1
|
Hoekstra M, Snip OSC, Janusz P, Bernabé Kleijn MNA, Truitt ER, Sullivan BD, Schmidt TA, Jay GD, Van Eck M. Recombinant human proteoglycan 4 lowers inflammation and atherosclerosis susceptibility in female low-density lipoprotein receptor knockout mice. J Physiol 2024; 602:1939-1951. [PMID: 38606903 DOI: 10.1113/jp286354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Recombinant human proteoglycan 4 (rhPRG4) is a macromolecular mucin-like glycoprotein that is classically studied as a lubricant within eyes and joints. Given that endogenously produced PRG4 is present within atherosclerotic lesions and genetic PRG4 deficiency increases atherosclerosis susceptibility in mice, in the current study we investigated the anti-atherogenic potential of chronic rhPRG4 treatment. Female low-density lipoprotein receptor knockout mice were fed an atherogenic Western-type diet for 6 weeks and injected three times per week intraperitoneally with 0.5 mg rhPRG4 or PBS as control. Treatment with rhPRG4 was associated with a small decrease in plasma-free cholesterol levels, without a change in cholesteryl ester levels. A marked increase in the number of peritoneal foam cells was detected in response to the peritoneal rhPRG4 administration, which could be attributed to elevated peritoneal leukocyte MSR1 expression levels. However, rhPRG4-treated mice exhibited significantly smaller aortic root lesions of 278 ± 21 × 103 μm2 compared with 339 ± 15 × 103 μm2 in the aortic root of control mice. The overall decreased atherosclerosis susceptibility coincided with a shift in the monocyte and macrophage polarization states towards the patrolling and anti-inflammatory M2-like phenotypes, respectively. Furthermore, rhPRG4 treatment significantly reduced macrophage gene expression levels as well as plasma protein levels of the pro-inflammatory/pro-atherogenic cytokine TNF-alpha. In conclusion, we have shown that peritoneal administration and subsequent systemic exposure to rhPRG4 beneficially impacts the inflammatory state and reduces atherosclerosis susceptibility in mice. Our findings highlight that PRG4 is not only a lubricant but also acts as an anti-inflammatory agent. KEY POINTS: Endogenously produced proteoglycan 4 is found in atherosclerotic lesions and its genetic deficiency in mice is associated with enhanced atherosclerosis susceptibility. In this study we investigated the anti-atherogenic potential of chronic treatment with recombinant human PRG4 in hypercholesterolaemic female low-density lipoprotein receptor knockout mice. We show that recombinant human PRG4 stimulates macrophage foam cell formation, but also dampens the pro-inflammatory state of monocyte/macrophages, eventually leading to a significant reduction in plasma TNF-alpha levels and a lowered atherosclerosis susceptibility. Our findings highlight that peritoneal recombinant human PRG4 treatment can execute effects both locally and systemically and suggest that it will be of interest to study whether rhPRG4 treatment is also able to inhibit the progression and/or induce regression of previously established atherosclerotic lesions.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| | - Olga S C Snip
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Philip Janusz
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mireia N A Bernabé Kleijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Gregory D Jay
- Department of Emergency Medicine, Warren Alpert Medical School and Division of Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| |
Collapse
|
2
|
Ma F, Huang X, Cai B. Linking MASLD to ACVD through Kupffer cells. NATURE CARDIOVASCULAR RESEARCH 2024; 3:258-259. [PMID: 39196120 DOI: 10.1038/s44161-024-00442-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Bae S, Moon YA. Deletion of Elovl5 leads to dyslipidemia and atherosclerosis in LDLR-deficient mice. Biochem Biophys Res Commun 2024; 690:149292. [PMID: 38000296 DOI: 10.1016/j.bbrc.2023.149292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease for which hepatic steatosis and atherogenic dyslipidemia are significant risk factors. We investigated the effects of endogenously generated very-long-chain polyunsaturated fatty acids (VL-PUFAs) on dyslipidemia and atherosclerosis development using mice that lack ELOVL5, a PUFA elongase that is required for the synthesis of arachidonic acid, EPA, and DHA from the essential fatty acids linoleic and linolenic acids, and the LDL receptor (LDLR). Elovl5-/-;Ldlr-/- mice manifest increased liver triglyceride and cholesterol concentrations due to the activation of sterol regulatory element binding protein-1, a transcription factor that activates enzymes required for de novo lipogenesis. Plasma levels of triglycerides and cholesterol in VLDL, IDL, and LDL were markedly elevated in Elovl5-/-;Ldlr-/- mice fed a chow and the mice exhibited marked aortic atherosclerotic plaques. Bone marrow-derived monocytes from wild-type (WT) and Elovl5-/- mice were polarized to M1 and M2 macrophages, and the effects of ELOVL5 on inflammatory activity were determined. There were no differences in most of the markers tested for M1 and M2 polarized cells between WT and Elovl5-/- cells, except for a slight increase in PGE2 secretion in Elovl5-/- cells, likely due to elevated Cox-2 expression. These results suggest that the deletion of Elovl5 leads to hepatic steatosis and dyslipidemia, which are the major factors in severe atherosclerosis in Elovl5-/-;Ldlr-/- mice.
Collapse
Affiliation(s)
- Sijeong Bae
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea.
| |
Collapse
|
4
|
Cheng J, Huang H, Chen Y, Wu R. Nanomedicine for Diagnosis and Treatment of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304294. [PMID: 37897322 PMCID: PMC10754137 DOI: 10.1002/advs.202304294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/11/2023] [Indexed: 10/30/2023]
Abstract
With the changing disease spectrum, atherosclerosis has become increasingly prevalent worldwide and the associated diseases have emerged as the leading cause of death. Due to their fascinating physical, chemical, and biological characteristics, nanomaterials are regarded as a promising tool to tackle enormous challenges in medicine. The emerging discipline of nanomedicine has filled a huge application gap in the atherosclerotic field, ushering a new generation of diagnosis and treatment strategies. Herein, based on the essential pathogenic contributors of atherogenesis, as well as the distinct composition/structural characteristics, synthesis strategies, and surface design of nanoplatforms, the three major application branches (nanodiagnosis, nanotherapy, and nanotheranostic) of nanomedicine in atherosclerosis are elaborated. Then, state-of-art studies containing a sequence of representative and significant achievements are summarized in detail with an emphasis on the intrinsic interaction/relationship between nanomedicines and atherosclerosis. Particularly, attention is paid to the biosafety of nanomedicines, which aims to pave the way for future clinical translation of this burgeoning field. Finally, this comprehensive review is concluded by proposing unresolved key scientific issues and sharing the vision and expectation for the future, fully elucidating the closed loop from atherogenesis to the application paradigm of nanomedicines for advancing the early achievement of clinical applications.
Collapse
Affiliation(s)
- Jingyun Cheng
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Hui Huang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| | - Rong Wu
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
5
|
van der Sluis RJ, van den Aardweg T, Sijsenaar TJP, Van Eck M, Hoekstra M. Metyrapone Treatment Protects Low-Density Lipoprotein Receptor Knockout Mice against Hypercorticosteronemia Development without Changing Atherosclerosis Susceptibility. Biomolecules 2023; 13:1287. [PMID: 37759687 PMCID: PMC10526766 DOI: 10.3390/biom13091287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The steroid 11beta-hydroxylase inhibitor metyrapone is able to effectively reverse the hypercortisolemia detected in human Cushing's Syndrome patients. In this current preclinical study, we investigated whether metyrapone monotherapy can also reverse the hypercortisolemia-associated increase in atherosclerotic cardiovascular disease risk. In this instance, female low-density lipoprotein receptor knockout mice fed a cholic acid-containing high cholesterol/high fat diet to induce the development of hypercorticosteronemia and atherosclerotic lesions were treated twice daily with 100 mg/kg metyrapone for 4 weeks. Metyrapone effectively protected against hypercorticosteronemia development with endpoint plasma corticosterone levels remaining 43% lower than in controls (p < 0.01). Gene expression analysis in livers and adrenals validated that glucocorticoid receptor signaling was also reduced. Importantly, metyrapone treatment did not impact plasma cholesterol levels or alter atherosclerotic plaque areas or lesional collagen contents. However, metyrapone induced significant systemic lymphocytopenia as evident from marked decreases in splenic white pulp contents and thymus weights (-48% and -41%, respectively; p < 0.001). In conclusion, we have shown that treatment with metyrapone diminishes hypercorticosteronemia without affecting atherosclerosis susceptibility in cholic acid-containing high cholesterol/high fat diet-fed low-density lipoprotein receptor knockout mice. These preclinical findings highlight that restoring plasma glucocorticoid levels to normal is not necessarily sufficient to overcome the cardiovascular co-morbidities associated with human Cushing's disease.
Collapse
Affiliation(s)
- Ronald J. van der Sluis
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands; (R.J.v.d.S.); (M.V.E.)
| | - Tim van den Aardweg
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands; (R.J.v.d.S.); (M.V.E.)
| | - Timothy J. P. Sijsenaar
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands; (R.J.v.d.S.); (M.V.E.)
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands; (R.J.v.d.S.); (M.V.E.)
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands; (R.J.v.d.S.); (M.V.E.)
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| |
Collapse
|
6
|
Lee WE, Genetzakis E, Figtree GA. Novel Strategies in the Early Detection and Treatment of Endothelial Cell-Specific Mitochondrial Dysfunction in Coronary Artery Disease. Antioxidants (Basel) 2023; 12:1359. [PMID: 37507899 PMCID: PMC10376062 DOI: 10.3390/antiox12071359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Although elevated cholesterol and other recognised cardiovascular risk factors are important in the development of coronary artery disease (CAD) and heart attack, the susceptibility of humans to this fatal process is distinct from other animals. Mitochondrial dysfunction of cells in the arterial wall, particularly the endothelium, has been strongly implicated in the pathogenesis of CAD. In this manuscript, we review the established evidence and mechanisms in detail and explore the potential opportunities arising from analysing mitochondrial function in patient-derived cells such as endothelial colony-forming cells easily cultured from venous blood. We discuss how emerging technology and knowledge may allow us to measure mitochondrial dysfunction as a potential biomarker for diagnosis and risk management. We also discuss the "pros and cons" of animal models of atherosclerosis, and how patient-derived cell models may provide opportunities to develop novel therapies relevant for humans. Finally, we review several targets that potentially alleviate mitochondrial dysfunction working both via direct and indirect mechanisms and evaluate the effect of several classes of compounds in the cardiovascular context.
Collapse
Affiliation(s)
- Weiqian E. Lee
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Elijah Genetzakis
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
7
|
Yin F, Lin P, Yu WQ, Shen N, Li Y, Guo SD. The Cordyceps militaris-Derived Polysaccharide CM1 Alleviates Atherosclerosis in LDLR (-/-) Mice by Improving Hyperlipidemia. Front Mol Biosci 2021; 8:783807. [PMID: 34966782 PMCID: PMC8710727 DOI: 10.3389/fmolb.2021.783807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/26/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerotic cardiovascular disease has a high mortality worldwide. Our lab previously purified a polysaccharide designated as CM1 with (1→4)-β-D-Glcp and (1→2)-α-D-Manp glycosyls as the backbone. In this study, we investigated the anti-atherosclerosis effect of CM1 and the underlying mechanisms of action in a low-density lipoprotein receptor knockout (LDLR(-/-) mouse model. It was found that CM1 significantly decreased the formation of atherosclerotic plaques. Mechanistically, CM1 enhanced plasma level of apolipoprotein A-I and decreased the plasma levels of triglyceride, apolipoprotein B, and total cholesterol. In the absence of LDLR, CM1 elevated the expression of very low-density lipoprotein receptor for liver uptake of plasma apolipoprotein B-containing particles and reduced hepatic triglyceride synthesis by inhibiting sterol regulatory element binding protein 1c. CM1 improved lipids excretion by increasing the liver X receptor α/ATP-binding cassette G5 pathway in small intestine. CM1 reduced lipogenesis and lipolysis by inhibiting peroxisome proliferator-activated receptor γ and adipose triglyceride lipase in epididymal fat. Furthermore, CM1 improved lipid profile in C57BL/6J mice. Collectively, CM1 can modulate lipid metabolism by multiple pathways, contributing to reduced plasma lipid level and formation of atherosclerotic plaques in LDLR(-/-) mice. This molecule could be explored as a potential compound for prevention and treatment of hyperlipidemia and atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
8
|
Hoekstra M, Ren B, Laurila PP, Hildebrand RB, Soronen J, Frodermann V, Li Z, Boon MR, Geerling JJ, Rensen PCN, Jauhiainen M, Van Eck M. Hematopoietic upstream stimulating factor 1 deficiency is associated with increased atherosclerosis susceptibility in LDL receptor knockout mice. Sci Rep 2021; 11:16419. [PMID: 34385562 PMCID: PMC8361089 DOI: 10.1038/s41598-021-95858-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Total body upstream stimulatory factor 1 (USF1) deficiency in mice is associated with brown adipose tissue activation and a marked protection against the development of obesity and atherosclerotic lesions. Functional expression of USF1 has also been detected in monocytes and monocyte-derived macrophages. In the current study we therefore tested whether selective hematopoietic USF1 deficiency can also beneficially impact the development of atherosclerosis. For this purpose, LDL receptor knockout mice were transplanted with bone marrow from USF1 knockout mice or their wild-type littermate controls and subsequently fed a Western-type diet for 20 weeks to stimulate atherosclerotic lesion development. Strikingly, absence of USF1 function in bone marrow-derived cells was associated with exacerbated blood leukocyte (+ 100%; P < 0.01) and peritoneal leukocyte (+ 50%; P < 0.05) lipid loading and an increased atherosclerosis susceptibility (+ 31%; P < 0.05). These effects could be attributed to aggravated hyperlipidemia, i.e. higher plasma free cholesterol (+ 33%; P < 0.001) and cholesteryl esters (+ 39%; P < 0.001), and the development of hepatosteatosis. In conclusion, we have shown that hematopoietic USF1 deficiency is associated with an increased atherosclerosis susceptibility in LDL receptor knockout mice. These findings argue against a contribution of macrophage-specific USF1 deficiency to the previously described beneficial effect of total body USF1 deficiency on atherosclerosis susceptibility in mice.
Collapse
Affiliation(s)
- Menno Hoekstra
- Gorlaeus Laboratories, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| | - Baoyan Ren
- Gorlaeus Laboratories, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Pirkka-Pekka Laurila
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland.,Genomics and Biobank Unit, National Institute for Health and Welfare, Biomedicum 1, Helsinki, Finland.,Institute for Molecular Medicine Finland, FIMM, Helsinki, Finland
| | - Reeni B Hildebrand
- Gorlaeus Laboratories, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Jarkko Soronen
- Genomics and Biobank Unit, National Institute for Health and Welfare, Biomedicum 1, Helsinki, Finland.,Pharmaceuticals Division, Bayer Oy BOF-PH-MRA-MA, Medical Affairs PO, Espoo, Finland
| | - Vanessa Frodermann
- Gorlaeus Laboratories, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Zhuang Li
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëtte R Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Janine J Geerling
- Gorlaeus Laboratories, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Miranda Van Eck
- Gorlaeus Laboratories, Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
9
|
Jurtz VI, Skovbjerg G, Salinas CG, Roostalu U, Pedersen L, Hecksher-Sørensen J, Rolin B, Nyberg M, van de Bunt M, Ingvorsen C. Deep learning reveals 3D atherosclerotic plaque distribution and composition. Sci Rep 2020; 10:21523. [PMID: 33299076 PMCID: PMC7726562 DOI: 10.1038/s41598-020-78632-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Complications of atherosclerosis are the leading cause of morbidity and mortality worldwide. Various genetically modified mouse models are used to investigate disease trajectory with classical histology, currently the preferred methodology to elucidate plaque composition. Here, we show the strength of light-sheet fluorescence microscopy combined with deep learning image analysis for characterising and quantifying plaque burden and composition in whole aorta specimens. 3D imaging is a non-destructive method that requires minimal ex vivo handling and can be up-scaled to large sample sizes. Combined with deep learning, atherosclerotic plaque in mice can be identified without any ex vivo staining due to the autofluorescent nature of the tissue. The aorta and its branches can subsequently be segmented to determine how anatomical position affects plaque composition and progression. Here, we find the highest plaque accumulation in the aortic arch and brachiocephalic artery. Simultaneously, aortas can be stained for markers of interest (for example the pan immune cell marker CD45) and quantified. In ApoE-/- mice we observe that levels of CD45 reach a plateau after which increases in plaque volume no longer correlate to immune cell infiltration. All underlying code is made publicly available to ease adaption of the method.
Collapse
MESH Headings
- Animals
- Aorta/pathology
- Aortic Diseases
- Apolipoproteins E/analysis
- Atherosclerosis/complications
- Atherosclerosis/pathology
- Deep Learning
- Disease Models, Animal
- Female
- Image Processing, Computer-Assisted/methods
- Imaging, Three-Dimensional/methods
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence/methods
- Plaque, Atherosclerotic/diagnostic imaging
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Receptors, LDL/analysis
Collapse
Affiliation(s)
| | - Grethe Skovbjerg
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- Gubra, 2970, Hoersholm, Denmark
| | | | | | - Louise Pedersen
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- University of Copenhagen, 1017, Copenhagen, Denmark
| | | | - Bidda Rolin
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
- Gubra, 2970, Hoersholm, Denmark
| | - Michael Nyberg
- Novo Nordisk A/S, Novo Nordisk Park, 2760, Maaloev, Denmark
| | | | | |
Collapse
|
10
|
Scavenging of reactive dicarbonyls with 2-hydroxybenzylamine reduces atherosclerosis in hypercholesterolemic Ldlr -/- mice. Nat Commun 2020; 11:4084. [PMID: 32796843 PMCID: PMC7429830 DOI: 10.1038/s41467-020-17915-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 07/27/2020] [Indexed: 12/21/2022] Open
Abstract
Lipid peroxidation generates reactive dicarbonyls including isolevuglandins (IsoLGs) and malondialdehyde (MDA) that covalently modify proteins. Humans with familial hypercholesterolemia (FH) have increased lipoprotein dicarbonyl adducts and dysfunctional HDL. We investigate the impact of the dicarbonyl scavenger, 2-hydroxybenzylamine (2-HOBA) on HDL function and atherosclerosis in Ldlr−/− mice, a model of FH. Compared to hypercholesterolemic Ldlr−/− mice treated with vehicle or 4-HOBA, a nonreactive analogue, 2-HOBA decreases atherosclerosis by 60% in en face aortas, without changing plasma cholesterol. Ldlr−/− mice treated with 2-HOBA have reduced MDA-LDL and MDA-HDL levels, and their HDL display increased capacity to reduce macrophage cholesterol. Importantly, 2-HOBA reduces the MDA- and IsoLG-lysyl content in atherosclerotic aortas versus 4-HOBA. Furthermore, 2-HOBA reduces inflammation and plaque apoptotic cells and promotes efferocytosis and features of stable plaques. Dicarbonyl scavenging with 2-HOBA has multiple atheroprotective effects in a murine FH model, supporting its potential as a therapeutic approach for atherosclerotic cardiovascular disease. Hypercholesterolemia is associated with lipid peroxidation induced reactive dicarbonyl adducts. Here the authors show that the dicarbonyl scavenger, 2-hydroxybenzylamine(2-HOBA), decreases reactive dicarbonyl modifications of LDL and HDL, improves HDL function, reduces atherosclerosis and promotes features of stable plaques in a mouse model of hypercholestrolemia.
Collapse
|
11
|
Abstract
Atherosclerosis is a chronic inflammatory vascular disease and the predominant cause of heart attack and ischemic stroke. Despite the well-known sexual dimorphism in the incidence and complications of atherosclerosis, there are relatively limited data in the clinical and preclinical literature to rigorously address mechanisms underlying sex as a biological variable in atherosclerosis. In multiple histological and imaging studies, overall plaque burden and markers of inflammation appear to be greater in men than women and are predictive of cardiovascular events. However, while younger women are relatively protected from cardiovascular disease, by the seventh decade, the incidence of myocardial infarction in women ultimately surpasses that of men, suggesting an interaction between sex and age. Most preclinical studies in animal atherosclerosis models do not examine both sexes, and even in those that do, well-powered direct statistical comparisons for sex as an independent variable remain rare. This article reviews the available data. Overall, male animals appear to have more inflamed yet smaller plaques compared to female animals. Plaque inflammation is often used as a surrogate end point for plaque vulnerability in animals. The available data support the notion that rather than plaque size, plaque inflammation may be more relevant in assessing sex-specific mechanisms since the findings correlate with the sex difference in ischemic events and mortality and thus may be more reflective of the human condition. Overall, the number of preclinical studies directly comparing plaque inflammation between the sexes is extremely limited relative to the vast literature exploring atherosclerosis mechanisms. Failure to include both sexes and to address age in mechanistic atherosclerosis studies are missed opportunities to uncover underlying sex-specific mechanisms. Understanding the mechanisms driving sex as a biological variable in atherosclerotic disease is critical to future precision medicine strategies to mitigate what is still the leading cause of death of men and women worldwide.
Collapse
Affiliation(s)
- Joshua J. Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Joshua A. Beckman
- Cardiovascular Division, Vanderbilt University Medical Center, Nashville, TN
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| |
Collapse
|
12
|
Sofra X, Badami S. Adverse Effects of Sedentary Lifestyles: Inflammation, and High-Glucose Induced Oxidative Stress—A Double Blind Randomized Clinical Trial on Diabetic and Prediabetic Patients. Health (London) 2020. [DOI: 10.4236/health.2020.128076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
13
|
Mushenkova NV, Summerhill VI, Silaeva YY, Deykin AV, Orekhov AN. Modelling of atherosclerosis in genetically modified animals. Am J Transl Res 2019; 11:4614-4633. [PMID: 31497187 PMCID: PMC6731422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Atherosclerosis is a lipid-driven, chronic inflammatory disease that leads to plaque formation at specific sites of the arterial tree. Being the common cause of many cardiovascular disorders, atherosclerosis makes a tremendous impact on morbidity and mortality rates of cardiovascular diseases (CVDs) in countries with higher income. Animal models of atherosclerosis are utilized as useful tools for studying the aetiology, pathogenesis and complications of atherosclerosis, thus, providing a valuable platform for the efficacy testing of different pharmacological therapies and validation of imaging techniques. To date, a large variety of models is available. Pathophysiological changes can be induced in animals by either an atherogenic diet or genetic manipulations. The discussion of advantages and disadvantages of some murine, rabbit and porcine genetic models currently available for the atherosclerosis research is the scope of the following review.
Collapse
Affiliation(s)
| | - Volha I Summerhill
- Institute for Atherosclerosis Research, Skolkovo Innovative CentreMoscow 121609, Russia
| | - Yulia Yu Silaeva
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences34/5 Vavilova Street, Moscow 119334, Russia
| | - Alexey V Deykin
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences34/5 Vavilova Street, Moscow 119334, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative CentreMoscow 121609, Russia
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences34/5 Vavilova Street, Moscow 119334, Russia
| |
Collapse
|
14
|
Grajeda-Iglesias C, Aviram M. Specific Amino Acids Affect Cardiovascular Diseases and Atherogenesis via Protection against Macrophage Foam Cell Formation: Review Article. Rambam Maimonides Med J 2018; 9:RMMJ.10337. [PMID: 29944113 PMCID: PMC6115485 DOI: 10.5041/rmmj.10337] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The strong relationship between cardiovascular diseases (CVD), atherosclerosis, and endogenous or exogenous lipids has been recognized for decades, underestimating the contribution of other dietary components, such as amino acids, to the initiation of the underlying inflammatory disease. Recently, specific amino acids have been associated with incident cardiovascular disorders, suggesting their significant role in the pathogenesis of CVD. Special attention has been paid to the group of branched-chain amino acids (BCAA), leucine, isoleucine, and valine, since their plasma values are frequently found in high concentrations in individuals with CVD risk. Nevertheless, dietary BCAA, leucine in particular, have been associated with improved indicators of atherosclerosis. Therefore, their potential role in the process of atherogenesis and concomitant CVD development remains unclear. Macrophages play pivotal roles in the development of atherosclerosis. They can accumulate high amounts of circulating lipids, through a process known as macrophage foam cell formation, and initiate the atherogenesis process. We have recently screened for anti- or pro-atherogenic amino acids in the macrophage model system. Our study showed that glycine, cysteine, alanine, leucine, glutamate, and glutamine significantly affected macrophage atherogenicity mainly through modulation of the cellular triglyceride metabolism. The anti-atherogenic properties of glycine and leucine, and the pro-atherogenic effects of glutamine, were also confirmed in vivo. Further investigation is warranted to define the role of these amino acids in atherosclerosis and CVD, which may serve as a basis for the development of anti-atherogenic nutritional and therapeutic approaches.
Collapse
|
15
|
Nahon JE, Hoekstra M, Van Eck M. Total body proteoglycan 4 (Prg4) deficiency increases atherosclerosis susceptibility in apolipoprotein E knockout and low-density lipoprotein receptor knockout mice. Atherosclerosis 2018; 278:315-316. [PMID: 30477757 DOI: 10.1016/j.atherosclerosis.2018.06.883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Joya E Nahon
- Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Menno Hoekstra
- Leiden Academic Centre for Drug Research, Leiden, The Netherlands.
| | - Miranda Van Eck
- Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| |
Collapse
|
16
|
Goldberg IJ. 2017 George Lyman Duff Memorial Lecture: Fat in the Blood, Fat in the Artery, Fat in the Heart: Triglyceride in Physiology and Disease. Arterioscler Thromb Vasc Biol 2018; 38:700-706. [PMID: 29419410 PMCID: PMC5864527 DOI: 10.1161/atvbaha.117.309666] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Cholesterol is not the only lipid that causes heart disease. Triglyceride supplies the heart and skeletal muscles with highly efficient fuel and allows for the storage of excess calories in adipose tissue. Failure to transport, acquire, and use triglyceride leads to energy deficiency and even death. However, overabundance of triglyceride can damage and impair tissues. Circulating lipoprotein-associated triglycerides are lipolyzed by lipoprotein lipase (LpL) and hepatic triglyceride lipase. We inhibited these enzymes and showed that LpL inhibition reduces high-density lipoprotein cholesterol by >50%, and hepatic triglyceride lipase inhibition shifts low-density lipoprotein to larger, more buoyant particles. Genetic variations that reduce LpL activity correlate with increased cardiovascular risk. In contrast, macrophage LpL deficiency reduces macrophage function and atherosclerosis. Therefore, muscle and macrophage LpL have opposite effects on atherosclerosis. With models of atherosclerosis regression that we used to study diabetes mellitus, we are now examining whether triglyceride-rich lipoproteins or their hydrolysis by LpL affect the biology of established plaques. Following our focus on triglyceride metabolism led us to show that heart-specific LpL hydrolysis of triglyceride allows optimal supply of fatty acids to the heart. In contrast, cardiomyocyte LpL overexpression and excess lipid uptake cause lipotoxic heart failure. We are now studying whether interrupting pathways for lipid uptake might prevent or treat some forms of heart failure.
Collapse
Affiliation(s)
- Ira J Goldberg
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine.
| |
Collapse
|
17
|
Vongpromek R, Bos S, Ten Kate GJR, Bujo H, Jiang M, Nieman K, Schneider W, Roeters van Lennep JE, Verhoeven AJM, Sijbrands EJG, Mulder MT. Soluble LR11 associates with aortic root calcification in asymptomatic treated male patients with familial hypercholesterolemia. Atherosclerosis 2017. [PMID: 28637586 DOI: 10.1016/j.atherosclerosis.2017.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Despite statin treatment, a high prevalence of severe vascular calcification is found in patients with familial hypercholesterolemia (FH). We assessed the relation between the circulating soluble form of low-density lipoprotein receptor relative with 11 ligand-binding repeats (sLR11), a risk factor for cardiovascular disease, and vascular calcification in asymptomatic statin-treated heterozygous FH patients. METHODS In 123 asymptomatic heterozygous FH patients (age 40-69 years), aortic root (ARC), aortic valve (AVC) and coronary artery calcification (CAC) were determined with CT-based calcium scoring expressed in Agatston units. Plasma sLR11 levels were measured by sandwich ELISA. RESULTS Seventy-three patients displayed ARC, 48 had AVC and 96 CAC. Plasma sLR11 levels were positively correlated with the presence of ARC (r = 0.2, p = 0.03), but not with AVC or CAC. The correlation between sLR11 levels and ARC was restricted to male FH patients (r = 0.31, p = 0.006). Multivariate logistic analyses showed that the association of plasma sLR11 with the presence of ARC was independent of other determinants (Adjusted Odds Ratio, 2.01 (95% CI = 1.28-3.16) p = 0.002). CONCLUSIONS Plasma sLR11 is associated with ARC in male FH patients and may be mechanistically involved in the differential distribution of atherosclerotic lesions in the vasculature.
Collapse
Affiliation(s)
- Ranitha Vongpromek
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sven Bos
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gert-Jan R Ten Kate
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hideaki Bujo
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University, Sakura Medical Center, Sakura, Japan
| | - Meizi Jiang
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University, Sakura Medical Center, Sakura, Japan
| | - Koen Nieman
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wolfgang Schneider
- Department of Medical Biochemistry, Medical University of Vienna, Max. F. Perutz Laboratories, Vienna, Austria
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Adrie J M Verhoeven
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
18
|
Samsoondar JP, Burke AC, Sutherland BG, Telford DE, Sawyez CG, Edwards JY, Pinkosky SL, Newton RS, Huff MW. Prevention of Diet-Induced Metabolic Dysregulation, Inflammation, and Atherosclerosis in
Ldlr
−/−
Mice by Treatment With the ATP-Citrate Lyase Inhibitor Bempedoic Acid. Arterioscler Thromb Vasc Biol 2017; 37:647-656. [DOI: 10.1161/atvbaha.116.308963] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/17/2017] [Indexed: 02/05/2023]
Abstract
Objective—
Bempedoic acid (ETC-1002, 8-hydroxy-2,2,14,14-tetramethylpentadecanedioic acid) is a novel low-density lipoprotein cholesterol–lowering compound. In animals, bempedoic acid targets the liver where it inhibits cholesterol and fatty acid synthesis through inhibition of ATP-citrate lyase and through activation of AMP-activated protein kinase. In this study, we tested the hypothesis that bempedoic acid would prevent diet-induced metabolic dysregulation, inflammation, and atherosclerosis.
Approach and Results—
Ldlr
−/−
mice were fed a high-fat, high-cholesterol diet (42% kcal fat, 0.2% cholesterol) supplemented with bempedoic acid at 0, 3, 10 and 30 mg/kg body weight/day. Treatment for 12 weeks dose-dependently attenuated diet-induced hypercholesterolemia, hypertriglyceridemia, hyperglycemia, hyperinsulinemia, fatty liver and obesity. Compared to high-fat, high-cholesterol alone, the addition of bempedoic acid decreased plasma triglyceride (up to 64%) and cholesterol (up to 50%) concentrations, and improved glucose tolerance. Adiposity was significantly reduced with treatment. In liver, bempedoic acid prevented cholesterol and triglyceride accumulation, which was associated with increased fatty acid oxidation and reduced fatty acid synthesis. Hepatic gene expression analysis revealed that treatment significantly increased expression of genes involved in fatty acid oxidation while suppressing inflammatory gene expression. In full-length aorta, bempedoic acid markedly suppressed cholesteryl ester accumulation, attenuated the expression of proinflammatory M1 genes and attenuated the
iNos
/
Arg1
ratio. Treatment robustly attenuated atherosclerotic lesion development in the aortic sinus by 44%, with beneficial changes in morphology, characteristic of earlier-stage lesions.
Conclusions—
Bempedoic acid effectively prevents plasma and tissue lipid elevations and attenuates the onset of inflammation, leading to the prevention of atherosclerotic lesion development in a mouse model of metabolic dysregulation.
Collapse
Affiliation(s)
- Joshua P. Samsoondar
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Amy C. Burke
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Brian G. Sutherland
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Dawn E. Telford
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Cynthia G. Sawyez
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Jane Y. Edwards
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Stephen L. Pinkosky
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Roger S. Newton
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| | - Murray W. Huff
- From the Molecular Medicine Research Laboratory, Robarts Research Institute (J.P.S., A.C.B., B.G.S., D.E.T., C.G.S., J.Y.E., M.W.H.), Department of Biochemistry (J.P.S., A.C.B., M.W.H.), and Department of Medicine (D.E.T., C.G.S., J.Y.E., M.W.H.), The University of Western Ontario, London, Canada; and Esperion Therapeutics Inc, Ann Arbor, MI (S.L.P., R.S.N.)
| |
Collapse
|
19
|
Zha XQ, Zhang WN, Peng FH, Xue L, Liu J, Luo JP. Alleviating VLDL overproduction is an important mechanism for Laminaria japonica polysaccharide to inhibit atherosclerosis in LDLr -/- mice with diet-induced insulin resistance. Mol Nutr Food Res 2017; 61. [PMID: 27928899 DOI: 10.1002/mnfr.201600456] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 10/27/2016] [Accepted: 11/24/2016] [Indexed: 01/22/2023]
Abstract
SCOPE The overproduction of very low density lipoprotein (VLDL) is an important cause for initiation and development of atherosclerosis, which is highly associated with insulin signaling. The aim of this work is to verify whether the inhibition of VLDL overproduction is an underlying mechanism for a Laminaria japonica polysaccharide (LJP61A (where LJP is L. japonica)) to resist atherosclerosis. METHODS AND RESULTS LJP61A (50 and 200 mg/kg/day) was orally administered to a high-fat diet (HFD)-fed LDL receptor deficient mice for 14 weeks. LJP61A significantly attenuated insulin resistance, hepatic steatosis, atherosclerosis, and dyslipidemia. Meanwhile, LJP61A ameliorated the HFD-induced impairment of hepatic insulin signaling and reduced VLDL overproduction via regulating the expression of genes involved in the assembly and secretion of VLDL. To study the possibility that the inhibition of mammalian target of rapamycin complex 1 and stimulation of Forkhead box protein O1 (Foxo1) nuclear exclusion is a result of LJP61A via regulating insulin signaling, LJP61A was administrated to HepG2 cells in the presence or absence of mTOR inhibitor and Foxo1 inhibitor. Results showed that LJP61A alleviated VLDL overproduction via regulating insulin receptor substrate mediated phosphatidylinositide 3-kinase AKT mammalian target of rapamycin complex 1 and phosphatidylinositide 3-kinase AKT-Foxo1 signaling pathways. CONCLUSION These results suggested that LJP61A ameliorated HFD-induced insulin resistance to attenuate VLDL overproduction possibly via regulating insulin signaling, leading to the inhibition of atherosclerosis.
Collapse
Affiliation(s)
- Xue-Qiang Zha
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, China
- School of Food Science and Engineering, Hefei University of Technology, Hefei, China
| | - Wei-Nan Zhang
- School of Food Science and Engineering, Hefei University of Technology, Hefei, China
| | - Fu-Hua Peng
- School of Food Science and Engineering, Hefei University of Technology, Hefei, China
| | - Lei Xue
- School of Food Science and Engineering, Hefei University of Technology, Hefei, China
| | - Jian Liu
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, China
| | - Jian-Ping Luo
- School of Food Science and Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
20
|
von Scheidt M, Zhao Y, Kurt Z, Pan C, Zeng L, Yang X, Schunkert H, Lusis AJ. Applications and Limitations of Mouse Models for Understanding Human Atherosclerosis. Cell Metab 2017; 25:248-261. [PMID: 27916529 PMCID: PMC5484632 DOI: 10.1016/j.cmet.2016.11.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/26/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
Abstract
Most of the biological understanding of mechanisms underlying coronary artery disease (CAD) derives from studies of mouse models. The identification of multiple CAD loci and strong candidate genes in large human genome-wide association studies (GWASs) presented an opportunity to examine the relevance of mouse models for the human disease. We comprehensively reviewed the mouse literature, including 827 literature-derived genes, and compared it to human data. First, we observed striking concordance of risk factors for atherosclerosis in mice and humans. Second, there was highly significant overlap of mouse genes with human genes identified by GWASs. In particular, of the 46 genes with strong association signals in CAD GWASs that were studied in mouse models, all but one exhibited consistent effects on atherosclerosis-related phenotypes. Third, we compared 178 CAD-associated pathways derived from human GWASs with 263 from mouse studies and observed that the majority were consistent between the species.
Collapse
Affiliation(s)
- Moritz von Scheidt
- Deutsches Herzzentrum München, Technische Universität München, 80333 Munich, Germany
| | - Yuqi Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zeyneb Kurt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Departments of Medicine, Microbiology, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lingyao Zeng
- Deutsches Herzzentrum München, Technische Universität München, 80333 Munich, Germany
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, 80333 Munich, Germany; Deutsches Zentrum für Herz- und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Aldons J Lusis
- Departments of Medicine, Microbiology, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
21
|
Lee YT, Lin HY, Chan YWF, Li KHC, To OTL, Yan BP, Liu T, Li G, Wong WT, Keung W, Tse G. Mouse models of atherosclerosis: a historical perspective and recent advances. Lipids Health Dis 2017; 16:12. [PMID: 28095860 PMCID: PMC5240327 DOI: 10.1186/s12944-016-0402-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/27/2016] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis represents a significant cause of morbidity and mortality in both the developed and developing countries. Animal models of atherosclerosis have served as valuable tools for providing insights on its aetiology, pathophysiology and complications. They can be used for invasive interrogation of physiological function and provide a platform for testing the efficacy and safety of different pharmacological therapies. Compared to studies using human subjects, animal models have the advantages of being easier to manage, with controllable diet and environmental risk factors. Moreover, pathophysiological changes can be induced either genetically or pharmacologically to study the harmful effects of these interventions. There is no single ideal animal model, as different systems are suitable for different research objectives. A good understanding of the similarities and differences to humans enables effective extrapolation of data for translational application. In this article, we will examine the different mouse models for the study and elucidation of the pathophysiological mechanisms underlying atherosclerosis. We also review recent advances in the field, such as the role of oxidative stress in promoting endoplasmic reticulum stress, mitochondrial dysfunction and mitochondrial DNA damage, which can result in vascular inflammation and atherosclerosis. Finally, novel therapeutic approaches to reduce vascular damage caused by chronic inflammation using microRNA and nano-medicine technology, are discussed.
Collapse
Affiliation(s)
- Yee Ting Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR People’s Republic of China
| | - Hiu Yu Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR People’s Republic of China
| | | | | | - Olivia Tsz Ling To
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, SAR People’s Republic of China
| | - Bryan P Yan
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, SAR People’s Republic of China
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211 People’s Republic of China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211 People’s Republic of China
| | - Wing Tak Wong
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, SAR People’s Republic of China
| | - Wendy Keung
- Stem Cell & Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR People’s Republic of China
| | - Gary Tse
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, SAR People’s Republic of China
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR People’s Republic of China
| |
Collapse
|
22
|
Shewale SV, Brown AL, Bi X, Boudyguina E, Sawyer JK, Alexander-Miller MA, Parks JS. In vivo activation of leukocyte GPR120/FFAR4 by PUFAs has minimal impact on atherosclerosis in LDL receptor knockout mice. J Lipid Res 2016; 58:236-246. [PMID: 27811230 DOI: 10.1194/jlr.m072769] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Indexed: 01/15/2023] Open
Abstract
G protein-coupled receptor (GPR)120/FFA receptor (FFAR)4 (GPR120/FFAR4) activation by n-3 PUFAs attenuates inflammation, but its impact on atherosclerosis is unknown. We determined whether in vivo activation of leukocyte GPR120/FFAR4 by n-3 versus n-6 PUFAs is atheroprotective. Leukocyte GPR120/FFAR4 WT or KO mice in the LDL receptor KO background were generated by bone marrow transplantation. Mice were fed one of the four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) + 10% calories as: 1) PO, 2) fish oil (FO; 20:5 n-3 and 22:6 n-3 enriched), 3) echium oil (EO; 18:4 n-3 enriched), or 4) borage oil (BO; 18:3 n-6 enriched) for 16 weeks. Compared with PO, mice fed BO, EO, and FO had significantly reduced plasma cholesterol, TG, VLDL cholesterol, hepatic neutral lipid, and atherosclerosis that were equivalent for WT and KO mice. In BO-, EO-, and FO-fed mice, but not PO-fed mice, lack of leukocyte GPR120/FFAR4 resulted in neutrophilia, pro-inflammatory Ly6Chi monocytosis, increased aortic root monocyte recruitment, and increased hepatic inflammatory gene expression. In conclusion, leukocyte GPR120 expression has minimal effects on dietary PUFA-induced plasma lipid/lipoprotein reduction and atheroprotection, and there is no distinction between n-3 versus n-6 PUFAs in activating anti-inflammatory effects of leukocyte GPR120/FFAR4 in vivo.
Collapse
Affiliation(s)
- Swapnil V Shewale
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157.,Physiology/Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Amanda L Brown
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xin Bi
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Elena Boudyguina
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Janet K Sawyer
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | | | - John S Parks
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 .,Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
23
|
Getz GS, Reardon CA. Do the Apoe-/- and Ldlr-/- Mice Yield the Same Insight on Atherogenesis? Arterioscler Thromb Vasc Biol 2016; 36:1734-41. [PMID: 27386935 DOI: 10.1161/atvbaha.116.306874] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/24/2016] [Indexed: 02/02/2023]
Abstract
Murine models of atherosclerosis are useful for investigating the environmental and genetic influences on lesion formation and composition. Apoe(-/-) and Ldlr(-/-) mice are the 2 most extensively used models. The models differ in important ways with respect to the precise mechanism by which their absence enhances atherosclerosis, including differences in plasma lipoproteins. The majority of the gene function studies have utilized only 1 model, with the results being generalized to atherogenic mechanisms. In only a relatively few cases have studies been conducted in both atherogenic murine models. This review will discuss important differences between the 2 atherogenic models and will point out studies that have been performed in the 2 models where results are comparable and those where different results were obtained.
Collapse
Affiliation(s)
- Godfrey S Getz
- From the Department of Pathology (G.S.G.) and Ben May Institute for Cancer Biology (C.A.R.), University of Chicago, IL.
| | - Catherine A Reardon
- From the Department of Pathology (G.S.G.) and Ben May Institute for Cancer Biology (C.A.R.), University of Chicago, IL
| |
Collapse
|
24
|
Thacker SG, Rousset X, Esmail S, Zarzour A, Jin X, Collins HL, Sampson M, Stonik J, Demosky S, Malide DA, Freeman L, Vaisman BL, Kruth HS, Adelman SJ, Remaley AT. Increased plasma cholesterol esterification by LCAT reduces diet-induced atherosclerosis in SR-BI knockout mice. J Lipid Res 2015; 56:1282-95. [PMID: 25964513 PMCID: PMC4479333 DOI: 10.1194/jlr.m048629] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/27/2015] [Indexed: 12/15/2022] Open
Abstract
LCAT, a plasma enzyme that esterifies cholesterol, has been proposed to play an antiatherogenic role, but animal and epidemiologic studies have yielded conflicting results. To gain insight into LCAT and the role of free cholesterol (FC) in atherosclerosis, we examined the effect of LCAT over- and underexpression in diet-induced atherosclerosis in scavenger receptor class B member I-deficient [Scarab(-/-)] mice, which have a secondary defect in cholesterol esterification. Scarab(-/-)×LCAT-null [Lcat(-/-)] mice had a decrease in HDL-cholesterol and a high plasma ratio of FC/total cholesterol (TC) (0.88 ± 0.033) and a marked increase in VLDL-cholesterol (VLDL-C) on a high-fat diet. Scarab(-/-)×LCAT-transgenic (Tg) mice had lower levels of VLDL-C and a normal plasma FC/TC ratio (0.28 ± 0.005). Plasma from Scarab(-/-)×LCAT-Tg mice also showed an increase in cholesterol esterification during in vitro cholesterol efflux, but increased esterification did not appear to affect the overall rate of cholesterol efflux or hepatic uptake of cholesterol. Scarab(-/-)×LCAT-Tg mice also displayed a 51% decrease in aortic sinus atherosclerosis compared with Scarab(-/-) mice (P < 0.05). In summary, we demonstrate that increased cholesterol esterification by LCAT is atheroprotective, most likely through its ability to increase HDL levels and decrease pro-atherogenic apoB-containing lipoprotein particles.
Collapse
Affiliation(s)
- Seth G. Thacker
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xavier Rousset
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Safiya Esmail
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Abdalrahman Zarzour
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xueting Jin
- Experimental Atherosclerosis Section, Center for Molecular, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Maureen Sampson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - John Stonik
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephen Demosky
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Daniela A. Malide
- Light Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Lita Freeman
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Boris L. Vaisman
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Howard S. Kruth
- Experimental Atherosclerosis Section, Center for Molecular, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Alan T. Remaley
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
25
|
Shewale SV, Boudyguina E, Zhu X, Shen L, Hutchins PM, Barkley RM, Murphy RC, Parks JS. Botanical oils enriched in n-6 and n-3 FADS2 products are equally effective in preventing atherosclerosis and fatty liver. J Lipid Res 2015; 56:1191-205. [PMID: 25921305 DOI: 10.1194/jlr.m059170] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Echium oil (EO), which is enriched in 18:4 n-3, the immediate product of fatty acid desaturase 2 (FADS2) desaturation of 18:3 n-3, is as atheroprotective as fish oil (FO). The objective of this study was to determine whether botanical oils enriched in the FADS2 products 18:3 n-6 versus 18:4 n-3 are equally atheroprotective. LDL receptor KO mice were fed one of four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) plus 10% calories as: 1) PO; 2) borage oil (BO; 18:3 n-6 enriched); 3) EO (18:4 n-3 enriched); or 4) FO for 16 weeks. Mice fed BO, EO, and FO versus PO had significantly lower plasma total and VLDL cholesterol concentrations; hepatic neutral lipid content and inflammation, aortic CE content, aortic root intimal area and macrophage content; and peritoneal macrophage inflammation, CE content, and ex vivo chemotaxis. Atheromas lacked oxidized CEs despite abundant generation of macrophage 12/15 lipooxygenase-derived metabolites. We conclude that botanical oils enriched in 18:3 n-6 and 18:4 n-3 PUFAs beyond the rate-limiting FADS2 enzyme are equally effective in preventing atherosclerosis and hepatosteatosis compared with saturated/monounsaturated fat due to cellular enrichment of ≥20 PUFAs, reduced plasma VLDL, and attenuated macrophage inflammation.
Collapse
Affiliation(s)
- Swapnil V Shewale
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 Physiology/Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Elena Boudyguina
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xuewei Zhu
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Lulu Shen
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Patrick M Hutchins
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert M Barkley
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - John S Parks
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
26
|
Wang H, Zhang W, Wan J, Liu W, Yu B, Jin Q, Guan M. Microchip-based human serum atherogenic lipoprotein profile analysis. Anal Biochem 2014; 467:75-83. [DOI: 10.1016/j.ab.2014.08.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/24/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
|
27
|
van der Sluis RJ, van den Aardweg T, Reuwer AQ, Twickler MT, Boutillon F, Van Eck M, Goffin V, Hoekstra M. Prolactin receptor antagonism uncouples lipids from atherosclerosis susceptibility. J Endocrinol 2014; 222:341-50. [PMID: 25063756 DOI: 10.1530/joe-14-0343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The pituitary-derived hormone prolactin has been suggested to stimulate the development of atherosclerosis and cardiovascular disease through its effects on metabolism and inflammation. In this study, we aimed to challenge the hypothesis that inhibition of prolactin function may beneficially affect atherosclerosis burden. Hereto, atherosclerosis-susceptible LDL receptor (Ldlr) knockout mice were transplanted with bone marrow from transgenic mice expressing the pure prolactin receptor antagonist Del1-9-G129R-hPRL or their non-transgenic littermates as control. Recipient mice expressing Del1-9-G129R-hPRL exhibited a decrease in plasma cholesterol levels (-29%; P<0.05) upon feeding a Western-type diet (WTD), which could be attributed to a marked decrease (-47%; P<0.01) in the amount of cholesterol esters associated with pro-atherogenic lipoproteins VLDL/LDL. By contrast, Del1-9-G129R-hPRL-expressing mice did not display any change in the susceptibility for atherosclerosis after 12 weeks of WTD feeding. Both the absolute atherosclerotic lesion size (223 ± 33 × 10(3) μm(2) for Del1-9-G129R-hPRL vs 259 ± 32 × 10(3) μm(2) for controls) and the lesional macrophage and collagen contents were not different between the two groups of bone marrow recipients. Importantly, Del1-9-G129R-hPRL exposure increased levels of circulating neutrophils (+91%; P<0.05), lymphocytes (+55%; P<0.05), and monocytes (+43%; P<0.05), resulting in a 49% higher (P<0.01) total blood leukocyte count. In conclusion, we have shown that prolactin receptor signaling inhibition uncouples the plasma atherogenic index from atherosclerosis susceptibility in Ldlr knockout mice. Despite an associated decrease in VLDL/LDL cholesterol levels, application of the prolactin receptor antagonist Del1-9-G129R-hPRL does not alter the susceptibility for initial development of atherosclerotic lesions probably due to the parallel increase in circulating leukocyte concentrations.
Collapse
Affiliation(s)
- Ronald J van der Sluis
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Tim van den Aardweg
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Q Reuwer
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marcel T Twickler
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Florence Boutillon
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Miranda Van Eck
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincent Goffin
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Menno Hoekstra
- Division of BiopharmaceuticsGorlaeus Laboratories, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC Leiden, The NetherlandsLaboratory for Microbiology and Infection ControlAmphia Hospital, Breda, The NetherlandsDepartment EndocrinologyDiabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, BelgiumInsermUnit 1151,Prolactin/Growth Hormone Pathophysiology Laboratory, Faculty of Medicine, Institut Necker Enfants Malades (INEM), University Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
28
|
Affiliation(s)
- Xinghui Sun
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mark W Feinberg
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
29
|
Sontag TJ, Krishack PA, Lukens JR, Bhanvadia CV, Getz GS, Reardon CA. Apolipoprotein A-I protection against atherosclerosis is dependent on genetic background. Arterioscler Thromb Vasc Biol 2013; 34:262-9. [PMID: 24334873 DOI: 10.1161/atvbaha.113.302831] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Inbred mouse strains have different susceptibilities to experimental atherosclerosis. The C57BL/6 strain is among the most sensitive and has, therefore, been the most widely used in atherosclerosis studies, whereas many strains are resistant. The FVB/N strain is highly resistant to atherosclerosis on the apolipoprotein E (apoE)- and low-density lipoprotein (LDL) receptor-deficient backgrounds. High-density lipoprotein and its major apoprotein, apoA-I, have been shown to be protective against atherogenesis on the C57BL/6 background. We here examine the influence of genetic background on the atheroprotective nature of apoA-I. APPROACH AND RESULTS ApoE-deficient/apoA-I-deficient mice were generated in the C57BL/6 and FVB/N strains from apoE-deficient mice. After 6 to 10 weeks on a Western-type diet, plasma lipids and atherosclerotic lesion size were assessed. Macrophage recruitment, cholesterol regulation, and blood monocyte levels were examined as potential mechanisms driving lesion size differences. FVB/N knockout mice had higher plasma very-LDL/LDL cholesterol than their C57BL/6 counterparts. ApoA-I deficiency decreased very-LDL/LDL cholesterol in C57BL/6 mice but not in FVB/N mice. FVB/N single and double knockout mice had less lesion than C57BL/6 6 to 10 weeks on diet. ApoA-I deficiency augmented lesion development only in C57BL/6 mice. Macrophage recruitment to thioglycollate-treated peritoneum and diet-induced blood monocyte levels reflected the pattern of lesion development among the 4 genotypes. ApoA-I deficiency increased macrophage cholesterol content only in C57BL/6. FVB/N plasma was a better acceptor for macrophage cholesterol efflux than C57BL/6. CONCLUSIONS ApoA-I is atheroprotective only in certain genetic contexts. In the C57BL/6 context, but not FVB/N, apoA-I decreases inflammatory macrophage recruitment and monocytosis, contributors to lesion formation.
Collapse
|
30
|
Subramanian S, Turner MS, Ding Y, Goodspeed L, Wang S, Buckner JH, O'Brien K, Getz GS, Reardon CA, Chait A. Increased levels of invariant natural killer T lymphocytes worsen metabolic abnormalities and atherosclerosis in obese mice. J Lipid Res 2013; 54:2831-41. [PMID: 23922382 DOI: 10.1194/jlr.m041020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Obesity is a chronic inflammatory state characterized by infiltration of adipose tissue by immune cell populations, including T lymphocytes. Natural killer T (NKT) cells, a specialized lymphocyte subset recognizing lipid antigens, can be pro- or anti-inflammatory. Their role in adipose inflammation continues to be inconclusive and contradictory. In obesity, the infiltration of tissues by invariant NKT (iNKT) cells is decreased. We therefore hypothesized that an excess iNKT cell complement might improve metabolic abnormalities in obesity. Vα14 transgenic (Vα14tg) mice, with increased iNKT cell numbers, on a LDL receptor-deficient (Ldlr(-/-)) background and control Ldlr(-/-) mice were placed on an obesogenic diet for 16 weeks. Vα14tg.Ldlr(-/-) mice gained 25% more weight and had increased adiposity than littermate controls. Transgenic mice also developed greater dyslipidemia, hyperinsulinemia, insulin resistance, and hepatic triglyceride accumulation. Increased macrophage Mac2 immunostaining and proinflammatory macrophage gene expression suggested worsened adipose inflammation. Concurrently, these mice had increased atherosclerotic lesion area and aortic inflammation. Thus, increasing the complement of iNKT cells surprisingly exacerbated the metabolic, inflammatory, and atherosclerotic features of obesity. These findings suggest that the reduction of iNKT cells normally observed in obesity may represent a physiological attempt to compensate for this inflammatory condition.
Collapse
Affiliation(s)
- Savitha Subramanian
- Division of Metabolism, Endocrinology and Nutrition and University of Washington, Seattle, WA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tsimikas S, Miyanohara A, Hartvigsen K, Merki E, Shaw PX, Chou MY, Pattison J, Torzewski M, Sollors J, Friedmann T, Lai NC, Hammond HK, Getz GS, Reardon CA, Li AC, Banka CL, Witztum JL. Human oxidation-specific antibodies reduce foam cell formation and atherosclerosis progression. J Am Coll Cardiol 2013; 58:1715-27. [PMID: 21982317 DOI: 10.1016/j.jacc.2011.07.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 07/11/2011] [Accepted: 07/11/2011] [Indexed: 12/31/2022]
Abstract
OBJECTIVES We sought to assess the in vivo importance of scavenger receptor (SR)-mediated uptake of oxidized low-density lipoprotein (OxLDL) in atherogenesis and to test the efficacy of human antibody IK17-Fab or IK17 single-chain Fv fragment (IK17-scFv), which lacks immunologic properties of intact antibodies other than the ability to inhibit uptake of OxLDL by macrophages, to inhibit atherosclerosis. BACKGROUND The unregulated uptake of OxLDL by macrophage SR contributes to foam cell formation, but the importance of this pathway in vivo is uncertain. METHODS Cholesterol-fed low-density lipoprotein receptor knockout (LDLR(-/-)) mice were treated with intraperitoneal infusion of human IK17-Fab (2.5 mg/kg) 3 times per week for 14 weeks. Because anti-human antibodies developed in these mice, LDLR(-/-)/low-density lipoprotein receptor Rag 1 double-knockout mice (lacking the ability to make immunoglobulins due to loss of T- and B-cell function) were treated with an adenoviral vector encoding adenovirus expressed (Adv)-IK17-scFv or control adenovirus-enhanced green fluorescent protein vector intravenously every 2 weeks for 16 weeks. RESULTS In LDLR(-/-) mice, infusion of IK17-Fab was able to sustain IK17 plasma levels for the first 8 weeks, but these diminished afterward due to increasing murine anti-IK17 antibody titers. Despite this, after 14 weeks, a 29% decrease in en face atherosclerosis was noted compared with phosphate-buffered saline-treated mice. In LDLR(-/-)/low-density lipoprotein receptor Rag 1 double-knockout mice, sustained levels of plasma IK17-scFv was achieved by Adv-IK17-scFv-mediated hepatic expression, which led to a 46% reduction (p < 0.001) in en face atherosclerosis compared with adenovirus-enhanced green fluorescent protein vector. Importantly, peritoneal macrophages isolated from Adv-IK17-scFv treated mice had decreased lipid accumulation compared with adenovirus-enhanced green fluorescent protein-treated mice. CONCLUSIONS These data support an important role for SR-mediated uptake of OxLDL in the pathogenesis of atherosclerosis and demonstrate that oxidation-specific antibodies reduce the progression of atherosclerosis, suggesting their potential in treating cardiovascular disease in humans.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Klingenberg R, Gerdes N, Badeau RM, Gisterå A, Strodthoff D, Ketelhuth DFJ, Lundberg AM, Rudling M, Nilsson SK, Olivecrona G, Zoller S, Lohmann C, Lüscher TF, Jauhiainen M, Sparwasser T, Hansson GK. Depletion of FOXP3+ regulatory T cells promotes hypercholesterolemia and atherosclerosis. J Clin Invest 2013; 123:1323-34. [PMID: 23426179 DOI: 10.1172/jci63891] [Citation(s) in RCA: 294] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/20/2012] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease promoted by hyperlipidemia. Several studies support FOXP3-positive regulatory T cells (Tregs) as inhibitors of atherosclerosis; however, the mechanism underlying this protection remains elusive. To define the role of FOXP3-expressing Tregs in atherosclerosis, we used the DEREG mouse, which expresses the diphtheria toxin (DT) receptor under control of the Treg-specific Foxp3 promoter, allowing for specific ablation of FOXP3+ Tregs. Lethally irradiated, atherosclerosis-prone, low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice received DEREG bone marrow and were injected with DT to eliminate FOXP3(+) Tregs. Depletion of Tregs caused a 2.1-fold increase in atherosclerosis without a concomitant increase in vascular inflammation. These mice also exhibited a 1.7-fold increase in plasma cholesterol and an atherogenic lipoprotein profile with increased levels of VLDL. Clearance of VLDL and chylomicron remnants was hampered, leading to accumulation of cholesterol-rich particles in the circulation. Functional and protein analyses complemented by gene expression array identified reduced protein expression of sortilin-1 in liver and increased plasma enzyme activity of lipoprotein lipase, hepatic lipase, and phospholipid transfer protein as mediators of the altered lipid phenotype. These results demonstrate that FOXP3(+) Tregs inhibit atherosclerosis by modulating lipoprotein metabolism.
Collapse
Affiliation(s)
- Roland Klingenberg
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bubb KJ, Khambata RS, Ahluwalia A. Sexual dimorphism in rodent models of hypertension and atherosclerosis. Br J Pharmacol 2013; 167:298-312. [PMID: 22582712 DOI: 10.1111/j.1476-5381.2012.02036.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Approximately one third of all deaths are attributed to cardiovascular disease (CVD), making it the biggest killer worldwide. Despite a number of therapeutic options available, the burden of CVD morbidity continues to grow indicating the need for continued research to address this unmet need. In this respect, investigation of the mechanisms underlying the protection that premenopausal females enjoy from cardiovascular-related disease and mortality is of interest. In this review, we discuss the essential role that rodent animal models play in enabling this field of research. In particular, we focus our discussion on models of hypertension and atherosclerosis.
Collapse
Affiliation(s)
- Kristen J Bubb
- William Harvey Research Institute, Clinical Pharmacology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | | | | |
Collapse
|
34
|
Lu H, Wu C, Howatt DA, Balakrishnan A, Charnigo RJ, Cassis LA, Daugherty A. Differential effects of dietary sodium intake on blood pressure and atherosclerosis in hypercholesterolemic mice. J Nutr Biochem 2012; 24:49-53. [PMID: 22705323 DOI: 10.1016/j.jnutbio.2012.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 01/11/2023]
Abstract
The amount of dietary sodium intake regulates the renin angiotensin system (RAS) and blood pressure, both of which play critical roles in atherosclerosis. However, there are conflicting findings regarding the effects of dietary sodium intake on atherosclerosis. This study applied a broad range of dietary sodium concentrations to determine the concomitant effects of dietary sodium intake on the RAS, blood pressure, and atherosclerosis in mice. Eight-week-old male low-density lipoprotein receptor -/- mice were fed a saturated fat-enriched diet containing selected sodium concentrations (Na 0.01%, 0.1%, or 2% w/w) for 12 weeks. Mice in these three groups were all hypercholesterolemic, although mice fed Na 0.01% and Na 0.1% had higher plasma cholesterol concentrations than mice fed Na 2%. Mice fed Na 0.01% had greater abundances of renal renin mRNA than those fed Na 0.1% and 2%. Plasma renin concentrations were higher in mice fed Na 0.01% (14.2 ± 1.7 ng/ml/30 min) than those fed Na 0.1% or 2% (6.2 ± 0.6 and 5.8 ± 1.6 ng/ml per 30 min, respectively). However, systolic blood pressure at 12 weeks was higher in mice fed Na 2% (138 ± 3 mm Hg) than those fed Na 0.01% and 0.1% (129 ± 3 and 128 ± 4 mmHg, respectively). In contrast, mice fed Na 0.01% (0.17 ± 0.02 mm(2)) had larger atherosclerotic lesion areas in aortic roots than those fed Na 2% (0.09 ± 0.01 mm(2)), whereas lesion areas in mice fed Na 0.1% (0.12 ± 0.02 mm(2)) were intermediate between and not significantly different from those in Na 0.01% and Na 2% groups. In conclusion, while high dietary sodium intake led to higher systolic blood pressure, low dietary sodium intake augmented atherosclerosis in hypercholesterolemic mice.
Collapse
Affiliation(s)
- Hong Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Eberlé D, Kim RY, Luk FS, de Mochel NSR, Gaudreault N, Olivas VR, Kumar N, Posada JM, Birkeland AC, Rapp JH, Raffai RL. Apolipoprotein E4 domain interaction accelerates diet-induced atherosclerosis in hypomorphic Arg-61 apoe mice. Arterioscler Thromb Vasc Biol 2012; 32:1116-23. [PMID: 22441102 DOI: 10.1161/atvbaha.112.246389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Apolipoprotein (apo) E4 is an established risk factor for atherosclerosis, but the structural components underlying this association remain unclear. ApoE4 is characterized by 2 biophysical properties: domain interaction and molten globule state. Substituting Arg-61 for Thr-61 in mouse apoE introduces domain interaction without molten globule state, allowing us to delineate potential proatherogenic effects of domain interaction in vivo. METHODS AND RESULTS We studied atherosclerosis susceptibility of hypomorphic Apoe mice expressing either Thr-61 or Arg-61 apoE (ApoeT(h/h) or ApoeR(h/h)mice). On a chow diet, both mouse models were normolipidemic with similar levels of plasma apoE and lipoproteins. However, on a high-cholesterol diet, ApoeR(h/h) mice displayed increased levels of total plasma cholesterol and very-low-density lipoprotein as well as larger atherosclerotic plaques in the aortic root, arch, and descending aorta compared with ApoeT(h/h) mice. In addition, evidence of cellular dysfunction was identified in peritoneal ApoeR(h/h) macrophages which released lower amounts of apoE in culture medium and displayed increased expression of major histocompatibility complex class II molecules. CONCLUSIONS These data indicate that domain interaction mediates proatherogenic effects of apoE4 in part by modulating lipoprotein metabolism and macrophage biology. Pharmaceutical targeting of domain interaction could lead to new treatments for atherosclerosis in apoE4 individuals.
Collapse
Affiliation(s)
- Delphine Eberlé
- Department of Surgery, University of California San Francisco and VA Medical Center, San Francisco, CA 94121, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
van der Sluis RJ, van Puijvelde GH, Van Berkel TJ, Hoekstra M. Adrenalectomy stimulates the formation of initial atherosclerotic lesions: Reversal by adrenal transplantation. Atherosclerosis 2012; 221:76-83. [DOI: 10.1016/j.atherosclerosis.2011.12.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/12/2011] [Accepted: 12/16/2011] [Indexed: 01/26/2023]
|
37
|
Abstract
Atherosclerosis is a chronic inflammatory disorder that is the underlying cause of most cardiovascular disease. Both cells of the vessel wall and cells of the immune system participate in atherogenesis. This process is heavily influenced by plasma lipoproteins, genetics, and the hemodynamics of the blood flow in the artery. A variety of small and large animal models have been used to study the atherogenic process. No model is ideal as each has its own advantages and limitations with respect to manipulation of the atherogenic process and modeling human atherosclerosis or lipoprotein profile. Useful large animal models include pigs, rabbits, and nonhuman primates. Due in large part to the relative ease of genetic manipulation and the relatively short time frame for the development of atherosclerosis, murine models are currently the most extensively used. Although not all aspects of murine atherosclerosis are identical to humans, studies using murine models have suggested potential biological processes and interactions that underlie this process. As it becomes clear that different factors may influence different stages of lesion development, the use of mouse models with the ability to turn on or delete proteins or cells in tissue specific and temporal manner will be very valuable.
Collapse
Affiliation(s)
- Godfrey S Getz
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
38
|
Goldberg IJ, Eckel RH, McPherson R. Triglycerides and heart disease: still a hypothesis? Arterioscler Thromb Vasc Biol 2011; 31:1716-25. [PMID: 21527746 DOI: 10.1161/atvbaha.111.226100] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The purpose of this article is to review the basic and clinical science relating plasma triglycerides and cardiovascular disease. Although many aspects of the basic physiology of triglyceride production, its plasma transport, and its tissue uptake have been known for several decades, the relationship of plasma triglyceride levels to vascular disease is uncertain. Are triglyceride-rich lipoproteins, their influence on high-density lipoprotein and low-density lipoprotein, or the underlying diseases that lead to defects in triglyceride metabolism the culprit? Animal models have failed to confirm that anything other than early fatty lesions can be produced by triglyceride-rich lipoproteins. Metabolic products of triglyceride metabolism can be toxic to arterial cells; however, these studies are primarily in vitro. Correlative studies of fasting and postprandial triglycerides and genetic diseases implicate very-low-density lipoprotein and their remnants and chylomicron remnants in atherosclerosis development, but the concomitant alterations in other lipoproteins and other risk factors obscure any conclusions about direct relationships between disease and triglycerides. Genes that regulate triglyceride levels also correlate with vascular disease. Human intervention trials, however, have lacked an appropriately defined population and have produced outcomes without definitive conclusions. The time is more than ripe for new and creative approaches to understanding the relationship of triglycerides and heart disease.
Collapse
Affiliation(s)
- Ira J Goldberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | |
Collapse
|
39
|
Affiliation(s)
- Daniel Steinberg
- Department of Medicine, University of California, San Diego, La Jolla, Calif 92093, USA
| | | |
Collapse
|
40
|
Nookaew I, Gabrielsson BG, Holmäng A, Sandberg AS, Nielsen J. Identifying molecular effects of diet through systems biology: influence of herring diet on sterol metabolism and protein turnover in mice. PLoS One 2010; 5:e12361. [PMID: 20808764 PMCID: PMC2927425 DOI: 10.1371/journal.pone.0012361] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 07/06/2010] [Indexed: 12/20/2022] Open
Abstract
Background Changes in lifestyle have resulted in an epidemic development of obesity-related diseases that challenge the healthcare systems worldwide. To develop strategies to tackle this problem the focus is on diet to prevent the development of obesity-associated diseases such as cardiovascular disease (CVD). This will require methods for linking nutrient intake with specific metabolic processes in different tissues. Methodology/Principal Finding Low-density lipoprotein receptor-deficient (Ldlr −/−) mice were fed a high fat/high sugar diet to mimic a westernized diet, being a major reason for development of obesity and atherosclerosis. The diets were supplemented with either beef or herring, and matched in macronutrient contents. Body composition, plasma lipids and aortic lesion areas were measured. Transcriptomes of metabolically important tissues, e.g. liver, muscle and adipose tissue were analyzed by an integrated approach with metabolic networks to directly map the metabolic effects of diet in these different tissues. Our analysis revealed a reduction in sterol metabolism and protein turnover at the transcriptional level in herring-fed mice. Conclusion This study shows that an integrated analysis of transcriptome data using metabolic networks resulted in the identification of signature pathways. This could not have been achieved using standard clustering methods. In particular, this systems biology analysis could enrich the information content of biomedical or nutritional data where subtle changes in several tissues together affects body metabolism or disease progression. This could be applied to improve diets for subjects exposed to health risks associated with obesity.
Collapse
Affiliation(s)
- Intawat Nookaew
- Life Sciences/Systems Biology, Department of Chemical and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
41
|
Anzinger JJ, Chang J, Xu Q, Buono C, Li Y, Leyva FJ, Park BC, Greene LE, Kruth HS. Native low-density lipoprotein uptake by macrophage colony-stimulating factor-differentiated human macrophages is mediated by macropinocytosis and micropinocytosis. Arterioscler Thromb Vasc Biol 2010; 30:2022-31. [PMID: 20634472 DOI: 10.1161/atvbaha.110.210849] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To examine the pinocytotic pathways mediating native low-density lipoprotein (LDL) uptake by human macrophage colony-stimulating factor-differentiated macrophages (the predominant macrophage phenotype in human atherosclerotic plaques). METHODS AND RESULTS We identified the kinase inhibitor SU6656 and the Rho GTPase inhibitor toxin B as inhibitors of macrophage fluid-phase pinocytosis of LDL. Assessment of macropinocytosis by time-lapse microscopy revealed that both drugs almost completely inhibited macropinocytosis, although LDL uptake and cholesterol accumulation by macrophages were only partially inhibited (approximately 40%) by these agents. Therefore, we investigated the role of micropinocytosis in mediating LDL uptake in macrophages and identified bafilomycin A1 as an additional partial inhibitor (approximately 40%) of macrophage LDL uptake that targeted micropinocytosis. When macrophages were incubated with both bafilomycin A1 and SU6656, inhibition of LDL uptake was additive (reaching 80%), showing that these inhibitors target different pathways. Microscopic analysis of fluid-phase uptake pathways in these macrophages confirmed that LDL uptake occurs through both macropinocytosis and micropinocytosis. CONCLUSIONS Our findings show that human macrophage colony-stimulating factor-differentiated macrophages take up native LDL by macropinocytosis and micropinocytosis, underscoring the importance of both pathways in mediating LDL uptake by these cells.
Collapse
Affiliation(s)
- Joshua J Anzinger
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md 20892-1422, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mulvihill EE, Assini JM, Sutherland BG, DiMattia AS, Khami M, Koppes JB, Sawyez CG, Whitman SC, Huff MW. Naringenin decreases progression of atherosclerosis by improving dyslipidemia in high-fat-fed low-density lipoprotein receptor-null mice. Arterioscler Thromb Vasc Biol 2010; 30:742-8. [PMID: 20110573 DOI: 10.1161/atvbaha.109.201095] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Naringenin is a citrus flavonoid that potently inhibits the assembly and secretion of apolipoprotein B100-containing lipoproteins in cultured hepatocytes and improves the dyslipidemia and insulin resistance in a mouse model of the metabolic syndrome. In the present study, we used low-density lipoprotein receptor-null mice fed a high-fat diet (Western, TD96125) to test the hypothesis that naringenin prevents atherosclerosis. METHODS AND RESULTS Three groups (chow, Western, and Western plus naringenin) were fed ad libitum for 6 months. The Western diet increased fasting plasma triglyceride (TG) (5-fold) and cholesterol (8-fold) levels compared with chow, whereas the addition of naringenin significantly decreased both lipids by 50%. The Western-fed mice developed extensive atherosclerosis in the aortic sinus because plaque area was increased by 10-fold compared with chow-fed animals. Quantitation of fat-soluble dye (Sudan IV)-stained aortas, prepared en face, revealed that Western-fed mice also had a 10-fold increase in plaque deposits throughout the arch and in the abdominal sections of the aorta, compared with chow. Atherosclerosis in both areas was significantly decreased by more than 70% in naringenin-treated mice. Consistent with quantitation of aortic lesions, the Western-fed mice had a significant 6-fold increase in cholesterol and a 4-fold increase in TG deposition in the aorta compared with chow-fed mice. Both were reduced more than 50% by naringenin. The Western diet induced extensive hepatic steatosis, with a 10-fold increase in both TG and cholesteryl ester mass compared with chow. The addition of naringenin decreased both liver TG and cholesteryl ester mass by 80%. The hyperinsulinemia and obesity that developed in Western-fed mice was normalized by naringenin to levels observed in chow-fed mice. CONCLUSIONS These in vivo studies demonstrate that the citrus flavonoid naringenin ameliorates the dyslipidemia in Western-fed low-density lipoprotein receptor-null mice, leading to decreased atherosclerosis; and suggests a potential therapeutic strategy for the hyperlipidemia and increased risk of atherosclerosis associated with insulin resistance.
Collapse
Affiliation(s)
- Erin E Mulvihill
- Vascular Biology Group, Robarts Research Institute, The University of Western Ontario, 100 Perth Dr, London, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yasuda T, Ishida T, Rader DJ. Update on the Role of Endothelial Lipase in High-Density Lipoprotein Metabolism, Reverse Cholesterol Transport, and Atherosclerosis. Circ J 2010; 74:2263-70. [DOI: 10.1253/circj.cj-10-0934] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tomoyuki Yasuda
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, University of Pennsylvania School of Medicine
- Division of Cardiovascular Medicine, Kobe University Graduate school of Medicine
| | - Tatsuro Ishida
- Division of Cardiovascular Medicine, Kobe University Graduate school of Medicine
| | - Daniel J. Rader
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, University of Pennsylvania School of Medicine
| |
Collapse
|
44
|
Lu Y, Guo J, Di Y, Zong Y, Qu S, Tian J. Proteomic analysis of the triglyceride-rich lipoprotein-laden foam cells. Mol Cells 2009; 28:175-81. [PMID: 19756395 DOI: 10.1007/s10059-009-0120-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 07/01/2009] [Accepted: 07/22/2009] [Indexed: 11/29/2022] Open
Abstract
In hypertriglyceridaemic individuals, atherosclerogenesis is associated with the increased concentrations of very low density lipoprotein (VLDL) and VLDL-associated remnant particles. In vitro studies have suggested that VLDL induces foam cells formation. To reveal the changes of the proteins expression in the process of foam cells formation induced by VLDL, we performed a proteomic analysis of the foam cells based on the stimulation of differentiated THP-1 cells with VLDL. Using two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser-desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) analysis, 14 differentially expressed proteins, containing 8 up-regulated proteins and 6 down-regulated proteins were identified. The proteins are involved in energy metabolism, oxidative stress, cell growth, differentiation and apoptosis, such as adipose differentiation-related protein (ADRP), enolase, S100A11, heat shock protein 27 and so on. In addition, the expression of some selected proteins was confirmed by Western blot and RT-PCR analysis. The results suggest that VLDL not only induces lipid accumulation, but also brings about foam cells diverse characteristics by altering the expression of various proteins.
Collapse
Affiliation(s)
- Yanjun Lu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | | | | | | | | | | |
Collapse
|
45
|
Peng D, Hiipakka RA, Xie JT, Reardon CA, Getz GS, Liao S. Differential effects of activation of liver X receptor on plasma lipid homeostasis in wild-type and lipoprotein clearance-deficient mice. Atherosclerosis 2009; 208:126-33. [PMID: 19632679 DOI: 10.1016/j.atherosclerosis.2009.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 06/29/2009] [Accepted: 07/01/2009] [Indexed: 10/20/2022]
Abstract
The effects of liver X receptor (LXR) agonists on plasma lipid homeostasis, especially triglyceride metabolism are controversial. Here we examined the effect of long-term activation of LXR on plasma lipid homeostasis in wild-type C57BL/6 and LDL receptor deficient (LDLR-/-) mice given the LXR agonist T0901317 for 4 weeks. LXR agonist treatment of wild-type mice decreased plasma total triglycerides by 35% due to a significant reduction of plasma VLDL triglycerides. In contrast, in LDLR-/- mice T0901317 treatment increased plasma total cholesterol and triglycerides. An increase in the level of smaller VLDL particles was also observed in T0901317-treated LDLR-/- mice. The changes in circulating lipoprotein profiles in response to T0901317 treatment in these two animal models reflect the balance between synthesis and secretion on the one hand and lipolysis and clearance on the other. In both models there was both an increase in VLDL production and secretion and in an increase in LPL production and activity in T0901317-treated animals. In wild-type mice lipolysis and clearance predominates, while in the absence of the LDLR, which plays a major role in the clearance of apoB-containing lipoproteins, the increased output predominates. The generation of elevated levels of small VLDL particles due to increased lipolysis may represent an additional risk factor for atherosclerosis.
Collapse
Affiliation(s)
- Dacheng Peng
- Ben May Department for Cancer Research, University of Chicago, 929 E 57th Street, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|