51
|
Zimmerman B, Kundu P, Rooney WD, Raber J. The Effect of High Fat Diet on Cerebrovascular Health and Pathology: A Species Comparative Review. Molecules 2021; 26:3406. [PMID: 34199898 PMCID: PMC8200075 DOI: 10.3390/molecules26113406] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023] Open
Abstract
In both humans and animal models, consumption of a high-saturated-fat diet has been linked to vascular dysfunction and cognitive impairments. Laboratory animals provide excellent models for more invasive high-fat-diet-related research. However, the physiological differences between humans and common animal models in terms of how they react metabolically to high-fat diets need to be considered. Here, we review the factors that may affect the translatability of mechanistic research in animal models, paying special attention to the effects of a high-fat diet on vascular outcomes. We draw attention to the dissociation between metabolic syndrome and dyslipidemia in rodents, unlike the state in humans, where the two commonly occur. We also discuss the differential vulnerability between species to the metabolic and vascular effects of macronutrients in the diet. Findings from animal studies are better interpreted as modeling specific aspects of dysfunction. We conclude that the differences between species provide an opportunity to explore why some species are protected from the detrimental aspects of high-fat-diet-induced dysfunction, and to translate these findings into benefits for human health.
Collapse
Affiliation(s)
- Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
| | - William D. Rooney
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
52
|
Proprotein Convertase Subtilisin/Kexin Type 9 Loss-of-Function Is Detrimental to the Juvenile Host With Septic Shock. Crit Care Med 2021; 48:1513-1520. [PMID: 32769621 DOI: 10.1097/ccm.0000000000004487] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Proprotein convertase subtilisin/kexin type 9 is a central regulator of lipid metabolism and has been implicated in regulating the host response to sepsis. Proprotein convertase subtilisin/kexin type 9 loss-of-function is associated with improved sepsis outcomes in the adult host through increased hepatic bacterial clearance. Thus, there is interest in leveraging proprotein convertase subtilisin/kexin type 9 inhibitors as a therapeutic strategy in adults with sepsis. We sought to validate this association in children with septic shock and in a juvenile murine model of sepsis. DESIGN Prospectively enrolled cohort of children with septic shock; experimental mice. SETTING Seventeen participating institutions; research laboratory. PATIENTS AND SUBJECTS Five-hundred twenty-two children with septic shock; juvenile (14 d old) and adult (10-14 wk) mice with constitutive proprotein convertase subtilisin/kexin type 9 null and wildtype control mice (C57BL/6). INTERVENTIONS Proprotein convertase subtilisin/kexin type 9 single-nucleotide polymorphisms, serum proprotein convertase subtilisin/kexin type 9, and lipid profiles in patients. Cecal slurry murine model of sepsis; survival studies in juvenile and adult mice, assessment of lipoprotein fractions, bacterial burden, and inflammation in juvenile mice. MEASUREMENTS AND MAIN RESULTS PCSK9 loss-of-function genetic variants were independently associated with increased odds of complicated course and mortality in children with septic shock. PCSK9, low-density lipoprotein, and high-density lipoprotein concentrations were lower among patients with complicated course relative to those without. PCSK9 concentrations negatively correlated with proinflammatory cytokine interleukin-8. Proprotein convertase subtilisin/kexin type 9 loss-of-function decreased survival in juvenile mice, but increased survival in adult mice with sepsis. PCSK9 loss-of-function resulted in low lipoproteins and decreased hepatic bacterial burden in juvenile mice. CONCLUSIONS In contrast to the adult host, proprotein convertase subtilisin/kexin type 9 loss-of-function is detrimental to the juvenile host with septic shock. PCSK9 loss-of-function, in the context of low lipoproteins, may result in reduced hepatic bacterial clearance in the juvenile host with septic shock. Our data indicate that children should be excluded in sepsis clinical trials involving proprotein convertase subtilisin/kexin type 9 inhibitors.
Collapse
|
53
|
Syed S, Nissilä E, Ruhanen H, Fudo S, Gaytán MO, Sihvo SP, Lorey MB, Metso J, Öörni K, King SJ, Oommen OP, Jauhiainen M, Meri S, Käkelä R, Haapasalo K. Streptococcus pneumoniae pneumolysin and neuraminidase A convert high-density lipoproteins into pro-atherogenic particles. iScience 2021; 24:102535. [PMID: 34124613 PMCID: PMC8175417 DOI: 10.1016/j.isci.2021.102535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/19/2021] [Accepted: 05/10/2021] [Indexed: 11/19/2022] Open
Abstract
High-density lipoproteins (HDLs) are a group of different subpopulations of sialylated particles that have an essential role in the reverse cholesterol transport (RCT) pathway. Importantly, changes in the protein and lipid composition of HDLs may lead to the formation of particles with reduced atheroprotective properties. Here, we show that Streptococcus pneumoniae pneumolysin (PLY) and neuraminidase A (NanA) impair HDL function by causing chemical and structural modifications of HDLs. The proteomic, lipidomic, cellular, and biochemical analysis revealed that PLY and NanA induce significant changes in sialic acid, protein, and lipid compositions of HDL. The modified HDL particles have reduced cholesterol acceptor potential from activated macrophages, elevated levels of malondialdehyde adducts, and show significantly increased complement activating capacity. These results suggest that accumulation of these modified HDL particles in the arterial intima may present a trigger for complement activation, inflammatory response, and thereby promote atherogenic disease progression. S. pneumoniae molecules PLY and NanA target human high-density lipoprotein (HDL). These interactions result in major modifications in the HDL proteome and lipidome. Microbially modified HDL activates humoral and cell-mediated innate immune responses. The activated immune response mediates formation of pro-atherogenic epitopes on HDL.
Collapse
Affiliation(s)
- Shahan Syed
- Department of Bacteriology and Immunology, University of Helsinki, 00014 Helsinki, Finland
| | - Eija Nissilä
- Department of Bacteriology and Immunology, University of Helsinki, 00014 Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute for Life Science (HiLIFE) and Biocenter Finland, Helsinki 00014, Finland
| | - Satoshi Fudo
- Department of Bacteriology and Immunology, University of Helsinki, 00014 Helsinki, Finland
| | - Meztlli O. Gaytán
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Sanna P. Sihvo
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute for Life Science (HiLIFE) and Biocenter Finland, Helsinki 00014, Finland
| | | | - Jari Metso
- Minerva Foundation Institute for Medical Research, Biomedicum, 00290 Helsinki, Finland
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
| | | | - Samantha J. King
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Oommen P. Oommen
- Bioengineering and Nanomedicine Lab, Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, 33720 Tampere, Finland
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Biomedicum, 00290 Helsinki, Finland
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki, 00014 Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute for Life Science (HiLIFE) and Biocenter Finland, Helsinki 00014, Finland
| | - Karita Haapasalo
- Department of Bacteriology and Immunology, University of Helsinki, 00014 Helsinki, Finland
- Corresponding author
| |
Collapse
|
54
|
Tou JC, Gucciardi E, Young I. Lipid-modifying effects of lean fish and fish-derived protein consumption in humans: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2021; 80:91-112. [PMID: 33942085 DOI: 10.1093/nutrit/nuab003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/21/2020] [Accepted: 01/15/2021] [Indexed: 12/28/2022] Open
Abstract
CONTEXT Consumption of lean fish and fish-derived proteins were effective for improving lipid profiles in published studies; however, evidence remains inconclusive. OBJECTIVE To evaluate the effectiveness of lean fish or fish-derived protein on serum/plasma lipid and lipoprotein levels by conducting a systematic review of the literature and meta-analysis of available randomized controlled trials (RCTs). DATA SOURCES Medline (Ovid), Scopus, CINAHL, and Food and Nutritional Sciences databases were searched from the start date of each database to September 2019 to identify RCTs determining the effect of lean fish on lipid profile. STUDY SELECTION INCLUDED RCTs investigated lean fish and fish-derived proteins intake and determined at least 1 major lipid or lipoprotein measurement. DATA EXTRACTION Two reviewers independently evaluated 1217 studies against the inclusion and exclusion criteria. Relevant studies were assessed for risks of bias, and random-effects meta-analysis was conducted to generate average estimates of effect. RESULTS A total of 24 studies met the inclusion criteria. Meta-analysis of data from 18 to 21 eligible crossover and parallel-design RCTs with a total of 1392 to 1456 participants found triacylglycerol-lowering effects for lean fish compared with no fish consumption. Lean fish intake showed no significant differences related to total cholesterol or lipoprotein levels. Subanalysis showed that parallel-group RCTs tended to find greater reduction effects on circulating triacylglycerol than did crossover RCTs. CONCLUSION Additional better-designed, longer, and larger RCTs, particularly crossover RCTs, are needed to clarify the impact of lean fish and fish proteins on the serum/plasma lipid profile. Findings from such studies would enable practitioners to provide their patients evidence-based recommendations to meet the American Heart Association guidelines for fish consumption to reduce cardiovascular disease risk.
Collapse
Affiliation(s)
- Janet C Tou
- J.C. Tou is with the Human Nutrition and Foods, Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, USA. E. Gucciardi is with the School of Nutrition, Ryerson University, Toronto, Ontario, Canada. I. Young is with the School of Occupational Health and Public Health, Ryerson University, Toronto, Ontario, Canada
| | - Enza Gucciardi
- J.C. Tou is with the Human Nutrition and Foods, Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, USA. E. Gucciardi is with the School of Nutrition, Ryerson University, Toronto, Ontario, Canada. I. Young is with the School of Occupational Health and Public Health, Ryerson University, Toronto, Ontario, Canada
| | - Ian Young
- J.C. Tou is with the Human Nutrition and Foods, Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, USA. E. Gucciardi is with the School of Nutrition, Ryerson University, Toronto, Ontario, Canada. I. Young is with the School of Occupational Health and Public Health, Ryerson University, Toronto, Ontario, Canada
| |
Collapse
|
55
|
Trabjerg MS, Andersen DC, Huntjens P, Oklinski KE, Bolther L, Hald JL, Baisgaard AE, Mørk K, Warming N, Kullab UB, Kroese LJ, Pritchard CEJ, Huijbers IJ, Nieland JDV. Downregulating carnitine palmitoyl transferase 1 affects disease progression in the SOD1 G93A mouse model of ALS. Commun Biol 2021; 4:509. [PMID: 33931719 PMCID: PMC8087699 DOI: 10.1038/s42003-021-02034-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 03/26/2021] [Indexed: 02/03/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease characterized by death of motor neurons. The etiology and pathogenesis remains elusive despite decades of intensive research. Herein, we report that dysregulated metabolism plays a central role in the SOD1 G93A mouse model mimicking ALS. Specifically, we report that the activity of carnitine palmitoyl transferase 1 (CPT1) lipid metabolism is associated with disease progression. Downregulation of CPT1 activity by pharmacological and genetic methods results in amelioration of disease symptoms, inflammation, oxidative stress and mitochondrial function, whereas upregulation by high-fat diet or corticosterone results in a more aggressive disease progression. Finally, we show that downregulating CPT1 shifts the gut microbiota communities towards a protective phenotype in SOD1 G93A mice. These findings reveal that metabolism, and specifically CPT1 lipid metabolism plays a central role in the SOD1 G93A mouse model and shows that CPT1 might be a therapeutic target in ALS.
Collapse
Affiliation(s)
| | | | - Pam Huntjens
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - Luise Bolther
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Jonas Laugård Hald
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - Kasper Mørk
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Nikolaj Warming
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Ulla Bismark Kullab
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lona John Kroese
- Mouse Clinic for Cancer and Aging Research, Transgenic Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Colin Eliot Jason Pritchard
- Mouse Clinic for Cancer and Aging Research, Transgenic Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ivo Johan Huijbers
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
56
|
Tomé-Carneiro J, Crespo MC, López de Las Hazas MC, Visioli F, Dávalos A. Olive oil consumption and its repercussions on lipid metabolism. Nutr Rev 2021; 78:952-968. [PMID: 32299100 DOI: 10.1093/nutrit/nuaa014] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Consumption of highly processed foods, such as those high in trans fats and free sugars, coupled with sedentarism and chronic stress increases the risk of obesity and cardiometabolic disorders, while adherence to a Mediterranean diet is inversely associated with the prevalence of such diseases. Olive oil is the main source of fat in the Mediterranean diet. Data accumulated thus far show consumption of extra virgin, (poly)phenol-rich olive oil to be associated with specific health benefits. Of note, recommendations for consumption based on health claims refer to the phenolic content of extra virgin olive oil as beneficial. However, even though foods rich in monounsaturated fatty acids, such as olive oil, are healthier than foods rich in saturated and trans fats, their inordinate use can lead to adverse effects on health. The aim of this review was to summarize the data on olive oil consumption worldwide and to critically examine the literature on the potential adverse effects of olive oil and its main components, particularly any effects on lipid metabolism. As demonstrated by substantial evidence, extra virgin olive oil is healthful and should be preferentially used within the context of a balanced diet, but excessive consumption may lead to adverse consequences.
Collapse
Affiliation(s)
- João Tomé-Carneiro
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence UAM + CSIC, Madrid, Spain
| | - María Carmen Crespo
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence UAM + CSIC, Madrid, Spain
| | - María Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence UAM + CSIC, Madrid, Spain
| | - Francesco Visioli
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence UAM + CSIC, Madrid, Spain.,Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence UAM + CSIC, Madrid, Spain
| |
Collapse
|
57
|
Lin M, Li M, Zheng H, Sun H, Zhang J. Lipoprotein proteome profile: novel insight into hyperlipidemia. Clin Transl Med 2021; 11:e361. [PMID: 33931962 PMCID: PMC8032137 DOI: 10.1002/ctm2.361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 11/09/2022] Open
Affiliation(s)
- Miao Lin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Menglin Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Hao Zheng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Haidan Sun
- Core facility of instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jinlan Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
58
|
Hannan FM, Stevenson M, Bayliss AL, Stokes VJ, Stewart M, Kooblall KG, Gorvin CM, Codner G, Teboul L, Wells S, Thakker RV. Ap2s1 mutation causes hypercalcaemia in mice and impairs interaction between calcium-sensing receptor and adaptor protein-2. Hum Mol Genet 2021; 30:880-892. [PMID: 33729479 PMCID: PMC8165646 DOI: 10.1093/hmg/ddab076] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/09/2021] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
Adaptor protein 2 (AP2), a heterotetrameric complex comprising AP2α, AP2β2, AP2μ2 and AP2σ2 subunits, is ubiquitously expressed and involved in endocytosis and trafficking of membrane proteins, such as the calcium-sensing receptor (CaSR), a G-protein coupled receptor that signals via Gα11. Mutations of CaSR, Gα11 and AP2σ2, encoded by AP2S1, cause familial hypocalciuric hypercalcaemia types 1–3 (FHH1–3), respectively. FHH3 patients have heterozygous AP2S1 missense Arg15 mutations (p.Arg15Cys, p.Arg15His or p.Arg15Leu) with hypercalcaemia, which may be marked and symptomatic, and occasional hypophosphataemia and osteomalacia. To further characterize the phenotypic spectrum and calcitropic pathophysiology of FHH3, we used CRISPR/Cas9 genome editing to generate mice harboring the AP2S1 p.Arg15Leu mutation, which causes the most severe FHH3 phenotype. Heterozygous (Ap2s1+/L15) mice were viable, and had marked hypercalcaemia, hypermagnesaemia, hypophosphataemia, and increases in alkaline phosphatase activity and fibroblast growth factor-23. Plasma 1,25-dihydroxyvitamin D was normal, and no alterations in bone mineral density or bone turnover were noted. Homozygous (Ap2s1L15/L15) mice invariably died perinatally. Co-immunoprecipitation studies showed that the AP2S1 p.Arg15Leu mutation impaired protein–protein interactions between AP2σ2 and the other AP2 subunits, and also with the CaSR. Cinacalcet, a CaSR positive allosteric modulator, decreased plasma calcium and parathyroid hormone concentrations in Ap2s1+/L15 mice, but had no effect on the diminished AP2σ2-CaSR interaction in vitro. Thus, our studies have established a mouse model that is representative for FHH3 in humans, and demonstrated that the AP2S1 p.Arg15Leu mutation causes a predominantly calcitropic phenotype, which can be ameliorated by treatment with cinacalcet.
Collapse
Affiliation(s)
- Fadil M Hannan
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Mark Stevenson
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Asha L Bayliss
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Victoria J Stokes
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Michelle Stewart
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Kreepa G Kooblall
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Caroline M Gorvin
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Gemma Codner
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Lydia Teboul
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Sara Wells
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, UK
| |
Collapse
|
59
|
Koike T, Koike Y, Yang D, Guo Y, Rom O, Song J, Xu J, Chen Y, Wang Y, Zhu T, Garcia-Barrio MT, Fan J, Chen YE, Zhang J. Human apolipoprotein A-II reduces atherosclerosis in knock-in rabbits. Atherosclerosis 2021; 316:32-40. [PMID: 33296791 PMCID: PMC7770079 DOI: 10.1016/j.atherosclerosis.2020.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Apolipoprotein A-II (apoAII) is the second major apolipoprotein of the high-density lipoprotein (HDL) particle, after apoAI. Unlike apoAI, the biological and physiological functions of apoAII are unclear. We aimed to gain insight into the specific roles of apoAII in lipoprotein metabolism and atherosclerosis using a novel rabbit model. METHODS Wild-type (WT) rabbits are naturally deficient in apoAII, thus their HDL contains only apoAI. Using TALEN technology, we replaced the endogenous apoAI in rabbits through knock-in (KI) of human apoAII. The newly generated apoAII KI rabbits were used to study the specific function of apoAII, independent of apoAI. RESULTS ApoAII KI rabbits expressed exclusively apoAII without apoAI, as confirmed by RT-PCR and Western blotting. On a standard diet, the KI rabbits exhibited lower plasma triglycerides (TG, 52%, p < 0.01) due to accelerated clearance of TG-rich particles and higher lipoprotein lipase activity than the WT littermates. ApoAII KI rabbits also had higher plasma HDL-C (28%, p < 0.05) and their HDL was rich in apoE, apoAIV, and apoAV. When fed a cholesterol-rich diet for 16 weeks, apoAII KI rabbits were resistant to diet-induced hypertriglyceridemia and developed significantly less aortic atherosclerosis compared to WT rabbits. HDL isolated from rabbits with apoAII KI had similar cholesterol efflux capacity and anti-inflammatory effects as HDL isolated from the WT rabbits. CONCLUSIONS ApoAII KI rabbits developed less atherosclerosis than WT rabbits, possibly through increased plasma HDL-C, reduced TG and atherogenic lipoproteins. These results suggest that apoAII may serve as a potential target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Tomonari Koike
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA; Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Yamanashi, Japan
| | - Yui Koike
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yanhong Guo
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Oren Rom
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jun Song
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yajie Chen
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Yamanashi, Japan
| | - Yanli Wang
- Department of Pathology, Xi'an Medical University, Xi'an, China
| | - Tianqing Zhu
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Minerva T Garcia-Barrio
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Yamanashi, Japan.
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| |
Collapse
|
60
|
Härdtner C, Kornemann J, Krebs K, Ehlert CA, Jander A, Zou J, Starz C, Rauterberg S, Sharipova D, Dufner B, Hoppe N, Dederichs TS, Willecke F, Stachon P, Heidt T, Wolf D, von Zur Mühlen C, Madl J, Kohl P, Kaeser R, Boettler T, Pieterman EJ, Princen HMG, Ho-Tin-Noé B, Swirski FK, Robbins CS, Bode C, Zirlik A, Hilgendorf I. Inhibition of macrophage proliferation dominates plaque regression in response to cholesterol lowering. Basic Res Cardiol 2020; 115:78. [PMID: 33296022 PMCID: PMC7725697 DOI: 10.1007/s00395-020-00838-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/01/2020] [Indexed: 02/08/2023]
Abstract
Statins induce plaque regression characterized by reduced macrophage content in humans, but the underlying mechanisms remain speculative. Studying the translational APOE*3-Leiden.CETP mouse model with a humanized lipoprotein metabolism, we find that systemic cholesterol lowering by oral atorvastatin or dietary restriction inhibits monocyte infiltration, and reverses macrophage accumulation in atherosclerotic plaques. Contrary to current believes, none of (1) reduced monocyte influx (studied by cell fate mapping in thorax-shielded irradiation bone marrow chimeras), (2) enhanced macrophage egress (studied by fluorescent bead labeling and transfer), or (3) atorvastatin accumulation in murine or human plaque (assessed by mass spectrometry) could adequately account for the observed loss in macrophage content in plaques that undergo phenotypic regression. Instead, suppression of local proliferation of macrophages dominates phenotypic plaque regression in response to cholesterol lowering: the lower the levels of serum LDL-cholesterol and lipid contents in murine aortic and human carotid artery plaques, the lower the rates of in situ macrophage proliferation. Our study identifies macrophage proliferation as the predominant turnover determinant and an attractive target for inducing plaque regression.
Collapse
Affiliation(s)
- Carmen Härdtner
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Jan Kornemann
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Katja Krebs
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Carolin A Ehlert
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Alina Jander
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Jiadai Zou
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Christopher Starz
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Simon Rauterberg
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Diana Sharipova
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Bianca Dufner
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Natalie Hoppe
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Tsai-Sang Dederichs
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Florian Willecke
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Timo Heidt
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Constantin von Zur Mühlen
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Josef Madl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rafael Kaeser
- Department of Medicine II, Faculty of Medicine, Medical Center-University Freiburg, University of Freiburg, Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II, Faculty of Medicine, Medical Center-University Freiburg, University of Freiburg, Freiburg, Germany
| | - Elsbeth J Pieterman
- The Netherlands Organization for Applied Scientific Research (TNO)-Metabolic Health Research, Leiden, Netherlands
| | - Hans M G Princen
- The Netherlands Organization for Applied Scientific Research (TNO)-Metabolic Health Research, Leiden, Netherlands
| | - Benoît Ho-Tin-Noé
- INSERM Unit 1148, University Paris Diderot, and Laboratory for Vascular Translational Science, Sorbonne Paris Cité, Paris, France
| | - Filip K Swirski
- Center of Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Clinton S Robbins
- Peter Munk Cardiac Centre, University Health Network, Toronto, Canada
| | - Christoph Bode
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany
| | - Andreas Zirlik
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany.,Department of Cardiology, University of Graz, Graz, Austria
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, 55 Hugstetter St, 79106, Freiburg, Germany.
| |
Collapse
|
61
|
Beaufrère H, Gardhouse SM, Wood RD, Stark KD. The plasma lipidome of the Quaker parrot (Myiopsitta monachus). PLoS One 2020; 15:e0240449. [PMID: 33259543 PMCID: PMC7707497 DOI: 10.1371/journal.pone.0240449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/25/2020] [Indexed: 11/19/2022] Open
Abstract
Dyslipidemias and lipid-accumulation disorders are common in captive parrots, in particular in Quaker parrots. Currently available diagnostic tests only measure a fraction of blood lipids and have overall problematic cross-species applicability. Comprehensively analyzing lipids in the plasma of parrots is the first step to better understand their lipid metabolism in health and disease, as well as to explore new lipid biomarkers. The plasma lipidome of 12 Quaker parrots was investigated using UHPLC-MS/MS with both targeted and untargeted methods. Targeted methods on 6 replicates measured 432 lipids comprised of sterol, cholesterol ester, bile acid, fatty acid, acylcarnitine, glycerolipid, glycerophospholipid, and sphingolipid panels. For untargeted lipidomics, precursor ion mass-to-charge ratios were matched to corresponding lipids using the LIPIDMAPS structure database and LipidBlast at the sum composition or acyl species level of information. Sterol lipids and glycerophospholipids constituted the majority of plasma lipids on a molar basis. The most common lipids detected with the targeted methods included free cholesterol, CE(18:2), CE(20:4) for sterol lipids; PC(36:2), PC(34:2), PC(34:1) for glycerophospholipids; TG(52:3), TG(54:4), TG(54:5), TG(52:2) for glycerolipids; SM(d18:1/16:0) for sphingolipids; and palmitic acid for fatty acyls. Over a thousand different lipid species were detected by untargeted lipidomics. Sex differences in the plasma lipidome were observed using heatmaps, principal component analysis, and discriminant analysis. This report presents the first comprehensive database of plasma lipid species in psittacine birds and paves the way for further research into blood lipid diagnostics and the impact of diet, diseases, and drugs on the parrot plasma lipidome.
Collapse
Affiliation(s)
- Hugues Beaufrère
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
- * E-mail:
| | - Sara M. Gardhouse
- Health Sciences Centre, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - R. Darren Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Ken D. Stark
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
62
|
Zec MM, Krga I, Takić M, Debeljak-Martačić J, Korićanac G, Ranković S, Popović T, Pantelić M, Glibetic M. Walnut Consumption Induces Tissue-Specific Omega-6/Omega-3 Decrease in High-Fructose-Fed Wistar Rats. ACS OMEGA 2020; 5:28136-28145. [PMID: 33163796 PMCID: PMC7643199 DOI: 10.1021/acsomega.0c03784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/08/2020] [Indexed: 05/08/2023]
Abstract
Increased dietary, blood, and tissue n-6/n-3 fatty acid ratios are associated with obesity and metabolic syndrome. Due to Westernized dietary patterns, the increasing n-6/n-3 ratio is of growing concern worldwide, and dietary strategies aimed at its lowering are of public health importance. Walnuts are rich in dietary fats, and their consumption promotes cardiometabolic health. This study aimed to examine the effect of 6-week walnut consumption on tissue-specific n-6/n-3 ratio and fatty acid metabolic conversion in fructose-fed rats with a cluster of metabolic disorders. Male Wistar rats were fed a standard diet with or without 10% fructose in drinking water for 9 weeks. Diets of half of the animals were then supplemented with walnuts (2.4 g/day) for 6 weeks, upon which fatty acid profiles were determined in plasma, liver, adipose tissue, and kidney total lipids. Results showed that walnuts induced significant decreases in the n-6/n-3 content of total lipid pool in plasma and examined tissues, irrespective of metabolic burden. Walnut intervention decreased plasma and liver palmitoleic/palmitic, arachidonic/linoleic, and docosahexaenoic/α-linolenic acid ratios. It also modulated individual fatty acid levels by reducing arachidonic and palmitic acid and increasing α-linolenic, eicosapentaenoic, and docosapentaenoic acid in plasma and most tissues. Our study demonstrated that 6-week consumption of walnuts favorably modulated n-6/n-3 plasma and tissue ratio in male Wistar rats regardless of high-fructose feeding, underscoring the promising potential of walnuts in both prevention and treatment of the metabolic syndrome.
Collapse
Affiliation(s)
- Manja M. Zec
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Irena Krga
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Marija Takić
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Jasmina Debeljak-Martačić
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Goran Korićanac
- Laboratory
for Molecular Biology and Endocrinology, Vinča Institute of
Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11001, Serbia
| | - Slavica Ranković
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Tamara Popović
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Marija Pantelić
- Laboratory
for Molecular Biology and Endocrinology, Vinča Institute of
Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11001, Serbia
| | - Maria Glibetic
- Centre
of Excellence in Nutrition and Metabolism Research, Institute for
Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
63
|
Early Pro-Inflammatory Remodeling of HDL Proteome in a Model of Diet-Induced Obesity: 2H 2O-Metabolic Labeling-Based Kinetic Approach. Int J Mol Sci 2020; 21:ijms21207472. [PMID: 33050482 PMCID: PMC7656294 DOI: 10.3390/ijms21207472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
Mice fed a high-fat diet for 12 weeks or longer develop hyperglycemia, insulin resistance, dyslipidemia, and fatty liver. Additionally, a high-fat diet induces inflammation that remodels and affects the anti-inflammatory and antiatherogenic property of the high-density lipoprotein (HDL). However, the precise time course of metabolic disease progression and HDL remodeling remains unclear. Short-term (four weeks) high-fat feeding (60% fat calories) was performed in wild-type male C57BL/6J mice to gain insights into the early metabolic disease processes in conjunction with a HDL proteome dynamics analysis using a heavy water metabolic labeling approach. The high-fat diet-fed mice developed hyperglycemia, impaired glucose tolerance, hypercholesterolemia without hypertriglyceridemia or hepatic steatosis. A plasma HDL proteome dynamics analysis revealed increased turnover rates (and reduced half-lives) of several acute-phase response proteins involved in innate immunity, including complement C3 (12.77 ± 0.81 vs. 9.98 ± 1.20 h, p < 0.005), complement factor B (12.71 ± 1.01 vs. 10.85 ± 1.04 h, p < 0.05), complement Factor H (19.60 ± 1.84 vs. 16.80 ± 1.58 h, p < 0.05), and complement factor I (25.25 ± 1.29 vs. 19.88 ± 1.50 h, p < 0.005). Our findings suggest that an early immune response-induced inflammatory remodeling of the plasma HDL proteome precedes the diet-induced steatosis and dyslipidemia.
Collapse
|
64
|
Nevado RM, Hamczyk MR, Gonzalo P, Andrés-Manzano MJ, Andrés V. Premature Vascular Aging with Features of Plaque Vulnerability in an Atheroprone Mouse Model of Hutchinson-Gilford Progeria Syndrome with Ldlr Deficiency. Cells 2020; 9:cells9102252. [PMID: 33049978 PMCID: PMC7601818 DOI: 10.3390/cells9102252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 10/04/2020] [Indexed: 12/21/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is among the most devastating of the laminopathies, rare genetic diseases caused by mutations in genes encoding nuclear lamina proteins. HGPS patients age prematurely and die in adolescence, typically of atherosclerosis-associated complications. The mechanisms of HGPS-related atherosclerosis are not fully understood due to the scarcity of patient-derived samples and the availability of only one atheroprone mouse model of the disease. Here, we generated a new atherosusceptible model of HGPS by crossing progeroid LmnaG609G/G609G mice, which carry a disease-causing mutation in the Lmna gene, with Ldlr−/− mice, a commonly used preclinical atherosclerosis model. Ldlr−/−LmnaG609G/G609G mice aged prematurely and had reduced body weight and survival. Compared with control mice, Ldlr−/−LmnaG609G/G609G mouse aortas showed a higher atherosclerosis burden and structural abnormalities typical of HGPS patients, including vascular smooth muscle cell depletion in the media, adventitial thickening, and elastin structure alterations. Atheromas of Ldlr−/−LmnaG609G/G609G mice had features of unstable plaques, including the presence of erythrocytes and iron deposits and reduced smooth muscle cell and collagen content. Ldlr−/−LmnaG609G/G609G mice faithfully recapitulate vascular features found in patients and thus provide a new tool for studying the mechanisms of HGPS-related atherosclerosis and for testing therapies.
Collapse
MESH Headings
- Aging, Premature/metabolism
- Aging, Premature/physiopathology
- Animals
- Aorta/metabolism
- Atherosclerosis/metabolism
- Atherosclerosis/physiopathology
- Disease Models, Animal
- Female
- Lamin Type A/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Nuclear Lamina/metabolism
- Plaque, Atherosclerotic/metabolism
- Progeria/metabolism
- Progeria/physiopathology
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
Collapse
Affiliation(s)
- Rosa M. Nevado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; (R.M.N.); (P.G.); (M.J.A.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Magda R. Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; (R.M.N.); (P.G.); (M.J.A.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain;
| | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; (R.M.N.); (P.G.); (M.J.A.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Jesús Andrés-Manzano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; (R.M.N.); (P.G.); (M.J.A.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; (R.M.N.); (P.G.); (M.J.A.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-91-453-1200
| |
Collapse
|
65
|
Michel JB. Phylogenic Determinants of Cardiovascular Frailty, Focus on Hemodynamics and Arterial Smooth Muscle Cells. Physiol Rev 2020; 100:1779-1837. [DOI: 10.1152/physrev.00022.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of the circulatory system from invertebrates to mammals has involved the passage from an open system to a closed in-parallel system via a closed in-series system, accompanying the increasing complexity and efficiency of life’s biological functions. The archaic heart enables pulsatile motion waves of hemolymph in invertebrates, and the in-series circulation in fish occurs with only an endothelium, whereas mural smooth muscle cells appear later. The present review focuses on evolution of the circulatory system. In particular, we address how and why this evolution took place from a closed, flowing, longitudinal conductance at low pressure to a flowing, highly pressurized and bifurcating arterial compartment. However, although arterial pressure was the latest acquired hemodynamic variable, the general teleonomy of the evolution of species is the differentiation of individual organ function, supported by specific fueling allowing and favoring partial metabolic autonomy. This was achieved via the establishment of an active contractile tone in resistance arteries, which permitted the regulation of blood supply to specific organ activities via its localized function-dependent inhibition (active vasodilation). The global resistance to viscous blood flow is the peripheral increase in frictional forces caused by the tonic change in arterial and arteriolar radius, which backscatter as systemic arterial blood pressure. Consequently, the arterial pressure gradient from circulating blood to the adventitial interstitium generates the unidirectional outward radial advective conductance of plasma solutes across the wall of conductance arteries. This hemodynamic evolution was accompanied by important changes in arterial wall structure, supported by smooth muscle cell functional plasticity, including contractility, matrix synthesis and proliferation, endocytosis and phagocytosis, etc. These adaptive phenotypic shifts are due to epigenetic regulation, mainly related to mechanotransduction. These paradigms actively participate in cardio-arterial pathologies such as atheroma, valve disease, heart failure, aneurysms, hypertension, and physiological aging.
Collapse
|
66
|
The marine compound and elongation factor 1A1 inhibitor, didemnin B, provides benefit in western diet-induced non-alcoholic fatty liver disease. Pharmacol Res 2020; 161:105208. [PMID: 32977024 DOI: 10.1016/j.phrs.2020.105208] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
Inhibition of eukaryotic elongation factor 1A1 (EEF1A1) with the marine compound didemnin B decreases lipotoxic HepG2 cell death in vitro and improves early stage non-alcoholic fatty liver disease (NAFLD) in young genetically obese mice. However, the effects of didemnin B on NAFLD in a model of long-term diet-induced obesity are not known. We investigated the effects of didemnin B on NAFLD severity and metabolic parameters in western diet-induced obese mice, and on the cell types that contribute to liver inflammation and fibrosis in vitro. Male 129S6 mice were fed either standard chow or western diet for 26 weeks, followed by intervention with didemnin B (50 μg/kg) or vehicle by intraperitoneal (i.p.) injection once every 3 days for 14 days. Didemnin B decreased liver and plasma triglycerides, improved oral glucose tolerance, and decreased NAFLD severity. Moreover, didemnin B moderately increased hepatic expression of genes involved in ER stress response (Perk, Chop), and fatty acid oxidation (Fgf21, Cpt1a). In vitro, didemnin B decreased THP-1 monocyte proliferation, disrupted THP-1 monocyte-macrophage differentiation, decreased THP-1 macrophage IL-1β secretion, and decreased hepatic stellate cell (HSteC) proliferation and collagen secretion under both basal and lipotoxic (high fatty acid) conditions. Thus, didemnin B improves hepatic steatosis, glucose tolerance, and blood lipids in obesity, in association with moderate, possibly hormetic, upregulation of pathways involved in cell stress response and energy balance in the liver. Furthermore, it decreases the activity of the cell types implicated in liver inflammation and fibrosis in vitro. These findings highlight the therapeutic potential of partial protein synthesis inhibition in the treatment of NAFLD.
Collapse
|
67
|
Fanjul V, Jorge I, Camafeita E, Macías Á, González‐Gómez C, Barettino A, Dorado B, Andrés‐Manzano MJ, Rivera‐Torres J, Vázquez J, López‐Otín C, Andrés V. Identification of common cardiometabolic alterations and deregulated pathways in mouse and pig models of aging. Aging Cell 2020; 19:e13203. [PMID: 32729659 PMCID: PMC7511870 DOI: 10.1111/acel.13203] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/10/2020] [Accepted: 07/03/2020] [Indexed: 12/15/2022] Open
Abstract
Aging is the main risk factor for cardiovascular and metabolic diseases, which have become a global concern as the world population ages. These diseases and the aging process are exacerbated in Hutchinson–Gilford progeria syndrome (HGPS or progeria). Here, we evaluated the cardiometabolic disease in animal models of premature and normal aging with the aim of identifying alterations that are shared or specific to each condition. Despite differences in body composition and metabolic markers, prematurely and normally aging mice developed heart failure and similar cardiac electrical abnormalities. High‐throughput proteomics of the hearts of progeric and normally aged mice revealed altered protein oxidation and glycation, as well as dysregulated pathways regulating energy metabolism, proteostasis, gene expression, and cardiac muscle contraction. These results were corroborated in the hearts of progeric pigs, underscoring the translational potential of our findings, which could help in the design of strategies to prevent or slow age‐related cardiometabolic disease.
Collapse
Affiliation(s)
- Víctor Fanjul
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Oncología Universidad de Oviedo Oviedo Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Inmaculada Jorge
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Emilio Camafeita
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Cristina González‐Gómez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Ana Barettino
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - María Jesús Andrés‐Manzano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - José Rivera‐Torres
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| | - Carlos López‐Otín
- Departamento de Bioquímica y Biología Molecular Facultad de Medicina Instituto Universitario de Oncología Universidad de Oviedo Oviedo Spain
- Centro de Investigación Biomédica en Red Enfermedades Cáncer (CIBERONC) Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV) Spain
| |
Collapse
|
68
|
Delgadillo-Puga C, Noriega LG, Morales-Romero AM, Nieto-Camacho A, Granados-Portillo O, Rodríguez-López LA, Alemán G, Furuzawa-Carballeda J, Tovar AR, Cisneros-Zevallos L, Torre-Villalvazo I. Goat's Milk Intake Prevents Obesity, Hepatic Steatosis and Insulin Resistance in Mice Fed A High-Fat Diet by Reducing Inflammatory Markers and Increasing Energy Expenditure and Mitochondrial Content in Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21155530. [PMID: 32752280 PMCID: PMC7432599 DOI: 10.3390/ijms21155530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/26/2022] Open
Abstract
Goat's milk is a rich source of bioactive compounds (peptides, conjugated linoleic acid, short chain fatty acids, monounsaturated and polyunsaturated fatty acids, polyphenols such as phytoestrogens and minerals among others) that exert important health benefits. However, goat's milk composition depends on the type of food provided to the animal and thus, the abundance of bioactive compounds in milk depends on the dietary sources of the goat feed. The metabolic impact of goat milk rich in bioactive compounds during metabolic challenges such as a high-fat (HF) diet has not been explored. Thus, we evaluated the effect of milk from goats fed a conventional diet, a conventional diet supplemented with 30% Acacia farnesiana (AF) pods or grazing on metabolic alterations in mice fed a HF diet. Interestingly, the incorporation of goat's milk in the diet decreased body weight and body fat mass, improved glucose tolerance, prevented adipose tissue hypertrophy and hepatic steatosis in mice fed a HF diet. These effects were associated with an increase in energy expenditure, augmented oxidative fibers in skeletal muscle, and reduced inflammatory markers. Consequently, goat's milk can be considered a non-pharmacologic strategy to improve the metabolic alterations induced by a HF diet. Using the body surface area normalization method gave a conversion equivalent daily human intake dose of 1.4 to 2.8 glasses (250 mL per glass/day) of fresh goat milk for an adult of 60 kg, which can be used as reference for future clinical studies.
Collapse
Affiliation(s)
- Claudia Delgadillo-Puga
- Departamento de Nutrición Animal Dr. Fernando Pérez-Gil Romo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico
- Correspondence: (C.D.-P.); (I.T.-V.); Tel.: +52-55-54870900 (C.D.-P. & I.T.-V.)
| | - Lilia G. Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Aurora M. Morales-Romero
- Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico 04510, Mexico;
| | - Antonio Nieto-Camacho
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico 04510, Mexico;
| | - Omar Granados-Portillo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Leonardo A. Rodríguez-López
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Gabriela Alemán
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Janette Furuzawa-Carballeda
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico;
| | - Armando R. Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Luis Cisneros-Zevallos
- Department of Horticultural Sciences, Texas A&M University, College Station, TX 77843-2133, USA;
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
- Correspondence: (C.D.-P.); (I.T.-V.); Tel.: +52-55-54870900 (C.D.-P. & I.T.-V.)
| |
Collapse
|
69
|
Gómez C, Stücheli S, Kratschmar DV, Bouitbir J, Odermatt A. Development and Validation of a Highly Sensitive LC-MS/MS Method for the Analysis of Bile Acids in Serum, Plasma, and Liver Tissue Samples. Metabolites 2020; 10:E282. [PMID: 32660078 PMCID: PMC7408441 DOI: 10.3390/metabo10070282] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023] Open
Abstract
Bile acids control lipid homeostasis by regulating uptake from food and excretion. Additionally, bile acids are bioactive molecules acting through receptors and modulating various physiological processes. Impaired bile acid homeostasis is associated with several diseases and drug-induced liver injury. Individual bile acids may serve as disease and drug toxicity biomarkers, with a great demand for improved bile acid quantification methods. We developed, optimized, and validated an LC-MS/MS method for quantification of 36 bile acids in serum, plasma, and liver tissue samples. The simultaneous quantification of important free and taurine- and glycine-conjugated bile acids of human and rodent species has been achieved using a simple workflow. The method was applied to a mouse model of statin-induced myotoxicity to assess a possible role of bile acids. Treatment of mice for three weeks with 5, 10, and 25 mg/kg/d simvastatin, causing adverse skeletal muscle effects, did not alter plasma and liver tissue bile acid profiles, indicating that bile acids are not involved in statin-induced myotoxicity. In conclusion, the established LC-MS/MS method enables uncomplicated sample preparation and quantification of key bile acids in serum, plasma, and liver tissue of human and rodent species to facilitate future studies of disease mechanisms and drug-induced liver injury.
Collapse
Affiliation(s)
| | | | | | | | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland; (C.G.); (S.S.); (D.V.K.); (J.B.)
| |
Collapse
|
70
|
Vedder VL, Aherrahrou Z, Erdmann J. Dare to Compare. Development of Atherosclerotic Lesions in Human, Mouse, and Zebrafish. Front Cardiovasc Med 2020; 7:109. [PMID: 32714944 PMCID: PMC7344238 DOI: 10.3389/fcvm.2020.00109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases, such as atherosclerosis, are the leading cause of death worldwide. Although mice are currently the most commonly used model for atherosclerosis, zebrafish are emerging as an alternative, especially for inflammatory and lipid metabolism studies. Here, we review the history of in vivo atherosclerosis models and highlight the potential for future studies on inflammatory responses in lipid deposits in zebrafish, based on known immune reactions in humans and mice, in anticipation of new zebrafish models with more advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Viviana L Vedder
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
71
|
White Z, Hakim CH, Theret M, Yang NN, Rossi F, Cox D, Francis GA, Straub V, Selby K, Panagiotopoulos C, Duan D, Bernatchez P. High prevalence of plasma lipid abnormalities in human and canine Duchenne and Becker muscular dystrophies depicts a new type of primary genetic dyslipidemia. J Clin Lipidol 2020; 14:459-469.e0. [PMID: 32593511 DOI: 10.1016/j.jacl.2020.05.098] [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: 01/28/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are allelic X-linked recessive muscle diseases caused by mutations in the DMD gene, with DMD being the more severe form. We have recently shown that increased plasma low-density lipoprotein-associated cholesterol causes severe muscle wasting in the mdx mouse, a mild DMD model, which suggested that plasma lipids may play a critical role in DMD. We have also observed that loss of dystrophin in mice causes unexpected elevations in plasma lipoprotein levels. OBJECTIVE The objectives of the study were to determine whether patients with DMD and BMD also present with clinically relevant plasma lipoprotein abnormalities and to mitigate the presence of confounders (medications and lifestyle) by analyzing the plasma from patients with DMD/BMD and unmedicated dogs with DMD, the most relevant model of DMD. METHODS Levels of low-density lipoprotein-associated cholesterol, high-density lipoprotein cholesterol, and triglycerides were analyzed in patients with DMD and BMD and female carriers. Samples from unmedicated, ambulatory dogs with DMD, unaffected carriers, and normal controls were also analyzed. RESULTS We report that 97% and 64% of all pediatric patients with DMD (33 of 36) and BMD (6 of 11) are dyslipidemic, along with an unusually high incidence in adult patients with BMD. All dogs with DMD showed plasma lipid abnormalities that progressively worsened with age. Most strikingly, unaffected carrier dogs also showed plasma lipid abnormalities similar to affected dogs with DMD. Dyslipidemia is likely not secondary to liver damage as unaffected carriers showed no plasma aminotransferase elevation. CONCLUSIONS The high incidence of plasma lipid abnormalities in dystrophin-deficient plasma may depict a new type of genetic dyslipidemia. Abnormal lipid levels in dystrophinopathic samples in the absence of muscle damage suggest a primary state of dyslipidemia. Whether dyslipidemia plays a causal role in patients with DMD warrants further investigation, which could lead to new diagnostic and therapeutic options.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada; Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO; National Center for Advancing Translational Sciences, NIH, Rockville, MD
| | | | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD
| | - Fabio Rossi
- Biomedical Research Centre, UBC, Vancouver, Canada
| | - Dan Cox
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Gordon A Francis
- Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada; Department of Medicine, UBC, Vancouver, Canada
| | - Volker Straub
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Kathryn Selby
- Department of Pediatrics, University of British Columbia (UBC), BC Children's Hospital Research Institute, Vancouver, Canada
| | - Constadina Panagiotopoulos
- Department of Pediatrics, University of British Columbia (UBC), BC Children's Hospital Research Institute, Vancouver, Canada
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO; Department of Pediatrics, University of British Columbia (UBC), BC Children's Hospital Research Institute, Vancouver, Canada; Department of Neurology, University of Missouri, Columbia, MO; Department of Bioengineering, Faculty of Medicine, University of Missouri, Columbia, MO; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO.
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada; Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada.
| |
Collapse
|
72
|
Koca RB, Güven O, Çelik MS, Fıratlı E. Wetting properties of blood lipid fractions on different titanium surfaces. Int J Implant Dent 2020; 6:16. [PMID: 32399791 PMCID: PMC7218032 DOI: 10.1186/s40729-020-00213-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/23/2020] [Indexed: 01/20/2023] Open
Abstract
Background Blood is the first tissue contacting the implant surface and starting the biological interactions to enhance osseointegration and stimulate bone formation with the progenitor cytokines, chemokines, and growth factors. The coagulation cascade initiates the first step of osseointegration between implant and neighboring tissues. The wound healing may be inadequate unless the blood wets the implant surface properly. Wettability is one of the most important features of the implant surface while lipid level constitutes a milestone that may change the energy of blood, which determines its distribution on implant material. Thus, the aim of this study was to evaluate the effect of lipid component of blood as cholesterol and its treatment on their wetting behavior of titanium surfaces. Methods Five surface groups were formed including grade 4 titanium-machined, grade 4 titanium-SLA, grade 4 titanium-SLActive, Roxolid-SLA, and Roxolid-SLActive. In healthy, hyperlipidemic, and treatment situations, blood was taken from eight rabbits and dropped to the disc surfaces. Contact angles were measured between the blood samples and disc surfaces. Results A significant difference was found between both machined and SLActive surfaces, SLA and SLActive surfaces in the hyperlipidemic period, and only Roxolid-SLA and SLActive surfaces during the treatment period. When evaluated according to time, only grade 4-machined and Grade 4-SLA surfaces showed a significant difference. Conclusions Our findings indicated that each period has its own characteristics and showed the importance of cholesterol in blood structure on applicability of implant surfaces.
Collapse
Affiliation(s)
- Revan Birke Koca
- Department of Periodontology, Faculty of Dentistry, University of Kyrenia, 99320, Kyrenia, Cyprus.
| | - Onur Güven
- Department of Mining Engineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Mehmet Sabri Çelik
- Department of Mineral Processing Engineering, Faculty of Mines, İstanbul Technical University, İstanbul, Turkey.,Harran University Rectorate, Şanlıurfa, Turkey
| | - Erhan Fıratlı
- Department of Periodontology, Faculty of Dentistry, Istanbul University, İstanbul, Turkey
| |
Collapse
|
73
|
Hamczyk MR, Andrés V. Vascular smooth muscle cell loss underpins the accelerated atherosclerosis in Hutchinson-Gilford progeria syndrome. Nucleus 2020; 10:28-34. [PMID: 30900948 PMCID: PMC6527384 DOI: 10.1080/19491034.2019.1589359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lamin A, a product of the LMNA gene, is an essential nuclear envelope component in most differentiated cells. Mutations in LMNA have been linked to premature aging disorders, including Hutchinson-Gilford progeria syndrome (HGPS). HGPS is caused by progerin, an aberrant form of lamin A that leads to premature death, typically from the complications of atherosclerotic disease. A key characteristic of HGPS is a severe loss of vascular smooth muscle cells (VSMCs) in the arteries. Various mouse models of HGPS have been created, but few of them feature VSMC depletion and none develops atherosclerosis, the death-causing symptom of the disease in humans. We recently generated a mouse model that recapitulates most features of HGPS, including VSMC loss and accelerated atherosclerosis. Furthermore, by generating cell-type–specific HGPS mouse models, we have demonstrated a central role of VSMC loss in progerin-induced atherosclerosis and premature death.
Collapse
Affiliation(s)
- Magda R Hamczyk
- a Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Vascular Pathophysiology Area , Centro Nacional de Investigaciones Cardiovasculares (CNIC) , Madrid , Spain.,b CIBER de Enfermedades Cardiovasculares (CIBERCV) , Spain.,c Departamento de Bioquímica y Biología Molecular , Universidad de Oviedo , Oviedo , Spain
| | - Vicente Andrés
- a Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Vascular Pathophysiology Area , Centro Nacional de Investigaciones Cardiovasculares (CNIC) , Madrid , Spain.,b CIBER de Enfermedades Cardiovasculares (CIBERCV) , Spain
| |
Collapse
|
74
|
Uceda DE, Zhu XY, Woollard JR, Ferguson CM, Patras I, Carlson DF, Asirvatham SJ, Lerman A, Lerman LO. Accumulation of Pericardial Fat Is Associated With Alterations in Heart Rate Variability Patterns in Hypercholesterolemic Pigs. Circ Arrhythm Electrophysiol 2020; 13:e007614. [PMID: 32189516 DOI: 10.1161/circep.119.007614] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Heart rate variability (HRV) and pulse rate variability are indices of autonomic cardiac modulation. Increased pericardial fat is associated with worse cardiovascular outcomes. We hypothesized that progressive increases in pericardial fat volume and inflammation prospectively dampen HRV in hypercholesterolemic pigs. METHODS WT (wild type) or PCSK9 (proprotein convertase subtilisin-like/kexin type-9) gain-of-function Ossabaw mini-pigs were studied in vivo before and after 3 and 6 months of a normal diet (WT-normal diet, n=4; PCSK9-normal diet, n=6) or high-fat diet (HFD; WT-HFD, n=3; PCSK9-HFD, n=6). The arterial pulse waveform was obtained from an arterial telemetry transmitter to analyze HRV indices, including SD (SD of all pulse-to-pulse intervals over a single 5-minute period), root mean square of successive differences, proportion >50 ms of normal-to-normal R-R intervals, and the calculated ratio of low-to-high frequency distributions (low-frequency power/high-frequency power). Pericardial fat volumes were evaluated using multidetector computed tomography and its inflammation by gene expression of TNF (tumor necrosis factor)-α. Plasma lipid panel and norepinephrine level were also measured. RESULTS At diet completion, hypercholesterolemic PCSK9-HFD had significantly (P<0.05 versus baseline) depressed HRV (SD of all pulse-to-pulse intervals over a single 5-minute period, root mean square of successive differences, proportion >50 ms, high-frequency power, low-frequency power), and both HFD groups had higher sympathovagal balance (SD of all pulse-to-pulse intervals over a single 5-minute period/root mean square of successive differences, low-frequency power/high-frequency power) compared with normal diet. Pericardial fat volumes and LDL (low-density lipoprotein) cholesterol concentrations correlated inversely with HRV and directly with sympathovagal balance, while sympathovagal balance correlated directly with plasma norepinephrine. Pericardial fat TNF-α expression was upregulated in PCSK9-HFD, colocalized with nerve fibers, and correlated inversely with root mean square of successive differences and proportion >50 ms. CONCLUSIONS Progressive pericardial fat expansion and inflammation are associated with a fall in HRV in Ossabaw mini-pigs, implying aggravated autonomic imbalance. Hence, pericardial fat accumulation is associated with alterations in HRV and the autonomic nervous system. Visual Overview: A visual overview is available for this article.
Collapse
Affiliation(s)
- Domingo E Uceda
- Division of Nephrology and Hypertension (D.E.U., X.-Y.Z., J.R.W., C.M.F., I.P., L.O.L.), Mayo Clinic, Rochester, MN
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension (D.E.U., X.-Y.Z., J.R.W., C.M.F., I.P., L.O.L.), Mayo Clinic, Rochester, MN
| | - John R Woollard
- Division of Nephrology and Hypertension (D.E.U., X.-Y.Z., J.R.W., C.M.F., I.P., L.O.L.), Mayo Clinic, Rochester, MN
| | - Christopher M Ferguson
- Division of Nephrology and Hypertension (D.E.U., X.-Y.Z., J.R.W., C.M.F., I.P., L.O.L.), Mayo Clinic, Rochester, MN
| | - Ioannis Patras
- Division of Nephrology and Hypertension (D.E.U., X.-Y.Z., J.R.W., C.M.F., I.P., L.O.L.), Mayo Clinic, Rochester, MN
| | | | - Samuel J Asirvatham
- Department of Cardiovascular Diseases (S.J.A., A.L., L.O.L.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Diseases (S.J.A., A.L., L.O.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- Division of Nephrology and Hypertension (D.E.U., X.-Y.Z., J.R.W., C.M.F., I.P., L.O.L.), Mayo Clinic, Rochester, MN.,Department of Cardiovascular Diseases (S.J.A., A.L., L.O.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
75
|
Zhang T, Shi H, Liu N, Tian J, Zhao X, Steer CJ, Han Q, Song G. Activation of microRNA-378a-3p biogenesis promotes hepatic secretion of VLDL and hyperlipidemia by modulating ApoB100-Sortilin1 axis. Am J Cancer Res 2020; 10:3952-3966. [PMID: 32226531 PMCID: PMC7086368 DOI: 10.7150/thno.39578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Rationale: Hyperlipidemia is a major risk factor of atherosclerosis and cardiovascular diseases (CVD). As a standard-of-care approach for hyperlipidemia, statins only reduce the risk of coronary artery disease by 20-40%, underscoring the importance of identifying molecular pathways for the design of drugs against this disorder. Alterations in microRNA (miRNA) expression have been reported in patients with hyperlipidemia and CVD. This study was designed to determine the mechanism of dysregulated miR-378a-3p under the status of hyperlipidemia and evaluate how miR-378a-3p regulates hepatic secretion of VLDL. Methods: Wild-type mice kept on a high fat diet were injected with miR-378a-3p inhibitor or a mini-circle expression system containing miR-378a precursor to study loss and gain-of functions of miR-378a-3p. Mice were treated with Triton WR1339 and 35S-methionine/cysteine to determine the effect of miR-378a-3p on hepatic secretion of VLDL. Database mining, luciferase assay, and ChIP (chromatin immunoprecipitation) were used to study the mechanism of dysregulated miR-378a-3p biogenesis. Results: miR-378a-3p expression is significantly increased in livers of hyperlipidemic mice. Sort1 (sortilin 1) was identified as a direct target of miR-378a-3p. By inhibiting the function of sortilin 1 as a transmembrane trafficking receptor, miR-378a-3p stabilized ApoB100 and promoted ApoB100 secretion in vitro. Liver-specific expression of miR-378a-3p stabilized ApoB100 and facilitated hepatic secretion of VLDL, which subsequently increased levels of VLDL/LDL cholesterol as well as triglycerides. In contrast, antagonizing miR-378a-3p using its inhibitor increased hepatic expression of Sort1 and reduced hepatic export of VLDL with its consequent effects of serum lipid levels. Additional knockdown of up-regulated Sort1 in livers of mice offset the effects of miR-378a-3p inhibitor, suggesting that Sort1 was indispensable for miR-378a-3p to promote secretion of VLDL and thereby high levels of circulating VLDL/LDL cholesterol and triglycerides. Furthermore, oncogenic E2F1 (E2F transcription factor 1) was identified as a transcriptional activator of miR-378a-3p. E2f1 knockdown, through reducing miR-378a-3p, impaired secretion of VLDL and reduced levels of VLDL/LDL cholesterol and triglycerides. Conclusions: This study defines a novel pathway of E2F1-miR-378a-3p-SORT1-ApoB100 that controls levels of circulating VLDL/LDL cholesterol and triglycerides by modulating degradation and secretion of ApoB100, and suggests the use of miR-378a-3p as a potential therapeutic target for dyslipidemia.
Collapse
|
76
|
A small molecule inhibitor of PCSK9 that antagonizes LDL receptor binding via interaction with a cryptic PCSK9 binding groove. Bioorg Med Chem 2020; 28:115344. [DOI: 10.1016/j.bmc.2020.115344] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
|
77
|
Sarkar P, Chattopadhyay A. Cholesterol interaction motifs in G protein-coupled receptors: Slippery hot spots? WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1481. [PMID: 32032482 DOI: 10.1002/wsbm.1481] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/28/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) are cell membrane associated signaling hubs that orchestrate a multitude of cellular functions upon binding to a diverse variety of extracellular ligands. Since GPCRs are integral membrane proteins with seven-transmembrane domain architecture, their function, organization and dynamics are intimately regulated by membrane lipids, such as cholesterol. Cholesterol is an extensively studied lipids in terms of its effects on GPCR structure and function. One of the possible mechanisms underlying modulation of GPCR function by cholesterol is via specific interaction of GPCRs with membrane cholesterol. These interactions of GPCRs with membrane cholesterol are often attributed to structural features of GPCRs that could facilitate their preferential association with cholesterol. In this backdrop, cholesterol interaction motifs represent putative interaction sites on GPCRs that could facilitate cholesterol-sensitive function of these receptors. In this review, we provide an overview of cholesterol interaction motifs found in GPCRs, which have been identified through a combination of crystallography, bioinformatics analysis, and functional studies. In addition, we will highlight, using specific examples, why mere presence of a cholesterol interaction motif at a given site may not directly implicate its role in interaction with membrane cholesterol. We therefore believe that experimental approaches, followed by functional analysis of cholesterol sensitivity of GPCRs, would provide a better understanding of the role played by these motifs in cholesterol-sensitive function. We envision that a comprehensive knowledge of cholesterol interaction sites in GPCRs would allow us to develop a better understanding of GPCR structure-function paradigm, and could be useful in future therapeutics. This article is categorized under: Models of Systems Properties and Processes > Mechanistic Models Analytical and Computational Methods > Computational Methods Laboratory Methods and Technologies > Macromolecular Interactions, Methods.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | |
Collapse
|
78
|
Tavernier G, Caspar-Bauguil S, Viguerie N. Apolipoprotein M: new connections with diet, adipose tissue and metabolic syndrome. Curr Opin Lipidol 2020; 31:8-14. [PMID: 31815756 DOI: 10.1097/mol.0000000000000654] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To focus on state-of-the-art knowledge on the apolipoprotein M (ApoM) physiology and physiopathology regarding metabolism. RECENT FINDINGS In humans, the ApoM was recently described as secreted by adipocytes. Obesity, metabolic syndrome and type 2 diabetes are associated with low circulating ApoM and adipose tissue APOM expression. Dieting-induced weight loss enhances adipose tissue expression and secretion, and exercise training increases plasma ApoM. The ApoM is a chaperone for the bioactive sphingolipid, sphingosine-1-phosphate (S1P), which has a specific role in inflammation. Its association with S1P in the inhibition of brown adipose tissue activity and subsequent insulin sensitivity was reported with the model of ApoM-deficient mouse. SUMMARY The adipose tissue is an endocrine organ responsible for obesity-related comorbidities. Obesity and dieting impact the adipose tissue secretory profile. The recent demonstration of ApoM being secreted by healthy adipocytes questions about the possible role of this adipose production in metabolic diseases. Low-circulating ApoM is associated with unhealthy metabolic phenotype. The lower circulating apoM during metabolic syndrome might be a cause of obesity-related comorbidities. Lifestyle interventions enhance ApoM production. Whether it acts in combination to S1P or other small lipidic molecules deserves further investigations.
Collapse
Affiliation(s)
- Geneviève Tavernier
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases (I2MC)
- University of Toulouse, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Paul Sabatier University
| | - Sylvie Caspar-Bauguil
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases (I2MC)
- University of Toulouse, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Paul Sabatier University
- Departments of Clinical Biochemistry and Nutrition, Toulouse University Hospitals, Toulouse, France
| | - Nathalie Viguerie
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases (I2MC)
- University of Toulouse, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Paul Sabatier University
| |
Collapse
|
79
|
Andreadou I, Schulz R, Badimon L, Adameová A, Kleinbongard P, Lecour S, Nikolaou PE, Falcão-Pires I, Vilahur G, Woudberg N, Heusch G, Ferdinandy P. Hyperlipidaemia and cardioprotection: Animal models for translational studies. Br J Pharmacol 2020; 177:5287-5311. [PMID: 31769007 DOI: 10.1111/bph.14931] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Hyperlipidaemia is a well-established risk factor for cardiovascular diseases and therefore, many animal model have been developed to mimic the human abnormal elevation of blood lipid levels. In parallel, extensive research for the alleviation of ischaemia/reperfusion injury has revealed that hyperlipidaemia is a major co-morbidity that attenuates the cardioprotective effect of conditioning strategies (preconditioning, postconditioning and remote conditioning) and that of pharmacological interventions by interfering with cardioprotective signalling pathways. In the present review article, we summarize the existing data on animal models of hypercholesterolaemia (total, low density and HDL abnormalities) and hypertriglyceridaemia used in ischaemia/reperfusion injury and protection from it. We also provide recommendations on preclinical animal models to be used for translations of the cardioprotective strategies into clinical practice. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic.,Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovak Republic
| | - Petra Kleinbongard
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Ines Falcão-Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Nicholas Woudberg
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gerd Heusch
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
80
|
Kumar NG, Contaifer D, Madurantakam P, Carbone S, Price ET, Van Tassell B, Brophy DF, Wijesinghe DS. Dietary Bioactive Fatty Acids as Modulators of Immune Function: Implications on Human Health. Nutrients 2019; 11:E2974. [PMID: 31817430 PMCID: PMC6950193 DOI: 10.3390/nu11122974] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/05/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Diet is major modifiable risk factor for cardiovascular disease that can influence the immune status of the individual and contribute to persistent low-grade inflammation. In recent years, there has been an increased appreciation of the role of polyunsaturated fatty acids (PUFA) in improving immune function and reduction of systemic inflammation via the modulation of pattern recognition receptors (PRR) on immune cells. Extensive research on the use of bioactive lipids such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and their metabolites have illustrated the importance of these pro-resolving lipid mediators in modulating signaling through PRRs. While their mechanism of action, bioavailability in the blood, and their efficacy for clinical use forms an active area of research, they are found widely administered as marine animal-based supplements like fish oil and krill oil to promote health. The focus of this review will be to discuss the effect of these bioactive fatty acids and their metabolites on immune cells and the resulting inflammatory response, with a brief discussion about modern methods for their analysis using mass spectrometry-based methods.
Collapse
Affiliation(s)
- Naren Gajenthra Kumar
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Daniel Contaifer
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Parthasarathy Madurantakam
- Department of General Practice, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA 23220, USA;
- VCU Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Elvin T. Price
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Benjamin Van Tassell
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Donald F. Brophy
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Dayanjan S. Wijesinghe
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
- da Vinci Center, Virginia Commonwealth University, Richmond, VA 23220, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University School of Pharmacy, Richmond, VA 23298, USA
| |
Collapse
|
81
|
Boretti FS, Burla B, Deuel J, Gao L, Wenk MR, Liesegang A, Sieber-Ruckstuhl NS. Serum lipidome analysis of healthy beagle dogs receiving different diets. Metabolomics 2019; 16:1. [PMID: 31797205 PMCID: PMC6890591 DOI: 10.1007/s11306-019-1621-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/22/2019] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Food and dietary ingredients have significant effects on metabolism and health. OBJECTIVE To evaluate whether and how different diets affected the serum lipidomic profile of dogs. METHODS Sixteen healthy beagles were fed a commercial dry diet for 3 months (control diet). After an overnight fasting period, a blood sample was taken for serum lipidomic profile analysis, and each dog was then randomly assigned to one of two groups. Group 1 was fed a commercial diet (Diet 1) and group 2 was fed a self-made, balanced diet supplemented with linseed oil and salmon oil (Diet 2) for 3 months. After an overnight fasting period, a blood sample was taken from each dog. Serum cholesterol and triacylglycerol analyses were performed and the serum lipidomic profiles were analyzed using targeted liquid chromatography-mass spectrometry. RESULTS Dogs fed the supplemented self-made diet (Diet 2) had significantly higher omega-3 fatty acid-containing lipids species and significantly lower saturated and mono- and di-unsaturated lipid species. Concentrations of sphingosine 1-phosphate species S1P d16:1 and S1P d17:1 were significantly increased after feeding Diet 2. CONCLUSION This study found that different diets had significant effects on the dog's serum lipidomic profile. Therefore, in studies that include lipidomic analyses, diet should be included as a confounding factor.
Collapse
Affiliation(s)
- Felicitas S Boretti
- Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Bo Burla
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Jeremy Deuel
- Divison of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Liang Gao
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Annette Liesegang
- Institute of Animal Nutrition, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Nadja S Sieber-Ruckstuhl
- Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
82
|
Torres Fernandez ED, Huffman AM, Syed M, Romero DG, Yanes Cardozo LL. Effect of GLP-1 Receptor Agonists in the Cardiometabolic Complications in a Rat Model of Postmenopausal PCOS. Endocrinology 2019; 160:2787-2799. [PMID: 31593246 PMCID: PMC6825516 DOI: 10.1210/en.2019-00450] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism and ovulatory dysfunction. Women with PCOS have an elevated prevalence of cardiometabolic risk factors that worsen after menopause. Liraglutide (Lira), a glucagon-like peptide-1 receptor agonist, has shown beneficial metabolic effects in small clinic trials in reproductive-age women with PCOS. We have shown that chronic hyperandrogenemia in an experimental model of postmenopausal PCOS is associated with an adverse cardiometabolic profile and upregulation of the intrarenal renin-angiotensin system (RAS). We analyzed the effect of Lira in the cardiometabolic profile, intrarenal RAS, and blood pressure (BP) in postmenopausal PCOS. Four-week-old female Sprague Dawley rats were treated with DHT or placebo for 17 months. Lira administration during the last 3 weeks caused a bigger reduction in food intake, body weight, fat mass, and homeostasis model assessment of insulin resistance index in PCOS than in control rats. Moreover, Lira improved dyslipidemia and elevated leptin levels in PCOS. In contrast, Lira decreased intrarenal expression of RAS components only in the control group. Lira transiently increased heart rate and decreased BP in control rats. However, Lira did not modify BP but increased heart rate in PCOS. The angiotensin-converting-enzyme inhibitor enalapril abolished the BP differences between PCOS and control rats. However, Lira coadministration with enalapril further reduced BP only in control rats. In summary, Lira has beneficial effects for several cardiometabolic risk factors in postmenopausal PCOS. However, hyperandrogenemia blunted the BP-lowering effect of Lira in postmenopausal PCOS. Androgen-induced activation of intrarenal RAS may play a major role mediating increases in BP in postmenopausal PCOS.
Collapse
Affiliation(s)
- Edgar D Torres Fernandez
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alexandra M Huffman
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Maryam Syed
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
- Correspondence: Licy L. Yanes Cardozo, MD, Departments of Cell & Molecular Biology and Medicine (Endocrinology), University of Mississippi Medical Center, 2500 North State Street, Jackson, Mississippi 39216. E-mail:
| |
Collapse
|
83
|
Kanasaki A, Jiang Z, Mizokami T, Shirouchi B, Iida T, Nagata Y, Sato M. Dietary d-allulose alters cholesterol metabolism in Golden Syrian hamsters partly by reducing serum PCSK9 levels. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
84
|
Thierer JH, Ekker SC, Farber SA. The LipoGlo reporter system for sensitive and specific monitoring of atherogenic lipoproteins. Nat Commun 2019; 10:3426. [PMID: 31366908 PMCID: PMC6668417 DOI: 10.1038/s41467-019-11259-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/28/2019] [Indexed: 12/18/2022] Open
Abstract
Apolipoprotein-B (ApoB) is the structural component of atherogenic lipoproteins, lipid-rich particles that drive atherosclerosis by accumulating in the vascular wall. As atherosclerotic cardiovascular disease is the leading cause of death worldwide, there is an urgent need to develop new strategies to prevent lipoproteins from causing vascular damage. Here we report the LipoGlo system, which uses a luciferase enzyme (NanoLuc) fused to ApoB to monitor several key determinants of lipoprotein atherogenicity including particle abundance, size, and localization. Using LipoGlo, we comprehensively characterize the lipoprotein profile of individual larval zebrafish and collect images of atherogenic lipoprotein localization in an intact organism. We report multiple extravascular lipoprotein localization patterns, as well as identify Pla2g12b as a potent regulator of lipoprotein size. ApoB-fusion proteins thus represent a sensitive and specific approach to study atherogenic lipoproteins and their genetic and small molecule modifiers.
Collapse
Affiliation(s)
- James H Thierer
- Carnegie Institution for Science Department of Embryology, 3520 San Martin Drive, Baltimore, MD, 21218, USA
- Johns Hopkins University Department of Biology, 3400N Charles Street, Baltimore, MD, 21218, USA
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Steven A Farber
- Carnegie Institution for Science Department of Embryology, 3520 San Martin Drive, Baltimore, MD, 21218, USA.
- Johns Hopkins University Department of Biology, 3400N Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
85
|
Rumora AE, LoGrasso G, Hayes JM, Mendelson FE, Tabbey MA, Haidar JA, Lentz SI, Feldman EL. The Divergent Roles of Dietary Saturated and Monounsaturated Fatty Acids on Nerve Function in Murine Models of Obesity. J Neurosci 2019; 39:3770-3781. [PMID: 30886017 PMCID: PMC6510336 DOI: 10.1523/jneurosci.3173-18.2019] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/23/2019] [Accepted: 02/08/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathy is the most common complication of prediabetes and diabetes and presents as distal-to-proximal loss of peripheral nerve function in the lower extremities. Neuropathy progression and disease severity in prediabetes and diabetes correlates with dyslipidemia in man and murine models of disease. Dyslipidemia is characterized by elevated levels of circulating saturated fatty acids (SFAs) that associate with the progression of neuropathy. Increased intake of monounsaturated fatty acid (MUFA)-rich diets confers metabolic health benefits; however, the impact of fatty acid saturation in neuropathy is unknown. This study examines the differential effect of SFAs and MUFAs on the development of neuropathy and the molecular mechanisms underlying the progression of the complication. Male mice Mus musculus fed a high-fat diet rich in SFAs developed robust peripheral neuropathy. This neuropathy was completely reversed by switching the mice from the SFA-rich high-fat diet to a MUFA-rich high-fat diet; nerve conduction velocities and intraepidermal nerve fiber density were restored. A MUFA oleate also prevented the impairment of mitochondrial transport and protected mitochondrial membrane potential in cultured sensory neurons treated with mixtures of oleate and the SFA palmitate. Moreover, oleate also preserved intracellular ATP levels, prevented apoptosis induced by palmitate treatment, and promoted lipid droplet formation in sensory neurons, suggesting that lipid droplets protect sensory neurons from lipotoxicity. Together, these results suggest that MUFAs reverse the progression of neuropathy by protecting mitochondrial function and transport through the formation of intracellular lipid droplets in sensory neurons.SIGNIFICANCE STATEMENT There is a global epidemic of prediabetes and diabetes, disorders that represent a continuum of metabolic disturbances in lipid and glucose metabolism. In the United States, 80 million individuals have prediabetes and 30 million have diabetes. Neuropathy is the most common complication of both disorders, carries a high morbidity, and, despite its prevalence, has no treatments. We report that dietary intervention with monounsaturated fatty acids reverses the progression of neuropathy and restores nerve function in high-fat diet-fed murine models of peripheral neuropathy. Furthermore, the addition of the monounsaturated fatty acid oleate to sensory neurons cultured under diabetic conditions shows that oleate prevents impairment of mitochondrial transport and mitochondrial dysfunction through a mechanism involving formation of axonal lipid droplets.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stephen I Lentz
- Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | | |
Collapse
|
86
|
Changes in the Canine Plasma Lipidome after Short- and Long-Term Excess Glucocorticoid Exposure. Sci Rep 2019; 9:6015. [PMID: 30979907 PMCID: PMC6461633 DOI: 10.1038/s41598-019-42190-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 03/20/2019] [Indexed: 02/07/2023] Open
Abstract
Glucocorticoids (GCs) are critical regulators of metabolic control in mammals and their aberrant function has been linked to several pathologies. GCs are widely used in human and veterinary clinical practice as potent anti-inflammatory and immune suppressive agents. Dyslipidaemia is a frequently observed consequence of GC treatment, typified by increased lipolysis, lipid mobilization, liponeogenesis, and adipogenesis. Dogs with excess GC show hyperlipidaemia, hypertension, and a higher risk of developing type 2 diabetes mellitus, but the risk of developing atherosclerotic lesions is low as compared to humans. This study aimed to examine alterations in the canine plasma lipidome in a model of experimentally induced short-term and long-term GC excess. Both treatments led to significant plasma lipidome alterations, which were more pronounced after long-term excess steroid exposure. In particular, monohexosylceramides, phosphatidylinositols, ether phosphatidylcholines, acyl phosphatidylcholines, triacylglycerols and sphingosine 1-phosphates showed significant changes. The present study highlights the hitherto unknown effects of GCs on lipid metabolism, which will be important in the further elucidation of the role and function of GCs as drugs and in metabolic and cardiovascular diseases.
Collapse
|
87
|
Fatakia SN, Sarkar P, Chattopadhyay A. A collage of cholesterol interaction motifs in the serotonin 1A receptor: An evolutionary implication for differential cholesterol interaction. Chem Phys Lipids 2019; 221:184-192. [PMID: 30822391 DOI: 10.1016/j.chemphyslip.2019.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/22/2022]
Abstract
The serotonin1A receptor is a representative member of the G protein-coupled receptor (GPCR) superfamily and acts as an important drug target. In our previous work, we comprehensively demonstrated that membrane cholesterol is necessary in the organization, dynamics and function of the serotonin1A receptor. In this context, analysis of high-resolution GPCR crystal structures in general and in silico studies of the serotonin1A receptor in particular, have suggested the presence of cholesterol interaction sites (hotspots) in various regions of the receptor. In this work, we have identified an evolutionarily conserved collage of four categories of cholesterol interaction motifs associated with transmembrane helix V and the adjacent intracellular loop 3 fragment of the vertebrate serotonin1A receptor. This collage of motifs represents a total of twenty diverse context-dependent cholesterol interaction configurations. We envision that the gamut of cholesterol interaction sites, characterized by sequence plasticity in cholesterol interaction, could be relevant in receptor-cholesterol interaction in membranes of varying cholesterol content and organization, as found in diverse cell types. We conclude that an evolutionarily conserved mechanism of GPCR-cholesterol interaction allows the serotonin1A receptor to adapt to diverse membrane cholesterol levels during natural evolution.
Collapse
Affiliation(s)
- Sarosh N Fatakia
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India.
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
88
|
Rodriguez-Duarte J, Galliussi G, Dapueto R, Rossello J, Malacrida L, Kamaid A, Schopfer FJ, Escande C, López GV, Batthyány C. A novel nitroalkene-α-tocopherol analogue inhibits inflammation and ameliorates atherosclerosis in Apo E knockout mice. Br J Pharmacol 2019; 176:757-772. [PMID: 30588602 DOI: 10.1111/bph.14561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is characterized by chronic low-grade inflammation with concomitant lipid accumulation in the arterial wall. Anti-inflammatory and anti-atherogenic properties have been described for a novel class of endogenous nitroalkenes (nitrated-unsaturated fatty acids), formed during inflammation and digestion/absorption processes. The lipid-associated antioxidant α-tocopherol is transported systemically by LDL particles including to the atheroma lesions. To capitalize on the overlapping and complementary salutary properties of endogenous nitroalkenes and α-tocopherol, we designed and synthesized a novel nitroalkene-α-tocopherol analogue (NATOH) to address chronic inflammation and atherosclerosis, particularly at the lesion sites. EXPERIMENTAL APPROACH We synthesized NATOH, determined its electrophilicity and antioxidant capacity and studied its effects over pro-inflammatory and cytoprotective pathways in macrophages in vitro. Moreover, we demonstrated its incorporation into lipoproteins and tissue both in vitro and in vivo, and determined its effect on atherosclerosis and inflammatory responses in vivo using the Apo E knockout mice model. KEY RESULTS NATOH exhibited similar antioxidant capacity to α-tocopherol and, due to the presence of the nitroalkenyl group, like endogenous nitroalkenes, it exerted electrophilic reactivity. NATOH was incorporated in vivo into the VLDL/LDL lipoproteins particles to reach the atheroma lesions. Furthermore, oral administration of NATOH down-regulated NF-κB-dependent expression of pro-inflammatory markers (including IL-1β and adhesion molecules) and ameliorated atherosclerosis in Apo E knockout mice. CONCLUSIONS AND IMPLICATIONS In toto, the data demonstrate a novel pharmacological strategy for the prevention of atherosclerosis based on a creative, natural and safe drug delivery system of a non-conventional anti-inflammatory compound (NATOH) with significant potential for clinical application.
Collapse
Affiliation(s)
- Jorge Rodriguez-Duarte
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Germán Galliussi
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Rosina Dapueto
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Jessica Rossello
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Leonel Malacrida
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Pathophysiology Department, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Andrés Kamaid
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Gloria V López
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| |
Collapse
|
89
|
Abstract
Metabolic syndrome is a complex disorder that comprises several other complex disorders, including obesity, hypertension, dyslipidemia, and diabetes. There are several rat models that encompass component features of MetS. Some models are inbred strains selected for one or more traits underlying MetS; others are population models with genetic risk for MetS traits, are induced by environmental stressors such as diet, are spontaneous monogenic mutant models, or are congenic strains derived from a combination of these models. Together they can be studied to identify the genetic and physiological underpinnings of MetS to identify candidate genes or mechanisms for study in human MetS subjects.
Collapse
Affiliation(s)
- Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
90
|
O'Brien PD, Hinder LM, Rumora AE, Hayes JM, Dauch JR, Backus C, Mendelson FE, Feldman EL. Juvenile murine models of prediabetes and type 2 diabetes develop neuropathy. Dis Model Mech 2018; 11:dmm.037374. [PMID: 30446513 PMCID: PMC6307897 DOI: 10.1242/dmm.037374] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/09/2018] [Indexed: 12/19/2022] Open
Abstract
Peripheral neuropathy (neuropathy) is a common complication of obesity and type 2 diabetes in children and adolescents. To model this complication in mice, 5-week-old male C57BL/6J mice were fed a high-fat diet to induce diet-induced obesity (DIO), a model of prediabetes, and a cohort of these animals was injected with low-dose streptozotocin (STZ) at 12 weeks of age to induce hyperglycemia and type 2 diabetes. Neuropathy assessments at 16, 24 and 36 weeks demonstrated that DIO and DIO-STZ mice displayed decreased motor and sensory nerve conduction velocities as early as 16 weeks, hypoalgesia by 24 weeks and cutaneous nerve fiber loss by 36 weeks, relative to control mice fed a standard diet. Interestingly, neuropathy severity was similar in DIO and DIO-STZ mice at all time points despite significantly higher fasting glucose levels in the DIO-STZ mice. These mouse models provide critical tools to better understand the underlying pathogenesis of prediabetic and diabetic neuropathy from youth to adulthood, and support the idea that hyperglycemia alone does not drive early neuropathy. This article has an associated First Person interview with the first author of the paper. Summary: The mouse models described in this paper provide critical tools to better understand the underlying pathogenesis of prediabetic and diabetic neuropathy from youth to adulthood.
Collapse
Affiliation(s)
- Phillipe D O'Brien
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Lucy M Hinder
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Amy E Rumora
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Jacqueline R Dauch
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Carey Backus
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Faye E Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
91
|
Sharma D, Hill AE, Christopher MM. Hypercholesterolemia and hypertriglyceridemia as biochemical markers of disease in companion rabbits. Vet Clin Pathol 2018; 47:589-602. [PMID: 30556916 DOI: 10.1111/vcp.12676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/07/2018] [Accepted: 06/08/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inflammation has important effects on lipid metabolism, but the relationship between hyperlipidemia, inflammation, and disease remains unknown in rabbits. While rabbits are sensitive to dietary hypercholesterolemia, the etiology of hyperlipidemia when fed non-atherogenic diets is uncertain. OBJECTIVES This study aimed to determine the association between hypercholesterolemia and patient characteristics, diseases, and select CBC and biochemistry analytes in rabbits, and to measure plasma lipoprotein lipid fractions in rabbits with inflammatory and other diseases. METHODS Complete blood count and plasma biochemistry data, including total cholesterol concentrations, were evaluated in 531 companion rabbits. Lipoprotein cholesterol fractions (non-high-density lipoprotein cholesterol [non-HDLc] and high-density lipoprotein [HDLc]) and triglycerides were measured using a colorimetric enzymatic assay in archived plasma from a subset of 267 rabbits. Rabbits were categorized by age, sex, spay/neuter status, breed, diet status (fed atherogenic dietary components or not), the organ system affected by disease, and the pathologic process. RESULTS Cholesterol was associated with fibrinogen (P = 0.01), globulins (P < 0.01), and heterophil (P < 0.01) concentrations. Adjusting for diet, rabbits with severe infection or sepsis (odds ratio [OR] = 13.25, 95% CI = 5.83-30.12), renal failure (OR = 14.42, 95% CI = 5.69-36.54), and hepatopathy (OR = 8.55, 95% CI = 3.55-20.62) had increased risks of hypercholesterolemia. Increased non-HDLc and triglyceride concentrations were also associated with these three disease states (P < 0.05). CONCLUSIONS Hyperlipidemia is associated with biochemical and CBC markers of inflammation, and with severe infection or sepsis, renal failure, and hepatopathy. Independent of diet, increased cholesterol, non-HDLc, and triglycerides are indicators of disease in companion rabbits.
Collapse
Affiliation(s)
- Diya Sharma
- Department of Pathology, Microbiology and Immunology, University of California-Davis School of Veterinary Medicine, Davis, California
| | - Ashley E Hill
- California Animal Health and Food Safety Laboratory System, University of California-Davis, Davis, California
| | - Mary M Christopher
- Department of Pathology, Microbiology and Immunology, University of California-Davis School of Veterinary Medicine, Davis, California
| |
Collapse
|
92
|
Multifaceted Effect of Rubus Occidentalis on Hyperglycemia and Hypercholesterolemia in Mice with Diet-Induced Metabolic Diseases. Nutrients 2018; 10:nu10121846. [PMID: 30513715 PMCID: PMC6315456 DOI: 10.3390/nu10121846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 11/17/2022] Open
Abstract
Metabolic syndrome is characterized by a combination of several metabolic disorders, including obesity, hyperglycemia, and hyperlipidemia. A simultaneous occurrence is one of the most crucial features of metabolic syndrome; therefore, we selected an animal model in which this would be reflected. We fed C57BL/6N mice a high-fat diet for 23 weeks to develop metabolic syndrome and examined the efficacy of Rubus occidentalis (RO) for hyperglycemia and hypercholesterolemia. Oral administration of RO for 16 weeks improved hyperglycemia as indicated by significantly decreased fasting glucose levels and a glucose tolerance test. Improvements were also observed in hypercholesterolemia, in which significant decreases in serum total cholesterol, non-high-density lipoprotein (non-HDL) cholesterol, apolipoprotein A-1, and apolipoprotein B levels were observed. The time comparison of major biomarkers, observed at the initiation and termination of the experimental period, consistently supported the beneficial effects of RO on each metabolic phenotype. In addition, RO treatment attenuated the excessive fat accumulation in hepatic and adipose tissue by decreasing the size and number of lipid droplets. These results suggested that RO simultaneously exerted antihyperglycemic and antihyperlipidemic effects in mice with diet-induced metabolic syndrome.
Collapse
|
93
|
Curcio CA. Soft Drusen in Age-Related Macular Degeneration: Biology and Targeting Via the Oil Spill Strategies. Invest Ophthalmol Vis Sci 2018; 59:AMD160-AMD181. [PMID: 30357336 PMCID: PMC6733535 DOI: 10.1167/iovs.18-24882] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AMD is a major cause of legal blindness in older adults approachable through multidisciplinary research involving human tissues and patients. AMD is a vascular-metabolic-inflammatory disease, in which two sets of extracellular deposits, soft drusen/basal linear deposit (BLinD) and subretinal drusenoid deposit (SDD), confer risk for end-stages of atrophy and neovascularization. Understanding how deposits form can lead to insights for new preventions and therapy. The topographic correspondence of BLinD and SDD with cones and rods, respectively, suggest newly realized exchange pathways among outer retinal cells and across Bruch's membrane and the subretinal space, in service of highly evolved, eye-specific physiology. This review focuses on soft drusen/BLinD, summarizing evidence that a major ultrastructural component is large apolipoprotein B,E-containing, cholesterol-rich lipoproteins secreted by the retinal pigment epithelium (RPE) that offload unneeded lipids of dietary and outer segment origin to create an atherosclerosis-like progression in the subRPE-basal lamina space. Clinical observations and an RPE cell culture system combine to suggest that soft drusen/BLinD form when secretions of functional RPE back up in the subRPE-basal lamina space by impaired egress across aged Bruch's membrane-choriocapillary endothelium. The soft drusen lifecycle includes growth, anterior migration of RPE atop drusen, then collapse, and atrophy. Proof-of-concept studies in humans and animal models suggest that targeting the “Oil Spill in Bruch's membrane” offers promise of treating a process in early AMD that underlies progression to both end-stages. A companion article addresses the antecedents of soft drusen within the biology of the macula.
Collapse
Affiliation(s)
- Christine A Curcio
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
94
|
Modulation of aquaporin gene expression by n-3 long-chain PUFA lipid structures in white and brown adipose tissue from hamsters. Br J Nutr 2018; 120:1098-1106. [DOI: 10.1017/s0007114518002519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractEPA (20 : 5n-3) and DHA (22 : 6n-3) fatty acids have weight-reducing properties with physiological activity depending on their molecular structure – that is, as TAG or ethyl esters (EE). Aquaporins (AQP) are membrane protein channels recognised as important players in fat metabolism, but their differential expression in white adipose tissue (WAT) and brown adipose tissue (BAT), as well as their modulation by dietary n-3 long-chain PUFA (LCPUFA) such as EPA and DHA, has never been investigated. In this study, the transcriptional profiles of AQP3, AQP5, AQP7 and selected lipid markers of WAT (subcutaneous and visceral) and BAT (interscapular) from hamsters fed diets containing n-3 LCPUFA in different lipid structures such as fish oil (FO, rich in EPA and DHA in the TAG form) and FO-EE (rich in EPA and DHA in the EE form) were used and compared with linseed oil (LSO) as the reference group. A clear effect of fat depot was observed for AQP3 and leptin (LEP), with the lowest values of mRNA found in BAT relative to WAT. The opposite occurred for PPARα. AQP7 was affected by diet, with FO-fed hamsters having higher mRNA levels compared with LSO-fed hamsters. The relative gene expression of AQP5, adiponectin (ADIPO), GLUT4 and PPARγ was influenced by both fat tissue and diet. Taken together, our results revealed a differential expression profile of AQP and some markers of lipid metabolism in both WAT and BAT in response to feeding n-3 LCPUFA in two different structural formats: TAG v. EE.
Collapse
|
95
|
Kang C, LeRoith D, Gallagher EJ. Diabetes, Obesity, and Breast Cancer. Endocrinology 2018; 159:3801-3812. [PMID: 30215698 PMCID: PMC6202853 DOI: 10.1210/en.2018-00574] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022]
Abstract
The rates of obesity and diabetes are increasing worldwide, whereas the age of onset for both obesity and diabetes are decreasing steadily. Obesity and diabetes are associated with multiple factors that contribute to the increased risk of a number of different cancers, including breast cancer. These factors are hyperinsulinemia, elevated IGFs, hyperglycemia, dyslipidemia, adipokines, inflammatory cytokines, and the gut microbiome. In this review, we discuss the current understanding of the complex signaling pathways underlying these multiple factors involved in the obesity/diabetes-breast cancer link, with a focus particularly on the roles of the insulin/IGF system and dyslipidemia in preclinical breast cancer models. We review some of the therapeutic strategies to target these metabolic derangements in cancer. Future research directions and potential therapeutic strategies are also discussed.
Collapse
Affiliation(s)
- Chifei Kang
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
96
|
Thompson GR. Atherosclerosis in cholesterol-fed rabbits and in homozygous and heterozygous LDL receptor-deficient humans. Atherosclerosis 2018; 276:148-154. [DOI: 10.1016/j.atherosclerosis.2018.07.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/10/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022]
|
97
|
van Leeuwen EM, Emri E, Merle BMJ, Colijn JM, Kersten E, Cougnard-Gregoire A, Dammeier S, Meester-Smoor M, Pool FM, de Jong EK, Delcourt C, Rodrigez-Bocanegra E, Biarnés M, Luthert PJ, Ueffing M, Klaver CCW, Nogoceke E, den Hollander AI, Lengyel I. A new perspective on lipid research in age-related macular degeneration. Prog Retin Eye Res 2018; 67:56-86. [PMID: 29729972 DOI: 10.1016/j.preteyeres.2018.04.006] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 12/15/2022]
Abstract
There is an urgency to find new treatment strategies that could prevent or delay the onset or progression of AMD. Different classes of lipids and lipoproteins metabolism genes have been associated with AMD in a multiple ways, but despite the ever-increasing knowledge base, we still do not understand fully how circulating lipids or local lipid metabolism contribute to AMD. It is essential to clarify whether dietary lipids, systemic or local lipoprotein metabolismtrafficking of lipids in the retina should be targeted in the disease. In this article, we critically evaluate what has been reported in the literature and identify new directions needed to bring about a significant advance in our understanding of the role for lipids in AMD. This may help to develop potential new treatment strategies through targeting the lipid homeostasis.
Collapse
Affiliation(s)
- Elisabeth M van Leeuwen
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eszter Emri
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Benedicte M J Merle
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Johanna M Colijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eveline Kersten
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Audrey Cougnard-Gregoire
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | - Sascha Dammeier
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Magda Meester-Smoor
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Eiko K de Jong
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Cécile Delcourt
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000, Bordeaux, France
| | | | | | | | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Caroline C W Klaver
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| | - Everson Nogoceke
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Imre Lengyel
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
98
|
Shinozawa E, Amano Y, Yamakawa H, Haba M, Shimada M, Tozawa R. Antidyslipidemic potential of a novel farnesoid X receptor antagonist in a hamster model of dyslipidemia: Comparative studies of other nonstatin agents. Pharmacol Res Perspect 2018. [PMID: 29541476 PMCID: PMC5842406 DOI: 10.1002/prp2.390] [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] [Indexed: 01/21/2023] Open
Abstract
We attempted to clarify the therapeutic capability of antagonists of the farnesoid X receptor (FXR), a nuclear receptor that regulates lipid and bile acid metabolism. Herein, we report the antidyslipidemic effects of a novel synthesized FXR antagonist, compound‐T1, utilizing a dyslipidemic hamster model. Compound‐T1 selectively inhibited chenodeoxycholic acid‐induced FXR activation (IC50, 2.1 nmol·L−1). A hamster model of diet‐induced hyperlipidemia was prepared to investigate the antidyslipidemic effects of compound‐T1 through comparative studies of the nonstatin lipid‐modulating agents ezetimibe, cholestyramine, and torcetrapib. In the hamster model, compound‐T1 (6 mg·kg−1·day−1, p.o.) increased the level of plasma high‐density lipoprotein (HDL)‐cholesterol (+22.2%) and decreased the levels of plasma non‐HDL‐cholesterol (−43.6%) and triglycerides (−31.1%). Compound‐T1 also increased hepatic cholesterol 7α‐hydroxylase expression and fecal bile acid excretion, and decreased hepatic cholesterol content. Moreover, the hamster model could reflect clinical results of other nonstatin agents. Torcetrapib especially increased large HDL particles compared with compound‐T1. Additionally, in the human hepatoma Huh‐7 cells, compound‐T1 enhanced apolipoprotein A‐I secretion at a concentration close to its IC50 value for FXR. Our results indicated the usefulness of the hamster model in evaluating FXR antagonists and nonstatin agents. Notably, compound‐T1 exhibited beneficial effects on both blood non‐HDL‐cholesterol and HDL‐cholesterol, which are thought to involve enhancement of cholesterol catabolism and apolipoprotein A‐I production. These findings aid the understanding of the antidyslipidemic potential of FXR antagonists with a unique lipid and bile acid modulation.
Collapse
Affiliation(s)
- Emiko Shinozawa
- Research Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Yuichiro Amano
- Research Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Hiroko Yamakawa
- Research Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Megumi Haba
- Research Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Mitsuyuki Shimada
- Research Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Ryuichi Tozawa
- Research Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| |
Collapse
|
99
|
Hamczyk MR, Villa-Bellosta R, Gonzalo P, Andrés-Manzano MJ, Nogales P, Bentzon JF, López-Otín C, Andrés V. Vascular Smooth Muscle-Specific Progerin Expression Accelerates Atherosclerosis and Death in a Mouse Model of Hutchinson-Gilford Progeria Syndrome. Circulation 2018; 138:266-282. [PMID: 29490993 PMCID: PMC6075893 DOI: 10.1161/circulationaha.117.030856] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 02/15/2018] [Indexed: 01/21/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Progerin, an aberrant protein that accumulates with age, causes the rare genetic disease Hutchinson-Gilford progeria syndrome (HGPS). Patients who have HGPS exhibit ubiquitous progerin expression, accelerated aging and atherosclerosis, and die in their early teens, mainly of myocardial infarction or stroke. The mechanisms underlying progerin-induced atherosclerosis remain unexplored, in part, because of the lack of appropriate animal models. Methods: We generated an atherosclerosis-prone model of HGPS by crossing apolipoprotein E–deficient (Apoe–/–) mice with LmnaG609G/G609G mice ubiquitously expressing progerin. To induce progerin expression specifically in macrophages or vascular smooth muscle cells (VSMCs), we crossed Apoe–/–LmnaLCS/LCS mice with LysMCre and SM22αCre mice, respectively. Progerin expression was evaluated by polymerase chain reaction and immunofluorescence. Cardiovascular alterations were determined by immunofluorescence and histology in male mice fed normal chow or a high-fat diet. In vivo low-density lipoprotein retention was assessed by intravenous injection of fluorescently labeled human low-density lipoprotein. Cardiac electric defects were evaluated by electrocardiography. Results: Apoe–/–LmnaG609G/G609G mice with ubiquitous progerin expression exhibited a premature aging phenotype that included failure to thrive and shortened survival. In addition, high-fat diet–fed Apoe–/–LmnaG609G/G609G mice developed a severe vascular pathology, including medial VSMC loss and lipid retention, adventitial fibrosis, and accelerated atherosclerosis, thus resembling most aspects of cardiovascular disease observed in patients with HGPS. The same vascular alterations were also observed in Apoe–/–LmnaLCS/LCSSM22αCre mice expressing progerin specifically in VSMCs, but not in Apoe–/–LmnaLCS/LCSLysMCre mice with macrophage-specific progerin expression. Moreover, Apoe–/–LmnaLCS/LCSSM22αCre mice had a shortened lifespan despite the lack of any overt aging phenotype. Aortas of ubiquitously and VSMC-specific progerin-expressing mice exhibited increased retention of fluorescently labeled human low-density lipoprotein, and atheromata in both models showed vulnerable plaque features. Immunohistopathological examination indicated that Apoe–/–LmnaLCS/LCSSM22αCre mice, unlike Apoe–/–LmnaG609G/G609G mice, die of atherosclerosis-related causes. Conclusions: We have generated the first mouse model of progerin-induced atherosclerosis acceleration, and demonstrate that restricting progerin expression to VSMCs is sufficient to accelerate atherosclerosis, trigger plaque vulnerability, and reduce lifespan. Our results identify progerin-induced VSMC death as a major factor triggering atherosclerosis and premature death in HGPS.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (M.R.H., M.J.A.-M., V.A.)
| | - Ricardo Villa-Bellosta
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.)
| | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.)
| | - María J Andrés-Manzano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (M.R.H., M.J.A.-M., V.A.)
| | - Paula Nogales
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.)
| | - Jacob F Bentzon
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (J.F.B.)
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Spain (C.L.-O.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain (C.L.-O.). The present affiliation for Dr Villa-Bellosta is Fundación Instituto de Investigación Sanitaria Fundación Jiménez Díaz (FIIS-FJD), Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.R.H., R.V.-B., P.G., M.J.A.-M., P.N., J.F.B., V.A.).
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (M.R.H., M.J.A.-M., V.A.)
| |
Collapse
|
100
|
Dietary supplementation with hybrid palm oil alters liver function in the common Marmoset. Sci Rep 2018; 8:2765. [PMID: 29426919 PMCID: PMC5807430 DOI: 10.1038/s41598-018-21151-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022] Open
Abstract
Hybrid palm oil, which contains higher levels of oleic acid and lower saturated fatty acids in comparison with African palm oil, has been proposed to be somehow equivalent to extra virgin olive oil. However, the biological effects of its consumption are poorly described. Here we have explored the effects of its overconsumption on lipid metabolism in a non-human primate model, the common marmoset. Dietary supplementation of marmoset with hyperlipidic diet containing hybrid palm oil for 3 months did not modify plasma lipids levels, but increased glucose levels as compared to the supplementation with African palm oil. Liver volume was unexpectedly found to be more increased in marmosets consuming hybrid palm oil than in those consuming African palm oil. Hepatic total lipid content and circulating transaminases were dramatically increased in animals consuming hybrid palm oil, as well as an increased degree of fibrosis. Analysis of liver miRNAs showed a selective modulation of certain miRNAs by hybrid palm oil, some of which were predicted to target genes involved in cell adhesion molecules and peroxisomal pathways. Our data suggest that consumption of hybrid palm oil should be monitored carefully, as its overconsumption compared to that of African palm oil could involve important alterations to hepatic metabolism.
Collapse
|