1
|
Xiao Q, Wang J, Wang L, Ding H. APOA1/C3/A4/A5 Gene Cluster at 11q23.3 and Lipid Metabolism Disorders: From Epigenetic Mechanisms to Clinical Practices. Biomedicines 2024; 12:1224. [PMID: 38927431 PMCID: PMC11201263 DOI: 10.3390/biomedicines12061224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
The APOA1/C3/A4/A5 cluster is an essential component in regulating lipoprotein metabolism and maintaining plasma lipid homeostasis. A genome-wide association analysis and Mendelian randomization have revealed potential associations between genetic variants within this cluster and lipid metabolism disorders, including hyperlipidemia and cardiovascular events. An enhanced understanding of the complexity of gene regulation has led to growing recognition regarding the role of epigenetic variation in modulating APOA1/C3/A4/A5 gene expression. Intensive research into the epigenetic regulatory patterns of the APOA1/C3/A4/A5 cluster will help increase our understanding of the pathogenesis of lipid metabolism disorders and facilitate the development of new therapeutic approaches. This review discusses the biology of how the APOA1/C3/A4/A5 cluster affects circulating lipoproteins and the current progress in the epigenetic regulation of the APOA1/C3/A4/A5 cluster.
Collapse
Affiliation(s)
- Qianqian Xiao
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.X.); (J.W.); (L.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jing Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.X.); (J.W.); (L.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Luyun Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.X.); (J.W.); (L.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.X.); (J.W.); (L.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| |
Collapse
|
2
|
Calhoon D, Sang L, Bezwada D, Kim N, Basu A, Hsu SC, Pimentel A, Brooks B, La K, Serrano AP, Cassidy DL, Cai L, Toffessi-Tcheuyap V, Margulis V, Cai F, Brugarolas J, Weiss RJ, DeBerardinis RJ, Birsoy K, Garcia-Bermudez J. Glycosaminoglycan-mediated lipoprotein uptake protects cancer cells from ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593939. [PMID: 38765991 PMCID: PMC11101130 DOI: 10.1101/2024.05.13.593939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Lipids are essential for tumours because of their structural, energetic, and signaling roles. While many cancer cells upregulate lipid synthesis, growing evidence suggests that tumours simultaneously intensify the uptake of circulating lipids carried by lipoproteins. Which mechanisms promote the uptake of extracellular lipids, and how this pool of lipids contributes to cancer progression, are poorly understood. Here, using functional genetic screens, we find that lipoprotein uptake confers resistance to lipid peroxidation and ferroptotic cell death. Lipoprotein supplementation robustly inhibits ferroptosis across numerous cancer types. Mechanistically, cancer cells take up lipoproteins through a pathway dependent on sulfated glycosaminoglycans (GAGs) linked to cell-surface proteoglycans. Tumour GAGs are a major determinant of the uptake of both low and high density lipoproteins. Impairment of glycosaminoglycan synthesis or acute degradation of surface GAGs decreases the uptake of lipoproteins, sensitizes cells to ferroptosis and reduces tumour growth in mice. We also find that human clear cell renal cell carcinomas, a distinctively lipid-rich tumour type, display elevated levels of lipoprotein-derived antioxidants and the GAG chondroitin sulfate than non-malignant human kidney. Altogether, our work identifies lipoprotein uptake as an essential anti-ferroptotic mechanism for cancer cells to overcome lipid oxidative stress in vivo, and reveals GAG biosynthesis as an unexpected mediator of this process.
Collapse
Affiliation(s)
- Dylan Calhoon
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- These authors contributed equally to this work
| | - Lingjie Sang
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- These authors contributed equally to this work
| | - Divya Bezwada
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nathaniel Kim
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amrita Basu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Sheng-Chieh Hsu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anastasia Pimentel
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bailey Brooks
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Konnor La
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Ana Paulina Serrano
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel L Cassidy
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ling Cai
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Peter O’Donnell School of Public Health, University of Texas Southwestern, Dallas, TX, USA
| | - Vanina Toffessi-Tcheuyap
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Feng Cai
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James Brugarolas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ryan J Weiss
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kivanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Javier Garcia-Bermudez
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
Matsuura H, Akahane S, Kaido T, Kamijo T, Sakamoto K, Yamauchi K. Apolipoprotein E isoforms and their Cys-thiol modifications impact LRP1-mediated metabolism of triglyceride-rich lipoproteins. FEBS Lett 2024; 598:347-362. [PMID: 38279679 DOI: 10.1002/1873-3468.14803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/14/2023] [Accepted: 12/15/2023] [Indexed: 01/28/2024]
Abstract
The low-density lipoprotein (LDL) receptor-related protein (LRP)1 participates in the metabolism of apolipoprotein (apo) E-containing lipoproteins (apoE-LP). We investigated the effects of modifications of cysteine (Cys)-thiol of apoE on LRP1-mediated metabolism. Among the three isoforms, apoE2-LP exhibited the lowest affinity for LRP1 but was significantly catabolized, whereas apoE4-LP was sufficiently bound to LRP1 but showed the lowest catabolic capability. The reduction enhanced the binding and suppressed the catabolism of apoE3-LP, but had no effect on apoE2-LP. The formation of disulfide-linked complexes with apoAII suppressed binding, but enhanced the catabolism of apoE2-LP. Redox modifications of apoE-Cys-thiol may modulate the LRP1-mediated metabolism of apoE2- or apoE3-LP, but not apoE4-LP. The failure of this function may be involved in the pathophysiology of dyslipidemia.
Collapse
Affiliation(s)
- Hiroto Matsuura
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Shogo Akahane
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Takahiro Kaido
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Tomu Kamijo
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Kenta Sakamoto
- Department of Laboratory Medicine, University of Yamanashi Hospital, Japan
| | - Kazuyoshi Yamauchi
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
- Department of Biomedical Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| |
Collapse
|
4
|
Li M, Pedersen LC, Xu D. Targeting heparan sulfate-protein interactions with oligosaccharides and monoclonal antibodies. Front Mol Biosci 2023; 10:1194293. [PMID: 37275960 PMCID: PMC10235622 DOI: 10.3389/fmolb.2023.1194293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/10/2023] [Indexed: 06/07/2023] Open
Abstract
Heparan sulfate-binding proteins (HSBPs) are structurally diverse extracellular and membrane attached proteins that interact with HS under normal physiological conditions. Interactions with HS offer an additional level of control over the localization and function of HSBPs, which enables them to behave in a more refined manner. Because all cell signaling events start at the cell membrane, and cell-cell communication relies on translocation of soluble factors across the extracellular matrix, HS occupies an apical position in cellular signal transduction by interacting with hundreds of growth factors, cytokines, chemokines, enzymes, enzyme inhibitors, receptors and adhesion molecules. These extracellular and membrane proteins can play important roles in physiological and pathological conditions. For most HS-binding proteins, the interaction with HS represents an essential element in regulating their normal physiological functions. Such dependence on HS suggests that manipulating HS-protein interactions could be explored as a therapeutic strategy to selectively antagonize/activate HS-binding proteins. In this review, we will discuss current understanding of the diverse nature of HS-HSBP interactions, and the latest advancements in targeting the HS-binding site of HSBPs using structurally-defined HS oligosaccharides and monoclonal antibodies.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, NY, United States
| | - Lars C. Pedersen
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Ding Xu
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, NY, United States
| |
Collapse
|
5
|
Zhou J, Wang Y, Huang G, Yang M, Zhu Y, Jin C, Jing D, Ji K, Shi Y. LilrB3 is a putative cell surface receptor of APOE4. Cell Res 2023; 33:116-130. [PMID: 36588123 PMCID: PMC9892561 DOI: 10.1038/s41422-022-00759-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 01/03/2023] Open
Abstract
The three isoforms of apolipoprotein E (APOE2, APOE3, and APOE4) only differ in two amino acid positions but exert quite different immunomodulatory effects. The underlying mechanism of such APOE isoform dependence remains enigmatic. Here we demonstrate that APOE4, but not APOE2, specifically interacts with the leukocyte immunoglobulin-like receptor B3 (LilrB3). Two discrete immunoglobin-like domains of the LilrB3 extracellular domain (ECD) recognize a positively charged surface patch on the N-terminal domain (NTD) of APOE4. The atomic structure reveals how two APOE4 molecules specifically engage two LilrB3 molecules, bringing their intracellular signaling motifs into close proximity through formation of a hetero-tetrameric complex. Consistent with our biochemical and structural analyses, APOE4, but not APOE2, activates human microglia cells (HMC3) into a pro-inflammatory state in a LilrB3-dependent manner. Together, our study identifies LilrB3 as a putative immune cell surface receptor for APOE4, but not APOE2, and may have implications for understanding the biological functions as well as disease relevance of the APOE isoforms.
Collapse
Affiliation(s)
- Jiayao Zhou
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Yumeng Wang
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Min Yang
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yumin Zhu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui, China
| | - Chen Jin
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Dan Jing
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Kai Ji
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yigong Shi
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
6
|
Schultheis N, Becker R, Berhanu G, Kapral A, Roseman M, Shah S, Connell A, Selleck S. Regulation of autophagy, lipid metabolism, and neurodegenerative pathology by heparan sulfate proteoglycans. Front Genet 2023; 13:1012706. [PMID: 36699460 PMCID: PMC9870329 DOI: 10.3389/fgene.2022.1012706] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Heparan sulfate modified proteins or proteoglycans (HSPGs) are an abundant class of cell surface and extracellular matrix molecules. They serve important co-receptor functions in the regulation of signaling as well as membrane trafficking. Many of these activities directly affect processes associated with neurodegeneration including uptake and export of Tau protein, disposition of Amyloid Precursor Protein-derived peptides, and regulation of autophagy. In this review we focus on the impact of HSPGs on autophagy, membrane trafficking, mitochondrial quality control and biogenesis, and lipid metabolism. Disruption of these processes are a hallmark of Alzheimer's disease (AD) and there is evidence that altering heparan sulfate structure and function could counter AD-associated pathological processes. Compromising presenilin function in several systems has provided instructive models for understanding the molecular and cellular underpinnings of AD. Disrupting presenilin function produces a constellation of cellular deficits including accumulation of lipid, disruption of autophagosome to lysosome traffic and reduction in mitochondrial size and number. Inhibition of heparan sulfate biosynthesis has opposing effects on all these cellular phenotypes, increasing mitochondrial size, stimulating autophagy flux to lysosomes, and reducing the level of intracellular lipid. These findings suggest a potential mechanism for countering pathology found in AD and related disorders by altering heparan sulfate structure and influencing cellular processes disrupted broadly in neurodegenerative disease. Vertebrate and invertebrate model systems, where the cellular machinery of autophagy and lipid metabolism are conserved, continue to provide important translational guideposts for designing interventions that address the root cause of neurodegenerative pathology.
Collapse
Affiliation(s)
- Nicholas Schultheis
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Robert Becker
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Gelila Berhanu
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Alexander Kapral
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Matthew Roseman
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Shalini Shah
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Alyssa Connell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
| | - Scott Selleck
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
7
|
Zhang X, Zhao Y, Liu L, He Y. Syndecan-1: A Novel Diagnostic and Therapeutic Target in Liver Diseases. Curr Drug Targets 2023; 24:1155-1165. [PMID: 37957867 DOI: 10.2174/0113894501250057231102061624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/12/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023]
Abstract
Syndecan-1 (SDC-1), known as a coreceptor of various growth factors or an integrin binding partner, regulates various cell behaviours. Under certain pathological conditions, SDC-1 is shed from the cell surface and plays a protective or pathogenic role in various diseases. In the liver, SDC-1 is highly expressed in hepatocytes, where it is localized on the basolateral surface. It is critical to the cellular and molecular functions of hepatocytes, including their attachment to hepatitis viruses. Previous studies have reported that SDC-1 may function as a novel and promising diagnostic and therapeutic marker for various liver diseases, such as drug-induced liver injury, liver fibrosis, and liver cancer. In this review, we summarize related research and highlight the mechanisms by which SDC-1 participates in the pathogenesis of liver diseases, as well as its potential diagnostic and therapeutic applications. This review is expected to lay the foundation for further therapeutic strategies to target SDC-1 in liver diseases.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Infectious Diseases, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National Regional Infectious Diseases Center Co-constructed by National Health Commission of PRC and People's Government of Shaanxi Province, Xi'an, China
| | - Yalei Zhao
- Department of Infectious Diseases, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National Regional Infectious Diseases Center Co-constructed by National Health Commission of PRC and People's Government of Shaanxi Province, Xi'an, China
| | - Liangru Liu
- Department of Infectious Diseases, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National Regional Infectious Diseases Center Co-constructed by National Health Commission of PRC and People's Government of Shaanxi Province, Xi'an, China
| | - Yingli He
- Department of Infectious Diseases, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National Regional Infectious Diseases Center Co-constructed by National Health Commission of PRC and People's Government of Shaanxi Province, Xi'an, China
| |
Collapse
|
8
|
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: 10] [Impact Index Per Article: 5.0] [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
|
9
|
Impaired mitophagy in Sanfilippo a mice causes hypertriglyceridemia and brown adipose tissue activation. J Biol Chem 2022; 298:102159. [PMID: 35750212 PMCID: PMC9364035 DOI: 10.1016/j.jbc.2022.102159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/25/2022] Open
Abstract
Lysosomal storage diseases result in various developmental and physiological complications, including cachexia. To study the causes for the negative energy balance associated with cachexia, we assessed the impact of sulfamidase deficiency and heparan sulfate storage on energy homeostasis and metabolism in a mouse model of type IIIa mucopolysaccharidosis (MPS IIIa, Sanfilippo A syndrome). At 12-weeks of age, MPS IIIa mice exhibited fasting and postprandial hypertriglyceridemia compared with wildtype mice, with a reduction of white and brown adipose tissues. Partitioning of dietary [3H]triolein showed a marked increase in intestinal uptake and secretion, whereas hepatic production and clearance of triglyceride-rich lipoproteins did not differ from wildtype controls. Uptake of dietary triolein was also elevated in brown adipose tissue (BAT), and notable increases in beige adipose tissue occurred, resulting in hyperthermia, hyperphagia, hyperdipsia, and increased energy expenditure. Furthermore, fasted MPS IIIa mice remained hyperthermic when subjected to low temperature but became cachexic and profoundly hypothermic when treated with a lipolytic inhibitor. We demonstrated that the reliance on increased lipid fueling of BAT was driven by a reduced ability to generate energy from stored lipids within the depot. These alterations arose from impaired autophagosome-lysosome fusion, resulting in increased mitochondria content in beige and BAT. Finally, we show that increased mitochondria content in BAT and postprandial dyslipidemia was partially reversed upon 5-week treatment with recombinant sulfamidase. We hypothesize that increased BAT activity and persistent increases in energy demand in MPS IIIa mice contribute to the negative energy balance observed in patients with MPS IIIa.
Collapse
|
10
|
Ramms B, Patel S, Sun X, Pessentheiner AR, Ducasa GM, Mullick AE, Lee RG, Crooke RM, Tsimikas S, Witztum JL, Gordts PL. Interventional hepatic apoC-III knockdown improves atherosclerotic plaque stability and remodeling by triglyceride lowering. JCI Insight 2022; 7:e158414. [PMID: 35653195 PMCID: PMC9310539 DOI: 10.1172/jci.insight.158414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Apolipoprotein C-III (apoC-III) is a critical regulator of triglyceride metabolism and correlates positively with hypertriglyceridemia and cardiovascular disease (CVD). It remains unclear if therapeutic apoC-III lowering reduces CVD risk and if the CVD correlation depends on the lipid-lowering or antiinflammatory properties. We determined the impact of interventional apoC-III lowering on atherogenesis using an apoC-III antisense oligonucleotide (ASO) in 2 hypertriglyceridemic mouse models where the intervention lowers plasma triglycerides and in a third lipid-refractory model. On a high-cholesterol Western diet apoC-III ASO treatment did not alter atherosclerotic lesion size but did attenuate advanced and unstable plaque development in the triglyceride-responsive mouse models. No lesion size or composition improvement was observed with apoC-III ASO in the lipid-refractory mice. To circumvent confounding effects of continuous high-cholesterol feeding, we tested the impact of interventional apoC-III lowering when switching to a cholesterol-poor diet after 12 weeks of Western diet. In this diet switch regimen, apoC-III ASO treatment significantly reduced plasma triglycerides, atherosclerotic lesion progression, and necrotic core area and increased fibrous cap thickness in lipid-responsive mice. Again, apoC-III ASO treatment did not alter triglyceride levels, lesion development, and lesion composition in lipid-refractory mice after the diet switch. Our findings suggest that interventional apoC-III lowering might be an effective strategy to reduce atherosclerosis lesion size and improve plaque stability when lipid lowering is achieved.
Collapse
Affiliation(s)
- Bastian Ramms
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Chemistry, Biochemistry I, Bielefeld University, Bielefeld, Germany
| | - Sohan Patel
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Xiaoli Sun
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Pharmacology, Mays Cancer Center, Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | - G. Michelle Ducasa
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | | | | | | | - Sotirios Tsimikas
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Joseph L. Witztum
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Philip L.S.M. Gordts
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
11
|
Apolipoprotein A5, a unique modulator of fasting and postprandial triglycerides. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159185. [DOI: 10.1016/j.bbalip.2022.159185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/30/2022] [Accepted: 05/13/2022] [Indexed: 11/19/2022]
|
12
|
Ahmed S, Pande AH, Sharma SS. Therapeutic potential of ApoE-mimetic peptides in CNS disorders: Current perspective. Exp Neurol 2022; 353:114051. [DOI: 10.1016/j.expneurol.2022.114051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 02/07/2023]
|
13
|
Heeren J, Scheja L. Metabolic-associated fatty liver disease and lipoprotein metabolism. Mol Metab 2021; 50:101238. [PMID: 33892169 PMCID: PMC8324684 DOI: 10.1016/j.molmet.2021.101238] [Citation(s) in RCA: 244] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/01/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease, or as recently proposed 'metabolic-associated fatty liver disease' (MAFLD), is characterized by pathological accumulation of triglycerides and other lipids in hepatocytes. This common disease can progress from simple steatosis to steatohepatitis, and eventually end-stage liver diseases. MAFLD is closely related to disturbances in systemic energy metabolism, including insulin resistance and atherogenic dyslipidemia. SCOPE OF REVIEW The liver is the central organ in lipid metabolism by secreting very low density lipoproteins (VLDL) and, on the other hand, by internalizing fatty acids and lipoproteins. This review article discusses recent research addressing hepatic lipid synthesis, VLDL production, and lipoprotein internalization as well as the lipid exchange between adipose tissue and the liver in the context of MAFLD. MAJOR CONCLUSIONS Liver steatosis in MAFLD is triggered by excessive hepatic triglyceride synthesis utilizing fatty acids derived from white adipose tissue (WAT), de novo lipogenesis (DNL) and endocytosed remnants of triglyceride-rich lipoproteins. In consequence of high hepatic lipid content, VLDL secretion is enhanced, which is the primary cause of complex dyslipidemia typical for subjects with MAFLD. Interventions reducing VLDL secretory capacity attenuate dyslipidemia while they exacerbate MAFLD, indicating that the balance of lipid storage versus secretion in hepatocytes is a critical parameter determining disease outcome. Proof of concept studies have shown that promoting lipid storage and energy combustion in adipose tissues reduces hepatic lipid load and thus ameliorates MAFLD. Moreover, hepatocellular triglyceride synthesis from DNL and WAT-derived fatty acids can be targeted to treat MAFLD. However, more research is needed to understand how individual transporters, enzymes, and their isoforms affect steatosis and dyslipidemia in vivo, and whether these two aspects of MAFLD can be selectively treated. Processing of cholesterol-enriched lipoproteins appears less important for steatosis. It may, however, modulate inflammation and consequently MAFLD progression.
Collapse
Affiliation(s)
- Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
14
|
The Importance of Lipoprotein Lipase Regulation in Atherosclerosis. Biomedicines 2021; 9:biomedicines9070782. [PMID: 34356847 PMCID: PMC8301479 DOI: 10.3390/biomedicines9070782] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [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
|
15
|
Cabodevilla AG, Tang S, Lee S, Mullick AE, Aleman JO, Hussain MM, Sessa WC, Abumrad NA, Goldberg IJ. Eruptive xanthoma model reveals endothelial cells internalize and metabolize chylomicrons, leading to extravascular triglyceride accumulation. J Clin Invest 2021; 131:e145800. [PMID: 34128469 PMCID: PMC8203467 DOI: 10.1172/jci145800] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Although tissue uptake of fatty acids from chylomicrons is primarily via lipoprotein lipase (LpL) hydrolysis of triglycerides (TGs), studies of patients with genetic LpL deficiency suggest additional pathways deliver dietary lipids to tissues. Despite an intact endothelial cell (EC) barrier, hyperchylomicronemic patients accumulate chylomicron-derived lipids within skin macrophages, leading to the clinical finding eruptive xanthomas. We explored whether an LpL-independent pathway exists for transfer of circulating lipids across the EC barrier. We found that LpL-deficient mice had a marked increase in aortic EC lipid droplets before and after a fat gavage. Cultured ECs internalized chylomicrons, which were hydrolyzed within lysosomes. The products of this hydrolysis fueled lipid droplet biogenesis in ECs and triggered lipid accumulation in cocultured macrophages. EC chylomicron uptake was inhibited by competition with HDL and knockdown of the scavenger receptor-BI (SR-BI). In vivo, SR-BI knockdown reduced TG accumulation in aortic ECs and skin macrophages of LpL-deficient mice. Thus, ECs internalize chylomicrons, metabolize them in lysosomes, and either store or release their lipids. This latter process may allow accumulation of TGs within skin macrophages and illustrates a pathway that might be responsible for creation of eruptive xanthomas.
Collapse
Affiliation(s)
- Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Songtao Tang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Sungwoon Lee
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jose O Aleman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - M Mahmood Hussain
- Diabetes and Obesity Center, NYU-Long Island School of Medicine, Mineola, New York, USA
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nada A Abumrad
- Nutritional Sciences, Department of Medicine and Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
16
|
Wolkowicz P, White CR, Anantharamaiah GM. Apolipoprotein Mimetic Peptides: An Emerging Therapy against Diabetic Inflammation and Dyslipidemia. Biomolecules 2021; 11:biom11050627. [PMID: 33922449 PMCID: PMC8146922 DOI: 10.3390/biom11050627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity has achieved epidemic status in the United States, resulting in an increase in type 2 diabetes mellitus, dyslipidemia, and cardiovascular disease. Numerous studies have shown that inflammation plays a key role in the development of insulin resistance and diabetic complications. HDL cholesterol levels are inversely associated with coronary heart disease in humans. The beneficial effect of HDL is due, in part, to apolipoproteins A-I and E, which possess anti-inflammatory properties. The functional quality of HDL, however, may be reduced in the context of diabetes. Thus, raising levels of functional HDL is an important target for reducing inflammation and diabetic complications. Apo A-I possesses eight alpha-helical sequences, most of which form class A amphipathic helical structures. Peptides belonging to this class inhibit atherogenesis in several mouse models. Additional peptides based on structural components of apoE have been shown to mediate a rapid clearance of atherogenic lipoproteins in dyslipidemic mice. In this review, we discuss the efficacy of apolipoprotein mimetic peptides in improving lipoprotein function, reducing inflammation, and reversing insulin resistance and cardiometabolic disease processes in diabetic animals.
Collapse
|
17
|
Wolska A, Reimund M, Sviridov DO, Amar MJ, Remaley AT. Apolipoprotein Mimetic Peptides: Potential New Therapies for Cardiovascular Diseases. Cells 2021; 10:597. [PMID: 33800446 PMCID: PMC8000854 DOI: 10.3390/cells10030597] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
Since the seminal breakthrough of treating diabetic patients with insulin in the 1920s, there has been great interest in developing other proteins and their peptide mimetics as therapies for a wide variety of other medical disorders. Currently, there are at least 60 different peptides that have been approved for human use and over 150 peptides that are in various stages of clinical development. Peptides mimetic of the major proteins on lipoproteins, namely apolipoproteins, have also been developed first as tools for understanding apolipoprotein structure and more recently as potential therapeutics. In this review, we discuss the biochemistry, peptide mimetics design and clinical trials for peptides based on apoA-I, apoE and apoC-II. We primarily focus on applications of peptide mimetics related to cardiovascular diseases. We conclude with a discussion on the limitations of peptides as therapeutic agents and the challenges that need to be overcome before apolipoprotein mimetic peptides can be developed into new drugs.
Collapse
Affiliation(s)
- Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (M.R.); (D.O.S.); (M.J.A.); (A.T.R.)
| | | | | | | | | |
Collapse
|
18
|
Gómez Toledo A, Sorrentino JT, Sandoval DR, Malmström J, Lewis NE, Esko JD. A Systems View of the Heparan Sulfate Interactome. J Histochem Cytochem 2021; 69:105-119. [PMID: 33494649 PMCID: PMC7841697 DOI: 10.1369/0022155420988661] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022] Open
Abstract
Heparan sulfate proteoglycans consist of a small family of proteins decorated with one or more covalently attached heparan sulfate glycosaminoglycan chains. These chains have intricate structural patterns based on the position of sulfate groups and uronic acid epimers, which dictate their ability to engage a large repertoire of heparan sulfate-binding proteins, including extracellular matrix proteins, growth factors and morphogens, cytokines and chemokines, apolipoproteins and lipases, adhesion and growth factor receptors, and components of the complement and coagulation system. This review highlights recent progress in the characterization of the so-called "heparan sulfate interactome," with a major focus on systems-wide strategies as a tool for discovery and characterization of this subproteome. In addition, we compiled all heparan sulfate-binding proteins reported in the literature to date and grouped them into a few major functional classes by applying a networking approach.
Collapse
Affiliation(s)
- Alejandro Gómez Toledo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
- Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, Lund, Sweden
| | - James T Sorrentino
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, California
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | - Daniel R Sandoval
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
| | - Johan Malmström
- Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, California
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
19
|
Jin W, Zhang F, Linhardt RJ. Glycosaminoglycans in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1325:189-204. [PMID: 34495536 DOI: 10.1007/978-3-030-70115-4_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glycosaminoglycans (GAGs) are linear polysaccharides that consist of alternating disaccharides sequences of uronic acids and/or galactose hexamino sugars most of which are sulfated. GAGs are ubiquitously expressed on the cell surface, in the intracellular milieu and in the extracellular matrix of all animal cells. Thus, GAGs exhibit many essential roles in a variety of physiological and pathological processes. The targets of GAGs are GAG-binding proteins and related proteins that are of significant interest to both the academic community and in the pharmaceutical industry. In this review, the structures of GAGs, their binding proteins, and analogs are presented that further the development of GAGs and their analogs for the treatment of neurodegenerative diseases agents.
Collapse
Affiliation(s)
- Weihua Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China.,Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA. .,Department of Biological Science, Departments of Chemistry and Chemical Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.
| |
Collapse
|
20
|
Enikanolaiye A, Ruston J, Zeng R, Taylor C, Schrock M, Buchovecky CM, Shendure J, Acar E, Justice MJ. Suppressor mutations in Mecp2-null mice implicate the DNA damage response in Rett syndrome pathology. Genome Res 2020; 30:540-552. [PMID: 32317254 PMCID: PMC7197480 DOI: 10.1101/gr.258400.119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/20/2020] [Indexed: 12/31/2022]
Abstract
Mutations in X-linked methyl-CpG-binding protein 2 (MECP2) cause Rett syndrome (RTT). To identify functional pathways that could inform therapeutic entry points, we carried out a genetic screen for secondary mutations that improved phenotypes in Mecp2/Y mice after mutagenesis with N-ethyl-N-nitrosourea (ENU). Here, we report the isolation of 106 founder animals that show suppression of Mecp2-null traits from screening 3177 Mecp2/Y genomes. Whole-exome sequencing, genetic crosses, and association analysis identified 22 candidate genes. Additional lesions in these candidate genes or pathway components associate variant alleles with phenotypic improvement in 30 lines. A network analysis shows that 63% of the genes cluster into the functional categories of transcriptional repression, chromatin modification, or DNA repair, delineating a pathway relationship with MECP2. Many mutations lie in genes that modulate synaptic signaling or lipid homeostasis. Mutations in genes that function in the DNA damage response (DDR) also improve phenotypes in Mecp2/Y mice. Association analysis was successful in resolving combinatorial effects of multiple loci. One line, which carries a suppressor mutation in a gene required for cholesterol synthesis, Sqle, carries a second mutation in retinoblastoma binding protein 8, endonuclease (Rbbp8, also known as CtIP), which regulates a DDR choice in double-stranded break (DSB) repair. Cells from Mecp2/Y mice have increased DSBs, so this finding suggests that the balance between homology-directed repair and nonhomologous end joining is important for neuronal cells. In this and other lines, two suppressor mutations confer greater improvement than one alone, suggesting that combination therapies could be effective in RTT.
Collapse
Affiliation(s)
- Adebola Enikanolaiye
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Julie Ruston
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Rong Zeng
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Christine Taylor
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Marijke Schrock
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Christie M Buchovecky
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, Washington 98195, USA
- Howard Hughes Medical Institute, Seattle, Washington 98195, USA
| | - Elif Acar
- The Centre for Phenogenomics, Toronto, Ontario, M5T 3H7, Canada
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - Monica J Justice
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- The Centre for Phenogenomics, Toronto, Ontario, M5T 3H7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
21
|
Takanashi M, Kimura T, Li C, Tanaka M, Matsuhashi A, Yoshida H, Noda A, Xu P, Takase S, Okazaki S, Iizuka Y, Kumagai H, Ikeda Y, Gotoda T, Takahashi M, Yagyu H, Ishibashi S, Yamauchi T, Kadowaki T, Liang G, Okazaki H. Critical Role of SREBP-1c Large-VLDL Pathway in Environment-Induced Hypertriglyceridemia of Apo AV Deficiency. Arterioscler Thromb Vasc Biol 2020; 39:373-386. [PMID: 30700132 DOI: 10.1161/atvbaha.118.311931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective- APOA5 variants are strongly associated with hypertriglyceridemia, as well as increased risks of cardiovascular disease and acute pancreatitis. Hypertriglyceridemia in apo AV dysfunction often aggravates by environmental factors such as high-carbohydrate diets or aging. To date, the molecular mechanisms by which these environmental factors induce hypertriglyceridemia are poorly defined, leaving the high-risk hypertriglyceridemia condition undertreated. Previously, we reported that LXR (liver X receptor)-SREBP (sterol regulatory element-binding protein)-1c pathway regulates large-VLDL (very low-density lipoprotein) production induced by LXR agonist. However, the pathophysiological relevance of the finding remains unknown. Approach and Results- Here, we reconstitute the environment-induced hypertriglyceridemia phenotype of human APOA5 deficiency in Apoa5-/- mice and delineate the role of SREBP-1c in vivo by generating Apoa5-/- ;Srebp-1c-/- mice. The Apoa5-/- mice, which showed moderate hypertriglyceridemia on a chow diet, developed severe hypertriglyceridemia on high-carbohydrate feeding or aging as seen in patients with human apo AV deficiency. These responses were nearly completely abolished in the Apoa5-/- ;Srebp-1c-/- mice. Further mechanistic studies revealed that in response to these environmental factors, SREBP-1c was activated to increase triglyceride synthesis and to permit the incorporation of triglyceride into abnormally large-VLDL particles, which require apo AV for efficient clearance. Conclusions- Severe hypertriglyceridemia develops only when genetic factors (apo AV deficiency) and environmental effects (SREBP-1c activation) coexist. We demonstrate that the regulated production of large-sized VLDL particles via SREBP-1c determines plasma triglyceride levels in apo AV deficiency. Our findings explain the long-standing enigma of the late-onset hypertriglyceridemia phenotype of apo AV deficiency and suggest a new approach to treat hypertriglyceridemia by targeting genes that mediate environmental effects.
Collapse
Affiliation(s)
- Mikio Takanashi
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Takeshi Kimura
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Chengcheng Li
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Masaki Tanaka
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Ako Matsuhashi
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Hiroki Yoshida
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Akari Noda
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Pengfei Xu
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Satoru Takase
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Sachiko Okazaki
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Yoko Iizuka
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Hidetoshi Kumagai
- Department of Cardiovascular Medicine (H.K., Y. Ikeda), Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Japan
| | - Yuichi Ikeda
- Department of Cardiovascular Medicine (H.K., Y. Ikeda), Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Japan
| | - Takanari Gotoda
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (Manabu Takahashi, S.I.)
| | - Hiroaki Yagyu
- Department of Endocrinology and Metabolism, Mito Medical Center, Tsukuba University Hospital, Mito, Ibaraki, Japan (H. Yagyu)
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (Manabu Takahashi, S.I.)
| | - Toshimasa Yamauchi
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Takashi Kadowaki
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Guosheng Liang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX (G.L., H.O.)
| | - Hiroaki Okazaki
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX (G.L., H.O.)
| |
Collapse
|
22
|
Jana S, Zhang H, Lopaschuk GD, Freed DH, Sergi C, Kantor PF, Oudit GY, Kassiri Z. Disparate Remodeling of the Extracellular Matrix and Proteoglycans in Failing Pediatric Versus Adult Hearts. J Am Heart Assoc 2019; 7:e010427. [PMID: 30371322 PMCID: PMC6404896 DOI: 10.1161/jaha.118.010427] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background Dilated cardiomyopathy (DCM) is a common cause of heart failure in adult and pediatric patients, but the underlying mechanism may vary in adults and children, with few studies conducted to date. The objective of the present study was to determine whether differential remodeling of the extracellular matrix contributes to the differences between pediatric and adult DCM hearts. Methods and Results Explanted hearts were procured from adult (age, 46–61 years) and pediatric (age, 2–8) patients with DCM‐related heart failure and nonfailing control hearts. Fibrillar and nonfibrillar extracellular matrix (proteoglycans, glycosaminoglycans, glycoprotein), their regulatory enzymes (matrix metalloproteinases, disintegrin and metalloproteinases, and disintegrin and metalloproteinases with a thrombospondin domain), and their inhibitors (tissue inhibitor of metalloproteinases) were assessed. Pediatric DCM hearts exhibited less fibrosis compared with adult DCMs. Total glycosaminoglycans increased similarly in both DCM groups but exhibited a significantly lower affinity for transforming growth factor‐β in adult DCMs versus pediatric DCMs, resulting in increased bioavailability of transforming growth factor‐β1 and a significantly higher activity of the Smad2/3 pathway in adult DCMs. Glycosylated biglycan and versican, and cleaved thrombospondin‐1 increased in both DCMs. Protein expression of disintegrin and metalloproteinases with thrombospondin domains (‐1, ‐2, ‐4, ‐7) and disintegrin and metalloproteinases (‐12, ‐15, ‐17, ‐19) were altered differently in pediatric and adult control and failing hearts. Total matrix metalloproteinase activity increased in both DCMs. Tissue inhibitor of metalloproteinase levels were altered similarly with heart failure in both age groups, and only tissue inhibitor of metalloproteinase 3 decreased in both DCM groups. Conclusions Differential remodeling of glycosaminoglycans in pediatric DCMs versus adult DCMs could underlie the enhanced activation of the transforming growth factor‐β pathway, leading to more fibrosis in adult DCM hearts. The distinct remodeling of the fibrillar and nonfibrillar extracellular matrix between pediatric and adult DCM hearts highlights a distinct pathophysiological basis for these cohorts.
Collapse
Affiliation(s)
- Sayantan Jana
- 1 Department of Physiology Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Hao Zhang
- 2 Department of Medicine Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Gary D Lopaschuk
- 3 Department of Pediatrics Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Darren H Freed
- 1 Department of Physiology Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,5 Division of Cardiac Surgery Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Consolato Sergi
- 4 Department of Laboratory Medicine and Pathology Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Paul F Kantor
- 3 Department of Pediatrics Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Gavin Y Oudit
- 2 Department of Medicine Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Zamaneh Kassiri
- 1 Department of Physiology Faculty of Medicine and Dentistry University of Alberta Edmonton AB.,6 Cardiovascular Research Centre Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| |
Collapse
|
23
|
Anower-E-Khuda F, Singh G, Deng Y, Gordts PLSM, Esko JD. Triglyceride-rich lipoprotein binding and uptake by heparan sulfate proteoglycan receptors in a CRISPR/Cas9 library of Hep3B mutants. Glycobiology 2019; 29:582-592. [PMID: 31094413 PMCID: PMC6639542 DOI: 10.1093/glycob/cwz037] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/01/2019] [Accepted: 05/13/2019] [Indexed: 12/20/2022] Open
Abstract
Binding and uptake of triglyceride-rich lipoproteins (TRLs) in mice depend on heparan sulfate and the hepatic proteoglycan, syndecan-1 (SDC1). Alteration of glucosamine N-sulfation by deletion of glucosamine N-deacetylase-N-sulfotransferase 1 (Ndst1) and 2-O-sulfation of uronic acids by deletion of uronyl 2-O-sulfotransferase (Hs2st) led to diminished lipoprotein metabolism, whereas inactivation of glucosaminyl 6-O-sulfotransferase 1 (Hs6st1), which encodes one of the three 6-O-sulfotransferases, had little effect on lipoprotein binding. However, other studies have suggested that 6-O-sulfation may be important for TRL binding and uptake. In order to explain these discrepant findings, we used CRISPR/Cas9 gene editing to create a library of mutants in the human hepatoma cell line, Hep3B. Inactivation of EXT1 encoding the heparan sulfate copolymerase, NDST1 and HS2ST dramatically reduced binding of TRLs. Inactivation of HS6ST1 had no effect, but deletion of HS6ST2 reduced TRL binding. Compounding mutations in HS6ST1 and HS6ST2 did not exacerbate this effect indicating that HS6ST2 is the dominant 6-O-sulfotransferase and that binding of TRLs indeed depends on 6-O-sulfation of glucosamine residues. Uptake studies showed that TRL internalization was also affected in 6-O-sulfation deficient cells. Interestingly, genetic deletion of SDC1 only marginally impacted binding of TRLs but reduced TRL uptake to the same extent as treating the cells with heparin lyases. These findings confirm that SDC1 is the dominant endocytic proteoglycan receptor for TRLs in human Hep3B cells and that binding and uptake of TRLs depend on SDC1 and N- and 2-O-sulfation as well as 6-O-sulfation of heparan sulfate chains catalyzed by HS6ST2.
Collapse
Affiliation(s)
| | | | - Yiping Deng
- Department of Cellular and Molecular Medicine
- Juventas Cell Therapy Ltd, Beijing, China
| | - Philip L S M Gordts
- Department of Medicine
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
Affiliation(s)
- Gissette Reyes-Soffer
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Henry N Ginsberg
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
25
|
Cui Y, Cui XD, Xu M, Fang M, Cai MJ. Serum apolipoprotein C3 levels are negatively associated with hepatitis B virus DNA in HBeAg-negative chronic hepatitis B patients. Lipids Health Dis 2019; 18:138. [PMID: 31186008 PMCID: PMC6560873 DOI: 10.1186/s12944-019-1084-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/04/2019] [Indexed: 01/07/2023] Open
Abstract
Background Hepatitis B virus (HBV) infection remains a global health issue associated with substantial morbidity and mortality. Serum apolipoprotein C3 (ApoC3) and apolipoprotein A5 (ApoA5) levels were decreased in chronic hepatitis B (CHB) patients, however the relationship between ApoC3 or ApoA5 and HBV DNA load remains elusive. Methods A total of 384 CHB patients including 194 HBsAg(+) HBeAg(−) and 190 HBsAg(+) HBeAg(+) and 154 healthy individuals were recruited in our study. Serum levels of alanine aminotransferase (ALT), aspartate transaminase (AST), total cholesterol (Chol), triglycerides (TG), apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), high-density lipoproteins cholesterol (HDL-C), low-density lipoproteins cholesterol (LDL-C) and lipoprotein a (Lpa) were examined in an automatic biochemical analyzer. Apolipoprotein A5 (ApoA5) and apolipoprotein C3 (ApoC3) were detected via ELISA. Results Serum ApoA1, ApoB, ApoC3 and ApoA5 levels were reduced in CHB patients. In HBeAg(−) CHB patients, plasma ApoC3 levels were negatively associated with HBV DNA load (r = 0.219, P < 0.001). But no correlation between ApoA5 and HBV DNA load was observed in CHB patients. Conclusions These data showed that HBV infection inhibits lipid metabolism and ApoC3 is negatively associated with HBV DNA load in HBeAg (−) CHB patients. These findings provided new evidence about the link between ApoC3-related lipid metabolism and immune response.
Collapse
Affiliation(s)
- Yu Cui
- Department of Obstetrics, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Xiang-Dan Cui
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Xu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, No. 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Min Fang
- Department of Gynaecology, Qingdao Women's and Children's Hospital, Qingdao University, Qingdao, China
| | - Mei-Juan Cai
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, No. 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
26
|
Ramms B, Patel S, Nora C, Pessentheiner AR, Chang MW, Green CR, Golden GJ, Secrest P, Krauss RM, Metallo CM, Benner C, Alexander VJ, Witztum JL, Tsimikas S, Esko JD, Gordts PLSM. ApoC-III ASO promotes tissue LPL activity in the absence of apoE-mediated TRL clearance. J Lipid Res 2019; 60:1379-1395. [PMID: 31092690 PMCID: PMC6672034 DOI: 10.1194/jlr.m093740] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/10/2019] [Indexed: 11/21/2022] Open
Abstract
Hypertriglyceridemia results from accumulation of triglyceride (TG)-rich lipoproteins (TRLs) in the circulation and is associated with increased CVD risk. ApoC-III is an apolipoprotein on TRLs and a prominent negative regulator of TG catabolism. We recently established that in vivo apoC-III predominantly inhibits LDL receptor-mediated and LDL receptor-related protein 1-mediated hepatic TRL clearance and that apoC-III-enriched TRLs are preferentially cleared by syndecan-1 (SDC1). In this study, we determined the impact of apoE, a common ligand for all three receptors, on apoC-III metabolism using apoC-III antisense oligonucleotide (ASO) treatment in mice lacking apoE and functional SDC1 (Apoe−/−Ndst1f/fAlb-Cre+). ApoC-III ASO treatment significantly reduced plasma TG levels in Apoe−/−Ndst1f/fAlb-Cre+ mice without reducing hepatic VLDL production or improving hepatic TRL clearance. Further analysis revealed that apoC-III ASO treatment lowered plasma TGs in Apoe−/−Ndst1f/fAlb-Cre+ mice, which was associated with increased LPL activity in white adipose tissue in the fed state. Finally, clinical data confirmed that ASO-mediated lowering of APOC-III via volanesorsen can reduce plasma TG levels independent of the APOE isoform genotype. Our data indicate that apoE determines the metabolic impact of apoC-III as we establish that apoE is essential to mediate inhibition of TRL clearance by apoC-III and that, in the absence of functional apoE, apoC-III inhibits tissue LPL activity.
Collapse
Affiliation(s)
- Bastian Ramms
- Departments of Cellular and Molecular Medicine,University of California, San Diego, La Jolla, CA.,Medicine, University of California, San Diego, La Jolla, CA.,Department of Chemistry, Biochemistry I, Bielefeld University, Bielefeld, Germany
| | - Sohan Patel
- Medicine, University of California, San Diego, La Jolla, CA
| | - Chelsea Nora
- Medicine, University of California, San Diego, La Jolla, CA
| | | | - Max W Chang
- Departments of Cellular and Molecular Medicine,University of California, San Diego, La Jolla, CA
| | - Courtney R Green
- Bioengineering, University of California, San Diego, La Jolla, CA
| | - Gregory J Golden
- Departments of Cellular and Molecular Medicine,University of California, San Diego, La Jolla, CA.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA
| | - Patrick Secrest
- Departments of Cellular and Molecular Medicine,University of California, San Diego, La Jolla, CA
| | | | | | - Christopher Benner
- Departments of Cellular and Molecular Medicine,University of California, San Diego, La Jolla, CA
| | | | | | | | - Jeffrey D Esko
- Departments of Cellular and Molecular Medicine,University of California, San Diego, La Jolla, CA.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA
| | - Philip L S M Gordts
- Medicine, University of California, San Diego, La Jolla, CA .,Bioengineering, University of California, San Diego, La Jolla, CA
| |
Collapse
|
27
|
Morton AM, Furtado JD, Mendivil CO, Sacks FM. Dietary unsaturated fat increases HDL metabolic pathways involving apoE favorable to reverse cholesterol transport. JCI Insight 2019; 4:124620. [PMID: 30944249 DOI: 10.1172/jci.insight.124620] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND HDL that contains apolipoprotein E (apoE) is a subspecies especially active in steps in reverse cholesterol transport, a process that brings cholesterol from peripheral cells to the liver. Here, we studied the effect of dietary unsaturated fat compared with carbohydrate on the metabolism of HDL containing apoE. METHODS We enrolled 9 adults who were overweight or obese and had below-average HDL-cholesterol in a crossover study of a high-fat diet, primarily unsaturated, and a low-fat, high-carbohydrate diet. A metabolic tracer study was performed after each diet period. RESULTS Dietary fat increased the secretion, metabolism, and clearance of HDL subspecies containing apoE. Dietary fat increased the rate of clearance of large cholesterol-rich HDL containing apoE and increased their conversion to small HDL containing apoE, indicating selective cholesterol ester delivery to the liver. The high-unsaturated-fat diet did not affect the metabolism of HDL lacking apoE. CONCLUSION HDL containing apoE is a diet-responsive metabolic pathway that renders HDL more biologically active in reverse cholesterol transport. This may be a mechanism by which unsaturated fat protects against coronary heart disease. Protein-based HDL subspecies such as HDL containing apoE may be used to identify additional atheroprotective treatment targets not evident in the total HDL-cholesterol measurement. TRIAL REGISTRATION ClinicalTrials.gov NCT01399632. FUNDING NIH and the National Center for Advancing Translational Science.
Collapse
Affiliation(s)
- Allyson M Morton
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jeremy D Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Carlos O Mendivil
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Wight TN. A role for proteoglycans in vascular disease. Matrix Biol 2018; 71-72:396-420. [PMID: 29499356 PMCID: PMC6110991 DOI: 10.1016/j.matbio.2018.02.019] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022]
Abstract
The content of proteoglycans (PGs) is low in the extracellular matrix (ECM) of vascular tissue, but increases dramatically in all phases of vascular disease. Early studies demonstrated that glycosaminoglycans (GAGs) including chondroitin sulfate (CS), dermatan sulfate (DS), keratan sulfate (KS) and heparan sulfate (HS) accumulate in vascular lesions in both humans and in animal models in areas of the vasculature that are susceptible to disease initiation (such as at branch points) and are frequently coincident with lipid deposits. Later studies showed the GAGs were covalently attached to specific types of core proteins that accumulate in vascular lesions. These molecules include versican (CSPG), biglycan and decorin (DS/CSPGs), lumican and fibromodulin (KSPGs) and perlecan (HSPG), although other types of PGs are present, but in lesser quantities. While the overall molecular design of these macromolecules is similar, there is tremendous structural diversity among the different PG families creating multiple forms that have selective roles in critical events that form the basis of vascular disease. PGs interact with a variety of different molecules involved in disease pathogenesis. For example, PGs bind and trap serum components that accumulate in vascular lesions such as lipoproteins, amyloid, calcium, and clotting factors. PGs interact with other ECM components and regulate, in part, ECM assembly and turnover. PGs interact with cells within the lesion and alter the phenotypes of both resident cells and cells that invade the lesion from the circulation. A number of therapeutic strategies have been developed to target specific PGs involved in key pathways that promote vascular disease. This review will provide a historical perspective of this field of research and then highlight some of the evidence that defines the involvement of PGs and their roles in the pathogenesis of vascular disease.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, United States.
| |
Collapse
|
29
|
Zhao RR, Ackers-Johnson M, Stenzig J, Chen C, Ding T, Zhou Y, Wang P, Ng SL, Li PY, Teo G, Rudd PM, Fawcett JW, Foo RS. Targeting Chondroitin Sulfate Glycosaminoglycans to Treat Cardiac Fibrosis in Pathological Remodeling. Circulation 2018; 137:2497-2513. [DOI: 10.1161/circulationaha.117.030353] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/21/2017] [Indexed: 12/21/2022]
Abstract
Background:
Heart failure is a leading cause of mortality and morbidity, and the search for novel therapeutic approaches continues. In the monogenic disease mucopolysaccharidosis VI, loss-of-function mutations in arylsulfatase B lead to myocardial accumulation of chondroitin sulfate (CS) glycosaminoglycans, manifesting as myriad cardiac symptoms. Here, we studied changes in myocardial CS in nonmucopolysaccharidosis failing hearts and assessed its generic role in pathological cardiac remodeling.
Methods:
Healthy and diseased human and rat left ventricles were subjected to histological and immunostaining methods to analyze glycosaminoglycan distribution. Glycosaminoglycans were extracted and analyzed for quantitative and compositional changes with Alcian blue assay and liquid chromatography–mass spectrometry. Expression changes in 20 CS-related genes were studied in 3 primary human cardiac cell types and THP-1–derived macrophages under each of 9 in vitro stimulatory conditions. In 2 rat models of pathological remodeling induced by transverse aortic constriction or isoprenaline infusion, recombinant human arylsulfatase B (rhASB), clinically used as enzyme replacement therapy in mucopolysaccharidosis VI, was administered intravenously for 7 or 5 weeks, respectively. Cardiac function, myocardial fibrosis, and inflammation were assessed by echocardiography and histology. CS-interacting molecules were assessed with surface plasmon resonance, and a mechanism of action was verified in vitro.
Results:
Failing human hearts displayed significant perivascular and interstitial CS accumulation, particularly in regions of intense fibrosis. Relative composition of CS disaccharides remained unchanged. Transforming growth factor–β induced CS upregulation in cardiac fibroblasts. CS accumulation was also observed in both the pressure-overload and the isoprenaline models of pathological remodeling in rats. Early treatment with rhASB in the transverse aortic constriction model and delayed treatment in the isoprenaline model proved rhASB to be effective at preventing cardiac deterioration and augmenting functional recovery. Functional improvement was accompanied by reduced myocardial inflammation and overall fibrosis. Tumor necrosis factor–α was identified as a direct binding partner of CS glycosaminoglycan chains, and rhASB reduced tumor necrosis factor–α—induced inflammatory gene activation in vitro in endothelial cells and macrophages.
Conclusions:
CS glycosaminoglycans accumulate during cardiac pathological remodeling and mediate myocardial inflammation and fibrosis. rhASB targets CS effectively as a novel therapeutic approach for the treatment of heart failure.
Collapse
Affiliation(s)
- Rong-Rong Zhao
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
| | - Matthew Ackers-Johnson
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
| | - Justus Stenzig
- Genome Institute of Singapore (J.S., S.L.N., R.S.Y.F.)
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.S.)
| | - Chen Chen
- Bioprocessing Technology Institute (C.C., G.T., P.M.R.), Agency for Science, Technology and Research
| | - Tao Ding
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
| | - Yue Zhou
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
| | - Peipei Wang
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
| | - Shi Ling Ng
- Genome Institute of Singapore (J.S., S.L.N., R.S.Y.F.)
| | - Peter Y. Li
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
| | - Gavin Teo
- Bioprocessing Technology Institute (C.C., G.T., P.M.R.), Agency for Science, Technology and Research
| | - Pauline M. Rudd
- Bioprocessing Technology Institute (C.C., G.T., P.M.R.), Agency for Science, Technology and Research
- Glycoscience Group, National Institute for Bioprocessing, Research and Training, Dublin, Ireland (P.M.R.)
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, United Kingdom (J.W.F.)
| | - Roger S.Y. Foo
- Cardiovascular Research Institute, National University of Singapore (R.R.Z., M.A.-J., T.D., Y.Z., P.W., P.Y.L., R.S.Y.F.)
- Genome Institute of Singapore (J.S., S.L.N., R.S.Y.F.)
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Apolipoprotein (apo) C-III is a key player in triglyceride-rich lipoprotein metabolism and strongly associated with elevated plasma triglyceride levels. Several new studies added important insights on apoC-III and its physiological function confirming its promise as a valid therapeutic target. RECENT FINDINGS APOC3 is expressed in liver and intestine and regulates triglyceride-rich lipoprotein (TRL) catabolism and anabolism. The transcriptional regulation in both organs requires different regulatory elements. Clinical and preclinical studies established that apoC-III raises plasma triglyceride levels predominantly by inhibiting hepatic TRL clearance. Mechanistic insights into missense variants indicate accelerated renal clearance of apoC-III variants resulting in enhanced TRL catabolism. In contrast, an APOC3 gain-of-function variant enhances de novo lipogenesis and hepatic TRL production. Multiple studies confirmed the correlation between increased apoC-III levels and cardiovascular disease. This has opened up new therapeutic avenues allowing targeting of specific apoC-III properties in triglyceride metabolism. SUMMARY Novel in vivo models and APOC3 missense variants revealed unique mechanisms by which apoC-III inhibits TRL catabolism. Clinical trials with Volanesorsen, an APOC3 antisense oligonucleotide, report very promising lipid-lowering outcomes. However, future studies will need to address if acute apoC-III lowering will have the same clinical benefits as a life-long reduction.
Collapse
Affiliation(s)
- Bastian Ramms
- Department of Cellular and Molecular Medicine
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, San Diego, California, USA
- Department of Chemistry, Biochemistry I, Bielefeld University, Bielefeld, Germany
| | - Philip L S M Gordts
- Department of Cellular and Molecular Medicine
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, San Diego, California, USA
| |
Collapse
|
31
|
Gordts PLSM, Esko JD. The heparan sulfate proteoglycan grip on hyperlipidemia and atherosclerosis. Matrix Biol 2018; 71-72:262-282. [PMID: 29803939 DOI: 10.1016/j.matbio.2018.05.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
Heparan sulfate proteoglycans are found at the cell surface and in the extracellular matrix, where they interact with a plethora of proteins involved in lipid homeostasis and inflammation. Over the last decade, new insights have emerged regarding the mechanism and biological significance of these interactions in the context of cardiovascular disease. The majority of cardiovascular disease-related deaths are caused by complications of atherosclerosis, a disease that results in narrowing of the arterial lumen, thereby reducing blood flow to critical levels in vital organs, such as the heart and brain. Here, we discuss novel insights into how heparan sulfate proteoglycans modulate risk factors such as hyperlipidemia and inflammation that drive the initiation and progression of atherosclerotic plaques to their clinical critical endpoint.
Collapse
Affiliation(s)
- Philip L S M Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA.
| | - Jeffrey D Esko
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
32
|
Morton AM, Koch M, Mendivil CO, Furtado JD, Tjønneland A, Overvad K, Wang L, Jensen MK, Sacks FM. Apolipoproteins E and CIII interact to regulate HDL metabolism and coronary heart disease risk. JCI Insight 2018; 3:98045. [PMID: 29467335 DOI: 10.1172/jci.insight.98045] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/17/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Subspecies of HDL contain apolipoprotein E (apoE) and/or apoCIII. Both proteins have properties that could affect HDL metabolism. The relation between HDL metabolism and risk of coronary heart disease (CHD) is not well understood. METHODS Eighteen participants were given a bolus infusion of [D3]L-leucine to label endogenous proteins on HDL. HDL was separated into subspecies containing apoE and/or apoCIII and then into 4 sizes. Metabolic rates for apoA-I in HDL subspecies and sizes were determined by interactive modeling. The concentrations of apoE in HDL that contain or lack apoCIII were measured in a prospective study in Denmark including 1,949 incident CHD cases during 9 years. RESULTS HDL containing apoE but not apoCIII is disproportionately secreted into the circulation, actively expands while circulating, and is quickly cleared. These are key metabolic steps in reverse cholesterol transport, which may protect against atherosclerosis. ApoCIII on HDL strongly attenuates these metabolic actions of HDL apoE. In the epidemiological study, the relation between HDL apoE concentration and CHD significantly differed depending on whether apoCIII was present. HDL apoE was associated significantly with lower risk of CHD only in the HDL subspecies lacking apoCIII. CONCLUSIONS ApoE and apoCIII on HDL interact to affect metabolism and CHD. ApoE promotes metabolic steps in reverse cholesterol transport and is associated with lower risk of CHD. ApoCIII, when coexisting with apoE on HDL, abolishes these benefits. Therefore, differences in metabolism of HDL subspecies pertaining to reverse cholesterol transport are reflected in differences in association with CHD. TRIAL REGISTRATION Clinicaltrials.gov NCT01399632. FUNDING This work was supported by NIH grant R01HL095964 to FMS and by a grant to the Harvard Clinical and Translational Science Center (8UL1TR0001750) from the National Center for Advancing Translational Science.
Collapse
Affiliation(s)
- Allyson M Morton
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Manja Koch
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Carlos O Mendivil
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Medicine, Universidad de los Andes, Bogotá, Colombia.,Section of Endocrinology, Department of Internal Medicine, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Jeremy D Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Kim Overvad
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark.,Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Liyun Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Majken K Jensen
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Lipase inhibitor orlistat prevents hepatitis B virus infection by targeting an early step in the virus life cycle. Antiviral Res 2018; 151:4-7. [PMID: 29309795 DOI: 10.1016/j.antiviral.2018.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/19/2017] [Accepted: 01/04/2018] [Indexed: 12/31/2022]
Abstract
Hepatitis B Virus (HBV) is a strictly hepatotropic pathogen which is very efficiently targeted to the liver and into its host cell, the hepatocyte. The sodium taurocholate co-transporting polypeptide (NTCP) has been identified as a key virus entry receptor, but the early steps in the virus life cycle are still only barely understood. Here, we investigated the effect of lipase inhibition and lipoprotein uptake on HBV infection using differentiated HepaRG cells and primary human hepatocytes. We found that an excess of triglyceride rich lipoprotein particles in vitro diminished HBV infection and a reduced hepatic virus uptake in vivo if apolipoprotein E is lacking indicating virus transport along with lipoproteins to target hepatocytes. Moreover, we showed that HBV infection of hepatocytes was inhibited by the broadly active lipase inhibitor orlistat, approved as a therapeutic agent which blocks neutral lipid hydrolysis activity. Orlistat treatment targets HBV infection at a post-entry step and inhibited HBV infection during virus inoculation strongly in a dose-dependent manner. In contrast, orlistat had no effect on HBV gene expression or replication or when added after HBV infection. Taken together, our data indicate that HBV connects to the hepatotropic lipoprotein metabolism and that inhibition of cellular hepatic lipase(s) may allow to target early steps of HBV infection.
Collapse
|
34
|
Giordano-Mooga S, Datta G, Wolkowicz P, Garber DW, Palgunachari M, White CR, Anantharamaiah G. The Apolipoprotein E Mimetic Peptide AEM-2 Attenuates Mitochondrial Injury And Apoptosis In Human THP-1 Macrophages. CURRENT TOPICS IN PEPTIDE & PROTEIN RESEARCH 2018; 19:15-25. [PMID: 29955206 PMCID: PMC6019287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cardiovascular disease, specifically atherosclerosis, is exacerbated by hypercholesterolemia. Current therapies that target lipid lowering, however, are not effective in all patients. Apolipoprotein E (apoE) plays an important role in mediating the clearance of plasma cholesterol and also exerts numerous cytoprotective responses. Our laboratory has synthesized novel therapeutics that mimic the ability of apoE to decrease plasma cholesterol. The apoE mimetic peptide AEM-2 is a dual domain peptide composed of an amphipathic helical region that binds phospholipids and a positively charged region that mediates the hepatic clearance of lipoproteins. Administration of AEM-2 to apoE null mice reduced plasma cholesterol concentration by 80% one hour post-administration. Since apoE is also known to exert anti-inflammatory effects that are independent of its ability to lower cholesterol, we tested effects of AEM-2 on lipopolysaccharide-induced responses in human THP-1 macrophages. Pre-treatment of THP-1 cells with AEM-2 significantly reduced the LPS-induced secretion of IL-6 and TNFα. Since LPS administration is associated with an increase in mitochondrial injury, we monitored effects of AEM-2 on mitochondrial function. AEM-2 significantly reduced mitochondrial superoxide formation, prevented the LPS-induced decrease in mitochondrial membrane potential and attenuated the release of cytochrome c. AEM-2 also inhibited the activities of initiator caspases 8 and 9 and effector caspase 3. The attenuation of apoptosis in AEM-2 treated cells was associated with an increase in cellular autophagy. These data suggest that AEM-2 attenuates cellular injury in LPS-treated THP-1 macrophages and facilitates the removal of cellular debris and damaged organelles via induction of autophagy.
Collapse
Affiliation(s)
- Samantha Giordano-Mooga
- Department of Clinical and Diagnostic Sciences, University of Alabama at Birmingham, Birmingham, AL
| | - Geeta Datta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Paul Wolkowicz
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - David W. Garber
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - C. Roger White
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | |
Collapse
|
35
|
Zhu C, Zhu H, Song H, Xu L, Li L, Liu F, Liu X. Hepatitis B virus inhibits the in vivo and in vitro synthesis and secretion of apolipoprotein C3. Lipids Health Dis 2017; 16:213. [PMID: 29132372 PMCID: PMC5683573 DOI: 10.1186/s12944-017-0607-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/05/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) infection in the body can damage liver cells and cause disorders in blood lipid metabolism. Apolipoprotein C3 (ApoC3) plays an important role in the regulation of lipid metabolism, but no study on the HBV regulation of ApoC3 has been reported. This purpose of this study was to investigate the effect of HBV on ApoC3 expression and its regulatory mechanism. METHODS The expression levels of ApoC3 mRNA and protein in the human hepatoma cell lines HepG2 and HepG2.2.15 were determined using real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot. The HepG2 cells were co-transfected with the ApoC3 gene promoter and either HBV-infected clone pHBV1.3 or its individual genes. The changes in luciferase activity were assayed. The expression levels of ApoC3 mRNA and protein were determined using RT-qPCR and Western blot. The content of ApoC3 in the supernatant of the cultured cells was determined using an enzyme-linked immunosorbent assay (ELISA). The sera were collected from 149 patients with HBV infection and 102 healthy subjects at physical examination as the normal controls. The serological levels of ApoC3 in the HBV group and the normal control group were determined using ELISA. The contents of serum triglyceride (TG) and very-low-density lipoprotein (VLDL) in the HBV patients and the normal control were determined using an automatic biochemical analyser. RESULTS The expression levels of ApoC3 mRNA and protein were lower in the HepG2.2.15 cells than in the HepG2 cells. pHBV1.3 and its X gene could inhibit the activity of the ApoC3 promoter and its mRNA and protein expression. The serum levels of ApoC3, VLDL and TG were 65.39 ± 7.48 μg/ml, 1.24 ± 0.49 mmol/L, and 0.46 ± 0.10 mmol/L in the HBV patients and 41.02 ± 6.88 μg/ml, 0.76 ± 0.21 mmol/L, 0.29 ± 0.05 mmol/L in the normal controls, respectively, statistical analysis revealed significantly lower serum levels of ApoC3, VLDL and TG in HBV patients than in the normal controls (P < 0.05). CONCLUSION HBV can inhibit the in vivo and in vitro synthesis and secretion of ApoC3.
Collapse
Affiliation(s)
- Chengliang Zhu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Hengcheng Zhu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Hui Song
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Limin Xu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Longxuan Li
- Department of Neurology, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Fang Liu
- The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | - Xinghui Liu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai, 200135, China.
| |
Collapse
|
36
|
Wolska A, Dunbar RL, Freeman LA, Ueda M, Amar MJ, Sviridov DO, Remaley AT. Apolipoprotein C-II: New findings related to genetics, biochemistry, and role in triglyceride metabolism. Atherosclerosis 2017; 267:49-60. [PMID: 29100061 DOI: 10.1016/j.atherosclerosis.2017.10.025] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/03/2017] [Accepted: 10/19/2017] [Indexed: 02/08/2023]
Abstract
Apolipoprotein C-II (apoC-II) is a small exchangeable apolipoprotein found on triglyceride-rich lipoproteins (TRL), such as chylomicrons (CM) and very low-density lipoproteins (VLDL), and on high-density lipoproteins (HDL), particularly during fasting. ApoC-II plays a critical role in TRL metabolism by acting as a cofactor of lipoprotein lipase (LPL), the main enzyme that hydrolyses plasma triglycerides (TG) on TRL. Here, we present an overview of the role of apoC-II in TG metabolism, emphasizing recent novel findings regarding its transcriptional regulation and biochemistry. We also review the 24 genetic mutations in the APOC2 gene reported to date that cause hypertriglyceridemia (HTG). Finally, we describe the clinical presentation of apoC-II deficiency and assess the current therapeutic approaches, as well as potential novel emerging therapies.
Collapse
Affiliation(s)
- Anna Wolska
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Richard L Dunbar
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; ICON plc, North Wales, PA, USA; Cardiometabolic and Lipid Clinic, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Lita A Freeman
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Masako Ueda
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Marcelo J Amar
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Denis O Sviridov
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Leonova EI, Sadovnikova ES, Shaykhutdinova ER, Galzitskaya OV, Murashev AN, Solonin AS. Hepatic and Aortic Arch Expression and Serum Levels of Syndecan-1 in ApoE -/- Mice. Open Biochem J 2017; 11:77-93. [PMID: 29151984 PMCID: PMC5676011 DOI: 10.2174/1874091x01711010077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/03/2017] [Accepted: 06/09/2017] [Indexed: 12/17/2022] Open
Abstract
Background: Heparan sulfate proteoglycan (HSPG) syndecan-1 (Sdc1) acts as a receptor for triglyceride-rich lipoproteins (TRLs), growth factors, chemokines and enzymes. Due to the disordered structure, its function is as diverse as its ligands. In this paper, we have analyzed hepatic and aortic arch expression of Sdc1 in ApoE-/- mice and examined their association with biochemical changes in plasma during the atheroma formation. Methods: ApoE knockout (ApoE-/-) mice as a model of atherosclerosis were used. Plasma chemistry parameters were estimated by automatic biochemical analyzer. The ELISA test was used to detect soluble Sdc1. The mRNA level of syndecan-1 in liver cells and aortic arch was determined by real time PCR. Results: The Sdc1 mRNA level in liver cells was 1.5-2.5 times higher in ApoE-/- mice compared to the wild-type species and increased with age, whereas it remained at the same level in wild-type mice upon aging. Furthermore, the plasma cholesterol level was 4-6 times higher in ApoE-/- mice compared to the wild type; in contrast, triglyceride (TG) remained at the same level. Simultaneously, the expression of Sdc1 in the aortic arch of ApoE-/- mice decreases with age; however, it increases in wild-type mice of the same age. We determined that the Sdc1 mRNA expression in liver cells is significantly higher compared to the cells of aortic arch. In addition, our research demonstrated that the level of soluble Sdc1 slightly increased with age and did not depend on mouse genotype; yet, the total amount of soluble Sdc1 was higher in ApoE-/- mice. Conclusion: Our data suggest that the level of soluble Sdc1 in serum of mice can be associated with chronic inflammation. In addition, we hypothesized that a compensatory increase in the Sdc1 expression in ApoE-/- mice may prevent accumulation of triglycerides in serum, yet having no effect on cholesterol accumulation.
Collapse
Affiliation(s)
- Elena I Leonova
- Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, Moscow Region, Pushchino, 142290, Russia
| | - Elena S Sadovnikova
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Elvira R Shaykhutdinova
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Oxana V Galzitskaya
- Institute of Protein Research, Russian Academy of Sciences, Moscow Region, Pushchino, Russia
| | - Arkady N Murashev
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Alexandr S Solonin
- Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, Moscow Region, Pushchino, 142290, Russia
| |
Collapse
|
38
|
Abstract
Heparin and heparan sulfate glycosaminoglycans are long, linear polysaccharides that are made up of alternating dissacharide sequences of sulfated uronic acid and amino sugars. Unlike heparin, which is only found in mast cells, heparan sulfate is ubiquitously expressed on the cell surface and in the extracellular matrix of all animal cells. These negatively-charged glycans play essential roles in important cellular functions such as cell growth, adhesion, angiogenesis, and blood coagulation. These biomolecules are also involved in pathophysiological conditions such as pathogen infection and human disease. This review discusses past and current methods for targeting these complex biomolecules as a novel therapeutic strategy to treating disorders such as cancer, neurodegenerative diseases, and infection.
Collapse
Affiliation(s)
- Ryan J Weiss
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, USA.
| |
Collapse
|
39
|
Association of apolipoprotein A5 gene variants with metabolic syndrome in Tunisian population. ANNALES D'ENDOCRINOLOGIE 2017. [DOI: 10.1016/j.ando.2017.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
40
|
Guardiola M, Ribalta J. Update on APOA5 Genetics: Toward a Better Understanding of Its Physiological Impact. Curr Atheroscler Rep 2017; 19:30. [DOI: 10.1007/s11883-017-0665-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
41
|
Moriarty PM, Varvel SA, Gordts PLSM, McConnell JP, Tsimikas S. Lipoprotein(a) Mass Levels Increase Significantly According to APOE Genotype: An Analysis of 431 239 Patients. Arterioscler Thromb Vasc Biol 2017; 37:580-588. [PMID: 28062489 DOI: 10.1161/atvbaha.116.308704] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 12/21/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Lipoprotein(a) [Lp(a)] levels are genetically determined by hepatocyte apolipoprotein(a) synthesis, but catabolic pathways also influence circulating levels. APOE genotypes have different affinities for the low-density lipoprotein (LDL) receptor and LDL-related protein-1, with ε2 having the weakest binding to LDL receptor at <2% relative to ε3 and ε4. APPROACH AND RESULTS: APOE genotypes (ε2/ε2, ε2/ε3, ε2/ε4, ε3/ε3, ε3/ε4, and ε4/ε4), Lp(a) mass, directly measured Lp(a)-cholesterol levels, and a variety of apoB-related lipoproteins were measured in 431 239 patients. The prevalence of APOE traits were ε2: 7.35%, ε3: 77.56%, and ε4: 15.09%. Mean (SD) Lp(a) levels were 65% higher in ε4/ε4 compared with ε2/ε2 genotypes and increased significantly according to APOE genotype: ε2/ε2: 23.4 (29.2), ε2/ε3: 31.3 (38.0), ε2/ε4: 32.8 (38.5), ε3/ε3: 33.2 (39.1), ε3/ε4: 35.5 (41.6), and ε4/ε4: 38.5 (44.1) mg/dL (P<0.0001). LDL-cholesterol, apoB, Lp(a)-cholesterol, LDL-cholesterol corrected for Lp(a)-cholesterol content, LDL-particle number, and small, dense LDL also had similar patterns. Patients with LDL-cholesterol ≥250 mg/dL, who are more likely to have LDL receptor mutations and reduced affinity for apoB, had higher Lp(a) levels across all apoE isoforms, but particularly in patients with ε2 alleles, compared with LDL <250 mg/dL. The lowest Lp(a) mass levels were present in patients with ε2 isoforms and lowest LDL-cholesterol. CONCLUSIONS APOE genotypes strongly influence Lp(a) and apoB-related lipoprotein levels. This suggests that differences in affinity of apoE proteins for lipoprotein clearance receptors may affect Lp(a) catabolism, suggesting a competition between Lp(a) and apoE protein for similar receptors.
Collapse
Affiliation(s)
- Patrick M Moriarty
- From the Division of Clinical Pharmacology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City (P.M.M.); Salveo Diagnostics, Inc, Richmond, VA (S.A.V., J.P.M.); Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center (P.L.S.M.G.), Department of Medicine, Division of Endocrinology and Metabolism (P.L.S.M.G.), and Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center (S.T.), University of California San Diego, La Jolla
| | - Stephen A Varvel
- From the Division of Clinical Pharmacology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City (P.M.M.); Salveo Diagnostics, Inc, Richmond, VA (S.A.V., J.P.M.); Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center (P.L.S.M.G.), Department of Medicine, Division of Endocrinology and Metabolism (P.L.S.M.G.), and Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center (S.T.), University of California San Diego, La Jolla
| | - Philip L S M Gordts
- From the Division of Clinical Pharmacology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City (P.M.M.); Salveo Diagnostics, Inc, Richmond, VA (S.A.V., J.P.M.); Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center (P.L.S.M.G.), Department of Medicine, Division of Endocrinology and Metabolism (P.L.S.M.G.), and Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center (S.T.), University of California San Diego, La Jolla
| | - Joseph P McConnell
- From the Division of Clinical Pharmacology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City (P.M.M.); Salveo Diagnostics, Inc, Richmond, VA (S.A.V., J.P.M.); Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center (P.L.S.M.G.), Department of Medicine, Division of Endocrinology and Metabolism (P.L.S.M.G.), and Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center (S.T.), University of California San Diego, La Jolla
| | - Sotirios Tsimikas
- From the Division of Clinical Pharmacology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City (P.M.M.); Salveo Diagnostics, Inc, Richmond, VA (S.A.V., J.P.M.); Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center (P.L.S.M.G.), Department of Medicine, Division of Endocrinology and Metabolism (P.L.S.M.G.), and Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center (S.T.), University of California San Diego, La Jolla.
| |
Collapse
|
42
|
Gordts PLSM, Nock R, Son NH, Ramms B, Lew I, Gonzales JC, Thacker BE, Basu D, Lee RG, Mullick AE, Graham MJ, Goldberg IJ, Crooke RM, Witztum JL, Esko JD. ApoC-III inhibits clearance of triglyceride-rich lipoproteins through LDL family receptors. J Clin Invest 2016; 126:2855-66. [PMID: 27400128 DOI: 10.1172/jci86610] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/12/2016] [Indexed: 02/05/2023] Open
Abstract
Hypertriglyceridemia is an independent risk factor for cardiovascular disease, and plasma triglycerides (TGs) correlate strongly with plasma apolipoprotein C-III (ApoC-III) levels. Antisense oligonucleotides (ASOs) for ApoC-III reduce plasma TGs in primates and mice, but the underlying mechanism of action remains controversial. We determined that a murine-specific ApoC-III-targeting ASO reduces fasting TG levels through a mechanism that is dependent on low-density lipoprotein receptors (LDLRs) and LDLR-related protein 1 (LRP1). ApoC-III ASO treatment lowered plasma TGs in mice lacking lipoprotein lipase (LPL), hepatic heparan sulfate proteoglycan (HSPG) receptors, LDLR, or LRP1 and in animals with combined deletion of the genes encoding HSPG receptors and LDLRs or LRP1. However, the ApoC-III ASO did not lower TG levels in mice lacking both LDLR and LRP1. LDLR and LRP1 were also required for ApoC-III ASO-induced reduction of plasma TGs in mice fed a high-fat diet, in postprandial clearance studies, and when ApoC-III-rich or ApoC-III-depleted lipoproteins were injected into mice. ASO reduction of ApoC-III had no effect on VLDL secretion, heparin-induced TG reduction, or uptake of lipids into heart and skeletal muscle. Our data indicate that ApoC-III inhibits turnover of TG-rich lipoproteins primarily through a hepatic clearance mechanism mediated by the LDLR/LRP1 axis.
Collapse
|
43
|
Julve J, Martín-Campos JM, Escolà-Gil JC, Blanco-Vaca F. Chylomicrons: Advances in biology, pathology, laboratory testing, and therapeutics. Clin Chim Acta 2016; 455:134-48. [PMID: 26868089 DOI: 10.1016/j.cca.2016.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/01/2016] [Accepted: 02/06/2016] [Indexed: 01/17/2023]
Abstract
The adequate absorption of lipids is essential for all mammalian species due to their inability to synthesize some essential fatty acids and fat-soluble vitamins. Chylomicrons (CMs) are large, triglyceride-rich lipoproteins that are produced in intestinal enterocytes in response to fat ingestion, which function to transport the ingested lipids to different tissues. In addition to the contribution of CMs to postprandial lipemia, their remnants, the degradation products following lipolysis by lipoprotein lipase, are linked to cardiovascular disease. In this review, we will focus on the structure-function and metabolism of CMs. Second, we will analyze the impact of gene defects reported to affect CM metabolism and, also, the role of CMs in other pathologies, such as atherothrombotic cardiovascular disease and diabetes mellitus. Third, we will provide an overview of the laboratory tests currently used to study CM disorders, and, finally, we will highlight current treatments in diseases affecting CMs.
Collapse
Affiliation(s)
- Josep Julve
- Institut de Recerca de l'HSCSP - Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.
| | - Jesús M Martín-Campos
- Institut de Recerca de l'HSCSP - Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l'HSCSP - Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
| | - Francisco Blanco-Vaca
- Institut de Recerca de l'HSCSP - Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain; Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica, Barcelona, Spain
| |
Collapse
|
44
|
Extracellular matrix component signaling in cancer. Adv Drug Deliv Rev 2016; 97:28-40. [PMID: 26519775 DOI: 10.1016/j.addr.2015.10.013] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Cell responses to the extracellular matrix depend on specific signaling events. These are important from early development, through differentiation and tissue homeostasis, immune surveillance, and disease pathogenesis. Signaling not only regulates cell adhesion cytoskeletal organization and motility but also provides survival and proliferation cues. The major classes of cell surface receptors for matrix macromolecules are the integrins, discoidin domain receptors, and transmembrane proteoglycans such as syndecans and CD44. Cells respond not only to specific ligands, such as collagen, fibronectin, or basement membrane glycoproteins, but also in terms of matrix rigidity. This can regulate the release and subsequent biological activity of matrix-bound growth factors, for example, transforming growth factor-β. In the environment of tumors, there may be changes in cell populations and their receptor profiles as well as matrix constitution and protein cross-linking. Here we summarize roles of the three major matrix receptor types, with emphasis on how they function in tumor progression.
Collapse
|
45
|
Update on the molecular biology of dyslipidemias. Clin Chim Acta 2016; 454:143-85. [DOI: 10.1016/j.cca.2015.10.033] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/24/2015] [Accepted: 10/30/2015] [Indexed: 12/20/2022]
|
46
|
Forte TM, Sharma V, Ryan RO. Apolipoprotein A-V gene therapy for disease prevention / treatment: a critical analysis. J Biomed Res 2015; 30:88-93. [PMID: 26679785 PMCID: PMC4820885 DOI: 10.7555/jbr.30.20150059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 06/06/2015] [Accepted: 09/05/2015] [Indexed: 12/17/2022] Open
Abstract
Apolipoprotein (apo) A-V is a novel member of the class of exchangeable apo's involved in triacylglycerol (TG) homeostasis. Whereas a portion of hepatic-derived apoA-V is secreted into plasma and functions to facilitate lipoprotein lipase-mediated TG hydrolysis, another portion is recovered intracellularly, in association with cytosolic lipid droplets. Loss of apoA-V function is positively correlated with elevated plasma TG and increased risk of cardiovascular disease. Single nucleotide polymorphisms (SNP) in the APOA5 locus can affect transcription efficiency or introduce deleterious amino acid substitutions. Likewise, rare mutations in APOA5 that compromise functionality are associated with increased plasma TG and premature myocardial infarction. Genetically engineered mouse models and human population studies suggest that, in certain instances, supplementation with wild type (WT) apoA-V may have therapeutic benefit. It is hypothesized that individuals that manifest elevated plasma TG owing to deleterious APOA5 SNPs or rare mutations would respond to WT apoA-V supplementation with improved plasma TG clearance. On the other hand, subjects with hypertriglyceridemia of independent origin (unrelated to apoA-V function) may not respond to apoA-V augmentation in this manner. Improvement in the ability to identify individuals predicted to benefit, advances in gene transfer technology and the strong connection between HTG and heart disease, point to apoA-V supplementation as a viable disease prevention / therapeutic strategy. Candidates would include individuals that manifest chronic TG elevation, have low plasma apoA-V due to an APOA5 mutation/polymorphism and not have deleterious mutations/polymorphisms in other genes known to influence plasma TG levels.
Collapse
Affiliation(s)
- Trudy M Forte
- Center for Prevention of Obesity, Diabetes and Cardiovascular Disease, Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Vineeta Sharma
- Center for Prevention of Obesity, Diabetes and Cardiovascular Disease, Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Robert O Ryan
- Center for Prevention of Obesity, Diabetes and Cardiovascular Disease, Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA;
| |
Collapse
|
47
|
Weiss RJ, Gordts PLSM, Le D, Xu D, Esko JD, Tor Y. Small molecule antagonists of cell-surface heparan sulfate and heparin-protein interactions. Chem Sci 2015; 6:5984-5993. [PMID: 28133533 PMCID: PMC5267326 DOI: 10.1039/c5sc01208b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 07/21/2015] [Indexed: 01/13/2023] Open
Abstract
Surfen, bis-2-methyl-4-amino-quinolyl-6-carbamide, was previously reported as a small molecule antagonist of heparan sulfate (HS), a key cell-surface glycosaminoglycan found on all mammalian cells. To generate structure-activity relationships, a series of rationally designed surfen analogs was synthesized, where its dimeric structure, exocyclic amines, and urea linker region were modified to probe the role of each moiety in recognizing HS. An in vitro assay monitoring inhibition of fibroblast growth factor 2 binding to wild-type CHO cells was utilized to quantify interactions with cell surface HS. The dimeric molecular structure of surfen and its aminoquinoline ring systems was essential for its interaction with HS, and certain dimeric analogs displayed higher inhibitory potency than surfen and were also shown to block downstream FGF signaling in mouse embryonic fibroblast cells. These molecules were also able to antagonize other HS-protein interactions including the binding of soluble RAGE to HS. Importantly, selected molecules were shown to neutralize heparin and other heparinoids, including the synthetic pentasaccharide fondaparinux, in a factor Xa chromogenic assay and in vivo in mice. These results suggest that small molecule antagonists of heparan sulfate and heparin can be of therapeutic potential for the treatment of disorders involving glycosaminoglycan-protein interactions.
Collapse
Affiliation(s)
- Ryan J. Weiss
- Department of Chemistry and Biochemistry , University of California , San Diego , La Jolla , CA 92093-0358 , USA .
| | - Philip L. S. M. Gordts
- Cellular and Molecular Medicine , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| | - Dzung Le
- Department of Medicine , University of California , San Diego , La Jolla , CA 92093-0612 , USA
- Glycobiology Research and Training Center , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| | - Ding Xu
- Department of Oral Biology , University at Buffalo , Buffalo , NY 14260-1660 , USA
| | - Jeffrey D. Esko
- Cellular and Molecular Medicine , University of California , San Diego , La Jolla , CA 92093-0687 , USA
- Glycobiology Research and Training Center , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry , University of California , San Diego , La Jolla , CA 92093-0358 , USA .
- Glycobiology Research and Training Center , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Randomized clinical trials have assessed the effects of several classes of drugs on plasma cholesterol levels in patients with coronary artery disease. Agents including niacin, fibrates and statins significantly lower LDL-cholesterol, but tolerance issues and undesirable side-effects are common. Residual risk may also be present in patients with persistently low HDL-cholesterol despite a reduction in LDL-cholesterol. Recent trials of drugs that increase circulating HDL-cholesterol have also been disappointing. RECENT FINDINGS Ongoing efforts target the development of new pharmacotherapies to reduce circulating levels of atherogenic lipoproteins. The goal of this review is to discuss recent advances in the treatment of coronary artery disease and other vascular diseases characterized by an increase in circulating atherogenic lipoproteins. These include the development of inhibitors of ATP citrate lyase and proprotein convertase subtilisin/kexin type 9. We also discuss recent developments in HDL therapy, including the clinical assessment of cholesteryl ester transfer protein inhibitors and apolipoprotein E mimetic peptides. SUMMARY Several new classes of drug are undergoing clinical evaluation that show promise for atherogenic lipoprotein reduction in patients who are statin intolerant.
Collapse
Affiliation(s)
- C Roger White
- aDepartment of Medicine, Division of Cardiovascular Disease bDivision of Gerontology, Geriatric Medicine and Palliative Care cDivision of Biochemistry and Molecular Genetics dUniversity of Alabama at Birmingham, Birmingham, Alabama eLipimetiX Development, LLC, Natick, Massachusetts, USA
| | | | | |
Collapse
|
49
|
Zhang LS, Xu M, Yang Q, Ryan RO, Howles P, Tso P. Apolipoprotein A-V deficiency enhances chylomicron production in lymph fistula mice. Am J Physiol Gastrointest Liver Physiol 2015; 308:G634-42. [PMID: 25617349 PMCID: PMC4385892 DOI: 10.1152/ajpgi.00339.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/16/2015] [Indexed: 01/31/2023]
Abstract
Apolipoprotein A-V (apoA-V), a liver-synthesized apolipoprotein discovered in 2001, strongly modulates fasting plasma triglycerides (TG). Little is reported on the effect of apoA-V on postprandial plasma TG, an independent predictor for atherosclerosis. Overexpressing apoA-V in mice suppresses postprandial TG, but mechanisms focus on increased lipolysis or clearance of remnant particles. Unknown is whether apoA-V suppresses the absorption of dietary lipids by the gut. This study examines how apoA-V deficiency affects the steady-state absorption and lymphatic transport of dietary lipids in chow-fed mice. Using apoA-V knockout (KO, n = 8) and wild-type (WT, n = 8) lymph fistula mice, we analyzed the uptake and lymphatic transport of lipids during a continuous infusion of an emulsion containing [(3)H]triolein and [(14)C]cholesterol. ApoA-V KO mice showed a twofold increase in (3)H (P < 0.001) and a threefold increase in (14)C (P < 0.001) transport into the lymph compared with WT. The increased lymphatic transport was accompanied by a twofold reduction (P < 0.05) in mucosal (3)H, suggesting that apoA-V KO mice more rapidly secreted [(3)H]TG out of the mucosa into the lymph. ApoA-V KO mice also produced chylomicrons more rapidly than WT (P < 0.05), as measured by the transit time of [(14)C]oleic acid from the intestinal lumen to lymph. Interestingly, apoA-V KO mice produced a steadily increasing number of chylomicron particles over time, as measured by lymphatic apoB output. The data suggest that apoA-V suppresses the production of chylomicrons, playing a previously unknown role in lipid metabolism that may contribute to the postprandial hypertriglyceridemia associated with apoA-V deficiency.
Collapse
Affiliation(s)
- Linda S. Zhang
- 1Children's Hospital Oakland Research Institute, Oakland, California
| | - Min Xu
- 1Children's Hospital Oakland Research Institute, Oakland, California
| | - Qing Yang
- 1Children's Hospital Oakland Research Institute, Oakland, California
| | - Robert O. Ryan
- 2Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| | - Philip Howles
- 1Children's Hospital Oakland Research Institute, Oakland, California
| | - Patrick Tso
- 1Children's Hospital Oakland Research Institute, Oakland, California
| |
Collapse
|
50
|
Giammanco A, Cefalù AB, Noto D, Averna MR. The pathophysiology of intestinal lipoprotein production. Front Physiol 2015; 6:61. [PMID: 25852563 PMCID: PMC4367171 DOI: 10.3389/fphys.2015.00061] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/14/2015] [Indexed: 12/12/2022] Open
Abstract
Intestinal lipoprotein production is a multistep process, essential for the absorption of dietary fats and fat-soluble vitamins. Chylomicron assembly begins in the endoplasmic reticulum with the formation of primordial, phospholipids-rich particles that are then transported to the Golgi for secretion. Several classes of transporters play a role in the selective uptake and/or export of lipids through the villus enterocytes. Once secreted in the lymph stream, triglyceride-rich lipoproteins (TRLs) are metabolized by Lipoprotein lipase (LPL), which catalyzes the hydrolysis of triacylglycerols of very low density lipoproteins (VLDLs) and chylomicrons, thereby delivering free fatty acids to various tissues. Genetic mutations in the genes codifying for these proteins are responsible of different inherited disorders affecting chylomicron metabolism. This review focuses on the molecular pathways that modulate the uptake and the transport of lipoproteins of intestinal origin and it will highlight recent findings on TRLs assembly.
Collapse
Affiliation(s)
| | | | | | - Maurizio R. Averna
- Dipartimento Biomedico di Medicina Interna e Specialistica, Università degli Studi di PalermoPalermo, Italy
| |
Collapse
|