1
|
Hong NE, Chaplin A, Di L, Ravodina A, Bevan GH, Gao H, Asase C, Gangwar RS, Cameron MJ, Mignery M, Cherepanova O, Finn AV, Nayak L, Pieper AA, Maiseyeu A. Nanoparticle-based itaconate treatment recapitulates low-cholesterol/low-fat diet-induced atherosclerotic plaque resolution. Cell Rep 2024; 43:114911. [PMID: 39466775 PMCID: PMC11648168 DOI: 10.1016/j.celrep.2024.114911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/22/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Current pharmacologic treatments for atherosclerosis do not completely protect patients; additional protection can be achieved by dietary modifications, such as a low-cholesterol/low-fat diet (LCLFD), that mediate plaque stabilization and inflammation reduction. However, this lifestyle modification can be challenging for patients. Unfortunately, incomplete understanding of the underlying mechanisms has thwarted efforts to mimic the protective effects of a LCLFD. Here, we report that the tricarboxylic acid cycle intermediate itaconate (ITA), produced by plaque macrophages, is key to diet-induced plaque resolution. ITA is produced by immunoresponsive gene 1 (IRG1), which we observe is highly elevated in myeloid cells of vulnerable plaques and absent from early or stable plaques in mice and humans. We additionally report development of an ITA-conjugated lipid nanoparticle that accumulates in plaque and bone marrow myeloid cells, epigenetically reduces inflammation via H3K27ac deacetylation, and reproduces the therapeutic effects of LCLFD-induced plaque resolution in multiple atherosclerosis models.
Collapse
Affiliation(s)
- Natalie E Hong
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alice Chaplin
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lin Di
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Anastasia Ravodina
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Graham H Bevan
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Huiyun Gao
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Courteney Asase
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Roopesh Singh Gangwar
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Division of Allergy and Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Matthew Mignery
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Olga Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aloke V Finn
- Department of Internal Medicine, Cardiovascular Division, University of Maryland School of Medicine, Baltimore, MD, USA; CVPath Institute, Inc., Gaithersburg, MD, USA
| | - Lalitha Nayak
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Hematology & Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
2
|
Izquierdo MC, Cabodevilla AG, Basu D, Nasias D, Kanter JE, Ho W, Gjini J, Fisher EA, Kim J, Lee W, Bornfeldt KE, Goldberg IJ. Hyperchylomicronemia causes endothelial cell inflammation and increases atherosclerosis. RESEARCH SQUARE 2024:rs.3.rs-5451391. [PMID: 39649171 PMCID: PMC11623764 DOI: 10.21203/rs.3.rs-5451391/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
The effect of increased triglycerides (TGs) as an independent factor in atherosclerosis development has been contentious, in part, because severe hypertriglyceridemia associates with low levels of low-density lipoprotein cholesterol (LDL-C). To test whether hyperchylomicronemia, in the absence of markedly reduced LDL-C levels, contributes to atherosclerosis, we created mice with induced whole-body lipoprotein lipase (LpL) deficiency combined with LDL receptor (LDLR) deficiency. On an atherogenic Western-type diet (WD), male and female mice with induced global LpL deficiency (iLpl -/-) and LDLR knockdown (Ldlr kd ) developed hypertriglyceridemia and elevated cholesterol levels; all the increased cholesterol was in chylomicrons or large VLDL. After 12 weeks on a WD, atherosclerotic lesions both in the brachiocephalic artery and the aortic root were more severe in iLpl -/- /Ldlr kd mice compared to the control Ldlr kd mice. One likely mechanism for this is that exposure of the aorta to hyperchylomicronemia led to endothelial cell inflammation. Thus, our data show that intact chylomicrons contribute to atherosclerosis, explain the association of postprandial lipemia and vascular disease, and prove that hyperchylomicronemia is not benign.
Collapse
Affiliation(s)
- Maria Concepcion Izquierdo
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Ainara G. Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Dimitris Nasias
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Jenny E. Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Winnie Ho
- Keenan Centre for Biomedical Research, St. Michael’s Hospital and Division of Critical Care, Department of Medicine, University of Toronto, Canada
| | - Jana Gjini
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey Kim
- Comparative Medicine Research Unit, University of Louisville School of Medicine, Louisville, KY
| | - Warren Lee
- Keenan Centre for Biomedical Research, St. Michael’s Hospital and Division of Critical Care, Department of Medicine, University of Toronto, Canada
| | - Karin E. Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
3
|
Tong J, Wang Z, Zhang J, Gao R, Liu X, Liao Y, Guo X, Wei Y. Advanced Applications of Nanomaterials in Atherosclerosis Diagnosis and Treatment: Challenges and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58072-58099. [PMID: 39432384 DOI: 10.1021/acsami.4c13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Atherosclerosis-induced coronary artery disease is a major cause of cardiovascular mortality. Clinically, conservative treatment strategies for atherosclerosis still focus on lifestyle interventions and the use of lipid-lowering and anticoagulant medications. Despite achieving some therapeutic effects, these approaches are limited by low bioavailability, long intervention periods, and significant side effects. With the advancement of nanotechnology, nanomaterials have demonstrated extraordinary potential in the biomedical field. Their excellent biocompatibility, surface modifiability, and high targeting capability not only enable efficient diagnosis of plaque progression but also allow precise drug delivery within atherosclerotic plaques, significantly enhancing drug bioavailability and reducing systemic side effects. Here, we systematically review the current research progress of nanomaterials in the field of atherosclerosis to summarize not only the types of nanomaterials but also their applications in both the diagnosis and treatment of atherosclerosis. Notably, in the context of plaque therapy, we provide a comprehensive overview of current nanomaterial applications based on their targeted therapeutic systems for different cell types within plaques. Additionally, we address the persistent challenge of clinical translation of nanomaterials by summarizing current issues and providing directions for innovation and improvement in nanomaterial design. Overall, we believe that this review systematically summarizes the applications and challenges of biomedical nanomaterials in atherosclerosis diagnosis and therapy, thereby offering insights and references for the development of therapeutic materials for atherosclerosis.
Collapse
Affiliation(s)
- Junran Tong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangfei Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
4
|
Smith TKT, Ghorbani P, LeBlond ND, Nunes JRC, O'Dwyer C, Ambursley N, Fong-McMaster C, Minarrieta L, Burkovsky LA, El-Hakim R, Trzaskalski NA, Locatelli CAA, Stotts C, Pember C, Rayner KJ, Kemp BE, Loh K, Harper ME, Mulvihill EE, St-Pierre J, Fullerton MD. AMPK-mediated regulation of endogenous cholesterol synthesis does not affect atherosclerosis in a murine Pcsk9-AAV model. Atherosclerosis 2024; 397:117608. [PMID: 38880706 DOI: 10.1016/j.atherosclerosis.2024.117608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND AND AIMS Dysregulated cholesterol metabolism is a hallmark of atherosclerotic cardiovascular diseases, yet our understanding of how endogenous cholesterol synthesis affects atherosclerosis is not clear. The energy sensor AMP-activated protein kinase (AMPK) phosphorylates and inhibits the rate-limiting enzyme in the mevalonate pathway HMG-CoA reductase (HMGCR). Recent work demonstrated that when AMPK-HMGCR signaling was compromised in an Apoe-/- model of hypercholesterolemia, atherosclerosis was exacerbated due to elevated hematopoietic stem and progenitor cell mobilization and myelopoiesis. We sought to validate the significance of the AMPK-HMGCR signaling axis in atherosclerosis using a non-germline hypercholesterolemia model with functional ApoE. METHODS Male and female HMGCR S871A knock-in (KI) mice and wild-type (WT) littermate controls were made atherosclerotic by intravenous injection of a gain-of-function Pcsk9D374Y-adeno-associated virus followed by high-fat and high-cholesterol atherogenic western diet feeding for 16 weeks. RESULTS AMPK activation suppressed endogenous cholesterol synthesis in primary bone marrow-derived macrophages from WT but not HMGCR KI mice, without changing other parameters of cholesterol regulation. Atherosclerotic plaque area was unchanged between WT and HMGCR KI mice, independent of sex. Correspondingly, there were no phenotypic differences observed in hematopoietic progenitors or differentiated immune cells in the bone marrow, blood, or spleen, and no significant changes in systemic markers of inflammation. When lethally irradiated female mice were transplanted with KI bone marrow, there was similar plaque content relative to WT. CONCLUSIONS Given previous work, our study demonstrates the importance of preclinical atherosclerosis model comparison and brings into question the importance of AMPK-mediated control of cholesterol synthesis in atherosclerosis.
Collapse
Affiliation(s)
- Tyler K T Smith
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Nicholas D LeBlond
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Julia R C Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Conor O'Dwyer
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Nia Ambursley
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Claire Fong-McMaster
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Lucía Minarrieta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Leah A Burkovsky
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rama El-Hakim
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Natasha A Trzaskalski
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Cassandra A A Locatelli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Cameron Stotts
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ciara Pember
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Katey J Rayner
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Bruce E Kemp
- Protein Chemistry and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Kim Loh
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Erin E Mulvihill
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada; Centre for Catalysis Research and Innovation, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada.
| |
Collapse
|
5
|
Feng X, Peng D, Qiu Y, Guo Q, Zhang X, Li Z, Pan C. Identification and Validation of Aging- and Endoplasmic Reticulum Stress-Related Genes in Periodontitis Using a Competing Endogenous RNA Network. Inflammation 2024:10.1007/s10753-024-02124-0. [PMID: 39136902 DOI: 10.1007/s10753-024-02124-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 01/03/2025]
Abstract
Periodontitis is a multifactorial chronic inflammatory disease that destroy periodontium. Apart from microbial infection and host immune responses, emerging evidence shows aging and endoplasmic reticulum stress (ER stress) play a key role in periodontitis pathogenesis. The aim of this study is to identify aging-related genes (ARGs) and endoplasmic reticulum stress-related genes (ERGs) in periodontitis. Data were obtained from the Gene Expression Omnibus (GEO), Human Ageing Genomic Resources (HAGR) and GeneCards databases to identify differentially expressed mRNAs/miRNAs/lncRNAs (DEmRNAs/DEmiRNAs/DElncRNAs), ARGs and ERGs, respectively. We used the MultiMiR database for the reverse prediction of miRNAs and predicted miRNA-lncRNA interactions using the STARBase database. Afterwards, we constructed a mRNA-miRNA-lncRNA ceRNA network. A total of 10 hub genes, namely LCK, LYN, CXCL8, IL6, HCK, IL1B, BTK, CXCL12, GNAI1 and FCER1G, and 5 DEmRNAs-ARGs-ERGs were then discovered. Further, weighted gene co-expression network analysis (WGCNA) and single sample gene set enrichment analysis (ssGSEA) were performed to explore co-expression modules and immune infiltration respectively. Finally, we used transmission electron microscope (TEM), inverted fluorescence microscopy, quantitative real-time polymerase chain reaction (qRT-PCR) and Western Blot to verify the bioinformatic results in periodontal ligament stem cells (PDLSCs) infected with Porphyromonas gingivalis (P. gingivalis). The experimental results broadly confirmed the accuracy of bioinformatic analysis. The present study established an aging- and ER stress-related ceRNA network in periodontitis, contributing to a deeper understanding of the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Xinran Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Da Peng
- School and Hospital of Stomatology, China Medical University, Shenyang, 110002, China
| | - Yunjing Qiu
- School of Nursing & Midwifery, Faculty of Health, University of Technology Sydney, Sydney, 2007, Australia
| | - Qian Guo
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhixuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Chunling Pan
- School and Hospital of Stomatology, China Medical University, Shenyang, 110002, China.
| |
Collapse
|
6
|
Poznyak AV, Yakovlev AA, Popov MА, Zhuravlev AD, Sukhorukov VN, Orekhov AN. WITHDRAWN: Coronary atherosclerotic plaque regression strategies. J Biomed Res 2024; 39:1-21. [PMID: 38808553 DOI: 10.7555/jbr.37.20230223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Ahead of Print article withdrawn by publisher.
Collapse
Affiliation(s)
| | - Alexey A Yakovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 109240, Russia
| | - Mikhail А Popov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Alexander D Zhuravlev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Vasily N Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| |
Collapse
|
7
|
Wang Y, Tinsley B, Spolitu S, Zadroga JA, Agarwal H, Sarecha AK, Ozcan L. Geranylgeranyl isoprenoids and hepatic Rap1a regulate basal and statin-induced expression of PCSK9. J Lipid Res 2024; 65:100515. [PMID: 38309417 PMCID: PMC10910342 DOI: 10.1016/j.jlr.2024.100515] [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: 10/26/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
LDL-C lowering is the main goal of atherosclerotic cardiovascular disease prevention, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition is now a validated therapeutic strategy that lowers serum LDL-C and reduces coronary events. Ironically, the most widely used medicine to lower cholesterol, statins, has been shown to increase circulating PCSK9 levels, which limits their efficacy. Here, we show that geranylgeranyl isoprenoids and hepatic Rap1a regulate both basal and statin-induced expression of PCSK9 and contribute to LDL-C homeostasis. Rap1a prenylation and activity is inhibited upon statin treatment, and statin-mediated PCSK9 induction is dependent on geranylgeranyl synthesis and hepatic Rap1a. Accordingly, treatment of mice with a small-molecule activator of Rap1a lowered PCSK9 protein and plasma cholesterol and inhibited statin-mediated PCSK9 induction in hepatocytes. The mechanism involves inhibition of the downstream RhoA-ROCK pathway and regulation of PCSK9 at the post-transcriptional level. These data further identify Rap1a as a novel regulator of PCSK9 protein and show that blocking Rap1a prenylation through lowering geranylgeranyl levels contributes to statin-mediated induction of PCSK9.
Collapse
Affiliation(s)
- Yating Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Brea Tinsley
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefano Spolitu
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - John A Zadroga
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Heena Agarwal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
8
|
Pinos I, Coronel J, Albakri A, Blanco A, McQueen P, Molina D, Sim J, Fisher EA, Amengual J. β-Carotene accelerates the resolution of atherosclerosis in mice. eLife 2024; 12:RP87430. [PMID: 38319073 PMCID: PMC10945528 DOI: 10.7554/elife.87430] [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] [Indexed: 02/07/2024] Open
Abstract
β-Carotene oxygenase 1 (BCO1) catalyzes the cleavage of β-carotene to form vitamin A. Besides its role in vision, vitamin A regulates the expression of genes involved in lipid metabolism and immune cell differentiation. BCO1 activity is associated with the reduction of plasma cholesterol in humans and mice, while dietary β-carotene reduces hepatic lipid secretion and delays atherosclerosis progression in various experimental models. Here we show that β-carotene also accelerates atherosclerosis resolution in two independent murine models, independently of changes in body weight gain or plasma lipid profile. Experiments in Bco1-/- mice implicate vitamin A production in the effects of β-carotene on atherosclerosis resolution. To explore the direct implication of dietary β-carotene on regulatory T cells (Tregs) differentiation, we utilized anti-CD25 monoclonal antibody infusions. Our data show that β-carotene favors Treg expansion in the plaque, and that the partial inhibition of Tregs mitigates the effect of β-carotene on atherosclerosis resolution. Our data highlight the potential of β-carotene and BCO1 activity in the resolution of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Johana Coronel
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Asma'a Albakri
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Amparo Blanco
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Patrick McQueen
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Donald Molina
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - JaeYoung Sim
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University Grossman School of Medicine, NYU Langone Medical CenterNew YorkUnited States
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| |
Collapse
|
9
|
Pinos I, Coronel J, Albakri A, Blanco A, McQueen P, Molina D, Sim J, Fisher EA, Amengual J. β-carotene accelerates the resolution of atherosclerosis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.07.531563. [PMID: 36945561 PMCID: PMC10028884 DOI: 10.1101/2023.03.07.531563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
β-carotene oxygenase 1 (BCO1) catalyzes the cleavage of β-carotene to form vitamin A. Besides its role in vision, vitamin A regulates the expression of genes involved in lipid metabolism and immune cell differentiation. BCO1 activity is associated with the reduction of plasma cholesterol in humans and mice, while dietary β-carotene reduces hepatic lipid secretion and delays atherosclerosis progression in various experimental models. Here we show that β-carotene also accelerates atherosclerosis resolution in two independent murine models, independently of changes in body weight gain or plasma lipid profile. Experiments in Bco1-/- mice implicate vitamin A production in the effects of β-carotene on atherosclerosis resolution. To explore the direct implication of dietary β-carotene on regulatory T cells (Tregs) differentiation, we utilized anti-CD25 monoclonal antibody infusions. Our data show that β-carotene favors Treg expansion in the plaque, and that the partial inhibition of Tregs mitigates the effect of β-carotene on atherosclerosis resolution. Our data highlight the potential of β-carotene and BCO1 activity in the resolution of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Johana Coronel
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| | - Asma'a Albakri
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Amparo Blanco
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Patrick McQueen
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Donald Molina
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| | - JaeYoung Sim
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University Grossman School of Medicine, NYU Langone Medical Center, NY
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| |
Collapse
|
10
|
Huang D, Zuo Y, Zhang C, Sun G, Jing Y, Lei J, Ma S, Sun S, Lu H, Cai Y, Zhang W, Gao F, Peng Xiang A, Belmonte JCI, Liu GH, Qu J, Wang S. A single-nucleus transcriptomic atlas of primate testicular aging reveals exhaustion of the spermatogonial stem cell reservoir and loss of Sertoli cell homeostasis. Protein Cell 2023; 14:888-907. [PMID: 36929025 PMCID: PMC10691849 DOI: 10.1093/procel/pwac057] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2022] Open
Abstract
The testis is pivotal for male reproduction, and its progressive functional decline in aging is associated with infertility. However, the regulatory mechanism underlying primate testicular aging remains largely elusive. Here, we resolve the aging-related cellular and molecular alterations of primate testicular aging by establishing a single-nucleus transcriptomic atlas. Gene-expression patterns along the spermatogenesis trajectory revealed molecular programs associated with attrition of spermatogonial stem cell reservoir, disturbed meiosis and impaired spermiogenesis along the sequential continuum. Remarkably, Sertoli cell was identified as the cell type most susceptible to aging, given its deeply perturbed age-associated transcriptional profiles. Concomitantly, downregulation of the transcription factor Wilms' Tumor 1 (WT1), essential for Sertoli cell homeostasis, was associated with accelerated cellular senescence, disrupted tight junctions, and a compromised cell identity signature, which altogether may help create a hostile microenvironment for spermatogenesis. Collectively, our study depicts in-depth transcriptomic traits of non-human primate (NHP) testicular aging at single-cell resolution, providing potential diagnostic biomarkers and targets for therapeutic interventions against testicular aging and age-related male reproductive diseases.
Collapse
Grants
- 2022M712216 National Key Research and Development Program of China
- 81921006, 82125011, 92149301, 92168201, 91949209, 92049304, 92049116, 32121001, 82192863, 82122024, 82071588, 32000500, 31900523, 82201714, 82271600, 82201727 National Natural Science Foundation of China
- 11000022T000000461062 Beijing-affiliated Medical Research
- CAS-WX2021SF-0301, CAS-WX2021SF-0101, CAS-WX2022SDC-XK14 Youth Innovation Promotion Association
- CAS-WX2021SF-0301 Youth Innovation Promotion Association
Collapse
Affiliation(s)
- Daoyuan Huang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuesheng Zuo
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
| | - Chen Zhang
- The Fifth People’s Hospital of Chongqing, Chongqing 400062, China
| | - Guoqiang Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Jing
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- Aging Biomarker Consortium, China
| | - Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
| | - Huifen Lu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
- Sino-Danish Center for Education and Research, Beijing 101408, China
- Aging Biomarker Consortium, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510000, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510000, China
| | | | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- Aging Biomarker Consortium, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- Aging Biomarker Consortium, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- The Fifth People’s Hospital of Chongqing, Chongqing 400062, China
- Aging Biomarker Consortium, China
| |
Collapse
|
11
|
Assini JM, Clark JR, Youssef A, Xing C, Doerfler AM, Park SH, Saxena L, Yaseen AB, Børen J, Gros R, Bao G, Lagor WR, Boffa MB, Koschinsky ML. High levels of lipoprotein(a) in transgenic mice exacerbate atherosclerosis and promote vulnerable plaque features in a sex-specific manner. Atherosclerosis 2023; 384:117150. [PMID: 37290980 DOI: 10.1016/j.atherosclerosis.2023.05.019] [Citation(s) in RCA: 1] [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] [Received: 02/28/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND AIMS Despite increased clinical interest in lipoprotein(a) (Lp(a)), many questions remain about the molecular mechanisms by which it contributes to atherosclerotic cardiovascular disease. Existing murine transgenic (Tg) Lp(a) models are limited by low plasma levels of Lp(a) and have not consistently shown a pro-atherosclerotic effect of Lp(a). METHODS We generated Tg mice expressing both human apolipoprotein(a) (apo(a)) and human apoB-100, with pathogenic levels of plasma Lp(a) (range 87-250 mg/dL). Female and male Lp(a) Tg mice (Tg(LPA+/0;APOB+/0)) and human apoB-100-only controls (Tg(APOB+/0)) (n = 10-13/group) were fed a high-fat, high-cholesterol diet for 12 weeks, with Ldlr knocked down using an antisense oligonucleotide. FPLC was used to characterize plasma lipoprotein profiles. Plaque area and necrotic core size were quantified and immunohistochemical assessment of lesions using a variety of cellular and protein markers was performed. RESULTS Male and female Tg(LPA+/0;APOB+/0) and Tg(APOB+/0) mice exhibited proatherogenic lipoprotein profiles with increased cholesterol-rich VLDL and LDL-sized particles and no difference in plasma total cholesterol between genotypes. Complex lesions developed in the aortic sinus of all mice. Plaque area (+22%), necrotic core size (+25%), and calcified area (+65%) were all significantly increased in female Tg(LPA+/0;APOB+/0) mice compared to female Tg(APOB+/0) mice. Immunohistochemistry of lesions demonstrated that apo(a) deposited in a similar pattern as apoB-100 in Tg(LPA+/0;APOB+/0) mice. Furthermore, female Tg(LPA+/0;APOB+/0) mice exhibited less organized collagen deposition as well as 42% higher staining for oxidized phospholipids (OxPL) compared to female Tg(APOB+/0) mice. Tg(LPA+/0;APOB+/0) mice had dramatically higher levels of plasma OxPL-apo(a) and OxPL-apoB compared to Tg(APOB+/0) mice, and female Tg(LPA+/0;APOB+/0) mice had higher plasma levels of the proinflammatory cytokine MCP-1 (+3.1-fold) compared to female Tg(APOB+/0) mice. CONCLUSIONS These data suggest a pro-inflammatory phenotype exhibited by female Tg mice expressing Lp(a) that appears to contribute to the development of more severe lesions with greater vulnerable features.
Collapse
Affiliation(s)
- Julia M Assini
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Justin R Clark
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Chuce Xing
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Alexandria M Doerfler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, Houston, USA
| | - Lavanya Saxena
- Department of Bioengineering, Rice University, Houston, USA
| | - Adam B Yaseen
- Department of Bioengineering, Rice University, Houston, USA
| | - Jan Børen
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Robert Gros
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| | - Gang Bao
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Department of Bioengineering, Rice University, Houston, USA
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada.
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
12
|
Wang Y, Tinsley B, Spolitu S, Zadroga JA, Agarwal H, Sarecha AK, Ozcan L. Geranylgeranyl Isoprenoids and Hepatic Rap1a Regulate Basal and Statin-Induced Expression of PCSK9. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563509. [PMID: 37961667 PMCID: PMC10634727 DOI: 10.1101/2023.10.23.563509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Low-density lipoprotein cholesterol (LDL-C) lowering is the main goal of atherosclerotic cardiovascular disease prevention, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition is now a validated therapeutic strategy that lowers serum LDL-C and reduces coronary events. Ironically, the most widely used medicine to lower cholesterol, statins, has been shown to increase circulating PCSK9 levels, which limits their efficacy. Here, we show that geranylgeranyl isoprenoids and hepatic Rap1a regulate both basal and statin induced expression of PCSK9 and contribute to LDL-C homeostasis. Rap1a prenylation and activity is inhibited upon statin treatment, and statin mediated PCSK9 induction is dependent on geranylgeranyl synthesis and hepatic Rap1a. Accordingly, treatment of mice with a small molecule activator of Rap1a lowered PCSK9 protein and plasma cholesterol and inhibited statin mediated PCSK9 induction in hepatocytes. The mechanism involves inhibition of the downstream RhoA-ROCK pathway and regulation of PCSK9 at the post transcriptional level. These data further identify Rap1a as a novel regulator of PCSK9 protein and show that blocking Rap1a prenylation through lowering geranylgeranyl levels contributes to statin-mediated induction of PCSK9.
Collapse
|
13
|
Burr SD, Chen Y, Hartley CP, Zhao X, Liu J. Replacement of saturated fatty acids with linoleic acid in western diet attenuates atherosclerosis in a mouse model with inducible ablation of hepatic LDL receptor. Sci Rep 2023; 13:16832. [PMID: 37803087 PMCID: PMC10558454 DOI: 10.1038/s41598-023-44030-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023] Open
Abstract
Dietary saturate fatty acids (SFAs) have been consistently linked to atherosclerosis and obesity, both of which are characterized by chronic inflammation and impaired lipid metabolism. In comparison, the effects of linoleic acid (LA), the predominant polyunsaturated fatty acid in the Western diet, seem to diverge. Data from human studies suggest a positive association between high dietary intake of LA and the improvement of cardiovascular risk. However, excessive LA intake has been implicated in the development of obesity. Concerns have also been raised on the potential pro-inflammatory properties of LA metabolites. Herein, by utilizing a mouse model with liver-specific Ldlr knockdown, we directly determined the effects of replacing SFAs with LA in a Western diet on the development of obesity and atherosclerosis. Specifically, mice treated with a Ldlr ASO were placed on a Western diet containing either SFA-rich butter (WD-B) or LA-rich corn oil (WD-CO) for 12 weeks. Despite of showing no changes in body weight gain or adiposity, mice on WD-CO exhibited significantly less atherosclerotic lesions compared to those on WD-B diet. Reduced lesion formation in the WD-CO-fed mice corresponded with a reduction of plasma triglyceride and cholesterol content, especially in VLDL and LDL, and ApoB protein levels. Although it increased expression of proinflammatory cytokines TNF-α and IL-6 in the liver, WD-CO did not appear to affect hepatic injury or damage when compared to WD-B. Collectively, our results indicate that replacing SFAs with LA in a Western diet could reduce the development of atherosclerosis independently of obesity.
Collapse
Affiliation(s)
- Stephanie D Burr
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic in Rochester, Guggenheim Building 14-11A, 222 3Rd Avenue SW, Rochester, MN, 55905, USA
| | - Yongbin Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Christopher P Hartley
- Department of Laboratory Medicine and Pathology, Mayo Clinic in Rochester, Rochester, MN, 55905, USA
| | - Xianda Zhao
- Department of Laboratory Medicine and Pathology, Mayo Clinic in Rochester, Rochester, MN, 55905, USA
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic in Rochester, Guggenheim Building 14-11A, 222 3Rd Avenue SW, Rochester, MN, 55905, USA.
| |
Collapse
|
14
|
Al Thani NA, Hasan M, Yalcin HC. Use of Animal Models for Investigating Cardioprotective Roles of SGLT2 Inhibitors. J Cardiovasc Transl Res 2023; 16:975-986. [PMID: 37052784 PMCID: PMC10615955 DOI: 10.1007/s12265-023-10379-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023]
Abstract
Sodium-glucose co-transporter 2 (SGLT2) inhibitors represent one type of new-generation type 2 diabetes (T2DM) drug treatment. The mechanism of action of an SGLT2 inhibitor (SGLT2i) in treating T2DM depends on lowering blood glucose levels effectively via increasing the glomerular excretion of glucose. A good number of randomized clinical trials revealed that SGLT2is significantly prevented heart failure (HF) and cardiovascular death in T2DM patients. Despite ongoing clinical trials in HF patients without T2DM, there have been a limited number of translational studies on the cardioprotective properties of SGLT2is. As the cellular mechanism behind the cardiac benefits of SGLT2is is still to be elucidated, animal models are used to better understand the pathways behind the cardioprotective mechanism of SGLT2i. In this review, we summarize the animal models constructed to study the cardioprotective mechanisms of SGLT2is to help deliver a more comprehensive understanding of the in vivo work that has been done in this field and to help select the most optimal animal model to use when studying the different cardioprotective effects of SGLT2is.
Collapse
Affiliation(s)
- Najlaa A Al Thani
- Research and Development Department, Barzan Holdings, P. O. Box 7178, Doha, Qatar
| | - Maram Hasan
- Biomedical Research Center, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, P. O. Box 2713, Doha, Qatar.
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar.
| |
Collapse
|
15
|
Dai W, Zhang H, Lund H, Zhang Z, Castleberry M, Rodriguez M, Kuriakose G, Gupta S, Lewandowska M, Powers HR, Valmiki S, Zhu J, Shapiro AD, Hussain MM, López JA, Sorci-Thomas MG, Silverstein RL, Ginsberg HN, Sahoo D, Tabas I, Zheng Z. Intracellular tPA-PAI-1 interaction determines VLDL assembly in hepatocytes. Science 2023; 381:eadh5207. [PMID: 37651538 PMCID: PMC10697821 DOI: 10.1126/science.adh5207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/13/2023] [Indexed: 09/02/2023]
Abstract
Apolipoprotein B (apoB)-lipoproteins initiate and promote atherosclerotic cardiovascular disease. Plasma tissue plasminogen activator (tPA) activity is negatively associated with atherogenic apoB-lipoprotein cholesterol levels in humans, but the mechanisms are unknown. We found that tPA, partially through the lysine-binding site on its Kringle 2 domain, binds to the N terminus of apoB, blocking the interaction between apoB and microsomal triglyceride transfer protein (MTP) in hepatocytes, thereby reducing very-low-density lipoprotein (VLDL) assembly and plasma apoB-lipoprotein cholesterol levels. Plasminogen activator inhibitor 1 (PAI-1) sequesters tPA away from apoB and increases VLDL assembly. Humans with PAI-1 deficiency have smaller VLDL particles and lower plasma levels of apoB-lipoprotein cholesterol. These results suggest a mechanism that fine-tunes VLDL assembly by intracellular interactions among tPA, PAI-1, and apoB in hepatocytes.
Collapse
Affiliation(s)
- Wen Dai
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Heng Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Hayley Lund
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ziyu Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | - Maya Rodriguez
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- College of Arts and Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sweta Gupta
- Indiana Hemophilia and Thrombosis Center, Indianapolis, IN 46260, USA
| | | | - Hayley R. Powers
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Swati Valmiki
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amy D. Shapiro
- Indiana Hemophilia and Thrombosis Center, Indianapolis, IN 46260, USA
| | - M. Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | - José A. López
- Bloodworks Research Institute, Seattle, WA 98102, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Mary G. Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Roy L. Silverstein
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Daisy Sahoo
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ze Zheng
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
16
|
Poznyak AV, Sukhorukov VN, Eremin II, Nadelyaeva II, Orekhov AN. Diagnostics of atherosclerosis: Overview of the existing methods. Front Cardiovasc Med 2023; 10:1134097. [PMID: 37229223 PMCID: PMC10203409 DOI: 10.3389/fcvm.2023.1134097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/11/2023] [Indexed: 05/27/2023] Open
Abstract
Atherosclerosis was and remains an extremely common and serious health problem. Since the elderly are most at risk of cardiovascular risk, and the average life expectancy is increasing, the spread of atherosclerosis and its consequences increases as well. One of the features of atherosclerosis is its asymptomaticity. This factor makes it difficult to make a timely diagnosis. This entails the lack of timely treatment and even prevention. To date, in the arsenal of physicians, there is only a limited set of methods to suspect and fully diagnose atherosclerosis. In this review, we have tried to briefly describe the most common and effective methods for diagnosing atherosclerosis.
Collapse
|
17
|
Huang R, Zhang L, Li X, Liu F, Cheng X, Ran H, Wang Z, Li Y, Feng Y, Liang L, Su W, Melgiri ND, Sun Y. Anti-CXCR2 antibody-coated nanoparticles with an erythrocyte-platelet hybrid membrane layer for atherosclerosis therapy. J Control Release 2023; 356:610-622. [PMID: 36898531 DOI: 10.1016/j.jconrel.2023.02.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/20/2023] [Accepted: 02/26/2023] [Indexed: 03/12/2023]
Abstract
Atherosclerosis is the leading cause of mortality globally. RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NPs), which biologically mimic platelets in vivo, display evidence of anti-atherosclerotic activity. The efficacy of a targeted RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NP)-based approach was investigated as a primary preventive measure against atherosclerosis. A ligand-receptor interactome analysis conducted with circulating platelets and monocytes derived from CAD patients and healthy controls identified CXCL8-CXCR2 as a key platelet ligand-monocyte receptor dyad in CAD patients. Based on this analysis, a novel anti-CXCR2 [RBC-P]NP that specifically binds to CXCR2 and blocks the interaction between CXCL8 and CXCR2 was engineered and characterized. Administering anti-CXCR2 [RBC-P]NPs to Western diet-fed Ldlr-/- mice led to diminished plaque size, necrosis, and intraplaque macrophage accumulation relative to control [RBC-P]NPs or vehicle. Importantly, anti-CXCR2 [RBC-P]NPs demonstrated no adverse bleeding/hemorrhagic effects. A series of in vitro experiments was conducted to characterize anti-CXCR2 [RBC-P]NP's mechanism of action in plaque macrophages. Mechanistically, anti-CXCR2 [RBC-P]NPs inhibited p38α (Mapk14)-mediated, pro-inflammatory M1 skewing and corrected efferocytosis in plaque macrophages. This targeted [RBC-P]NP-based approach, in which the cardioprotective effects of anti-CXCR2 [RBC-P]NP therapy overweighs its bleeding/hemorrhagic risks, could potentially be used to proactively manage atherosclerotic progression in at-risk populations.
Collapse
Affiliation(s)
- Rongzhong Huang
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Lujun Zhang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xingsheng Li
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Fan Liu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Xiaoxiao Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Zhigang Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Yongyong Li
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Yuxing Feng
- Department of Rehabilitation and Pain Medicine, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Liwen Liang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Wenhua Su
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, China
| | - N D Melgiri
- Impactys Foundation for Biomedical Research, San Diego, CA, USA
| | - Yang Sun
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China.
| |
Collapse
|
18
|
Ilyas I, Little PJ, Liu Z, Xu Y, Kamato D, Berk BC, Weng J, Xu S. Mouse models of atherosclerosis in translational research. Trends Pharmacol Sci 2022; 43:920-939. [PMID: 35902281 DOI: 10.1016/j.tips.2022.06.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 06/12/2022] [Accepted: 06/17/2022] [Indexed: 12/21/2022]
Abstract
Atherosclerotic cardiovascular disease (CVD), the major cause of premature human mortality, is a chronic and progressive metabolic and inflammatory disease in large- and medium-sized arteries. Mouse models are widely used to gain mechanistic insights into the pathogenesis of atherosclerosis and have facilitated the discovery of anti-atherosclerotic drugs. Despite promising preclinical studies, many drug candidates have not translated to clinical use because of the complexity of disease patho-mechanisms including lipid metabolic traits and inflammatory, genetic, and hemodynamic factors. We review the current preclinical utility and translation potential of traditional [apolipoprotein E (APOE)- and low-density lipoprotein (LDL) receptor (LDLR)-deficient mice] and emerging mouse models that include partial carotid ligation and AAV8-Pcsk9-D377Y injection in atherosclerosis research and drug discovery. This article represents an important resource in atherosclerosis research.
Collapse
Affiliation(s)
- Iqra Ilyas
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Zhiping Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yanyong Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Pathology of School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, Australia
| | - Bradford C Berk
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China; Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China; Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
19
|
Liu W, Östberg NK, Yalcinkaya M, Dou H, Endo-Umeda K, Tang Y, Hou X, Xiao T, Filder T, Abramowicz S, Yang YG, Soehnlein O, Tall AR, Wang N. Erythroid lineage Jak2V617F expression promotes atherosclerosis through erythrophagocytosis and macrophage ferroptosis. J Clin Invest 2022; 132:155724. [PMID: 35587375 PMCID: PMC9246386 DOI: 10.1172/jci155724] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Elevated hematocrit is associated with cardiovascular risk; however, the causality and mechanisms are unclear. The JAK2V617F (Jak2VF) mutation increases cardiovascular risk in myeloproliferative disorders and in clonal hematopoiesis. Jak2VF mice with elevated WBCs, platelets, and RBCs display accelerated atherosclerosis and macrophage erythrophagocytosis. To investigate whether selective erythroid Jak2VF expression promotes atherosclerosis, we developed hyperlipidemic erythropoietin receptor Cre mice that express Jak2VF in the erythroid lineage (VFEpoR mice). VFEpoR mice without elevated blood cell counts showed increased atherosclerotic plaque necrosis, erythrophagocytosis, and ferroptosis. Selective induction of erythrocytosis with low-dose erythropoietin further exacerbated atherosclerosis with prominent ferroptosis, lipid peroxidation, and endothelial damage. VFEpoR RBCs had reduced antioxidant defenses and increased lipid hydroperoxides. Phagocytosis of human or murine WT or JAK2VF RBCs by WT macrophages induced ferroptosis, which was prevented by the ferroptosis inhibitor liproxstatin-1. Liproxstatin-1 reversed increased atherosclerosis, lipid peroxidation, ferroptosis, and endothelial damage in VFEpoR mice and in Jak2VF chimeric mice simulating clonal hematopoiesis, but had no impact in controls. Erythroid lineage Jak2VF expression led to qualitative and quantitative defects in RBCs that exacerbated atherosclerosis. Phagocytosis of RBCs by plaque macrophages promoted ferroptosis, suggesting a therapeutic target for reducing RBC-mediated cardiovascular risk.
Collapse
Affiliation(s)
- Wenli Liu
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Nataliya K Östberg
- Physiology and Pharmacology (FyFA), Karolinska Institute, Stockholm, Sweden
| | - Mustafa Yalcinkaya
- Division of Molecular Medicine, Department of Medicine, Columbia Unicersity, New York, United States of America
| | - Huijuan Dou
- Division of Molecular Medicine, Department of Medicine, Columbia Unicersity, New York, United States of America
| | - Kaori Endo-Umeda
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | - Yang Tang
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Xintong Hou
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Trevor Filder
- Division of Molecular Medicine, Department of Medicine, Columbia Unicersity, New York, United States of America
| | - Sandra Abramowicz
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Yong-Guang Yang
- Institute of Immunology, The First Hospital of Jilin University, Changchun, China
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Munich, Germany
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| |
Collapse
|
20
|
Pig and Mouse Models of Hyperlipidemia and Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:379-411. [PMID: 35237978 DOI: 10.1007/978-1-0716-1924-7_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is a chronic inflammatory disorder that is the underlying cause of most cardiovascular disease. Resident cells of the artery wall and cells of the immune system participate in atherogenesis. This process is influenced by plasma lipoproteins, genetics, and the hemodynamics of the blood flow in the artery. A variety of animal models have been used to study the pathophysiology and mechanisms that contribute to atherosclerotic lesion formation. No model is ideal as each has its own advantages and limitations with respect to manipulation of the atherogenic process and modeling human atherosclerosis and lipoprotein profile. In this chapter we will discuss pig and mouse models of experimental atherosclerosis. The similarity of pig lipoprotein metabolism and the pathophysiology of the lesions in these animals with that of humans is a major advantage. While a few genetically engineered pig models have been generated, the ease of genetic manipulation in mice and the relatively short time frame for the development of atherosclerosis has made them the most extensively used model. Newer approaches to induce hypercholesterolemia in mice have been developed that do not require germline modifications. These approaches will facilitate studies on atherogenic mechanisms.
Collapse
|
21
|
Bjørklund MM, Bernal JA, Bentzon JF. Atherosclerosis Induced by Adeno-Associated Virus Encoding Gain-of-Function PCSK9. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:461-473. [PMID: 35237981 DOI: 10.1007/978-1-0716-1924-7_27] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induction of atherosclerosis in mice with one or more genetic alterations (e.g., conditional deletion of a gene of interest) has traditionally required crossbreeding with Apoe or Ldlr deficient mice to achieve sufficient hypercholesterolemia. However, this procedure is time consuming and generates a surplus of mice with genotypes that are irrelevant for experiments. Several alternative methods exist that obviate the need to work in mice with germline-encoded hypercholesterolemia. In this chapter, we detail an efficient and increasingly used method to induce hypercholesterolemia in mice through adeno-associated virus-mediated transfer of the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene.
Collapse
Affiliation(s)
- Martin Mæng Bjørklund
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jacob F Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark.
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
22
|
Gomes D, Wang S, Goodspeed L, Turk KE, Wietecha T, Liu Y, Bornfeldt KE, O'Brien KD, Chait A, den Hartigh LJ. Comparison between genetic and pharmaceutical disruption of Ldlr expression for the development of atherosclerosis. J Lipid Res 2022; 63:100174. [PMID: 35101425 PMCID: PMC8953673 DOI: 10.1016/j.jlr.2022.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/15/2022] Open
Abstract
Antisense oligonucleotides (ASOs) against Ldl receptor (Ldlr-ASO) represent a promising strategy to promote hypercholesterolemic atherosclerosis in animal models without the need for complex breeding strategies. Here, we sought to characterize and contrast atherosclerosis in mice given Ldlr-ASO with those bearing genetic Ldlr deficiency. To promote atherosclerosis, male and female C57Bl6/J mice were either given weekly injections of Ldlr-ASO (5 mg/kg once per week) or genetically deficient in Ldlr (Ldlr-/-). Mice consumed either standard rodent chow or a diet high in saturated fat and sucrose with 0.15% added cholesterol for 16 weeks. While both models of Ldlr deficiency promoted hypercholesterolemia, Ldlr-/- mice exhibited nearly 2-fold higher cholesterol levels than Ldlr-ASO mice, reflected by increased VLDL and LDL levels. Consistent with this, the en face atherosclerotic lesion area was 3-fold and 3.6-fold greater in male and female mice with genetic Ldlr deficiency, respectively, as compared with the modest atherosclerosis observed following Ldlr-ASO treatment. Aortic sinus lesion sizes, fibrosis, smooth muscle actin, and necrotic core areas were also larger in Ldlr-/- mice, suggesting a more advanced phenotype. Despite a more modest effect on hypercholesterolemia, Ldlr-ASO induced greater hepatic inflammatory gene expression, macrophage accumulation, and histological lobular inflammation than was observed in Ldlr-/- mice. We conclude Ldlr-ASO is a promising tool for the generation of complex rodent models with which to study atherosclerosis but does not promote comparable levels of hypercholesterolemia or atherosclerosis as Ldlr-/- mice and increases hepatic inflammation. Thus, genetic Ldlr deficiency may be a superior model, depending on the proposed use.
Collapse
Affiliation(s)
- Diego Gomes
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Shari Wang
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Leela Goodspeed
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Katherine E Turk
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Tomasz Wietecha
- Diabetes Institute, University of Washington, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Yongjun Liu
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Karin E Bornfeldt
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kevin D O'Brien
- Diabetes Institute, University of Washington, Seattle, WA, USA; Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Alan Chait
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Laura J den Hartigh
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
23
|
Altabas V, Biloš LSK. The Role of Endothelial Progenitor Cells in Atherosclerosis and Impact of Anti-Lipemic Treatments on Endothelial Repair. Int J Mol Sci 2022; 23:ijms23052663. [PMID: 35269807 PMCID: PMC8910333 DOI: 10.3390/ijms23052663] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/16/2022] [Accepted: 02/26/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complications are associated with advanced atherosclerosis. Although atherosclerosis is still regarded as an incurable disease, at least in its more advanced stages, the discovery of endothelial progenitor cells (EPCs), with their ability to replace old and injured cells and differentiate into healthy and functional mature endothelial cells, has shifted our view of atherosclerosis as an incurable disease, and merged traditional theories of atherosclerosis pathogenesis with evolving concepts of vascular biology. EPC alterations are involved in the pathogenesis of vascular abnormalities in atherosclerosis, but many questions remain unanswered. Many currently available drugs that impact cardiovascular morbidity and mortality have shown a positive effect on EPC biology. This review examines the role of endothelial progenitor cells in atherosclerosis development, and the impact standard antilipemic drugs, including statins, fibrates, and ezetimibe, as well as more novel treatments such as proprotein convertase subtilisin/kexin type 9 (PCSK9) modulating agents and angiopoietin-like proteins (Angtpl3) inhibitors have on EPC biology.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
- Correspondence: ; Tel.: +385-1-3787-692
| | | |
Collapse
|
24
|
Hassani KM. Investigation of the Effect of Atorvastatin on Skeletal Muscles in Male Rats and the Involved Mechanisms. ARCHIVES OF RAZI INSTITUTE 2022; 77:285-291. [PMID: 35891762 PMCID: PMC9288636 DOI: 10.22092/ari.2021.356683.1895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/04/2021] [Indexed: 06/15/2023]
Abstract
It has been approved that atrovastatin is a preferred treatment for hyperlipidemia. One of the atrovastatin drawbacks would be the detrimental effects on skeletal muscles. Therefore, the current study was designed to evaluate all the skeletal muscles alteration in rats' after administration of atrovastatin and identification the mechanisms involved in these structural alterations in the skeletal muscles. A total of 12 healthy adult male rats (Rattus norvegicus) were randomly divided into two groups (n=6). The control group (G1) included rats that received distilled water as the placebo, and the treatment group (G2) included animals that were treated with atorvastatin (80 mg/kg/day) dissolved in distilled water and administrated by a gastric tube for eight weeks. At the end of the experiment, trapezius and vastus medialis muscle tissues were sampled and fixed with 10% formalin for histopathological studies. Atorvastatin administration gave rise to morphological changes in the skeletal muscle fibers and the nerve fibers, including atrophied myofibers, infarction, irregular arrangement of myonuclei, disappearance of nuclei from their normal peripheral position with acute skeletal muscular infarction, and infiltration of accumulated inflammatory cells. Atorvastatin has been revealed to have several adverse effects on the skeletal muscle and the nerve supply. Based on the data in the current study, it is evident that atorvastatin administration for less than two months resulted in some sorts of myotoxic structural changes and apoptosis as evident by deformity and lack of striation degeneration of nuclei, as well as splitting of the muscle fibers in the adult male rats' skeletal muscle.
Collapse
Affiliation(s)
- K M Hassani
- College of Science, University of Misan, Maysan, Iraq
| |
Collapse
|
25
|
|
26
|
Tomas L, Prica F, Schulz C. Trafficking of Mononuclear Phagocytes in Healthy Arteries and Atherosclerosis. Front Immunol 2021; 12:718432. [PMID: 34759917 PMCID: PMC8573388 DOI: 10.3389/fimmu.2021.718432] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Monocytes and macrophages play essential roles in all stages of atherosclerosis – from early precursor lesions to advanced stages of the disease. Intima-resident macrophages are among the first cells to be confronted with the influx and retention of apolipoprotein B-containing lipoproteins at the onset of hypercholesterolemia and atherosclerosis development. In this review, we outline the trafficking of monocytes and macrophages in and out of the healthy aorta, as well as the adaptation of their migratory behaviour during hypercholesterolemia. Furthermore, we discuss the functional and ontogenetic composition of the aortic pool of mononuclear phagocytes and its link to the atherosclerotic disease process. The development of mouse models of atherosclerosis regression in recent years, has enabled scientists to investigate the behaviour of monocytes and macrophages during the resolution of atherosclerosis. Herein, we describe the dynamics of these mononuclear phagocytes upon cessation of hypercholesterolemia and how they contribute to the restoration of tissue homeostasis. The aim of this review is to provide an insight into the trafficking, fate and disease-relevant dynamics of monocytes and macrophages during atherosclerosis, and to highlight remaining questions. We focus on the results of rodent studies, as analysis of cellular fates requires experimental manipulations that cannot be performed in humans but point out findings that could be replicated in human tissues. Understanding of the biology of macrophages in atherosclerosis provides an important basis for the development of therapeutic strategies to limit lesion formation and promote plaque regression.
Collapse
Affiliation(s)
- Lukas Tomas
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Filip Prica
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
27
|
Murray JM, Pfeffer P, Seifert R, Hermann A, Handke J, Kummer L, Janssen H, Weigand MA, Schlemmer HP, Larmann J, Kleesiek J. Vesseg: An Open-Source Tool for Deep Learning-Based Atherosclerotic Plaque Quantification in Histopathology Images-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:2516-2522. [PMID: 34380331 DOI: 10.1161/atvbaha.121.316124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: Manual plaque segmentation in microscopy images is a time-consuming process in atherosclerosis research and potentially subject to unacceptable user-to-user variability and observer bias. We address this by releasing Vesseg a tool that includes state-of-the-art deep learning models for atherosclerotic plaque segmentation. Approach and Results: Vesseg is a containerized, extensible, open-source, and user-oriented tool. It includes 2 models, trained and tested on 1089 hematoxylin-eosin stained mouse model atherosclerotic brachiocephalic artery sections. The models were compared to 3 human raters. Vesseg can be accessed at https://vesseg .online or downloaded. The models show mean Soerensen-Dice scores of 0.91+/-0.15 for plaque and 0.97+/-0.08 for lumen pixels. The mean accuracy is 0.98+/-0.05. Vesseg is already in active use, generating time savings of >10 minutes per slide. Conclusions: Vesseg brings state-of-the-art deep learning methods to atherosclerosis research, providing drastic time savings, while allowing for continuous improvement of models and the underlying pipeline.
Collapse
Affiliation(s)
- Jacob M Murray
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.M.M., H.-P.S., J.K.)
- Heidelberg University, Germany (J.M.M.)
- Institute for AI in Medicine (IKIM), University Medicine Essen, Germany (J.M.M., J.K.)
| | - Phillip Pfeffer
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Robert Seifert
- Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, Germany (R.S., J.K.)
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany (R.S.)
- Department of Nuclear Medicine, University Hospital Münster, Germany (R.S.)
| | - Alexander Hermann
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Jessica Handke
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Laura Kummer
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Henrike Janssen
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Markus A Weigand
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Heinz-Peter Schlemmer
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.M.M., H.-P.S., J.K.)
| | - Jan Larmann
- Department of Anesthesiology, University of Heidelberg, Germany (P.P., A.H., J.H., L.K., H.J., M.A.W., J.L.)
| | - Jens Kleesiek
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.M.M., H.-P.S., J.K.)
- Institute for AI in Medicine (IKIM), University Medicine Essen, Germany (J.M.M., J.K.)
- Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, Germany (R.S., J.K.)
| |
Collapse
|
28
|
Opoku E, Traughber CA, Zhang D, Iacano AJ, Khan M, Han J, Smith JD, Gulshan K. Gasdermin D Mediates Inflammation-Induced Defects in Reverse Cholesterol Transport and Promotes Atherosclerosis. Front Cell Dev Biol 2021; 9:715211. [PMID: 34395445 PMCID: PMC8355565 DOI: 10.3389/fcell.2021.715211] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023] Open
Abstract
Activation of inflammasomes, such as Nlrp3 and AIM2, can exacerbate atherosclerosis in mice and humans. Gasdermin D (GsdmD) serves as a final executor of inflammasome activity, by generating membrane pores for the release of mature Interleukin-1beta (IL-1β). Inflammation dampens reverse cholesterol transport (RCT) and promotes atherogenesis, while anti-IL-1β antibodies were shown to reduce cardiovascular disease in humans. Though Nlrp3/AIM2 and IL-1β nexus is an emerging atherogenic pathway, the direct role of GsdmD in atherosclerosis is not yet fully clear. Here, we used in vivo Nlrp3 inflammasome activation to show that the GsdmD-/- mice release ∼80% less IL-1β vs. Wild type (WT) mice. The GsdmD-/- macrophages were more resistant to Nlrp3 inflammasome mediated reduction in cholesterol efflux, showing ∼26% decrease vs. ∼60% reduction in WT macrophages. GsdmD expression in macrophages exacerbated foam cell formation in an IL-1β dependent fashion. The GsdmD-/- mice were resistant to Nlrp3 inflammasome mediated defect in RCT, with ∼32% reduction in plasma RCT vs. ∼57% reduction in WT mice, ∼17% reduction in RCT to liver vs. 42% in WT mice, and ∼37% decrease in RCT to feces vs. ∼61% in WT mice. The LDLr antisense oligonucleotides (ASO) induced hyperlipidemic mouse model showed the role of GsdmD in promoting atherosclerosis. The GsdmD-/- mice exhibit ∼42% decreased atherosclerotic lesion area in females and ∼33% decreased lesion area in males vs. WT mice. The atherosclerotic plaque-bearing sections stained positive for the cleaved N-terminal fragment of GsdmD, indicating cleavage of GsdmD in atherosclerotic plaques. Our data show that GsdmD mediates inflammation-induced defects in RCT and promotes atherosclerosis.
Collapse
Affiliation(s)
- Emmanuel Opoku
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Cynthia Alicia Traughber
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States,Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States,Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - David Zhang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Amanda J. Iacano
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Mariam Khan
- Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States,Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Juying Han
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Jonathan D. Smith
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Kailash Gulshan
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States,Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States,Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States,*Correspondence: Kailash Gulshan, ;
| |
Collapse
|
29
|
Weinstock A, Rahman K, Yaacov O, Nishi H, Menon P, Nikain CA, Garabedian ML, Pena S, Akbar N, Sansbury BE, Heffron SP, Liu J, Marecki G, Fernandez D, Brown EJ, Ruggles KV, Ramsey SA, Giannarelli C, Spite M, Choudhury RP, Loke P, Fisher EA. Wnt signaling enhances macrophage responses to IL-4 and promotes resolution of atherosclerosis. eLife 2021; 10:e67932. [PMID: 33720008 PMCID: PMC7994001 DOI: 10.7554/elife.67932] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a disease of chronic inflammation. We investigated the roles of the cytokines IL-4 and IL-13, the classical activators of STAT6, in the resolution of atherosclerosis inflammation. Using Il4-/-Il13-/- mice, resolution was impaired, and in control mice, in both progressing and resolving plaques, levels of IL-4 were stably low and IL-13 was undetectable. This suggested that IL-4 is required for atherosclerosis resolution, but collaborates with other factors. We had observed increased Wnt signaling in macrophages in resolving plaques, and human genetic data from others showed that a loss-of-function Wnt mutation was associated with premature atherosclerosis. We now find an inverse association between activation of Wnt signaling and disease severity in mice and humans. Wnt enhanced the expression of inflammation resolving factors after treatment with plaque-relevant low concentrations of IL-4. Mechanistically, activation of the Wnt pathway following lipid lowering potentiates IL-4 responsiveness in macrophages via a PGE2/STAT3 axis.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Karishma Rahman
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Or Yaacov
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Hitoo Nishi
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Prashanthi Menon
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Cyrus A Nikain
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Michela L Garabedian
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Stephanie Pena
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Brian E Sansbury
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Sean P Heffron
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
- NYU Center for the Prevention of Cardiovascular Disease, New York University Grossman School of MedicineNew YorkUnited States
| | - Jianhua Liu
- Department of Surgery, Mount Sinai School of MedicineNew YorkUnited States
| | - Gregory Marecki
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Dawn Fernandez
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Emily J Brown
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
| | - Kelly V Ruggles
- Division of Translational Medicine, Department of Medicine, New York University Langone Health, Institute for Systems Genetics, New York University Grossman School of MedicineNew YorkUnited States
| | - Stephen A Ramsey
- Department of Biomedical Sciences, School of Electrical Engineering and Computer Science, Oregon State UniversityCorvallisUnited States
| | - Chiara Giannarelli
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- The Precision Immunology Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Microbiology (Parasitology), New York University School of MedicineNew YorkUnited States
| | - Matthew Spite
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - P'ng Loke
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaUnited States
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Program, New York University Grossman School of MedicineNew YorkUnited States
- NYU Center for the Prevention of Cardiovascular Disease, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Cell Biology and Microbiology, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
30
|
Yu H. Atherosclerotic Plaque Regression: Experimental Approaches and Therapeutic Advances. Trends Cell Biol 2021; 31:424-427. [PMID: 33726967 DOI: 10.1016/j.tcb.2021.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Reversal of atherosclerosis has been well documented in humans on intensive lifestyle changes or lipid-lowering therapies. The development of mouse models has greatly advanced our understanding of molecular mechanisms underlying this biological process. I seek to summarize the established mouse models and highlight the recent therapeutic progress on plaque regression.
Collapse
Affiliation(s)
- Haojie Yu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
31
|
Zhang K, Zheng J, Chen Y, Dong J, Li Z, Chiang YP, He M, Huang Q, Tang H, Jiang XC. Inducible phospholipid transfer protein deficiency ameliorates atherosclerosis. Atherosclerosis 2021; 324:9-17. [PMID: 33798923 DOI: 10.1016/j.atherosclerosis.2021.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis progression and regression studies are related to its prevention and treatment. Although we have gained extensive knowledge on germline phospholipid transfer protein (PLTP) deficiency, the effect of inducible PLTP deficiency in atherosclerosis remains unexplored. METHODS We generated inducible PLTP (iPLTP)-knockout (KO) mice and measured their plasma lipid levels after feeding a normal chow or a Western-type diet. Adenovirus associated virus-proprotein convertase subtilisin/kexin type 9 (AAV-PCSK9) was used to induce hypercholesterolemia in the mice. Collars were placed around the common carotid arteries, and atherosclerosis progression and regression in the carotid arteries and aortic roots were evaluated. RESULTS On a normal chow diet, iPLTP-KO mice exhibited decreased cholesterol, phospholipid, apoA-I, and apoB levels compared with control mice. Furthermore, the overall amount of high-density lipoprotein (HDL) particles was reduced in these mice, but this effect was more profound for larger HDL particles. On a Western-type diet, iPLTP-KO mice again exhibited reduced levels of all tested lipids, even though the basal lipid levels were increased. Additionally, these mice displayed significantly reduced atherosclerotic plaque sizes with increased plaque stability. Importantly, inducible PLTP deficiency significantly ameliorated atherosclerosis by reducing the size of established plaques and the number of macrophages in the plaques without causing lipid accumulation in the liver. CONCLUSIONS Induced PLTP deficiency in adult mice reduces plasma total cholesterol and triglycerides, prevents atherosclerosis progression, and promotes atherosclerosis regression. Thus, PLTP inhibition is a promising therapeutic approach for atherosclerosis.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiao Zheng
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Beijing University of Chinese Medicine, Beijing, China
| | | | | | - Zhiqiang Li
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA
| | - Yeun-Po Chiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Mulin He
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | | | | | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA.
| |
Collapse
|
32
|
Josefs T, Basu D, Vaisar T, Arets B, Kanter JE, Huggins LA, Hu Y, Liu J, Clouet-Foraison N, Heinecke JW, Bornfeldt KE, Goldberg IJ, Fisher EA. Atherosclerosis Regression and Cholesterol Efflux in Hypertriglyceridemic Mice. Circ Res 2021; 128:690-705. [PMID: 33530703 DOI: 10.1161/circresaha.120.317458] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Tatjana Josefs
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine.,Department of Internal Medicine, MUMC, Maastricht, the Netherlands (T.J., B.A.).,CARIM, MUMC, Maastricht, the Netherlands (T.J., B.A.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine.,Department of Internal Medicine, MUMC, Maastricht, the Netherlands (T.J., B.A.).,CARIM, MUMC, Maastricht, the Netherlands (T.J., B.A.)
| | - Tomas Vaisar
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | | | - Jenny E Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Jianhua Liu
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine
| | - Noemie Clouet-Foraison
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Jay W Heinecke
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Karin E Bornfeldt
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Edward A Fisher
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine
| |
Collapse
|
33
|
Choi SH, Agatisa-Boyle C, Gonen A, Kim A, Kim J, Alekseeva E, Tsimikas S, Miller YI. Intracellular AIBP (Apolipoprotein A-I Binding Protein) Regulates Oxidized LDL (Low-Density Lipoprotein)-Induced Mitophagy in Macrophages. Arterioscler Thromb Vasc Biol 2021; 41:e82-e96. [PMID: 33356389 PMCID: PMC8105271 DOI: 10.1161/atvbaha.120.315485] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Atherosclerotic lesions are often characterized by accumulation of OxLDL (oxidized low-density lipoprotein), which is associated with vascular inflammation and lesion vulnerability to rupture. Extracellular AIBP (apolipoprotein A-I binding protein; encoded by APOA1BP gene), when secreted, promotes cholesterol efflux and regulates lipid rafts dynamics, but its role as an intracellular protein in mammalian cells remains unknown. The aim of this work was to determine the function of intracellular AIBP in macrophages exposed to OxLDL and in atherosclerotic lesions. Approach and Results: Using a novel monoclonal antibody against human and mouse AIBP, which are highly homologous, we demonstrated robust AIBP expression in human and mouse atherosclerotic lesions. We observed significantly reduced autophagy in bone marrow-derived macrophages, isolated from Apoa1bp-/- compared with wild-type mice, which were exposed to OxLDL. In atherosclerotic lesions from Apoa1bp-/- mice subjected to Ldlr knockdown and fed a Western diet, autophagy was reduced, whereas apoptosis was increased, when compared with that in wild-type mice. AIBP expression was necessary for efficient control of reactive oxygen species and cell death and for mitochondria quality control in macrophages exposed to OxLDL. Mitochondria-localized AIBP, via its N-terminal domain, associated with E3 ubiquitin-protein ligase PARK2 (Parkin), MFN (mitofusin)1, and MFN2, but not BNIP3 (Bcl2/adenovirus E1B 19-kDa-interacting protein-3), and regulated ubiquitination of MFN1 and MFN2, key components of mitophagy. CONCLUSIONS These data suggest that intracellular AIBP is a new regulator of autophagy in macrophages. Mitochondria-localized AIBP augments mitophagy and participates in mitochondria quality control, protecting macrophages against cell death in the context of atherosclerosis.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Colin Agatisa-Boyle
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Ayelet Gonen
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Alisa Kim
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Jungsu Kim
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Elena Alekseeva
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Sotirios Tsimikas
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Yury I. Miller
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
34
|
Li Z, Zhao P, Zhang Y, Wang J, Wang C, Liu Y, Yang G, Yuan L. Exosome-based Ldlr gene therapy for familial hypercholesterolemia in a mouse model. Am J Cancer Res 2021; 11:2953-2965. [PMID: 33456582 PMCID: PMC7806494 DOI: 10.7150/thno.49874] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
Familial hypercholesterolemia (FH), with high LDL (low-density lipoprotein) cholesterol levels, is due to inherited mutations in genes, such as low-density lipoprotein receptor (LDLR). Development of therapeutic strategies for FH, which causes atherosclerosis and cardiovascular disease, is urgently needed. Methods: Mice with low-density lipoprotein receptor (Ldlr) deletion (Ldlr-/- mice) were used as an FH model. Ldlr mRNA was encapsulated into exosomes by forced expression of Ldlr in the donor AML12 (alpha mouse liver) cells, and the resultant exosomes were denoted as ExoLdlr. In vivo distribution of exosomes was analyzed by fluorescence labeling and imaging. The delivery efficiency of Ldlr mRNA was analyzed by qPCR and Western blotting. Therapeutic effects of ExoLdlr were examined in Ldlr-/- mice by blood lipids and Oil Red O staining. Results: The encapsulated mRNA was stable and could be translated into functional protein in the recipient cells. Following tail vein injection, exosomes were mainly delivered into the liver, producing abundant LDLR protein, resembling the endogenous expression profile in the wild-type mouse. Compared with control exosomes, ExoLdlr treatment significantly decreased lipid deposition in the liver and lowered the serum LDL-cholesterol level. Significantly, the number and size of atherosclerotic plaques and inflammation were reduced in the ExoLdlr-treated mice. Conclusions: We have shown that exosome-mediated Ldlr mRNA delivery effectively restored receptor expression, treating the disorders in the Ldlr-/- mouse. Our study provided a new therapeutic approach for the treatment of FH patients and managing atherosclerosis.
Collapse
|
35
|
Abstract
The development of potent cholesterol-reducing medications in the last decade of the twentieth century has altered the approach to prevention and treatment of cardiovascular disease (CVD). Initial experience with statins, and more recently with the addition of PCSK9 inhibitors, has proven that human CVD, like that in animal models, can be halted and regressed. Available clinical data show that the lower the achieved level of low-density lipoprotein cholesterol, the greater the regression of disease. Investigative studies are now aimed to understand those factors that both accelerate and impede this healing process. Some of these are likely to be modifiable, and the future of atherosclerotic CVD treatment is likely to be early screening, use of measures to repair atherosclerotic arteries, and prevention of most CVD events.
Collapse
Affiliation(s)
- Ira J Goldberg
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA;
| | - Gaurav Sharma
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA;
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA;
| |
Collapse
|
36
|
Silva JL, Leocádio PCL, Reis JM, Campos GP, Capettini LSA, Foureaux G, Ferreira AJ, Windmöller CC, Santos FA, Oriá RB, Crespo-López ME, Alvarez-Leite JI. Oral methylmercury intoxication aggravates cardiovascular risk factors and accelerates atherosclerosis lesion development in ApoE knockout and C57BL/6 mice. Toxicol Res 2020; 37:311-321. [PMID: 34295795 DOI: 10.1007/s43188-020-00066-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/09/2020] [Accepted: 09/23/2020] [Indexed: 01/08/2023] Open
Abstract
Methylmercury (MeHg) intoxication is associated with hypertension, hypercholesterolemia, and atherosclerosis by mechanisms that are not yet fully understood. We investigated the effects of MeHg intoxication in atherosclerosis-prone (ApoE-KO) and resistant C57BL/6 mice. Mice were submitted to carotid stenosis surgery (to induce atherosclerosis faster) and received water or MeHg solution (20 mg/L) for 15 days. Tail plethysmography was performed before and after MeHg exposure. Food and MeHg solution intakes were monitored weekly. On the 15th day, mice were submitted to intravital fluorescence microscopy of mesenteric vasculature to observe in vivo leukocyte rolling and adhesion. Results showed that despite the high hair and liver Hg concentrations in the MeHg group, food and water (or MeHg solution) consumption and liver function marker levels were similar to those in controls. MeHg exposure increased total cholesterol, the atherogenic (non-HDL) fraction and systolic and diastolic blood pressure. MeHg exposure also induced inflammation, as seen by the increased rolling and adhered leukocytes in the mesenteric vasculature. Atherosclerosis lesions were more extensive in the aorta and carotid sites of MeHg-ApoE knockout mice. Surprisingly, MeHg exposure also induced atherosclerosis lesions in C57BL/6 mice, which are resistant to atherosclerosis formation. We concluded that MeHg intoxication might represent a risk for cardiovascular diseases since it accelerates atherogenesis by exacerbating several independent risk factors.
Collapse
Affiliation(s)
- Janayne L Silva
- Departamento de Bioquímica e Imunologia ICB/UFMG Caixa Postal 486, Belo Horizonte, MG CEP 30161-970 Brazil
| | - Paola C L Leocádio
- Departamento de Nutrição e Saúde, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Jonas M Reis
- Departamento de Bioquímica e Imunologia ICB/UFMG Caixa Postal 486, Belo Horizonte, MG CEP 30161-970 Brazil
| | - Gianne P Campos
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Luciano S A Capettini
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Giselle Foureaux
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Anderson J Ferreira
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Cláudia C Windmöller
- Departamento de Química, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Flávia A Santos
- Departamento de Morfologia, Universidade Federal Do Ceará, Fortaleza, Ceará Brazil
| | - Reinaldo B Oriá
- Departamento de Morfologia, Universidade Federal Do Ceará, Fortaleza, Ceará Brazil
| | - Maria E Crespo-López
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará Brazil
| | - Jacqueline I Alvarez-Leite
- Departamento de Bioquímica e Imunologia ICB/UFMG Caixa Postal 486, Belo Horizonte, MG CEP 30161-970 Brazil
| |
Collapse
|
37
|
Barrett TJ, Schlegel M, Zhou F, Gorenchtein M, Bolstorff J, Moore KJ, Fisher EA, Berger JS. Platelet regulation of myeloid suppressor of cytokine signaling 3 accelerates atherosclerosis. Sci Transl Med 2020; 11:11/517/eaax0481. [PMID: 31694925 DOI: 10.1126/scitranslmed.aax0481] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/19/2019] [Accepted: 10/04/2019] [Indexed: 12/19/2022]
Abstract
Platelets are best known as mediators of hemostasis and thrombosis; however, their inflammatory effector properties are increasingly recognized. Atherosclerosis, a chronic vascular inflammatory disease, represents the interplay between lipid deposition in the artery wall and unresolved inflammation. Here, we reveal that platelets induce monocyte migration and recruitment into atherosclerotic plaques, resulting in plaque platelet-macrophage aggregates. In Ldlr -/- mice fed a Western diet, platelet depletion decreased plaque size and necrotic area and attenuated macrophage accumulation. Platelets drive atherogenesis by skewing plaque macrophages to an inflammatory phenotype, increasing myeloid suppressor of cytokine signaling 3 (SOCS3) expression and reducing the Socs1:Socs3 ratio. Platelet-induced Socs3 expression regulates plaque macrophage reprogramming by promoting inflammatory cytokine production (Il6, Il1b, and Tnfa) and impairing phagocytic capacity, dysfunctions that contribute to unresolved inflammation and sustained plaque growth. Translating our data to humans with cardiovascular disease, we found that women with, versus without, myocardial infarction have up-regulation of SOCS3, lower SOCS1:SOCS3, and increased monocyte-platelet aggregate. A second cohort of patients with lower extremity atherosclerosis demonstrated that SOCS3 and the SOCS1:SOCS3 ratio correlated with platelet activity and inflammation. Collectively, these data provide a causative link between platelet-mediated myeloid inflammation and dysfunction, SOCS3, and cardiovascular disease. Our findings define an atherogenic role of platelets and highlight how, in the absence of thrombosis, platelets contribute to inflammation.
Collapse
Affiliation(s)
- Tessa J Barrett
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Martin Schlegel
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Felix Zhou
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Mike Gorenchtein
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Jennifer Bolstorff
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Kathryn J Moore
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Edward A Fisher
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Jeffrey S Berger
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
38
|
Kim K, Yu J, Kang JK, Morrow JP, Pajvani UB. Liver-selective γ-secretase inhibition ameliorates diet-induced hepatic steatosis, dyslipidemia and atherosclerosis. Biochem Biophys Res Commun 2020; 527:979-984. [PMID: 32439159 DOI: 10.1016/j.bbrc.2020.04.157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 01/05/2023]
Abstract
Hepatic γ-secretase regulates low-density lipoprotein receptor (LDLR) cleavage and degradation, affecting clearance of plasma triglyceride (TG)-rich lipoproteins (TRLs). In this study, we investigated whether γ-secretase inhibition modulates risk of Western (high-fat/sucrose and high-cholesterol)-type diet (WTD)-induced hepatic steatosis, dyslipidemia and atherosclerosis. We evaluated liver and plasma lipids in WTD-fed mice with hepatocyte-specific ablation of the non-redundant γ-secretase-targeting subunit Nicastrin (L-Ncst). In parallel, we investigated the effect of liver-selective Ncst antisense oligonucleotides (ASO) on lipid metabolism and atherosclerosis in wildtype (WT) and ApoE knockout (ApoE-/-) mice fed normal chow or WTD. WTD-fed L-Ncst and Ncst ASO-treated WT mice showed reduced total cholesterol and LDL-cholesterol (LDL-C), as well as reduced hepatic lipid content as compared to Cre- and control ASO-treated WT mice. Treatment of WTD-fed ApoE-/- mice with Ncst ASO markedly lowered total and LDL cholesterol, hepatic TG and attenuated atherosclerotic lesions in the aorta, as compared to control ASO-treated mice. L-Ncst and Ncst ASO similarly showed reduced plasma glucose as compared to control mice. In conclusion, inhibition of hepatic γ-secretase reduces plasma glucose, and attenuates WTD-induced dyslipidemia, hepatic fat accumulation and atherosclerosis, suggesting potential pleiotropic application for diet-induced metabolic dysfunction.
Collapse
Affiliation(s)
- KyeongJin Kim
- Department of Medicine, Columbia University, New York, NY, 10032, USA; Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, 22212, South Korea.
| | - Junjie Yu
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Jin Ku Kang
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - John P Morrow
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
39
|
Sari G, Meester EJ, van der Zee LC, Wouters K, van Lennep JR, Peppelenbosch M, Boonstra A, Van der Heiden K, Mulder MMT, Vanwolleghem T. A mouse model of humanized liver shows a human-like lipid profile, but does not form atherosclerotic plaque after western type diet. Biochem Biophys Res Commun 2020; 524:510-515. [PMID: 32014257 DOI: 10.1016/j.bbrc.2020.01.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/11/2020] [Indexed: 01/12/2023]
Abstract
Mouse models are a crucial and often used tool to provide insight into the underlying mechanisms of human atherosclerosis. However, mice profoundly differ from humans in lipoprotein synthesis and metabolism, key factors in atherosclerotic plaque formation. Mouse models often require genetic and dietary modifications to mimic human pathophysiology, shifting from a high-density lipoprotein to an low-density lipoprotein dominant lipoprotein profile. We examined the suitability of mice with a humanized liver as a model for lipoprotein studies and studies on plaque formation, given the central role of hepatocytes in lipoprotein synthesis and metabolism. Our results show a progressive humanization of the mouse liver and a humanized lipoprotein profile. However, no atherosclerotic plaque formation was observed in the studied time frame, despite presence of functional macrophages and application of a high cholesterol western-type diet. The humanized-liver mouse model therefore might require further modifications to induce atherosclerosis, yet seems a valuable model for in vivo studies on lipoprotein metabolism.
Collapse
Affiliation(s)
- Gulce Sari
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Eric J Meester
- Department of Biomedical Engineering, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Leonie C van der Zee
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, MUMC+, Maastricht, the Netherlands
| | - Jeanine R van Lennep
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maikel Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kim Van der Heiden
- Department of Biomedical Engineering, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Monique M T Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thomas Vanwolleghem
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands; Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp and Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.
| |
Collapse
|
40
|
Pennig J, Scherrer P, Gissler MC, Anto-Michel N, Hoppe N, Füner L, Härdtner C, Stachon P, Wolf D, Hilgendorf I, Mullick A, Bode C, Zirlik A, Goldberg IJ, Willecke F. Glucose lowering by SGLT2-inhibitor empagliflozin accelerates atherosclerosis regression in hyperglycemic STZ-diabetic mice. Sci Rep 2019; 9:17937. [PMID: 31784656 PMCID: PMC6884628 DOI: 10.1038/s41598-019-54224-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/26/2019] [Indexed: 12/12/2022] Open
Abstract
Diabetes worsens atherosclerosis progression and leads to a defect in repair of arteries after cholesterol reduction, a process termed regression. Empagliflozin reduces blood glucose levels via inhibition of the sodium glucose cotransporter 2 (SGLT-2) in the kidney and has been shown to lead to a marked reduction in cardiovascular events in humans. To determine whether glucose lowering by empagliflozin accelerates atherosclerosis regression in a mouse model, male C57BL/6J mice were treated intraperitoneally with LDLR- and SRB1- antisense oligonucleotides and fed a high cholesterol diet for 16 weeks to induce severe hypercholesterolemia and atherosclerosis progression. At week 14 all mice were rendered diabetic by streptozotocin (STZ) injections. At week 16 a baseline group was sacrificed and displayed substantial atherosclerosis of the aortic root. In the remaining mice, plasma cholesterol was lowered by switching to chow diet and treatment with LDLR sense oligonucleotides to induce atherosclerosis regression. These mice then received either empagliflozin or vehicle for three weeks. Atherosclerotic plaques in the empagliflozin treated mice were significantly smaller, showed decreased lipid and CD68+ macrophage content, as well as greater collagen content. Proliferation of plaque resident macrophages and leukocyte adhesion to the vascular wall were significantly decreased in empagliflozin-treated mice. In summary, plasma glucose lowering by empagliflozin improves plaque regression in diabetic mice.
Collapse
Affiliation(s)
- Jan Pennig
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Philipp Scherrer
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Mark Colin Gissler
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Nathaly Anto-Michel
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Natalie Hoppe
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Lisa Füner
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Carmen Härdtner
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Adam Mullick
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Christoph Bode
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Andreas Zirlik
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany.,Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Ira J Goldberg
- Department of Medicine, New York University Langone Health, New York, NY, USA
| | - Florian Willecke
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany. .,Klinik für Allgemeine und Interventionelle Kardiologie/Angiologie, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bochum, Germany.
| |
Collapse
|
41
|
Abstract
Macrophages play a central role in the development of atherosclerotic cardiovascular disease (ASCVD), which encompasses coronary artery disease, peripheral artery disease, cerebrovascular disease, and aortic atherosclerosis. In each vascular bed, macrophages contribute to the maintenance of the local inflammatory response, propagate plaque development, and promote thrombosis. These central roles, coupled with their plasticity, makes macrophages attractive therapeutic targets in stemming the development of and stabilizing existing atherosclerosis. In the context of ASCVD, classically activated M1 macrophages initiate and sustain inflammation, and alternatively activated M2 macrophages resolve inflammation. However, this classification is now considered an oversimplification, and a greater understanding of plaque macrophage physiology in ASCVD is required to aid in the development of therapeutics to promote ASCVD regression. Reviewed herein are the macrophage phenotypes and molecular regulators characteristic of ASCVD regression, and the current murine models of ASCVD regression.
Collapse
Affiliation(s)
- Tessa J. Barrett
- From the Division of Cardiology, Department of Medicine, New York University
| |
Collapse
|
42
|
Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J, Moore KJ, Fisher EA. Single-Cell RNA Sequencing of Visceral Adipose Tissue Leukocytes Reveals that Caloric Restriction Following Obesity Promotes the Accumulation of a Distinct Macrophage Population with Features of Phagocytic Cells. IMMUNOMETABOLISM 2019; 1:e190008. [PMID: 31396408 PMCID: PMC6687332 DOI: 10.20900/immunometab20190008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity can lead to type 2 diabetes and is an epidemic. A major contributor to its adverse effects is inflammation of the visceral adipose tissue (VAT). Life-long caloric restriction (CR), in contrast, results in extended lifespan, enhanced glucose tolerance/insulin sensitivity, and other favorable phenotypes. The effects of CR following obesity are incompletely established, but studies show multiple benefits. Many leukocyte types, macrophages predominantly, reside in VAT in homeostatic and pathological states. CR following obesity transiently increases VAT macrophage content prior to resolution of inflammation and obesity, suggesting that macrophage content and phenotype play critical roles. Here, we examined the heterogeneity of VAT leukocytes and the effects of obesity and CR. In general, our single-cell RNA-sequencing data demonstrate that macrophages are the most abundant and diverse subpopulation of leukocytes in VAT. Obesity induced significant transcriptional changes in all 15 leukocyte subpopulations, with many genes showing coordinated changes in expression across the leukocyte subpopulations. Additionally, obese VAT displayed expansion of one major macrophage subpopulation, which, in silico, was enriched in lipid binding and metabolic processes. This subpopulation returned from dominance in obesity to lean proportions after only 2 weeks of CR, although the pattern of gene expression overall remained similar. Surprisingly, CR VAT is dominated by a different macrophage subpopulation, which is absent in lean conditions. This subpopulation is enriched in genes related to phagocytosis and we postulate that its function includes clearance of dead cells, as well as excess lipids, contributing to limiting VAT inflammation and restoration of the homeostatic state.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Emily J Brown
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Michela L Garabedian
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Stephanie Pena
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Monika Sharma
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Juan Lafaille
- Department of Microbiology and Immunology, NYU School of Medicine, New York, NY 10016, USA
| | - Kathryn J Moore
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
- Department of Microbiology and Immunology, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
43
|
Hussain MM, Goldberg IJ. Human MicroRNA-33b Promotes Atherosclerosis in Apoe -/- Mice. Arterioscler Thromb Vasc Biol 2019; 38:2272-2275. [PMID: 30354227 DOI: 10.1161/atvbaha.118.311617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- M Mahmood Hussain
- From the Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, NY; and Division of Endocrinology, Diabetes, and Metabolism, NYU School of Medicine, NYU Langone Health, New York
| | - Ira J Goldberg
- From the Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, NY; and Division of Endocrinology, Diabetes, and Metabolism, NYU School of Medicine, NYU Langone Health, New York
| |
Collapse
|
44
|
Williams R. Circulation Research "In This Issue" Anthology. Circ Res 2019; 124:e123-e148. [PMID: 31170049 DOI: 10.1161/res.0000000000000275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Burke AC, Sutherland BG, Telford DE, Morrow MR, Sawyez CG, Edwards JY, Huff MW. Naringenin enhances the regression of atherosclerosis induced by a chow diet in Ldlr -/- mice. Atherosclerosis 2019; 286:60-70. [PMID: 31102954 DOI: 10.1016/j.atherosclerosis.2019.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/31/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Naringenin is a citrus-derived flavonoid with lipid-lowering and insulin-sensitizing effects leading to athero-protection in Ldlr-/- mice fed a high-fat diet. However, the ability of naringenin to promote atherosclerosis regression is unknown. In the present study, we assessed the capacity of naringenin to enhance regression in Ldlr-/- mice with diet-induced intermediate atherosclerosis intervened with a chow diet. METHODS Male Ldlr-/- mice were fed a high-fat, cholesterol-containing (HFHC) diet for 12 weeks to induce intermediate atherosclerosis and metabolic dysfunction. Subsequently, a group of these mice were sacrificed for baseline analyses and the remainder either 1) continued on the HFHC diet, 2) switched to a chow diet or 3) switched to chow diet supplemented with naringenin. RESULTS After 12 weeks induction, intermediate lesions developed in the aortic sinus. Intervention with chow alone slowed lesion growth, while intervention with naringenin-supplemented chow completely halted lesion growth. Lesions were characterized by features of improved morphology. Compared to chow alone, naringenin reduced plaque macrophages and modestly increased smooth muscle cells. Investigating processes that contributed to improved plaque morphology, we showed naringenin further reduced plasma triglycerides and cholesterol compared to chow alone. Furthermore, elevated monocytosis and myelopoiesis were further corrected by intervention with naringenin compared to chow alone. Metabolically, naringenin enhanced the correction of insulin resistance, hepatic steatosis and obesity compared to chow alone, potentially contributing to enhanced regression. CONCLUSIONS Naringenin supplementation to chow enhances atherosclerosis regression in male Ldlr-/- mice. These studies further underscore the potential therapeutic utility of naringenin.
Collapse
Affiliation(s)
- Amy C Burke
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada; Department of Biochemistry, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada
| | - Brian G Sutherland
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada
| | - Dawn E Telford
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada; Department of Medicine, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada
| | - Marisa R Morrow
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada
| | - Cynthia G Sawyez
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada; Department of Medicine, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada
| | - Jane Y Edwards
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada; Department of Medicine, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada
| | - Murray W Huff
- Molecular Medicine, Robarts Research Institute, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada; Department of Biochemistry, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada; Department of Medicine, The University of Western Ontario, 1151 Richmond St N., London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
46
|
Weinstock A, Fisher EA. Methods to Study Monocyte and Macrophage Trafficking in Atherosclerosis Progression and Resolution. Methods Mol Biol 2019; 1951:153-165. [PMID: 30825151 DOI: 10.1007/978-1-4939-9130-3_12] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Monocytes are circulating cells imperative to the response against pathogens. Upon infection, they are quickly recruited to the affected tissue where they can differentiate into specialized phagocytes and antigen-presenting cells. Additionally, monocytes play a vital role in chronic inflammation, where they can promote and enhance inflammation or induce its resolution. There are two major subsets of monocytes, "inflammatory" and "nonclassical," which are believed to have distinct functions. In atherosclerosis, both types of monocytes are constantly recruited to lesions, where they contribute to plaque formation and atherosclerosis progression. Surprisingly, these cells can also be recruited to lesions and promote resolution of atherosclerosis. Tracking these cells in various disease stages may inform about the dynamic changes occurring in the inflamed and resolving tissues. In this chapter we will discuss methods for differential labeling of the two monocyte subsets in order to examine their dynamics in inflammation.
Collapse
Affiliation(s)
- Ada Weinstock
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA
| | - Edward A Fisher
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
47
|
Gao W, Li X, Liu Z, Fu W, Sun Y, Cao W, Tong L, Tang B. A Redox-Responsive Self-Assembled Nanoprobe for Photoacoustic Inflammation Imaging to Assess Atherosclerotic Plaque Vulnerability. Anal Chem 2018; 91:1150-1156. [PMID: 30497260 DOI: 10.1021/acs.analchem.8b04912] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inflammation triggered by oxidative stress is the main determinant of atherosclerotic plaque disruption, which is the leading cause of myocardial infarctions and strokes. Hence, noninvasive mapping of alterations in redox status in vivo is highly desirable for accurate assessment of plaque inflammatory activity and vulnerability. Herein, two types of near-infrared fluorescence probes, specific for glutathione (GSH)/hydrogen peroxide (H2O2) redox couple, were used to introduce the self-assembly of bovine serum albumin (BSA), forming a BSA-Cy-Mito nanoprobe for in vivo photoacoustic imaging of redox status. Such BSA-based self-assemblies on one hand processed good biocompatibility and long blood circulation for high EPR effect and plaque accumulation and on the other hand displayed strong GSH- and H2O2-dependent absorbance at 765 and 680 nm, which enabled simultaneous photoacoustic detection of GSH/H2O2 with high specificity and sensitivity. Using BSA-Cy-Mito as an in vivo GSH/H2O2 indicator, accurate detection of the redox-related inflammatory process was realized both in oxidized low-density lipoprotein (ox-LDL)-activated macrophages and high fat diet-fed apolipoprotein E-deficient (ApoE-/-) mice. Systemic administration of BSA-Cy-Mito further enabled differentiation of vulnerable plaques from stable ones based on their different redox states. Therefore, this sensitive redox-responsive PA nanoprobe may be a powerful tool for early identification of rupture-prone plaques and help in implementing successful preventative therapeutic strategies.
Collapse
Affiliation(s)
- Wen Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| | - Xiang Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| | - Zhenhua Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| | - Wei Fu
- Department of Pharmacy , ZiBo Central Hospital , Zibo 255000 , P. R. China
| | - Yuhui Sun
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| | - Wenhua Cao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| | - Lili Tong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences , Shandong Normal University , Jinan 250014 , P. R. China
| |
Collapse
|
48
|
Cervadoro A, Palomba R, Vergaro G, Cecchi R, Menichetti L, Decuzzi P, Emdin M, Luin S. Targeting Inflammation With Nanosized Drug Delivery Platforms in Cardiovascular Diseases: Immune Cell Modulation in Atherosclerosis. Front Bioeng Biotechnol 2018; 6:177. [PMID: 30542650 PMCID: PMC6277804 DOI: 10.3389/fbioe.2018.00177] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis (AS) is a disorder of large and medium-sized arteries; it consists in the formation of lipid-rich plaques in the intima and inner media, whose pathophysiology is mostly driven by inflammation. Currently available interventions and therapies for treating atherosclerosis are not always completely effective; side effects associated with treatments, mainly caused by immunodepression for anti-inflammatory molecules, limit the systemic administration of these and other drugs. Given the high degree of freedom in the design of nanoconstructs, in the last decades researchers have put high effort in the development of nanoparticles (NPs) formulations specifically designed for either drug delivery, visualization of atherosclerotic plaques, or possibly the combination of both these and other functionalities. Here we will present the state of the art of these subjects, the knowledge of which is necessary to rationally address the use of NPs for prevention, diagnosis, and/or treatment of AS. We will analyse the work that has been done on: (a) understanding the role of the immune system and inflammation in cardiovascular diseases, (b) the pathological and biochemical principles in atherosclerotic plaque formation, (c) the latest advances in the use of NPs for the recognition and treatment of cardiovascular diseases, (d) the cellular and animal models useful to study the interactions of NPs with the immune system cells.
Collapse
Affiliation(s)
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Giuseppe Vergaro
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Roberta Cecchi
- NEST Laboratory, Scuola Normale Superiore, Pisa, Italy.,Center for Nanotechnology Innovation (CNI@NEST), Istituto Italiano di Tecnologia, Pisa, Italy
| | | | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Michele Emdin
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Pisa, Italy.,NEST Laboratory, Istituto Nanoscienze, CNR, Pisa, Italy
| |
Collapse
|
49
|
Jarrett KE, Lee C, De Giorgi M, Hurley A, Gillard BK, Doerfler AM, Li A, Pownall HJ, Bao G, Lagor WR. Somatic Editing of Ldlr With Adeno-Associated Viral-CRISPR Is an Efficient Tool for Atherosclerosis Research. Arterioscler Thromb Vasc Biol 2018; 38:1997-2006. [PMID: 30026278 PMCID: PMC6202188 DOI: 10.1161/atvbaha.118.311221] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/27/2018] [Indexed: 01/23/2023]
Abstract
Objective- Atherosclerosis studies in Ldlr knockout mice require breeding to homozygosity and congenic status on C57BL6/J background, a process that is both time and resource intensive. We aimed to develop a new method for generating atherosclerosis through somatic deletion of Ldlr in livers of adult mice. Approach and Results- Overexpression of PCSK9 (proprotein convertase subtilisin/kexin type 9) is currently used to study atherosclerosis, which promotes degradation of LDLR (low-density lipoprotein receptor) in the liver. We sought to determine whether CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats-associated 9) could also be used to generate atherosclerosis through genetic disruption of Ldlr in adult mice. We engineered adeno-associated viral (AAV) vectors expressing Staphylococcus aureus Cas9 and a guide RNA targeting the Ldlr gene (AAV-CRISPR). Both male and female mice received either (1) saline, (2) AAV-CRISPR, or (3) AAV-hPCSK9 (human PCSK9)-D374Y. A fourth group of germline Ldlr-KO mice was included for comparison. Mice were placed on a Western diet and followed for 20 weeks to assess plasma lipids, PCSK9 protein levels, atherosclerosis, and editing efficiency. Disruption of Ldlr with AAV-CRISPR was robust, resulting in severe hypercholesterolemia and atherosclerotic lesions in the aorta. AAV-hPCSK9 also produced hypercholesterolemia and atherosclerosis as expected. Notable sexual dimorphism was observed, wherein AAV-CRISPR was superior for Ldlr removal in male mice, while AAV-hPCSK9 was more effective in female mice. Conclusions- This all-in-one AAV-CRISPR vector targeting Ldlr is an effective and versatile tool to model atherosclerosis with a single injection and provides a useful alternative to the use of germline Ldlr-KO mice.
Collapse
Affiliation(s)
- Kelsey E. Jarrett
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ciaran Lee
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ayrea Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Baiba K. Gillard
- Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medicine, New York, NY 10065, USA
| | - Alexandria M. Doerfler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ang Li
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Henry J. Pownall
- Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medicine, New York, NY 10065, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - William R. Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
50
|
Burke AC, Sutherland BG, Telford DE, Morrow MR, Sawyez CG, Edwards JY, Drangova M, Huff MW. Intervention with citrus flavonoids reverses obesity and improves metabolic syndrome and atherosclerosis in obese Ldlr-/- mice. J Lipid Res 2018; 59:1714-1728. [PMID: 30008441 DOI: 10.1194/jlr.m087387] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/12/2018] [Indexed: 12/11/2022] Open
Abstract
Obesity and its associated metabolic dysfunction and cardiovascular disease risk represent a leading cause of adult morbidity worldwide. Currently available pharmacological therapies for obesity have had limited success in reversing existing obesity and metabolic dysregulation. Previous prevention studies demonstrated that the citrus flavonoids, naringenin and nobiletin, protect against obesity and metabolic dysfunction in Ldlr-/- mice fed a high-fat cholesterol-containing (HFHC) diet. However, their effects in an intervention model are unknown. In this report, we show that, in Ldlr-/- mice with diet-induced obesity, citrus flavonoid supplementation to a HFHC diet reversed existing obesity and adipocyte size and number through enhanced energy expenditure and increased hepatic fatty acid oxidation. Caloric intake was unaffected and no evidence of white adipose tissue browning was observed. Reversal of adiposity was accompanied by improvements in hyperlipidemia, insulin sensitivity, hepatic steatosis, and a modest reduction in blood monocytes. Together, this resulted in atherosclerotic lesions that were unchanged in size, but characterized by reduced macrophage content, consistent with a more stable plaque phenotype. These studies further suggest potential therapeutic utility of citrus flavonoids, especially in the context of existing obesity, metabolic dysfunction, and cardiovascular disease.
Collapse
Affiliation(s)
- Amy C Burke
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7; Robarts Research Institute, Departments of Biochemistry University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Brian G Sutherland
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Dawn E Telford
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7; Medicine, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Marisa R Morrow
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Cynthia G Sawyez
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7; Medicine, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Jane Y Edwards
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7; Medicine, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Maria Drangova
- Imaging Research Laboratories, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Murray W Huff
- Molecular Medicine University of Western Ontario, London, Ontario, Canada N6A 5B7; Robarts Research Institute, Departments of Biochemistry University of Western Ontario, London, Ontario, Canada N6A 5B7; Medicine, University of Western Ontario, London, Ontario, Canada N6A 5B7.
| |
Collapse
|