. These findings could account for the increased liver damage observed in female Ppargc1af/+Alb-cre+/0 mice; while, compensatory increases in PPARG coactivator 1β could prevent oxidative damage associated with complete loss of PGC1A expression in Ppargc1af/fAlb-cre+/0 female mice. CONCLUSIONS In mice, loss of estrogen signaling contributes to oxidative damage caused by low levels of PGC1A in liver, exacerbating steatohepatitis associated with diets high in fructose and fat.
Collapse
Affiliation(s)
- Aurèle Besse-Patin
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Medicine, University of Montreal, Montreal, Québec, Canada
| | - Mélissa Léveillé
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Medicine, University of Montreal, Montreal, Québec, Canada
| | - Daniel Oropeza
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Bich N Nguyen
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Québec, Canada; University of Montreal Health Network, Montreal, Québec, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada
| | - Jennifer L Estall
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Medicine, University of Montreal, Montreal, Québec, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
37
|
Susan-Resiga D, Girard E, Kiss RS, Essalmani R, Hamelin J, Asselin MC, Awan Z, Butkinaree C, Fleury A, Soldera A, Dory YL, Baass A, Seidah NG. The Proprotein Convertase Subtilisin/Kexin Type 9-resistant R410S Low Density Lipoprotein Receptor Mutation: A NOVEL MECHANISM CAUSING FAMILIAL HYPERCHOLESTEROLEMIA. J Biol Chem 2016; 292:1573-1590. [PMID: 27998977 DOI: 10.1074/jbc.m116.769430] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 12/20/2016] [Indexed: 12/22/2022] Open
Abstract
Familial hypercholesterolemia (FH) is characterized by severely elevated low density lipoprotein (LDL) cholesterol. Herein, we identified an FH patient presenting novel compound heterozygote mutations R410S and G592E of the LDL receptor (LDLR). The patient responded modestly to maximum rosuvastatin plus ezetimibe therapy, even in combination with a PCSK9 monoclonal antibody injection. Using cell biology and molecular dynamics simulations, we aimed to define the underlying mechanism(s) by which these LDLR mutations affect LDL metabolism and lead to hypercholesterolemia. Our data showed that the LDLR-G592E is a class 2b mutant, because it mostly failed to exit the endoplasmic reticulum and was degraded. Even though LDLR-R410S and LDLR-WT were similar in levels of cell surface and total receptor and bound equally well to LDL or extracellular PCSK9, the LDLR-R410S was resistant to exogenous PCSK9-mediated degradation in endosomes/lysosomes and showed reduced LDL internalization and degradation relative to LDLR-WT. Evidence is provided for a tighter association of LDL with LDLR-R410S at acidic pH, a reduced LDL delivery to late endosomes/lysosomes, and an increased release in the medium of the bound/internalized LDL, as compared with LDLR-WT. These data suggested that LDLR-R410S recycles loaded with its LDL-cargo. Our findings demonstrate that LDLR-R410S represents an LDLR loss-of-function through a novel class 8 FH-causing mechanism, thereby rationalizing the observed phenotype.
Collapse
Affiliation(s)
- Delia Susan-Resiga
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Emmanuelle Girard
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Robert Scott Kiss
- the Department of Medicine, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Rachid Essalmani
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Josée Hamelin
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Marie-Claude Asselin
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Zuhier Awan
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Chutikarn Butkinaree
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada
| | - Alexandre Fleury
- the Laboratory of Supramolecular Chemistry, Department of Chemistry, University of Sherbrooke, Sherbrooke, Quebec J1K 2R1, Canada
| | - Armand Soldera
- the Laboratory of Supramolecular Chemistry, Department of Chemistry, University of Sherbrooke, Sherbrooke, Quebec J1K 2R1, Canada
| | - Yves L Dory
- the Laboratory of Supramolecular Chemistry, Department of Chemistry, University of Sherbrooke, Sherbrooke, Quebec J1K 2R1, Canada
| | - Alexis Baass
- Nutrition, Metabolism, and Atherosclerosis Clinic, Institut de Recherches Cliniques de Montréal, affiliated with University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Nabil G Seidah
- From the Laboratory of Biochemical Neuroendocrinology, Montreal, Quebec H2W 1R7, Canada.
| |
Collapse
|
38
|
Seidah NG, Abifadel M, Prost S, Boileau C, Prat A. The Proprotein Convertases in Hypercholesterolemia and Cardiovascular Diseases: Emphasis on Proprotein Convertase Subtilisin/Kexin 9. Pharmacol Rev 2016; 69:33-52. [DOI: 10.1124/pr.116.012989] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
39
|
Lebeau P, Al-Hashimi A, Sood S, Lhoták Š, Yu P, Gyulay G, Paré G, Chen SRW, Trigatti B, Prat A, Seidah NG, Austin RC. Endoplasmic Reticulum Stress and Ca2+ Depletion Differentially Modulate the Sterol Regulatory Protein PCSK9 to Control Lipid Metabolism. J Biol Chem 2016; 292:1510-1523. [PMID: 27909053 DOI: 10.1074/jbc.m116.744235] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/08/2016] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence implicates endoplasmic reticulum (ER) stress as a mediator of impaired lipid metabolism, thereby contributing to fatty liver disease and atherosclerosis. Previous studies demonstrated that ER stress can activate the sterol regulatory element-binding protein-2 (SREBP2), an ER-localized transcription factor that directly up-regulates sterol regulatory genes, including PCSK9 Given that PCSK9 contributes to atherosclerosis by targeting low density lipoprotein (LDL) receptor (LDLR) degradation, this study investigates a novel mechanism by which ER stress plays a role in lipid metabolism by examining its ability to modulate PCSK9 expression. Herein, we demonstrate the existence of two independent effects of ER stress on PCSK9 expression and secretion. In cultured HuH7 and HepG2 cells, agents or conditions that cause ER Ca2+ depletion, including thapsigargin, induced SREBP2-dependent up-regulation of PCSK9 expression. In contrast, a significant reduction in the secreted form of PCSK9 protein was observed in the media from both thapsigargin- and tunicamycin (TM)-treated HuH7 cells, mouse primary hepatocytes, and in the plasma of TM-treated C57BL/6 mice. Furthermore, TM significantly increased hepatic LDLR expression and reduced plasma LDL concentrations in mice. Based on these findings, we propose a model in which ER Ca2+ depletion promotes the activation of SREBP2 and subsequent transcription of PCSK9. However, conditions that cause ER stress regardless of their ability to dysregulate ER Ca2+ inhibit PCSK9 secretion, thereby reducing PCSK9-mediated LDLR degradation and promoting LDLR-dependent hepatic cholesterol uptake. Taken together, our studies provide evidence that the retention of PCSK9 in the ER may serve as a potential strategy for lowering LDL cholesterol levels.
Collapse
Affiliation(s)
- Paul Lebeau
- From the Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Hamilton Healthcare and Hamilton Centre for Kidney Research, Hamilton, Ontario L8N 4A6
| | - Ali Al-Hashimi
- From the Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Hamilton Healthcare and Hamilton Centre for Kidney Research, Hamilton, Ontario L8N 4A6
| | - Sudesh Sood
- From the Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Hamilton Healthcare and Hamilton Centre for Kidney Research, Hamilton, Ontario L8N 4A6
| | - Šárka Lhoták
- From the Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Hamilton Healthcare and Hamilton Centre for Kidney Research, Hamilton, Ontario L8N 4A6
| | - Pei Yu
- the Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario L8L 2X2.,the Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8
| | - Gabriel Gyulay
- From the Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Hamilton Healthcare and Hamilton Centre for Kidney Research, Hamilton, Ontario L8N 4A6
| | - Guillaume Paré
- the Population Health Research Institute and the Departments of Medicine, Epidemiology and Pathology, McMaster University, Hamilton, Ontario L8L 2X2
| | - S R Wayne Chen
- the Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 2T9, and
| | - Bernardo Trigatti
- the Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario L8L 2X2.,the Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8
| | - Annik Prat
- the Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Nabil G Seidah
- the Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Richard C Austin
- From the Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Hamilton Healthcare and Hamilton Centre for Kidney Research, Hamilton, Ontario L8N 4A6, .,the Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario L8L 2X2
| |
Collapse
|
40
|
Lavoie JM. Dynamics of hepatic and intestinal cholesterol and bile acid pathways: The impact of the animal model of estrogen deficiency and exercise training. World J Hepatol 2016; 8:961-975. [PMID: 27621762 PMCID: PMC4990760 DOI: 10.4254/wjh.v8.i23.961] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/25/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023] Open
Abstract
Plasma cholesterol level is determined by a complex dynamics that involves transport lipoproteins which levels are tightly dependent on how the liver and the intestine regulate cholesterol and biliary acid metabolism. Regulation of cholesterol and biliary acids by the liver and the intestine is in turn coupled to a large array of enzymes and transporters that largely influence the inflow and the outflow of cholesterol and biliary acids through these organs. The activity of the key regulators of cholesterol and biliary acids may be influenced by several external factors such as pharmacological drugs and the nutritional status. In recent years, more information has been gathered about the impact of estrogens on regulation of cholesterol in the body. Exposure to high levels of estrogens has been reported to promote cholesterol gallstone formation and women are twice as likely as men to develop cholesterol gallstones. The impact of estrogen withdrawal, such as experienced by menopausal women, is therefore of importance and more information on how the absence of estrogens influence cholesterol regulation is started to come out, especially through the use of animal models. An interesting alternative to metabolic deterioration due to estrogen deficiency is exercise training. The present review is intended to summarize the present information that links key regulators of cholesterol and biliary acid pathways in liver and intestine to the absence of estrogens in an animal model and to discuss the potential role of exercise training as an alternative.
Collapse
|
41
|
Chański W, González-Prendes R, Castelló A, Jordana J, Manunza A, Quintanilla R, Amills M. An association analysis between a missense polymorphism at the pig PCSK9 gene and serum lipid and meat quality traits in Duroc pigs. Livest Sci 2016. [DOI: 10.1016/j.livsci.2016.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Combined Effects of Rosuvastatin and Exercise on Gene Expression of Key Molecules Involved in Cholesterol Metabolism in Ovariectomized Rats. PLoS One 2016; 11:e0159550. [PMID: 27442011 PMCID: PMC4956224 DOI: 10.1371/journal.pone.0159550] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study was to investigate the effects of three weeks of rosuvastatin (Ros) treatment alone and in combination with voluntary training (Tr) on expression of genes involved in cholesterol metabolism (LDLR, PCSK9, LRP-1, SREBP-2, IDOL, ACAT-2 and HMGCR) in the liver of eight week-old ovariectomized (Ovx) rats. Sprague Dawley rats were Ovx or sham-operated (Sham) and kept sedentary for 8 weeks under a standard diet. Thereafter, rats were transferred for three weeks in running wheel cages for Tr or kept sedentary (Sed) with or without Ros treatment (5mg/kg/day). Six groups were formed: Sham-Sed treated with saline (Sal) or Ros (Sham-Sed-Sal; Sham-Sed-Ros), Ovx-Sed treated with Sal or Ros (Ovx-Sed-Sal; Ovx-Sed-Ros), Ovx trained treated with Sal or Ros (Ovx-Tr-Sal; Ovx-Tr-Ros). Ovx-Sed-Sal rats depicted higher (P < 0.05) body weight, plasma total cholesterol (TC) and LDL-C, and liver TC content compared to Sham-Sed-Sal rats. In contrast, mRNA levels of liver PCSK9, LDLR, LRP-1 as well as plasma PCSK9 concentrations and protein levels of LRP-1 were reduced (P < 0.01) in Ovx-Sed-Sal compared to Sham-Sed-Sal rats. However, protein levels of LDLR increased (P < 0.05) in Ovx-Sed-Sal compared to Sham-Sed-Sal rats. Treatment of Ovx rats with Ros increased (P < 0.05) mRNA and protein levels of LRP-1 and PCSK9 but not mRNA levels of LDLR, while its protein abundance was reduced at the level of Sham rats. As a result, plasma LDL-C was not reduced. Exercise alone did not affect the expression of any of these markers in Ovx rats. Overall, Ros treatment corrected Ovx-induced decrease in gene expression of markers of cholesterol metabolism in liver of Ovx rats, but without reducing plasma LDL-C concentrations. Increased plasma PCSK9 levels could be responsible for the reduction of liver LDLR protein abundance and the absence of reduction of plasma LDL-C after Ros treatment.
Collapse
|
43
|
Weider E, Susan-Resiga D, Essalmani R, Hamelin J, Asselin MC, Nimesh S, Ashraf Y, Wycoff KL, Zhang J, Prat A, Seidah NG. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Single Domain Antibodies Are Potent Inhibitors of Low Density Lipoprotein Receptor Degradation. J Biol Chem 2016; 291:16659-71. [PMID: 27284008 DOI: 10.1074/jbc.m116.717736] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 01/14/2023] Open
Abstract
Single domain antibodies (sdAbs) correspond to the antigen-binding domains of camelid antibodies. They have the same antigen-binding properties and specificity as monoclonal antibodies (mAbs) but are easier and cheaper to produce. We report here the development of sdAbs targeting human PCSK9 (proprotein convertase subtilisin/kexin type 9) as an alternative to anti-PCSK9 mAbs. After immunizing a llama with human PCSK9, we selected four sdAbs that bind PCSK9 with a high affinity and produced them as fusion proteins with a mouse Fc. All four sdAb-Fcs recognize the C-terminal Cys-His-rich domain of PCSK9. We performed multiple cellular assays and demonstrated that the selected sdAbs efficiently blocked PCSK9-mediated low density lipoprotein receptor (LDLR) degradation in cell lines, in human hepatocytes, and in mouse primary hepatocytes. We further showed that the sdAb-Fcs do not affect binding of PCSK9 to the LDLR but rather block its induced cellular LDLR degradation. Pcsk9 knock-out mice expressing a human bacterial artificial chromosome (BAC) transgene were generated, resulting in plasma levels of ∼300 ng/ml human PCSK9. Mice were singly or doubly injected with the best sdAb-Fc and analyzed at day 4 or 11, respectively. After 4 days, mice exhibited a 32 and 44% decrease in the levels of total cholesterol and apolipoprotein B and ∼1.8-fold higher liver LDLR protein levels. At 11 days, the equivalent values were 24 and 46% and ∼2.3-fold higher LDLR proteins. These data constitute a proof-of-principle for the future usage of sdAbs as PCSK9-targeting drugs that can efficiently reduce LDL-cholesterol, and as tools to study the Cys-His-rich domain-dependent sorting the PCSK9-LDLR complex to lysosomes.
Collapse
Affiliation(s)
- Elodie Weider
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Delia Susan-Resiga
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Rachid Essalmani
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Josée Hamelin
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Marie-Claude Asselin
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Surendra Nimesh
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Yahya Ashraf
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Keith L Wycoff
- Planet Biotechnology Inc., Hayward, California 94545-2740, and
| | - Jianbing Zhang
- the Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario K1A 0R6, Canada
| | - Annik Prat
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada
| | - Nabil G Seidah
- From the Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec H2W 1R7, Canada,
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW High levels of LDL-cholesterol (LDL-C) are directly associated with devastating cardiovascular complications. Statins downregulate cholesterol synthesis and upregulate hepatic mRNA levels of LDL receptor (LDLR) and proprotein convertase subtilisin-kexin 9 (PCSK9), a validated enhancer of LDLR protein degradation. Herein, we summarize recent discoveries of the biological properties of PCSK9 in both health and disease states. RECENT FINDINGS PCSK9 downregulation of the LDLR protein likely explains the observed protective effect of the loss of PCSK9 in reducing lipoprotein(a) and incidence of septic shock. Injectable inhibitory PCSK9 monoclonal antibodies are now prescribed to hypercholesterolemic patients that do not reach target levels of LDL-C with available drugs. PCSK9 also reduces the levels of other receptors, for example, VLDL receptor (VLDLR), ApoER2, CD36, and CD81. The efficacy of the upregulation of LDLR and VLDLR cell surface levels in the absence of PCSK9 is both tissue and sex dependent. As LDLR, CD81, and VLDLR are hepatitis C receptors, PCSK9 may protect against certain viral infections. SUMMARY New functions of PCSK9 and other receptor targets are beginning to emerge to explain the observed changes in LDL-C and triglycerides. The effect of PCSK9 loss-of-function on glucose metabolism, factors that regulate the expression of PCSK9, and the roles of PCSK9 in other tissues, for example, intestine, kidney, and brain require further investigations.
Collapse
Affiliation(s)
- Nabil G Seidah
- Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with the Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
45
|
Levenson AE, Haas ME, Miao J, Brown RJ, de Ferranti SD, Muniyappa R, Biddinger SB. Effect of Leptin Replacement on PCSK9 in ob/ob Mice and Female Lipodystrophic Patients. Endocrinology 2016; 157:1421-9. [PMID: 26824363 PMCID: PMC4816729 DOI: 10.1210/en.2015-1624] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Leptin treatment has beneficial effects on plasma lipids in patients with lipodystrophy, but the underlying mechanism is unknown. Proprotein convertase subtilisin/kexin type 9 (PCSK9) decreases low-density lipoprotein (LDL) clearance, promotes hypercholesterolemia, and has recently emerged as a novel therapeutic target. To determine the effect of leptin on PCSK9, we treated male and female ob/ob mice with leptin for 4 days via sc osmotic pumps (∼24 μg/d). Leptin reduced body weight and food intake in all mice, but the effects of leptin on plasma PCSK9 and lipids differed markedly between the sexes. In male mice, leptin suppressed PCSK9 but had no effect on plasma triglycerides or cholesterol. In female mice, leptin suppressed plasma triglycerides and cholesterol but had no effect on plasma PCSK9. In parallel, we treated female lipodystrophic patients (8 females, ages 5-23 y) with sc metreleptin injections (∼4.4 mg/d) for 4-6 months. In this case, leptin reduced plasma PCSK9 by 26% (298 ± 109 vs 221 ± 102 ng/mL; n = 8; P = .008), and the change in PCSK9 was correlated with a decrease in LDL cholesterol (r(2) = 0.564, P = .03). In summary, in leptin-deficient ob/ob mice, the effects of leptin on PCSK9 and plasma lipids appeared to be independent of one another and strongly modified by sex. On the other hand, in lipodystrophic females, leptin treatment reduced plasma PCSK9 in parallel with LDL cholesterol.
Collapse
Affiliation(s)
- Amy E Levenson
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mary E Haas
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ji Miao
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Rebecca J Brown
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Sarah D de Ferranti
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ranganath Muniyappa
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Sudha B Biddinger
- Division of Endocrinology (A.E.L., M.E.H., J.M., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Diabetes, Endocrinology, and Obesity Branch (R.J.B., R.M.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Department of Cardiology (S.D.d.F.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|