1
|
Felix MN, Waerner T, Lakatos D, Reisinger B, Fischer S, Garidel P. Polysorbates degrading enzymes in biotherapeutics - a current status and future perspectives. Front Bioeng Biotechnol 2025; 12:1490276. [PMID: 39867473 PMCID: PMC11760601 DOI: 10.3389/fbioe.2024.1490276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/02/2024] [Indexed: 01/28/2025] Open
Abstract
Polysorbates, in particular polysorbate (PS) 20 and 80, are the most commonly used surfactants for stabilising biotherapeutics produced by biotechnological processes. PSs are derived from ethoxylated sorbitan (a derivative of sorbitol) esterified with fatty acids of varying chain length and degree of saturation. In the past, these surfactants have been reported to have specific liabilities. Chemical (oxidations and hydrolyses) and enzymatic degradations have been reported to affect the stability of PS in drug products. Specifically, the presence of trace amounts (sub-ppm) of certain host cell proteins (HCPs) can induce enzymatic PS degradation, which can lead to the release of free fatty acids during storage over time. Enzymatic polysorbate degradation may impair the functionality of the surfactant in stabilising therapeutic proteins, leading to the formation of visible and/or sub-visible particles in biopharmaceutical drug products. This review summarises the enzymes currently known to be involved in the degradation of polysorbate in mammalian biotechnological processes for therapeutic proteins. In recent years, advanced analytical methods have been developed to qualify and quantify the PS-degrading enzymes. Most of these assays are based on mass spectrometry with a preceding HCP enrichment approach. Efforts were made to measure the enzyme activity and correlate it with observed PS degradation. The impact on drug product quality attributes, including fatty acid solubility and phase separation, up to the formation of visible particles, and the potential induction of protein and protein/fatty acid mixed particles as well as the sensitivity of specific PS quality towards enzymatic degradation, was considered. Various drug substance (DS) mitigation strategies related to the occurrence of PS degrading enzymes are discussed as amongst them the generation of stable HCP knockout cell lines, which are also carefully analysed. The underlying opinion article reflects the undergoing discussions related to PS degrading enzymes and focusses on (i) impact on drug product, (ii) analytics for identification/quantification (characterisation) of the PS degrading enzymes, (iii) enzyme activity (iv) currently identified enzymes, and (v) potential mitigation strategies to avoid enzymatic PS degradation during DS manufacturing.
Collapse
Affiliation(s)
- Marius Nicolaus Felix
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Thomas Waerner
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Daniel Lakatos
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Bernd Reisinger
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Simon Fischer
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Patrick Garidel
- Pharmaceutical Development Biologicals, TIP, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| |
Collapse
|
2
|
Perera SD, Wang J, McIntyre AD, Hegele RA. Lipoprotein Lipase: Structure, Function, and Genetic Variation. Genes (Basel) 2025; 16:55. [PMID: 39858602 PMCID: PMC11764694 DOI: 10.3390/genes16010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Biallelic rare pathogenic loss-of-function (LOF) variants in lipoprotein lipase (LPL) cause familial chylomicronemia syndrome (FCS). Heterozygosity for these same variants is associated with a highly variable plasma triglyceride (TG) phenotype ranging from normal to severe hypertriglyceridemia (HTG), with longitudinal variation in phenotype severity seen often in a given carrier. Here, we provide an updated overview of genetic variation in LPL in the context of HTG, with a focus on disease-causing and/or disease-associated variants. We provide a curated list of 300 disease-causing variants discovered in LPL, as well as an exon-by-exon breakdown of the LPL gene and protein, highlighting the impact of variants and the various functional residues of domains of the LPL protein. We also provide a curated list of variants of unknown or uncertain significance, many of which may be upgraded to pathogenic/likely pathogenic classification should an additional case and/or segregation data be reported. Finally, we also review the association between benign/likely benign variants in LPL, many of which are common polymorphisms, and the TG phenotype.
Collapse
Affiliation(s)
- Shehan D. Perera
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
| | - Jian Wang
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
| | - Adam D. McIntyre
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
| | - Robert A. Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond Street North, London, ON N6A 5B7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond Street North, London, ON N6A 5B7, Canada
| |
Collapse
|
3
|
Risti R, Reimund M, Seeba NN, Lõokene A. A negatively charged cluster in the disordered acidic domain of GPIHBP1 provides selectivity in the interaction with lipoprotein lipase. Sci Rep 2024; 14:19639. [PMID: 39179764 PMCID: PMC11344153 DOI: 10.1038/s41598-024-70468-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
GPIHBP1 is a membrane protein of endothelial cells that transports lipoprotein lipase (LPL), the key enzyme in plasma triglyceride metabolism, from the interstitial space to its site of action on the capillary lumen. An intrinsically disordered highly negatively charged N-terminal domain of GPIHBP1 contributes to the interaction with LPL. In this work, we investigated whether the plethora of heparin-binding proteins with positively charged regions found in human plasma affect this interaction. We also wanted to know whether the role of the N-terminal domain is purely non-specific and supportive for the interaction between LPL and full-length GPIHBP1, or whether it participates in the specific recognition mechanism. Using surface plasmon resonance, affinity chromatography, and FRET, we were unable to identify any plasma component, besides LPL, that bound the N-terminus with detectable affinity or affected its interaction with LPL. By examining different synthetic peptides, we show that the high affinity of the LPL/N-terminal domain interaction is ensured by at least ten negatively charged residues, among which at least six must sequentially arranged. We conclude that the association of LPL with the N-terminal domain of GPIHBP1 is highly specific and human plasma does not contain components that significantly affect this complex.
Collapse
Affiliation(s)
- Robert Risti
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Mart Reimund
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Natjan-Naatan Seeba
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Aivar Lõokene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia.
| |
Collapse
|
4
|
Wang Q, Miao X, Hu M, Xu F, Tang G, He Y, Song Z, Zhao W, Niu X, Leng S. Nonlinear relationship between serum 25-hydroxyvitamin D and lipid profile in Chinese adults. Front Nutr 2024; 11:1388017. [PMID: 38933885 PMCID: PMC11199867 DOI: 10.3389/fnut.2024.1388017] [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: 02/19/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Background Previous studies on the liner associations between serum 25-hydroxyvitamin D [25(OH)D] levels and lipid profiles revealed ambiguous findings. The current study therefore tried to elucidate the possible non-linear associations between 25(OH)D and lipid profiles. Methods This study involved 8,516 adult participants (aged 18-74 years, males N = 3,750, females N = 4,766) recruited from the Dalian health management cohort (DHMC). The risk (OR) for specific dyslipidemias was estimated across the serum 25(OH)D levels and the cut-off value for serum 25(OH)D were determined by using logistic regression, restricted cubic spline, and piecewise linear regression methods, adjusted for age, sex, season, and ultraviolet index. Results In this study, a high prevalence of 25(OH)D deficiency was observed in the participants (65.05%). The level of 25(OH)D showed the inverse U-shaped correlations with the risks (ORs) of abnormal lipid profile, with inflection points observed at 23.7 ng/ml for hypercholesterolemia, 24.3 ng/ml for hypertriglyceridemia, 18.5 ng/ml for hyper-low-density lipoprotein cholesterolemia, 23.3 ng/ml for hypo-high-density lipoprotein cholesterolemia, 23.3 ng/ml for hyper-non-high-density lipoprotein cholesterol, and 24.3 ng/ml for high remnant cholesterol. The stratified analyses showed that the risk for most dyslipidemias related to deficiency of 25(OH)D was particularly increased among females aged 50-74 (except for hypertriglyceridemia, where the highest risk was among men aged 50-74 years), during winter/spring or under low/middle ultraviolet index environments. Conclusions Nonlinear inverse U-shaped associations were observed between 25(OH)D levels and abnormal lipid profile. The risk was particularly increased among females aged 50-74, during winter/spring period or under lower ultraviolet index environments. In vitamin D deficient subjects [25(OH)D <20 ng/ml], a positive association of serum vitamin D levels with the risk for dyslipidemia was observed, which needs a further.
Collapse
Affiliation(s)
- Qianqian Wang
- Health Management Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Xinlei Miao
- Health Management Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Manling Hu
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Fei Xu
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Guimin Tang
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Yangxuan He
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ziping Song
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Wan Zhao
- Health Management Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiangjun Niu
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Song Leng
- Health Management Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
5
|
Papadopoulou P, van der Pol R, van Hilten N, van Os WL, Pattipeiluhu R, Arias-Alpizar G, Knol RA, Noteborn W, Moradi MA, Ferraz MJ, Aerts JMFG, Sommerdijk N, Campbell F, Risselada HJ, Sevink GJA, Kros A. Phase-Separated Lipid-Based Nanoparticles: Selective Behavior at the Nano-Bio Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310872. [PMID: 37988682 DOI: 10.1002/adma.202310872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 11/23/2023]
Abstract
The membrane-protein interface on lipid-based nanoparticles influences their in vivo behavior. Better understanding may evolve current drug delivery methods toward effective targeted nanomedicine. Previously, the cell-selective accumulation of a liposome formulation in vivo is demonstrated, through the recognition of lipid phase-separation by triglyceride lipases. This exemplified how liposome morphology and composition can determine nanoparticle-protein interactions. Here, the lipase-induced compositional and morphological changes of phase-separated liposomes-which bear a lipid droplet in their bilayer- are investigated, and the mechanism upon which lipases recognize and bind to the particles is unravelled. The selective lipolytic degradation of the phase-separated lipid droplet is observed, while nanoparticle integrity remains intact. Next, the Tryptophan-rich loop of the lipase is identified as the region with which the enzymes bind to the particles. This preferential binding is due to lipid packing defects induced on the liposome surface by phase separation. In parallel, the existing knowledge that phase separation leads to in vivo selectivity, is utilized to generate phase-separated mRNA-LNPs that target cell-subsets in zebrafish embryos, with subsequent mRNA delivery and protein expression. Together, these findings can expand the current knowledge on selective nanoparticle-protein communications and in vivo behavior, aspects that will assist to gain control of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Panagiota Papadopoulou
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Rianne van der Pol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Niek van Hilten
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Winant L van Os
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Roy Pattipeiluhu
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Gabriela Arias-Alpizar
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Renzo Aron Knol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Willem Noteborn
- NeCEN, Leiden University, Einsteinweg 55, Leiden, 2333 AL, The Netherlands
| | - Mohammad-Amin Moradi
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Maria Joao Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | | | - Nico Sommerdijk
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
- Department of Medical BioSciences and Radboud Technology Center - Electron Microscopy, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Herre Jelger Risselada
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
- Department of Physics, Technical University Dortmund, 44221, Dortmund, Germany
| | - Geert Jan Agur Sevink
- Department of Biophysical Organic Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| |
Collapse
|
6
|
Alves M, Laranjeira F, Correia-da-Silva G. Understanding Hypertriglyceridemia: Integrating Genetic Insights. Genes (Basel) 2024; 15:190. [PMID: 38397180 PMCID: PMC10887881 DOI: 10.3390/genes15020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertriglyceridemia is an exceptionally complex metabolic disorder characterized by elevated plasma triglycerides associated with an increased risk of acute pancreatitis and cardiovascular diseases such as coronary artery disease. Its phenotype expression is widely heterogeneous and heavily influenced by conditions as obesity, alcohol consumption, or metabolic syndromes. Looking into the genetic underpinnings of hypertriglyceridemia, this review focuses on the genetic variants in LPL, APOA5, APOC2, GPIHBP1 and LMF1 triglyceride-regulating genes reportedly associated with abnormal genetic transcription and the translation of proteins participating in triglyceride-rich lipoprotein metabolism. Hypertriglyceridemia resulting from such genetic abnormalities can be categorized as monogenic or polygenic. Monogenic hypertriglyceridemia, also known as familial chylomicronemia syndrome, is caused by homozygous or compound heterozygous pathogenic variants in the five canonical genes. Polygenic hypertriglyceridemia, also known as multifactorial chylomicronemia syndrome in extreme cases of hypertriglyceridemia, is caused by heterozygous pathogenic genetic variants with variable penetrance affecting the canonical genes, and a set of common non-pathogenic genetic variants (polymorphisms, using the former nomenclature) with well-established association with elevated triglyceride levels. We further address recent progress in triglyceride-lowering treatments. Understanding the genetic basis of hypertriglyceridemia opens new translational opportunities in the scope of genetic screening and the development of novel therapies.
Collapse
Affiliation(s)
- Mara Alves
- Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Francisco Laranjeira
- CGM—Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António (CHUdSA), 4099-028 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-346 Porto, Portugal
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-600 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO Applied Molecular Biosciences Unit and Associate Laboratory i4HB—Institute for Health and Bioeconomy Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
7
|
Wheless A, Gunn KH, Neher SB. Macromolecular Interactions of Lipoprotein Lipase (LPL). Subcell Biochem 2024; 104:139-179. [PMID: 38963487 DOI: 10.1007/978-3-031-58843-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Lipoprotein lipase (LPL) is a critical enzyme in humans that provides fuel to peripheral tissues. LPL hydrolyzes triglycerides from the cores of lipoproteins that are circulating in plasma and interacts with receptors to mediate lipoprotein uptake, thus directing lipid distribution via catalytic and non-catalytic functions. Functional losses in LPL or any of its myriad of regulators alter lipid homeostasis and potentially affect the risk of developing cardiovascular disease-either increasing or decreasing the risk depending on the mutated protein. The extensive LPL regulatory network tunes LPL activity to allocate fatty acids according to the energetic needs of the organism and thus is nutritionally responsive and tissue dependent. Multiple pharmaceuticals in development manipulate or mimic these regulators, demonstrating their translational importance. Another facet of LPL biology is that the oligomeric state of the enzyme is also central to its regulation. Recent structural studies have solidified the idea that LPL is regulated not only by interactions with other binding partners but also by self-associations. Here, we review the complexities of the protein-protein and protein-lipid interactions that govern LPL structure and function.
Collapse
Affiliation(s)
- Anna Wheless
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn H Gunn
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Stony Brook University, Stony Brook, USA
| | - Saskia B Neher
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
Jiang S, Ren Z, Yang Y, Liu Q, Zhou S, Xiao Y. The GPIHBP1-LPL complex and its role in plasma triglyceride metabolism: Insights into chylomicronemia. Biomed Pharmacother 2023; 169:115874. [PMID: 37951027 DOI: 10.1016/j.biopha.2023.115874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/13/2023] Open
Abstract
GPIHBP1 is a protein found in the endothelial cells of capillaries that is anchored by glycosylphosphatidylinositol and binds to high-density lipoproteins. GPIHBP1 attaches to lipoprotein lipase (LPL), subsequently carrying the enzyme and anchoring it to the capillary lumen. Enabling lipid metabolism is essential for the marginalization of lipoproteins alongside capillaries. Studies underscore the significance of GPIHBP1 in transporting, stabilizing, and aiding in the marginalization of LPL. The intricate interplay between GPIHBP1 and LPL has provided novel insights into chylomicronemia in recent years. Mutations hindering the formation or reducing the efficiency of the GPIHBP1-LPL complex are central to the onset of chylomicronemia. This review delves into the structural nuances of the GPIHBP1-LPL interaction, the consequences of mutations in the complex leading to chylomicronemia, and cutting-edge advancements in chylomicronemia treatment.
Collapse
Affiliation(s)
- Shali Jiang
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Zhuoqun Ren
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Yutao Yang
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China.
| |
Collapse
|
9
|
Deng L, Kersten S, Stienstra R. Triacylglycerol uptake and handling by macrophages: From fatty acids to lipoproteins. Prog Lipid Res 2023; 92:101250. [PMID: 37619883 DOI: 10.1016/j.plipres.2023.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Macrophages are essential innate immune cells and form our first line of immune defense. Also known as professional phagocytes, macrophages interact and take up various particles, including lipids. Defective lipid handling can drive excessive lipid accumulation leading to foam cell formation, a key feature of various cardiometabolic conditions such as atherosclerosis, non-alcoholic fatty liver disease, and obesity. At the same time, intracellular lipid storage and foam cell formation can also be viewed as a protective and anti-lipotoxic mechanism against a lipid-rich environment and associated elevated lipid uptake. Traditionally, foam cell formation has primarily been linked to cholesterol uptake via native and modified low-density lipoproteins. However, other lipids, including non-esterified fatty acids and triacylglycerol (TAG)-rich lipoproteins (very low-density lipoproteins and chylomicrons), can also interact with macrophages. Recent studies have identified multiple pathways mediating TAG uptake and processing by macrophages, including endocytosis and receptor/transporter-mediated internalization and transport. This review will present the current knowledge of how macrophages take up different lipids and lipoprotein particles and address how TAG-rich lipoproteins are processed intracellularly. Understanding how macrophages take up and process different lipid species such as TAG is necessary to design future therapeutic interventions to correct excessive lipid accumulation and associated co-morbidities.
Collapse
Affiliation(s)
- Lei Deng
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
10
|
Silbernagel G, Chen YQ, Rief M, Kleber ME, Hoffmann MM, Stojakovic T, Stang A, Sarzynski MA, Bouchard C, März W, Qian YW, Scharnagl H, Konrad RJ. Inverse association between apolipoprotein C-II and cardiovascular mortality: role of lipoprotein lipase activity modulation. Eur Heart J 2023:7156982. [PMID: 37155355 DOI: 10.1093/eurheartj/ehad261] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 02/20/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
AIMS Apolipoprotein C-II (ApoC-II) is thought to activate lipoprotein lipase (LPL) and is therefore a possible target for treating hypertriglyceridemia. Its relationship with cardiovascular risk has not been investigated in large-scale epidemiologic studies, particularly allowing for apolipoprotein C-III (ApoC-III), an LPL antagonist. Furthermore, the exact mechanism of ApoC-II-mediated LPL activation is unclear. METHODS AND RESULTS ApoC-II was measured in 3141 LURIC participants of which 590 died from cardiovascular diseases during a median (inter-quartile range) follow-up of 9.9 (8.7-10.7) years. Apolipoprotein C-II-mediated activation of the glycosylphosphatidylinositol high-density lipoprotein binding protein 1 (GPIHBP1)-LPL complex was studied using enzymatic activity assays with fluorometric lipase and very low-density lipoprotein (VLDL) substrates. The mean ApoC-II concentration was 4.5 (2.4) mg/dL. The relationship of ApoC-II quintiles with cardiovascular mortality exhibited a trend toward an inverse J-shape, with the highest risk in the first (lowest) quintile and lowest risk in the middle quintile. Compared with the first quintile, all other quintiles were associated with decreased cardiovascular mortality after multivariate adjustments including ApoC-III as a covariate (all P < 0.05). In experiments using fluorometric substrate-based lipase assays, there was a bell-shaped relationship for the effect of ApoC-II on GPIHBP1-LPL activity when exogenous ApoC-II was added. In ApoC-II-containing VLDL substrate-based lipase assays, GPIHBP1-LPL enzymatic activity was almost completely blocked by a neutralizing anti-ApoC-II antibody. CONCLUSION The present epidemiologic data suggest that increasing low circulating ApoC-II levels may reduce cardiovascular risk. This conclusion is supported by the observation that optimal ApoC-II concentrations are required for maximal GPIHBP1-LPL enzymatic activity.
Collapse
Affiliation(s)
- Günther Silbernagel
- Division of Vascular Medicine, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Auenbruggerplatz 15 Graz, Austria
| | - Yan Q Chen
- Lilly Research Laboratories, Eli Lilly and Company, 893 Delaware St, Indianapolis, IN 46225, USA
| | - Martin Rief
- Division of General Anaesthesiology, Emergency and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Auenbruggerplatz 5 Graz, Austria
| | - Marcus E Kleber
- Department of Internal Medicine 5 (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Michael M Hoffmann
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, 8036 Graz, Auenbruggerplatz 15 Graz, Austria
| | - Andreas Stang
- Institut für Medizinische Informatik, Biometrie und Epidemiologie, Universitätsklinikum Essen, Hufelandstraße 55, 45122 Essen, Germany
- School of Public Health, Department of Epidemiology, Boston University, 715 Albany St, Boston, MA 02118, USA
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina, 921 Assembly St, Columbia, SC 29201, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA
| | - Winfried März
- Department of Internal Medicine 5 (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Auenbruggerpl. 15, Graz, Austria
- Synlab Academy, Synlab Holding Germany GmbH, P5, 7 (Street) 68161 Mannheim, Germany
| | - Yue-Wei Qian
- Lilly Research Laboratories, Eli Lilly and Company, 893 Delaware St, Indianapolis, IN 46225, USA
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Auenbruggerpl. 15, Graz, Austria
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, 893 Delaware St, Indianapolis, IN 46225, USA
| |
Collapse
|
11
|
Gunn KH, Neher SB. Structure of dimeric lipoprotein lipase reveals a pore adjacent to the active site. Nat Commun 2023; 14:2569. [PMID: 37142573 PMCID: PMC10160067 DOI: 10.1038/s41467-023-38243-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/24/2023] [Indexed: 05/06/2023] Open
Abstract
Lipoprotein lipase (LPL) hydrolyzes triglycerides from circulating lipoproteins, releasing free fatty acids. Active LPL is needed to prevent hypertriglyceridemia, which is a risk factor for cardiovascular disease (CVD). Using cryogenic electron microscopy (cryoEM), we determined the structure of an active LPL dimer at 3.9 Å resolution. This structure reveals an open hydrophobic pore adjacent to the active site residues. Using modeling, we demonstrate that this pore can accommodate an acyl chain from a triglyceride. Known LPL mutations that lead to hypertriglyceridemia localize to the end of the pore and cause defective substrate hydrolysis. The pore may provide additional substrate specificity and/or allow unidirectional acyl chain release from LPL. This structure also revises previous models on how LPL dimerizes, revealing a C-terminal to C-terminal interface. We hypothesize that this active C-terminal to C-terminal conformation is adopted by LPL when associated with lipoproteins in capillaries.
Collapse
Affiliation(s)
- Kathryn H Gunn
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Saskia B Neher
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
12
|
Risti R, Gunn KH, Hiis-Hommuk K, Seeba NN, Karimi H, Villo L, Vendelin M, Neher SB, Lõokene A. Combined action of albumin and heparin regulates lipoprotein lipase oligomerization, stability, and ligand interactions. PLoS One 2023; 18:e0283358. [PMID: 37043509 PMCID: PMC10096250 DOI: 10.1371/journal.pone.0283358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/07/2023] [Indexed: 04/13/2023] Open
Abstract
Lipoprotein lipase (LPL), a crucial enzyme in the intravascular hydrolysis of triglyceride-rich lipoproteins, is a potential drug target for the treatment of hypertriglyceridemia. The activity and stability of LPL are influenced by a complex ligand network. Previous studies performed in dilute solutions suggest that LPL can appear in various oligomeric states. However, it was not known how the physiological environment, that is blood plasma, affects the action of LPL. In the current study, we demonstrate that albumin, the major protein component in blood plasma, has a significant impact on LPL stability, oligomerization, and ligand interactions. The effects induced by albumin could not solely be reproduced by the macromolecular crowding effect. Stabilization, isothermal titration calorimetry, and surface plasmon resonance studies revealed that albumin binds to LPL with affinity sufficient to form a complex in both the interstitial space and the capillaries. Negative stain transmission electron microscopy and raster image correlation spectroscopy showed that albumin, like heparin, induced reversible oligomerization of LPL. However, the albumin induced oligomers were structurally different from heparin-induced filament-like LPL oligomers. An intriguing observation was that no oligomers of either type were formed in the simultaneous presence of albumin and heparin. Our data also suggested that the oligomer formation protected LPL from the inactivation by its physiological regulator angiopoietin-like protein 4. The concentration of LPL and its environment could influence whether LPL follows irreversible inactivation and aggregation or reversible LPL oligomer formation, which might affect interactions with various ligands and drugs. In conclusion, the interplay between albumin and heparin could provide a mechanism for ensuring the dissociation of heparan sulfate-bound LPL oligomers into active LPL upon secretion into the interstitial space.
Collapse
Affiliation(s)
- Robert Risti
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Kathryn H. Gunn
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kristofer Hiis-Hommuk
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Natjan-Naatan Seeba
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Hamed Karimi
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Ly Villo
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Tallinn, Estonia
| | - Saskia B. Neher
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Aivar Lõokene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
13
|
Gunn KH, Neher SB. Structure of Dimeric Lipoprotein Lipase Reveals a Pore for Hydrolysis of Acyl Chains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.21.533650. [PMID: 36993689 PMCID: PMC10055231 DOI: 10.1101/2023.03.21.533650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Lipoprotein lipase (LPL) hydrolyzes triglycerides from circulating lipoproteins, releasing free fatty acids. Active LPL is needed to prevent hypertriglyceridemia, which is a risk factor for cardiovascular disease (CVD). Using cryogenic electron microscopy (cryoEM), we determined the structure of an active LPL dimer at 3.9 Ã… resolution. This is the first structure of a mammalian lipase with an open, hydrophobic pore adjacent to the active site. We demonstrate that the pore can accommodate an acyl chain from a triglyceride. Previously, it was thought that an open lipase conformation was defined by a displaced lid peptide, exposing the hydrophobic pocket surrounding the active site. With these previous models after the lid opened, the substrate would enter the active site, be hydrolyzed and then released in a bidirectional manner. It was assumed that the hydrophobic pocket provided the only ligand selectivity. Based on our structure, we propose a new model for lipid hydrolysis, in which the free fatty acid product travels unidirectionally through the active site pore, entering and exiting opposite sides of the protein. By this new model, the hydrophobic pore provides additional substrate specificity and provides insight into how LPL mutations in the active site pore may negatively impact LPL activity, leading to chylomicronemia. Structural similarity of LPL to other human lipases suggests that this unidirectional mechanism could be conserved but has not been observed due to the difficulty of studying lipase structure in the presence of an activating substrate. We hypothesize that the air/water interface formed during creation of samples for cryoEM triggered interfacial activation, allowing us to capture, for the first time, a fully open state of a mammalian lipase. Our new structure also revises previous models on how LPL dimerizes, revealing an unexpected C-terminal to C-terminal interface. The elucidation of a dimeric LPL structure highlights the oligomeric diversity of LPL, as now LPL homodimer, heterodimer, and helical filament structures have been elucidated. This diversity of oligomerization may provide a form of regulation as LPL travels from secretory vesicles in the cell, to the capillary, and eventually to the liver for lipoprotein remnant uptake. We hypothesize that LPL dimerizes in this active C-terminal to C-terminal conformation when associated with mobile lipoproteins in the capillary.
Collapse
|
14
|
Sun X, Yu Z, Liang C, Xie S, Wen J, Wang H, Wang J, Yang Y, Han R. Developmental changes in proteins of casein micelles in goat milk using data-independent acquisition-based proteomics methods during the lactation cycle. J Dairy Sci 2022; 106:47-60. [PMID: 36333141 DOI: 10.3168/jds.2022-22032] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/12/2022] [Indexed: 11/05/2022]
Abstract
Casein micelles (CM) play an important role in milk secretion, stability, and processing. The composition and content of milk proteins are affected by physiological factors, which have been widely investigated. However, the variation in CM proteins in goat milk throughout the lactation cycle has yet to be fully clarified. In the current study, milk samples were collected at d 1, 3, 30, 90, 150, and 240 of lactation from 15 dairy goats. The size of CM was determined using laser light scattering, and CM proteins were separated, digested, and identified using data-independent acquisition (DIA) and data-dependent acquisition (DDA)-based proteomics approaches. According to clustering and principal component analysis, protein profiles identified using DIA were similar to those identified using the DDA approach. Significant differences in the abundance of 115 proteins during the lactation cycle were identified using the DIA approach. Developmental changes in the CM proteome corresponding to lactation stages were revealed: levels of lecithin cholesterol acyltransferase, folate receptor α, and prominin 2 increased from 1 to 240 d, whereas levels of growth/differentiation factor 8, peptidoglycan-recognition protein, and 45 kDa calcium-binding protein decreased in the same period. In addition, lipoprotein lipase, glycoprotein IIIb, and α-lactalbumin levels increased from 1 to 90 d and then decreased to 240 d, which is consistent with the change in CM size. Protein-protein interaction analysis showed that fibronectin, albumin, and apolipoprotein E interacted more with other proteins at the central node. These findings indicate that changes in the CM proteome during lactation could be related to requirements of newborn development, as well as mammary gland development, and may thus contribute to elucidating the physical and chemical properties of CM.
Collapse
Affiliation(s)
- Xueheng Sun
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Zhongna Yu
- Haidu College, Qingdao Agricultural University, Laiyang 265200, Shandong, China
| | - Chuozi Liang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Shubin Xie
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Jing Wen
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Hexiang Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Jun Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Yongxin Yang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Rongwei Han
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China.
| |
Collapse
|
15
|
Wen Y, Chen YQ, Konrad RJ. The Regulation of Triacylglycerol Metabolism and Lipoprotein Lipase Activity. Adv Biol (Weinh) 2022; 6:e2200093. [PMID: 35676229 DOI: 10.1002/adbi.202200093] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/03/2022] [Indexed: 01/28/2023]
Abstract
Triacylglycerol (TG) metabolism is tightly regulated to maintain a pool of TG within circulating lipoproteins that can be hydrolyzed in a tissue-specific manner by lipoprotein lipase (LPL) to enable the delivery of fatty acids to adipose or oxidative tissues as needed. Elevated serum TG concentrations, which result from a deficiency of LPL activity or, more commonly, an imbalance in the regulation of tissue-specific LPL activities, have been associated with an increased risk of atherosclerotic cardiovascular disease through multiple studies. Among the most critical LPL regulators are the angiopoietin-like (ANGPTL) proteins ANGPTL3, ANGPTL4, and ANGPTL8, and a number of different apolipoproteins including apolipoprotein A5 (ApoA5), apolipoprotein C2 (ApoC2), and apolipoprotein C3 (ApoC3). These ANGPTLs and apolipoproteins work together to orchestrate LPL activity and therefore play pivotal roles in TG partitioning, hydrolysis, and utilization. This review summarizes the mechanisms of action, epidemiological findings, and genetic data most relevant to these ANGPTLs and apolipoproteins. The interplay between these important regulators of TG metabolism in both fasted and fed states is highlighted with a holistic view toward understanding key concepts and interactions. Strategies for developing safe and effective therapeutics to reduce circulating TG by selectively targeting these ANGPTLs and apolipoproteins are also discussed.
Collapse
Affiliation(s)
- Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Yan Q Chen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| |
Collapse
|
16
|
Dijk W, Di Filippo M, Kooijman S, van Eenige R, Rimbert A, Caillaud A, Thedrez A, Arnaud L, Pronk A, Garçon D, Sotin T, Lindenbaum P, Ozcariz Garcia E, Pais de Barros JP, Duvillard L, Si-Tayeb K, Amigo N, Le Questel JY, Rensen PC, Le May C, Moulin P, Cariou B. Identification of a Gain-of-Function LIPC Variant as a Novel Cause of Familial Combined Hypocholesterolemia. Circulation 2022; 146:724-739. [PMID: 35899625 PMCID: PMC9439636 DOI: 10.1161/circulationaha.121.057978] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease is the main cause of mortality worldwide and is strongly influenced by circulating low-density lipoprotein (LDL) cholesterol levels. Only a few genes causally related to plasma LDL cholesterol levels have been identified so far, and only 1 gene, ANGPTL3, has been causally related to combined hypocholesterolemia. Here, our aim was to elucidate the genetic origin of an unexplained combined hypocholesterolemia inherited in 4 generations of a French family. METHODS Using next-generation sequencing, we identified a novel dominant rare variant in the LIPC gene, encoding for hepatic lipase, which cosegregates with the phenotype. We characterized the impact of this LIPC-E97G variant on circulating lipid and lipoprotein levels in family members using nuclear magnetic resonance-based lipoprotein profiling and lipidomics. To uncover the mechanisms underlying the combined hypocholesterolemia, we used protein homology modeling, measured triglyceride lipase and phospholipase activities in cell culture, and studied the phenotype of APOE*3.Leiden.CETP mice after LIPC-E97G overexpression. RESULTS Family members carrying the LIPC-E97G variant had very low circulating levels of LDL cholesterol and high-density lipoprotein cholesterol, LDL particle numbers, and phospholipids. The lysophospholipids/phospholipids ratio was increased in plasma of LIPC-E97G carriers, suggestive of an increased lipolytic activity on phospholipids. In vitro and in vivo studies confirmed that the LIPC-E97G variant specifically increases the phospholipase activity of hepatic lipase through modification of an evolutionarily conserved motif that determines substrate access to the hepatic lipase catalytic site. Mice overexpressing human LIPC-E97G recapitulated the combined hypocholesterolemic phenotype of the family and demonstrated that the increased phospholipase activity promotes catabolism of triglyceride-rich lipoproteins by different extrahepatic tissues but not the liver. CONCLUSIONS We identified and characterized a novel rare variant in the LIPC gene in a family who presents with dominant familial combined hypocholesterolemia. This gain-of-function variant makes LIPC the second identified gene, after ANGPTL3, causally involved in familial combined hypocholesterolemia. Our mechanistic data highlight the critical role of hepatic lipase phospholipase activity in LDL cholesterol homeostasis and suggest a new LDL clearance mechanism.
Collapse
Affiliation(s)
- Wieneke Dijk
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Mathilde Di Filippo
- UF Dyslipidémies, Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiStites, Hospices Civils de Lyon, Bron, France (M.D.F.).,CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France (M.D.F., P.M.)
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (S.K., R.v.E., A.P., P.C.N.R.)
| | - Robin van Eenige
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (S.K., R.v.E., A.P., P.C.N.R.)
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Amandine Caillaud
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Aurélie Thedrez
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Lucie Arnaud
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Amanda Pronk
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (S.K., R.v.E., A.P., P.C.N.R.)
| | - Damien Garçon
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Thibaud Sotin
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Pierre Lindenbaum
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | | | - Jean-Paul Pais de Barros
- Lipidomic Platform, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France (J.-P.P.d.B.)
| | - Laurence Duvillard
- University of Burgundy, INSERM LNC UMR1231, Dijon, France (L.D.).,CHU Dijon, Department of Biochemistry, Dijon, France (L.D.)
| | - Karim Si-Tayeb
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Nuria Amigo
- Biosfer Teslab, Reus, Spain (E.O.G., N.A.).,Department of Basic Medical Sciences, Rovira I Virgili University, IISPV, CIBERDEM, Reus, Spain (N.A.)
| | | | - Patrick C.N. Rensen
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (S.K., R.v.E., A.P., P.C.N.R.)
| | - Cédric Le May
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| | - Philippe Moulin
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France (M.D.F., P.M.).,Fédération d’endocrinologie, maladies métaboliques, diabète et nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France (P.M.)
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, France (W.D., A.R., A.C., A.T., L.A., D.G., T.S., P.L., K.S.-T., C.L.M., B.C.)
| |
Collapse
|
17
|
Young SG, Song W, Yang Y, Birrane G, Jiang H, Beigneux AP, Ploug M, Fong LG. A protein of capillary endothelial cells, GPIHBP1, is crucial for plasma triglyceride metabolism. Proc Natl Acad Sci U S A 2022; 119:e2211136119. [PMID: 36037340 PMCID: PMC9457329 DOI: 10.1073/pnas.2211136119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
GPIHBP1, a protein of capillary endothelial cells (ECs), is a crucial partner for lipoprotein lipase (LPL) in the lipolytic processing of triglyceride-rich lipoproteins. GPIHBP1, which contains a three-fingered cysteine-rich LU (Ly6/uPAR) domain and an intrinsically disordered acidic domain (AD), captures LPL from within the interstitial spaces (where it is secreted by parenchymal cells) and shuttles it across ECs to the capillary lumen. Without GPIHBP1, LPL remains stranded within the interstitial spaces, causing severe hypertriglyceridemia (chylomicronemia). Biophysical studies revealed that GPIHBP1 stabilizes LPL structure and preserves LPL activity. That discovery was the key to crystallizing the GPIHBP1-LPL complex. The crystal structure revealed that GPIHBP1's LU domain binds, largely by hydrophobic contacts, to LPL's C-terminal lipid-binding domain and that the AD is positioned to project across and interact, by electrostatic forces, with a large basic patch spanning LPL's lipid-binding and catalytic domains. We uncovered three functions for GPIHBP1's AD. First, it accelerates the kinetics of LPL binding. Second, it preserves LPL activity by inhibiting unfolding of LPL's catalytic domain. Third, by sheathing LPL's basic patch, the AD makes it possible for LPL to move across ECs to the capillary lumen. Without the AD, GPIHBP1-bound LPL is trapped by persistent interactions between LPL and negatively charged heparan sulfate proteoglycans (HSPGs) on the abluminal surface of ECs. The AD interrupts the HSPG interactions, freeing LPL-GPIHBP1 complexes to move across ECs to the capillary lumen. GPIHBP1 is medically important; GPIHBP1 mutations cause lifelong chylomicronemia, and GPIHBP1 autoantibodies cause some acquired cases of chylomicronemia.
Collapse
Affiliation(s)
- Stephen G. Young
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Wenxin Song
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Ye Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Gabriel Birrane
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Anne P. Beigneux
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen 2200N, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Loren G. Fong
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| |
Collapse
|
18
|
Sustar U, Groselj U, Khan SA, Shafi S, Khan I, Kovac J, Bizjan BJ, Battelino T, Sadiq F. A homozygous variant in the GPIHBP1 gene in a child with severe hypertriglyceridemia and a systematic literature review. Front Genet 2022; 13:983283. [PMID: 36051701 PMCID: PMC9424485 DOI: 10.3389/fgene.2022.983283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Due to nonspecific symptoms, rare dyslipidaemias are frequently misdiagnosed, overlooked, and undertreated, leading to increased risk for severe cardiovascular disease, pancreatitis and/or multiple organ failures before diagnosis. Better guidelines for the recognition and early diagnosis of rare dyslipidaemias are urgently required. Methods: Genomic DNA was isolated from blood samples of a Pakistani paediatric patient with hypertriglyceridemia, and from his parents and siblings. Next-generation sequencing (NGS) was performed, and an expanded dyslipidaemia panel was employed for genetic analysis. Results: The NGS revealed the presence of a homozygous missense pathogenic variant c.230G>A (NM_178172.6) in exon 3 of the GPIHBP1 (glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1) gene resulting in amino acid change p.Cys77Tyr (NP_835466.2). The patient was 5.5 years old at the time of genetic diagnosis. The maximal total cholesterol and triglyceride levels were measured at the age of 10 months (850.7 mg/dl, 22.0 mmol/L and 5,137 mg/dl, 58.0 mmol/L, respectively). The patient had cholesterol deposits at the hard palate, eruptive xanthomas, lethargy, poor appetite, and mild splenomegaly. Both parents and sister were heterozygous for the familial variant in the GPIHBP1 gene. Moreover, in the systematic review, we present 62 patients with pathogenic variants in the GPIHBP1 gene and clinical findings, associated with hyperlipoproteinemia. Conclusion: In a child with severe hypertriglyceridemia, we identified a pathogenic variant in the GPIHBP1 gene causing hyperlipoproteinemia (type 1D). In cases of severe elevations of plasma cholesterol and/or triglycerides genetic testing for rare dyslipidaemias should be performed as soon as possible for optimal therapy and patient management.
Collapse
Affiliation(s)
- Ursa Sustar
- Department of Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urh Groselj
- Department of Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Urh Groselj, ; Fouzia Sadiq,
| | - Sabeen Abid Khan
- Department of Paediatrics, Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Saeed Shafi
- Department of Anatomy, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Iqbal Khan
- Department of Vascular Surgery, Shifa International Hospital, Islamabad, Pakistan
- Department of Vascular Surgery, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Jernej Kovac
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Barbara Jenko Bizjan
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fouzia Sadiq
- Directorate of Research, Shifa Tameer-e-Millat University, Islamabad, Pakistan
- *Correspondence: Urh Groselj, ; Fouzia Sadiq,
| |
Collapse
|
19
|
Stewart V, Ronald PC. Sulfotyrosine residues: interaction specificity determinants for extracellular protein-protein interactions. J Biol Chem 2022; 298:102232. [PMID: 35798140 PMCID: PMC9372746 DOI: 10.1016/j.jbc.2022.102232] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/28/2022] Open
Abstract
Tyrosine sulfation, a post-translational modification, can determine and often enhance protein–protein interaction specificity. Sulfotyrosyl residues (sTyrs) are formed by the enzyme tyrosyl-protein sulfotransferase during protein maturation in the Golgi apparatus and most often occur singly or as a cluster within a six-residue span. With both negative charge and aromatic character, sTyr facilitates numerous atomic contacts as visualized in binding interface structural models, thus there is no discernible binding site consensus. Found exclusively in secreted proteins, in this review, we discuss the four broad sequence contexts in which sTyr has been observed: first, a solitary sTyr has been shown to be critical for diverse high-affinity interactions, such as between peptide hormones and their receptors, in both plants and animals. Second, sTyr clusters within structurally flexible anionic segments are essential for a variety of cellular processes, including coreceptor binding to the HIV-1 envelope spike protein during virus entry, chemokine interactions with receptors, and leukocyte rolling cell adhesion. Third, a subcategory of sTyr clusters is found in conserved acidic sequences termed hirudin-like motifs that enable proteins to interact with thrombin; consequently, many proven and potential therapeutic proteins derived from blood-consuming invertebrates depend on sTyrs for their activity. Finally, several proteins that interact with collagen or similar proteins contain one or more sTyrs within an acidic residue array. Refined methods to direct sTyr incorporation in peptides synthesized both in vitro and in vivo, together with continued advances in mass spectrometry and affinity detection, promise to accelerate discoveries of sTyr occurrence and function.
Collapse
Affiliation(s)
- Valley Stewart
- Department of Microbiology & Molecular Genetics, University of California, Davis, USA.
| | - Pamela C Ronald
- Department of Plant Pathology, University of California, Davis, USA; Genome Center, University of California, Davis, USA.
| |
Collapse
|
20
|
Heo Y, Lee I, Moon S, Yun JH, Kim EY, Park SY, Park JH, Kim WT, Lee W. Crystal Structures of the Plant Phospholipase A1 Proteins Reveal a Unique Dimerization Domain. Molecules 2022; 27:molecules27072317. [PMID: 35408716 PMCID: PMC9000616 DOI: 10.3390/molecules27072317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Phospholipase is an enzyme that hydrolyzes various phospholipid substrates at specific ester bonds and plays important roles such as membrane remodeling, as digestive enzymes, and the regulation of cellular mechanism. Phospholipase proteins are divided into following the four major groups according to the ester bonds they cleave off: phospholipase A1 (PLA1), phospholipase A2 (PLA2), phospholipase C (PLC), and phospholipase D (PLD). Among the four phospholipase groups, PLA1 has been less studied than the other phospholipases. Here, we report the first molecular structures of plant PLA1s: AtDSEL and CaPLA1 derived from Arabidopsis thaliana and Capsicum annuum, respectively. AtDSEL and CaPLA1 are novel PLA1s in that they form homodimers since PLAs are generally in the form of a monomer. The dimerization domain at the C-terminal of the AtDSEL and CaPLA1 makes hydrophobic interactions between each monomer, respectively. The C-terminal domain is also present in PLA1s of other plants, but not in PLAs of mammals and fungi. An activity assay of AtDSEL toward various lipid substrates demonstrates that AtDSEL is specialized for the cleavage of sn-1 acyl chains. This report reveals a new domain that exists only in plant PLA1s and suggests that the domain is essential for homodimerization.
Collapse
Affiliation(s)
- Yunseok Heo
- Structural Biochemistry & Molecular Biophysics Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea; (Y.H.); (I.L.); (S.M.); (J.-H.Y.); (J.-H.P.)
| | - Inhwan Lee
- Structural Biochemistry & Molecular Biophysics Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea; (Y.H.); (I.L.); (S.M.); (J.-H.Y.); (J.-H.P.)
| | - Sunjin Moon
- Structural Biochemistry & Molecular Biophysics Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea; (Y.H.); (I.L.); (S.M.); (J.-H.Y.); (J.-H.P.)
| | - Ji-Hye Yun
- Structural Biochemistry & Molecular Biophysics Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea; (Y.H.); (I.L.); (S.M.); (J.-H.Y.); (J.-H.P.)
- PCG-Biotech, Ltd., 508 KBIZ DMC Tower, Sangam-ro, Seoul 03929, Korea
| | - Eun Yu Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea;
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama 230-0045, Japan;
| | - Jae-Hyun Park
- Structural Biochemistry & Molecular Biophysics Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea; (Y.H.); (I.L.); (S.M.); (J.-H.Y.); (J.-H.P.)
| | - Woo Taek Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea;
- Correspondence: (W.T.K.); (W.L.)
| | - Weontae Lee
- Structural Biochemistry & Molecular Biophysics Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea; (Y.H.); (I.L.); (S.M.); (J.-H.Y.); (J.-H.P.)
- PCG-Biotech, Ltd., 508 KBIZ DMC Tower, Sangam-ro, Seoul 03929, Korea
- Correspondence: (W.T.K.); (W.L.)
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Lipoprotein lipase (LPL) is the rate-limiting enzyme for intravascular processing of circulating triglyceride-rich lipoproteins (TRLs). One emerging strategy for therapeutic lowering of plasma triglyceride levels aims at increasing the longevity of LPL activity by attenuating its inhibition from angiopoietin-like proteins (ANGPTL) 3, 4 and 8. This mini-review focuses on recent insights into the molecular mechanisms underpinning the regulation of LPL activity in the intravascular unit by ANGPTLs with special emphasis on ANGPTL4. RECENT FINDINGS Our knowledge on the molecular interplays between LPL, its endothelial transporter GPIHBP1, and its inhibitor(s) ANGPTL4, ANGPTL3 and ANGPTL8 have advanced considerably in the last 2 years and provides an outlined on how these proteins regulate the activity and compartmentalization of LPL. A decisive determinant instigating this control is the inherent protein instability of LPL at normal body temperature, a property that is reciprocally impacted by the binding of GPIHBP1 and ANGPTLs. Additional layers in this complex LPL regulation is provided by the different modulation of ANGPTL4 and ANGPTL3 activities by ANGPTL8 and the inhibition of ANGPTL3/8 complexes by apolipoprotein A5 (APOA5). SUMMARY Posttranslational regulation of LPL activity in the intravascular space is essential for the differential partitioning of TRLs across tissues and their lipolytic processing in response to nutritional cues.
Collapse
Affiliation(s)
- Michael Ploug
- Finsen Laboratory, Rigshospitalet
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Wang S, Cheng Y, Shi Y, Zhao W, Gao L, Fang L, Jin X, Han X, Sun Q, Li G, Zhao J, Xu C. Identification and Characterization of Two Novel Compounds: Heterozygous Variants of Lipoprotein Lipase in Two Pedigrees With Type I Hyperlipoproteinemia. Front Endocrinol (Lausanne) 2022; 13:874608. [PMID: 35923617 PMCID: PMC9339609 DOI: 10.3389/fendo.2022.874608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/03/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Type I hyperlipoproteinemia, characterized by severe hypertriglyceridemia, is caused mainly by loss-of-function mutation of the lipoprotein lipase (LPL) gene. To date, more than 200 mutations in the LPL gene have been reported, while only a limited number of mutations have been evaluated for pathogenesis. OBJECTIVE This study aims to explore the molecular mechanisms underlying lipoprotein lipase deficiency in two pedigrees with type 1 hyperlipoproteinemia. METHODS We conducted a systematic clinical and genetic analysis of two pedigrees with type 1 hyperlipoproteinemia. Postheparin plasma of all the members was used for the LPL activity analysis. In vitro studies were performed in HEK-293T cells that were transiently transfected with wild-type or variant LPL plasmids. Furthermore, the production and activity of LPL were analyzed in cell lysates or culture medium. RESULTS Proband 1 developed acute pancreatitis in youth, and her serum triglycerides (TGs) continued to be at an ultrahigh level, despite the application of various lipid-lowering drugs. Proband 2 was diagnosed with type 1 hyperlipoproteinemia at 9 months of age, and his serum TG levels were mildly elevated with treatment. Two novel compound heterozygous variants of LPL (c.3G>C, p. M1? and c.835_836delCT, p. L279Vfs*3, c.188C>T, p. Ser63Phe and c.662T>C, p. Ile221Thr) were identified in the two probands. The postheparin LPL activity of probands 1 and 2 showed decreases of 72.22 ± 9.46% (p<0.01) and 54.60 ± 9.03% (p<0.01), respectively, compared with the control. In vitro studies showed a substantial reduction in the expression or enzyme activity of LPL in the LPL variants. CONCLUSIONS Two novel compound heterozygous variants of LPL induced defects in the expression and function of LPL and caused type I hyperlipoproteinemia. The functional characterization of these variants was in keeping with the postulated LPL mutant activity.
Collapse
Affiliation(s)
- Shuping Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Endocrinology and Metabolism, Dongying People’s Hospital, Dongying, China
| | - Yiping Cheng
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yingzhou Shi
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wanyi Zhao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ling Gao
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Li Fang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaolong Jin
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoyan Han
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiuying Sun
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guimei Li
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- *Correspondence: Jiajun Zhao, ; Guimei Li, ; Chao Xu,
| | - Jiajun Zhao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Jiajun Zhao, ; Guimei Li, ; Chao Xu,
| | - Chao Xu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Jiajun Zhao, ; Guimei Li, ; Chao Xu,
| |
Collapse
|
23
|
Jayaraman S, Pérez A, Miñambres I, Sánchez-Quesada JL, Gursky O. Heparin binding triggers human VLDL remodeling by circulating lipoprotein lipase: Relevance to VLDL functionality in health and disease. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159064. [PMID: 34610468 PMCID: PMC8595799 DOI: 10.1016/j.bbalip.2021.159064] [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: 07/20/2021] [Revised: 09/13/2021] [Accepted: 09/29/2021] [Indexed: 02/06/2023]
Abstract
Hydrolysis of VLDL triacylglycerol (TG) by lipoprotein lipase (LpL) is a major step in energy metabolism and VLDL-to-LDL maturation. Most functional LpL is anchored to the vascular endothelium, yet a small amount circulates on TG-rich lipoproteins. As circulating LpL has low catalytic activity, its role in VLDL remodeling is unclear. We use pre-heparin plasma and heparin-sepharose affinity chromatography to isolate VLDL fractions from normolipidemic, hypertriglyceridemic, or type-2 diabetic subjects. LpL is detected only in the heparin-bound fraction. Transient binding to heparin activates this VLDL-associated LpL, which hydrolyses TG, leading to gradual VLDL remodeling into IDL/LDL and HDL-size particles. The products and the timeframe of this remodeling closely resemble VLDL-to-LDL maturation in vivo. Importantly, the VLDL fraction that does not bind heparin is not remodeled. This relatively inert LpL-free VLDL is rich in TG and apoC-III, poor in apoE and apoC-II, shows impaired functionality as a substrate for the exogenous LpL or CETP, and likely has prolonged residence time in blood, which is expected to promote atherogenesis. This non-bound VLDL fraction increases in hypertriglyceridemia and in type-2 diabetes but decreases upon diabetes treatment that restores the glycemic control. In stark contrast, heparin binding by LDL increases in type-2 diabetes triggering pro-atherogenic LDL modifications. Therefore, the effects of heparin binding are associated negatively with atherogenesis for VLDL but positively for LDL. Collectively, the results reveal that binding to glycosaminoglycans initiates VLDL remodeling by circulating LpL, and suggest heparin binding as a marker of VLDL functionality and a readout for treatment of metabolic disorders.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA.,Corresponding author.
| | - Antonio Pérez
- Endocrinology Department of the Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain
| | - Inka Miñambres
- Endocrinology Department of the Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain.,Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau, CIBERDEM, Barcelona, Spain
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
24
|
Jiang H, Pu Y, Li ZH, Liu W, Deng Y, Liang R, Zhang XM, Zuo HD. Adiponectin, May Be a Potential Protective Factor for Obesity-Related Osteoarthritis. Diabetes Metab Syndr Obes 2022; 15:1305-1319. [PMID: 35510046 PMCID: PMC9058006 DOI: 10.2147/dmso.s359330] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/08/2022] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease in elderly individuals and seriously affects quality of life. OA has often been thought to be caused by body weight load, but studies have increasingly shown that OA is an inflammation-mediated metabolic disease. The current existing evidence suggests that OA is associated with obesity-related chronic inflammation as well as abnormal lipid metabolism in obesity, such as fatty acids (FA) and triglycerides. Adiponectin, a cytokine secreted by adipose tissue, can affect the progression of OA by regulating obesity-related inflammatory factors. However, the specific molecular mechanism has not been fully elucidated. According to previous research, adiponectin can promote the metabolism of FA and triglycerides, which indicates that it is a potential protective factor for OA through many mechanisms. This article aims to review the mechanisms of chronic inflammation, FA and triglycerides in OA, as well as the potential mechanisms of adiponectin in regulating chronic inflammation and promoting FA and triglyceride metabolism. Therefore, adiponectin may have a protective effect on obesity-related OA, which could provide new insight into adiponectin and the related mechanisms in OA.
Collapse
Affiliation(s)
- Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Yu Pu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Zeng-Hui Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Wei Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Yan Deng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Rui Liang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
- Correspondence: Hou-Dong Zuo, Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China, Tel +86-817-2587621, Email
| |
Collapse
|
25
|
Lund Winther AM, Kristensen KK, Kumari A, Ploug M. Expression and one-step purification of active lipoprotein lipase contemplated by biophysical considerations. J Lipid Res 2021; 62:100149. [PMID: 34780727 DOI: 10.1016/j.jlr.2021.100149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/17/2022] Open
Abstract
Lipoprotein lipase (LPL) is essential for intravascular lipid metabolism and is of high medical relevance. Since LPL is notoriously unstable, there is an unmet need for a robust expression system producing high quantities of active and pure recombinant human LPL. We showed previously that bovine LPL purified from milk is unstable at body temperature (Tm is 34.8 °C), but in the presence of the endothelial transporter glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1) LPL is stabile (Tm increases to 57.6 °C). Building on this information, we now designed an expression system for human LPL using Drosophila S2 cells grown in suspension at high cell density and at an advantageous temperature of 25 °C. We co-transfected S2 cells with human LPL, LMF1 and soluble GPIHBP1 to provide an efficient chaperoning and stabilization of LPL in all compartments during synthesis and after secretion into the conditioned medium. For LPL purification, we used heparin-Sepharose affinity chromatography, which disrupted LPL-GPIHBP1 complexes causing GPIHBP1 to elute with the flow-through of the conditioned media. This one-step purification procedure yielded high quantities of pure and active LPL (4‒28 mg/L). Purification of several human LPL variants (furin-cleavage resistant mutant R297A, active-site mutant S132A, and lipid-binding-deficient mutant W390A-W393A-W394A) as well as murine LPL underscores the versatility and robustness of this protocol. Notably, we were able to produce and purify LPL containing the cognate furin-cleavage site. This method provides an efficient and cost-effective approach to produce large quantities of LPL for biophysical and large-scale drug discovery studies.
Collapse
Affiliation(s)
- Anne-Marie Lund Winther
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Kristian Kølby Kristensen
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Anni Kumari
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Kristensen KK, Leth-Espensen KZ, Kumari A, Grønnemose AL, Lund-Winther AM, Young SG, Ploug M. GPIHBP1 and ANGPTL4 Utilize Protein Disorder to Orchestrate Order in Plasma Triglyceride Metabolism and Regulate Compartmentalization of LPL Activity. Front Cell Dev Biol 2021; 9:702508. [PMID: 34336854 PMCID: PMC8319833 DOI: 10.3389/fcell.2021.702508] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Intravascular processing of triglyceride-rich lipoproteins (TRLs) is crucial for delivery of dietary lipids fueling energy metabolism in heart and skeletal muscle and for storage in white adipose tissue. During the last decade, mechanisms underlying focal lipolytic processing of TRLs along the luminal surface of capillaries have been clarified by fresh insights into the functions of lipoprotein lipase (LPL); LPL's dedicated transporter protein, glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1); and its endogenous inhibitors, angiopoietin-like (ANGPTL) proteins 3, 4, and 8. Key discoveries in LPL biology include solving the crystal structure of LPL, showing LPL is catalytically active as a monomer rather than as a homodimer, and that the borderline stability of LPL's hydrolase domain is crucial for the regulation of LPL activity. Another key discovery was understanding how ANGPTL4 regulates LPL activity. The binding of ANGPTL4 to LPL sequences adjacent to the catalytic cavity triggers cooperative and sequential unfolding of LPL's hydrolase domain resulting in irreversible collapse of the catalytic cavity and loss of LPL activity. Recent studies have highlighted the importance of the ANGPTL3-ANGPTL8 complex for endocrine regulation of LPL activity in oxidative organs (e.g., heart, skeletal muscle, brown adipose tissue), but the molecular mechanisms have not been fully defined. New insights have also been gained into LPL-GPIHBP1 interactions and how GPIHBP1 moves LPL to its site of action in the capillary lumen. GPIHBP1 is an atypical member of the LU (Ly6/uPAR) domain protein superfamily, containing an intrinsically disordered and highly acidic N-terminal extension and a disulfide bond-rich three-fingered LU domain. Both the disordered acidic domain and the folded LU domain are crucial for the stability and transport of LPL, and for modulating its susceptibility to ANGPTL4-mediated unfolding. This review focuses on recent advances in the biology and biochemistry of crucial proteins for intravascular lipolysis.
Collapse
Affiliation(s)
- Kristian Kølby Kristensen
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Zinck Leth-Espensen
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Anni Kumari
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Anne Louise Grønnemose
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie Lund-Winther
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Stephen G Young
- Departments of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
The Importance of Lipoprotein Lipase Regulation in Atherosclerosis. Biomedicines 2021; 9:biomedicines9070782. [PMID: 34356847 PMCID: PMC8301479 DOI: 10.3390/biomedicines9070782] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
Lipoprotein lipase (LPL) plays a major role in the lipid homeostasis mainly by mediating the intravascular lipolysis of triglyceride rich lipoproteins. Impaired LPL activity leads to the accumulation of chylomicrons and very low-density lipoproteins (VLDL) in plasma, resulting in hypertriglyceridemia. While low-density lipoprotein cholesterol (LDL-C) is recognized as a primary risk factor for atherosclerosis, hypertriglyceridemia has been shown to be an independent risk factor for cardiovascular disease (CVD) and a residual risk factor in atherosclerosis development. In this review, we focus on the lipolysis machinery and discuss the potential role of triglycerides, remnant particles, and lipolysis mediators in the onset and progression of atherosclerotic cardiovascular disease (ASCVD). This review details a number of important factors involved in the maturation and transportation of LPL to the capillaries, where the triglycerides are hydrolyzed, generating remnant lipoproteins. Moreover, LPL and other factors involved in intravascular lipolysis are also reported to impact the clearance of remnant lipoproteins from plasma and promote lipoprotein retention in capillaries. Apolipoproteins (Apo) and angiopoietin-like proteins (ANGPTLs) play a crucial role in regulating LPL activity and recent insights into LPL regulation may elucidate new pharmacological means to address the challenge of hypertriglyceridemia in atherosclerosis development.
Collapse
|
28
|
Saji RS, Prasana JC, Muthu S, George J. Experimental and theoretical spectroscopic (FT-IR, FT-Raman, UV-VIS) analysis, natural bonding orbitals and molecular docking studies on 2-bromo-6-methoxynaphthalene: A potential anti-cancer drug. Heliyon 2021; 7:e07213. [PMID: 34169166 PMCID: PMC8209084 DOI: 10.1016/j.heliyon.2021.e07213] [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: 03/26/2021] [Revised: 05/07/2021] [Accepted: 06/01/2021] [Indexed: 10/28/2022] Open
Abstract
The vibrational, electronic and charge transfer studies on 2-bromo-6-methoxynaphthalene (2BMN) were done using DFT method with B3LYP/6-311++G(d,p) theory using GAUSSIAN 09W software. Theoretical and experimental investigations on FT-IR and FT Raman were executed on 2BMN. The calculated vibrational wavenumbers were scaled using suitable scaling factors and vibrational assignments were done to all modes of vibrations using Potential Energy Distribution (PED). Frontier Molecular Orbitals were calculated using TD-DFT method and the HOMO-LUMO energy gap was also obtained. Other electronic properties and global parameters for 2BMN were found using the HOMO-LUMO energy values. An energy gap of 4.208 eV shows the stability of the molecule. The reactive sites were predicted using Molecular Electrostatic Potential (MEP), Electron Localization Function (ELF) and Fukui calculations. Hence all electrophilic sites and nucleophilic areas of the molecule were determined. The delocalization of electron density was studied using NBO calculations. The intramolecular transitions and stability of structure were explained using in detail using the former. As the compound satisfies drug-like properties and has a softness value (indicating its less toxic nature), it may be used as a pharmaceutical product. Molecular docking studies were made and the protein-ligand binding properties were discussed. It was found out that title compound exhibits anti-cancer activities. The low binding energy predicts that the compound may be modified as a drug for treating Cancer.
Collapse
Affiliation(s)
- Rinnu Sara Saji
- Department of Physics, Madras Christian College, East Tambaram 600059, Tamil Nadu, India.,University of Madras, Chennai, 600005, Tamil Nadu, India
| | | | - S Muthu
- Department of Physics, Arignar Anna Government Arts College, Cheyyar, 604407, Tamil Nadu, India.,Department of Physics, Puratchi Thalaivar Dr. M. G. R Govt. Arts and Science College, Uthiramerur, 603406, Tamil Nadu, India
| | - Jacob George
- Department of Physics, Madras Christian College, East Tambaram 600059, Tamil Nadu, India.,University of Madras, Chennai, 600005, Tamil Nadu, India
| |
Collapse
|
29
|
Shaik A, Rosenson RS. Genetics of Triglyceride-Rich Lipoproteins Guide Identification of Pharmacotherapy for Cardiovascular Risk Reduction. Cardiovasc Drugs Ther 2021; 35:677-690. [PMID: 33710501 DOI: 10.1007/s10557-021-07168-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Despite aggressive reduction of low-density lipoprotein cholesterol (LDL-C), there is a residual risk of cardiovascular disease (CVD). Hypertriglyceridemia is known to be associated with increased CVD risk, independently of LDL-C. Triglycerides are one component of the heterogenous class of triglyceride-rich lipoproteins (TGRLs). METHODS/RESULTS Growing evidence from biology, epidemiology, and genetics supports the contribution of TGRLs to the development of CVD via a number of mechanisms, including through proinflammatory, proapoptotic, and procoagulant pathways. CONCLUSION New genetics-guided pharmacotherapies to reduce levels of triglycerides and TGRLs and thus reduce risk of CVD have been developed and will be discussed here.
Collapse
Affiliation(s)
- Aleesha Shaik
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert S Rosenson
- Cardiometabolics Unit, Zena and Michael A Wiener Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
30
|
Yang Q, Pu N, Li XY, Shi XL, Chen WW, Zhang GF, Hu YP, Zhou J, Chen FX, Li BQ, Tong ZH, Férec C, Cooper DN, Chen JM, Li WQ. Digenic Inheritance and Gene-Environment Interaction in a Patient With Hypertriglyceridemia and Acute Pancreatitis. Front Genet 2021; 12:640859. [PMID: 34040631 PMCID: PMC8143378 DOI: 10.3389/fgene.2021.640859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
The etiology of hypertriglyceridemia (HTG) and acute pancreatitis (AP) is complex. Herein, we dissected the underlying etiology in a patient with HTG and AP. The patient had a 20-year history of heavy alcohol consumption and an 8-year history of mild HTG. He was hospitalized for alcohol-triggered AP, with a plasma triglyceride (TG) level up to 21.4 mmol/L. A temporary rise in post-heparin LPL concentration (1.5–2.5 times of controls) was noted during the early days of AP whilst LPL activity was consistently low (50∼70% of controls). His TG level rapidly decreased to normal in response to treatment, and remained normal to borderline high during a ∼3-year follow-up period during which he had abstained completely from alcohol. Sequencing of the five primary HTG genes (i.e., LPL, APOC2, APOA5, GPIHBP1 and LMF1) identified two heterozygous variants. One was the common APOA5 c.553G > T (p.Gly185Cys) variant, which has been previously associated with altered TG levels as well as HTG-induced acute pancreatitis (HTG-AP). The other was a rare variant in the LPL gene, c.756T > G (p.Ile252Met), which was predicted to be likely pathogenic and found experimentally to cause a 40% loss of LPL activity without affecting either protein synthesis or secretion. We provide evidence that both a gene-gene interaction (between the common APOA5 variant and the rare LPL variant) and a gene-environment interaction (between alcohol and digenic inheritance) might have contributed to the development of mild HTG and alcohol-triggered AP in the patient, thereby improving our understanding of the complex etiology of HTG and HTG-AP.
Collapse
Affiliation(s)
- Qi Yang
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Na Pu
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiao-Yao Li
- Department of Intensive Care Unit, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiao-Lei Shi
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei-Wei Chen
- Department of Gastroenterology, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Guo-Fu Zhang
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yue-Peng Hu
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jing Zhou
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fa-Xi Chen
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bai-Qiang Li
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhi-Hui Tong
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Claude Férec
- Univ Brest, INSERM, EFS, UMR 1078, GGB, Brest, France.,Service de Génétique Médicale et de Biologie de la Reproduction, CHRU Brest, Brest, France
| | - David N Cooper
- School of Medicine, Institute of Medical Genetics, Cardiff University, Cardiff, United Kingdom
| | - Jian-Min Chen
- Univ Brest, INSERM, EFS, UMR 1078, GGB, Brest, France
| | - Wei-Qin Li
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
31
|
Jin N, Matter WF, Michael LF, Qian Y, Gheyi T, Cano L, Perez C, Lafuente C, Broughton HB, Espada A. The Angiopoietin-Like Protein 3 and 8 Complex Interacts with Lipoprotein Lipase and Induces LPL Cleavage. ACS Chem Biol 2021; 16:457-462. [PMID: 33656326 DOI: 10.1021/acschembio.0c00954] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipoprotein lipase (LPL) is the key enzyme that hydrolyzes triglycerides from triglyceride-rich lipoproteins. Angiopoietin-like proteins (ANGPTL) 3, 4, and 8 are well-characterized protein inhibitors of LPL. ANGPTL8 forms a complex with ANGPTL3, and the complex is a potent endogenous inhibitor of LPL. However, the nature of the structural interaction between ANGPTL3/8 and LPL is unknown. To probe the conformational changes in LPL induced by ANGPTL3/8, we found that HDX-MS detected significantly altered deuteration in the lid region, ApoC2 binding site, and furin cleavage region of LPL in the presence of ANGPTL3/8. Supporting this HDX structural evidence, we found that ANGPTL3/8 inhibits LPL enzymatic activities and increases LPL cleavage. ANGPTL3/8-induced effects on LPL activity and LPL cleavage are much stronger than those of ANGPTL3 or ANGPTL8 alone. ANGPTL3/8-mediated LPL cleavage is blocked by both an ANGPTL3 antibody and a furin inhibitor. Knock-down of furin expression by siRNA significantly reduced ANGPT3/8-induced cleavage of LPL. Our data suggest ANGPTL3/8 promotes furin-mediated LPL cleavage.
Collapse
Affiliation(s)
- Najia Jin
- Diabetes and Complications Therapeutic Area, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - William F. Matter
- Diabetes and Complications Therapeutic Area, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Laura F. Michael
- Diabetes and Complications Therapeutic Area, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Yuewei Qian
- Laboratory for Experimental Medicine, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Tarun Gheyi
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Leticia Cano
- Centro de Investigación Lilly S.A., 28108 Alcobendas, Spain
| | - Carlos Perez
- Centro de Investigación Lilly S.A., 28108 Alcobendas, Spain
| | - Celia Lafuente
- Centro de Investigación Lilly S.A., 28108 Alcobendas, Spain
| | | | - Alfonso Espada
- Centro de Investigación Lilly S.A., 28108 Alcobendas, Spain
| |
Collapse
|
32
|
ANGPLT3 in cardio-metabolic disorders. Mol Biol Rep 2021; 48:2729-2739. [PMID: 33677817 DOI: 10.1007/s11033-021-06248-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/22/2021] [Indexed: 01/15/2023]
Abstract
Dyslipidemia is associated with numerous health problems that include the combination of insulin resistance, hypertension and obesity, which is always grouped together asmetabolic syndrome. Given that metabolic syndrome leads to a high mortality and poses serious risks to human health worldwide, it is vital to explore the mechanisms whereby dyslipidemia modulates the risk and the severity of cardio-metabolic disorders. Recently, a specific secretory protein family, named angiopoietin-like protein (ANGPTL), is considered as one of the significant biomarkers which facilitate the development of angiogenesis. Among the eight proteins of ANGPTL family, ANGPTL3 has been demonstrated as an essential modulator of lipid catabolism within circulation by inhibiting the activity of lipoprotein lipase (LPL) and endothelial lipase (EL). Consistent with these notions, mice with ANGPTL3 gene-deficiency presented reduced circulating levels of low density lipoprotein cholesterol (LDL-C) and lower risk of atherosclerosis. On the other hand, participants carrying homozygous loss-of function (LOF) mutation in ANGPTL3 gene also displayed lower circulating LDL-C levels and atherosclerotic risk. In the current review, we summarized the recent understanding of ANGPTL3 in controlling the risk and the development of dyslipidemia and its related cardio-metabolic disorders. Moreover, we also provided the perspectives which potentially suggested that ANGPTL3 could be considered as a promising target in treating metabolic syndrome.
Collapse
|
33
|
Abumrad NA, Cabodevilla AG, Samovski D, Pietka T, Basu D, Goldberg IJ. Endothelial Cell Receptors in Tissue Lipid Uptake and Metabolism. Circ Res 2021; 128:433-450. [PMID: 33539224 DOI: 10.1161/circresaha.120.318003] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.
Collapse
Affiliation(s)
- Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Dmitri Samovski
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Terri Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| |
Collapse
|
34
|
Wu SA, Kersten S, Qi L. Lipoprotein Lipase and Its Regulators: An Unfolding Story. Trends Endocrinol Metab 2021; 32:48-61. [PMID: 33277156 PMCID: PMC8627828 DOI: 10.1016/j.tem.2020.11.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Lipoprotein lipase (LPL) is one of the most important factors in systemic lipid partitioning and metabolism. It mediates intravascular hydrolysis of triglycerides packed in lipoproteins such as chylomicrons and very-low-density lipoprotein (VLDL). Since its initial discovery in the 1940s, its biology and pathophysiological significance have been well characterized. Nonetheless, several studies in the past decade, with recent delineation of LPL crystal structure and the discovery of several new regulators such as angiopoietin-like proteins (ANGPTLs), glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), lipase maturation factor 1 (LMF1) and Sel-1 suppressor of Lin-12-like 1 (SEL1L), have completely transformed our understanding of LPL biology.
Collapse
Affiliation(s)
- Shuangcheng Alivia Wu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48105, USA.
| | - Sander Kersten
- Nutrition Metabolism and Genomics group, Wageningen University, Wageningen, The Netherlands
| | - Ling Qi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48105, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA.
| |
Collapse
|
35
|
D. Bruce K, Tang M, Reigan P, H. Eckel R. Genetic Variants of Lipoprotein Lipase and Regulatory Factors Associated with Alzheimer's Disease Risk. Int J Mol Sci 2020; 21:ijms21218338. [PMID: 33172164 PMCID: PMC7664401 DOI: 10.3390/ijms21218338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Lipoprotein lipase (LPL) is a key enzyme in lipid and lipoprotein metabolism. The canonical role of LPL involves the hydrolysis of triglyceride-rich lipoproteins for the provision of FFAs to metabolic tissues. However, LPL may also contribute to lipoprotein uptake by acting as a molecular bridge between lipoproteins and cell surface receptors. Recent studies have shown that LPL is abundantly expressed in the brain and predominantly expressed in the macrophages and microglia of the human and murine brain. Moreover, recent findings suggest that LPL plays a direct role in microglial function, metabolism, and phagocytosis of extracellular factors such as amyloid- beta (Aβ). Although the precise function of LPL in the brain remains to be determined, several studies have implicated LPL variants in Alzheimer's disease (AD) risk. For example, while mutations shown to have a deleterious effect on LPL function and expression (e.g., N291S, HindIII, and PvuII) have been associated with increased AD risk, a mutation associated with increased bridging function (S447X) may be protective against AD. Recent studies have also shown that genetic variants in endogenous LPL activators (ApoC-II) and inhibitors (ApoC-III) can increase and decrease AD risk, respectively, consistent with the notion that LPL may play a protective role in AD pathogenesis. Here, we review recent advances in our understanding of LPL structure and function, which largely point to a protective role of functional LPL in AD neuropathogenesis.
Collapse
Affiliation(s)
- Kimberley D. Bruce
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.T.); (R.H.E.)
- Correspondence:
| | - Maoping Tang
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.T.); (R.H.E.)
| | - Philip Reigan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Robert H. Eckel
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.T.); (R.H.E.)
| |
Collapse
|
36
|
Abstract
The enzyme lipoprotein lipase (LPL) is responsible for breaking down triglycerides in the blood. Mutations in LPL cause a rare but debilitating disorder characterized by excessive plasma triglyceride levels for which treatment options are limited. Nimonkar et al. now present a fusion protein between LPL and its physiological transporter GBIHBP1 that is highly active and largely resistant to physiological inhibitors of LPL. Injecting this fusion protein effectively lowers plasma triglycerides in mice and represents a promising new approach for lowering triglycerides in patients with familial chylomicronemia syndrome.
Collapse
Affiliation(s)
- Philip M M Ruppert
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| |
Collapse
|
37
|
A novel long noncoding RNA, ENSGALG00000021686, regulates the intracellular transport of fatty acids by targeting the FABP3 gene in chicken. Biochem Biophys Res Commun 2020; 528:706-712. [PMID: 32507601 DOI: 10.1016/j.bbrc.2020.05.133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022]
Abstract
Fatty acids (FAs) are essential for the vital movement of humans and animals. Their metabolism is, in part, regulated by FABP3. In our previous study, a novel lncRNA (ENSGALG00000021686, L21686) was identified, and FABP3 was predicted as its target gene. Here, using chicken myocytes, lymphocytes, and different tissues, L21686 target on the FABP3 gene, FABP3 mRNA expression, and their effect on FA metabolism are explored. The results show that the highest expression of L21686 is in muscle tissue, a significant energy-consuming tissue. L21686 expression is consistent with FABP3 mRNA expression. We also show that under the different treatments, the levels of FABP3 mRNA and protein in myocytes and lymphocytes change in tandem with L21686 expression. Moreover, the dual-luciferase reporter assay provided direct evidence that L21686 targets the FABP3 gene. Finally, it was found that the content of free FAs increases along with the up-regulation of L21686 and the FABP3 gene. Malonyl CoA content does not change under the different treatments, suggesting that L21686 regulates the intake of extracellular FAs in chicken. Further, the changes in lipoprotein lipase (LPL), sterol-regulatory element binding protein 1 (SREBP-1), fatty acid synthase (FASN), and acetyl-CoA carboxylase (ACC) mRNA levels support this view. In summary, our data show that the new lncRNA (L21686) regulates the intake of extracellular FAs in chicken cells in vitro by targeting the expression of the FABP3 gene. Our findings will help to establish the groundwork and provide a new clue for deciphering the regulation of FAs metabolism in chicken.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW To discuss the recent developments in structure, function and physiology of lipoprotein lipase (LpL) and the regulators of LpL, which are being targeted for therapy. RECENT FINDINGS Recent studies have revealed the long elusive crystal structure of LpL and its interaction with glycosylphosphatidylinositol anchored high-density lipoprotein binding protein 1 (GPIHBP1). New light has been shed on LpL being active as a monomer, which brings into questions previous thinking that LpL inhibitors like angiopoietin-like 4 (ANGPTL4) and stabilizers like LMF1 work on disrupting or maintaining LpL in dimer form. There is increasing pharmaceutical interest in developing targets to block LpL inhibitors like ANGPTL3. Other approaches to reducing circulating triglyceride levels have been using an apoC2 mimetic and reducing apoC3. SUMMARY Lipolysis of triglyceride-rich lipoproteins by LpL is a central event in lipid metabolism, releasing fatty acids for uptake by tissues and generating low-density lipoprotein and expanding high-density lipoprotein. Recent mechanistic insights into the structure and function of LpL have added to our understanding of triglyceride metabolism. This has also led to heightened interest in targeting its posttranslational regulators, which can be the next generation of lipid-lowering agents used to prevent hypertriglyceridemic pancreatitis and, hopefully, cardiovascular disease.
Collapse
Affiliation(s)
- Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | | |
Collapse
|
39
|
The structure of helical lipoprotein lipase reveals an unexpected twist in lipase storage. Proc Natl Acad Sci U S A 2020; 117:10254-10264. [PMID: 32332168 DOI: 10.1073/pnas.1916555117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lipases are enzymes necessary for the proper distribution and utilization of lipids in the human body. Lipoprotein lipase (LPL) is active in capillaries, where it plays a crucial role in preventing dyslipidemia by hydrolyzing triglycerides from packaged lipoproteins. Thirty years ago, the existence of a condensed and inactive LPL oligomer was proposed. Although recent work has shed light on the structure of the LPL monomer, the inactive oligomer remained opaque. Here we present a cryo-EM reconstruction of a helical LPL oligomer at 3.8-Å resolution. Helix formation is concentration-dependent, and helices are composed of inactive dihedral LPL dimers. Heparin binding stabilizes LPL helices, and the presence of substrate triggers helix disassembly. Superresolution fluorescent microscopy of endogenous LPL revealed that LPL adopts a filament-like distribution in vesicles. Mutation of one of the helical LPL interaction interfaces causes loss of the filament-like distribution. Taken together, this suggests that LPL is condensed into its inactive helical form for storage in intracellular vesicles.
Collapse
|
40
|
Unfolding of monomeric lipoprotein lipase by ANGPTL4: Insight into the regulation of plasma triglyceride metabolism. Proc Natl Acad Sci U S A 2020; 117:4337-4346. [PMID: 32034094 DOI: 10.1073/pnas.1920202117] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The binding of lipoprotein lipase (LPL) to GPIHBP1 focuses the intravascular hydrolysis of triglyceride-rich lipoproteins on the surface of capillary endothelial cells. This process provides essential lipid nutrients for vital tissues (e.g., heart, skeletal muscle, and adipose tissue). Deficiencies in either LPL or GPIHBP1 impair triglyceride hydrolysis, resulting in severe hypertriglyceridemia. The activity of LPL in tissues is regulated by angiopoietin-like proteins 3, 4, and 8 (ANGPTL). Dogma has held that these ANGPTLs inactivate LPL by converting LPL homodimers into monomers, rendering them highly susceptible to spontaneous unfolding and loss of enzymatic activity. Here, we show that binding of an LPL-specific monoclonal antibody (5D2) to the tryptophan-rich lipid-binding loop in the carboxyl terminus of LPL prevents homodimer formation and forces LPL into a monomeric state. Of note, 5D2-bound LPL monomers are as stable as LPL homodimers (i.e., they are not more prone to unfolding), but they remain highly susceptible to ANGPTL4-catalyzed unfolding and inactivation. Binding of GPIHBP1 to LPL alone or to 5D2-bound LPL counteracts ANGPTL4-mediated unfolding of LPL. In conclusion, ANGPTL4-mediated inactivation of LPL, accomplished by catalyzing the unfolding of LPL, does not require the conversion of LPL homodimers into monomers. Thus, our findings necessitate changes to long-standing dogma on mechanisms for LPL inactivation by ANGPTL proteins. At the same time, our findings align well with insights into LPL function from the recent crystal structure of the LPL•GPIHBP1 complex.
Collapse
|
41
|
Morelli MB, Chavez C, Santulli G. Angiopoietin-like proteins as therapeutic targets for cardiovascular disease: focus on lipid disorders. Expert Opin Ther Targets 2020; 24:79-88. [PMID: 31856617 DOI: 10.1080/14728222.2020.1707806] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Angiopoietin-like (ANGPTL) proteins belong to a family of eight secreted factors that are structurally related to proteins that modulate angiogenesi, commonly known as angiopoietins. Specifically, ANGPTL3, ANGPTL4, and ANGPTL8 (the 'ANGPT L3-4-8 triad'), have surfaced as principal regulators of plasma lipid metabolism by functioning as potent inhibitors of lipoprotein lipase. The targeting of these proteins may open up future therapeutic avenues for metabolic and cardiovascular disease.Areas covered: This article systematically summarizes the compelling literature describing the mechanistic roles of ANGPTL3, 4, and 8 in lipid metabolism, emphasizing their importance in determining the risk of cardiovascular disease. We shed light on population-based studies linking loss-of-function variations in ANGPTL3, 4, and 8 with decreased risk of metabolic conditions and cardiovascular disorders. We also discuss how the strategies aiming at targeting the ANGPT L3-4-8 triad could offer therapeutic benefit in the clinical scenario.Expert opinion: Monoclonal antibodies and antisense oligonucleotides that target ANGPTL3, 4, and 8 are potentially an efficient therapeutic strategy for hypertriglyceridemia and cardiovascular risk reduction, especially in patients with limited treatment options. These innovative therapeutical approaches are at an embryonic stage in development and hence further investigations are necessary for eventual use in humans.
Collapse
Affiliation(s)
- Marco Bruno Morelli
- Department of Medicine; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA.,Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), The "Norman Fleischer" Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, NY, New York, USA
| | - Christopher Chavez
- Department of Medicine; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Gaetano Santulli
- Department of Medicine; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA.,Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), The "Norman Fleischer" Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, NY, New York, USA.,Department of Advanced Biomedical Sciences and International Translational Research and Medical Education Consortium (ITME), "Federico II" University, Naples, Italy
| |
Collapse
|
42
|
Ruscica M, Zimetti F, Adorni MP, Sirtori CR, Lupo MG, Ferri N. Pharmacological aspects of ANGPTL3 and ANGPTL4 inhibitors: New therapeutic approaches for the treatment of atherogenic dyslipidemia. Pharmacol Res 2020; 153:104653. [PMID: 31931117 DOI: 10.1016/j.phrs.2020.104653] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/24/2022]
Abstract
Among the determinants of atherosclerotic cardiovascular disease (ASCVD), genetic and experimental evidence has provided data on a major role of angiopoietin-like proteins 3 and 4 (ANGPTL3 and ANGPTL4) in regulating the activity of lipoprotein lipase (LPL), antagonizing the hydrolysis of triglycerides (TG). Indeed, beyond low-density lipoprotein cholesterol (LDL-C), ASCVD risk is also dependent on a cluster of metabolic abnormalities characterized by elevated fasting and post-prandial levels of TG-rich lipoproteins and their remnants. In a head-to-head comparison between murine models for ANGPTL3 and ANGPTL4, the former was found to be a better pharmacological target for the treatment of hypertriglyceridemia. In humans, loss-of-function mutations of ANGPTL3 are associated with a marked reduction of plasma levels of VLDL, low-density lipoprotein (LDL) and high-density lipoprotein (HDL). Carriers of loss-of-function mutations of ANGPTL4 show instead lower TG-rich lipoproteins and a modest but significant increase of HDL. The relevance of ANGPTL3 and ANGPTL4 as new therapeutic targets is proven by the development of monoclonal antibodies or antisense oligonucleotides. Studies in animal models, including non-human primates, have demonstrated that short-term treatment with monoclonal antibodies against ANGPTL3 and ANGPTL4 induces activation of LPL and a marked reduction of plasma TG-rich-lipoproteins, apparently without any major side effects. Inhibition of both targets also partially reduces LDL-C, independent of the LDL receptor. Similar evidence has been observed with the antisense oligonucleotide ANGPTL3-LRX. The genetic studies have paved the way for the development of new ANGPTL3 and 4 antagonists for the treatment of atherogenic dyslipidemias. Conclusive data of phase 2 and 3 clinical trials are still needed in order to define their safety and efficacy profile.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| | - Francesca Zimetti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Maria Pia Adorni
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Cesare R Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Maria Giovanna Lupo
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padua, Italy
| | - Nicola Ferri
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padua, Italy
| |
Collapse
|
43
|
Basu D, Bornfeldt KE. Hypertriglyceridemia and Atherosclerosis: Using Human Research to Guide Mechanistic Studies in Animal Models. Front Endocrinol (Lausanne) 2020; 11:504. [PMID: 32849290 PMCID: PMC7423973 DOI: 10.3389/fendo.2020.00504] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Human studies support a strong association between hypertriglyceridemia and atherosclerotic cardiovascular disease (CVD). However, whether a causal relationship exists between hypertriglyceridemia and increased CVD risk is still unclear. One plausible explanation for the difficulty establishing a clear causal role for hypertriglyceridemia in CVD risk is that lipolysis products of triglyceride-rich lipoproteins (TRLs), rather than the TRLs themselves, are the likely mediators of increased CVD risk. This hypothesis is supported by studies of rare mutations in humans resulting in impaired clearance of such lipolysis products (remnant lipoprotein particles; RLPs). Several animal models of hypertriglyceridemia support this hypothesis and have provided additional mechanistic understanding. Mice deficient in lipoprotein lipase (LPL), the major vascular enzyme responsible for TRL lipolysis and generation of RLPs, or its endothelial anchor GPIHBP1, are severely hypertriglyceridemic but develop only minimal atherosclerosis as compared with animal models deficient in apolipoprotein (APO) E, which is required to clear TRLs and RLPs. Likewise, animal models convincingly show that increased clearance of TRLs and RLPs by LPL activation (achieved by inhibition of APOC3, ANGPTL3, or ANGPTL4 action, or increased APOA5) results in protection from atherosclerosis. Mechanistic studies suggest that RLPs are more atherogenic than large TRLs because they more readily enter the artery wall, and because they are enriched in cholesterol relative to triglycerides, which promotes pro-atherogenic effects in lesional cells. Other mechanistic studies show that hepatic receptors (LDLR and LRP1) and APOE are critical for RLP clearance. Thus, studies in animal models have provided additional mechanistic insight and generally agree with the hypothesis that RLPs derived from TRLs are highly atherogenic whereas hypertriglyceridemia due to accumulation of very large TRLs in plasma is not markedly atherogenic in the absence of TRL lipolysis products.
Collapse
Affiliation(s)
- Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY, United States
| | - Karin E. Bornfeldt
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
- Department of Pathology, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
- *Correspondence: Karin E. Bornfeldt
| |
Collapse
|
44
|
Koerner CM, Roberts BS, Neher SB. Endoplasmic reticulum quality control in lipoprotein metabolism. Mol Cell Endocrinol 2019; 498:110547. [PMID: 31442546 PMCID: PMC6814580 DOI: 10.1016/j.mce.2019.110547] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 12/26/2022]
Abstract
Lipids play a critical role in energy metabolism, and a suite of proteins is required to deliver lipids to tissues. Several of these proteins require an intricate endoplasmic reticulum (ER) quality control (QC) system and unique secondary chaperones for folding. Key examples include apolipoprotein B (apoB), which is the primary scaffold for many lipoproteins, dimeric lipases, which hydrolyze triglycerides from circulating lipoproteins, and the low-density lipoprotein receptor (LDLR), which clears cholesterol-rich lipoproteins from the circulation. ApoB requires specialized proteins for lipidation, dimeric lipases lipoprotein lipase (LPL) and hepatic lipase (HL) require a transmembrane maturation factor for secretion, and the LDLR requires several specialized, domain-specific chaperones. Deleterious mutations in these proteins or their chaperones may result in dyslipidemias, which are detrimental to human health. Here, we review the ER quality control systems that ensure secretion of apoB, LPL, HL, and LDLR with a focus on the specialized chaperones required by each protein.
Collapse
Affiliation(s)
- Cari M Koerner
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, USA
| | - Benjamin S Roberts
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, USA
| | - Saskia B Neher
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
45
|
Nimonkar AV, Weldon S, Godbout K, Panza D, Hanrahan S, Cubbon R, Xu F, Trauger JW, Gao J, Voznesensky A. A lipoprotein lipase-GPI-anchored high-density lipoprotein-binding protein 1 fusion lowers triglycerides in mice: Implications for managing familial chylomicronemia syndrome. J Biol Chem 2019; 295:2900-2912. [PMID: 31645434 PMCID: PMC7062184 DOI: 10.1074/jbc.ra119.011079] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/15/2019] [Indexed: 02/04/2023] Open
Abstract
Lipoprotein lipase (LPL) is central to triglyceride metabolism. Severely compromised LPL activity causes familial chylomicronemia syndrome (FCS), which is associated with very high plasma triglyceride levels and increased risk of life-threatening pancreatitis. Currently, no approved pharmacological intervention can acutely lower plasma triglycerides in FCS. Low yield, high aggregation, and poor stability of recombinant LPL have thus far prevented development of enzyme replacement therapy. Recently, we showed that LPL monomers form 1:1 complexes with the LPL transporter glycosylphosphatidylinositol-anchored high-density lipoprotein–binding protein 1 (GPIHBP1) and solved the structure of the complex. In the present work, we further characterized the monomeric LPL/GPIHBP1 complex and its derivative, the LPL–GPIHBP1 fusion protein, with the goal of contributing to the development of an LPL enzyme replacement therapy. Fusion of LPL to GPIHBP1 increased yields of recombinant LPL, prevented LPL aggregation, stabilized LPL against spontaneous inactivation, and made it resistant to inactivation by the LPL antagonists angiopoietin-like protein 3 (ANGPTL3) or ANGPTL4. The high stability of the fusion protein enabled us to identify LPL amino acids that interact with ANGPTL4. Additionally, the LPL–GPIHBP1 fusion protein exhibited high enzyme activity in in vitro assays. Importantly, both intravenous and subcutaneous administrations of the fusion protein lowered triglycerides in several mouse strains without causing adverse effects. These results indicate that the LPL–GPIHBP1 fusion protein has potential for use as a therapeutic for managing FCS.
Collapse
Affiliation(s)
- Amitabh V Nimonkar
- Cardiovascular and Metabolic Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Stephen Weldon
- Novartis Biologics Center, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Kevin Godbout
- Novartis Biologics Center, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Darrell Panza
- Cardiovascular and Metabolic Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Susan Hanrahan
- Cardiovascular and Metabolic Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Rose Cubbon
- Cardiovascular and Metabolic Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Fangmin Xu
- Protein Analytics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - John W Trauger
- Cardiovascular and Metabolic Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Jiaping Gao
- Cardiovascular and Metabolic Disease Area, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Andrei Voznesensky
- Novartis Biologics Center, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139.
| |
Collapse
|
46
|
Young SG, Fong LG, Beigneux AP, Allan CM, He C, Jiang H, Nakajima K, Meiyappan M, Birrane G, Ploug M. GPIHBP1 and Lipoprotein Lipase, Partners in Plasma Triglyceride Metabolism. Cell Metab 2019; 30:51-65. [PMID: 31269429 PMCID: PMC6662658 DOI: 10.1016/j.cmet.2019.05.023] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lipoprotein lipase (LPL), identified in the 1950s, has been studied intensively by biochemists, physiologists, and clinical investigators. These efforts uncovered a central role for LPL in plasma triglyceride metabolism and identified LPL mutations as a cause of hypertriglyceridemia. By the 1990s, with an outline for plasma triglyceride metabolism established, interest in triglyceride metabolism waned. In recent years, however, interest in plasma triglyceride metabolism has awakened, in part because of the discovery of new molecules governing triglyceride metabolism. One such protein-and the focus of this review-is GPIHBP1, a protein of capillary endothelial cells. GPIHBP1 is LPL's essential partner: it binds LPL and transports it to the capillary lumen; it is essential for lipoprotein margination along capillaries, allowing lipolysis to proceed; and it preserves LPL's structure and activity. Recently, GPIHBP1 was the key to solving the structure of LPL. These developments have transformed the models for intravascular triglyceride metabolism.
Collapse
Affiliation(s)
- Stephen G Young
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Loren G Fong
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Anne P Beigneux
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher M Allan
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cuiwen He
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Haibo Jiang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; School of Molecular Sciences, University of Western Australia, Crawley 6009, Australia
| | - Katsuyuki Nakajima
- Department of Clinical Laboratory Medicine, Gunma University Graduate School of Department of Medicine, Maebashi, Gunma 371-0805, Japan
| | - Muthuraman Meiyappan
- Discovery Therapeutics, Takeda Pharmaceutical Company Ltd., Cambridge, MA 02142, USA
| | - Gabriel Birrane
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, Copenhagen DK-2200, Denmark; Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK-2200, Denmark.
| |
Collapse
|