1
|
Xue J, Allaband C, Zuffa S, Poulsen O, Meadows J, Zhou D, Dorrestein PC, Knight R, Haddad GG. Gut microbiota and derived metabolites mediate obstructive sleep apnea induced atherosclerosis. Gut Microbes 2025; 17:2474142. [PMID: 40025767 PMCID: PMC11881840 DOI: 10.1080/19490976.2025.2474142] [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: 12/04/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia/hypercapnia (IHC), affects predominantly obese individuals, and increases atherosclerosis risk. Since we and others have implicated gut microbiota and metabolites in atherogenesis, we dissected their contributions to OSA-induced atherosclerosis. Atherosclerotic lesions were compared between conventionally-reared specific pathogen free (SPF) and germ-free (GF) Apoe-/- mice following a high fat high cholesterol diet (HFHC), with and without IHC conditions. The fecal microbiota and metabolome were profiled using 16S rRNA gene amplicon sequencing and untargeted tandem mass spectrometry (LC-MS/MS) respectively. Phenotypic data showed that HFHC significantly increased atherosclerosis as compared to regular chow (RC) in both aorta and pulmonary artery (PA) of SPF mice. IHC exacerbated lesions in addition to HFHC. Differential abundance analysis of gut microbiota identified an enrichment of Akkermansiaceae and a depletion of Muribaculaceae (formerly S24-7) family members in the HFHC-IHC group. LC-MS/MS showed a dysregulation of bile acid profiles with taurocholic acid, taurodeoxycholic acid, and 12-ketodeoxycholic acid enriched in the HFHC-IHC group, long-chain N-acyl amides, and phosphatidylcholines. Interestingly, GF Apoe-/- mice markedly reduced atherosclerotic formation relative to SPF Apoe-/- mice in the aorta under HFHC/IHC conditions. In contrast, microbial colonization did not show a significant impact on the atherosclerotic progression in PA. In summary, this research demonstrated that (1) IHC acts cooperatively with HFHC to induce atherosclerosis; (2) gut microbiota modulate atherogenesis, induced by HFHC/IHC, in the aorta not in PA; (3) different analytical methods suggest that a specific imbalance between Akkermansiaceae and Muribaculaceae bacterial families mediate OSA-induced atherosclerosis; and (4) derived bile acids, such as deoxycholic acid and lithocholic acid, regulate atherosclerosis in OSA. The knowledge obtained provides novel insights into the potential therapeutic approaches to prevent and treat OSA-induced atherosclerosis.
Collapse
Affiliation(s)
- Jin Xue
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Celeste Allaband
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, San Diego, CA, USA
| | - Orit Poulsen
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Jason Meadows
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Dan Zhou
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Pieter C. Dorrestein
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Gabriel G. Haddad
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
- The Division of Respiratory Medicine, Rady Children’s Hospital, San Diego, CA, USA
| |
Collapse
|
2
|
Wang H, Kc P, Zhang K, Materne C, Lhomme M, Galier S, Ichou F, Neves C, Lehuen A, Haas JT, Salem JE, Guerin M, Lesnik P. MAIT Cells Promote Cholesterol Excretion Pathways Mitigating Atherosclerosis. Circ Res 2025; 136:968-981. [PMID: 40135347 DOI: 10.1161/circresaha.124.325841] [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: 11/08/2024] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous clinical studies have indicated reduced circulating mucosal-associated invariant T (MAIT) cells in individuals with coronary artery disease. However, the precise role and underlying mechanisms of MAIT cells in this context remain unclear. Immune homeostasis plays a pivotal role in the development of atherosclerosis. This study explores the impact of MAIT cells on atherosclerosis. METHODS Vα19+/- Ldlr-/- mice, characterized by a high MAIT cell frequency, and MAIT cell deficient MR1-/- (major histocompatibility complex-related molecule 1) Ldlr-/- mice and their respective controls were used. Starting at 6 weeks of age, mice were subjected to a 1% cholesterol diet for 16 weeks. Additionally, the study analyzed circulating MAIT cell frequency and cholesterol levels in 68 patients with hypercholesterolemia. RESULTS In Vα19+/- Ldlr-/- mice, increased MAIT cells demonstrated a protective effect against atherosclerosis by reducing VLDL-C (very-low-density lipoprotein cholesterol) levels through heightened cholesterol excretion. This effect was accompanied by elevated jejunal ABCB1a, ABCG5, and ABCG8 expression, mediated by augmented levels of Liver X receptor transcription and activation, likely through intestinal IL-22 (interleukin-22) signaling. Conversely, cholesterol reduction mediated by intestinal cholesterol excretion was blocked by inhibition of MAIT cells. Moreover, MAIT cell-deficient MR1-/- Ldlr-/- mice exhibited elevated total cholesterol levels and increased atherosclerotic lesions. In patients with hypercholesterolemia, circulating MAIT cell frequency displayed negative correlations with VLDL-C levels and positive correlations with HDL-C (high-density lipoprotein cholesterol) levels. CONCLUSIONS Our findings demonstrate a new mechanism for plasma VLDL-C clearance by MAIT cell-mediated cholesterol excretion. The results provide further evidence that immunity is involved in cholesterol homeostasis. Targeting intestinal immunity to regulate cholesterol homeostasis holds promise as a new cholesterol-lowering modality to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Hua Wang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Pukar Kc
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Kaidi Zhang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Clément Materne
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Sophie Galier
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Farid Ichou
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Carolina Neves
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Agnès Lehuen
- Université Paris Cité, Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique UMR 8104, Inflamex Laboratory, Paris, France (A.L.)
| | - Joel T Haas
- Université de Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, Lille, France (J.T.H.)
| | - Joe-Elie Salem
- INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France (J.-E.S.)
| | - Maryse Guerin
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Philippe Lesnik
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| |
Collapse
|
3
|
Zhang L, Al-Ammari A, Zhu D, Zhang H, Zhou P, Zhi X, Ding W, Li X, Yu Q, Gai Y, Ma X, Chen C, Zuo C, Zhang J, Zhu W, Sun D. A nanovaccine for immune activation and prophylactic protection of atherosclerosis in mouse models. Nat Commun 2025; 16:2111. [PMID: 40025093 PMCID: PMC11873251 DOI: 10.1038/s41467-025-57467-5] [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: 10/08/2023] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Vaccines offer prophylactic treatments against atherosclerosis by eliciting effector T cell and antibody responses, which require effective delivery of antigen and adjuvant to activate dendritic cells (DC). Here we show that individual conjugation of antigen p210 and adjuvant CpG oligodeoxynucleotides onto superparamagnetic iron oxide nanoparticles formulates a nanovaccine cocktail that activates DCs for antigen cross-presentation and induction of co-stimulatory signals, cytokines and CD8+ effector/effector memory T cell responses. This nanovaccine modulates the DCs in the draining lymph nodes, activates both CD4+ and CD8+ T cells, elicits memory responses, and induces both anti-p210 IgM and IgG antibodies to suppress atherosclerosis. Lastly, three intradermal vaccinations of this nanovaccine mitigate the atherosclerosis development in the ApoE-/- mice. Our nanovaccine design and preclinical data thus presents a potential candidate for prophylactic treatment for atherosclerosis.
Collapse
Affiliation(s)
- Lei Zhang
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Abdulrahman Al-Ammari
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, PR China
| | - Danxuan Zhu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Peng Zhou
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Xu Zhi
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, PR China
| | - Weixiao Ding
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Xinmeng Li
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Qingqing Yu
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Yuwen Gai
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Xiaoling Ma
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Chuntao Chen
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Chao Zuo
- School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, PR China
| | - Jiaan Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, PR China.
| | - Wanying Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, PR China.
| | - Dongping Sun
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, PR China.
| |
Collapse
|
4
|
Li S, Gao H, Wang H, Zhao X, Pan D, Pacheco-Fernández I, Ma M, Liu J, Hirvonen J, Liu Z, Santos HA. Tailored polysaccharide entrapping metal-organic framework for RNAi therapeutics and diagnostics in atherosclerosis. Bioact Mater 2025; 43:376-391. [PMID: 39399834 PMCID: PMC11470791 DOI: 10.1016/j.bioactmat.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024] Open
Abstract
Metal-organic frameworks (MOFs) hold promise as theranostic carriers for atherosclerosis. However, to further advance their therapeutic effects with higher complexity and functionality, integrating multiple components with complex synthesis procedures are usually involved. Here, we reported a facile and general strategy to prepare multifunctional anti-atherosclerosis theranostic platform in a single-step manner. A custom-designed multifunctional polymer, poly(butyl methacrylate-co-methacrylic acid) branched phosphorylated β-glucan (PBMMA-PG), can effectively entrap different MOFs via coordination, simultaneously endow the MOF with enhanced stability, lesional macrophages selectivity and enhanced endosome escape. Sequential ex situ characterization and computational studies elaborated the potential mechanism. This facile post-synthetic modification granted the administered nanoparticles atherosclerotic tropism by targeting Dectin-1+ macrophages, enhancing in situ MR signal intensity by 72 %. Delivery of siNLRP3 effectively mitigated NLRP3 inflammasomes activation, resulting a 43 % reduction of plaque area. Overall, the current study highlights a simple and general approach for fabricating a MOF-based theranostic platform towards atherosclerosis conditioning, which may also expand to other indications targeting the lesional macrophages.
Collapse
Affiliation(s)
- Sen Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Han Gao
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Haoji Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Xiaolin Zhao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, 210009, PR China
| | - Idaira Pacheco-Fernández
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Jianjun Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Zehua Liu
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
5
|
Su S, Chen Z, Ke Q, Kocher O, Krieger M, Kang PM. Nanoparticle-Directed Antioxidant Therapy Can Ameliorate Disease Progression in a Novel, Diet-Inducible Model of Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:2476-2488. [PMID: 39417229 PMCID: PMC11602363 DOI: 10.1161/atvbaha.124.321030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Oxidative stress plays a crucial role in the pathogenesis of coronary artery disease. In cardiovascular research using murine models, the generation and maintenance of models with robust coronary arterial atherosclerosis has been challenging. METHODS We characterized a new mouse model in which the last 3 amino acids of the carboxyl terminus of the HDL (high-density lipoprotein) receptor (SR-B1 [scavenger receptor, class B, type 1]) were deleted in a low-density lipoprotein receptor knockout (LDLR-/-) mouse model (SR-B1ΔCT/LDLR-/-) fed an atherogenic diet. We also tested the therapeutic effects of an oxidative stress-targeted nanoparticle in atherogenic diet-fed SR-B1ΔCT/LDLR-/- mice. RESULTS The SR-B1ΔCT/LDLR-/- mice fed an atherogenic diet had occlusive coronary artery atherosclerosis, impaired cardiac function, and a dramatically lower survival rate, compared with LDLR-/- mice fed the same diet. As SR-B1ΔCT/LDLR-/- mice do not exhibit female infertility or low pup yield, they are far easier and less costly to use than the previously described SR-B1-based models of coronary artery disease. We found that treatment with the targeted nanoparticles improved the cardiac functions and corrected hematologic abnormalities caused by the atherogenic diet in SR-B1ΔCT/LDLR-/- mice but did not alter the distinctive plasma lipid levels. CONCLUSIONS The SR-B1ΔCT/LDLR-/- mice developed diet-inducible, fatal atherosclerotic coronary artery disease, which could be ameliorated by targeted nanoparticle therapy. Our study provides new tools for the development of cardiovascular therapies.
Collapse
Affiliation(s)
- Shi Su
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhifen Chen
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Xue J, Allaband C, Zuffa S, Poulsen O, Meadows J, Zhou D, Dorrestein PC, Knight R, Haddad GG. Gut Microbiota and Derived Metabolites Mediate Obstructive Sleep Apnea Induced Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624205. [PMID: 39605650 PMCID: PMC11601605 DOI: 10.1101/2024.11.18.624205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia/hypercapnia (IHC), affects predominantly obese individuals, and increases atherosclerosis risk. Since we and others have implicated gut microbiota and metabolites in atherogenesis, we dissected their contributions to OSA-induced atherosclerosis. Results Atherosclerotic lesions were compared between conventionally-reared specific pathogen free (SPF) and germ-free (GF) ApoE -/- mice following a high fat high cholesterol diet (HFHC), with and without IHC conditions. The fecal microbiota and metabolome were profiled using 16S rRNA gene amplicon sequencing and untargeted tandem mass spectrometry (LC-MS/MS) respectively. Phenotypic data showed that HFHC significantly increased atherosclerosis as compared to regular chow (RC) in both aorta and pulmonary artery (PA) of SPF mice. IHC exacerbated lesions in addition to HFHC. Differential abundance analysis of gut microbiota identified an enrichment of Akkermansiaceae and a depletion of Muribaculaceae (formerly S24-7) family members in the HFHC-IHC group. LC-MS/MS showed a dysregulation of bile acid profiles with taurocholic acid, taurodeoxycholic acid, and 12-ketodeoxycholic acid enriched in the HFHC-IHC group, long-chain N-acyl amides, and phosphatidylcholines. Interestingly, GF ApoE -/- mice markedly reduced atherosclerotic formation relative to SPF ApoE -/- mice in the aorta under HFHC/IHC conditions. In contrast, microbial colonization did not show a significant impact on the atherosclerotic progression in PA. Conclusions In summary, this research demonstrated that (1) IHC acts cooperatively with HFHC to induce atherosclerosis; (2) gut microbiota modulate atherogenesis, induced by HFHC/IHC, in the aorta not in PA; (3) different analytical methods suggest that a specific imbalance between Akkermansiaceae and Muribaculaceae bacterial families mediate OSA-induced atherosclerosis; and (4) derived bile acids, such as deoxycholic acid and lithocholic acid, regulate atherosclerosis in OSA. The knowledge obtained provides novel insights into the potential therapeutic approaches to prevent and treat OSA-induced atherosclerosis.
Collapse
|
7
|
Maffia P, Mauro C, Case A, Kemper C. Canonical and non-canonical roles of complement in atherosclerosis. Nat Rev Cardiol 2024; 21:743-761. [PMID: 38600367 DOI: 10.1038/s41569-024-01016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Cardiovascular diseases are the leading cause of death globally, and atherosclerosis is the major contributor to the development and progression of cardiovascular diseases. Immune responses have a central role in the pathogenesis of atherosclerosis, with the complement system being an acknowledged contributor. Chronic activation of liver-derived and serum-circulating canonical complement sustains endothelial inflammation and innate immune cell activation, and deposition of complement activation fragments on inflamed endothelial cells is a hallmark of atherosclerotic plaques. However, increasing evidence indicates that liver-independent, cell-autonomous and non-canonical complement activities are underappreciated contributors to atherosclerosis. Furthermore, complement activation can also have atheroprotective properties. These specific detrimental or beneficial contributions of the complement system to the pathogenesis of atherosclerosis are dictated by the location of complement activation and engagement of its canonical versus non-canonical functions in a temporal fashion during atherosclerosis progression. In this Review, we summarize the classical and the emerging non-classical roles of the complement system in the pathogenesis of atherosclerosis and discuss potential strategies for therapeutic modulation of complement for the prevention and treatment of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance (ARUA) & The Guild, Accra, Ghana
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Ayden Case
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Complement and Inflammation Research Section, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
8
|
Chang Z, Zhou Y, Dong L, Qiao LR, Yang H, Xu GK. Deciphering the complex mechanics of atherosclerotic plaques: A hybrid hierarchical theory-microrheology approach. Acta Biomater 2024; 189:399-412. [PMID: 39307259 DOI: 10.1016/j.actbio.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024]
Abstract
Understanding the viscoelastic properties of atherosclerotic plaques at rupture-prone scales is crucial for assessing their vulnerability. Here, we develop a Hybrid Hierarchical theory-Microrheology (HHM) approach, enabling the analysis of multiscale mechanical variations and distribution changes in regional tissue viscoelasticity within plaques across different spatial scales. We disclose a universal two-stage power-law rheology in plaques, characterized by distinct power-law exponents (αshort and αlong), which serve as mechanical indexes for plaque components and assessing mechanical gradients. We further propose a self-similar hierarchical theory that effectively delineates plaque heterogeneity from the cytoplasm, cell, to tissue levels. Moreover, our proposed multi-layer perceptron model addresses the viscoelastic heterogeneity and gradients within plaques, offering a promising diagnostic strategy for identifying unstable plaques. These findings not only advance our understanding of plaque mechanics but also pave the way for innovative diagnostic approaches in cardiovascular disease management. STATEMENT OF SIGNIFICANCE: Our study pioneers a Hybrid Hierarchical theory-Microrheology (HHM) approach to dissect the intricate viscoelasticity of atherosclerotic plaques, focusing on distinct components including cap fibrosis, lipid pools, and intimal fibrosis. We unveil a universal two-stage power-law rheology capturing mechanical variations across plaque structures. The proposed hierarchical model adeptly captures viscoelasticity changes from cytoplasm, cell to tissue levels. Based on the newly proposed markers, we further develop a machine learning (ML) diagnostic model that sets precise criteria for evaluating plaque components and heterogeneity. This work not only reveals the comprehensive mechanical heterogeneity within plaques but also introduces a mechanical marker-based ML strategy for assessing plaque conditions, offering a significant leap towards understanding and diagnosing atherosclerotic risks.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, China
| | - Le Dong
- School of Artificial Intelligence, Xidian University, Xi'an 710071, China
| | - Lin-Ru Qiao
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, China.
| | - Guang-Kui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
9
|
Park SH, Kang MK, Kim DY, Lim SS, Kang YH. Dietary ellagic acid blocks inflammation-associated atherosclerotic plaque formation in cholesterol-fed apoE-deficient mice. Nutr Res Pract 2024; 18:617-632. [PMID: 39398881 PMCID: PMC11464280 DOI: 10.4162/nrp.2024.18.5.617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/09/2024] [Accepted: 07/05/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND/OBJECTIVES Atherosclerosis particularly due to high circulating level of low-density lipoprotein is a major cause of cardiovascular diseases. Ellagic acid is a natural polyphenolic compound rich in pomegranates and berries. Our previous study showed that ellagic acid improved functionality of reverse cholesterol transport in murine model of atherosclerosis. The aim of this study is to investigate whether ellagic acid inhibited inflammation-associated atherosclerotic plaque formation in cholesterol-fed apolipoprotein E (apoE)-knockout (KO) mice. MATERIALS/METHODS Wild type mice and apoE-KO mice were fed a cholesterol-rich Paigen diet for 10 weeks to induce severe atherosclerosis. Concurrently, 10 mg/kg ellagic acid was orally administered to the apoE-KO mice. Plaque lesion formation and lipid deposition were examined by staining with hematoxylin and eosin, Sudan IV and oil red O. RESULTS The plasma leukocyte profile of cholesterol-fed mice was not altered by apoE deficiency. Oral administration of ellagic acid attenuated plaque lesion formation and lipid deposition in the aorta tree of apoE-KO mice. Ellagic acid substantially reduced plasma levels of soluble vascular cell adhesion molecule and interferon-γ in Paigen diet-fed apoE-KO mice. When 10 mg/kg ellagic acid was administered to cholesterol-fed apoE-KO mice, the levels of CD68 and MCP-1 were strongly reduced in aorta vessels. The protein expression level of nitric oxide synthase-2 (NOS2) in the aorta was highly enhanced by supplementation of ellagic acid to apoE-KO mice, but the expression level of heme oxygenase-1 (HO-1) in the aorta was reduced. Furthermore, ellagic acid diminished the increased aorta expression of the inflammatory adhesion molecules in cholesterol-fed apoE-KO mice. The treatment of ellagic acid inhibited the scavenger receptor-B1 expression in the aorta of apoE-KO mice, while the cholesterol efflux-related transporters were not significantly changed. CONCLUSION These results suggest that ellagic acid may be an atheroprotective compound by attenuating apoE deficiency-induced vascular inflammation and reducing atherosclerotic plaque lesion formation.
Collapse
Affiliation(s)
- Sin-Hye Park
- Department of Food Science and Nutrition and Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Min-Kyung Kang
- Department of Food and Nutrition, Andong National University, Andong 36729, Korea
| | - Dong Yeon Kim
- Department of Food and Nutrition, Andong National University, Andong 36729, Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition and Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition and Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
10
|
Jin Y, Kozan D, Young ED, Hensley MR, Shen MC, Wen J, Moll T, Anderson JL, Kozan H, Rawls JF, Farber SA. A high-cholesterol zebrafish diet promotes hypercholesterolemia and fasting-associated liver steatosis. J Lipid Res 2024; 65:100637. [PMID: 39218217 PMCID: PMC11913794 DOI: 10.1016/j.jlr.2024.100637] [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: 10/19/2023] [Revised: 07/22/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Zebrafish are an ideal model organism to study lipid metabolism and to elucidate the molecular underpinnings of human lipid-associated disorders. Unlike murine models, to which various standardized high lipid diets such as a high-cholesterol diet (HCD) are available, there has yet to be a uniformly adopted zebrafish HCD protocol. In this study, we have developed an improved HCD protocol and thoroughly tested its impact on zebrafish lipid deposition and lipoprotein regulation in a dose- and time-dependent manner. The diet stability, reproducibility, and fish palatability were also validated. Fish fed HCD developed hypercholesterolemia as indicated by significantly elevated ApoB-containing lipoproteins (ApoB-LPs) and increased plasma levels of cholesterol and cholesterol esters. Feeding of the HCD to larvae for 8 days produced hepatic steatosis that became more stable and sever after 1 day of fasting and was associated with an opaque liver phenotype (dark under transmitted light). Unlike larvae, adult fish fed HCD for 14 days followed by a 3-day fast did not develop a stable fatty liver phenotype, though the fish had higher ApoB-LP levels in plasma and an upregulated lipogenesis gene fasn in adipose tissue. In conclusion, our HCD zebrafish protocol represents an effective and reliable approach for studying the temporal characteristics of the physiological and biochemical responses to high levels of dietary cholesterol and provides insights into the mechanisms that may underlie fatty liver disease.
Collapse
Affiliation(s)
- Yang Jin
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Aas, Norway
| | - Darby Kozan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Eric D Young
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Division of Gastrointestinal and Liver Pathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Monica R Hensley
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Meng-Chieh Shen
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Jia Wen
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Tabea Moll
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer L Anderson
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Hannah Kozan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Steven A Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
11
|
Tehlivets O, Almer G, Brunner MS, Lechleitner M, Sommer G, Kolb D, Leitinger G, Diwoky C, Wolinski H, Habisch H, Opriessnig P, Bogoni F, Pernitsch D, Kavertseva M, Bourgeois B, Kukilo J, Tehlivets YG, Schwarz AN, Züllig T, Bubalo V, Schauer S, Groselj-Strele A, Hoefler G, Rechberger GN, Herrmann M, Eller K, Rosenkranz AR, Madl T, Frank S, Holzapfel GA, Kratky D, Mangge H, Hörl G. Homocysteine contributes to atherogenic transformation of the aorta in rabbits in the absence of hypercholesterolemia. Biomed Pharmacother 2024; 178:117244. [PMID: 39116783 DOI: 10.1016/j.biopha.2024.117244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Atherosclerosis, the leading cause of cardiovascular disease, cannot be sufficiently explained by established risk factors, including cholesterol. Elevated plasma homocysteine (Hcy) is an independent risk factor for atherosclerosis and is closely linked to cardiovascular mortality. However, its role in atherosclerosis has not been fully clarified yet. We have previously shown that rabbits fed a diet deficient in B vitamins and choline (VCDD), which are required for Hcy degradation, exhibit an accumulation of macrophages and lipids in the aorta, aortic stiffening and disorganization of aortic collagen in the absence of hypercholesterolemia, and an aggravation of atherosclerosis in its presence. In the current study, plasma Hcy levels were increased by intravenous injections of Hcy into balloon-injured rabbits fed VCDD (VCDD+Hcy) in the absence of hypercholesterolemia. While this treatment did not lead to thickening of aortic wall, intravenous injections of Hcy into rabbits fed VCDD led to massive accumulation of VLDL-triglycerides as well as significant impairment of vascular reactivity of the aorta compared to VCDD alone. In the aorta intravenous Hcy injections into VCDD-fed rabbits led to fragmentation of aortic elastin, accumulation of elastin-specific electron-dense inclusions, collagen disorganization, lipid degradation, and autophagolysosome formation. Furthermore, rabbits from the VCDD+Hcy group exhibited a massive decrease of total protein methylated arginine in blood cells and decreased creatine in blood cells, serum and liver compared to rabbits from the VCDD group. Altogether, we conclude that Hcy contributes to atherogenic transformation of the aorta not only in the presence but also in the absence of hypercholesterolemia.
Collapse
Affiliation(s)
- Oksana Tehlivets
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Division of General Radiology, Department of Radiology, Medical University of Graz, Graz, Austria.
| | - Gunter Almer
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Markus S Brunner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerhard Sommer
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria; Center for Medical Research, Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Clemens Diwoky
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hansjörg Habisch
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Peter Opriessnig
- Division of General Neurology, Department of Neurology, Medical University of Graz, Graz, Austria; Division of Pediatric Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Francesca Bogoni
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Dominique Pernitsch
- Center for Medical Research, Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Maria Kavertseva
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Benjamin Bourgeois
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Jelena Kukilo
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Yuriy G Tehlivets
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Andreas N Schwarz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Thomas Züllig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Vladimir Bubalo
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Silvia Schauer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Andrea Groselj-Strele
- Center for Medical Research, Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Markus Herrmann
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | | | - Tobias Madl
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria; Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Gerd Hörl
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| |
Collapse
|
12
|
Ferraro B. The SR-B1ΔCT/LDLR KO mouse: A new tool to shed light on coronary artery disease. Atherosclerosis 2024; 395:117564. [PMID: 38796408 DOI: 10.1016/j.atherosclerosis.2024.117564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/28/2024]
Affiliation(s)
- Bartolo Ferraro
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig- Maximilian-University Munich, Planegg-Martinsried, Germany; Medizinische Klinik und Poliklinik I, Klinikum der Universität, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
13
|
Santana TM, Caria SJ, Carlini GCG, Rogero MM, Donato J, Tavares MR, Castro IA. Trans-resveratrol reduced hepatic oxidative stress in an animal model without inducing an upregulation of nuclear factor erythroid 2-related factor 2. J Clin Biochem Nutr 2024; 75:40-45. [PMID: 39070534 PMCID: PMC11273272 DOI: 10.3164/jcbn.23-124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/09/2024] [Indexed: 07/30/2024] Open
Abstract
Trans-resveratrol, a widely used supplement for humans, aims to enhance the body's antioxidant defense. Studies suggest that it exerts anti-inflammatory and antioxidant effects by activating the nuclear factor erythroid 2-related factor 2 (Nrf2). In order to evaluate this hypothesis, LDLr(-/-) mice were fed a Western diet to induce liver inflammation and oxidative stress. One group was fed a diet containing 0.60 mg/day of trans-resveratrol (RESV), while another group received no dietary supplementation (CONT). Oxidative stress biomarkers and inflammatory cytokines were assessed in liver homogenates. It was observed that trans-resveratrol decreased hepatic oxidative stress by increasing the GSH/GSSG ratio and reducing malondialdehyde (MDA) concentration. However, the RESV group exhibited a reduction in Nrf2 relative expression compared to CONT. Additionally, trans-resveratrol supplementation reduced nuclear factor-κB (NF-κB) expression but led to an increase in IL-6, with no significant changes observed in tumor necrosis factor-α (TNF-α) and interleukin-10 (IL-10) concentrations. Overall, these findings indicate that the in vivo antioxidant impact induced by trans-resveratrol supplementation in hepatic tissue did not correlate with increase of inflammatory cytokines and Nrf2 relative expression. Further exploration of alternative mechanisms, such as direct radical scavenger activity, is warranted to elucidate the antioxidant effect.
Collapse
Affiliation(s)
- Tamires M. Santana
- LADAF. Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
- Food Research Center (FoRC), CEPID-FAPESP, Research Innovation and Dissemination Centers São Paulo Research Foundation, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
| | - Sarah J. Caria
- LADAF. Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
| | - Giovanna C. G. Carlini
- LADAF. Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
| | - Marcelo M. Rogero
- Food Research Center (FoRC), CEPID-FAPESP, Research Innovation and Dissemination Centers São Paulo Research Foundation, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
- Nutritional Genomics and Inflammation Laboratory, Department of Nutrition, School of Public Health, University of São Paulo, Av. Dr. Arnaldo, 715, São Paulo 01246-904, Brazil
| | - José Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Lineu Prestes, 2415, São Paulo 05508-900, Brazil
| | - Mariana R. Tavares
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Lineu Prestes, 2415, São Paulo 05508-900, Brazil
| | - Inar A. Castro
- LADAF. Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
- Food Research Center (FoRC), CEPID-FAPESP, Research Innovation and Dissemination Centers São Paulo Research Foundation, Av. Lineu Prestes, 580, B14, São Paulo 05508-900, Brazil
| |
Collapse
|
14
|
Li J, Yang L, Song J, Yan B, Morris AJ, Moseley H, Flight R, Wang C, Liu J, Weiss HL, Morris EF, Abdelhamid I, Gerl MJ, Melander O, Smyth S, Evers BM. Neurotensin accelerates atherosclerosis and increases circulating levels of short-chain and saturated triglycerides. Atherosclerosis 2024; 392:117479. [PMID: 38423808 PMCID: PMC11088984 DOI: 10.1016/j.atherosclerosis.2024.117479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND AND AIMS Obesity and type 2 diabetes are significant risk factors for atherosclerotic cardiovascular disease (CVD) worldwide, but the underlying pathophysiological links are poorly understood. Neurotensin (NT), a 13-amino-acid hormone peptide, facilitates intestinal fat absorption and contributes to obesity in mice fed a high-fat diet. Elevated levels of pro-NT (a stable NT precursor produced in equimolar amounts relative to NT) are associated with obesity, type 2 diabetes, and CVD in humans. Whether NT is a causative factor in CVD is unknown. METHODS Nt+/+ and Nt-/- mice were either injected with adeno-associated virus encoding PCSK9 mutants or crossed with Ldlr-/- mice and fed a Western diet. Atherosclerotic plaques were analyzed by en face analysis, Oil Red O and CD68 staining. In humans, we evaluated the association between baseline pro-NT and growth of carotid bulb thickness after 16.4 years. Lipidomic profiles were analyzed. RESULTS Atherosclerotic plaque formation is attenuated in Nt-deficient mice through mechanisms that are independent of reductions in circulating cholesterol and triglycerides but associated with remodeling of the plasma triglyceride pool. An increasing plasma concentration of pro-NT predicts atherosclerotic events in coronary and cerebral arteries independent of all major traditional risk factors, indicating a strong link between NT and atherosclerosis. This plasma lipid profile analysis confirms the association of pro-NT with remodeling of the plasma triglyceride pool in atherosclerotic events. CONCLUSIONS Our findings are the first to directly link NT to increased atherosclerosis and indicate the potential role for NT in preventive and therapeutic strategies for CVD.
Collapse
Affiliation(s)
- Jing Li
- University of Kentucky, Lexington, KY, 40536, USA
| | - Liping Yang
- University of Kentucky, Lexington, KY, 40536, USA
| | - Jun Song
- University of Kentucky, Lexington, KY, 40536, USA
| | - Baoxiang Yan
- University of Kentucky, Lexington, KY, 40536, USA
| | - Andrew J Morris
- University of Arkansas for Medical Sciences, Little Rock, AR, 77205, USA
| | | | | | - Chi Wang
- University of Kentucky, Lexington, KY, 40536, USA
| | - Jinpeng Liu
- University of Kentucky, Lexington, KY, 40536, USA
| | | | - Edward F Morris
- Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | | | | | | | - Susan Smyth
- University of Arkansas for Medical Sciences, Little Rock, AR, 77205, USA
| | - B Mark Evers
- University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
15
|
Gao JP, Zhang HP, Wei R, Guo W. A Novel Method for the Rat Model of Abdominal Aortic Aneurysm Induced by Retroperitoneal Implantation of an Osmotic Pump System With Lipopolysaccharide. Ann Vasc Surg 2024; 101:41-52. [PMID: 38154490 DOI: 10.1016/j.avsg.2023.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/17/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Few methods can cocurrently mimic the pathological characteristics and nature history of human abdominal aortic aneurysms (AAAs), especially for the exist of the self-healing tendency of rodents. This study tested a novel method for the AAA rat model induced by retroperitoneal implantation of an osmotic pump system with lipopolysaccharide (LPS) based on the hypothesis that chronic inflammation of perivascular adipose tissue directly influenced the development and progression of AAAs. METHODS 20 male Sprague-Dawley rats (10-month-old) fed with the Paigen diet were randomly divided into 4 groups: the blank group ×2, the sham group ×4, the empty capsule group ×4, and the LPS capsule group ×10. The LPS capsule group received implantations of the ALZET® osmotic pump capsule with LPS (3.6 μg/day) parallel to the abdominal aorta through a retroperitoneal approach. Two weeks later, 6 rats were randomly selected from the LPS capsule group to form the anti-inflammatory group and received implantations of another osmotic pump capsule with interleukin (IL)-10 (75 ng/day) through the same approach. The changes in abdominal aortic diameter were observed by ultrasound every 2 weeks, and samples were harvested for histopathologic and immunohistochemical analysis 6 weeks later. RESULTS Within the 6 weeks after modeling, the LPS capsule group showed sustained and significant aortic dilatation (P < 0.01), while the anti-inflammatory group showed a rapid and obvious shrinkage 2 weeks after the IL-10 osmotic pump capsule implantation (P < 0.01). The LPS capsule group presented excellent pathological mimicking of human AAAs and showed severe medial degeneration with the least elastic content among the 5 groups at the end of the sixth week (P < 0.05). Notably, the anti-inflammatory group showed perfect medial preservation with the most elastic content (P < 0.05) and the highest elastin/collagen ratio (P < 0.01) at the end of the study. Matrix metalloproteinases (MMP) 2 and 9 and toll-like receptor 2 showed strong expression in the LPS capsule group at the end of the sixth week, which was significantly higher than that in the blank group and sham group. Interestingly, the anti-inflammatory group showed a slightly higher MMP9 expression than the LPS capsule group though there was no statistical difference between them. CONCLUSIONS This novel method for the rat AAA model induced by retroperitoneal implantation of an osmotic pump capsule with LPS can concurrently mimic the histological characteristics and natural history of human AAAs. Further studies were needed to improve the osmotic pump system.
Collapse
Affiliation(s)
- Jiang-Ping Gao
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Hong-Peng Zhang
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Ren Wei
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Wei Guo
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
16
|
Sachan V, Le Dévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Resiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQH, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, Seidah NG. PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease. Metabolism 2024; 150:155736. [PMID: 37967646 DOI: 10.1016/j.metabol.2023.155736] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Epidemiological evidence links the proprotein convertase subtilisin/kexin 7 (PCSK7) to triglyceride (TG) metabolism. We associated the known PCSK7 gain-of-function non-coding SNP rs236918 with higher levels of plasma apolipoprotein B (apoB) and the loss-of-function coding variant p.Pro777Leu (SNP rs201598301) with lower apoB and TG. Herein, we aimed to unravel the in vivo role of liver PCSK7. METHODS We biochemically defined the functional role of PCSK7 in lipid metabolism using hepatic cell lines and Pcsk7-/- mice. Our findings were validated following subcutaneous administration of hepatocyte-targeted N-acetylgalactosamine (GalNAc)-antisense oligonucleotides (ASOs) against Pcsk7. RESULTS Independent of its proteolytic activity, membrane-bound PCSK7 binds apoB100 in the endoplasmic reticulum and enhances its secretion. Mechanistically, the loss of PCSK7/Pcsk7 leads to apoB100 degradation, triggering an unfolded protein response, autophagy, and β-oxidation, eventually reducing lipid accumulation in hepatocytes. Non-alcoholic fatty liver disease (NAFLD) was induced by a 12-week high fat/fructose/cholesterol diet in wild type (WT) and Pcsk7-/- mice that were then allowed to recover on a 4-week control diet. Pcsk7-/- mice recovered more effectively than WT mice from all NAFLD-related liver phenotypes. Finally, subcutaneous administration of GalNAc-ASOs targeting hepatic Pcsk7 to WT mice validated the above results. CONCLUSIONS Our data reveal hepatic PCSK7 as one of the major regulators of apoB, and its absence reduces apoB secretion from hepatocytes favoring its ubiquitination and degradation by the proteasome. This results in a cascade of events, eventually reducing hepatic lipid accumulation, thus supporting the notion of silencing PCSK7 mRNA in hepatocytes for targeting NAFLD.
Collapse
Affiliation(s)
- Vatsal Sachan
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Maïlys Le Dévéhat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Anna Roubtsova
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Rachid Essalmani
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Jean-Francois Laurendeau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Damien Garçon
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Delia Susan-Resiga
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Stéphanie Duval
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Sahar Mikaeeli
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Josée Hamelin
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Alexandra Evagelidis
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Michael Chong
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | | | - Zu-Hua Gao
- Department of Pathology, McGill University Health Centre, Montréal, QC, Canada
| | - Isabelle Robillard
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Matthieu Ruiz
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Vincent Quoc-Huy Trinh
- Departement of Pathology and Cellular Biology, Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, QC, Canada
| | - Jennifer L Estall
- Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - May Faraj
- Nutrition Department, Université de Montréal, Research Unit on Nutrition, Lipoproteins and Cardiometabolic Diseases, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, Canada
| | - Martin Sauvageau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Annik Prat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Robert S Kiss
- McGill University Health Centre Research Institute, Montréal, QC, Canada
| | - Nabil G Seidah
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
17
|
Subramaniam NK, Gagnon N, Makhani K, Kukolj N, Mouradian MH, Giles BH, Srikannan H, Fruh V, Meliker J, Wellenius GA, Mann KK. In vitro and in vivo approaches to assess atherosclerosis following exposure to low-dose mixtures of arsenic and cadmium. Toxicol Appl Pharmacol 2023; 481:116763. [PMID: 37980961 PMCID: PMC11414205 DOI: 10.1016/j.taap.2023.116763] [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: 08/17/2023] [Revised: 10/29/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Worldwide, millions of people are co-exposed to arsenic and cadmium. Environmental exposure to both metals is linked with a higher risk of atherosclerosis. While studies have characterized the pro-atherosclerotic effects of arsenic and cadmium as single agents, little is known about the potential effects of metal mixtures, particularly at low doses. Here, we used a combination of in vitro and in vivo models to assess the effects of low-dose metals individually and as mixtures on early events and plaque development associated with atherosclerosis. In vitro, we investigated early pro-atherogenic changes in macrophages and endothelial cells with metal treatments. The combined cytotoxic effects of both metals at low concentrations were dose interactive, specifically, synergistic in macrophages, but antagonistic in endothelial cells. Despite this differential behavior across cell types, the mixtures did not initiate early pro-atherogenic events: neither reactive oxygen species generation in macrophages nor adhesion molecule expression on endothelial cells. In vivo, we utilized the well-characterized hyperlipidemic apolipoprotein E knock-out (ApoE-/-) mouse model. Previously, we have shown that low concentrations of arsenic (down to 10 ppb) enhance atherosclerosis in ApoE-/- mice. This model has also been used with cadmium to demonstrate pro-atherogenic effects, although at concentrations above human-relevant exposures. In both sexes, there are some small increases in atherosclerotic lesion size, but very few changes in plaque constituents in the ApoE-/- mouse model. Together, these results suggests that low-dose metal mixtures are not significantly more pro-atherogenic than either metal alone.
Collapse
Affiliation(s)
- Nivetha K Subramaniam
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| | - Natascha Gagnon
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| | - Kiran Makhani
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| | - Nikola Kukolj
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| | - Michael H Mouradian
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| | - Braeden H Giles
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| | - Harinee Srikannan
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| | - Victoria Fruh
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA.
| | - Jaymie Meliker
- Program in Public Health, Department of Family, Population, & Preventive Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Gregory A Wellenius
- Center for Climate and Health, Boston University School of Public Health, Boston, MA, USA.
| | - Koren K Mann
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
18
|
Laurans L, Mouttoulingam N, Chajadine M, Lavelle A, Diedisheim M, Bacquer E, Creusot L, Suffee N, Esposito B, Melhem NJ, Le Goff W, Haddad Y, Paul JL, Rainteau D, Tedgui A, Ait-Oufella H, Zitvogel L, Sokol H, Taleb S. An obesogenic diet increases atherosclerosis through promoting microbiota dysbiosis-induced gut lymphocyte trafficking into the periphery. Cell Rep 2023; 42:113350. [PMID: 37897726 DOI: 10.1016/j.celrep.2023.113350] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
Although high-fat diet (HFD)-induced gut microbiota dysbiosis is known to affect atherosclerosis, the underlying mechanisms remain to be fully explored. Here, we show that the progression of atherosclerosis depends on a gut microbiota shaped by an HFD but not a high-cholesterol (HC) diet and, more particularly, on low fiber (LF) intake. Mechanistically, gut lymphoid cells impacted by HFD- or LF-induced microbiota dysbiosis highly proliferate in mesenteric lymph nodes (MLNs) and migrate from MLNs to the periphery, which fuels T cell accumulation within atherosclerotic plaques. This is associated with the induction of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) within plaques and the presence of enterotropic lymphocytes expressing β7 integrin. MLN resection or lymphocyte deficiency abrogates the pro-atherogenic effects of a microbiota shaped by LF. Our study shows a pathological link between a diet-shaped microbiota, gut immune cells, and atherosclerosis, suggesting that a diet-modulated microbiome might be a suitable therapeutic target to prevent atherosclerosis.
Collapse
Affiliation(s)
- Ludivine Laurans
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nirmala Mouttoulingam
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Mouna Chajadine
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Aonghus Lavelle
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), 37540 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades (INEM), Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, 75015 Paris, France
| | - Emilie Bacquer
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laura Creusot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Nadine Suffee
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Bruno Esposito
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nada Joe Melhem
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Wilfried Le Goff
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Yacine Haddad
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France
| | - Jean-Louis Paul
- Université Paris-Sud, Equipe d'Accueil 4529, UFR de Pharmacie, Chatenay-Malabry, France and Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Dominique Rainteau
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Clinical Metabolomics Department, 75012 Paris, France
| | - Alain Tedgui
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France; Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAe, Micalis & AgroParisTech, Jouy en Josas, France
| | - Soraya Taleb
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France.
| |
Collapse
|
19
|
Jin Y, Kozan D, Anderson JL, Hensley M, Shen MC, Wen J, Moll T, Kozan H, Rawls JF, Farber SA. A high-cholesterol zebrafish diet promotes hypercholesterolemia and fasting-associated liver triglycerides accumulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565134. [PMID: 37961364 PMCID: PMC10635069 DOI: 10.1101/2023.11.01.565134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Zebrafish are an ideal model organism to study lipid metabolism and to elucidate the molecular underpinnings of human lipid-associated disorders. In this study, we provide an improved protocol to assay the impact of a high-cholesterol diet (HCD) on zebrafish lipid deposition and lipoprotein regulation. Fish fed HCD developed hypercholesterolemia as indicated by significantly elevated ApoB-containing lipoproteins (ApoB-LP) and increased plasma levels of cholesterol and cholesterol esters. Feeding of the HCD to larvae (8 days followed by a 1 day fast) and adult female fish (2 weeks, followed by 3 days of fasting) was also associated with a fatty liver phenotype that presented as severe hepatic steatosis. The HCD feeding paradigm doubled the levels of liver triacylglycerol (TG), which was striking because our HCD was only supplemented with cholesterol. The accumulated liver TG was unlikely due to increased de novo lipogenesis or inhibited β-oxidation since no differentially expressed genes in these pathways were found between the livers of fish fed the HCD versus control diets. However, fasted HCD fish had significantly increased lipogenesis gene fasn in adipose tissue and higher free fatty acids (FFA) in plasma. This suggested that elevated dietary cholesterol resulted in lipid accumulation in adipocytes, which supplied more FFA during fasting, promoting hepatic steatosis. In conclusion, our HCD zebrafish protocol represents an effective and reliable approach for studying the temporal characteristics of the physiological and biochemical responses to high levels of dietary cholesterol and provides insights into the mechanisms that may underlie fatty liver disease.
Collapse
Affiliation(s)
- Yang Jin
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Aas, Norway
| | - Darby Kozan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Jennifer L Anderson
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
| | - Monica Hensley
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
| | - Meng-Chieh Shen
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
| | - Jia Wen
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, United States
| | - Tabea Moll
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Hannah Kozan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, United States
| | - Steven A. Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, United States
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
20
|
Yan C, Huang SH, Ding HF, Kwek E, Liu JH, Chen ZX, Ma KY, Chen ZY. Adverse effect of oxidized cholesterol exposure on colitis is mediated by modulation of gut microbiota. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132057. [PMID: 37467611 DOI: 10.1016/j.jhazmat.2023.132057] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Both cholesterol and oxidized cholesterol (OXC) are present in human diets. The incidence of inflammatory bowel diseases (IBDs) is increasing in the world. The present study was to investigate the mechanism by which OXC promotes colitis using C57BL/6 mice as a model. Results shown that more severe colitis was developed in OXC-treated mice with the administration of dextran sulfate sodium (DSS) in water. Direct effects of short-term OXC exposure on gut barrier or inflammation were not observed in healthy mice. However, OXC exposure could cause gut microbiota dysbiosis with a decrease in the relative abundance of short-train fatty acids (SCFAs)-producing bacteria (Lachnospiraceae_NK4A136_group and Blautia) and an increase in the abundance of some potential harmful bacteria (Bacteroides). OXC-induced symptoms of colitis were eliminated when mice were administered with antibiotic cocktails, indicating the promoting effect of OXC on DSS-induced colitis was mediated by its effect on gut microbiota. Moreover, bacteria-depleted mice colonized with gut microbiome from OXC-DSS-exposed mice exhibited a severe colitis, further proving the gut dysbiosis caused by OXC exposure was the culprit in exacerbating the colitis. It was concluded that dietary OXC exposure increased the susceptibility of colitis in mice by causing gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Chi Yan
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Shou-He Huang
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Hua-Fang Ding
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Erika Kwek
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Jian-Hui Liu
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Zi-Xing Chen
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Ka Ying Ma
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Zhen-Yu Chen
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China.
| |
Collapse
|
21
|
Lee WE, Genetzakis E, Figtree GA. Novel Strategies in the Early Detection and Treatment of Endothelial Cell-Specific Mitochondrial Dysfunction in Coronary Artery Disease. Antioxidants (Basel) 2023; 12:1359. [PMID: 37507899 PMCID: PMC10376062 DOI: 10.3390/antiox12071359] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Although elevated cholesterol and other recognised cardiovascular risk factors are important in the development of coronary artery disease (CAD) and heart attack, the susceptibility of humans to this fatal process is distinct from other animals. Mitochondrial dysfunction of cells in the arterial wall, particularly the endothelium, has been strongly implicated in the pathogenesis of CAD. In this manuscript, we review the established evidence and mechanisms in detail and explore the potential opportunities arising from analysing mitochondrial function in patient-derived cells such as endothelial colony-forming cells easily cultured from venous blood. We discuss how emerging technology and knowledge may allow us to measure mitochondrial dysfunction as a potential biomarker for diagnosis and risk management. We also discuss the "pros and cons" of animal models of atherosclerosis, and how patient-derived cell models may provide opportunities to develop novel therapies relevant for humans. Finally, we review several targets that potentially alleviate mitochondrial dysfunction working both via direct and indirect mechanisms and evaluate the effect of several classes of compounds in the cardiovascular context.
Collapse
Affiliation(s)
- Weiqian E. Lee
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Elijah Genetzakis
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
22
|
Mosavi SS, Rabizadeh S, Yadegar A, Seifouri S, Mohammadi F, Qahremani R, Salehi SS, Rajab A, Esteghamati A, Nakhjavani M. Therapeutic effects of resveratrol and Omega-3 in mice atherosclerosis: focus on histopathological changes. BMC Complement Med Ther 2023; 23:81. [PMID: 36932392 PMCID: PMC10024363 DOI: 10.1186/s12906-023-03899-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Resveratrol and omega-3 have been shown to prevent atherosclerosis. However, histopathological changes and their comparison have not been studied well. This study investigated the therapeutic effects of resveratrol and omega-3 in experimental atherosclerosis of mice. METHODS We divided sixty 6-week-old male C57BL/6 mice into six groups and followed for 10 weeks: (1) standard diet, (2) atherogenic diet, (3) atherogenic diet along with resveratrol from the start of the sixth week, (4) atherogenic diet along with omega-3 from the start of the sixth week, (5) standard diet along with resveratrol from the start of the sixth week, (6) standard diet along with omega-3 from the start of the sixth week. RESULTS The mice fed on an atherogenic diet had a larger fat area and a thicker aortic wall thickness than mice fed on a standard diet. The use of omega-3 and resveratrol in the mice with an atherogenic diet resulted in a significantly reduced fat area (p-value = 0.003), and resveratrol had a significantly higher effect. Omega-3 or resveratrol induced a significant reduction in aortic wall thickness in mice on an atherogenic diet, and there was no significant difference between them. Among the mice with a standard diet, this study did not observe any significant changes in the fat area or the aortic wall thickness with the consumption of omega-3 or resveratrol. CONCLUSIONS Resveratrol and omega-3 had a regressive and therapeutic role in atherosclerosis, with a more significant effect in favor of resveratrol.
Collapse
Affiliation(s)
- Shamsi Sadat Mosavi
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Soghra Rabizadeh
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Amirhossein Yadegar
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Sara Seifouri
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Fatemeh Mohammadi
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Reihane Qahremani
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Salome Sadat Salehi
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Armin Rajab
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Alireza Esteghamati
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Manouchehr Nakhjavani
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| |
Collapse
|
23
|
Rao X, Razavi M, Mihai G, Wei Y, Braunstein Z, Frieman MB, Sun XJ, Gong Q, Chen J, Zhao G, Liu Z, Quon MJ, Dong L, Rajagopalan S, Zhong J. Dipeptidyl Peptidase 4/Midline-1 Axis Promotes T Lymphocyte Motility in Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204194. [PMID: 36683148 PMCID: PMC10037965 DOI: 10.1002/advs.202204194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/04/2022] [Indexed: 06/17/2023]
Abstract
T cells play a crucial role in atherosclerosis, with its infiltration preceding the formation of atheroma. However, how T-cell infiltration is regulated in atherosclerosis remains largely unknown. Here, this work demonstrates that dipeptidyl peptidase-4 (DPP4) is a novel regulator of T-cell motility in atherosclerosis. Single-cell ribonucleic acid (RNA) sequencing and flow cytometry show that CD4+ T cells in atherosclerotic patients display a marked increase of DPP4. Lack of DPP4 in hematopoietic cells or T cells reduces T-cell infiltration and atherosclerotic plaque volume in atherosclerosis mouse models. Mechanistically, DPP4 deficiency reduces T-cell motility by suppressing the expression of microtubule associated protein midline-1 (Mid1) in T cells. Deletion of either DPP4 or Mid1 inhibits chemokine-induced shape change and motility, while restitution of Mid1 in Dpp4-/- T cell largely restores its migratory ability. Thus, DPP4/Mid1, as a novel regulator of T-cell motility, may be a potential inflammatory target in atherosclerosis.
Collapse
Affiliation(s)
- Xiaoquan Rao
- Division of CardiologyDepartment of Internal MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
- Cardiovascular Research InstituteCase Western Reserve UniversityClevelandOhio44106USA
| | - Michael Razavi
- Cardiovascular Research InstituteCase Western Reserve UniversityClevelandOhio44106USA
| | - Georgeta Mihai
- Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts02115USA
| | - Yingying Wei
- Cardiovascular Research InstituteCase Western Reserve UniversityClevelandOhio44106USA
| | | | - Matthew B. Frieman
- Department of Microbiology and ImmunologyUniversity of Maryland School of MedicineBaltimoreMaryland21201USA
| | - Xiao Jian Sun
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMaryland21201USA
| | - Quan Gong
- Department of ImmunologySchool of MedicineYangtze UniversityJingzhouHubei434023P. R. China
| | - Jun Chen
- Sinopharm Dongfeng General HospitalHubei University of MedicineShiyanHubei442008P. R. China
| | - Gang Zhao
- Department of CardiologyShandong Provincial Hospital affiliated to Shandong UniversityJinanShandong250021P. R. China
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
- Institute of Allergy and Clinical ImmunologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
| | - Michael J. Quon
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMaryland21201USA
| | - Lingli Dong
- Division of Rheumatology and ImmunologyDepartment of Internal MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
| | - Sanjay Rajagopalan
- Cardiovascular Research InstituteCase Western Reserve UniversityClevelandOhio44106USA
| | - Jixin Zhong
- Cardiovascular Research InstituteCase Western Reserve UniversityClevelandOhio44106USA
- Wexner Medical CenterThe Ohio State UniversityColumbusOhio43210USA
- Institute of Allergy and Clinical ImmunologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
- Division of Rheumatology and ImmunologyDepartment of Internal MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
| |
Collapse
|
24
|
Busnelli M, Manzini S, Colombo A, Franchi E, Lääperi M, Laaksonen R, Chiesa G. Effect of Diets on Plasma and Aorta Lipidome: A Study in the apoE Knockout Mouse Model. Mol Nutr Food Res 2023; 67:e2200367. [PMID: 36419336 DOI: 10.1002/mnfr.202200367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/20/2022] [Indexed: 11/27/2022]
Abstract
SCOPE Specific lipid molecules circulating in plasma at low concentrations have emerged as biomarkers of atherosclerotic risk. The aim of the present study is that of evaluating, in an athero-prone mouse model, how different diets can affect plasma and aorta lipidome. METHODS AND RESULTS Thirty-six apoE knockout mice are divided in three groups and feed 12 weeks with diets differing for cholesterol and fatty acid content. Atherosclerosis is measured at the aortic sinus and aorta. Lipids are quantified in plasma and aorta with mass spectrometry. The cholesterol content of the diets is the main driver of lipid accumulation in plasma and aorta. The fatty acid composition of the diets affects plasma levels both of essential (linoleic acid) and nonessential (myristic and arachidonic acid) ones. Lipidomics show a comparable distribution, in plasma and aorta, of the main lipid components of oxidized LDL, including cholesteryl esters and lysophosphatidylcholines. Interestingly, lactosylceramide, glucosyl/galactosylceramide, and individual ceramide species are found to accumulate in diseased aortic segments. CONCLUSION Both the cholesterol and fatty acid content of the diets profoundly affect plasma lipidome. Aorta lipidome is likewise affected with the accumulation of specific lipids known as markers of atherosclerosis.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | | | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, 02150, Finland.,Finnish Cardiovascular Research Center, University of Tampere, Tampere, 33520, Finland
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| |
Collapse
|
25
|
Effect of Linoleic Acid on Cholesterol Levels in a High-Fat Diet-Induced Hypercholesterolemia Rat Model. Metabolites 2022; 13:metabo13010053. [PMID: 36676979 PMCID: PMC9864559 DOI: 10.3390/metabo13010053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality worldwide, accounting for almost one-third of all deaths. The risk factors for developing this disease include high levels of serum total cholesterol (TC), triglycerides (TG), and low-density lipoprotein (LDL), alongside low levels of high-density lipoprotein (HDL). Dietary linoleic acid has been suggested to reduce these risk factors. This study aims to determine the effects of linoleic acid on cholesterol levels, liver function tests, and structural changes in liver tissue in comparison with fenofibrate in a hypercholesterolemic rat model. Thirty-six male Sprague Dawley rats (150-180 g) were divided into non-hypercholesterolemic and hypercholesterolemic groups. Hypercholesterolemia was induced in the rats by feeding them with a high-fat diet for two weeks. After two weeks, the non-hypercholesterolemic and hypercholesterolemic rats were equally divided into six groups (n = 6): control non-hypercholesterolemic rats, non-hypercholesterolemic rats treated with fenofibrate (60 mg/kg), non-hypercholesterolemic rats treated with linoleic acid (5 mg/kg), control hypercholesterolemic rats, hypercholesterolemic rats treated with fenofibrate (60 mg/kg), and hypercholesterolemic rats treated with linoleic acid (5 mg/kg). The changes in the rats' body weight, serum lipid profiles, atherogenic indices, and liver function test results were obtained. The rats' liver tissues were stained for histopathological analysis. The linoleic acid-treated hypercholesterolemic rats exhibited significantly reduced serum TC, TG, LDL, aspartate aminotransferase, and alanine aminotransferase levels, as well as increased HDL levels compared with the control hypercholesterolemic rats. These linoleic acid effects were comparable to those in the fenofibrate-treated hypercholesterolemic rats. In conclusion, linoleic acid possesses early anti-hypercholesterolemic properties, which may be due to the reductions in serum cholesterol levels and mild early structural changes in the liver tissues of hypercholesterolemic rats. Therefore, continued studies on linoleic acid in atherosclerotic and/or obese animal models are suggested.
Collapse
|
26
|
Orecchioni M, Wolf D, Suryawanshi V, Winkels H, Kobiyama K, Makings J, Kiosses WB, Ley K. Deleting interleukin-10 from myeloid cells exacerbates atherosclerosis in Apoe -/- mice. Cell Mol Life Sci 2022; 80:10. [PMID: 36496494 PMCID: PMC10141524 DOI: 10.1007/s00018-022-04649-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/05/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is initiated by subendothelial retention of lipoproteins and cholesterol, which triggers a non-resolving inflammatory process that over time leads to plaque progression in the artery wall. Myeloid cells and in particular macrophages are the primary drivers of the inflammatory response and plaque formation. Several immune cells including macrophages, T cells and B cells secrete the anti-inflammatory cytokine IL-10, known to be essential for the atherosclerosis protection. The cellular source of IL-10 in natural atherosclerosis progression is unknown. This study aimed to determine the main IL10-producing cell type in atherosclerosis. To do so, we crossed VertX mice, in which IRES-green fluorescent protein (eGFP) was placed downstream of exon 5 of the Il10 gene, with atherosclerosis-prone Apoe-/- mice. We found that myeloid cells express high levels of IL-10 in VertX Apoe-/- mice in both chow and western-diet fed mice. By single cell RNA sequencing and flow cytometry analysis, we identified resident and inflammatory macrophages in atherosclerotic plaques as the main IL-10 producers. To address whether IL-10 secreted by myeloid cells is essential for the protection, we utilized LyzMCre+Il10fl/fl mice crossed into the Apoe-/- background and confirmed that macrophages were unable to secrete IL-10. Chow and western diet-fed LyzMCre+Il10fl/fl Apoe-/- mice developed significantly larger atherosclerotic plaques as measured by en face morphometry than LyzMCre-Il10 fl/flApoe-/-. Flow cytometry and cytokine measurements suggest that the depletion of IL-10 in myeloid cells increases Th17 cells with elevated CCL2, and TNFα in blood plasma. We conclude that macrophage-derived IL-10 is critical for limiting atherosclerosis in mice.
Collapse
Affiliation(s)
- Marco Orecchioni
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA.
| | - Dennis Wolf
- Cardiology and Angiology I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vasantika Suryawanshi
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jeffrey Makings
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA
| | - William B Kiosses
- Histology and Microscopy Core Facility, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Immunology Center of Georgia (IMMCG), Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
27
|
May L, Bartolo B, Harrison D, Guzik T, Drummond G, Figtree G, Ritchie R, Rye KA, de Haan J. Translating atherosclerosis research from bench to bedside: navigating the barriers for effective preclinical drug discovery. Clin Sci (Lond) 2022; 136:1731-1758. [PMID: 36459456 PMCID: PMC9727216 DOI: 10.1042/cs20210862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. An ongoing challenge remains the development of novel pharmacotherapies to treat CVD, particularly atherosclerosis. Effective mechanism-informed development and translation of new drugs requires a deep understanding of the known and currently unknown biological mechanisms underpinning atherosclerosis, accompanied by optimization of traditional drug discovery approaches. Current animal models do not precisely recapitulate the pathobiology underpinning human CVD. Accordingly, a fundamental limitation in early-stage drug discovery has been the lack of consensus regarding an appropriate experimental in vivo model that can mimic human atherosclerosis. However, when coupled with a clear understanding of the specific advantages and limitations of the model employed, preclinical animal models remain a crucial component for evaluating pharmacological interventions. Within this perspective, we will provide an overview of the mechanisms and modalities of atherosclerotic drugs, including those in the preclinical and early clinical development stage. Additionally, we highlight recent preclinical models that have improved our understanding of atherosclerosis and associated clinical consequences and propose model adaptations to facilitate the development of new and effective treatments.
Collapse
Affiliation(s)
- Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN, U.S.A
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Gemma A. Figtree
- Kolling Research Institute, University of Sydney, Sydney, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney 2052, Australia
| | - Judy B. de Haan
- Cardiovascular Inflammation and Redox Biology Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Department Cardiometabolic Health, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
28
|
Yan C, Kwek E, Ding HF, He Z, Ma KY, Zhu H, Chen ZY. Dietary Oxidized Cholesterol Aggravates Chemically Induced Murine Colon Inflammation and Alters Gut Microbial Ecology. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13289-13301. [PMID: 36198042 DOI: 10.1021/acs.jafc.2c05001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Western diet with a higher intake of fat and cholesterol has been claimed as an intestinal inflammation trigger. Human diet contains both cholesterol and oxidized cholesterol. Oxidized cholesterol has been claimed to be associated with various inflammation diseases, but its effects on colitis and gut microbiome remain largely unknown. The present study was the first time to investigate the effect of the oxidized cholesterol on gut microbiota and dextran sodium sulfate-induced colitis using mice as a model. The results showed that oxidized cholesterol promoted colitis by exacerbating bleeding, body weight decrease, colon shortening, gut barrier damage, oxidative stress, and gut inflammation, whereas non-oxidized cholesterol had no effect. Meanwhile, oxidized cholesterol could adversely modulate the gut microbiota by increasing the relative abundance of pro-inflammatory bacteria (including Escherichia-Shigella and Bacteroides) and decreasing that of beneficial bacteria (Lachnospiraceae_NK4A136_group and Odoribacter). In addition, oxidized cholesterol significantly reduced the production of fecal short-chain fatty acids in colitis mice. It was concluded that oxidized cholesterol was a potential dietary factor of gut dysbiosis.
Collapse
Affiliation(s)
- Chi Yan
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Erika Kwek
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Hua-Fang Ding
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Zouyan He
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
- School of Public Health, Guanxi Medical University, Nanning 530021, China
| | - Ka Ying Ma
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Hanyue Zhu
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
- School of Food Science and Engineering/South China Food Safety Research Center, Foshan University, Foshan 528000, Guangdong, China
| | - Zhen-Yu Chen
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| |
Collapse
|
29
|
Almer G, Opriessnig P, Wolinski H, Sommer G, Diwoky C, Lechleitner M, Kolb D, Bubalo V, Brunner MS, Schwarz AN, Leitinger G, Schoiswohl G, Marsche G, Niedrist T, Schauer S, Oswald W, Groselj-Strele A, Paar M, Cvirn G, Hoefler G, Rechberger GN, Herrmann M, Frank S, Holzapfel GA, Kratky D, Mangge H, Hörl G, Tehlivets O. Deficiency of B vitamins leads to cholesterol-independent atherogenic transformation of the aorta. Biomed Pharmacother 2022; 154:113640. [PMID: 36081286 PMCID: PMC7617128 DOI: 10.1016/j.biopha.2022.113640] [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: 07/05/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022] Open
Abstract
Atherosclerosis, the leading cause of cardiovascular disease responsible for the majority of deaths worldwide, cannot be sufficiently explained by established risk factors, including hypercholesterolemia. Elevated plasma homocysteine is an independent risk factor for atherosclerosis and is strongly linked to cardiovascular mortality. However, the role of homocysteine in atherosclerosis is still insufficiently understood. Previous research in this area has been also hampered by the lack of reproducible in vivo models of atherosclerosis that resemble the human situation. Here, we have developed and applied an automated system for vessel wall injury that leads to more homogenous damage and more pronounced atherosclerotic plaque development, even at low balloon pressure. Our automated system helped to glean vital details of cholesterol-independent changes in the aortic wall of balloon-injured rabbits. We show that deficiency of B vitamins, which are required for homocysteine degradation, leads to atherogenic transformation of the aorta resulting in accumulation of macrophages and lipids, impairment of its biomechanical properties and disorganization of aortic collagen/elastin in the absence of hypercholesterolemia. A combination of B vitamin deficiency and hypercholesterolemia leads to thickening of the aorta, decreased aortic water diffusion, increased LDL-cholesterol and impaired vascular reactivity compared to any single condition. Our findings suggest that deficiency of B vitamins leads to atherogenic transformation of the aorta even in the absence of hypercholesterolemia and aggravates atherosclerosis development in its presence.
Collapse
Affiliation(s)
- Gunter Almer
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Peter Opriessnig
- Division of General Neurology, Department of Neurology, Medical University of Graz, Graz, Austria; Division of Pediatric Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gerhard Sommer
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Clemens Diwoky
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria; Center for Medical Research, Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Vladimir Bubalo
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Markus S Brunner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Andreas N Schwarz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gerd Leitinger
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Gabriele Schoiswohl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Pharmacology, Medical University of Graz, Graz, Austria
| | - Tobias Niedrist
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Silvia Schauer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Wolfgang Oswald
- Department of Surgery, Clinical Division of Vascular Surgery, Medical University of Graz, Graz, Austria
| | - Andrea Groselj-Strele
- Center for Medical Research, Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Margret Paar
- Otto Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Gerhard Cvirn
- Otto Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Markus Herrmann
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria; Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Gerd Hörl
- Otto Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Graz, Austria.
| | - Oksana Tehlivets
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Division of General Radiology, Department of Radiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
30
|
Braczko A, Kutryb-Zajac B, Jedrzejewska A, Krol O, Mierzejewska P, Zabielska-Kaczorowska M, Slominska EM, Smolenski RT. Cardiac Mitochondria Dysfunction in Dyslipidemic Mice. Int J Mol Sci 2022; 23:ijms231911488. [PMID: 36232794 PMCID: PMC9570391 DOI: 10.3390/ijms231911488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Dyslipidemia triggers many severe pathologies, including atherosclerosis and chronic inflammation. Several lines of evidence, including our studies, have suggested direct effects of dyslipidemia on cardiac energy metabolism, but details of these effects are not clear. This study aimed to investigate how mild dyslipidemia affects cardiac mitochondria function and vascular nucleotide metabolism. The analyses were performed in 3- and 6-month-old knock-out mice for low-density lipoprotein receptor (Ldlr−/−) and compared to wild-type C57Bl/6J mice (WT). Cardiac isolated mitochondria function was analyzed using Seahorse metabolic flux analyzer. The mechanical function of the heart was measured using echocardiography. The levels of fusion, fission, and mitochondrial biogenesis proteins were determined by ELISA kits, while the cardiac intracellular nucleotide concentration and vascular pattern of nucleotide metabolism ecto-enzymes were analyzed using reverse-phase high-performance liquid chromatography. We revealed the downregulation of mitochondrial complex I, together with a decreased activity of citrate synthase (CS), reduced levels of nuclear respiratory factor 1 and mitochondrial fission 1 protein, as well as lower intracellular adenosine and guanosine triphosphates’ pool in the hearts of 6-month Ldlr−/− mice vs. age-matched WT. The analysis of vascular ecto-enzyme pattern revealed decreased rate of extracellular adenosine monophosphate hydrolysis and increased ecto-adenosine deaminase activity (eADA) in 6-month Ldlr−/− vs. WT mice. No changes were observed in echocardiography parameters in both age groups of Ldlr−/− mice. Younger hyperlipidemic mice revealed no differences in cardiac mitochondria function, CS activity, intracellular nucleotides, mitochondrial biogenesis, and dynamics but exhibited minor changes in vascular eADA activity vs. WT. This study revealed that dysfunction of cardiac mitochondria develops during prolonged mild hyperlipidemia at the time point corresponding to the formation of early vascular alterations.
Collapse
Affiliation(s)
- Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
| | - Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
- Correspondence: (B.K.-Z.); (R.T.S.); Tel.: +48-58-349-14-14 (B.K.-Z.); +48-58-349-14-60 (R.T.S.)
| | - Agata Jedrzejewska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
| | - Oliwia Krol
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
| | - Paulina Mierzejewska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
| | - Magdalena Zabielska-Kaczorowska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
- Department of Physiology, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Ewa M. Slominska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St., 80-211 Gdansk, Poland
- Correspondence: (B.K.-Z.); (R.T.S.); Tel.: +48-58-349-14-14 (B.K.-Z.); +48-58-349-14-60 (R.T.S.)
| |
Collapse
|
31
|
Scavenger receptor-targeted plaque delivery of microRNA-coated nanoparticles for alleviating atherosclerosis. Proc Natl Acad Sci U S A 2022; 119:e2201443119. [PMID: 36122215 PMCID: PMC9522431 DOI: 10.1073/pnas.2201443119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis treatments by gene regulation are garnering attention, yet delivery of gene cargoes to atherosclerotic plaques remains inefficient. Here, we demonstrate that assembly of therapeutic oligonucleotides into a three-dimensional spherical nucleic acid nanostructure improves their systemic delivery to the plaque and the treatment of atherosclerosis. This noncationic nanoparticle contains a shell of microRNA-146a oligonucleotides, which regulate the NF-κB pathway, for achieving transfection-free cellular entry. Upon an intravenous injection into apolipoprotein E knockout mice fed with a high-cholesterol diet, this nanoparticle naturally targets class A scavenger receptor on plaque macrophages and endothelial cells, contributing to elevated delivery to the plaques (∼1.2% of the injected dose). Repeated injections of the nanoparticle modulate genes related to immune response and vascular inflammation, leading to reduced and stabilized plaques but without inducing severe toxicity. Our nanoparticle offers a safe and effective treatment of atherosclerosis and reveals the promise of nucleic acid nanotechnology for cardiovascular disease.
Collapse
|
32
|
Smith GA, Padmanabhan A, Lau BH, Pampana A, Li L, Lee CY, Pelonero A, Nishino T, Sadagopan N, Xia VQ, Jain R, Natarajan P, Wu RS, Black BL, Srivastava D, Shokat KM, Chorba JS. Cold shock domain-containing protein E1 is a posttranscriptional regulator of the LDL receptor. Sci Transl Med 2022; 14:eabj8670. [PMID: 36103516 PMCID: PMC10174261 DOI: 10.1126/scitranslmed.abj8670] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The low-density lipoprotein receptor (LDLR) controls cellular delivery of cholesterol and clears LDL from the bloodstream, protecting against atherosclerotic heart disease, the leading cause of death in the United States. We therefore sought to identify regulators of the LDLR beyond the targets of current therapies and known causes of familial hypercholesterolemia. We found that cold shock domain-containing protein E1 (CSDE1) enhanced hepatic LDLR messenger RNA (mRNA) decay via its 3' untranslated region and regulated atherogenic lipoproteins in vivo. Using parallel phenotypic genome-wide CRISPR interference screens in a tissue culture model, we identified 40 specific regulators of the LDLR that were not previously identified by observational human genetic studies. Among these, we demonstrated that, in HepG2 cells, CSDE1 regulated the LDLR at least as strongly as statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. In addition, we showed that hepatic gene silencing of Csde1 treated diet-induced dyslipidemia in mice to a similar degree as Pcsk9 silencing. These results suggest the therapeutic potential of targeting CSDE1 to manipulate the posttranscriptional regulation of the LDLR mRNA for the prevention of cardiovascular disease. Our approach of modeling a clinically relevant phenotype in a forward genetic screen, followed by mechanistic pharmacologic dissection and in vivo validation, may serve as a generalizable template for the identification of therapeutic targets in other human disease states.
Collapse
Affiliation(s)
- Geoffrey A Smith
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Arun Padmanabhan
- Division of Cardiology, UCSF Health, San Francisco, CA 94143, USA.,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Bryan H Lau
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Akhil Pampana
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.,Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Li Li
- Department of Medicine and Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clara Y Lee
- Division of Cardiology, UCSF Health, San Francisco, CA 94143, USA.,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Angelo Pelonero
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Tomohiro Nishino
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Nandhini Sadagopan
- Division of Cardiology, UCSF Health, San Francisco, CA 94143, USA.,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Vivian Q Xia
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Division of Cardiology, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
| | - Rajan Jain
- Department of Medicine and Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Cell and Developmental Biology, Institute of Regenerative Medicine, and Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.,Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Roland S Wu
- Division of Cardiology, UCSF Health, San Francisco, CA 94143, USA.,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA.,Departments of Pediatrics and Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA.,Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John S Chorba
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Division of Cardiology, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
| |
Collapse
|
33
|
Luo JY, Cheng CK, He L, Pu Y, Zhang Y, Lin X, Xu A, Lau CW, Tian XY, Ma RCW, Jo H, Huang Y. Endothelial UCP2 Is a Mechanosensitive Suppressor of Atherosclerosis. Circ Res 2022; 131:424-441. [PMID: 35899624 PMCID: PMC9390236 DOI: 10.1161/circresaha.122.321187] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Inflamed endothelial cells (ECs) trigger atherogenesis, especially at arterial regions experiencing disturbed blood flow. UCP2 (Uncoupling protein 2), a key mitochondrial antioxidant protein, improves endothelium-dependent relaxation in obese mice. However, whether UCP2 can be regulated by shear flow is unknown, and the role of endothelial UCP2 in regulating inflammation and atherosclerosis remains unclear. This study aims to investigate the mechanoregulation of UCP2 expression in ECs and the effect of UCP2 on endothelial inflammation and atherogenesis. METHODS In vitro shear stress simulation system was used to investigate the regulation of UCP2 expression by shear flow. EC-specific Ucp2 knockout mice were used to investigate the role of UCP2 in flow-associated atherosclerosis. RESULTS Shear stress experiments showed that KLF2 (Krüppel-like factor 2) mediates fluid shear stress-dependent regulation of UCP2 expression in human aortic and human umbilical vein ECs. Unidirectional shear stress, statins, and resveratrol upregulate whereas oscillatory shear stress and proinflammatory stimuli inhibit UCP2 expression through altered KLF2 expression. KLF2 directly binds to UCP2 promoter to upregulate its transcription in human umbilical vein ECs. UCP2 knockdown induced expression of genes involved in proinflammatory and profibrotic signaling, resulting in a proatherogenic endothelial phenotype. EC-specific Ucp2 deletion promotes atherogenesis and collagen production. Additionally, we found endothelial Ucp2 deficiency aggravates whereas adeno-associated virus-mediated EC-Ucp2 overexpression inhibits carotid atherosclerotic plaque formation in disturbed flow-enhanced atherosclerosis mouse model. RNA-sequencing analysis revealed FoxO1 (forkhead box protein O1) as the major proinflammatory transcriptional regulator activated by UCP2 knockdown, and FoxO1 inhibition reduced vascular inflammation and disturbed flow-enhanced atherosclerosis. We showed further that UCP2 level is critical for phosphorylation of AMPK (AMP-activated protein kinase), which is required for UCP2-induced inhibition of FoxO1. CONCLUSIONS Altogether, our studies uncover that UCP2 is novel mechanosensitive gene under the control of fluid shear stress and KLF2 in ECs. UCP2 expression is critical for endothelial proinflammatory response and atherogenesis. Therapeutic strategies enhancing UCP2 level may have therapeutic potential against atherosclerosis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China (J.-Y.L.)
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Chak Kwong Cheng
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Lei He
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Yujie Pu
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China (Y.Z.)
| | - Xiao Lin
- School of Life Sciences (X.L.), Chinese University of Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, China (A.X.)
| | - Chi Wai Lau
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Xiao Yu Tian
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics (R.C.W.M.), Chinese University of Hong Kong, China
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (H.J.)
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| |
Collapse
|
34
|
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
|
35
|
Domingo-Relloso A, Makhani K, Riffo-Campos AL, Tellez-Plaza M, Klein KO, Subedi P, Zhao J, Moon KA, Bozack AK, Haack K, Goessler W, Umans JG, Best LG, Zhang Y, Herreros-Martinez M, Glabonjat RA, Schilling K, Galvez-Fernandez M, Kent JW, Sanchez TR, Taylor KD, Craig Johnson W, Durda P, Tracy RP, Rotter JI, Rich SS, Berg DVD, Kasela S, Lappalainen T, Vasan RS, Joehanes R, Howard BV, Levy D, Lohman K, Liu Y, Daniele Fallin M, Cole SA, Mann KK, Navas-Acien A. Arsenic Exposure, Blood DNA Methylation, and Cardiovascular Disease. Circ Res 2022; 131:e51-e69. [PMID: 35658476 PMCID: PMC10203287 DOI: 10.1161/circresaha.122.320991] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/18/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Epigenetic dysregulation has been proposed as a key mechanism for arsenic-related cardiovascular disease (CVD). We evaluated differentially methylated positions (DMPs) as potential mediators on the association between arsenic and CVD. METHODS Blood DNA methylation was measured in 2321 participants (mean age 56.2, 58.6% women) of the Strong Heart Study, a prospective cohort of American Indians. Urinary arsenic species were measured using high-performance liquid chromatography coupled to inductively coupled plasma mass spectrometry. We identified DMPs that are potential mediators between arsenic and CVD. In a cross-species analysis, we compared those DMPs with differential liver DNA methylation following early-life arsenic exposure in the apoE knockout (apoE-/-) mouse model of atherosclerosis. RESULTS A total of 20 and 13 DMPs were potential mediators for CVD incidence and mortality, respectively, several of them annotated to genes related to diabetes. Eleven of these DMPs were similarly associated with incident CVD in 3 diverse prospective cohorts (Framingham Heart Study, Women's Health Initiative, and Multi-Ethnic Study of Atherosclerosis). In the mouse model, differentially methylated regions in 20 of those genes and DMPs in 10 genes were associated with arsenic. CONCLUSIONS Differential DNA methylation might be part of the biological link between arsenic and CVD. The gene functions suggest that diabetes might represent a relevant mechanism for arsenic-related cardiovascular risk in populations with a high burden of diabetes.
Collapse
Affiliation(s)
- Arce Domingo-Relloso
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
- Department of Statistics and Operations Research, University of Valencia, Spain
| | - Kiran Makhani
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Angela L. Riffo-Campos
- Millennium Nucleus on Sociomedicine (SocioMed) and Vicerrectoría Académica, Universidad de La Frontera, Temuco, Chile
- Department of Computer Science, ETSE, University of Valencia, Valencia, Spain
| | - Maria Tellez-Plaza
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Kathleen Oros Klein
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Pooja Subedi
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jinying Zhao
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Katherine A. Moon
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anne K. Bozack
- Department of Environmental Health Sciences, School of Public Health, University of California, Berkeley, USA
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Walter Goessler
- Institute of Chemistry - Analytical Chemistry for Health and Environment, University of Graz, Austria
| | | | - Lyle G. Best
- Missouri Breaks Industries and Research Inc., Eagle Butte, SD, USA
| | - Ying Zhang
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, OK, USA
| | | | - Ronald A. Glabonjat
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Kathrin Schilling
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Marta Galvez-Fernandez
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Jack W. Kent
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Tiffany R Sanchez
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Peter Durda
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Russell P. Tracy
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - David Van Den Berg
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Silva Kasela
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Tuuli Lappalainen
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Ramachandran S Vasan
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA; Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Department of Medicine, department of Epidemiology, Boston University Schools of medicine and Public health, Boston, MA, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | | | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | - Kurt Lohman
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Yongmei Liu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - M Daniele Fallin
- Departments of Mental Health and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Koren K. Mann
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
36
|
Ho F, Watson AMD, Elbatreek MH, Kleikers PWM, Khan W, Sourris KC, Dai A, Jha J, Schmidt HHHW, Jandeleit-Dahm KAM. Endothelial reactive oxygen-forming NADPH oxidase 5 is a possible player in diabetic aortic aneurysm but not atherosclerosis. Sci Rep 2022; 12:11570. [PMID: 35798762 PMCID: PMC9262948 DOI: 10.1038/s41598-022-15706-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis and its complications are major causes of cardiovascular morbidity and death. Apart from risk factors such as hypercholesterolemia and inflammation, the causal molecular mechanisms are unknown. One proposed causal mechanism involves elevated levels of reactive oxygen species (ROS). Indeed, early expression of the ROS forming NADPH oxidase type 5 (Nox5) in vascular endothelial cells correlates with atherosclerosis and aortic aneurysm. Here we test the pro-atherogenic Nox5 hypothesis using mouse models. Because Nox5 is missing from the mouse genome, a knock-in mouse model expressing human Nox5 in its physiological location of endothelial cells (eNOX5ki/ki) was tested as a possible new humanised mouse atherosclerosis model. However, whether just on a high cholesterol diet or by crossing in aortic atherosclerosis-prone ApoE−/− mice with and without induction of diabetes, Nox5 neither induced on its own nor aggravated aortic atherosclerosis. Surprisingly, however, diabetic ApoE−/− x eNOX5ki/ki mice developed aortic aneurysms more than twice as often correlating with lower vascular collagens, as assessed by trichrome staining, without changes in inflammatory gene expression, suggesting that endothelial Nox5 directly affects extracellular matrix remodelling associated with aneurysm formation in diabetes. Thus Nox5-derived reactive oxygen species are not a new independent mechanism of atherosclerosis but may enhance the frequency of abdominal aortic aneurysms in the context of diabetes. Together with similar clinical findings, our preclinical target validation opens up a first-in-class mechanism-based approach to treat or even prevent abdominal aortic aneurysms.
Collapse
Affiliation(s)
- Florence Ho
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Anna M D Watson
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, 75 commercial Road, Melbourne, VIC, 3004, Australia
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt. .,Department of Pharmacology and Personalised Medicine, MeHNS, Faculty of Health, Medicine & Life Science, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.
| | - Pamela W M Kleikers
- Department of Pharmacology and Personalised Medicine, MeHNS, Faculty of Health, Medicine & Life Science, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Karly C Sourris
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Aozhi Dai
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Jay Jha
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, Faculty of Health, Medicine & Life Science, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.
| | - Karin A M Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Institute for Clinical Diabetology, German Diabetes Centre, Leibniz Centre for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
| |
Collapse
|
37
|
Shen Y, Gu HM, Zhai L, Wang B, Qin S, Zhang DW. The role of hepatic Surf4 in lipoprotein metabolism and the development of atherosclerosis in apoE -/- mice. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159196. [PMID: 35803528 DOI: 10.1016/j.bbalip.2022.159196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022]
Abstract
Elevated plasma levels of low-density lipoprotein-C (LDL-C) increase the risk of atherosclerotic cardiovascular disease. Circulating LDL is derived from very low-density lipoprotein (VLDL) metabolism and cleared by LDL receptor (LDLR). We have previously demonstrated that cargo receptor Surfeit 4 (Surf4) mediates VLDL secretion. Inhibition of hepatic Surf4 impairs VLDL secretion, significantly reduces plasma LDL-C levels, and markedly mitigates the development of atherosclerosis in LDLR knockout (Ldlr-/-) mice. Here, we investigated the role of Surf4 in lipoprotein metabolism and the development of atherosclerosis in another commonly used mouse model of atherosclerosis, apolipoprotein E knockout (apoE-/-) mice. Adeno-associated viral shRNA was used to silence Surf4 expression mainly in the liver of apoE-/- mice. In apoE-/- mice fed a regular chow diet, knockdown of Surf4 expression significantly reduced triglyceride secretion and plasma levels of non-HDL cholesterol and triglycerides without causing hepatic lipid accumulation or liver damage. When Surf4 was knocked down in apoE-/- mice fed the Western-type diet, we observed a significant reduction in plasma levels of non-HDL cholesterol, but not triglycerides. Knockdown of Surf4 did not increase hepatic cholesterol and triglyceride levels or cause liver damage, but significantly diminished atherosclerosis lesions. Therefore, our findings indicate the potential of hepatic Surf4 inhibition as a novel therapeutic strategy to reduce the risk of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Yishi Shen
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hong-Mei Gu
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lei Zhai
- Institute of Atherosclerosis in Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Binxiang Wang
- Institute of Atherosclerosis in Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Shucun Qin
- Institute of Atherosclerosis in Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China.
| | - Da-Wei Zhang
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
38
|
Sheik SM, Bakthavatchalam P, Shenoy RP, Hadapad BS, Nayak M D, Biswas M, Bolar Suryakanth V. Anti-hyperglycemic, anti-hyperlipidemic, and anti-inflammatory effect of the drug Guggulutiktaka ghrita on high-fat diet-induced obese rats. J Ayurveda Integr Med 2022; 13:100583. [PMID: 35759880 PMCID: PMC9250015 DOI: 10.1016/j.jaim.2022.100583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Background Ayurveda is a holistic system of medicine and describes a vast array of herbs and herbal mixtures that are been demonstrated to possess efficacy in research investigations. Guggulutikthaka gritha (GTG) is one such drug evaluated for its role in skin and bone diseases. Objective In the current study, the hypoglycemic, hypolipidemic, and anti-inflammatory effect of the drug GTG was studied with the scope to treat dyslipidemia and thereby reduce the risk of cardiovascular disease. Materials and method The animals (Wistar rats) were fed a high-fat diet and dyslipidemia was induced. The control group was provided with a normal chow diet and had free access to water. The treatment with the drug GTG was given for 21 days after confirming dyslipidemia. The blood glucose was measured immediately using a glucometer. The serum was analyzed for lipid profile and Vascular Cell Adhesion Molecule – 1(VCAM 1) by ELISA method before and after treatment. The histopathology of the heart and liver was also performed. Results The abnormal change in lipid profile, blood glucose, and inflammatory marker along with the accumulation of intracellular fats in the arteries of the heart and liver confirmed dyslipidemia. A significant reduction in serum lipid profile (p < 0.05), blood glucose (p < 0.05), and VCAM 1 (p < 0.05) was noted after the treatment with significant histopathological changes in arteries of the heart and liver. Conclusion The study provides scientific validation on the drug GTG being effective in hyperglycemia, hyperlipidemia, and inflammation in dyslipidemia.
Collapse
Affiliation(s)
- Samreen M Sheik
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Pugazhandhi Bakthavatchalam
- Department of Anatomy, Melaka Manipal Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Revathi P Shenoy
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Basavaraj S Hadapad
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Deepak Nayak M
- Department of Pathology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Monalisa Biswas
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varashree Bolar Suryakanth
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
39
|
Burger F, Baptista D, Roth A, Brandt KJ, Miteva K. The E3 Ubiquitin Ligase Peli1 Deficiency Promotes Atherosclerosis Progression. Cells 2022; 11:cells11132014. [PMID: 35805095 PMCID: PMC9265341 DOI: 10.3390/cells11132014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Background: Atherosclerosis is a chronic inflammatory vascular disease and the main cause of death and morbidity. Emerging evidence suggests that ubiquitination plays an important role in the pathogenesis of atherosclerosis including control of vascular inflammation, vascular smooth muscle cell (VSMC) function and atherosclerotic plaque stability. Peli1 a type of E3 ubiquitin ligase has emerged as a critical regulator of innate and adaptive immunity, however, its role in atherosclerosis remains to be elucidated. Methods: Apoe−/− mice and Peli1-deficient Apoe−/− Peli1−/− mice were subject to high cholesterol diet. Post sacrifice, serum was collected, and atherosclerotic plaque size and parameters of atherosclerotic plaque stability were evaluated. Immunoprofiling and foam cell quantification were performed. Results: Peli1 deficiency does not affect atherosclerosis lesion burden and cholesterol levels, but promotes VSMCs foam cells formation, necrotic core expansion, collagen, and fibrous cap reduction. Apoe−/− Peli1−/− mice exhibit a storm of inflammatory cytokines, expansion of Th1, Th1, Th17, and Tfh cells, a decrease in regulatory T and B cells and induction of pro-atherogenic serum level of IgG2a and IgE. Conclusions: In the present study, we uncover a crucial role for Peli1 in atherosclerosis as an important regulator of inflammation and VSMCs phenotypic modulation and subsequently atherosclerotic plaque destabilization.
Collapse
|
40
|
Ldlr-Deficient Mice with an Atherosclerosis-Resistant Background Develop Severe Hyperglycemia and Type 2 Diabetes on a Western-Type Diet. Biomedicines 2022; 10:biomedicines10061429. [PMID: 35740449 PMCID: PMC9220196 DOI: 10.3390/biomedicines10061429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 01/10/2023] Open
Abstract
Apoe-/- and Ldlr-/- mice are two animal models extensively used for atherosclerosis research. We previously reported that Apoe-/- mice on certain genetic backgrounds, including C3H/HeJ (C3H), develop type 2 diabetes when fed a Western diet. We sought to characterize diabetes-related traits in C3H-Ldlr-/- mice through comparing with C3H-Apoe-/- mice. On a chow diet, Ldlr-/- mice had lower plasma total and non-HDL cholesterol levels but higher HDL levels than Apoe-/- mice. Fasting plasma glucose was much lower in Ldlr-/- than Apoe-/- mice (male: 122.5 ± 5.9 vs. 229.4 ± 17.5 mg/dL; female: 144.1 ± 12.4 vs. 232.7 ± 6.4 mg/dL). When fed a Western diet, Ldlr-/- and Apoe-/- mice developed severe hypercholesterolemia and also hyperglycemia with fasting plasma glucose levels exceeding 250 mg/dL. Both knockouts had similar non-HDL cholesterol and triglyceride levels, and their fasting glucose levels were also similar. Male Ldlr-/- mice exhibited greater glucose tolerance and insulin sensitivity compared to their Apoe-/- counterpart. Female mice showed similar glucose tolerance and insulin sensitivity though Ldlr-/- mice had higher non-fasting glucose levels. Male Ldlr-/- and Apoe-/- mice developed moderate obesity on the Western diet, but female mice did not. These results indicate that the Western diet and ensuing hyperlipidemia lead to the development of type 2 diabetes, irrespective of underlying genetic causes.
Collapse
|
41
|
La Rose AM, Groenen AG, Halmos B, Bazioti V, Rutten MG, Krishnamurthy KA, Koster MH, Kloosterhuis NJ, Smit M, Havinga R, Mithieux G, Rajas F, Kuipers F, Oosterveer MH, Westerterp M. Increased atherosclerosis in a mouse model of glycogen storage disease type 1a. Mol Genet Metab Rep 2022; 31:100872. [PMID: 35782606 PMCID: PMC9248218 DOI: 10.1016/j.ymgmr.2022.100872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 12/02/2022] Open
Abstract
Glycogen storage disease type 1a (GSD Ia) is an inborn error of carbohydrate metabolism. Despite severe hyperlipidemia, GSD Ia patients show limited atherogenesis compared to age-and-gender matched controls. Employing a GSD Ia mouse model that resembles the severe hyperlipidemia in patients, we here found increased atherogenesis in GSD Ia. These data provide a rationale for investigating atherogenesis in GSD Ia in a larger patient cohort.
Collapse
Affiliation(s)
- Anouk M. La Rose
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anouk G. Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn G.S. Rutten
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kishore A. Krishnamurthy
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rick Havinga
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maaike H. Oosterveer
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Corresponding author at: Department of Pediatrics, University Medical Center Groningen, ERIBA Building 3226 room 04.14, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
42
|
Gaul S, Shahzad K, Medert R, Gadi I, Mäder C, Schumacher D, Wirth A, Ambreen S, Fatima S, Boeckel JN, Khawaja H, Haas J, Brune M, Nawroth PP, Isermann B, Laufs U, Freichel M. Novel Nongenetic Murine Model of Hyperglycemia and Hyperlipidemia-Associated Aggravated Atherosclerosis. Front Cardiovasc Med 2022; 9:813215. [PMID: 35350534 PMCID: PMC8957812 DOI: 10.3389/fcvm.2022.813215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/02/2022] [Indexed: 01/24/2023] Open
Abstract
Objective Atherosclerosis, the main pathology underlying cardiovascular diseases is accelerated in diabetic patients. Genetic mouse models require breeding efforts which are time-consuming and costly. Our aim was to establish a new nongenetic model of inducible metabolic risk factors that mimics hyperlipidemia, hyperglycemia, or both and allows the detection of phenotypic differences dependent on the metabolic stressor(s). Methods and Results Wild-type mice were injected with gain-of-function PCSK9D377Y (proprotein convertase subtilisin/kexin type 9) mutant adeno-associated viral particles (AAV) and streptozotocin and fed either a high-fat diet (HFD) for 12 or 20 weeks or a high-cholesterol/high-fat diet (Paigen diet, PD) for 8 weeks. To evaluate atherosclerosis, two different vascular sites (aortic sinus and the truncus of the brachiocephalic artery) were examined in the mice. Combined hyperlipidemic and hyperglycemic (HGHCi) mice fed a HFD or PD displayed characteristic features of aggravated atherosclerosis when compared to hyperlipidemia (HCi HFD or PD) mice alone. Atherosclerotic plaques of HGHCi HFD animals were larger, showed a less stable phenotype (measured by the increased necrotic core area, reduced fibrous cap thickness, and less α-SMA-positive area) and had more inflammation (increased plasma IL-1β level, aortic pro-inflammatory gene expression, and MOMA-2-positive cells in the BCA) after 20 weeks of HFD. Differences between the HGHCi and HCi HFD models were confirmed using RNA-seq analysis of aortic tissue, revealing that significantly more genes were dysregulated in mice with combined hyperlipidemia and hyperglycemia than in the hyperlipidemia-only group. The HGHCi-associated genes were related to pathways regulating inflammation (increased Cd68, iNos, and Tnfa expression) and extracellular matrix degradation (Adamts4 and Mmp14). When comparing HFD with PD, the PD aggravated atherosclerosis to a greater extent in mice and showed plaque formation after 8 weeks. Hyperlipidemic and hyperglycemic mice fed a PD (HGHCi PD) showed less collagen (Sirius red) and increased inflammation (CD68-positive cells) within aortic plaques than hyperlipidemic mice (HCi PD). HGHCi-PD mice represent a directly inducible hyperglycemic atherosclerosis model compared with HFD-fed mice, in which atherosclerosis is severe by 8 weeks. Conclusion We established a nongenetically inducible mouse model allowing comparative analyses of atherosclerosis in HCi and HGHCi conditions and its modification by diet, allowing analyses of multiple metabolic hits in mice.
Collapse
Affiliation(s)
- Susanne Gaul
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Khurrum Shahzad
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Rebekka Medert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Ihsan Gadi
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Christina Mäder
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Dagmar Schumacher
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Angela Wirth
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Saira Ambreen
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Sameen Fatima
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Jes-Niels Boeckel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Hamzah Khawaja
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Jan Haas
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
- Department of Internal Medicine III, Heidelberg University, Heidelberg, Germany
| | - Maik Brune
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), Heidelberg University, Heidelberg, Germany
| | - Peter P. Nawroth
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), Heidelberg University, Heidelberg, Germany
| | - Berend Isermann
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| |
Collapse
|
43
|
Papanikolaou M, Crump SM, Abbott GW. The focal adhesion protein Testin modulates KCNE2 potassium channel β subunit activity. Channels (Austin) 2021; 15:229-238. [PMID: 33464998 PMCID: PMC7833772 DOI: 10.1080/19336950.2021.1874119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 11/25/2022] Open
Abstract
Coronary Artery Disease (CAD) typically kills more people globally each year than any other single cause of death. A better understanding of genetic predisposition to CAD and the underlying mechanisms will help to identify those most at risk and contribute to improved therapeutic approaches. KCNE2 is a functionally versatile, ubiquitously expressed potassium channel β subunit associated with CAD and cardiac arrhythmia susceptibility in humans and mice. Here, to identify novel KCNE2 interaction partners, we employed yeast two-hybrid screening of adult and fetal human heart libraries using the KCNE2 intracellular C-terminal domain as bait. Testin (encoded by TES), an endothelial cell-expressed, CAD-associated, focal adhesion protein, was identified as a high-confidence interaction partner for KCNE2. We confirmed physical association between KCNE2 and Testin in vitro by co-immunoprecipitation. Whole-cell patch clamp electrophysiology revealed that KCNE2 negative-shifts the voltage dependence and increases the rate of activation of the endothelial cell and cardiomyocyte-expressed Kv channel α subunit, Kv1.5 in CHO cells, whereas Testin did not alter Kv1.5 function. However, Testin nullified KCNE2 effects on Kv1.5 voltage dependence and gating kinetics. In contrast, Testin did not prevent KCNE2 regulation of KCNQ1 gating. The data identify a novel role for Testin as a tertiary ion channel regulatory protein. Future studies will address the potential role for KCNE2-Testin interactions in arterial and myocyte physiology and CAD.
Collapse
Affiliation(s)
- Maria Papanikolaou
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Shawn M. Crump
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
44
|
Lou X, Wang D, Gu Z, Li T, Ren L. Mechanism of microRNA regulating the progress of atherosclerosis in apoE-deficient mice. Bioengineered 2021; 12:10994-11006. [PMID: 34775883 PMCID: PMC8809940 DOI: 10.1080/21655979.2021.2004979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs play important roles in atherosclerogenesis and are important novel pharmaceutic targets in atherosclerosis management. The whole spectrum of miRNAs dysregulation is still under intense investigation. This study intends to identify more novel dysregulated microRNAs in atherosclerotic mice. Half of eight-week-old male ApoE-/- mice were fed with high-fat-diet for 12 weeks as a model mice, and the remaining half of ApoE-/- mice were fed with a normal-diet as a control. A serum lipid profile was performed with ELISA kits, and atherosclerotic lesions were assessed. Aortic tissues were dissected for gene expression profiling using a Multispecies miRNA 4.0 Array, and significant differentially expressed miRNAs were identified with fold change ≥ 2 and p < 0.05. Real-time quantitative PCR was used to validate microarray gene expression data on selected genes. Predicted target genes were extracted and subjected to bioinformatic analysis for molecular function and pathway enrichment analysis. Model mice showed a 15.32% atherosclerotic lesion compared to 1.52% in the control group. A total of 25 significant differentially expressed microRNAs were identified, with most of them (24/25) downregulated. Real-time quantitative PCR confirmed the GeneChip data. Bioinformatic analysis of predicted target genes identified high involvement of the PI3K/Akt/mTOR signaling pathway. Microarray profiling of miRNAs in high-fat-fed Model mice identified 25 differentially expressed miRNAs, including some novel miRNAs, and the PI3K/Akt/mTOR signaling pathway is highly enriched in the predicted target genes. The novel identified dysregulated miRNAs suggest a broader spectrum of miRNA dysregulation in the progression of atherosclerosis and provide more research and therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Xiaoqian Lou
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin, China
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Dawei Wang
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Zehui Gu
- Department of Pathology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Tengteng Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin, China
| |
Collapse
|
45
|
Dandan M, Han J, Mann S, Kim R, Mohammed H, Nyangau E, Hellerstein M. Turnover Rates of the Low-Density Lipoprotein Receptor and PCSK9: Added Dimension to the Cholesterol Homeostasis Model. Arterioscler Thromb Vasc Biol 2021; 41:2866-2876. [PMID: 34615375 DOI: 10.1161/atvbaha.121.316764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We measured the turnover rates of the LDLR (low-density lipoprotein receptor) and PCSK9 (proprotein convertase subtilisin/kexin type 9) in mice by metabolic labeling with heavy water and mass spectrometry. Approach and Results: In liver of mice fed high-cholesterol diets, LDLR mRNA levels and synthesis rates were markedly lower with complete suppression of cholesterol synthesis and higher cholesterol content, consistent with the Brown-Goldstein model of tissue cholesterol homeostasis. We observed markedly lower PCSK9 mRNA levels and synthesis rates in liver and lower concentrations and synthesis rates in plasma. Hepatic LDLR half-life (t½) was prolonged, consistent with an effect of reduced PCSK9, and resulted in no reduction in hepatic LDLR content despite reduced mRNA levels and LDLR synthesis rates. These changes in PCSK9 synthesis complement and expand the well-established model of tissue cholesterol homeostasis in mouse liver, in that reduced synthesis and levels of PCSK9 counterbalance lower LDLR synthesis by promoting less LDLR catabolism, thereby maintaining uptake of LDL cholesterol into liver despite high intracellular cholesterol concentrations. CONCLUSIONS Lower hepatic synthesis and secretion of PCSK9, an SREBP2 (sterol response element binding protein) target gene, results in longer hepatic LDLR t½ in response to cholesterol feeding in mice in the face of high intracellular cholesterol content. PCSK9 modulation opposes the canonical lowering of LDLR mRNA and synthesis by cholesterol surplus and preserves LDLR levels. The physiological and therapeutic implications of these opposing control mechanisms over liver LDLR are of interest and may reflect subservience of hepatic cholesterol homeostasis to whole body cholesterol needs.
Collapse
Affiliation(s)
- Mohamad Dandan
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Julia Han
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Sabrina Mann
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Rachael Kim
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Hussein Mohammed
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Edna Nyangau
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Marc Hellerstein
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| |
Collapse
|
46
|
Axmann M, Plochberger B, Mikula M, Weber F, Strobl WM, Stangl H. Plasma Membrane Lipids: An Important Binding Site for All Lipoprotein Classes. MEMBRANES 2021; 11:membranes11110882. [PMID: 34832111 PMCID: PMC8622984 DOI: 10.3390/membranes11110882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Cholesterol is one of the main constituents of plasma membranes; thus, its supply is of utmost importance. This review covers the known mechanisms of cholesterol transfer from circulating lipoprotein particles to the plasma membrane, and vice versa. To achieve homeostasis, the human body utilizes cellular de novo synthesis and extracellular transport particles for supply of cholesterol and other lipids via the blood stream. These lipoprotein particles can be classified according to their density: chylomicrons, very low, low, and high-density lipoprotein (VLDL, LDL, and HDL, respectively). They deliver and receive their lipid loads, most importantly cholesterol, to and from cells by several redundant routes. Defects in one of these pathways (e.g., due to mutations in receptors) usually are not immediately fatal. Several redundant pathways, at least temporarily, compensate for the loss of one or more of them, but the defects trigger systemic diseases, such as atherosclerosis later on. Recently, intracellular membrane–membrane contact sites were shown to be involved in intracellular cholesterol transfer and the plasma membrane itself has been proposed to act as a binding site for lipoprotein-mediated cargo unloading.
Collapse
Affiliation(s)
- Markus Axmann
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Birgit Plochberger
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Mario Mikula
- Center for Pathobiochemistry and Genetics, Institute for Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
| | - Florian Weber
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Witta Monika Strobl
- Center for Pathobiochemistry and Genetics, Institute for Medical Chemistry, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Institute for Medical Chemistry, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
- Correspondence:
| |
Collapse
|
47
|
Zhang Y, Fatima M, Hou S, Bai L, Zhao S, Liu E. Research methods for animal models of atherosclerosis (Review). Mol Med Rep 2021; 24:871. [PMID: 34713295 PMCID: PMC8569513 DOI: 10.3892/mmr.2021.12511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that threatens human health and lives by causing vascular stenosis and plaque rupture. Various animal models have been employed for elucidating the pathogenesis, drug development and treatment validation studies for atherosclerosis. To the best of our knowledge, the species used for atherosclerosis research include mice, rats, hamsters, rabbits, pigs, dogs, non-human primates and birds, among which the most commonly used ones are mice and rabbits. Notably, apolipoprotein E knockout (KO) or low-density lipoprotein receptor KO mice have been the most widely used animal models for atherosclerosis research since the late 20th century. Although the aforementioned animal models can form atherosclerotic lesions, they cannot completely simulate those in humans with respect to lesion location, lesion composition, lipoprotein composition and physiological structure. Hence, an appropriate animal model needs to be selected according to the research purpose. Additionally, it is necessary for atherosclerosis research to include quantitative analysis results of atherosclerotic lesion size and plaque composition. Laboratory animals can provide not only experimental tissues for in vivo studies but also cells needed for in vitro experiments. The present review first summarizes the common animal models and their practical applications, followed by focus on mouse and rabbit models and elucidating the methods to quantify atherosclerotic lesions. Finally, the methods of culturing endothelial cells, macrophages and smooth muscle cells were elucidated in detail and the experiments involved in atherosclerosis research were discussed.
Collapse
Affiliation(s)
- Yali Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Mahreen Fatima
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Siyuan Hou
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Liang Bai
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
48
|
Liu G, Lai P, Guo J, Wang Y, Xian X. Genetically-engineered hamster models: applications and perspective in dyslipidemia and atherosclerosis-related cardiovascular disease. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:92-110. [PMID: 37724074 PMCID: PMC10388752 DOI: 10.1515/mr-2021-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in both developed and developing countries, in which atherosclerosis triggered by dyslipidemia is the major pathological basis. Over the past 40 years, small rodent animals, such as mice, have been widely used for understanding of human atherosclerosis-related cardiovascular disease (ASCVD) with the advantages of low cost and ease of maintenance and manipulation. However, based on the concept of precision medicine and high demand of translational research, the applications of mouse models for human ASCVD study would be limited due to the natural differences in metabolic features between mice and humans even though they are still the most powerful tools in this research field, indicating that other species with biological similarity to humans need to be considered for studying ASCVD in future. With the development and breakthrough of novel gene editing technology, Syrian golden hamster, a small rodent animal replicating the metabolic characteristics of humans, has been genetically modified, suggesting that gene-targeted hamster models will provide new insights into the precision medicine and translational research of ASCVD. The purpose of this review was to summarize the genetically-modified hamster models with dyslipidemia to date, and their potential applications and perspective for ASCVD.
Collapse
Affiliation(s)
- George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Pingping Lai
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Jiabao Guo
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
49
|
Checkouri E, Blanchard V, Meilhac O. Macrophages in Atherosclerosis, First or Second Row Players? Biomedicines 2021; 9:biomedicines9091214. [PMID: 34572399 PMCID: PMC8465019 DOI: 10.3390/biomedicines9091214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages represent a cell type that has been widely described in the context of atherosclerosis since the earliest studies in the 17th century. Their role has long been considered to be preponderant in the onset and aggravation of atherosclerosis, in particular by participating in the establishment of a chronic inflammatory state by the release of pro-inflammatory cytokines and by uncontrolled engorgement of lipids resulting in the formation of foam cells and later of the necrotic core. However, recent evidence from mouse models using an elegant technique of tracing vascular smooth muscle cells (VSMCs) during plaque development revealed that resident VSMCs display impressive plastic properties in response to an arterial injury, allowing them to switch into different cell types within the plaque, including mesenchymal-like cells, macrophage-like cells and osteochondrogenic-like cells. In this review, we oppose the arguments in favor or against the influence of macrophages versus VSMCs in all stages of atherosclerosis including pre-atherosclerosis, formation of lipid-rich foam cells, development of the necrotic core and the fibrous cap as well as calcification and rupture of the plaque. We also analyze the relevance of animal models for the investigation of the pathophysiological mechanisms of atherosclerosis in humans, and discuss potential therapeutic strategies targeting either VSMCs or macrophage to prevent the development of cardiovascular events. Overall, although major findings have been made from animal models, efforts are still needed to better understand and therefore prevent the development of atherosclerotic plaques in humans.
Collapse
Affiliation(s)
- Eloïse Checkouri
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- Habemus Papam, Food Industry, 97470 Saint-Benoit, France
| | - Valentin Blanchard
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- Departments of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Olivier Meilhac
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- CHU de La Réunion, INSERM, CIC1410, 97500 Saint-Pierre, France
- Correspondence: ; Tel.: +33-262-93-8811
| |
Collapse
|
50
|
Tillie RJHA, De Bruijn J, Perales-Patón J, Temmerman L, Ghosheh Y, Van Kuijk K, Gijbels MJ, Carmeliet P, Ley K, Saez-Rodriguez J, Sluimer JC. Partial Inhibition of the 6-Phosphofructo-2-Kinase/Fructose-2,6-Bisphosphatase-3 (PFKFB3) Enzyme in Myeloid Cells Does Not Affect Atherosclerosis. Front Cell Dev Biol 2021; 9:695684. [PMID: 34458258 PMCID: PMC8387953 DOI: 10.3389/fcell.2021.695684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background The protein 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) is a key stimulator of glycolytic flux. Systemic, partial PFKFB3 inhibition previously decreased total plaque burden and increased plaque stability. However, it is unclear which cell type conferred these positive effects. Myeloid cells play an important role in atherogenesis, and mainly rely on glycolysis for energy supply. Thus, we studied whether myeloid inhibition of PFKFB3-mediated glycolysis in Ldlr–/–LysMCre+/–Pfkfb3fl/fl (Pfkfb3fl/fl) mice confers beneficial effects on plaque stability and alleviates cardiovascular disease burden compared to Ldlr–/–LysMCre+/–Pfkfb3wt/wt control mice (Pfkfb3wt/wt). Methods and Results Analysis of atherosclerotic human and murine single-cell populations confirmed PFKFB3/Pfkfb3 expression in myeloid cells, but also in lymphocytes, endothelial cells, fibroblasts and smooth muscle cells. Pfkfb3wt/wt and Pfkfb3fl/fl mice were fed a 0.25% cholesterol diet for 12 weeks. Pfkfb3fl/fl bone marrow-derived macrophages (BMDMs) showed 50% knockdown of Pfkfb3 mRNA. As expected based on partial glycolysis inhibition, extracellular acidification rate as a measure of glycolysis was partially reduced in Pfkfb3fl/fl compared to Pfkfb3wt/wt BMDMs. Unexpectedly, plaque and necrotic core size, as well as macrophage (MAC3), neutrophil (Ly6G) and collagen (Sirius Red) content were unchanged in advanced Pfkfb3fl/fl lesions. Similarly, early lesion plaque and necrotic core size and total plaque burden were unaffected. Conclusion Partial myeloid knockdown of PFKFB3 did not affect atherosclerosis development in advanced or early lesions. Previously reported positive effects of systemic, partial PFKFB3 inhibition on lesion stabilization, do not seem conferred by monocytes, macrophages or neutrophils. Instead, other Pfkfb3-expressing cells in atherosclerosis might be responsible, such as DCs, smooth muscle cells or fibroblasts.
Collapse
Affiliation(s)
- Renée J H A Tillie
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Jenny De Bruijn
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Javier Perales-Patón
- Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany.,Institute of Experimental Medicine and Systems Biology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Yanal Ghosheh
- La Jolla Institute for Immunology, San Diego, CA, United States
| | - Kim Van Kuijk
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Marion J Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven Cancer Institute, KU Leuven, Leuven, Belgium.,State Key Laboratory of Ophthalmology, Zhongshan Opthalmic Center, Sun Yat-sen University, Guangzhou, China.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Klaus Ley
- La Jolla Institute for Immunology, San Diego, CA, United States.,Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany.,Institute of Experimental Medicine and Systems Biology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,British Heart Foundation (BHF) Centre for Cardiovascular Sciences (CVS), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|