1
|
Arya P, Sharma V, Thapliyal S, Sagar R, Singh P. Preclinical models of atherosclerosis: An overview. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:535-542. [PMID: 38629090 PMCID: PMC11017846 DOI: 10.22038/ijbms.2024.74352.16148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/11/2023] [Indexed: 04/19/2024]
Abstract
Atherosclerosis is a primary cause of illness and death globally and its mechanism is still unclear. Different animal models have been created to evaluate the progression of atherosclerosis, allowing researchers to carefully control the circumstances of the experiment as well as the nutrition and environmental risk factors. To investigate the negative effects of various interventions, pathophysiological alterations might be generated utilizing genetic or pharmacological methods. These models' molecular and pathophysiological mechanisms have been clarified through experiments, and they have served as platforms for the creation of new drugs. Different models can be employed to address various research problems, each with its own benefits and drawbacks. In the current review study, various species of atherosclerosis models are discussed, along with the viability of using them in experiments.
Collapse
Affiliation(s)
- Priyanka Arya
- Galgotias College of Pharmacy, Greater Noida, U.P., India
| | - Vikram Sharma
- Galgotias College of Pharmacy, Greater Noida, U.P., India
| | - Surabhi Thapliyal
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh 249203, India
| | | | - Priyanka Singh
- Galgotias College of Pharmacy, Greater Noida, U.P., India
| |
Collapse
|
2
|
Yan J, Yang S, Han L, Ba X, Shen P, Lin W, Li T, Zhang R, Huang Y, Huang Y, Qin K, Wang Y, Tu S, Chen Z. Dyslipidemia in rheumatoid arthritis: the possible mechanisms. Front Immunol 2023; 14:1254753. [PMID: 37954591 PMCID: PMC10634280 DOI: 10.3389/fimmu.2023.1254753] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease, of which the leading cause of death is cardiovascular disease (CVD). The levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c), and high-density lipoprotein cholesterol (HDL-c) in RA decrease especially under hyperinflammatory conditions. It is conflictive with the increased risk of CVD in RA, which is called "lipid paradox". The systemic inflammation may explain this apparent contradiction. The increased systemic proinflammatory cytokines in RA mainly include interleukin-6(IL-6)、interleukin-1(IL-1)and tumor necrosis factor alpha(TNF-α). The inflammation of RA cause changes in the subcomponents and structure of HDL particles, leading to a weakened anti-atherosclerosis function and promoting LDL oxidation and plaque formation. Dysfunctional HDL can further worsen the abnormalities of LDL metabolism, increasing the risk of cardiovascular disease. However, the specific mechanisms underlying lipid changes in RA and increased CVD risk remain unclear. Therefore, this article comprehensively integrates the latest existing literature to describe the unique lipid profile of RA, explore the mechanisms of lipid changes, and investigate the impact of lipid changes on cardiovascular disease.
Collapse
Affiliation(s)
- Jiahui Yan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Sisi Yang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Pan Shen
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weiji Lin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ruiyuan Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yao Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yu Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Traughber CA, Iacano AJ, Neupane K, Khan MR, Opoku E, Nunn T, Prince A, Sangwan N, Hazen SL, Smith JD, Gulshan K. Impavido attenuates inflammation, reduces atherosclerosis, and alters gut microbiota in hyperlipidemic mice. iScience 2023; 26:106453. [PMID: 37020959 PMCID: PMC10067757 DOI: 10.1016/j.isci.2023.106453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/14/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Impavido (Miltefosine) is an FDA-approved drug for treating leishmaniasis and primary amebic meningoencephalitis. We have shown previously that Miltefosine increased cholesterol release and dampened Nlrp3 inflammasome assembly in macrophages. Here, we show that Miltefosine reduced LPS-induced choline uptake by macrophages, and attenuated Nlrp3 inflammasome assembly in mice. Miltefosine-fed mice showed reduced plasma IL-1β in a polymicrobial cecal slurry model of systemic inflammation. Miltefosine-fed mice showed increased reverse cholesterol transport to the plasma, liver, and feces. Hyperlipidemic apoE-/- mice fed with WTD + Miltefosine showed significantly reduced weight gain and markedly reduced atherosclerotic lesions versus mice fed with WTD. The 16S rDNA sequencing and analysis of gut microbiota showed marked alterations in the microbiota profile of Miltefosine-fed hyperlipidemic apoE-/- versus control, with the most notable changes in Romboutsia and Bacteriodes species. Taken together, these data indicate that Miltefosine causes pleiotropic effects on lipid metabolism, inflammasome activity, atherosclerosis, and the gut microbiota.
Collapse
|
4
|
Chagué C, Gautier T, Dal Zuffo L, Pais de Barros J, Wetzel A, Tarris G, Pallot G, Martin L, Valmary‐Degano S, Deckert V, Lagrost L, Daguindau E, Saas P. High-density lipoprotein infusion protects from acute graft-versus-host disease in experimental allogeneic hematopoietic cell transplantation. Am J Transplant 2022; 22:1350-1361. [PMID: 35038785 PMCID: PMC9306461 DOI: 10.1111/ajt.16960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 01/25/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a major limitation of the therapeutic potential of allogeneic hematopoietic cell transplantation. Lipopolysaccharides (LPS) derived from intestinal gram-negative bacteria are well-known aGVHD triggers and amplifiers. Here, we explored the LPS metabolism in aGVHD mouse models using an innovative quantification method. We demonstrated that systemic LPS accumulation after transplantation was due, at least partly, to a defect in its clearance through lipoprotein-mediated transport to the liver (i.e., the so-called reverse LPS transport). After transplantation, reduced circulating HDL concentration impaired LPS neutralization and elimination through biliary flux. Accordingly, HDL-deficient (Apoa1tm1Unc ) recipient mice developed exacerbated aGVHD. Repeated administration of HDL isolated from human plasma significantly decreased the mortality and the severity of aGVHD. While the potential role of HDL in scavenging circulating LPS was examined in this study, it appears that HDL plays a more direct immunomodulatory role by limiting or controlling aGVHD. Notably, HDL infusion mitigated liver aGVHD by diminishing immune infiltration (e.g., interferon-γ-secreting CD8+ T cells and non-resident macrophages), systemic and local inflammation (notably cholangitis). Hence, our results revealed the interest of HDL-based therapies in the prevention of aGVHD.
Collapse
Affiliation(s)
- Cécile Chagué
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance
| | - Thomas Gautier
- University Bourgogne Franche‐ComtéINSERMLNC UMR1231LabEX LipSTICDijonFrance
| | - Ludivine Dal Zuffo
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance
| | | | - Audrey Wetzel
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance
| | - Georges Tarris
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance,Service d’Anatomie et Cytologie PathologiquesCHU DijonDijonFrance
| | - Gaëtan Pallot
- University Bourgogne Franche‐ComtéINSERMLNC UMR1231LabEX LipSTICDijonFrance
| | - Laurent Martin
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance,Service d’Anatomie et Cytologie PathologiquesCHU DijonDijonFrance
| | | | - Valérie Deckert
- University Bourgogne Franche‐ComtéINSERMLNC UMR1231LabEX LipSTICDijonFrance
| | - Laurent Lagrost
- University Bourgogne Franche‐ComtéINSERMLNC UMR1231LabEX LipSTICDijonFrance
| | - Etienne Daguindau
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance,Service d’HématologieCHU BesançonBesançonFrance
| | - Philippe Saas
- University Bourgogne Franche‐ComtéINSERM, EFS BFCUMR1098 RIGHT Interactions Greffon‐Hôte‐Tumeur/Ingénierie Cellulaire et GéniqueLabEX LipSTICFHU INCREASEBesançonFrance
| |
Collapse
|
5
|
Arya P, Bhandari U, Sharma K, Bansal P. Anti-PCSK9 monoclonal antibody attenuates high-fat diet and zymosan-induced vascular inflammation in C57BL/6 mice by modulating TLR2/NF-ƙB signaling pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:577-585. [PMID: 35911646 PMCID: PMC9282737 DOI: 10.22038/ijbms.2022.60467.13404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 05/01/2022] [Indexed: 11/05/2022]
Abstract
Objectives Excess intake of a high-fatty diet (HFD) together with zymosan administration mediates vasculitis response which leads to impaired serum lipid levels and causes arterial stiffness. In the development of new cholesterol-lowering medications, PCSK9 inhibitor (proprotein convertase subtilisin/kexin type 9) is an emerging therapeutic. The goal of the present study was to see whether anti-PCSK9 mAb1 might prevent vasculitis in C57BL/6 mice by blocking TLR2/NF-B activation in HFD and Zymosan-induced vasculitis. Materials and Methods Protein-protein molecular docking was performed to validate the binding affinity of anti-PCSK9 mAb1 against TLR2. Under the experimental study, mice were randomly allocated to the following groups: Group I: standard mice diet (30 days) + Zymosan vehicle (sterile PBS solution of 5mg/ml on 8th day); Group II: HFD (30 days) + Zymosan ( single IP dose 80 mg/kg on day 8th); Group III: HFD+Zymosan + anti-PCSK9 mAb1 (6 mg/kg, s.c. on 10th and 20th days); Group IV: HFD+Zymosan+anti-PCSK9 mAb1 (10 mg/kg, s.c. on 10th and 20th days). Results In comparison with the low dose of anti-PCSK9 mAb1 (6 mg/kg), the high dose of anti-PCSK9 mAb1 (10 mg/kg) together with HFD and Zymosan inhibited vasculitis more effectively by decreasing aortic TLR2 and NF-B levels, reducing serum TNF- and IL-6, and up-regulating liver LDLR levels, which down-regulated serum LDL-C and improved serum lipids levels. Histopathological studies showed that anti-PCSK9 mAb1 treatment reduced plaque accumulation in the aorta of mice. Conclusion These findings indicate that anti-PCSK9 mAb1 has therapeutic potential in reducing HFD and Zymosan-induced vascular inflammation.
Collapse
Affiliation(s)
- Priyanka Arya
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi - 110062, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi - 110062, India,Corresponding author: Uma Bhandari. Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi - 110062, India.
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi-110017, India
| | - Priyanka Bansal
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi - 110062, India
| |
Collapse
|
6
|
Diab A, Valenzuela Ripoll C, Guo Z, Javaheri A. HDL Composition, Heart Failure, and Its Comorbidities. Front Cardiovasc Med 2022; 9:846990. [PMID: 35350538 PMCID: PMC8958020 DOI: 10.3389/fcvm.2022.846990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Although research on high-density lipoprotein (HDL) has historically focused on atherosclerotic coronary disease, there exists untapped potential of HDL biology for the treatment of heart failure. Anti-oxidant, anti-inflammatory, and endothelial protective properties of HDL could impact heart failure pathogenesis. HDL-associated proteins such as apolipoprotein A-I and M may have significant therapeutic effects on the myocardium, in part by modulating signal transduction pathways and sphingosine-1-phosphate biology. Furthermore, because heart failure is a complex syndrome characterized by multiple comorbidities, there are complex interactions between heart failure, its comorbidities, and lipoprotein homeostatic mechanisms. In this review, we will discuss the effects of heart failure and associated comorbidities on HDL, explore potential cardioprotective properties of HDL, and review novel HDL therapeutic targets in heart failure.
Collapse
|
7
|
Huang R, Hu Z, Chen X, Cao Y, Li H, Zhang H, Li Y, Liang L, Feng Y, Wang Y, Su W, Kong Z, Melgiri ND, Jiang L, Li X, Du J, Chen Y. The Transcription Factor SUB1 Is a Master Regulator of the Macrophage TLR Response in Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004162. [PMID: 34378353 PMCID: PMC8498911 DOI: 10.1002/advs.202004162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/24/2021] [Indexed: 12/26/2022]
Abstract
Toll-like receptor 2 and 4 (TLR2, TLR4) signaling is implicated in atherosclerotic plaque formation. The two-stage master regulator Virtual Inference of Protein-activity by Enriched Regulon (VIPER) analysis of macrophage TLR2 and TLR4 signature genes integrated with coexpression network genes derived from 371 patient-derived carotid specimens identifies activated RNA polymerase II transcriptional coactivator p15 (SUB1/Sub1, PC4) as a master regulon in the atherogenic TLR response. It is found that TLR2 and TLR4 signaling is proinflammatory and proatherosclerotic in chow-fed apolipoprotein E-deficient (ApoE-/- ) mice. Through transgenic myeloid-specific Sub1 knockout in ApoE-/- mice, it is discovered that these proatherosclerotic effects of TLR2 and TLR4 signaling are mediated by Sub1. Sub1 knockout in macrophages enhances anti-inflammatory M2 macrophage polarization and cholesterol efflux. Irradiated low density lipoprotein receptor-deficient (Ldlr-/- ) mice transplanted with Sub1-/- murine bone marrow display reduced atherosclerosis. Promoter analysis reveals Sub1-dependent activation of interferon regulatory factor 1 (Irf1) transcription in a casein kinase 2 (Ck2)-dependent manner, and Sub1-knockout macrophages display decreased Irf1 expression. Artificial Irf1 overexpression in Sub1-knockout macrophages enhances proinflammatory M1 skewing and lowers cholesterol clearance. In conclusion, the TLR master regulon Sub1, and its downstream effect on the transcription factor Irf1, promotes a proinflammatory M1 macrophage phenotype and enhances atherosclerotic burden in vivo.
Collapse
Affiliation(s)
- Rongzhong Huang
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zicheng Hu
- Institute of Ultrasound ImagingThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaorui Chen
- Department of Pulmonary and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Yu Cao
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Hongrong Li
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Hong Zhang
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Yongyong Li
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Liwen Liang
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Yuxing Feng
- Department of Rehabilitation and Pain MedicineThe Ninth People's Hospital of ChongqingChongqing400700China
| | - Ying Wang
- Department of Rehabilitation MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Wenhua Su
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Zerui Kong
- Department of Cardiothoracic SurgeryThe Affiliated Yan An Hospital of Kunming Medical UniversityKunming650000China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunming650500China
| | - ND Melgiri
- Impactys Foundation for Biomedical ResearchSan DiegoCA92121USA
| | - Lihong Jiang
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Xingsheng Li
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Jianlin Du
- Department of CardiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Yunqing Chen
- Department of CardiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| |
Collapse
|
8
|
Arya P, Nabi S, Bhandari U. Modulatory role of atorvastatin against high-fat diet and zymosan-induced activation of TLR2/NF-ƙB signaling pathway in C57BL/6 mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1023-1032. [PMID: 34804419 PMCID: PMC8591763 DOI: 10.22038/ijbms.2021.55460.12409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/11/2021] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Accumulated evidence provides a strong connection between the immune system and vascular inflammation. The innate immune system's main sensors are toll-like receptors (TLRs). Zymosan (Zym), a fungal product, induces an inflammatory response via activating TLR2 of the immune system. Atorvastatin, a potent statin, possesses pleiotropic effects including immunomodulatory, lipid-lowering, and anti-inflammatory. Therefore, the current study aimed to evaluate the protective role of atorvastatin against a high-fat diet (HFD) and Zym-induced vascular inflammation in C57BL/6 mice via modulation of TLR2/NF-ƙB signaling pathway. MATERIALS AND METHODS In silico study was conducted to confirm the binding affinity of atorvastatin against TLR2. Under in vivo study, mice were divided into four groups: Normal control: normal standard chow-diet fed for 30 days + Zym vehicle (sterile PBS, 5 mg/ml on 8th day); HFD (30 days) + Zym (80 mg/kg, IP, on 8th day); HFD/Zym + atorvastatin vehicle (0.5% CMC, p.o., from 10th to 30th day); HFD/Zym + atorvastatin (3.6 mg/kg, p.o., from 10th to 30th day). RESULTS Atorvastatin treatment along with HFD and Zym inhibited vascular inflammation by suppressing the levels of aortic TLR2, cardiac NF-ƙB and decrease in serum TNF-α and IL-6. Further, there was an increase in hepatic LDLR levels, resulting in a decrease in serum LDL-C and an increase in HDL-C levels. Histopathological examination of the aorta showed a reduction in plaque accumulation with the atorvastatin-treated group as compared with HFD and Zym-treated group. CONCLUSION Atorvastatin attenuates vascular inflammation mediated by HFD and Zym through suppression of TLR2, NF-ƙB, TNF-α, IL-6, and upregulation of LDLR levels.
Collapse
Affiliation(s)
- Priyanka Arya
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard (UGC approved deemed to be University, Govt. of India), New Delhi- 110062, India
| | - Sayima Nabi
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard (UGC approved deemed to be University, Govt. of India), New Delhi- 110062, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard (UGC approved deemed to be University, Govt. of India), New Delhi- 110062, India
| |
Collapse
|
9
|
Opoku E, Traughber CA, Zhang D, Iacano AJ, Khan M, Han J, Smith JD, Gulshan K. Gasdermin D Mediates Inflammation-Induced Defects in Reverse Cholesterol Transport and Promotes Atherosclerosis. Front Cell Dev Biol 2021; 9:715211. [PMID: 34395445 PMCID: PMC8355565 DOI: 10.3389/fcell.2021.715211] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023] Open
Abstract
Activation of inflammasomes, such as Nlrp3 and AIM2, can exacerbate atherosclerosis in mice and humans. Gasdermin D (GsdmD) serves as a final executor of inflammasome activity, by generating membrane pores for the release of mature Interleukin-1beta (IL-1β). Inflammation dampens reverse cholesterol transport (RCT) and promotes atherogenesis, while anti-IL-1β antibodies were shown to reduce cardiovascular disease in humans. Though Nlrp3/AIM2 and IL-1β nexus is an emerging atherogenic pathway, the direct role of GsdmD in atherosclerosis is not yet fully clear. Here, we used in vivo Nlrp3 inflammasome activation to show that the GsdmD-/- mice release ∼80% less IL-1β vs. Wild type (WT) mice. The GsdmD-/- macrophages were more resistant to Nlrp3 inflammasome mediated reduction in cholesterol efflux, showing ∼26% decrease vs. ∼60% reduction in WT macrophages. GsdmD expression in macrophages exacerbated foam cell formation in an IL-1β dependent fashion. The GsdmD-/- mice were resistant to Nlrp3 inflammasome mediated defect in RCT, with ∼32% reduction in plasma RCT vs. ∼57% reduction in WT mice, ∼17% reduction in RCT to liver vs. 42% in WT mice, and ∼37% decrease in RCT to feces vs. ∼61% in WT mice. The LDLr antisense oligonucleotides (ASO) induced hyperlipidemic mouse model showed the role of GsdmD in promoting atherosclerosis. The GsdmD-/- mice exhibit ∼42% decreased atherosclerotic lesion area in females and ∼33% decreased lesion area in males vs. WT mice. The atherosclerotic plaque-bearing sections stained positive for the cleaved N-terminal fragment of GsdmD, indicating cleavage of GsdmD in atherosclerotic plaques. Our data show that GsdmD mediates inflammation-induced defects in RCT and promotes atherosclerosis.
Collapse
Affiliation(s)
- Emmanuel Opoku
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Cynthia Alicia Traughber
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States,Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States,Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - David Zhang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Amanda J. Iacano
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Mariam Khan
- Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States,Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Juying Han
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Jonathan D. Smith
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Kailash Gulshan
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States,Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States,Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States,*Correspondence: Kailash Gulshan, ;
| |
Collapse
|
10
|
Su X, Zhang G, Cheng Y, Wang B. New insights into the emerging effects of inflammatory response on HDL particles structure and function. Mol Biol Rep 2021; 48:5723-5733. [PMID: 34319542 DOI: 10.1007/s11033-021-06553-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
According to the increasing results, it has been well-demonstrated that the chronic inflammatory response, including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism whereby inflammatory response up-regulates the risk of cardio-metabolic disorder disease is multifactorial; furthermore, the alterations in high density lipoprotein (HDL) structure and function which occur under the inflammatory response could play an important modulatory function. On the other hand, the serum concentrations of HDL cholesterol (HDL-C) have been shown to be reduced significantly under inflammatory status with remarked alterations in HDL particles. Nevertheless, the potential mechanism whereby the inflammatory response reduces serum HDL-C levels is not simply defined but reduces apolipoprotein A1 production. The alterations in HDL structure mediated by the inflammatory response has been also confirmed to decrease the ability of HDL particle to play an important role in reverse cholesterol transport and protect the LDL particles from oxidation. Recently, it has been shown that under the inflammatory condition, diverse alterations in HDL structure could be observed which lead to changes in HDL function. In the current review, the emerging effects of inflammatory response on HDL particles structure and function are well-summarized to elucidate the potential mechanism whereby different inflammatory status modulates the pathogenic development of dyslipidemia.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Guoming Zhang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Ye Cheng
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| | - Bin Wang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| |
Collapse
|
11
|
Xie B, He J, Liu Y, Liu T, Liu C. A meta-analysis of HDL cholesterol efflux capacity and concentration in patients with rheumatoid arthritis. Lipids Health Dis 2021; 20:18. [PMID: 33612101 PMCID: PMC7897392 DOI: 10.1186/s12944-021-01444-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/03/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Poor cholesterol efflux capacity (CEC) has been proposed to be an independent risk factor for cardiovascular diseases. However, current evidence is inconsistent, especially in rheumatoid arthritis (RA) patients. This meta-analysis aims to identify whether CEC is impaired or altered by drug therapy in RA. METHODS The PubMed/MEDLINE, Embase, Cochrane Library and ClinicalTrials.gov databases were browsed to identify studies on CEC in RA patients. The searches mainly focused on studies in human subjects that were published before November 14, 2020, without any language restrictions. The effect size was pooled by the standardized mean differences and mean differences (SMD & MD) as well as the corresponding 95% confidence intervals (CIs) in a random or fixed effect model. Heterogeneity across the studies was tested using Cochran's Q test and I2 statistic. Newcastle-Ottawa Scale and the Downs and Black scale (D&B) were applied to evaluate the quality of included studies. The GRADE-system with its 4-grade evidence scale was used to assess the quality of evidence. RESULTS A total of 11 eligible articles, including 6 observational and 5 interventional studies, were retrieved. The pooled results showed that in patients with RA, CEC was not significantly different than in healthy controls (SMD: -0.34, 95% CI: - 0.83 to 0.14), whereas the plasma HDL-C levels was significantly lower (MD: -3.91, 95% CI: - 7.15 to - 0.68). Furthermore, in the before-after studies, the CEC of RA patients (SMD: 0.20, 95% CI: 0.02 to 0.37) increased, but the plasma HDL-C levels (MD: 3.63, 95% CI: - 0.13 to 7.39) remained at a comparable quantity after anti-rheumatic treatment comparing with the baseline. In addition, the funnel plot of included studies displayed a lightly asymmetry, while Egger's and Begg's test did not suggest the existence of publication bias. The quality of evidence was rated according to GRADE as moderate to very low. CONCLUSION The current meta-analysis demonstrated that HDL-mediated CEC can be improved by the early control of inflammation and anti-rheumatic treatment in RA patients, which is independent of the plasma HDL-C levels. However, the results should be interpreted with caution because of low-quality and limited quantity of evidence. Future randomized controlled trials are needed to determine whether therapeutic strategies to enhance CEC in RA patients have beneficial effects for preventing CVD.
Collapse
Affiliation(s)
- Binbin Xie
- Department of Nutrition, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jiang He
- Department of Mathematics and Physics, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yong Liu
- Department of Laboratory Medicine, Hospital of Stomatology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ting Liu
- Department of Nutrition, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Chaoqun Liu
- Department of Nutrition, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
12
|
Arya P, Bhandari U. Involvement of the toll-like receptors-2/nuclear factor-kappa B signaling pathway in atherosclerosis induced by high-fat diet and zymosan A in C57BL/6 mice. Indian J Pharmacol 2020; 52:203-209. [PMID: 32874003 PMCID: PMC7446673 DOI: 10.4103/ijp.ijp_567_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 05/13/2020] [Accepted: 07/13/2020] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE: Accumulated evidence reported a link between the immune system, microbial infection, and the development of atherosclerosis. Excess intake of high-fat diet (HFD) increases blood lipid levels and induces inflammatory pathways whereas zymosan A (Zym), a microbial component, mediates inflammatory response through the stimulation of specific ligand of toll-like receptors (TLRs) of the immune system. The current research work was aimed to evaluate the mechanism behind atherosclerosis mediated by HFD and Zym in C57BL/6 mice. MATERIALS AND METHODS: The mice were orally fed with HFD for 30 days and Zym (80 mg/kg, single intraperitoneal injection on day 8th). On the 31st day, blood was withdrawn from overnight fasted mice by tail vein puncture and estimated for serum lipids and tumor necrosis factor-alpha (TNF-α). Animals were sacrificed, and cardiac, liver, and aortic tissues were isolated for the estimation of cardiac TLR-2, nuclear factor-kappa B (NF-ƙB); hepatic low-density lipoprotein receptors (LDLR); and base of aorta analyzed for histopathology. RESULTS: It was found that HFD and Zym administration increased arterial inflammation directly through modulation of the TLR-2/NF-ƙB pathway, thereby upregulate serum TNF-α, cardiac TLR-2, and NF-ƙB levels. Further, HFD and Zym treatment significantly increased serum lipid levels and marked decrease in LDLR protein expression in the liver when compared to normal control mice. Histopathological analysis showed the formation of atherosclerotic plaque. CONCLUSION: The study is first, to our current knowledge, to demonstrate the involvement of the TLR-2/NF-ƙB signaling pathway in atherosclerosis induced by HFD and Zym in C57BL/6 mice, resulting in increased degradation of LDLR protein, thereby, increasing the serum lipid levels.
Collapse
Affiliation(s)
- Priyanka Arya
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi, India
| |
Collapse
|
13
|
Castro-Alves VC, Shiga TM, Nascimento JROD. Polysaccharides from chayote enhance lipid efflux and regulate NLRP3 inflammasome priming in macrophage-like THP-1 cells exposed to cholesterol crystals. Int J Biol Macromol 2019; 127:502-510. [PMID: 30658148 DOI: 10.1016/j.ijbiomac.2019.01.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/24/2018] [Accepted: 01/10/2019] [Indexed: 01/14/2023]
Abstract
The contribution of dietary fiber to decrease the risk of atherosclerosis may occur through other mechanisms besides the increased excretion of cholesterol. Although macrophages are crucial for lipid clearance, the excessive uptake of cholesterol crystals (CC) by these cells induce NLRP3 inflammasome and foam cell formation. Thus, we investigated whether the water-soluble DF from chayote (WSP) regulate CC-pretreated macrophage-like THP-1 cells. Linkage analysis indicated that WSP is composed mainly of pectic homogalacturonan and highly branched type I rhamnogalacturonan as well as hemicellulosic material including glucomannan, xyloglucan, and glucurono(arabino)xylan. WSP reduced interleukin (IL)-1β and chemokine release in CC-pretreated macrophages. Notably, WSP also reduced lipid accumulation in cells previously exposed to CC. Furthermore, WSP upregulated liver X receptor alpha expression, which may account for increased lipid efflux, and reduced matrix metallopeptidase 9 expression. WSP also reduced active caspase-1 protein levels, and downregulated NLRP3 and IL-1β gene expression in CC-pretreated cells, suggesting that this polysaccharide fraction regulates the priming signals required for NLRP3 inflammasome activation. Thus, WSP regulate lipid efflux and suppress inflammasome priming in macrophages, suggesting that the health benefits of this dietary fiber could go beyond its physical properties on the gastrointestinal tract.
Collapse
Affiliation(s)
- Victor Costa Castro-Alves
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Tânia Misuzu Shiga
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - João Roberto Oliveira do Nascimento
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil; Food and Nutrition Research Center (NAPAN), University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
14
|
Wu X, Li C, Mariyam Z, Jiang P, Zhou M, Zeb F, Haq IU, Chen A, Feng Q. Acrolein-induced atherogenesis by stimulation of hepatic flavin containing monooxygenase 3 and a protection from hydroxytyrosol. J Cell Physiol 2018; 234:475-485. [PMID: 29953618 DOI: 10.1002/jcp.26600] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/15/2018] [Indexed: 12/24/2022]
Abstract
Acrolein, a highly toxic α, β-unsaturated aldehyde, promotes the progression of atherosclerosis in association with inflammatory signaling pathway and reverse cholesterol transport (RCT) process. Additionally, hepatic flavin containing monooxygenase 3 (FMO3) is involved in the pathogenesis of atherosclerosis by regulating cholesterol metabolism. Hydroxytyrosol (HT), as a major phenolic compound in olive oil, exerts anti-inflammatory and anti-atherogenic activities in vitro and animal models. The current study was designed to evaluate whether FMO3 participated in pro-atherogenic process by acrolein and HT showed protective effect during this process. Here, endothelial cells and macrophage Raw264.7 cells were used as the cell models. Following oxidized low-density lipoprotein (OX-LDL) treatment, acrolein exposure promoted foam cells formation in macrophage Raw264.7 cells. The expression of FMO3 and inflammatory makers such as phospho-NF-κB, IL-1β, TNFα as well as IL-6 were significantly increased. However, ATP-binding cassette transporters subfamily A member 1 (ABCA1), a major transporter in RCT process, was repressed by acrolein. In addition, FMO3 knockdown could suppress inflammatory markers and promote ABCA1 expression. Hydroxytyrosol (HT) was observed to reduce lipid accumulation, FMO3 expression as well as inflammatory response. Moreover, it promoted ABCA1 expression. Therefore, our findings indicated that acrolein-enhanced atherogenesis by increasing FMO3 which increased inflammatory responses and decreased ABCA1 in vitro can be alleviated by HT, which may have a therapeutic potential for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyue Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chaofeng Li
- Department of Cardiology, The Second Affiliated Hospital of Southeast University, Nanjing, China
| | - Zahula Mariyam
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Pan Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ming Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Falak Zeb
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ijaz Ul Haq
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Aochang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qing Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Suratanee A, Plaimas K. Network-based association analysis to infer new disease-gene relationships using large-scale protein interactions. PLoS One 2018; 13:e0199435. [PMID: 29949603 PMCID: PMC6021074 DOI: 10.1371/journal.pone.0199435] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/07/2018] [Indexed: 01/02/2023] Open
Abstract
Protein-protein interactions integrated with disease-gene associations represent important information for revealing protein functions under disease conditions to improve the prevention, diagnosis, and treatment of complex diseases. Although several studies have attempted to identify disease-gene associations, the number of possible disease-gene associations is very small. High-throughput technologies have been established experimentally to identify the association between genes and diseases. However, these techniques are still quite expensive, time consuming, and even difficult to perform. Thus, based on currently available data and knowledge, computational methods have served as alternatives to provide more possible associations to increase our understanding of disease mechanisms. Here, a new network-based algorithm, namely, Disease-Gene Association (DGA), was developed to calculate the association score of a query gene to a new possible set of diseases. First, a large-scale protein interaction network was constructed, and the relationship between two interacting proteins was calculated with regard to the disease relationship. Novel plausible disease-gene pairs were identified and statistically scored by our algorithm using neighboring protein information. The results yielded high performance for disease-gene prediction, with an F-measure of 0.78 and an AUC of 0.86. To identify promising candidates of disease-gene associations, the association coverage of genes and diseases were calculated and used with the association score to perform gene and disease selection. Based on gene selection, we identified promising pairs that exhibited evidence related to several important diseases, e.g., inflammation, lipid metabolism, inborn errors, xanthomatosis, cerebellar ataxia, cognitive deterioration, malignant neoplasms of the skin and malignant tumors of the cervix. Focusing on disease selection, we identified target genes that were important to blistering skin diseases and muscular dystrophy. In summary, our developed algorithm is simple, efficiently identifies disease–gene associations in the protein-protein interaction network and provides additional knowledge regarding disease-gene associations. This method can be generalized to other association studies to further advance biomedical science.
Collapse
Affiliation(s)
- Apichat Suratanee
- Department of Mathematics, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
- * E-mail: (AS); (KP)
| | - Kitiporn Plaimas
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- * E-mail: (AS); (KP)
| |
Collapse
|
16
|
Korolenko TA, Johnston TP, Machova E, Bgatova NP, Lykov AP, Goncharova NV, Nescakova Z, Shintyapina AB, Maiborodin IV, Karmatskikh OL. Hypolipidemic effect of mannans from C. albicans serotypes a and B in acute hyperlipidemia in mice. Int J Biol Macromol 2017; 107:2385-2394. [PMID: 29074085 DOI: 10.1016/j.ijbiomac.2017.10.111] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/31/2022]
Abstract
Mannans, which are biological macromolecules of polysaccharide origin and function as immunomodulators, have been shown to stimulate macrophages in vivo by interaction with the mannose receptor. Thus, they can be used to stimulate macrophages in order to effectively remove circulating atherogenic lipoproteins. Our primary aim was to evaluate the hypolipidemic potential of mannans from C. albicans serotype A (mannan A) and serotype B (mannan B) in a murine model of hyperlipidemia. Mannan A and mannan B were shown to significantly (p<0.05) stimulate both the proliferation (p <0.05) and nitric oxide production of murine peritoneal macrophages in vitro. Pre-treatment of CBA/Lac mice with mannan A prior to induction of hyperlipidemia significantly (p<0.001) reduced serum atherogenic LDL-cholesterol, total cholesterol, and triglycerides. Mannan B exhibited a similar, but more potent, hypolipidemic effect. Electron microscopic analysis of liver revealed a significant (p<0.001) decrease in the volume of lipid droplets when hyperlipidemic mice were pretreated by both mannans. In conclusion, our findings would suggest that both polysaccharide-based biological macromolecules evaluated in the present study, specifically, the natural immunomodulators (mannans A and B), appeared to function as effective lipid-lowering macromolecules, which could potentially serve as adjunct therapy to more conventional hypolipidemic medications such as a statin drug.
Collapse
Affiliation(s)
- T A Korolenko
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.
| | - T P Johnston
- Division of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO, United States.
| | - E Machova
- Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - N P Bgatova
- Scientific Institute of Clinical and Experimental Lymphology-filial of the Institute of Cytology and Genetic Siberian Branch of Russian Academy of Science, Novosibirsk, Russia.
| | - A P Lykov
- Scientific Institute of Clinical and Experimental Lymphology-filial of the Institute of Cytology and Genetic Siberian Branch of Russian Academy of Science, Novosibirsk, Russia.
| | - N V Goncharova
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.
| | - Z Nescakova
- Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - A B Shintyapina
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia.
| | - I V Maiborodin
- The Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.
| | - O L Karmatskikh
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.
| |
Collapse
|
17
|
Dumolt JH, Radhakrishnan SK, Moghadasian MH, Le K, Patel MS, Browne RW, Rideout TC. Maternal hypercholesterolemia enhances oxysterol concentration in mothers and newly weaned offspring but is attenuated by maternal phytosterol supplementation. J Nutr Biochem 2017; 52:10-17. [PMID: 29107136 DOI: 10.1016/j.jnutbio.2017.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/02/2017] [Accepted: 09/12/2017] [Indexed: 12/17/2022]
Abstract
In hypercholesterolemic pregnancies, the maternal environment is characterized by excessive levels of atherogenic lipids that may increase cardiovascular disease risk in mothers and their offspring. We examined the influence of maternal hypercholesterolemia and phytosterol (PS) intervention on the concentration and metabolism of oxysterols, bioactive oxygenated cholesterol derivatives that regulate arterial health and lesion progression, in mothers and their newly weaned offspring. Twenty-one female apoE-/- mice were randomly assigned to three different diets throughout gestation and lactation: (1) chow, (2) high cholesterol (CH; 0.15%) and (3) CH with added PS (2%, CH/PS). At the end of the lactation period, mothers and pups were euthanized for serum and hepatic oxysterol analyses, hepatic transcriptional profiling of hepatic sterol regulatory targets and atherosclerosis. Hypercholesterolemic dams and their pups demonstrated increased (P˂.05) serum oxysterols [including 24 hydroxycholesterol (HC), 25HC, 27HC, 7αHC, 7βHC and 7 ketocholesterol)] compared with the chow group that were normalized by maternal PS supplementation. Hepatic oxysterol concentrations followed a similar pattern of response in mothers but were not altered in newly weaned pups. Hepatic mRNA expression suggested a pattern of enhanced abca1/g1 high-density-lipoprotein-mediated efflux but a reduction in biliary abcg5/g8 export in both dams and their pups. Although arterial lesions were not apparent in newly weaned pups, CH dams demonstrated enhanced atherosclerosis that was reduced upon PS intervention. These results demonstrate that offspring from hypercholesterolemic pregnancies have enhanced circulating oxysterol concentrations and highlight the potential utility of PS as a lipid-lowering option during hypercholesterolemic pregnancies for which there are currently limited options.
Collapse
Affiliation(s)
- Jerad H Dumolt
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA 14214
| | - Sandhya K Radhakrishnan
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA 14214
| | - Mohammed H Moghadasian
- Department of Human Nutritional Sciences, University of Manitoba, and Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada, RH2 2A6
| | - Khuong Le
- Department of Human Nutritional Sciences, University of Manitoba, and Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada, RH2 2A6
| | - Mulchand S Patel
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA 14214
| | - Richard W Browne
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA 14214
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA 14214.
| |
Collapse
|
18
|
Preventing cardiovascular heart disease: Promising nutraceutical and non-nutraceutical treatments for cholesterol management. Pharmacol Res 2017; 120:219-225. [PMID: 28408313 DOI: 10.1016/j.phrs.2017.04.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/20/2017] [Accepted: 04/07/2017] [Indexed: 11/24/2022]
Abstract
Hypercholesterolemia is one of the major risk factors for the development of cardiovascular disease. Atherosclerosis resulting from hypercholesterolemia causes many serious cardiovascular diseases. Statins are generally accepted as a treatment of choice for lowering low-density lipoprotein (LDL) cholesterol, which reduces coronary heart disease morbidity and mortality. Since statin use can be associated with muscle problems and other adverse symptoms, non-adherence and discontinuation of statin therapy often leads to inadequate control of plasma cholesterol levels and increased cardiovascular risk. Moreover, there is compelling evidence on the presence of still considerable residual cardiovascular risk in statin-treated patients. Ezetimibe improves cholesterol-lowering efficacy and provides mild additional cardiovascular protection when combined with statin treatment. Despite a favorable safety profile compared to statins, ezetimibe-induced cholesterol-lowering is modest when used alone. Hence, there is a critical need to identity additional effective hypolipidemic agents that can be used either in combination with statins, or alone, if statins are not tolerated. Thus, hypolipidemic agents such as proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, apolipoprotein B-100 antisense oligonucleotides, cholesteryl ester transfer protein (CETP) inhibitors, and microsomal triglyceride transfer protein (MTTP) inhibitors, as well as yeast polysaccharides (beta-glucans and mannans) and compounds derived from natural sources (nutraceuticals) such as glucomannans, plant sterols, berberine, and red yeast rice are being used. In this review, we will discuss hypercholesterolemia, its impact on the development of cardiovascular disease (CVD), and the use of yeast polysaccharides, various nutraceuticals, and several therapeutic agents not derived from 'natural' sources, to treat hypercholesterolemia.
Collapse
|
19
|
Relapsing-remitting multiple sclerosis patients display an altered lipoprotein profile with dysfunctional HDL. Sci Rep 2017; 7:43410. [PMID: 28230201 PMCID: PMC5322497 DOI: 10.1038/srep43410] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 01/24/2017] [Indexed: 01/08/2023] Open
Abstract
Lipoproteins modulate innate and adaptive immune responses. In the chronic inflammatory disease multiple sclerosis (MS), reports on lipoprotein level alterations are inconsistent and it is unclear whether lipoprotein function is affected. Using nuclear magnetic resonance (NMR) spectroscopy, we analysed the lipoprotein profile of relapsing-remitting (RR) MS patients, progressive MS patients and healthy controls (HC). We observed smaller LDL in RRMS patients compared to healthy controls and to progressive MS patients. Furthermore, low-BMI (BMI ≤ 23 kg/m2) RRMS patients show increased levels of small HDL (sHDL), accompanied by larger, triglyceride (TG)-rich VLDL, and a higher lipoprotein insulin resistance (LP-IR) index. These alterations coincide with a reduced serum capacity to accept cholesterol via ATP-binding cassette (ABC) transporter G1, an impaired ability of HDL3 to suppress inflammatory activity of human monocytes, and modifications of HDL3’s main protein component ApoA-I. In summary, lipoprotein levels and function are altered in RRMS patients, especially in low-BMI patients, which may contribute to disease progression in these patients.
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The ability of HDL to promote cholesterol efflux from macrophages is a predictor of cardiovascular risk independent of HDL cholesterol levels. However, the molecular determinants of HDL cholesterol efflux capacity (CEC) are largely unknown. RECENT FINDINGS The term HDL defines a heterogeneous population of particles with distinct size, shape, protein, and lipid composition. Cholesterol efflux is mediated by multiple pathways that may be differentially modulated by HDL composition. Furthermore, different subpopulations of HDL particles mediate CEC via specific pathways, but the molecular determinants of CEC, either proteins or lipids, are unclear. Inflammation promotes a profound remodeling of HDL and impairs overall HDL CEC while improving ATP-binding cassette transporter G1-mediated efflux. This review discusses recent findings that connect HDL composition and CEC. SUMMARY Data from recent animal and human studies clearly show that multiple factors associate with CEC including individual proteins, lipid composition, as well as specific particle subpopulations. Although acute inflammation remodels HDL and impairs CEC, chronic inflammation has more subtle effects. Standardization of assays measuring HDL composition and CEC is a necessary prerequisite for understanding the factors controlling HDL CEC. Unraveling these factors may help the development of new therapeutic interventions improving HDL function.
Collapse
Affiliation(s)
| | - Tomas Vaisar
- Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
- Corresponding author: Tomas Vaisar, Diabetes Institute, Department of Medicine, University of Washington, 850 Republican St, Seattle, WA 98109, Ph: (206) 616-4972,
| |
Collapse
|
21
|
Korolenko T, Johnston TP, Lykov AP, Shintyapina AB, Khrapova MV, Goncharova NV, Korolenko E, Bgatova NP, Machova E, Nescakova Z, Sakhno LV. A comparative study of the hypolipidaemic effects of a new polysaccharide, mannan Candida albicans serotype A, and atorvastatin in mice with poloxamer 407-induced hyperlipidaemia. J Pharm Pharmacol 2016; 68:1516-1526. [DOI: 10.1111/jphp.12633] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/10/2016] [Indexed: 01/15/2023]
Abstract
Abstract
Objectives
We evaluated the hypolipidaemic effect of mannan Candida albicans serotype A, relative to atorvastatin, in a mouse model of hyperlipidaemia.
Methods
Mannan serotype A was investigated in vitro and in vivo to determine its effects on macrophage proliferation, nitric oxide (NO) production by cultured macrophages, serum and liver lipids, changes in liver morphology and serum chitotriosidase activity and its expression in the liver.
Key findings
Mannan serotype A stimulates the macrophage proliferation and NO production in murine peritoneal macrophages in vitro. The activity of serum chitotriosidase (an enzyme released from the activated macrophages) was found to be significantly increased in P-407-induced hyperlipidaemic mice pretreated with low-dose mannan compared with mice administered P-407 only. Mannan treatment in mice was shown to significantly increase the chitotriosidase expression in the liver of both non-hyperlipidaemic and P-407-induced hyperlipidaemic mice. Lastly, mice pretreated with mannan before the induction of hyperlipidaemia with P-407 showed a significant reduction in the serum concentration of atherogenic LDL cholesterol, total cholesterol, triglycerides and liver triglycerides.
Conclusions
It is suggested that mannan serotype A, like β-glucan, may represent another hypolipidaemic agent, which could potentially be used as an adjunctive therapy with conventional antihyperlipidaemic drugs (statins and fibrates) in humans.
Collapse
Affiliation(s)
- Tatyana Korolenko
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - Thomas P Johnston
- Division of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Alexander P Lykov
- Scientific Institute of Clinical and Experimental Lymphology, Novosibirsk, Russia
| | | | - Marina V Khrapova
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - Natalya V Goncharova
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | | | - Nataliya P Bgatova
- Scientific Institute of Clinical and Experimental Lymphology, Novosibirsk, Russia
| | - Eva Machova
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Zuzana Nescakova
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ludmila V Sakhno
- Scientific Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Studies have shown that chronic inflammatory disorders, such as rheumatoid arthritis, systemic lupus erythematosus, and psoriasis are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism by which inflammation increases cardiovascular disease is likely multifactorial but changes in HDL structure and function that occur during inflammation could play a role. RECENT FINDINGS HDL levels decrease with inflammation and there are marked changes in HDL-associated proteins. Serum amyloid A markedly increases whereas apolipoprotein A-I, lecithin:cholesterol acyltransferase, cholesterol ester transfer protein, paraoxonase 1, and apolipoprotein M decrease. The exact mechanism by which inflammation decreases HDL levels is not defined but decreases in apolipoprotein A-I production, increases in serum amyloid A, increases in endothelial lipase and secretory phospholipase A2 activity, and decreases in lecithin:cholesterol acyltransferase activity could all contribute. The changes in HDL induced by inflammation reduce the ability of HDL to participate in reverse cholesterol transport and protect LDL from oxidation. SUMMARY During inflammation multiple changes in HDL structure occur leading to alterations in HDL function. In the short term, these changes may be beneficial resulting in an increase in cholesterol in peripheral cells to improve host defense and repair but over the long term these changes may increase the risk of atherosclerosis.
Collapse
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section, Department of Veterans Affairs Medical Center, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
23
|
Kim MH, de Beer MC, Wroblewski JM, Charnigo RJ, Ji A, Webb NR, de Beer FC, van der Westhuyzen DR. Impact of individual acute phase serum amyloid A isoforms on HDL metabolism in mice. J Lipid Res 2016; 57:969-79. [PMID: 27018443 DOI: 10.1194/jlr.m062174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 01/12/2023] Open
Abstract
The acute phase (AP) reactant serum amyloid A (SAA), an HDL apolipoprotein, exhibits pro-inflammatory activities, but its physiological function(s) are poorly understood. Functional differences between SAA1.1 and SAA2.1, the two major SAA isoforms, are unclear. Mice deficient in either isoform were used to investigate plasma isoform effects on HDL structure, composition, and apolipoprotein catabolism. Lack of either isoform did not affect the size of HDL, normally enlarged in the AP, and did not significantly change HDL composition. Plasma clearance rates of HDL apolipoproteins were determined using native HDL particles. The fractional clearance rates (FCRs) of apoA-I, apoA-II, and SAA were distinct, indicating that HDL is not cleared as intact particles. The FCRs of SAA1.1 and SAA2.1 in AP mice were similar, suggesting that the selective deposition of SAA1.1 in amyloid plaques is not associated with a difference in the rates of plasma clearance of the isoforms. Although the clearance rate of SAA was reduced in the absence of the HDL receptor, scavenger receptor class B type I (SR-BI), it remained significantly faster compared with that of apoA-I and apoA-II, indicating a relatively minor role of SR-BI in SAA's rapid clearance. These studies enhance our understanding of SAA metabolism and SAA's effects on AP-HDL composition and catabolism.
Collapse
Affiliation(s)
- Myung-Hee Kim
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536
| | - Maria C de Beer
- Physiology, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Joanne M Wroblewski
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Richard J Charnigo
- Departments of Statistics and Biostatistics, University of Kentucky, Lexington, KY 40506
| | - Ailing Ji
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536 Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, KY 40536
| | - Frederick C de Beer
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Deneys R van der Westhuyzen
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536 Molecular and Cellular Biochemistry, University of Kentucky Medical Center, Lexington, KY 40536
| |
Collapse
|
24
|
Qin L, Zhu N, Ao BX, Liu C, Shi YN, Du K, Chen JX, Zheng XL, Liao DF. Caveolae and Caveolin-1 Integrate Reverse Cholesterol Transport and Inflammation in Atherosclerosis. Int J Mol Sci 2016; 17:429. [PMID: 27011179 PMCID: PMC4813279 DOI: 10.3390/ijms17030429] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 01/18/2023] Open
Abstract
Lipid disorder and inflammation play critical roles in the development of atherosclerosis. Reverse cholesterol transport is a key event in lipid metabolism. Caveolae and caveolin-1 are in the center stage of cholesterol transportation and inflammation in macrophages. Here, we propose that reverse cholesterol transport and inflammation in atherosclerosis can be integrated by caveolae and caveolin-1.
Collapse
Affiliation(s)
- Li Qin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Bao-Xue Ao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Chan Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Ya-Ning Shi
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Ke Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Jian-Xiong Chen
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS 39216, USA.
| | - Xi-Long Zheng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
- Department of Biochemistry & Molecular Biology, the Libin Cardiovascular Institute of Alberta, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada.
| | - Duan-Fang Liao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
25
|
Chen HH, Keyhanian K, Zhou X, Vilmundarson RO, Almontashiri NAM, Cruz SA, Pandey NR, Lerma Yap N, Ho T, Stewart CA, Huang H, Hari A, Geoffrion M, McPherson R, Rayner KJ, Stewart AFR. IRF2BP2 Reduces Macrophage Inflammation and Susceptibility to Atherosclerosis. Circ Res 2015. [PMID: 26195219 DOI: 10.1161/circresaha.114.305777] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RATIONALE Inflammation impairs macrophage cholesterol clearance from vascular tissues and promotes atherosclerosis. Inflammatory macrophages suppress expression of the transcription cofactor interferon regulatory factor 2-binding protein 2 (IRF2BP2), and genetic variants near IRF2BP2 associate with ischemic heart disease progression in humans. OBJECTIVES To test whether IRF2BP2 in macrophages affects atherosclerosis in mice and humans. METHODS AND RESULTS We generated mice that delete IRF2BP2 in macrophages. IRF2BP2-deficient macrophages worsened atherosclerosis in irradiated low-density lipoprotein receptor null-recipient mice and in apolipoprotein E null mice. IRF2BP2-deficient macrophages were inflammatory and had impaired cholesterol efflux because of their inability to activate the cholesterol transporter ABCA1 in response to cholesterol loading. Their expression of the anti-inflammatory transcription factor Krüppel-like factor 2 was markedly reduced. Promoter studies revealed that IRF2BP2 is required for MEF2-dependent activation of Krüppel-like factor 2. Importantly, restoring Krüppel-like factor 2 in IRF2BP2-deficient macrophages attenuated M1 inflammatory and rescued M2 anti-inflammatory gene activation and improved the cholesterol efflux deficit by restoring ABCA1 activation in response to cholesterol loading. In a cohort of 1066 angiographic cases and 1011 controls, homozygous carriers of a deletion polymorphism (rs3045215) in the 3' untranslated region sequence of human IRF2BP2 mRNA had a higher risk of coronary artery disease (recessive model, odds ratio [95% confidence interval]=1.560 [1.179-2.065], P=1.73E-03) and had lower IRF2BP2 (and Krüppel-like factor 2) protein levels in peripheral blood mononuclear cells. The effect of this deletion polymorphism to suppress protein expression was confirmed in luciferase reporter studies. CONCLUSION Ablation of IRF2BP2 in macrophages worsens atherosclerosis in mice, and a deletion variant that lowers IRF2BP2 expression predisposes to coronary artery disease in humans.
Collapse
Affiliation(s)
- Hsiao-Huei Chen
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.).
| | - Kianoosh Keyhanian
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Xun Zhou
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Ragnar O Vilmundarson
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Naif A M Almontashiri
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Shelly A Cruz
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Nihar R Pandey
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Nida Lerma Yap
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Tiffany Ho
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Chloe A Stewart
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Hua Huang
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Aswin Hari
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Michele Geoffrion
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Ruth McPherson
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Katey J Rayner
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.)
| | - Alexandre F R Stewart
- From the Department of Cellular and Molecular Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (H.-H.C., K.K., X.Z., S.A.C., N.R.P., C.A.S., H.H., A.H.); the Department of Biochemistry, Microbiology and Immunology, University of Ottawa, and the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.O.V., N.A.M.A., N.L.Y., T.H., M.G., R.M., K.J.R., A.F.R.S.); and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (H.H.C., R.M., A.F.R.S.).
| |
Collapse
|
26
|
Favari E, Chroni A, Tietge UJF, Zanotti I, Escolà-Gil JC, Bernini F. Cholesterol efflux and reverse cholesterol transport. Handb Exp Pharmacol 2015; 224:181-206. [PMID: 25522988 DOI: 10.1007/978-3-319-09665-0_4] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Both alterations of lipid/lipoprotein metabolism and inflammatory events contribute to the formation of the atherosclerotic plaque, characterized by the accumulation of abnormal amounts of cholesterol and macrophages in the artery wall. Reverse cholesterol transport (RCT) may counteract the pathogenic events leading to the formation and development of atheroma, by promoting the high-density lipoprotein (HDL)-mediated removal of cholesterol from the artery wall. Recent in vivo studies established the inverse relationship between RCT efficiency and atherosclerotic cardiovascular diseases (CVD), thus suggesting that the promotion of this process may represent a novel strategy to reduce atherosclerotic plaque burden and subsequent cardiovascular events. HDL plays a primary role in all stages of RCT: (1) cholesterol efflux, where these lipoproteins remove excess cholesterol from cells; (2) lipoprotein remodeling, where HDL undergo structural modifications with possible impact on their function; and (3) hepatic lipid uptake, where HDL releases cholesterol to the liver, for the final excretion into bile and feces. Although the inverse association between HDL plasma levels and CVD risk has been postulated for years, recently this concept has been challenged by studies reporting that HDL antiatherogenic functions may be independent of their plasma levels. Therefore, assessment of HDL function, evaluated as the capacity to promote cell cholesterol efflux may offer a better prediction of CVD than HDL levels alone. Consistent with this idea, it has been recently demonstrated that the evaluation of serum cholesterol efflux capacity (CEC) is a predictor of atherosclerosis extent in humans.
Collapse
Affiliation(s)
- Elda Favari
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Shen L, Peng H, Peng R, Fan Q, Zhao S, Xu D, Morisseau C, Chiamvimonvat N, Hammock BD. Inhibition of soluble epoxide hydrolase in mice promotes reverse cholesterol transport and regression of atherosclerosis. Atherosclerosis 2015; 239:557-65. [PMID: 25733327 PMCID: PMC4527317 DOI: 10.1016/j.atherosclerosis.2015.02.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 01/15/2015] [Accepted: 02/06/2015] [Indexed: 01/07/2023]
Abstract
Adipose tissue is the body largest free cholesterol reservoir and abundantly expresses ATP binding cassette transporter A1 (ABCA1), which maintains plasma high-density lipoprotein (HDL) levels. HDLs have a protective role in atherosclerosis by mediating reverse cholesterol transport (RCT). Soluble epoxide hydrolase (sEH) is a cytosolic enzyme whose inhibition has various beneficial effects on cardiovascular disease. The sEH is highly expressed in adipocytes, and it converts epoxyeicosatrienoic acids (EETs) into less bioactive dihydroxyeicosatrienoic acids. We previously showed that increasing EETs levels with a sEH inhibitor (sEHI) (t-AUCB) resulted in elevated ABCA1 expression and promoted ABCA1-mediated cholesterol efflux from 3T3-L1 adipocytes. The present study investigates the impacts of t-AUCB in mice deficient for the low density lipoprotein (LDL) receptor (Ldlr(-/-) mice) with established atherosclerotic plaques. The sEH inhibitor delivered in vivo for 4 weeks decreased the activity of sEH in adipose tissue, enhanced ABCA1 expression and cholesterol efflux from adipose depots, and consequently increased HDL levels. Furthermore, t-AUCB enhanced RCT to the plasma, liver, bile and feces. It also showed the reduction of plasma LDL-C levels. Consistently, t-AUCB-treated mice showed reductions in the size of atherosclerotic plaques. These studies establish that raising adipose ABCA1 expression, cholesterol efflux, and plasma HDL levels with t-AUCB treatment promotes RCT, decreasing LDL-C and atherosclerosis regression, suggesting that sEH inhibition may be a promising strategy to treat atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Li Shen
- Department of Cardiology, Internal Medicine, Xiangya Second Hospital, Central South University, Changsha, 410011, PR China
| | - Hongchun Peng
- Department of Orthopaedics and Emergency, Changsha Central Hospital, Changsha, 410011, PR China
| | - Ran Peng
- Department of Cardiology, Internal Medicine, Xiangya Second Hospital, Central South University, Changsha, 410011, PR China
| | - Qingsong Fan
- Department of Pathology, Xiangya Second Hospital, Central South University, Changsha, Hunan Province, 410001, PR China
| | - Shuiping Zhao
- Department of Cardiology, Internal Medicine, Xiangya Second Hospital, Central South University, Changsha, 410011, PR China
| | - Danyan Xu
- Department of Cardiology, Internal Medicine, Xiangya Second Hospital, Central South University, Changsha, 410011, PR China.
| | - Christophe Morisseau
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| |
Collapse
|
28
|
Yassine HN, Trenchevska O, He H, Borges CR, Nedelkov D, Mack W, Kono N, Koska J, Reaven PD, Nelson RW. Serum amyloid a truncations in type 2 diabetes mellitus. PLoS One 2015; 10:e0115320. [PMID: 25607823 PMCID: PMC4301920 DOI: 10.1371/journal.pone.0115320] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 11/21/2014] [Indexed: 12/16/2022] Open
Abstract
Serum Amyloid A (SAA) is an acute phase protein complex consisting of several abundant isoforms. The N- terminus of SAA is critical to its function in amyloid formation. SAA is frequently truncated, either missing an arginine or an arginine-serine dipeptide, resulting in isoforms that may influence the capacity to form amyloid. However, the relative abundance of truncated SAA in diabetes and chronic kidney disease is not known.
Collapse
Affiliation(s)
- Hussein N Yassine
- University of Southern California, Los Angeles, CA, United States of America
| | | | - Huijuan He
- University of Southern California, Los Angeles, CA, United States of America
| | - Chad R Borges
- Arizona State University, Tempe, AZ, United States of America
| | - Dobrin Nedelkov
- Arizona State University, Tempe, AZ, United States of America
| | - Wendy Mack
- University of Southern California, Los Angeles, CA, United States of America
| | - Naoko Kono
- University of Southern California, Los Angeles, CA, United States of America
| | - Juraj Koska
- Phoenix VA Health Care System, Phoenix, AZ, United States of America
| | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, AZ, United States of America
| | | |
Collapse
|
29
|
Berisha SZ, Brubaker G, Kasumov T, Hung KT, DiBello PM, Huang Y, Li L, Willard B, Pollard KA, Nagy LE, Hazen SL, Smith JD. HDL from apoA1 transgenic mice expressing the 4WF isoform is resistant to oxidative loss of function. J Lipid Res 2015; 56:653-664. [PMID: 25561462 DOI: 10.1194/jlr.m056754] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HDL functions are impaired by myeloperoxidase (MPO), which selectively targets and oxidizes human apoA1. We previously found that the 4WF isoform of human apoA1, in which the four tryptophan residues are substituted with phenylalanine, is resistant to MPO-mediated loss of function. The purpose of this study was to generate 4WF apoA1 transgenic mice and compare functional properties of the 4WF and wild-type human apoA1 isoforms in vivo. Male mice had significantly higher plasma apoA1 levels than females for both isoforms of human apoA1, attributed to different production rates. With matched plasma apoA1 levels, 4WF transgenics had a trend for slightly less HDL-cholesterol versus human apoA1 transgenics. While 4WF transgenics had 31% less reverse cholesterol transport (RCT) to the plasma compartment, equivalent RCT to the liver and feces was observed. Plasma from both strains had similar ability to accept cholesterol and facilitate ex vivo cholesterol efflux from macrophages. Furthermore, we observed that 4WF transgenic HDL was partially (∼50%) protected from MPO-mediated loss of function while human apoA1 transgenic HDL lost all ABCA1-dependent cholesterol acceptor activity. In conclusion, the structure and function of HDL from 4WF transgenic mice was not different than HDL derived from human apoA1 transgenic mice.
Collapse
Affiliation(s)
- Stela Z Berisha
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Greg Brubaker
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Takhar Kasumov
- Department of Gastroenterology and Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Kimberly T Hung
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Patricia M DiBello
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ying Huang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ling Li
- Department of Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Belinda Willard
- Department of Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Katherine A Pollard
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Laura E Nagy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| | - Jonathan D Smith
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195.
| |
Collapse
|
30
|
Dolganova OM, Rudina MI, Chrapova MV, Dushkin MI. The effect of cholesterol on macrophage-foam-cell generation upon zymosan-induced inflammation in mice. ACTA ACUST UNITED AC 2014. [DOI: 10.1134/s1990519x14030055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
De Beer MC, Wroblewski JM, Noffsinger VP, Rateri DL, Howatt DA, Balakrishnan A, Ji A, Shridas P, Thompson JC, van der Westhuyzen DR, Tannock LR, Daugherty A, Webb NR, De Beer FC. Deficiency of endogenous acute phase serum amyloid A does not affect atherosclerotic lesions in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2013; 34:255-61. [PMID: 24265416 DOI: 10.1161/atvbaha.113.302247] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although elevated plasma concentrations of serum amyloid A (SAA) are associated strongly with increased risk for atherosclerotic cardiovascular disease in humans, the role of SAA in the pathogenesis of lesion formation remains obscure. Our goal was to determine the impact of SAA deficiency on atherosclerosis in hypercholesterolemic mice. APPROACH AND RESULTS Apolipoprotein E-deficient (apoE(-/-)) mice, either wild type or deficient in both major acute phase SAA isoforms, SAA1.1 and SAA2.1, were fed a normal rodent diet for 50 weeks. Female mice, but not male apoE-/- mice deficient in SAA1.1 and SAA2.1, had a modest increase (22%; P≤0.05) in plasma cholesterol concentrations and a 53% increase in adipose mass compared with apoE-/- mice expressing SAA1.1 and SAA2.1 that did not affect the plasma cytokine levels or the expression of adipose tissue inflammatory markers. SAA deficiency did not affect lipoprotein cholesterol distributions or plasma triglyceride concentrations in either male or female mice. Atherosclerotic lesion areas measured on the intimal surfaces of the arch, thoracic, and abdominal regions were not significantly different between apoE-/- mice deficient in SAA1.1 and SAA2.1 and apoE-/- mice expressing SAA1.1 and SAA2.1 in either sex. To accelerate lesion formation, mice were fed a Western diet for 12 weeks. SAA deficiency had effect neither on diet-induced alterations in plasma cholesterol, triglyceride, or cytokine concentrations nor on aortic atherosclerotic lesion areas in either male or female mice. In addition, SAA deficiency in male mice had no effect on lesion areas or macrophage accumulation in the aortic roots. CONCLUSIONS The absence of endogenous SAA1.1 and 2.1 does not affect atherosclerotic lipid deposition in apolipoprotein E-deficient mice fed either normal or Western diets.
Collapse
Affiliation(s)
- Maria C De Beer
- From the Graduate Center for Nutritional Science (M.C.D.B., J.M.W., V.P.N., A.J., P.S., J.C.T., D.R.v.d.W., L.R.T., N.R.W., F.C.D.B.), Saha Cardiovascular Research Center (M.C.D.B., J.M.W., V.P.N., D.L.R., D.A.H., A.B., A.J., P.S., J.C.T., D.R.v.d.W., L.R.T., A.D., N.R.W., F.C.D.B.), and the Departments of Physiology (M.C.D.B.) and Internal Medicine (J.M.W., V.P.N., D.L.R., D.A.H., A.B., A.J., P.S., J.C.T., D.R.v.d.W., L.R.T., A.D., N.R.W., F.C.D.B.), University of Kentucky Medical Center, Lexington, KY; and Department of Veterans Affairs Medical Center, Lexington, KY (D.R.v.d.W., L.R.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Differential regulation of ABCA1 and macrophage cholesterol efflux by elaidic and oleic acids. Lipids 2013; 48:757-67. [PMID: 23800855 DOI: 10.1007/s11745-013-3808-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
Trans fatty acid consumption is associated with an increased risk of coronary heart disease. This increased risk has been attributed to decreased levels of HDL cholesterol and increased levels of LDL cholesterol. However, the mechanism by which trans fatty acid modulates cholesterol transit remains poorly defined. ATP-binding cassette transporter A1 (ABCA1)-mediated macrophage cholesterol efflux is the rate-limiting step initiating apolipoprotein A-I lipidation. In this study, elaidic acid, the most abundant trans fatty acid in partially hydrogenated vegetable oil, was shown to stabilize macrophage ABCA1 protein levels in comparison to that of its cis fatty acid isomer, oleic acid. The mechanism responsible for the disparate effects of oleic and elaidic acid on ABCA1 levels was through accelerated ABCA1 protein degradation in cells treated with oleic acid. In contrast, no apparent differences were observed in ABCA1 mRNA levels, and only minor changes were observed in Liver X receptor/Retinoic X receptor promoter activity in cells treated with elaidic and oleic acid. Efflux of both tracers and cholesterol mass revealed that elaidic acid slightly increased ABCA1-mediated cholesterol efflux, while oleic acid led to decreased ABCA1-mediated efflux. In conclusion, these studies show that cis and trans structural differences in 18 carbon n-9 monoenoic fatty acids variably impact cholesterol efflux through disparate effects on ABCA1 protein degradation.
Collapse
|
33
|
Activation of TRPV1 prevents OxLDL-induced lipid accumulation and TNF-α-induced inflammation in macrophages: role of liver X receptor α. Mediators Inflamm 2013; 2013:925171. [PMID: 23878415 PMCID: PMC3710635 DOI: 10.1155/2013/925171] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/26/2013] [Indexed: 11/18/2022] Open
Abstract
The transient receptor potential vanilloid type 1 (TRPV1) is crucial in the pathogenesis of atherosclerosis; yet its role and underlying mechanism in the formation of macrophage foam cells remain unclear. Here, we show increased TRPV1 expression in the area of foamy macrophages in atherosclerotic aortas of apolipoprotein E-deficient mice. Exposure of mouse bone-marrow-derived macrophages to oxidized low-density lipoprotein (oxLDL) upregulated the expression of TRPV1. In addition, oxLDL activated TRPV1 and elicited calcium (Ca2+) influx, which were abrogated by the pharmacological TRPV1 antagonist capsazepine. Furthermore, oxLDL-induced lipid accumulation in macrophages was ameliorated by TRPV1 agonists but exacerbated by TRPV1 antagonist. Treatment with TRPV1 agonists did not affect the internalization of oxLDL but promoted cholesterol efflux by upregulating the efflux ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. Moreover, the upregulation of ABC transporters was mainly through liver X receptor α- (LXRα-) dependent regulation of transcription. Moreover, the TNF-α-induced inflammatory response was alleviated by TRPV1 agonists but aggravated by the TRPV1 antagonist and LXRα siRNA in macrophages. Our data suggest that LXRα plays a pivotal role in TRPV1-activation-conferred protection against oxLDL-induced lipid accumulation and TNF-α-induced inflammation in macrophages.
Collapse
|
34
|
|
35
|
Triolo M, Annema W, Dullaart RPF, Tietge UJF. Assessing the functional properties of high-density lipoproteins: an emerging concept in cardiovascular research. Biomark Med 2013; 7:457-72. [DOI: 10.2217/bmm.13.35] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although plasma concentrations of high-density lipoprotein (HDL) cholesterol correlate inversely with the incidence of atherosclerotic cardiovascular disease, results from recent epidemiological, genetic and pharmacological intervention studies resulted in a shift of concept. Rather than HDL cholesterol mass levels, the functionality of HDL particles is increasingly regarded as potentially clinically important. This review provides an overview of four key functional properties of HDL, namely cholesterol efflux and reverse cholesterol transport; antioxidative activities; anti-inflammatory activities; and the ability of HDL to increase vascular nitric oxide production resulting in vasorelaxation. Currently available assays are put into context with different HDL isolation procedures yielding compositional heterogeneity of the particle. Gathered knowledge on the impact of different disease states on HDL function is discussed together with potential underlying causative factors modulating HDL functionalities. In addition, a perspective is provided regarding how a better understanding of the determinants of (dys)functional HDL might impact clinical practice and the future design of rational and specific therapeutic approaches targeting atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Michela Triolo
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Wijtske Annema
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Top Institute Food & Nutrition, Wageningen, The Netherlands
| | - Robin PF Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Uwe JF Tietge
- Top Institute Food & Nutrition, Wageningen, The Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
36
|
The Impairment of Macrophage-to-Feces Reverse Cholesterol Transport during Inflammation Does Not Depend on Serum Amyloid A. J Lipids 2013; 2013:283486. [PMID: 23431457 PMCID: PMC3572687 DOI: 10.1155/2013/283486] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 01/19/2023] Open
Abstract
Studies suggest that inflammation impairs reverse cholesterol transport (RCT). We investigated whether serum amyloid A (SAA) contributes to this impairment using an established macrophage-to-feces RCT model. Wild-type (WT) mice and mice deficient in SAA1.1 and SAA2.1 (SAAKO) were injected intraperitoneally with 3H-cholesterol-labeled J774 macrophages 4 hr after administration of LPS or buffered saline. 3H-cholesterol in plasma 4 hr after macrophage injection was significantly reduced in both WT and SAAKO mice injected with LPS, but this was not associated with a reduced capacity of serum from LPS-injected mice to promote macrophage cholesterol efflux in vitro. Hepatic accumulation of 3H-cholesterol was unaltered in either WT or SAAKO mice by LPS treatment. Radioactivity present in bile and feces of LPS-injected WT mice 24 hr after macrophage injection was reduced by 36% (P < 0.05) and 80% (P < 0.001), respectively. In contrast, in SAAKO mice, LPS did not significantly reduce macrophage-derived 3H-cholesterol in bile, and fecal excretion was reduced by only 45% (P < 0.05). Injection of cholesterol-loaded allogeneic J774 cells, but not syngeneic bone-marrow-derived macrophages, transiently induced SAA in C57BL/6 mice. Our study confirms reports that acute inflammation impairs steps in the RCT pathway and establishes that SAA plays only a minor role in this impairment.
Collapse
|
37
|
Fisher EA, Feig JE, Hewing B, Hazen SL, Smith JD. High-density lipoprotein function, dysfunction, and reverse cholesterol transport. Arterioscler Thromb Vasc Biol 2013; 32:2813-20. [PMID: 23152494 DOI: 10.1161/atvbaha.112.300133] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although high high-density lipoprotein (HDL)-cholesterol levels are associated with decreased cardiovascular risk in epidemiological studies, recent genetic and pharmacological findings have raised doubts about the beneficial effects of HDL. Raising HDL levels in animal models by infusion or overexpression of apolipoprotein A-I has shown clear vascular improvements, such as delayed atherosclerotic lesion progression and accelerated lesion regression, along with increased reverse cholesterol transport. Inflammation and other factors, such as myeloperoxidase-mediated oxidation, can impair HDL production and HDL function, with regard to its reverse cholesterol transport, antioxidant, and anti-inflammatory activities. Thus, tests of HDL function, which have not yet been developed as routine diagnostic assays, may prove useful and be a better predictor of cardiovascular risk than HDL-cholesterol levels.
Collapse
Affiliation(s)
- Edward A Fisher
- Department of Cardiovascular Medicine, New York University School of Medicine, New York, NY 444195, USA
| | | | | | | | | |
Collapse
|
38
|
Hung KT, Berisha SZ, Ritchey BM, Santore J, Smith JD. Red blood cells play a role in reverse cholesterol transport. Arterioscler Thromb Vasc Biol 2012; 32:1460-5. [PMID: 22499994 PMCID: PMC3360517 DOI: 10.1161/atvbaha.112.248971] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Reverse cholesterol transport (RCT) involves the removal of cholesterol from peripheral tissue for excretion in the feces. Here, we determined whether red blood cells (RBCs) can contribute to RCT. METHODS AND RESULTS We performed a series of studies in apolipoprotein AI-deficient mice where the high-density lipoprotein-mediated pathway of RCT is greatly diminished. RBCs carried a higher fraction of whole blood cholesterol than plasma in apolipoprotein AI-deficient mice, and as least as much of the labeled cholesterol derived from injected foam cells appeared in RBCs compared with plasma. To determine whether RBCs mediate RCT to the fecal compartment, we measured RCT in anemic and control apolipoprotein AI-deficient mice and found that anemia decreased RCT to the feces by over 35% after correcting for fecal mass. Transfusion of [(3)H]cholesterol-labeled RBCs led to robust delivery of the labeled cholesterol to the feces in apolipoprotein AI-deficient hosts. In wild-type mice, the majority of the blood cholesterol mass, as well as [(3)H]cholesterol derived from the injected foam cells, was found in plasma, and anemia did not significantly alter RCT to the feces after correction for fecal mass. CONCLUSIONS The RBC cholesterol pool is dynamic and facilitates RCT of peripheral cholesterol to the feces, particularly in the low high-density lipoprotein state.
Collapse
Affiliation(s)
- Kimberly T Hung
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
39
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2012; 19:142-7. [PMID: 22374141 DOI: 10.1097/med.0b013e3283520fe6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Azzam KM, Fessler MB. Crosstalk between reverse cholesterol transport and innate immunity. Trends Endocrinol Metab 2012; 23:169-78. [PMID: 22406271 PMCID: PMC3338129 DOI: 10.1016/j.tem.2012.02.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 01/30/2012] [Accepted: 02/01/2012] [Indexed: 02/06/2023]
Abstract
Although lipid metabolism and host defense are widely considered to be very divergent disciplines, compelling evidence suggests that host cell handling of self- and microbe-derived (e.g. lipopolysaccharide, LPS) lipids may have common evolutionary roots, and that they indeed may be inseparable processes. The innate immune response and the homeostatic network controlling cellular sterol levels are now known to regulate each other reciprocally, with important implications for several common diseases, including atherosclerosis. In the present review we discuss recent discoveries that provide new insight into the bidirectional crosstalk between reverse cholesterol transport and innate immunity, and highlight the broader implications of these findings for the development of therapeutics.
Collapse
Affiliation(s)
- Kathleen M Azzam
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
41
|
Regulation of reverse cholesterol transport - a comprehensive appraisal of available animal studies. Nutr Metab (Lond) 2012; 9:25. [PMID: 22458435 PMCID: PMC3366910 DOI: 10.1186/1743-7075-9-25] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 03/29/2012] [Indexed: 12/31/2022] Open
Abstract
Plasma levels of high density lipoprotein (HDL) cholesterol are strongly inversely correlated to the risk of atherosclerotic cardiovascular disease. A major recognized functional property of HDL particles is to elicit cholesterol efflux and consequently mediate reverse cholesterol transport (RCT). The recent introduction of a surrogate method aiming at determining specifically RCT from the macrophage compartment has facilitated research on the different components and pathways relevant for RCT. The current review provides a comprehensive overview of studies carried out on macrophage-specific RCT including a quick reference guide of available data. Knowledge and insights gained on the regulation of the RCT pathway are summarized. A discussion of methodological issues as well as of the respective relevance of specific pathways for RCT is also included.
Collapse
|
42
|
Inflammation modulates human HDL composition and function in vivo. Atherosclerosis 2012; 222:390-4. [PMID: 22456230 DOI: 10.1016/j.atherosclerosis.2012.02.032] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 01/31/2012] [Accepted: 02/21/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Inflammation may directly impair HDL functions, in particular reverse cholesterol transport (RCT), but limited data support this concept in humans. METHODS AND RESULTS We employed low-dose human endotoxemia to assess the effects of inflammation on HDL and RCT-related parameters in vivo. Endotoxemia induced remodelling of HDL with depletion of pre-β1a HDL particles determined by 2-D gel electrophoresis (-32.2±9.3% at 24 h, p<0.05) as well as small (-23.0±5.1%, p<0.01, at 24 h) and medium (-57.6±8.0% at 16 h, p<0.001) HDL estimated by nuclear magnetic resonance (NMR). This was associated with induction of class II secretory phospholipase A2 (~36 fold increase) and suppression of lecithin:cholesterol acyltransferase activity (-20.8±3.4% at 24 h, p<0.01) and cholesterol ester transfer protein mass (-22.2±6.8% at 24 h, p<0.001). The HDL fraction, isolated following endotoxemia, had reduced capacity to efflux cholesterol in vitro from SR-BI and ABCA1, but not ABCG1 transporter cell models. CONCLUSIONS These data support the concept that "atherogenic-HDL dysfunction" and impaired RCT occur in human inflammatory syndromes, largely independent of changes in plasma HDL-C and ApoA-I levels.
Collapse
|