1
|
Chaix A, Lin T, Ramms B, Cutler RG, Le T, Lopez C, Miu P, Pinto AFM, Saghatelian A, Playford MP, Mehta NN, Mattson MP, Gordts P, Witztum JL, Panda S. Time-Restricted Feeding Reduces Atherosclerosis in LDLR KO Mice but Not in ApoE Knockout Mice. Arterioscler Thromb Vasc Biol 2024; 44:2069-2087. [PMID: 39087348 PMCID: PMC11409897 DOI: 10.1161/atvbaha.124.320998] [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: 03/23/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Dyslipidemia increases cardiovascular disease risk, the leading cause of death worldwide. Under time-restricted feeding (TRF), wherein food intake is restricted to a consistent window of <12 hours, weight gain, glucose intolerance, inflammation, dyslipidemia, and hypercholesterolemia are all reduced in mice fed an obesogenic diet. LDLR (low-density lipoprotein receptor) mutations are a major cause of familial hypercholesterolemia and early-onset cardiovascular disease. METHODS We subjected benchmark preclinical models, mice lacking LDLR-knockout or ApoE knockout to ad libitum feeding of an isocaloric atherogenic diet either ad libitum or 9 hours TRF for up to 13 weeks and assessed disease development, mechanism, and global changes in hepatic gene expression and plasma lipids. In a regression model, a subset of LDLR-knockout mice were ad libitum fed and then subject to TRF. RESULTS TRF could significantly attenuate weight gain, hypercholesterolemia, and atherosclerosis in mice lacking the LDLR-knockout mice under experimental conditions of both prevention and regression. In LDLR-knockout mice, increased hepatic expression of genes mediating β-oxidation during fasting is associated with reduced VLDL (very-low-density lipoprotein) secretion and lipid accumulation. Additionally, increased sterol catabolism coupled with fecal loss of cholesterol and bile acids contributes to the atheroprotective effect of TRF. Finally, TRF alone or combined with a cholesterol-free diet can reduce atherosclerosis in LDLR-knockout mice. However, mice lacking ApoE, which is an important protein for hepatic lipoprotein reuptake do not respond to TRF. CONCLUSIONS In a preclinical animal model, TRF is effective in both the prevention and regression of atherosclerosis in LDLR knockout mice. The results suggest TRF alone or in combination with a low-cholesterol diet can be a lifestyle intervention for reducing cardiovascular disease risk in humans.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Terry Lin
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Bastian Ramms
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Roy G. Cutler
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. 21224
| | - Tiffani Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Catherine Lopez
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Phuong Miu
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Antonio F. M. Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States. 21205
| | - Philip Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Joseph L. Witztum
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Lead contact
| |
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
|
Bornfeldt KE. Apolipoprotein C3: form begets function. J Lipid Res 2024; 65:100475. [PMID: 37972731 PMCID: PMC10805671 DOI: 10.1016/j.jlr.2023.100475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023] Open
Abstract
Increased circulating levels of apolipoprotein C3 (APOC3) predict cardiovascular disease (CVD) risk in humans, and APOC3 promotes atherosclerosis in mouse models. APOC3's mechanism of action is due in large part to its ability to slow the clearance of triglyceride-rich lipoproteins (TRLs) and their remnants when APOC3 is carried by these lipoproteins. However, different pools and forms of APOC3 exert distinct biological effects or associations with atherogenic processes. Thus, lipid-free APOC3 induces inflammasome activation in monocytes whereas lipid particle-bound APOC3 does not. APOC3-enriched LDL binds better to the vascular glycosaminoglycan biglycan than does LDL depleted of APOC3. Patterns of APOC3 glycoforms predict CVD risk differently. The function of APOC3 bound to HDL is largely unknown. There is still much to learn about the mechanisms of action of different forms and pools of APOC3 in atherosclerosis and CVD, and whether APOC3 inhibition would prevent CVD risk in patients on LDL-cholesterol lowering medications.
Collapse
Affiliation(s)
- Karin E Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Trieger GW, Pessentheiner AR, Purcell SC, Green CR, DeForest N, Willert K, Majithia AR, Metallo CM, Godula K, Gordts PLSM. Glycocalyx engineering with heparan sulfate mimetics attenuates Wnt activity during adipogenesis to promote glucose uptake and metabolism. J Biol Chem 2023; 299:104611. [PMID: 36931394 PMCID: PMC10164900 DOI: 10.1016/j.jbc.2023.104611] [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: 11/15/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Adipose tissue plays a crucial role in maintaining metabolic homeostasis by storing lipids and glucose from circulation as intracellular fat. As peripheral tissues like adipose tissue become insulin resistant, decompensation of blood glucose levels occurs causing type 2 diabetes (T2D). Currently, modulating the glycocalyx, a layer of cell-surface glycans, is an underexplored pharmacological treatment strategy to improve glucose homeostasis in T2D patients. Here, we show a novel role for cell-surface heparan sulfate (HS) in establishing glucose uptake capacity and metabolic utilization in differentiated adipocytes. Using a combination of chemical and genetic interventions, we identified that HS modulates this metabolic phenotype by attenuating levels of Wnt signaling during adipogenesis. By engineering, the glycocalyx of pre-adipocytes with exogenous synthetic HS mimetics, we were able to enhance glucose clearance capacity after differentiation through modulation of Wnt ligand availability. These findings establish the cellular glycocalyx as a possible new target for therapeutic intervention in T2D patients by enhancing glucose clearance capacity independent of insulin secretion.
Collapse
Affiliation(s)
- Greg W Trieger
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA; Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, USA
| | - Ariane R Pessentheiner
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, USA
| | - Sean C Purcell
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Courtney R Green
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Natalie DeForest
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, USA
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | - Amit R Majithia
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, USA; Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Kamil Godula
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA.
| | - Philip L S M Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California, USA; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
5
|
Smith MM, Melrose J. Pentosan Polysulfate Affords Pleotropic Protection to Multiple Cells and Tissues. Pharmaceuticals (Basel) 2023; 16:437. [PMID: 36986536 PMCID: PMC10132487 DOI: 10.3390/ph16030437] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
Pentosan polysulfate (PPS), a small semi-synthetic highly sulfated heparan sulfate (HS)-like molecule, shares many of the interactive properties of HS. The aim of this review was to outline the potential of PPS as an interventional therapeutic protective agent in physiological processes affecting pathological tissues. PPS is a multifunctional molecule with diverse therapeutic actions against many disease processes. PPS has been used for decades in the treatment of interstitial cystitis and painful bowel disease, it has tissue-protective properties as a protease inhibitor in cartilage, tendon and IVD, and it has been used as a cell-directive component in bioscaffolds in tissue engineering applications. PPS regulates complement activation, coagulation, fibrinolysis and thrombocytopenia, and it promotes the synthesis of hyaluronan. Nerve growth factor production in osteocytes is inhibited by PPS, reducing bone pain in osteoarthritis and rheumatoid arthritis (OA/RA). PPS also removes fatty compounds from lipid-engorged subchondral blood vessels in OA/RA cartilage, reducing joint pain. PPS regulates cytokine and inflammatory mediator production and is also an anti-tumor agent that promotes the proliferation and differentiation of mesenchymal stem cells and the development of progenitor cell lineages that have proven to be useful in strategies designed to effect repair of the degenerate intervertebral disc (IVD) and OA cartilage. PPS stimulates proteoglycan synthesis by chondrocytes in the presence or absence of interleukin (IL)-1, and stimulates hyaluronan production by synoviocytes. PPS is thus a multifunctional tissue-protective molecule of potential therapeutic application for a diverse range of disease processes.
Collapse
Affiliation(s)
- Margaret M. Smith
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia;
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia;
- Graduate Schools of Biomedical Engineering, University of NSW, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern Campus, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
6
|
He T, Lykov N, Luo X, Wang H, Du Z, Chen Z, Chen S, Zhu L, Zhao Y, Tzeng C. Protective Effects of Lactobacillus gasseri against High-Cholesterol Diet-Induced Fatty Liver and Regulation of Host Gene Expression Profiles. Int J Mol Sci 2023; 24:ijms24032053. [PMID: 36768377 PMCID: PMC9917166 DOI: 10.3390/ijms24032053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Fatty liver is one of the most pervasive liver diseases worldwide. Probiotics play an important role in the progression of liver disease, but their effects on host regulation are poorly understood. This study investigated the protective effects of lactobacillus gasseri (L. gasseri) against high-cholesterol diet (HCD)-induced fatty liver injury using a zebrafish larvae model. Liver pathology, lipid accumulation, oxidative stress and hepatic inflammation were evaluated to demonstrate the changes in a spectrum of hepatic injury. Moreover, multiple indexes on host gene expression profiles were comprehensively characterized by RNA screening. The results showed that treatment with L. gasseri ameliorated HCD-induced morphological and histological alterations, lipid regulations, oxidative stress and macrophage aggregation in the liver of zebrafish larvae. Furthermore, the enrichment of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway revealed that the core pathways of L. gasseri regulation were interleukin-17 (IL-17) signaling, phosphoinositide 3-kinase (PI3K)-AKT signaling pathway, the regulation of lipolysis and adipocytes and fatty acid elongation and estrogen signaling. The genes at key junction nodes, hsp90aa1.1, kyat3, hsd17b7, irs2a, myl9b, ptgs2b, cdk21 and papss2a were significantly regulated by L. gasseri administration. To conclude, the current research extends our understanding of the protective effects of L. gasseri against fatty liver and provides potential therapeutic options for fatty liver treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ye Zhao
- Correspondence: (Y.Z.); (C.T.)
| | | |
Collapse
|
7
|
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
|
8
|
du Preez HN, Aldous C, Kruger HG, Johnson L. N-Acetylcysteine and Other Sulfur-Donors as a Preventative and Adjunct Therapy for COVID-19. Adv Pharmacol Pharm Sci 2022; 2022:4555490. [PMID: 35992575 PMCID: PMC9385285 DOI: 10.1155/2022/4555490] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/07/2022] [Indexed: 12/11/2022] Open
Abstract
The airway epithelial glycocalyx plays an important role in preventing severe acute respiratory syndrome coronavirus 2 entry into the epithelial cells, while the endothelial glycocalyx contributes to vascular permeability and tone, as well as modulating immune, inflammatory, and coagulation responses. With ample evidence in the scientific literature that coronavirus disease 2019 (COVID-19) is related to epithelial and endothelial dysfunction, preserving the glycocalyx should be the main focus of any COVID-19 treatment protocol. The most studied functional unit of the glycocalyx is the glycosaminoglycan heparan sulfate, where the degree and position of the sulfate groups determine the biological activity. N-acetylcysteine (NAC) and other sulfur donors contribute to the inorganic sulfate pool, the rate-limiting molecule in sulfation. NAC is not only a precursor to glutathione but also converts to hydrogen sulfide, inorganic sulfate, taurine, Coenzyme A, and albumin. By optimising inorganic sulfate availability, and therefore sulfation, it is proposed that COVID-19 can be prevented or at least most of the symptoms attenuated. A comprehensive COVID-19 treatment protocol is needed to preserve the glycocalyx in both the prevention and treatment of COVID-19. The use of NAC at a dosage of 600 mg bid for the prevention of COVID-19 is proposed, but a higher dosage of NAC (1200 mg bid) should be administered upon the first onset of symptoms. In the severe to critically ill, it is advised that IV NAC should be administered immediately upon hospital admission, and in the late stage of the disease, IV sodium thiosulfate should be considered. Doxycycline as a protease inhibitor will prevent shedding and further degradation of the glycocalyx.
Collapse
Affiliation(s)
- Heidi N du Preez
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Colleen Aldous
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Lin Johnson
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
9
|
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
|
10
|
Kinaneh S, Hijaze W, Mansour-Wattad L, Hammoud R, Zaidani H, Kabala A, Hamoud S. Heparanase Inhibition Prevents Liver Steatosis in E 0 Mice. J Clin Med 2022; 11:jcm11061672. [PMID: 35329997 PMCID: PMC8954723 DOI: 10.3390/jcm11061672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 12/22/2022] Open
Abstract
Background: Non-alcoholic fatty liver disease affects up to 30% of adults in the USA, and is associated with a higher incidence of chronic liver morbidity and mortality. Several molecular pathways are involved in the pathology of liver steatosis, including lipid uptake, lipogenesis, lipolysis, and beta-oxidation. The enzyme heparanase has been implicated in liver steatosis. Herein, we investigated the effect of heparanase inhibition on liver steatosis in E0 mice. Methods: In vivo experiments: Male wild-type mice fed with either chow diet (n = 4) or high-fat diet (n = 6), and male E0 mice fed with chow diet (n = 8) or high-fat diet (n = 33) were included. Mice on a high-fat diet were treated for 12 weeks with PG545 at low dose (6.4 mg/kg/week, ip, n = 6) or high dose (13.3 mg/kg/week, ip, n = 7), SST0001 (1.2 mg/mouse/day, ip, n = 6), or normal saline (control, n = 14). Animals were sacrificed two days after inducing peritonitis. Serum was analyzed for biochemical parameters. Mouse peritoneal macrophages (MPMs) were harvested and analyzed for lipid content. Livers were harvested for histopathological analysis of steatosis, lipid content, and the expression of steatosis-related factors at the mRNA level. In vitro experiments: MPMs were isolated from untreated E0 mice aged 8–10 weeks and were cultured and treated with either PG545 or SST0001, both at 50 µg/mL for 24 h, followed by assessment of mRNA expression of steatosis related factors. Results: Heparanase inhibition significantly attenuated the development of liver steatosis, as was evident by liver histology and lipid content. Serum analysis indicated lowering of cholesterol and triglycerides levels in mice treated with heparanase inhibitors. In liver tissue, assessment of mRNA expression of key factors in lipid uptake, lipolysis, lipogenesis, and beta-oxidation exhibited significant downregulation following PG545 treatment and to a lesser extent when SST0001 was applied. However, in vitro treatment of MPMs with PG545, but not SST0001, resulted in increased lipid content in these cells, which is opposed to their effect on MPMs of treated mice. This may indicate distinct regulatory pathways in the system or isolated macrophages following heparanase inhibition. Conclusion: Heparanase inhibition significantly attenuates the development of liver steatosis by decreasing tissue lipid content and by affecting the mRNA expression of key lipid metabolism regulators.
Collapse
Affiliation(s)
- Safa Kinaneh
- Department of Physiology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3200003, Israel; (S.K.); (A.K.)
| | - Walaa Hijaze
- Department of Emergency Medicine, Rambam Health Care Campus, Haifa 3109601, Israel; (W.H.); (H.Z.)
| | - Lana Mansour-Wattad
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Rawan Hammoud
- Faculty of Biotechnology, Hadassah Academic College, Jerusalem 9101001, Israel;
| | - Hisam Zaidani
- Department of Emergency Medicine, Rambam Health Care Campus, Haifa 3109601, Israel; (W.H.); (H.Z.)
| | - Aviva Kabala
- Department of Physiology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3200003, Israel; (S.K.); (A.K.)
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel;
- Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3200003, Israel
- Correspondence: ; Tel.: +972-4-7772300; Fax: +972-4-7771691
| |
Collapse
|
11
|
Mashima R, Okuyama T, Ohira M. Physiology and Pathophysiology of Heparan Sulfate in Animal Models: Its Biosynthesis and Degradation. Int J Mol Sci 2022; 23:1963. [PMID: 35216081 PMCID: PMC8876164 DOI: 10.3390/ijms23041963] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/17/2022] Open
Abstract
Heparan sulfate (HS) is a type of glycosaminoglycan that plays a key role in a variety of biological functions in neurology, skeletal development, immunology, and tumor metastasis. Biosynthesis of HS is initiated by a link of xylose to Ser residue of HS proteoglycans, followed by the formation of a linker tetrasaccharide. Then, an extension reaction of HS disaccharide occurs through polymerization of many repetitive units consisting of iduronic acid and N-acetylglucosamine. Subsequently, several modification reactions take place to complete the maturation of HS. The sulfation positions of N-, 2-O-, 6-O-, and 3-O- are all mediated by specific enzymes that may have multiple isozymes. C5-epimerization is facilitated by the epimerase enzyme that converts glucuronic acid to iduronic acid. Once these enzymatic reactions have been completed, the desulfation reaction further modifies HS. Apart from HS biosynthesis, the degradation of HS is largely mediated by the lysosome, an intracellular organelle with acidic pH. Mucopolysaccharidosis is a genetic disorder characterized by an accumulation of glycosaminoglycans in the body associated with neuronal, skeletal, and visceral disorders. Genetically modified animal models have significantly contributed to the understanding of the in vivo role of these enzymes. Their role and potential link to diseases are also discussed.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; (T.O.); (M.O.)
| | | | | |
Collapse
|
12
|
du Preez HN, Aldous C, Hayden MR, Kruger HG, Lin J. Pathogenesis of COVID-19 described through the lens of an undersulfated and degraded epithelial and endothelial glycocalyx. FASEB J 2021; 36:e22052. [PMID: 34862979 DOI: 10.1096/fj.202101100rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
The glycocalyx surrounds every eukaryotic cell and is a complex mesh of proteins and carbohydrates. It consists of proteoglycans with glycosaminoglycan side chains, which are highly sulfated under normal physiological conditions. The degree of sulfation and the position of the sulfate groups mainly determine biological function. The intact highly sulfated glycocalyx of the epithelium may repel severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) through electrostatic forces. However, if the glycocalyx is undersulfated and 3-O-sulfotransferase 3B (3OST-3B) is overexpressed, as is the case during chronic inflammatory conditions, SARS-CoV-2 entry may be facilitated by the glycocalyx. The degree of sulfation and position of the sulfate groups will also affect functions such as immune modulation, the inflammatory response, vascular permeability and tone, coagulation, mediation of sheer stress, and protection against oxidative stress. The rate-limiting factor to sulfation is the availability of inorganic sulfate. Various genetic and epigenetic factors will affect sulfur metabolism and inorganic sulfate availability, such as various dietary factors, and exposure to drugs, environmental toxins, and biotoxins, which will deplete inorganic sulfate. The role that undersulfation plays in the various comorbid conditions that predispose to coronavirus disease 2019 (COVID-19), is also considered. The undersulfated glycocalyx may not only increase susceptibility to SARS-CoV-2 infection, but would also result in a hyperinflammatory response, vascular permeability, and shedding of the glycocalyx components, giving rise to a procoagulant and antifibrinolytic state and eventual multiple organ failure. These symptoms relate to a diagnosis of systemic septic shock seen in almost all COVID-19 deaths. The focus of prevention and treatment protocols proposed is the preservation of epithelial and endothelial glycocalyx integrity.
Collapse
Affiliation(s)
- Heidi N du Preez
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Colleen Aldous
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Melvin R Hayden
- Division of Endocrinology Diabetes and Metabolism, Department of Internal Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA.,Diabetes and Cardiovascular Disease Center, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Johnson Lin
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
13
|
Mkhize BC, Mosili P, Ngubane PS, Sibiya NH, Khathi A. Diet-induced prediabetes: Effects on the activity of the renin-angiotensin-aldosterone system (RAAS) in selected organs. J Diabetes Investig 2021; 13:768-780. [PMID: 34619025 PMCID: PMC9077724 DOI: 10.1111/jdi.13690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/14/2021] [Accepted: 10/04/2021] [Indexed: 11/30/2022] Open
Abstract
Derangements often observed with type 2 diabetes (T2D) are associated with disturbances in renin-angiotensin-aldosterone system (RAAS) activity. A positive correlation between local RAAS activity and the complications observed in T2D has been noted. However, the detrimental ramifications due to moderate hyperglycemia noted in prediabetes and the affected organ system and mechanistic pathways are not elucidated. Hence, this study investigated the effects of diet-induced prediabetes on RAAS in various organs. MATERIALS AND METHODS Male Sprague-Dawley rats were separated into two groups: non-pre-diabetic (NPD) through exposure to standard rat chow and diet-induced prediabetic (PD) group by exposure to a high-fat high carbohydrate diet for 32 weeks. RAAS activity in the skeletal muscle, adipose tissue, liver, pancreas and heart was determined through the analysis of RAAS components such as; renin, angiotensinogen, angiotensin-converting enzyme (ACE) and angiotensin II type 1 receptor (AT1R) via PCR as well as the quantification of angiotensin II and aldosterone concentration. Furthermore, NADPH oxidase, SOD and GPx1 concentrations were determined in the skeletal muscle, pancreas and heart in addition to the hepatic triglycerides. RESULTS The RAAS components were elevated in the PD group when compared to the NPD. This was further accompanied by increased NADPH oxidase and reduced SOD and GPx1 concentrations in the selected organs, in addition to the elevated hepatic triglycerides concentration in the PD by comparison to NPD. CONCLUSION Due to these observed changes, we suggest that local RAAS activity in the prediabetic state in selected organs elicits the derangements noted in T2D.
Collapse
|
14
|
Chen T, Zhang Y, Dong Y, Zhang D, Xia L, Sun X, Li H, Han C, Wang H, Xu G. Mast cell and heparin promote adipogenesis in superficial fascia of rats. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159024. [PMID: 34389520 DOI: 10.1016/j.bbalip.2021.159024] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/13/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
Fascial adipocytes are recently identified as a unique population of adipose cells, which have different developmental origins, anatomical locations, cytological and functional characteristics compared with subcutaneous or visceral adipocytes. Superficial fascia in rats (also in pigs but not obviously in mice) contains numbers of lineage committed preadipocytes which possess adipogenic potential in vivo. The present study aimed to investigate the physiological factors that contribute to fascial adipogenesis in rats. We detected that mast cells, adipose progenitor cells, and mature adipocytes distributed in certain fascia areas were closely associated with each other, and numerous heparin-loaded granules released from mast cells were distributed around fascial preadipocytes. The culture supernatants of rat peritoneal mast cells and RBL-2H3 mast cells contained 20-30 μg/ml of heparin, effectively activated PPAR-responsive luciferase activity, promoted mRNA and protein expressions of key adipogenic genes, and hence increased adipogenic differentiation of fascia- or epididymal adipose-derived stromal cells. Adipogenic effects of mast cell supernatants were mimicked by heparin but not by histamine or 5-hydroxytryptamine, and were antagonized by protamine sulfate. Adipogenic effects of heparin may relate to its chain length of glucosamine units, because heparin stimulated stronger adipogenesis than dalteparin and enoxaparin with relatively short chains. In rats, local administration of heparin-loaded microspheres for 30 days induced adipogenesis in local areas of superficial fascia. Our findings suggested that mast cell and its granule heparin could serve as the endogenous physiological factors to initiate and accelerate local adipogenesis in superficial fascia, or in adipose tissue with the fascia naturally embedded inside.
Collapse
Affiliation(s)
- Tongsheng Chen
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Yanfei Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Yingyue Dong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Dandan Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Lisha Xia
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Xiaozhe Sun
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Hanxiao Li
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Chunmiao Han
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Huamin Wang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Guoheng Xu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China.
| |
Collapse
|
15
|
Jagannath S, Chilkunda ND. High cholesterol-supplemented diet during gestation and lactation alters liver glycosaminoglycans and associated lipoprotein receptors and results in fat accumulation in adulthood. Nutr Res 2021; 93:50-60. [PMID: 34365197 DOI: 10.1016/j.nutres.2021.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
In utero insults to growing fetus impact its health in adulthood. Glycosaminoglycans (GAGs) are involved in lipoprotein metabolism in the liver and vary both quantitively and qualitatively on feeding adult rats a diet rich in cholesterol. However, no reports are available to show the modulation of GAGs when the offspring are subjected to a high cholesterol diet in gestation and lactation stages. Hypercholesterolemia in pregnant rats was induced by feeding an AIN-93 diet supplemented with 0.5% cholesterol. The pups born to mothers fed with high cholesterol diet showed a significant increase in cholesterol and triglycerides accumulation in the liver. Quantitative changes in sulfated glycosaminoglycans (sGAGs), in particular of heparan sulfate, were observed across the developmental stages. Other players involved in lipoprotein metabolism, namely low-density lipoprotein receptor-related protein 1, apolipoprotein E, and low-density lipoprotein receptor expression levels, also showed differential changes across developmental stages. Interestingly, when pups from hypercholesterolemic mothers were fed a normal diet after weaning until adulthood, a considerable amount of fat accumulation in the liver was observed, implicating fetal exposure to early high cholesterol exposure on long term health.
Collapse
Affiliation(s)
- Sanjana Jagannath
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, JLB Road, Mysore 570020, Karnataka, India
| | - Nandini D Chilkunda
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, JLB Road, Mysore 570020, Karnataka, India.
| |
Collapse
|
16
|
Shrestha P, Adepu S, Vivès RR, Masri RE, Klooster A, Kaptein F, Dam W, Bakker SJL, van Goor H, van de Sluis B, van den Born J. Hypercholesterolemia in Progressive Renal Failure Is Associated with Changes in Hepatic Heparan Sulfate - PCSK9 Interaction. J Am Soc Nephrol 2021; 32:1371-1388. [PMID: 33758009 PMCID: PMC8259657 DOI: 10.1681/asn.2020091376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 02/04/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Dyslipidemia is an important risk factor in CKD. The liver clears triglyceride-rich lipoproteins (TRL) via LDL receptor (LDLR), LDLR-related protein-1 (LRP-1), and heparan sulfate proteoglycans (HSPGs), mostly syndecan-1. HSPGs also facilitate LDLR degradation by proprotein convertase subtilisin/kexin type 9 (PCSK9). Progressive renal failure affects the structure and activity of hepatic lipoprotein receptors, PCSK9, and plasma cholesterol. METHODS Uninephrectomy- and aging-induced CKD in normotensive Wistar rats and hypertensive Munich-Wistar-Frömter (MWF) rats. RESULTS Compared with 22-week-old sex- and strain-matched rats, 48-week-old uninephrectomized Wistar-CKD and MWF-CKD rats showed proteinuria, increased plasma creatinine, and hypercholesterolemia (all P<0.05), which were most apparent in hypertensive MWF-CKD rats. Hepatic PCSK9 expression increased in both CKD groups (P<0.05), with unusual sinusoidal localization, which was not seen in 22-week-old rats. Heparan sulfate (HS) disaccharide analysis, staining with anti-HS mAbs, and mRNA expression of HS polymerase exostosin-1 (Ext-1), revealed elongated HS chains in both CKD groups. Solid-phase competition assays showed that the PCSK9 interaction with heparin-albumin (HS-proteoglycan analogue) was critically dependent on polysaccharide chain length. VLDL binding to HS from CKD livers was reduced (P<0.05). Proteinuria and plasma creatinine strongly associated with plasma cholesterol, PCSK9, and HS changes. CONCLUSIONS Progressive CKD induces hepatic HS elongation, leading to increased interaction with PCSK9. This might reduce hepatic lipoprotein uptake and thereby induce dyslipidemia in CKD. Therefore, PCSK9/HS may be a novel target to control dyslipidemia.
Collapse
Affiliation(s)
- Pragyi Shrestha
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Saritha Adepu
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Romain R. Vivès
- University Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Energie Atomique (CEA), Institute of Structural Biology, Grenoble, France
| | - Rana El Masri
- University Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Energie Atomique (CEA), Institute of Structural Biology, Grenoble, France
| | - Astrid Klooster
- Department of Pathology, Pathology Friesland, Leeuwarden, The Netherlands
| | - Fleur Kaptein
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wendy Dam
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J. L. Bakker
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department Pediatrics, Section of Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
Lazaris V, Hatziri A, Symeonidis A, Kypreos KE. The Lipoprotein Transport System in the Pathogenesis of Multiple Myeloma: Advances and Challenges. Front Oncol 2021; 11:638288. [PMID: 33842343 PMCID: PMC8032975 DOI: 10.3389/fonc.2021.638288] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/10/2021] [Indexed: 01/02/2023] Open
Abstract
Multiple myeloma (MM) is an incurable neoplastic hematologic disorder characterized by malignant plasma cells, mainly in the bone marrow. MM is associated with multiple factors, such as lipid metabolism, obesity, and age-associated disease development. Although, the precise pathogenetic mechanisms remain unknown, abnormal lipid and lipoprotein levels have been reported in patients with MM. Interestingly, patients with higher APOA1 levels, the major apolipoprotein of high density lipoprotein (HDL), have better overall survival. The limited existing studies regarding serum lipoproteins in MM are inconclusive, and often contradictory. Nevertheless, it appears that deregulation of the lipoprotein transport system may facilitate the development of the disease. Here, we provide a critical review of the literature on the role of lipids and lipoproteins in MM pathophysiology. We also propose novel mechanisms, linking the development and progression of MM to the metabolism of blood lipoproteins. We anticipate that proteomic and lipidomic analyses of serum lipoproteins along with analyses of their functionality may improve our understanding and shed light on novel mechanistic aspects of MM pathophysiology.
Collapse
Affiliation(s)
- Vasileios Lazaris
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece.,Hematology Clinic, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece
| | - Aikaterini Hatziri
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece
| | - Argiris Symeonidis
- Hematology Clinic, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece
| | - Kyriakos E Kypreos
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
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
|
Faria-Ramos I, Poças J, Marques C, Santos-Antunes J, Macedo G, Reis CA, Magalhães A. Heparan Sulfate Glycosaminoglycans: (Un)Expected Allies in Cancer Clinical Management. Biomolecules 2021; 11:136. [PMID: 33494442 PMCID: PMC7911160 DOI: 10.3390/biom11020136] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
In an era when cancer glycobiology research is exponentially growing, we are witnessing a progressive translation of the major scientific findings to the clinical practice with the overarching aim of improving cancer patients' management. Many mechanistic cell biology studies have demonstrated that heparan sulfate (HS) glycosaminoglycans are key molecules responsible for several molecular and biochemical processes, impacting extracellular matrix properties and cellular functions. HS can interact with a myriad of different ligands, and therefore, hold a pleiotropic role in regulating the activity of important cellular receptors and downstream signalling pathways. The aberrant expression of HS glycan chains in tumours determines main malignant features, such as cancer cell proliferation, angiogenesis, invasion and metastasis. In this review, we devote particular attention to HS biological activities, its expression profile and modulation in cancer. Moreover, we highlight HS clinical potential to improve both diagnosis and prognosis of cancer, either as HS-based biomarkers or as therapeutic targets.
Collapse
Affiliation(s)
- Isabel Faria-Ramos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Juliana Poças
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Catarina Marques
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - João Santos-Antunes
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
- Gastroenterology Department, Centro Hospitalar S. João, 4200-319 Porto, Portugal
| | - Guilherme Macedo
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
- Gastroenterology Department, Centro Hospitalar S. João, 4200-319 Porto, Portugal
| | - Celso A. Reis
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
| |
Collapse
|
20
|
Arnold K, Liao YE, Liu J. Potential Use of Anti-Inflammatory Synthetic Heparan Sulfate to Attenuate Liver Damage. Biomedicines 2020; 8:E503. [PMID: 33207634 PMCID: PMC7697061 DOI: 10.3390/biomedicines8110503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/12/2020] [Accepted: 11/14/2020] [Indexed: 12/28/2022] Open
Abstract
Heparan sulfate is a highly sulfated polysaccharide abundant on the surface of hepatocytes and surrounding extracellular matrix. Emerging evidence demonstrates that heparan sulfate plays an important role in neutralizing the activities of proinflammatory damage associate molecular patterns (DAMPs) that are released from hepatocytes under pathological conditions. Unlike proteins and nucleic acids, isolation of homogenous heparan sulfate polysaccharides from biological sources is not possible, adding difficulty to study the functional role of heparan sulfate. Recent advancement in the development of a chemoenzymatic approach allows production of a large number of structurally defined oligosaccharides. These oligosaccharides are used to probe the physiological functions of heparan sulfate in liver damage under different pathological conditions. The findings provide a potential new therapeutic agent to treat liver diseases that are associated with excessive inflammation.
Collapse
Affiliation(s)
| | | | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; (K.A.); (Y.-E.L.)
| |
Collapse
|
21
|
Santamaria S. ADAMTS-5: A difficult teenager turning 20. Int J Exp Pathol 2020; 101:4-20. [PMID: 32219922 DOI: 10.1111/iep.12344] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/28/2019] [Accepted: 01/19/2020] [Indexed: 12/13/2022] Open
Abstract
A Disintegrin And Metalloproteinase with ThromboSpondin motif (ADAMTS)-5 was identified in 1999 as one of the enzymes responsible for cleaving aggrecan, the major proteoglycan in articular cartilage. Studies in vitro, ex vivo and in vivo have validated ADAMTS-5 as a target in osteoarthritis (OA), a disease characterized by extensive degradation of aggrecan. For this reason, it attracted the interest of many research groups aiming to develop a therapeutic treatment for OA patients. However, ADAMTS-5 proteoglycanase activity is not only involved in the dysregulated aggrecan proteolysis, which occurs in OA, but also in the physiological turnover of other related proteoglycans. In particular, versican, a major ADAMTS-5 substrate, plays an important structural role in heart and blood vessels and its proteolytic processing by ADAMTS-5 must be tightly regulated. On the occasion of the 20th anniversary of the discovery of ADAMTS-5, this review looks at the evidence for its detrimental role in OA, as well as its physiological turnover of cardiovascular proteoglycans. Moreover, the other potential functions of this enzyme are highlighted. Finally, challenges and emerging trends in ADAMTS-5 research are discussed.
Collapse
|
22
|
Morton RE, Liu Y, Izem L. ApoF knockdown increases cholesteryl ester transfer to LDL and impairs cholesterol clearance in fat-fed hamsters. J Lipid Res 2019; 60:1868-1879. [PMID: 31511396 DOI: 10.1194/jlr.ra119000171] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/10/2019] [Indexed: 02/04/2023] Open
Abstract
Cholesteryl ester transfer protein (CETP) regulates intravascular lipoprotein metabolism. In vitro studies indicate that ApoF alters CETP function by inhibiting its activity with LDL. To explore in vivo the complexities driving ApoF's effects on CETP, we developed a siRNA-based hamster model of ApoF knockdown. In both male and female hamsters on chow- or fat-fed diets, we measured lipoprotein levels and composition, determined CETP-mediated transfer of cholesteryl esters (CEs) between lipoproteins, and quantified reverse cholesterol transport (RCT). We found that apoF knockdown in chow-fed hamsters had no effect on lipoprotein levels or composition, but these ApoF-deficient lipoproteins supported 50-100% higher LDL CETP activity in vitro. ApoF knockdown in fat-fed male hamsters created a phenotype in which endogenous CETP-mediated CE transfer from HDL to LDL increased up to 2-fold, LDL cholesterol increased 40%, HDL declined 25%, LDL and HDL lipid compositions were altered, and hepatic LDLR gene expression was decreased. Diet-induced hypercholesterolemia obscured this phenotype on occasion. In fat-fed female hamsters, ApoF knockdown caused similar but smaller changes in plasma CETP activity and LDL cholesterol. Notably, ApoF knockdown impaired HDL RCT in fat-fed hamsters but increased sterol excretion in chow-fed animals. These in vivo data validate in vitro findings that ApoF regulates lipid transfer to LDL. The consequences of ApoF knockdown on lipoproteins and sterol excretion depend on the underlying lipid status. By minimizing the transfer of HDL-derived CE to LDL, ApoF helps control LDL cholesterol levels when LDL clearance mechanisms are limiting.
Collapse
Affiliation(s)
- Richard E Morton
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Yan Liu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Lahoucine Izem
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| |
Collapse
|
23
|
Sibiya S, Msibi B, Khathi A, Sibiya N, Booysen I, Ngubane P. The effect of dioxidovanadium complex (V) on hepatic function in streptozotocin-induced diabetic rats. Can J Physiol Pharmacol 2019; 97:1169-1175. [PMID: 31491333 DOI: 10.1139/cjpp-2019-0369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Diabetics are susceptible to hepatic dysfunction risks due to hyperglycaemia and insulin therapy. Conventional diabetes treatments improve glycaemic control; however, hepatic hazards associated with these agents remains a challenge. Accordingly, this study sought to investigate the effect of a dioxidovanadium complex (V) on the hepatic function in streptozotocin-induced diabetic rats. Sprague-Dawley rats (240-250 g) were divided into 4 groups (n = 6): nondiabetic control, diabetic control, insulin-treated, and vanadium complex groups. The dioxidovanadium (10, 20, and 40 mg/kg) was administered twice every 2nd day for 5 weeks and blood glucose concentration was monitored weekly. At the end of the experimental period, all the experimental groups were sacrificed, and then the lipid profile, liver superoxide dismutase, glutathione peroxidase and malondialdehyde, plasma alanine aminotransferase and aspartate aminotransferase, and C-reactive protein (CRP) concentration were measured. The administration of dioxidovanadium significantly alleviated hyperglycaemia with concomitant attenuation in oxidative stress as evidenced by reduced malondialdehyde concentrations. Furthermore, vanadium complex abolished diabetes-induced dyslipidaemia. Lastly, vanadium complex administration attenuated the increase in alanine aminotransferase, aspartate aminotransferase, and plasma C-reactive protein. These findings suggest that this metallo-compound (dioxidovanadium) may ameliorate liver dysfunction often observed in diabetes.
Collapse
Affiliation(s)
- Samukelisiwe Sibiya
- Schools of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Bonisiwe Msibi
- Schools of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- Schools of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
| | - Irvin Booysen
- School of Chemistry and Physics, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Phikelelani Ngubane
- Schools of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
24
|
Kegulian NC, Ramms B, Horton S, Trenchevska O, Nedelkov D, Graham MJ, Lee RG, Esko JD, Yassine HN, Gordts PLSM. ApoC-III Glycoforms Are Differentially Cleared by Hepatic TRL (Triglyceride-Rich Lipoprotein) Receptors. Arterioscler Thromb Vasc Biol 2019; 39:2145-2156. [PMID: 31390883 DOI: 10.1161/atvbaha.119.312723] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE ApoC-III (apolipoprotein C-III) glycosylation can predict cardiovascular disease risk. Higher abundance of disialylated (apoC-III2) over monosialylated (apoC-III1) glycoforms is associated with lower plasma triglyceride levels. Yet, it remains unclear whether apoC-III glycosylation impacts TRL (triglyceride-rich lipoprotein) clearance and whether apoC-III antisense therapy (volanesorsen) affects distribution of apoC-III glycoforms. Approach and Results: To measure the abundance of human apoC-III glycoforms in plasma over time, human TRLs were injected into wild-type mice and mice lacking hepatic TRL clearance receptors, namely HSPGs (heparan sulfate proteoglycans) or both LDLR (low-density lipoprotein receptor) and LRP1 (LDLR-related protein 1). ApoC-III was more rapidly cleared in the absence of HSPG (t1/2=25.4 minutes) than in wild-type animals (t1/2=55.1 minutes). In contrast, deficiency of LDLR and LRP1 (t1/2=56.1 minutes) did not affect clearance of apoC-III. After injection, a significant increase in the relative abundance of apoC-III2 was observed in HSPG-deficient mice, whereas the opposite was observed in mice lacking LDLR and LRP1. In patients, abundance of plasma apoC-III glycoforms was assessed after placebo or volanesorsen administration. Volanesorsen treatment correlated with a statistically significant 1.4-fold increase in the relative abundance of apoC-III2 and a 15% decrease in that of apoC-III1. The decrease in relative apoC-III1 abundance was strongly correlated with decreased plasma triglyceride levels in patients. CONCLUSIONS Our results indicate that HSPGs preferentially clear apoC-III2. In contrast, apoC-III1 is more effectively cleared by LDLR/LRP1. Clinically, the increase in the apoC-III2/apoC-III1 ratio on antisense lowering of apoC-III might reflect faster clearance of apoC-III1 because this metabolic shift associates with improved triglyceride levels.
Collapse
Affiliation(s)
- Natalie C Kegulian
- From the Department of Medicine, University of Southern California, Los Angeles (N.C.K., S.H., H.N.Y.)
| | - Bastian Ramms
- Department of Medicine (B.R., J.D.E., P.L.S.M.G.), University of California San Diego, La Jolla
- Department of Chemistry, Biochemistry I, Bielefeld University, Germany (B.R.)
| | - Steven Horton
- From the Department of Medicine, University of Southern California, Los Angeles (N.C.K., S.H., H.N.Y.)
| | | | - Dobrin Nedelkov
- The Biodesign Institute, Arizona State University, Tempe (O.T., D.N.)
| | - Mark J Graham
- Ionis Pharmaceuticals, Carlsbad, CA (M.J.G., R.G.L.)
| | - Richard G Lee
- Ionis Pharmaceuticals, Carlsbad, CA (M.J.G., R.G.L.)
| | - Jeffrey D Esko
- Department of Medicine (B.R., J.D.E., P.L.S.M.G.), University of California San Diego, La Jolla
- Glycobiology Research and Training Center (J.D.E., P.L.S.M.G.), University of California San Diego, La Jolla
| | - Hussein N Yassine
- From the Department of Medicine, University of Southern California, Los Angeles (N.C.K., S.H., H.N.Y.)
| | - Philip L S M Gordts
- Department of Medicine (B.R., J.D.E., P.L.S.M.G.), University of California San Diego, La Jolla
- Glycobiology Research and Training Center (J.D.E., P.L.S.M.G.), University of California San Diego, La Jolla
| |
Collapse
|
25
|
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
|
26
|
Poli M, Anower-E-Khuda F, Asperti M, Ruzzenenti P, Gryzik M, Denardo A, Gordts PLSM, Arosio P, Esko JD. Hepatic heparan sulfate is a master regulator of hepcidin expression and iron homeostasis in human hepatocytes and mice. J Biol Chem 2019; 294:13292-13303. [PMID: 31315930 PMCID: PMC6737225 DOI: 10.1074/jbc.ra118.007213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
Hepcidin is a liver-derived peptide hormone that controls systemic iron homeostasis. Its expression is regulated by the bone morphogenetic protein 6 (BMP6)/SMAD1/5/8 pathway and by the proinflammatory cytokine interleukin 6 (IL6). Proteoglycans that function as receptors of these signaling proteins in the liver are commonly decorated by heparan sulfate, but the potential role of hepatic heparan sulfate in hepcidin expression and iron homeostasis is unclear. Here, we show that modulation of hepatic heparan sulfate significantly alters hepcidin expression and iron metabolism both in vitro and in vivo. Specifically, enzymatic removal of heparan sulfate from primary human hepatocytes, CRISPR/Cas9 manipulation of heparan sulfate biosynthesis in human hepatoma cells, or pharmacological manipulation of heparan sulfate–protein interactions using sodium chlorate or surfen dramatically reduced baseline and BMP6/SMAD1/5/8-dependent hepcidin expression. Moreover inactivation of the heparan sulfate biosynthetic gene N-deacetylase and N-sulfotransferase 1 (Ndst1) in murine hepatocytes (Ndst1f/fAlbCre+) reduced hepatic hepcidin expression and caused a redistribution of systemic iron, leading to iron accumulation in the liver and serum of mice. Manipulation of heparan sulfate had a similar effect on IL6-dependent hepcidin expression in vitro and suppressed IL6-mediated iron redistribution induced by lipopolysaccharide in vivo. These results provide compelling evidence that hepatocyte heparan sulfate plays a key role in regulating hepcidin expression and iron homeostasis in mice and in human hepatocytes.
Collapse
Affiliation(s)
- Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Ferdous Anower-E-Khuda
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093
| | - Michela Asperti
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Paola Ruzzenenti
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Magdalena Gryzik
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Andrea Denardo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Philip L S M Gordts
- Glycobiology Research and Training Center, University of California San Diego, La Jolla, California 92093; Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, California 92093
| | - Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093; Glycobiology Research and Training Center, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
27
|
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
|
28
|
Shrestha P, van de Sluis B, Dullaart RP, van den Born J. Novel aspects of PCSK9 and lipoprotein receptors in renal disease-related dyslipidemia. Cell Signal 2019; 55:53-64. [DOI: 10.1016/j.cellsig.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/01/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
|
29
|
Proteoglycan 4 deficiency protects against glucose intolerance and fatty liver disease in diet-induced obese mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:494-501. [DOI: 10.1016/j.bbadis.2018.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
|
30
|
Heparan sulfate S-domains and extracellular sulfatases (Sulfs): their possible roles in protein aggregation diseases. Glycoconj J 2018; 35:387-396. [PMID: 30003471 DOI: 10.1007/s10719-018-9833-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/25/2018] [Accepted: 07/04/2018] [Indexed: 10/28/2022]
Abstract
Highly sulfated domains of heparan sulfate (HS), also known as HS S-domains, consist of repeated trisulfated disaccharide units [iduronic acid (2S)-glucosamine (NS, 6S)-]. The expression of HS S-domains at the cell surface is determined by two mechanisms: tightly regulated biosynthetic machinery and enzymatic remodeling by extracellular endoglucosamine 6-sulfatases, Sulf-1 and Sulf-2. Intracellular or extracellular deposits of misfolded and aggregated proteins are characteristic of protein aggregation diseases. Although proteins can aggregate alone, deposits of protein aggregates in vivo contain a number of proteinaceous and non-protein components. HS S-domains are one non-protein component of these aggregated deposits. HS S-domains are considered to be critical for signal transduction of several growth factors and several disease conditions, such as tumor progression, but their roles in protein aggregation diseases are not yet fully understood. This review summarizes the current understanding of the possible roles of HS S-domains and Sulfs in the formation and cytotoxicity of protein aggregates.
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
|
Kim K, Goldberg IJ, Graham MJ, Sundaram M, Bertaggia E, Lee SX, Qiang L, Haeusler RA, Metzger D, Chambon P, Yao Z, Ginsberg HN, Pajvani UB. γ-Secretase Inhibition Lowers Plasma Triglyceride-Rich Lipoproteins by Stabilizing the LDL Receptor. Cell Metab 2018; 27:816-827.e4. [PMID: 29576536 PMCID: PMC5884729 DOI: 10.1016/j.cmet.2018.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/14/2017] [Accepted: 02/14/2018] [Indexed: 12/31/2022]
Abstract
Excess plasma triglycerides (TGs) are a key component of obesity-induced metabolic syndrome. We have shown that γ-secretase inhibitor (GSI) treatment improves glucose tolerance due to inhibition of hepatic Notch signaling but found additional Notch-independent reduction of plasma TG-rich lipoproteins (TRLs) in GSI-treated, as well as hepatocyte-specific, γ-secretase knockout (L-Ncst) mice, which suggested a primary effect on hepatocyte TRL uptake. Indeed, we found increased VLDL and LDL particle uptake in L-Ncst hepatocytes and Ncst-deficient hepatoma cells, in part through reduced γ-secretase-mediated low-density lipoprotein receptor (LDLR) cleavage and degradation. To exploit this novel finding, we generated a liver-selective Nicastrin ASO, which recapitulated glucose and lipid improvements of L-Ncst mice, with increased levels of hepatocyte LDLR. Collectively, these results identify the role of hepatic γ-secretase to regulate LDLR and suggest that liver-specific GSIs may simultaneously improve multiple aspects of the metabolic syndrome.
Collapse
Affiliation(s)
- KyeongJin Kim
- Department of Medicine, Columbia University, New York, NY, USA
| | - Ira J Goldberg
- Department of Medicine, New York University, New York, NY, USA
| | | | - Meenakshi Sundaram
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Enrico Bertaggia
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Samuel X Lee
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Li Qiang
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Rebecca A Haeusler
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | | | | | - Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | | | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
33
|
Al-Sharea A, Murphy AJ, Huggins LA, Hu Y, Goldberg IJ, Nagareddy PR. SGLT2 inhibition reduces atherosclerosis by enhancing lipoprotein clearance in Ldlr -/- type 1 diabetic mice. Atherosclerosis 2018. [PMID: 29518749 DOI: 10.1016/j.atherosclerosis.2018.02.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND AIMS Leukocytosis, particularly monocytosis, has been shown to promote atherosclerosis in both diabetic and non-diabetic mouse models. We previously showed that hyperglycemia independently promotes monocytosis and impairs the resolution of atherosclerosis. Since patients with chronic diabetes often develop dyslipidemia and also have increased risk for atherosclerosis, we sought to examine how controlling blood glucose affects atherosclerosis development in the presence of severe hyperlipidemia. METHODS Diabetes was induced using streptozotocin (STZ) in low density lipoprotein receptor (Ldlr) knockout (Ldlr-/-) mice after which they were fed a high-cholesterol diet for 4 weeks. Control and diabetic mice were treated with vehicle or sodium glucose cotransporter inhibitor (SGLT2i, Phlorizin or Dapagliflozin) for the duration of the diet. RESULTS Induction of diabetes resulted in a dramatic increase in plasma cholesterol (TC) and triglyceride (TG) levels. These mice also exhibited an increased number of circulating monocytes and neutrophils. Monocytosis was driven by increased proliferation of progenitor cells in the bone marrow. Tighter glycemic control by SGLT2i treatment not only reduced monocytosis and atherosclerosis but also improved plasma lipoprotein profile. Interestingly, improved lipoprotein profile was not due to decreased TG synthesis or clearance via low density lipoprotein receptor-related protein (Lrp) 1 or scavenger receptor class B member (Scarb1) pathways, but likely mediated by heparin sulfate proteoglycans (HSPG)-dependent clearance mechanisms in the liver. Further examination of the liver revealed an important role for bile acid transporters (Abcg5, Abcg8) and cytochrome P450 enzymes in the clearance of hepatic cholesterol. CONCLUSIONS These data suggest that tighter glycemic control in diabetes can improve lipoprotein clearance exclusive of Ldlr, likely via HSPG and bile acid pathways, and has an overall net positive effect on atherosclerosis.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- Animals
- Atherosclerosis/blood
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Benzhydryl Compounds/pharmacology
- Biomarkers/blood
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Cholesterol, Dietary
- Cytochrome P-450 Enzyme System/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/drug therapy
- Glucosides/pharmacology
- Heparin/analogs & derivatives
- Heparin/metabolism
- Hyperlipidemias/blood
- Hyperlipidemias/drug therapy
- Hyperlipidemias/genetics
- Lipoproteins/blood
- Lipoproteins/metabolism
- Liver/drug effects
- Liver/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/metabolism
- Phlorhizin/pharmacology
- Proteoglycans/metabolism
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Streptozocin
Collapse
Affiliation(s)
| | - Andrew J Murphy
- Division of Preventive Medicine and Nutrition, Department of Medicine, Columbia University, New York, USA; Baker Heart and Diabetes Institute, Melbourne, Australia
| | - L A Huggins
- Division of Preventive Medicine and Nutrition, Department of Medicine, Columbia University, New York, USA; Dept. of Medicine, New York University, New York, USA
| | - Y Hu
- Division of Preventive Medicine and Nutrition, Department of Medicine, Columbia University, New York, USA; Dept. of Medicine, New York University, New York, USA
| | - Ira J Goldberg
- Division of Preventive Medicine and Nutrition, Department of Medicine, Columbia University, New York, USA; Dept. of Medicine, New York University, New York, USA
| | - Prabhakara R Nagareddy
- Division of Preventive Medicine and Nutrition, Department of Medicine, Columbia University, New York, USA; Dept. of Nutrition Sciences, University of Alabama at Birmingham, USA.
| |
Collapse
|
34
|
Ehrhardt N, Doche ME, Chen S, Mao HZ, Walsh MT, Bedoya C, Guindi M, Xiong W, Ignatius Irudayam J, Iqbal J, Fuchs S, French SW, Mahmood Hussain M, Arditi M, Arumugaswami V, Péterfy M. Hepatic Tm6sf2 overexpression affects cellular ApoB-trafficking, plasma lipid levels, hepatic steatosis and atherosclerosis. Hum Mol Genet 2018; 26:2719-2731. [PMID: 28449094 DOI: 10.1093/hmg/ddx159] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 04/21/2017] [Indexed: 12/15/2022] Open
Abstract
The human transmembrane 6 superfamily member 2 (TM6SF2) gene has been implicated in plasma lipoprotein metabolism, alcoholic and non-alcoholic fatty liver disease and myocardial infarction in multiple genome-wide association studies. To investigate the role of Tm6sf2 in metabolic homeostasis, we generated mice with elevated expression using adeno-associated virus (AAV)-mediated gene delivery. Hepatic overexpression of mouse Tm6sf2 resulted in phenotypes previously observed in Tm6sf2-deficient mice including reduced plasma lipid levels, diminished hepatic triglycerides secretion and increased hepatosteatosis. Furthermore, increased hepatic Tm6sf2 expression protected against the development of atherosclerosis in LDL-receptor/ApoB48-deficient mice. In cultured human hepatocytes, Tm6sf2 overexpression reduced apolipoprotein B secretion and resulted in its accumulation within the endoplasmic reticulum (ER) suggesting impaired ER-to-Golgi trafficking of pre-very low-density lipoprotein (VLDL) particles. Analysis of two metabolic trait-associated coding polymorphisms in the human TM6SF2 gene (rs58542926 and rs187429064) revealed that both variants impact TM6SF2 expression by affecting the rate of protein turnover. These data demonstrate that rs58542926 (E167K) and rs187429064 (L156P) are functional variants and suggest that they influence metabolic traits through altered TM6SF2 protein stability. Taken together, our results indicate that cellular Tm6sf2 level is an important determinant of VLDL metabolism and further implicate TM6SF2 as a causative gene underlying metabolic disease and trait associations at the 19p13.11 locus.
Collapse
Affiliation(s)
- Nicole Ehrhardt
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | | | - Shuang Chen
- Department of Biomedical Sciences.,Department of Pediatrics.,Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hui Z Mao
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Meghan T Walsh
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Candy Bedoya
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Maha Guindi
- Department of Pathology and Laboratory Medicine
| | - Weidong Xiong
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joseph Ignatius Irudayam
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jahangir Iqbal
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Sebastien Fuchs
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Samuel W French
- Department of Pathology and Laboratory Medicine.,Jonsson Comprehensive Cancer Center.,UCLA AIDS Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA.,Winthrop-University Hospital, Mineola, NY 11501, USA
| | - Moshe Arditi
- Department of Biomedical Sciences.,Department of Pediatrics.,Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Pediatrics
| | - Vaithilingaraja Arumugaswami
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Surgery
| | - Miklós Péterfy
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA.,Department of Biomedical Sciences.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
35
|
Phan AQ, Pacifici M, Esko JD. Advances in the pathogenesis and possible treatments for multiple hereditary exostoses from the 2016 international MHE conference. Connect Tissue Res 2018; 59:85-98. [PMID: 29099240 PMCID: PMC7604901 DOI: 10.1080/03008207.2017.1394295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multiple hereditary exostoses (MHE) is an autosomal dominant disorder that affects about 1 in 50,000 children worldwide. MHE, also known as hereditary multiple exostoses (HME) or multiple osteochondromas (MO), is characterized by cartilage-capped outgrowths called osteochondromas that develop adjacent to the growth plates of skeletal elements in young patients. These benign tumors can affect growth plate function, leading to skeletal growth retardation, or deformations, and can encroach on nerves, tendons, muscles, and other surrounding tissues and cause motion impairment, chronic pain, and early onset osteoarthritis. In about 2-5% of patients, the osteochondromas can become malignant and life threatening. Current treatments consist of surgical removal of the most symptomatic tumors and correction of the major skeletal defects, but physical difficulties and chronic pain usually continue and patients may undergo multiple surgeries throughout life. Thus, there is an urgent need to find new treatments to prevent or reverse osteochondroma formation. The 2016 International MHE Research Conference was convened to provide a forum for the presentation of the most up-to-date and advanced clinical and basic science data and insights in MHE and related fields; to stimulate the forging of new perspectives, collaborations, and venues of research; and to publicize key scientific findings within the biomedical research community and share insights and relevant information with MHE patients and their families. This report provides a description, review, and assessment of all the exciting and promising studies presented at the Conference and delineates a general roadmap for future MHE research targets and goals.
Collapse
Affiliation(s)
- Anne Q. Phan
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, 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
|
36
|
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
|
37
|
Effects of atorvastatin and diet interventions on atherosclerotic plaque inflammation and [18F]FDG uptake in Ldlr−/−Apob mice. Atherosclerosis 2017; 263:369-376. [DOI: 10.1016/j.atherosclerosis.2017.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
|
38
|
Diet-induced hypercholesterolemia alters liver glycosaminoglycans and associated-lipoprotein receptors in rats. J Physiol Biochem 2017; 73:539-550. [DOI: 10.1007/s13105-017-0583-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/18/2017] [Indexed: 10/19/2022]
|
39
|
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
|
40
|
Sustained Placental Growth Factor-2 Treatment Does Not Aggravate Advanced Atherosclerosis in Ischemic Cardiomyopathy. J Cardiovasc Transl Res 2017; 10:348-358. [DOI: 10.1007/s12265-017-9742-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/14/2017] [Indexed: 12/17/2022]
|
41
|
Abstract
Simple and complex carbohydrates (glycans) have long been known to play major metabolic, structural and physical roles in biological systems. Targeted microbial binding to host glycans has also been studied for decades. But such biological roles can only explain some of the remarkable complexity and organismal diversity of glycans in nature. Reviewing the subject about two decades ago, one could find very few clear-cut instances of glycan-recognition-specific biological roles of glycans that were of intrinsic value to the organism expressing them. In striking contrast there is now a profusion of examples, such that this updated review cannot be comprehensive. Instead, a historical overview is presented, broad principles outlined and a few examples cited, representing diverse types of roles, mediated by various glycan classes, in different evolutionary lineages. What remains unchanged is the fact that while all theories regarding biological roles of glycans are supported by compelling evidence, exceptions to each can be found. In retrospect, this is not surprising. Complex and diverse glycans appear to be ubiquitous to all cells in nature, and essential to all life forms. Thus, >3 billion years of evolution consistently generated organisms that use these molecules for many key biological roles, even while sometimes coopting them for minor functions. In this respect, glycans are no different from other major macromolecular building blocks of life (nucleic acids, proteins and lipids), simply more rapidly evolving and complex. It is time for the diverse functional roles of glycans to be fully incorporated into the mainstream of biological sciences.
Collapse
Affiliation(s)
- Ajit Varki
- Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center, University of California at San Diego, La Jolla, CA 92093-0687, USA
| |
Collapse
|
42
|
Affiliation(s)
- Che C. Colpitts
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques and Université de Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques and Université de Strasbourg and Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Hopitaux, Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
43
|
Viecili PRN, da Silva B, Hirsch GE, Porto FG, Parisi MM, Castanho AR, Wender M, Klafke JZ. Triglycerides Revisited to the Serial. Adv Clin Chem 2017; 80:1-44. [PMID: 28431638 DOI: 10.1016/bs.acc.2016.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review discusses the role of triglycerides (TGs) in the normal cardiovascular system as well as in the development and clinical manifestation of cardiovascular diseases. Regulation of TGs at the enzymatic and genetic level, in addition to their possible relevance as preclinical and clinical biomarkers, is discussed, culminating with a description of available and emerging treatments. Due to the high complexity of the subject and the vast amount of material in the literature, the objective of this review was not to exhaust the subject, but rather to compile the information to facilitate and improve the understanding of those interested in this topic. The main publications on the topic were sought out, especially those from the last 5 years. The data in the literature still give reason to believe that there is room for doubt regarding the use of TG as disease biomarkers; however, there is increasing evidence for the role of hypertriglyceridemia on the atherosclerotic inflammatory process, cardiovascular outcomes, and mortality.
Collapse
|
44
|
Bauters D, Spincemaille P, Geys L, Cassiman D, Vermeersch P, Bedossa P, Scroyen I, Lijnen HR. ADAMTS5 deficiency protects against non-alcoholic steatohepatitis in obesity. Liver Int 2016; 36:1848-1859. [PMID: 27254774 DOI: 10.1111/liv.13181] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/31/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Increased prevalence of obesity is paralleled by an increase in non-alcoholic steatohepatitis (NASH). We previously found that the expression of ADAMTS5 (A Disintegrin And Metalloproteinase with Thrombospondin type 1 motifs; member 5) is enhanced in expanding adipose tissue. However, no information is available on a potential role in liver pathology. We studied the effect of ADAMTS5 deficiency on NASH in mice. METHODS Wild-type (Adamts5+/+ ) and deficient (Adamts5-/- ) mice were kept on a standard- or high-fat diet (HFD) for 15 weeks. Alternatively, steatohepatitis was induced with methionine/choline-deficient (MCD) diet. RESULTS HFD feeding resulted in comparable body weights for both genotypes, but Adamts5-/- mice had approximately 40% lower liver weight (P = 0.0004). In the Adamts5-/- mice, the HFD as well as the MCD diet consistently induced less NASH with less fibrosis. The deteriorating effect of ADAMTS5 on the liver during diet-induced obesity may be due, at least in part, to proteolytic cleavage of the matrix components syndecan-1 and versican, thereby enhancing hepatic triglyceride clearance from the circulation. Plasma lipid levels were elevated in obese Adamts5-/- mice. There was no clear effect of ADAMTS5 deficiency on glycaemia or glucose tolerance, whereas insulin sensitivity was somewhat improved. Furthermore, Adamts5-/- mice were protected from hepatic mitochondrial dysfunction, as indicated by increased mitochondrial respiratory chain complex activity, higher ATP levels and higher expression of antioxidant enzymes. CONCLUSIONS Absence of ADAMTS5 preserves liver integrity in a diet-induced obesity model. Selective targeting of ADAMTS5 could provide a new therapeutic strategy for treatment/prevention of NASH.
Collapse
Affiliation(s)
- Dries Bauters
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Pieter Spincemaille
- Lab of Hepatology, University of Leuven, Leuven, Belgium.,Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Lotte Geys
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - David Cassiman
- Department of Hepatology and Metabolic Center, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Vermeersch
- Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium.,Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Pierre Bedossa
- Department of Pathology, Hôpital Beaujon, Clichy, France
| | - Ilse Scroyen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Henri R Lijnen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
45
|
Effect of pathological conditions on peritoneal macrophage glycosaminoglycans: Impact on cytoadherence. Int J Biol Macromol 2016; 92:1183-1190. [DOI: 10.1016/j.ijbiomac.2016.08.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 08/07/2016] [Accepted: 08/08/2016] [Indexed: 12/20/2022]
|
46
|
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
|
47
|
Shen J, Tong X, Sud N, Khound R, Song Y, Maldonado-Gomez MX, Walter J, Su Q. Low-Density Lipoprotein Receptor Signaling Mediates the Triglyceride-Lowering Action of
Akkermansia muciniphila
in Genetic-Induced Hyperlipidemia. Arterioscler Thromb Vasc Biol 2016; 36:1448-56. [DOI: 10.1161/atvbaha.116.307597] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/16/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Jing Shen
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Xuedong Tong
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Neetu Sud
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Rituraj Khound
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Yongyan Song
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Maria X. Maldonado-Gomez
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Jens Walter
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| | - Qiaozhu Su
- From the Departments of Nutrition and Health Sciences (J.S., X.T., N.S., R.K., Y.S., Q.S.) and Food Science and Technology (M.X.M.-G.), University of Nebraska-Lincoln; and Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada (J.W.)
| |
Collapse
|
48
|
Fonseca P, Vardaki I, Occhionero A, Panaretakis T. Metabolic and Signaling Functions of Cancer Cell-Derived Extracellular Vesicles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:175-99. [PMID: 27572129 DOI: 10.1016/bs.ircmb.2016.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles have gained tremendous attention in the recent years as a novel mechanism of cell to cell communication. There are several types of extracellular vesicles, including exosomes, microvesicles, exosome, like vesicles, apoptotic bodies that differ mainly in the mechanism of biogenesis and secretion. The most well studied type of extracellular vesicles are the exosomes which are endosome-derived vesicles with a diameter of 50-150nm and enriched in ESCRT proteins including Alix, TSG101, Hsp70, and tetraspanins. It is now well established that exosomes promote tumor growth, alter the tumor microenvironment, facilitate the dissemination of cancer cells in an organotropic manner, modulate immune responses, and mediate resistance to therapy. Exosomes have also been recently implicated in an emerging hallmark of cancer, the cancer cell metabolism. The metabolic state of the cell defines, to a certain extent, both the rate of secretion and the molecular content of tumor-derived exosomes. Furthermore, exosomes have been shown to possess intrinsic metabolic activity since they can synthesize ATP by glycolysis. It follows that exosomes carry a number of metabolic enzymes and metabolites, including lactate, PGE, LDH isoforms, pyruvate, and monocarboxylate transporters. Last but not the least, exosomes are implicated in fatty acid synthesis and cholesterol metabolism and are thought to be crucial for the transcellular metabolism procedure. Uptake of exosomes is thought to alter the intracellular metabolic state of the cell. In summary, we describe the state of the art on the role of metabolism in the secretion, uptake, and the biological effects of exosomes in the metabolism of recipient cells.
Collapse
Affiliation(s)
- P Fonseca
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - I Vardaki
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - A Occhionero
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - T Panaretakis
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| |
Collapse
|
49
|
Li JP, Kusche-Gullberg M. Heparan Sulfate: Biosynthesis, Structure, and Function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 325:215-73. [PMID: 27241222 DOI: 10.1016/bs.ircmb.2016.02.009] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Heparan sulfate (HS) proteoglycans (PGs) are ubiquitously expressed on cell surfaces and in the extracellular matrix of most animal tissues, having essential functions in development and homeostasis, as well as playing various roles in disease processes. The functions of HSPGs are mainly dependent on interactions between the HS-side chains with a variety of proteins including cytokines, growth factors, and their receptors. In a given HS polysaccharide, negatively charged sulfate and carboxylate groups are arranged in various types of domains, generated through strictly regulated biosynthetic reactions and with enormous potential for structural variability. The mode of HS-protein interactions is assessed through binding experiments using saccharides of defined composition in vitro, signaling assays in cell models where HS structures are manipulated, and targeted disruption of genes for biosynthetic enzymes in animals (mouse, zebrafish, Drosophila, and Caenorhabditis elegans) followed by phenotype analysis. Whereas some protein ligands appear to require strictly defined HS structure, others bind to variable saccharide domains without apparent dependence on distinct saccharide sequence. These findings raise intriguing questions concerning the functional significance of regulation in HS biosynthesis and the potential for development of therapeutics targeting HS-protein interactions.
Collapse
Affiliation(s)
- J-P Li
- Department of Medical Biochemistry and Microbiology, University of Uppsala, Uppsala, Sweden; SciLifeLab, University of Uppsala, Uppsala, Sweden.
| | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW This article summarizes the current evidence to support a role of sulfatase 2 (SULF2) in triglyceride-rich lipoprotein (TRL) metabolism and angiogenesis. RECENT FINDINGS Heparan sulfate proteoglycans (HSPG) are involved in the hepatic clearance of TRLs in mice and in humans. Different genetically modified mouse models have been instrumental to provide evidence that syndecan1, the core protein of HSPG, but also the degree of sulfation of the heparin sulfate chain, attached to syndecan 1, is important for hepatic TRL metabolism. Studies in humans demonstrate the regulating role of SULF2 in the hepatic uptake of TRL by HSPG and demonstrate the importance of 6-O-sulfation, modulated by SULF2, for HSPG function. The role of SULF2 in angiogenesis is illustrated by increased SULF2 mRNA expression in the stalk cells of angiogenic vascular sprouts that use fatty acids derived from TRL as a source for biomass production. Interestingly, SULF2 also interferes with HSPG-vascular endothelial growth factor binding, which impacts upon the angiogenic properties of stalk cells. SUMMARY SULF2 is a multifaceted protein involved in TRL homeostasis and angiogenesis. Future investigations should focus on the potential benefits of targeting SULF2 in atherosclerosis and angiogenesis.
Collapse
Affiliation(s)
- Marchien G Dallinga
- aDepartment of Ophthalmology bDepartment of Vascular cDepartment of Experimental Vascular Medicine, Academic Medical Center Amsterdam, University of Amsterdam, The Netherlands
| | | |
Collapse
|