1
|
Gu Y, Bi X, Liu X, Qian Q, Wen Y, Hua S, Fu Q, Zheng Y, Sun S. Roles of ABCA1 in Chronic Obstructive Pulmonary Disease. COPD 2025; 22:2493701. [PMID: 40302380 DOI: 10.1080/15412555.2025.2493701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the common chronic respiratory diseases, which causes a heavy burden to patients and society. Increasing studies suggest that ABCA1 plays an important role in COPD. ABCA1 belongs to a large class of ATP-binding (ABC) transporters. It is not only involved in the reverse transport of cholesterol, but also in the regulation of apoptosis, pyroptosis, cellular inflammation and cellular immunity. Meanwhile, ABCA1 is involved in several signaling pathways, such as SREBP pathway, LXR pathway, MAPK pathway, p62/mTOR pathway, CTRP1 pathway and so on. In addition, the ABCA1 participates in the disorder of lipid metabolism in COPD by regulating the formation of RCT and HDL, regulates the inflammation of COPD by removing excess cholesterol in macrophages, and promotes the differentiation of COPD phenotype into emphysema type. Accordingly, the ABCA1 may be a therapeutic target for COPD.
Collapse
Affiliation(s)
- Ying Gu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaoqing Bi
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaofei Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Qingqing Qian
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Qiaoli Fu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yuanyuan Zheng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Abulaban AA, Al-Kuraishy HM, Al-Gareeb AI, Ahmed EA, Fawzy MN, Alruwaili M, Alexiou A, Papadakis M, Batiha GES. Role of liver X receptor in multiple sclerosis: A long furtive life behind a barrier. Brain Res Bull 2025; 224:111333. [PMID: 40185420 DOI: 10.1016/j.brainresbull.2025.111333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Liver X receptors (LXRs) are nuclear receptors that function as transcription factors regulating cholesterol metabolism and are implicated in multiple sclerosis (MS) pathogenesis. This mini-review aims to elucidate the potential role of LXRs in MS neuropathology. MS is the most prevalent inflammatory and demyelinating disease of the central nervous system (CNS), impacting both the brain and spinal cord. Furthermore, alterations in brain cholesterol metabolism in MS can modify the functional activity and immune response of LXRs, which are implicated in MS neuropathology. Dysregulation of LXRs and cholesterol homeostasis is associated with the pathogenesis of MS. LXRs play a critical role in regulating the myelination of nerve sheaths, and defects in LXR function may contribute to the progression of MS. LXRs have immunomodulatory effects, including inhibition of the proliferation of lymphocytes, preventing contact of self-antigens to T cells, and regulating the apoptotic process of T cells. LXRs regulate the activity of microglia, which have pro-inflammatory and anti-inflammatory properties involved in immune regulation and clearance of debris as well as the remyelination process. LXRs regulate the functional activity of glial cells and prevent glial cell-mediated neurodegeneration. LXRs have an important role in the regulation of neuroinflammation during MS neuropathology. LXRs may prevent the progression of neuroinflammation in MS by inhibiting the NF-κB and NLRP3 inflammasome signaling pathways. In conclusion, LXRs play a crucial role in MS neuropathology by mitigating neuroinflammation. These findings proposed that LXR agonists, through modulation of cholesterol homeostasis and inflammatory response, could be effective in the management of MS.
Collapse
Affiliation(s)
- Ahmad A Abulaban
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Division of Neurology, King Abdulaziz Medical City, Ministry of the National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq.
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt.
| |
Collapse
|
3
|
Das A, Manna R, Chowdhury D, Sharma D, Bodakhe SH. Oxymatrine impedes Alzheimer's progression via the attenuation of hypercholesterolemia and fibrosis. Metab Brain Dis 2025; 40:187. [PMID: 40244482 DOI: 10.1007/s11011-025-01606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 04/05/2025] [Indexed: 04/18/2025]
Abstract
This study highlights the potential therapeutic benefits of oxymatrine (OMT), a quinolizidine alkaloid found in Sophora flavescens, for Alzheimer's disease (AD). This study connects the dots between metabolic and neuronal origins by exploring the effects of oxymatrine in slowing down hypercholesterolemic and fibrotic changes that contribute to cognitive deficits. In our study, laboratory rats were fed a high-cholesterol diet for eight weeks. Cognitive abilities were assessed weekly using Hebb's Williams Maze and Radial arm mazes. Additionally, intraperitoneal doses of OMT were administered (20 mg/kg, 40 mg/kg, and 80 mg/kg) for 21 days. Furthermore, using ELISA, plasma and brain oxysterols, transforming growth factor β, amyloid β, matrix metalloproteinase- 9, claudin- 5, and ATP Binding Cassette Transporter A1 levels were measured biweekly. High-density lipoprotein, low-density lipoprotein, aspartate aminotransferase, and alanine transaminase levels were estimated using diagnostic kits. The findings demonstrate that The administration of oxymatrine to experimental animals resulted in a dose-dependent synergistic decline in several biomarkers, including oxysterols, transforming growth factor β, amyloid β, matrix metalloproteinase- 9, low-density lipoprotein, aspartate aminotransferase, and alanine transaminase levels. At the same time, a concomitant increase in the levels of Claudin- 5, ATP Binding Cassette transporter A1, high-density lipoprotein, and antioxidants in the same animals was observed, especially at a dose of 80 mg/kg. This study aims to establish a link between metabolic and neural origins by investigating the effects of oxymatrine in reducing the progression of hypercholesterolemia and fibrosis, which contribute to cognitive impairment in AD. The research explores how oxymatrine regulates mediators involved in oxysterol production and fibrotic alterations in AD. Preliminary results suggest that oxymatrine has the potential to significantly delay the development and progression of AD, offering a promising treatment alternative for those affected by the disease. The findings of the present study strongly suggest that OMT effectively retards the progression of AD, which is commonly associated with the intake of high-cholesterol diets. Subsequent investigations ought to examine the molecular mechanisms behind oxymatrine's interaction with oxysterols and lipid metabolism, including sophisticated imaging methodologies and metabolomics. Longitudinal studies are essential to evaluate the long-term efficacy and safety of oxymatrine in both animal models and people. Exploring its possible synergistic effects with current medications may yield more effective therapeutic techniques. Identifying biomarkers for personalised medication may also be beneficial. Clinical trials and research on oxymatrine's potential as a prophylactic medication may yield significant insights.
Collapse
Affiliation(s)
- Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, India
| | - Rahul Manna
- Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, India
| | - Dilip Sharma
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, India.
| |
Collapse
|
4
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
5
|
Wang X, Cheng Z, Tai W, Shi M, Ayazi M, Liu Y, Sun L, Yu C, Fan Z, Guo B, He X, Sun D, Young W, Ren Y. Targeting foamy macrophages by manipulating ABCA1 expression to facilitate lesion healing in the injured spinal cord. Brain Behav Immun 2024; 119:431-453. [PMID: 38636566 DOI: 10.1016/j.bbi.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Spinal cord injury (SCI) triggers a complex cascade of events, including myelin loss, neuronal damage, neuroinflammation, and the accumulation of damaged cells and debris at the injury site. Infiltrating bone marrow derived macrophages (BMDMϕ) migrate to the epicenter of the SCI lesion, where they engulf cell debris including abundant myelin debris to become pro-inflammatory foamy macrophages (foamy Mϕ), participate neuroinflammation, and facilitate the progression of SCI. This study aimed to elucidate the cellular and molecular mechanisms underlying the functional changes in foamy Mϕ and their potential implications for SCI. Contusion at T10 level of the spinal cord was induced using a New York University (NYU) impactor (5 g rod from a height of 6.25 mm) in male mice. ABCA1, an ATP-binding cassette transporter expressed by Mϕ, plays a crucial role in lipid efflux from foamy cells. We observed that foamy Mϕ lacking ABCA1 exhibited increased lipid accumulation and a higher presence of lipid-accumulated foamy Mϕ as well as elevated pro-inflammatory response in vitro and in injured spinal cord. We also found that both genetic and pharmacological enhancement of ABCA1 expression accelerated lipid efflux from foamy Mϕ, reduced lipid accumulation and inhibited the pro-inflammatory response of foamy Mϕ, and accelerated clearance of cell debris and necrotic cells, which resulted in functional recovery. Our study highlights the importance of understanding the pathologic role of foamy Mϕ in SCI progression and the potential of ABCA1 as a therapeutic target for modulating the inflammatory response, promoting lipid metabolism, and facilitating functional recovery in SCI.
Collapse
Affiliation(s)
- Xi Wang
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China; College of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| | - Zhijian Cheng
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Wenjiao Tai
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Mingjun Shi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Yang Liu
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Caiyong Yu
- Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an 710032, China
| | - Bin Guo
- Department of Pathology, Guizhou Medical University, Guiyang 550025, China
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dongming Sun
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Yi Ren
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA.
| |
Collapse
|
6
|
Cusato J, Mulasso A, Ferrara M, Manca A, Antonucci M, Accardo G, Palermiti A, Bianco G, Chiara F, Mula J, Maddalone MG, Tettoni MC, Cuomo S, Trevisan G, Bonora S, Di Perri G, Lupo C, Rainoldi A, D’Avolio A. Studying the Changes in Physical Functioning and Oxidative Stress-Related Molecules in People Living with HIV after Switching from Triple to Dual Therapy. Antioxidants (Basel) 2024; 13:518. [PMID: 38790623 PMCID: PMC11117521 DOI: 10.3390/antiox13050518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Physical activity could increase the production of oxidative stress biomarkers, affecting the metabolism and excretion of antiretroviral drugs and, consequently, the clinical outcome. Nowadays, people living with HIV (PLWH) are mostly switching from triple to dual therapy, but no data are available in terms of physical functioning and oxidative stress. The aim of this study was to evaluate if some antioxidant biomarkers and physical functioning tests could be different according to triple or dual antiretroviral therapy. METHODS PLWH were evaluated at baseline (BL), while treated with three drugs, and six months after the switch to dual therapy. Physical functioning was quantified using validated tools. Mitochondrial and cytosol antioxidant molecules were evaluated through liquid chromatography. RESULTS Twenty-five patients were analyzed. A statistically significant difference between triple and dual therapy was found for mitochondrial glutathione, but not for physical tests. Evaluating differences between physically active and inactive individuals, the following statistically significant differences were suggested, considering triple therapy (mitochondrial n-formyl-methionine p = 0.022, triglycerides p = 0.023) and double therapy (mitochondrial glycine p = 0.035, cytosol glutamic acid p = 0.007, cytosol s-adenosylmethionine p = 0.021). CONCLUSIONS For the first time, this study suggests possible differences in terms of antioxidant molecules and physical functioning in PLWH switching from triple to dual therapy.
Collapse
Affiliation(s)
- Jessica Cusato
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| | - Anna Mulasso
- NeuroMuscolarFunction|Research Group, Department of Medical Sciences, University of Turin, 10128 Turin, Italy; (A.M.); (S.C.); (C.L.); (A.R.)
| | - Micol Ferrara
- ASL Città di Torino, Amedeo di Savoia Hospital, 10149 Turin, Italy; (M.F.); (M.A.); (M.C.T.)
| | - Alessandra Manca
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| | - Miriam Antonucci
- ASL Città di Torino, Amedeo di Savoia Hospital, 10149 Turin, Italy; (M.F.); (M.A.); (M.C.T.)
| | - Guido Accardo
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (G.A.); (G.T.); (S.B.); (G.D.P.)
| | - Alice Palermiti
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| | - Gianluca Bianco
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| | - Francesco Chiara
- Laboratory of Clinical Pharmacology S. Luigi A.O.U., Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole, Orbassano, 10043 Turin, Italy;
| | - Jacopo Mula
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| | - Maria Grazia Maddalone
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| | - Maria Cristina Tettoni
- ASL Città di Torino, Amedeo di Savoia Hospital, 10149 Turin, Italy; (M.F.); (M.A.); (M.C.T.)
| | - Simone Cuomo
- NeuroMuscolarFunction|Research Group, Department of Medical Sciences, University of Turin, 10128 Turin, Italy; (A.M.); (S.C.); (C.L.); (A.R.)
| | - Giulia Trevisan
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (G.A.); (G.T.); (S.B.); (G.D.P.)
| | - Stefano Bonora
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (G.A.); (G.T.); (S.B.); (G.D.P.)
| | - Giovanni Di Perri
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (G.A.); (G.T.); (S.B.); (G.D.P.)
| | - Corrado Lupo
- NeuroMuscolarFunction|Research Group, Department of Medical Sciences, University of Turin, 10128 Turin, Italy; (A.M.); (S.C.); (C.L.); (A.R.)
| | - Alberto Rainoldi
- NeuroMuscolarFunction|Research Group, Department of Medical Sciences, University of Turin, 10128 Turin, Italy; (A.M.); (S.C.); (C.L.); (A.R.)
| | - Antonio D’Avolio
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy; (J.C.); (A.P.); (G.B.); (J.M.); (M.G.M.)
| |
Collapse
|
7
|
Alnaaim SA, Al-Kuraishy HM, Alexiou A, Papadakis M, Saad HM, Batiha GES. Role of Brain Liver X Receptor in Parkinson's Disease: Hidden Treasure and Emerging Opportunities. Mol Neurobiol 2024; 61:341-357. [PMID: 37606719 PMCID: PMC10791998 DOI: 10.1007/s12035-023-03561-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease due to the degeneration of dopaminergic neurons (DNs) in the substantia nigra (SN). The liver X receptor (LXR) is involved in different neurodegenerative diseases. Therefore, the objective of the present review was to clarify the possible role of LXR in PD neuropathology. LXRs are the most common nuclear receptors of transcription factors that regulate cholesterol metabolism and have pleiotropic effects, including anti-inflammatory effects and reducing intracellular cholesterol accumulation. LXRs are highly expressed in the adult brain and act as endogenous sensors for intracellular cholesterol. LXRs have neuroprotective effects against the development of neuroinflammation in different neurodegenerative diseases by inhibiting the expression of pro-inflammatory cytokines. LXRs play an essential role in mitigating PD neuropathology by reducing the expression of inflammatory signaling pathways, neuroinflammation, oxidative stress, mitochondrial dysfunction, and enhancement of BDNF signaling.In conclusion, LXRs, through regulating brain cholesterol homeostasis, may be effectual in PD. Also, inhibition of node-like receptor pyrin 3 (NLRP3) inflammasome and nuclear factor kappa B (NF-κB) by LXRs could effectively prevent neuroinflammation in PD. Taken together, LXRs play a crucial role in PD neuropathology by inhibiting neuroinflammation and associated degeneration of DNs.
Collapse
Affiliation(s)
- Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, ALmustansiriyiah University, Baghdad, 14132, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Wien, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt
| |
Collapse
|
8
|
Fan J, To KKW, Chen ZS, Fu L. ABC transporters affects tumor immune microenvironment to regulate cancer immunotherapy and multidrug resistance. Drug Resist Updat 2023; 66:100905. [PMID: 36463807 DOI: 10.1016/j.drup.2022.100905] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022]
Abstract
Multidrug resistance (MDR) is the phenomenon in which cancer cells simultaneously develop resistance to a broad spectrum of structurally and mechanistically unrelated drugs. MDR severely hinders the effective treatment of cancer and is the major cause of chemotherapy failure. ATP-binding cassette (ABC) transporters are extensively expressed in various body tissues, and actively transport endogenous and exogenous substrates through biological membranes. Overexpression of ABC transporters is frequently observed in MDR cancer cells, which promotes efflux of chemotherapeutic drugs and reduces their intracellular accumulation. Increasing evidence suggests that ABC transporters regulate tumor immune microenvironment (TIME) by transporting various cytokines, thus controlling anti-tumor immunity and sensitivity to anticancer drugs. On the other hand, the expression of various ABC transporters is regulated by cytokines and other immune signaling molecules. Targeted inhibition of ABC transporter expression or function can enhance the efficacy of immune checkpoint inhibitors by promoting anticancer immune microenvironment. This review provides an update on the recent research progress in this field.
Collapse
Affiliation(s)
- Jingyi Fan
- State Key Laboratory of Oncology in South China;Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing 100038, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States.
| | - Liwu Fu
- State Key Laboratory of Oncology in South China;Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
9
|
Meszaros M, Bikov A. Obstructive Sleep Apnoea and Lipid Metabolism: The Summary of Evidence and Future Perspectives in the Pathophysiology of OSA-Associated Dyslipidaemia. Biomedicines 2022; 10:2754. [PMID: 36359273 PMCID: PMC9687681 DOI: 10.3390/biomedicines10112754] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Obstructive sleep apnoea (OSA) is associated with cardiovascular and metabolic comorbidities, including hypertension, dyslipidaemia, insulin resistance and atherosclerosis. Strong evidence suggests that OSA is associated with an altered lipid profile including elevated levels of triglyceride-rich lipoproteins and decreased levels of high-density lipoprotein (HDL). Intermittent hypoxia; sleep fragmentation; and consequential surges in the sympathetic activity, enhanced oxidative stress and systemic inflammation are the postulated mechanisms leading to metabolic alterations in OSA. Although the exact mechanisms of OSA-associated dyslipidaemia have not been fully elucidated, three main points have been found to be impaired: activated lipolysis in the adipose tissue, decreased lipid clearance from the circulation and accelerated de novo lipid synthesis. This is further complicated by the oxidisation of atherogenic lipoproteins, adipose tissue dysfunction, hormonal changes, and the reduced function of HDL particles in OSA. In this comprehensive review, we summarise and critically evaluate the current evidence about the possible mechanisms involved in OSA-associated dyslipidaemia.
Collapse
Affiliation(s)
- Martina Meszaros
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Andras Bikov
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9MT, UK
| |
Collapse
|
10
|
Li G, Zhao CY, Wu Q, Kang Z, Zhang JT, Guan SY, Jin HW, Zhang YB, Na XL. Di(2-ethylhexyl) phthalate disturbs cholesterol metabolism through oxidative stress in rat liver. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 95:103958. [PMID: 35970509 DOI: 10.1016/j.etap.2022.103958] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/29/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is widely used and has been implicated in hepatotoxicity, although the mechanism is unclear. Here, we investigated the effect of DEHP on hepatic cholesterol metabolism in SD rats exposed to 0 and 300 mg/kg/day DEHP for 12 weeks. An RNA-Seq analysis was performed to describe the hepatic responses to long-term DEHP exposure in combination with serological and oxidative stress parameter measurements. DEHP increased the serum levels of total cholesterol (TC), high-density lipoprotein (HDL), and alanine transaminase (ALT). Moreover, DEHP increased the content of malondialdehyde (MDA) and decreased antioxidant enzyme activities in the liver. Transcriptomic results revealed that DEHP dramatically changed the cholesterol metabolism pathway and oxidation-reduction process and depressed gene expression involved in cholesterol efflux and monooxygenase activity. Total antioxidant capacity (T-AOC) positively correlated with Abcg5 and Abcg8. Overall, this study showed the mechanisms underlying hepatotoxicity caused by DEHP, providing new insights into understanding DEHP poisoning.
Collapse
Affiliation(s)
- Gang Li
- Department of Environmental Hygiene, Public Health College, Harbin Medical University, Harbin 150086, Heilongjiang Province, China; Department of Preventive Medicine, Public Health College, Qiqihar Medical University, Qiqihar 161006, Heilongjiang Province, China
| | - Chen-Yang Zhao
- Department of Environmental Hygiene, Public Health College, Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Qian Wu
- Department of Environmental Hygiene, Public Health College, Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Zhen Kang
- Department of Environment Hygiene Harbin Center for Disease Control and Prevention, Harbin 150086, Heilongjiang Province, China
| | - Jia-Tai Zhang
- Department of Environmental Hygiene, Public Health College, Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Si-Yuan Guan
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, Fujian Province, China
| | - Hong-Wei Jin
- Guangming District Center for Disease Control and Prevention, Guangming District, Shenzhen 518106, Guangdong Province, China
| | - Yun-Bo Zhang
- Department of Environmental Hygiene, Public Health College, Harbin Medical University, Harbin 150086, Heilongjiang Province, China.
| | - Xiao-Lin Na
- Department of Environmental Hygiene, Public Health College, Harbin Medical University, Harbin 150086, Heilongjiang Province, China.
| |
Collapse
|
11
|
Shoji M, Okamoto R, Unno T, Harada K, Kubo M, Fukuyama Y, Kuzuhara T. Transcriptome analysis of PC12 cells reveals that trans-banglene upregulates RT1-CE1 and downregulates abca1 in the neurotrophic pathway. Biol Pharm Bull 2022; 45:1784-1790. [PMID: 36155550 DOI: 10.1248/bpb.b22-00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Trans(t)-banglene and cis(c)-banglene possess neurotrophin-like activity in rat neurons. However, the molecular mechanisms underlying t-banglene-induced neurotrophic activity in rat and human neurons remain unclear. Here, we performed transcriptome analysis in PC12 cells, a rat adrenal gland pheochromocytoma cell line treated with t-banglene, using comprehensive RNA sequencing. The differentially expressed gene analysis of the sequencing data revealed that the expression of RT1 class I, locus CE1 (RT1-CE1) was upregulated, and that of ATP binding cassette subfamily A member 1 (abca1), myosin light chain 6, and hippocampus abundant transcript 1 was downregulated in t-banglene-treated PC12 cells, with statistically significant differences. We also confirmed the RT1-CE1 upregulation and abca1 downregulation in t-banglene-treated PC12 cells by reverse transcription quantitative real-time polymerase chain reaction. RT1-CEl is a major histocompatibility complex class I (MHCI) protein. ABCAl is a major cholesterol transporter that regulates efflux of intracellular cholesterol and phospholipids. Thus, our results suggest an exciting link between MHCI, cholesterol regulation, and neural development.
Collapse
Affiliation(s)
- Masaki Shoji
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Risa Okamoto
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Taishi Unno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Kenichi Harada
- Laboratory of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Miwa Kubo
- Laboratory of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Yoshiyasu Fukuyama
- Laboratory of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Takashi Kuzuhara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
12
|
Inhibition of Xenobiotics Transporters’ Efflux Ability after Nanoplastics Exposure in Larval Japanese Medaka. WATER 2022. [DOI: 10.3390/w14060863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Nanoplastics can enter into the aquatic environment as primary nano-sized or fragmented from larger-sized plastic particles, and their ecological effects and environmental fate have aroused increasing public concerns. Here, we identified the disruption of ATP-binding cassette (ABC) efflux after polystyrene (PS) nanoplastics (76 ± 7 nm) exposure in larval Japanese medaka (Oryzias latipes). Nanoplastics (0.001–10 μg/mL) caused 3–6-fold higher lipid peroxidation in fish larvae than the control, with concomitant downregulated expression of efflux transporter-related genes (abcb6a, abcc2, abcg2). Two probes of rhodamine (indicative of p-glycoprotein function for parent compounds’ efflux, P-gp) and fluorescein (indicative of multidrug resistance-associated protein function for metabolites’ efflux, MRP) were further used to verify the inhibited ABC efflux ability, via rhodamine and fluorescein bioaccumulation results. Three-fold higher accumulation of rhodamine was observed following treatment with 10 μg/mL of nanoplastics. Excessive accumulation also occurred for fluorescein, with 1.7–1.8-fold higher concentrations than controls in larvae treated with 0.01–0.1 μg/mL of nanoplastics. Although the inhibition of ABC transporters diminished after two hours of depuration, the co-existence of nanoplastics and other contaminants still raises concerns. Collectively, this study suggests that nanoplastics can negatively impact ABC transporters’ efflux ability and could cause unanticipated accumulation of co-existing organic pollutants in aquatic organisms.
Collapse
|
13
|
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: 1.0] [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
|
14
|
Xu F, Shen L, Chen H, Wang R, Zang T, Qian J, Ge J. circDENND1B Participates in the Antiatherosclerotic Effect of IL-1β Monoclonal Antibody in Mouse by Promoting Cholesterol Efflux via miR-17-5p/Abca1 Axis. Front Cell Dev Biol 2021; 9:652032. [PMID: 33996813 PMCID: PMC8116881 DOI: 10.3389/fcell.2021.652032] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Inflammation is a crucial mediator of atherosclerosis, and several therapeutic methods that focus on inflammatory cytokines, including interleukin-1β (IL-1β), have proven effective in preventing atherogenesis. Circular RNAs (circRNAs) are a subclass of non-coding RNAs (ncRNAs) that can exert critical functions in the regulation of atherosclerosis. Here, using circRNA sequencing, we revealed that circRNA circDENND1B (mmu_circ_0000081) is a promising novel mediator of atherosclerosis in mouse. The expression of circDENND1B is negatively related to the progression of atherosclerosis and foam cell formation, and the upregulation of circDENND1B significantly alleviates foam cell formation induced by ox-LDL by promoting cholesterol efflux. Moreover, circDENND1B participates in the anti-atherosclerotic effect of IL-1β monoclonal antibody (IL-1β mAb), both in vivo and in vitro. With bioinformatic prediction and RNA pull-down assays, we determined that circDENND1B sponges mmu-miR-17-5p to promote Abca1 expression in cells treated with IL-1β mAb. Our study revealed that circDENND1B, a novel regulator of cholesterol efflux, is a potential therapeutic target in atherosclerosis and provides new insights into the interaction between inflammation and cholesterol transport.
Collapse
Affiliation(s)
- Fei Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Han Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Rui Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Tongtong Zang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, China.,National Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
15
|
Uekita H, Yamamoto H, Niinaga R, Yamane N, Yoshii M, Yamauchi-Takihara K, Kihara S. Reciprocal association of serum Mac-2 binding protein and HDL-cholesterol concentrations. Clin Chim Acta 2021; 516:142-148. [PMID: 33571485 DOI: 10.1016/j.cca.2021.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Mac-2 binding protein (Mac-2BP) is used as a serum biomarker of nonalcoholic steatohepatitis, considered to be a liver phenotype of metabolic syndrome (MetS). In this study, we investigated the serum Mac-2BP concentrations-correlated MetS-related clinical parameters in vivo, and the underlying mechanism in vitro. MATERIALS & METHODS We enrolled 54 healthy Japanese men who underwent health examination at Osaka University Health Care Center in this study. Physical and serum biochemical parameters were obtained from all the subjects. In the cultured HepG2 cells, the effects of interferon (IFN)-γ on the expression of Mac-2BP, apolipoprotein (apo) A-I, and ATP binding cassette transporter A1 (ABCA1) were studied. RESULTS Serum Mac-2BP concentrations correlated negatively with HDL-C, and positively with body mass index and systolic blood pressure in univariate analysis. These results suggested the association between Mac-2BP and MetS, although none of these 3 parameters had significant correlation with serum Mac-2BP concentrations in multivariate analysis. In HepG2 cells, IFN-γ stimulation resulted in the increased Mac-2BP and the decreased ABCA1 and apo A-I mRNA concentrations, while Mac-2BP had no effects on ABCA1 and apo A-I concentrations. CONCLUSIONS The serum Mac-2BP concentrations are negatively correlated with HDL-C concentrations in healthy subjects, as a result of chronic inflammation.
Collapse
Affiliation(s)
- Hiromi Uekita
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Medical Technology, Faculty of Health Sciences, Kansai University of Health Sciences, Osaka, Japan
| | - Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Ryu Niinaga
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Noriko Yamane
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Manami Yoshii
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
16
|
Li J, Meng Q, Fu Y, Yu X, Ji T, Chao Y, Chen Q, Li Y, Bian H. Novel insights: Dynamic foam cells derived from the macrophage in atherosclerosis. J Cell Physiol 2021; 236:6154-6167. [PMID: 33507545 DOI: 10.1002/jcp.30300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis can be regarded as a chronic disease derived from the interaction between disordered lipoproteins and an unsuitable immune response. The evolution of foam cells is not only a significant pathological change in the early stage of atherosclerosis but also a key stage in the occurrence and development of atherosclerosis. The formation of foam cells is mainly caused by the imbalance among lipids uptake, lipids treatment, and reverse cholesterol transport. Although a large number of studies have summarized the source of foam cells and the mechanism of foam cells formation, we propose a new idea about foam cells in atherosclerosis. Rather than an isolated microenvironment, the macrophage multiple lipid uptake pathways, lipid internalization, lysosome, mitochondria, endoplasmic reticulum, neutral cholesterol ester hydrolase (NCEH), acyl-coenzyme A-cholesterol acyltransferase (ACAT), and reverse cholesterol transport are mutually influential, and form a dynamic process under multi-factor regulation. The macrophage takes on different uptake lipid statuses depending on multiple uptake pathways and intracellular lipids, lipid metabolites versus pro-inflammatory factors. Except for NCEH and ACAT, the lipid internalization of macrophages also depends on multicellular organelles including the lysosome, mitochondria, and endoplasmic reticulum, which are associated with each other. A dynamic balance between esterification and hydrolysis of cholesterol for macrophages is essential for physiology and pathology. Therefore, we propose that the foam cell in the process of atherosclerosis may be dynamic under multi-factor regulation, and collate this study to provide a holistic and dynamic idea of the foam cell.
Collapse
Affiliation(s)
- Jun Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinghai Meng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Fu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xichao Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingting Ji
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Chao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huimin Bian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
Song J, Han D, Lee H, Kim DJ, Cho JY, Park JH, Seok SH. A Comprehensive Proteomic and Phosphoproteomic Analysis of Retinal Pigment Epithelium Reveals Multiple Pathway Alterations in Response to the Inflammatory Stimuli. Int J Mol Sci 2020; 21:ijms21093037. [PMID: 32344885 PMCID: PMC7246457 DOI: 10.3390/ijms21093037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/31/2022] Open
Abstract
Overwhelming and persistent inflammation of retinal pigment epithelium (RPE) induces destructive changes in the retinal environment. However, the precise mechanisms remain unclear. In this study, we aimed to investigate RPE-specific biological and metabolic responses against intense inflammation and identify the molecular characteristics determining pathological progression. We performed quantitative analyses of the proteome and phosphoproteome of the human-derived RPE cell line ARPE-19 after treatment with lipopolysaccharide (LPS) for 45 min or 24 h using the latest isobaric tandem-mass tags (TMTs) labeling approach. This approach led to the identification of 8984 proteins, of which 261 showed a 1.5-fold change in abundance after 24 h of treatment with LPS. A parallel phosphoproteome analysis identified 20,632 unique phosphopeptides from 3207 phosphoproteins with 3103 phosphorylation sites. Integrated proteomic and phosphoproteomic analyses showed significant downregulation of proteins related to mitochondrial respiration and cell cycle checkpoint, while proteins related to lipid metabolism, amino acid metabolism, cell-matrix adhesion, and endoplasmic reticulum (ER) stress were upregulated after LPS stimulation. Further, phosphorylation events in multiple pathways, including MAPKK and Wnt/β-catenin signalings, were identified as involved in LPS-triggered pathobiology. In essence, our findings reveal multiple integrated signals exerted by RPE under inflammation and are expected to give insight into the development of therapeutic interventions for RPE disorders.
Collapse
Affiliation(s)
- Juha Song
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea;
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul 08826, Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (D.H.); (H.L.)
| | - Heonyi Lee
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (D.H.); (H.L.)
| | - Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea; (D.J.K.); (J.-Y.C.)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea; (D.J.K.); (J.-Y.C.)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea
| | - Jae-Hak Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul 08826, Korea
- Correspondence: (J.-H.P.); (S.H.S.); Tel.: +82-2-880-1256 (J.-H.P.); +82-2-740-8302 (S.H.S.); Fax: +82-2-763-5206 (S.H.S.)
| | - Seung Hyeok Seok
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea;
- Department of Biomedical Sciences, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea
- Correspondence: (J.-H.P.); (S.H.S.); Tel.: +82-2-880-1256 (J.-H.P.); +82-2-740-8302 (S.H.S.); Fax: +82-2-763-5206 (S.H.S.)
| |
Collapse
|
18
|
ABC Transporters, Cholesterol Efflux, and Implications for Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:67-83. [DOI: 10.1007/978-981-15-6082-8_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Luo J, Wang X, Jiang X, Liu C, Li Y, Han X, Zuo X, Li Y, Li N, Xu Y, Si S. Rutaecarpine derivative R3 attenuates atherosclerosis via inhibiting NLRP3 inflammasome-related inflammation and modulating cholesterol transport. FASEB J 2019; 34:1398-1411. [PMID: 31914630 DOI: 10.1096/fj.201900903rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022]
Abstract
Atherosclerosis is a chronic disease characterized by lipid deposition and inflammatory response. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome-facilitated inflammatory responses are crucial in the pathogenesis of atherosclerosis, and thus new therapeutic approaches are emerging that target NLRP3 and inflammation. Here, we explored the anti-atherosclerotic effect and mechanisms of a new rutaecarpine derivative, 5-deoxy-rutaecarpine (R3) in vitro and in vivo. R3 treatment attenuated atherosclerosis development and increased plaque stability in Apoe-/- mice fed a high-fat diet, and decreased levels of inflammatory mediators, such as interleukin-1β, in the serum of Apoe-/- mice and in oxidized low-density lipoprotein (ox-LDL)-stimulated murine macrophages. R3 treatment inhibited NLRP3 inflammasome activation in the livers of Apoe-/- mice and ox-LDL-stimulated murine macrophages by inhibiting NF-κB and MAPK pathways. Additionally, R3 significantly decreased total cholesterol in the serum and livers of Apoe-/- mice and promoted cholesterol efflux in murine macrophages through upregulating protein expression of ATP-binding cassette subfamily A member 1 and scavenger receptor class B type I/human CD36 and lysosomal integral membrane protein-II analogous-1. Our results demonstrated that R3 prevented atherosclerotic progression via attenuating NLRP3 inflammasome-related inflammation and modulating cholesterol transport.
Collapse
Affiliation(s)
- Jinque Luo
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinhai Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongzhen Li
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaowan Han
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuan Zuo
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yining Li
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ni Li
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyi Si
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Indicaxanthin from Opuntia ficus indica (L. Mill) Inhibits Oxidized LDL-Mediated Human Endothelial Cell Dysfunction through Inhibition of NF- κB Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3457846. [PMID: 30911345 PMCID: PMC6398026 DOI: 10.1155/2019/3457846] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023]
Abstract
Oxidized low-density lipoproteins (oxLDL) play a pivotal role in the etiopathogenesis of atherosclerosis through the activation of inflammatory signaling events eventually leading to endothelial dysfunction and senescence. In the present work, we investigated the effects of indicaxanthin, a bioavailable, redox-modulating phytochemical from Opuntia ficus indica fruits, with anti-inflammatory activity, against oxLDL-induced endothelial dysfunction. Human umbilical vein cord cells (HUVEC) were stimulated with human oxLDL, and the effects of indicaxanthin were evaluated in a range between 5 and 20 μM, consistent with its plasma level after a fruit meal (7 μM). Pretreatment with indicaxanthin significantly and concentration-dependently inhibited oxLDL-induced cytotoxicity; ICAM-1, VCAM-1, and ELAM-1 increase; and ABC-A1 decrease of both protein and mRNA levels. From a mechanistic perspective, we also provided evidence that the protective effects of indicaxanthin were redox-dependent and related to the pigment efficacy to inhibit NF-κB transcriptional activity. In conclusion, here we demonstrate indicaxanthin as a novel, dietary phytochemical, able to exert significant protective vascular effects in vitro, at nutritionally relevant concentrations.
Collapse
|
21
|
Vendrov AE, Sumida A, Canugovi C, Lozhkin A, Hayami T, Madamanchi NR, Runge MS. NOXA1-dependent NADPH oxidase regulates redox signaling and phenotype of vascular smooth muscle cell during atherogenesis. Redox Biol 2018; 21:101063. [PMID: 30576919 PMCID: PMC6302039 DOI: 10.1016/j.redox.2018.11.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/16/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Increased reactive oxygen species (ROS) production and inflammation are key factors in the pathogenesis of atherosclerosis. We previously reported that NOX activator 1 (NOXA1) is the critical functional homolog of p67phox for NADPH oxidase activation in mouse vascular smooth muscle cells (VSMC). Here we investigated the effects of systemic and SMC-specific deletion of Noxa1 on VSMC phenotype during atherogenesis in mice. Neointimal hyperplasia following endovascular injury was lower in Noxa1-deficient mice versus the wild-type following endovascular injury. Noxa1 deletion in Apoe-/- or Ldlr-/- mice fed a Western diet showed 50% reduction in vascular ROS and 30% reduction in aortic atherosclerotic lesion area and aortic sinus lesion volume (P < 0.01). SMC-specific deletion of Noxa1 in Apoe-/- mice (Noxa1SMC-/-/Apoe-/-) similarly decreased vascular ROS levels and atherosclerotic lesion size. TNFα-induced ROS generation, proliferation and migration were significantly attenuated in Noxa1-deficient versus wild-type VSMC. Immunofluorescence analysis of atherosclerotic lesions showed a significant decrease in cells positive for CD68 and myosin11 (22% versus 9%) and Mac3 and α-actin (17% versus 5%) in the Noxa1SMC-/-/Apoe-/- versus Apoe-/- mice. The expression of transcription factor KLF4, a modulator of VSMC phenotype, and its downstream targets – VCAM1, CCL2, and MMP2 – were significantly reduced in the lesions of Noxa1SMC-/-/Apoe-/- versus Apoe-/- mice as well as in oxidized phospholipids treated Noxa1SMC-/- versus wild-type VSMC. Our data support an important role for NOXA1-dependent NADPH oxidase activity in VSMC plasticity during restenosis and atherosclerosis, augmenting VSMC proliferation and migration and KLF4-mediated transition to macrophage-like cells, plaque inflammation, and expansion. NOXA1 is a VSMC-specific regulator of NADPH oxidase 1 activity and downstream cell signaling. NOX1 NADPH oxidase-dependent ROS generation is required for VSMC proliferation and migration after endovascular injury. NOXA1-dependent NOX1 activation of KLF4 in atherosclerotic lesions induces SMC phenotypic switch to macrophage-like cells. Atherosclerotic lesion macrophage-like cells promote plaque inflammation, matrix remodeling and increase volume expansion.
Collapse
Affiliation(s)
- Aleksandr E Vendrov
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arihiro Sumida
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chandrika Canugovi
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrey Lozhkin
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takayuki Hayami
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R Madamanchi
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marschall S Runge
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Nessaibia I, Fouache A, Lobaccaro JMA, Tahraoui A, Trousson A, Souidi M. Stress as an immunomodulator: liver X receptors maybe the answer. Inflammopharmacology 2018; 27:15-25. [PMID: 30467620 DOI: 10.1007/s10787-018-0546-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/03/2018] [Indexed: 02/07/2023]
Abstract
Stress is a reflex response, both psychological and physiological, of the body to a difficult situation that requires adaptation. Stress is at the intersection of the objective event and the subjective event. The physiological mechanisms involved in chronic stress are numerous and can contribute to a wide variety of disorders, in all systems including the immune system. Stress modifies the Th1/Th2 balance via the HPA axis and a set of immune mediators. This will make the body more vulnerable to external infections in a scientific way while others claim the opposite, stress could be considered immune stimulatory. The development of synthetic LXR ligands such as T0901317 and GW3965 as well as an understanding of the direct involvement of these receptors in the regulation of proopiomelanocortin (POMC) gene expression and indirectly by producing a variety of cytokines in a stressor response, will open in the near future new therapeutic methods against the undesirable effects of stress on the behavior of the immune system.
Collapse
Affiliation(s)
- Issam Nessaibia
- CNRS UMR 6293, Laboratory GReD, INSERM U 1103, Clermont Auvergne University, 28 Place Henri Dunant, 63000, Clermont-Ferrand, France.
- Laboratory of Applied Neuro-Endocrinology, Department of Biology, Badji-Mokhtar University, Annaba, Algeria.
| | - Allan Fouache
- CNRS UMR 6293, Laboratory GReD, INSERM U 1103, Clermont Auvergne University, 28 Place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Jean-Marc A Lobaccaro
- CNRS UMR 6293, Laboratory GReD, INSERM U 1103, Clermont Auvergne University, 28 Place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Abdelkrim Tahraoui
- Laboratory of Applied Neuro-Endocrinology, Department of Biology, Badji-Mokhtar University, Annaba, Algeria
| | - Amalia Trousson
- CNRS UMR 6293, Laboratory GReD, INSERM U 1103, Clermont Auvergne University, 28 Place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Maâmar Souidi
- Institut de radioprotection et de sûreté nucléaire, Direction de la radioprotection de l'homme, IRSN, Fontenay-aux-Roses Cedex, France
| |
Collapse
|
23
|
Varghese JF, Patel R, Yadav UCS. Sterol regulatory element binding protein (SREBP) -1 mediates oxidized low-density lipoprotein (oxLDL) induced macrophage foam cell formation through NLRP3 inflammasome activation. Cell Signal 2018; 53:316-326. [PMID: 30389501 DOI: 10.1016/j.cellsig.2018.10.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 11/27/2022]
Abstract
Macrophage foam cell formation (FCF) has long been known to play a critical role during atherosclerotic plaque development. In the presence of atherogenic molecules such as oxidized low-density lipoprotein (oxLDL) macrophages accumulate massive amounts of lipid through uptake. However, in the presence of oxLDL mechanism of dysregulated lipid homeostasis in the macrophages remains largely unknown. Herein we have investigated the role of Sterol regulatory element binding protein (SREBP)-1 in oxLDL-induced inflammation and altered lipid homeostasis in macrophages. The U937 monocytes and monocyte-derived macrophages (MDMs) were stimulated with different doses of oxLDL. MTT assay to study the effect of oxLDL on cell viability, Oil-Red-O (ORO) staining to observe cytosolic lipid accumulation, semi-quantitative PCR and Western blotting to analyze mRNA and protein expressions, respectively, and spectrophotometric assay to measure the lipid synthesizing enzyme's activity were performed. Our results indicate that oxLDL increased proliferation in monocytes and decreased the viability in MDMs in a time- and dose-dependent manner. The oxLDL (100 μg/ml) enhanced lipid accumulation via increased expressions of SREBP-1 and its downstream proteins such as fatty acid synthase (FAS) and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) at both RNA and protein levels in monocytes as well as in MDMs. Inhibiting SREBP-1 by a synthetic inhibitor prevented excessive lipid accumulation by downregulating the expression of its downstream proteins. Further, oxLDL increased reactive oxygen species (ROS) levels, NLRP3 inflammasome activation and active interleukin 1β (IL-1β) release in both the cell types. The oxLDL-induced NLRP3 could be responsible for SREBP-1 and downstream proteins overexpression as siRNA silencing of NLRP3 decreased SERBP-1 levels. In summary, we have demonstrated that SREBP-1 could be a key player in oxLDL-induced excessive lipid accumulation leading to macrophage FCF via ROS-mediated NLRP3/IL-1β/SREBP-1 pathway.
Collapse
Affiliation(s)
- Johnna F Varghese
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Rohit Patel
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Umesh C S Yadav
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India.
| |
Collapse
|
24
|
He X, Chen X, Wang L, Wang W, Liang Q, Yi L, Wang Y, Gao Q. Metformin ameliorates Ox-LDL-induced foam cell formation in raw264.7 cells by promoting ABCG-1 mediated cholesterol efflux. Life Sci 2018; 216:67-74. [PMID: 30218721 DOI: 10.1016/j.lfs.2018.09.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/29/2018] [Accepted: 09/12/2018] [Indexed: 01/22/2023]
Abstract
AIMS The accumulation of lipids in macrophages contributes to the development of atherosclerosis. Cholesterol efflux of lipid-loaded macrophages mediated by ATP binding cassette (ABC) cholesterol transporters, on the other hand, has been shown to attenuate atherosclerosis progression in patients with unknown mechanism. We therefore sought to test the effect of metformin that reduced cardiovascular risk in diabetic patients independent of its hypoglycemia effect on cholesterol transport in murine raw264.7 macrophages. MATERIALS AND METHODS Mouse raw264.7 macrophages were loaded with Ox-LDL (50 μg/ml) for 24 h before incubated with metformin (15 μM) for 24 h. Foam cell formation was assessed by Oil red staining and BIODIPY fluorescent staining as well as cholesterol-ester quantification by commercial kit. Cholesterol uptake and expression of scavenger receptors were detected by flow-cytometry. Cholesterol efflux capacity was measured by fluorescent plate-reader and ABC transporters were detected by Western Blots. Cytokines were detected by ELISA in supernatants and normalized by cellular lysates. KEY FINDINGS Our results showed that metformin decreased oxidized low-density lipoprotein (Ox-LDL)-induced cholesterol accumulation and foam cell formation by increasing cholesterol efflux to HDL, which was associated with an upregulation of ABC transporter ABCG-1. Moreover, metformin increased Ox-LDL-impaired IL-10 secretion, an important anti-foam cell cytokine in atherosclerosis. SIGNIFICANCE Our data highlighted the therapeutic potential of targeting macrophage cholesterol efflux with new or existing drugs for the possible reduction of foam cell formation in the prevention and treatment of diabetes-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Xuan He
- Medical School of Nanjing University, Nanjing 210093, China
| | - Xiufang Chen
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Lei Wang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Wenqing Wang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Qiao Liang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Long Yi
- Medical School of Nanjing University, Nanjing 210093, China
| | - Yong Wang
- Medical School of Nanjing University, Nanjing 210093, China
| | - Qian Gao
- Medical School of Nanjing University, Nanjing 210093, China.
| |
Collapse
|
25
|
Tumurkhuu G, Dagvadorj J, Porritt RA, Crother TR, Shimada K, Tarling EJ, Erbay E, Arditi M, Chen S. Chlamydia pneumoniae Hijacks a Host Autoregulatory IL-1β Loop to Drive Foam Cell Formation and Accelerate Atherosclerosis. Cell Metab 2018; 28:432-448.e4. [PMID: 29937375 PMCID: PMC6125162 DOI: 10.1016/j.cmet.2018.05.027] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 01/02/2018] [Accepted: 05/29/2018] [Indexed: 01/07/2023]
Abstract
Pathogen burden accelerates atherosclerosis, but the mechanisms remain unresolved. Activation of the NLRP3 inflammasome is linked to atherogenesis. Here we investigated whether Chlamydia pneumoniae (C.pn) infection engages NLRP3 in promoting atherosclerosis. C.pn potentiated hyperlipidemia-induced inflammasome activity in cultured macrophages and in foam cells in atherosclerotic lesions of Ldlr-/- mice. C.pn-induced acceleration of atherosclerosis was significantly dependent on NLRP3 and caspase-1. We discovered that C.pn-induced extracellular IL-1β triggers a negative feedback loop to inhibit GPR109a and ABCA1 expression and cholesterol efflux, leading to accumulation of intracellular cholesterol and foam cell formation. Gpr109a and Abca1 were both upregulated in plaque lesions in Nlrp3-/- mice in both hyperlipidemic and C.pn infection models. Mature IL-1β and cholesterol may compete for access to the ABCA1 transporter to be exported from macrophages. C.pn exploits this metabolic-immune crosstalk, which can be modulated by NLRP3 inhibitors to alleviate atherosclerosis.
Collapse
Affiliation(s)
- Gantsetseg Tumurkhuu
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jargalsaikhan Dagvadorj
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rebecca A Porritt
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Timothy R Crother
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kenichi Shimada
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elizabeth J Tarling
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ebru Erbay
- Department of Molecular Biology and Genetics and National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Moshe Arditi
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Shuang Chen
- Departments of Pediatrics and Medicine, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
26
|
ApoA-1 Mimetic Peptide ELK-2A2K2E Decreases Inflammatory Factor Levels Through the ABCA1-JAK2-STAT3-TTP Axis in THP-1–Derived Macrophages. J Cardiovasc Pharmacol 2018; 72:60-67. [DOI: 10.1097/fjc.0000000000000594] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Chen L, Yao Q, Xu S, Wang H, Qu P. Inhibition of the NLRP3 inflammasome attenuates foam cell formation of THP-1 macrophages by suppressing ox-LDL uptake and promoting cholesterol efflux. Biochem Biophys Res Commun 2018; 495:382-387. [DOI: 10.1016/j.bbrc.2017.11.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/12/2022]
|
28
|
Affiliation(s)
- Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Srinivasa T Reddy
- Department Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
29
|
Şeker Karatoprak G, Göger F, Yerer MB, Koşar M. Chemical composition and biological investigation of Pelargonium endlicherianum root extracts. PHARMACEUTICAL BIOLOGY 2017; 55:1608-1618. [PMID: 28407721 PMCID: PMC7012040 DOI: 10.1080/13880209.2017.1314511] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 03/09/2017] [Accepted: 03/29/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT Pelargonium endlicherianum Fenzl. (Geraniaceae) roots and flowers are traditionally used in Turkey as a decoction treatment against intestinal parasites. Neither the chemical composition nor the potential bioactivity of the plant roots has been studied before. OBJECTIVES The phenolic content and effects of P. endlicherianum root extracts on antioxidant enzyme levels on A549 cells were studied for the first time. MATERIALS AND METHODS The chemical composition was analyzed via spectrophotometric and chromatographic (HPLC MS/MS and HPLC) techniques. The antioxidant activity was determined at different concentrations ranging from 0.001 to 2 mg/mL using DPPH• and ABTS•+ radical scavenging activity, β-carotene-linoleic acid co-oxidation assay, protection of 2-deoxyribose and bovine brain-derived phospholipids against a hydroxyl radical-mediated degradation assay. Glutathione peroxidase and superoxide dismutase activities were also studied as well as the effects of the extracts on nitric oxide levels on IL-1β stimulated A549 cells. RESULTS The key parameters for the most active ethyl acetate extract included the following: DPPH• IC50: 0.23 mg/mL, TEAC/ABTS: 2.17 mmol/L Trolox, reduction: 0.41 mmol/g AsscE, and protection of lipid peroxidation IC50: 0.05 mg/mL. Furthermore, the ethyl acetate extract increased the SOD level significantly compared to control group (4.48 U/mL) at concentrations of 100 and 200 μg/mL SOD, 5.50 and 5.67 U/mL, respectively. Apocynin was identified as the major component, and the ethyl acetate fraction was found to be rich in phenolic compounds. DISCUSSION AND CONCLUSION Pelargonium endlicherianum root extracts displayed antioxidant activity and increased the antioxidant enzyme levels in IL-1β stimulated A549 cells, while decreasing the NO levels.
Collapse
Affiliation(s)
- Gökçe Şeker Karatoprak
- Department of Pharmacognosy, Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Fatih Göger
- Department of Pharmacognosy, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Mükerrem Betül Yerer
- Department of Pharmacognosy, Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Müberra Koşar
- Department of Pharmacognosy, Faculty of Pharmacy, Eastern Mediterranean University, Gazimağusa, Mersin, Turkey
| |
Collapse
|
30
|
Pérez‐Baos S, Barrasa JI, Gratal P, Larrañaga‐Vera A, Prieto‐Potin I, Herrero‐Beaumont G, Largo R. Tofacitinib restores the inhibition of reverse cholesterol transport induced by inflammation: understanding the lipid paradox associated with rheumatoid arthritis. Br J Pharmacol 2017; 174:3018-3031. [PMID: 28646516 PMCID: PMC5573422 DOI: 10.1111/bph.13932] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 06/06/2017] [Accepted: 06/17/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Patients with active rheumatoid arthritis (RA) have increased cardiovascular mortality, paradoxically associated with reduced circulating lipid levels. The JAK inhibitor tofacitinib ameliorates systemic and joint inflammation in RA with a concomitant increase in serum lipids. We analysed the effect of tofacitinib on the lipid profile of hyperlipidaemic rabbits with chronic arthritis (CA) and on the changes in reverse cholesterol transport (RCT) during chronic inflammation. EXPERIMENTAL APPROACH CA was induced in previously immunized rabbits, fed a high-fat diet, by administering four intra-articular injections of ovalbumin. A group of rabbits received tofacitinib (10 mg·kg-1 ·day-1 ) for 2 weeks. Systemic and synovial inflammation and lipid content were evaluated. For in vitro studies, THP-1-derived macrophages were exposed to high lipid concentrations and then stimulated with IFNγ in the presence or absence of tofacitinib in order to study mediators of RCT. KEY RESULTS Tofacitinib decreased systemic and synovial inflammation and increased circulating lipid levels. Although it did not modify synovial macrophage density, it reduced the lipid content within synovial macrophages. In foam macrophages in culture, IFNγ further stimulated intracellular lipid accumulation, while the JAK/STAT inhibition provoked by tofacitinib induced lipid release by increasing the levels of cellular liver X receptor α and ATP-binding cassette transporter (ABCA1) synthesis. CONCLUSIONS AND IMPLICATIONS Active inflammation could be associated with lipid accumulation within macrophages of CA rabbits. JAK inhibition induced lipid release through RCT activation, providing a plausible explanation for the effect of tofacitinib on the lipid profile of RA patients.
Collapse
Affiliation(s)
- S Pérez‐Baos
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - J I Barrasa
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
| | - P Gratal
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - A Larrañaga‐Vera
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - I Prieto‐Potin
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - G Herrero‐Beaumont
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - R Largo
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| |
Collapse
|
31
|
Mani P, Uno K, Duong M, Wolski K, Spalding S, Husni ME, Nicholls SJ. HDL function and subclinical atherosclerosis in juvenile idiopathic arthritis. Cardiovasc Diagn Ther 2016; 6:34-43. [PMID: 26885490 DOI: 10.3978/j.issn.2223-3652.2015.12.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Increasing evidence suggests that inflammation adversely impacts the protective properties of high-density lipoproteins (HDL) and progression of atherosclerosis. The impact of early chronic inflammatory conditions on HDL function and vascular risk has not been well investigated. METHODS We compared measures of HDL particle distribution and functionality, in addition to measures of carotid intima-medial thickness (cIMT) in patients with juvenile idiopathic arthritis (JIA) and age matched controls. RESULTS JIA patients demonstrated lower levels of HDL cholesterol [47.0 (40.0, 56.0) vs. 56.0 (53.0, 61.0) mg/dL, P=0.04], total HDL [29.5 (27.9, 32.3) vs. 32.9 (31.6, 36.3) mg/dL, P=0.05] and large HDL [5.1 (3.7, 7.3) vs. 8.0 (6.7, 9.7) mg/dL, P=0.04] particles. In association JIA patients demonstrated greater cholesterol efflux mediated via ATP binding cassette A1 (ABCA1) [17.3% (12.8, 19.7) vs. 10.0% (5.8, 16.0), P=0.05] and less efflux mediated via ATP binding cassette G-1 (ABCG1) [3.2% (2.0, 3.9) vs. 4.8% (3.5, 5.8), P=0.01] and SR-B1 [6.9% (6.0, 8.4) vs. 9.1% (8.6, 10.2), P=0.002] compared with controls. Exposure of macrophages to serum from JIA patients resulted in a smaller increase in mRNA expression of ABCA1 (2.0±0.95 vs. 7.1±5.7 fold increase, P=0.01) and greater increases in expression of ABCG1 [1.4 (0.9, 1.5) vs. 0.8 (0.7, 1.1) fold increase, P=0.04] and SR-B1 (1.3±0.47 vs. 0.7±0.3 fold increase, P=0.001) compared with controls. Arylesterase (128.9±27.6 vs. 152.0±45.2 umoles/min/mL, P=0.04) activity and endothelial cell migration (491.2±68.9 vs. 634.2±227.4 cells/field, P=0.01) were less in JIA patients. No differences in cIMT were observed between JIA patients and controls. CONCLUSIONS The presence of JIA was associated with alterations in HDL particle distribution, cholesterol efflux and non-lipid transporting activities. The ultimate implication of these findings for cardiovascular risk requires further investigation.
Collapse
Affiliation(s)
- Preethi Mani
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kiyoko Uno
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - MyNgan Duong
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kathy Wolski
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Steven Spalding
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - M Elaine Husni
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stephen J Nicholls
- 1 Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA ; 2 Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA ; 3 South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia ; 4 Department of Rheumatology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
32
|
Xue XH, Shi FF, Chen T, Wei W, Zhou XM, Chen LD. Inhibition of ERK1/2 improves lipid balance in rat macrophages via ABCA1/G1 and CD36. Mol Med Rep 2015; 13:1533-40. [PMID: 26707062 DOI: 10.3892/mmr.2015.4697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 12/07/2015] [Indexed: 11/06/2022] Open
Abstract
ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1), and macrophage scavenger receptor, cluster of differentiation (CD)36, function as key mediators of cholesterol efflux and influx from macrophages. In addition, they are associated with foam cell formation and the development of atherosclerosis (AS). The aim of the present study was to investigate the effects of extracellular signal-regulated kinases 1/2 (ERK1/2) inhibition on lipid balance in oxidized-low-density lipoprotein (Ox-LDL)-stimulated rat macrophages, and to examine the role of ERK1/2 inhibitors in AS. Rat peritoneal macrophages were treated with Ox-LDL alone or in combination with an ERK1/2 inhibitor, U0126, and untreated cells served as controls. Ox-LDL-induced lipid accumulation was detected by DiI fluorescence and oil red O staining. In addition, the mRNA and protein expression levels of ABCA1, ABCG1 and CD36 were determined using polymerase chain reaction and western blotting, respectively. Treatment with Ox-LDL significantly increased lipid accumulation and upregulated the mRNA and protein expression levels of ABCA1, ABCG1 and CD36 in macrophages. The addition of U0126 resulted in a marked reduction of lipid deposition, upregulation of ABCA1/G1 expression and suppression of CD36 expression in Ox-LDL-stimulated macrophages. The results of the present study indicated a novel association between ERK1/2 signaling and lipid metabolism, thus suggesting that inhibition of ERK1/2 may be considered a promising therapeutic strategy against AS.
Collapse
Affiliation(s)
- Xie-Hua Xue
- Department of Neurology, Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Feng-Fei Shi
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Tong Chen
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Wei Wei
- Department of Neurology, Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Xiao-Mao Zhou
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Li-Dian Chen
- Institute of Rehabilitation Medicine, Rehabilitation Technology Collaborative Innovation Center, College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| |
Collapse
|
33
|
IRAK1 mediates TLR4-induced ABCA1 downregulation and lipid accumulation in VSMCs. Cell Death Dis 2015; 6:e1949. [PMID: 26512959 PMCID: PMC5399175 DOI: 10.1038/cddis.2015.212] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/23/2015] [Accepted: 06/30/2015] [Indexed: 12/14/2022]
Abstract
The activation of Toll-like receptor 4 (TLR4) signaling has an important role in promoting lipid accumulation and pro-inflammatory effects in vascular smooth muscle cells (VSMCs), which facilitate atherosclerosis development and progression. Previous studies have demonstrated that excess lipid accumulation in VSMCs is due to an inhibition of the expression of ATP-binding cassette transporter A1 (ABCA1), an important molecular mediator of lipid efflux from VSMCs. However, the underlying molecular mechanisms of this process are unclear. The purpose of this study was to disclose the underlying molecular mechanisms of TLR4 signaling in regulating ABCA1 expression. Primary cultured VSMCs were stimulated with 50 μg/ml oxidized low-density lipoprotein (oxLDL). We determined that enhancing TLR4 signaling using oxLDL significantly downregulated ABCA1 expression and induced lipid accumulation in VSMCs. However, TLR4 knockout significantly rescued oxLDL-induced ABCA1 downregulation and lipid accumulation. In addition, IL-1R-associated kinase 1 (IRAK1) was involved in the effects of TLR4 signaling on ABCA1 expression and lipid accumulation. Silencing IRAK1 expression using a specific siRNA reversed TLR4-induced ABCA1 downregulation and lipid accumulation in vitro. These results were further confirmed by our in vivo experiments. We determined that enhancing TLR4 signaling by administering a 12-week-long high-fat diet (HFD) to mice significantly increased IRAK1 expression, which downregulated ABCA1 expression and induced lipid accumulation. In addition, TLR4 knockout in vivo reversed the effects of the HFD on IRAK1 and ABCA1 expression, as well as on lipid accumulation. In conclusion, IRAK1 is involved in TLR4-mediated downregulation of ABCA1 expression and lipid accumulation in VSMCs.
Collapse
|
34
|
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: 5.5] [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
|
35
|
ABCA1 expression in macrophages of allogeneic hematopoietic stem cell transplantation patients with severe infection undergoing continuous blood purification. Int J Artif Organs 2015; 38:83-8. [PMID: 25744199 DOI: 10.5301/ijao.5000388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND Excessive activation of the inflammatory mediator cascade after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients is associated with high mortality. Many studies have shown that continuous blood purification (CBP) could improve the prognosis of allo-HSCT patients with severe infection. However, the exact mechanism remains unclear. The aim of this study was to observe the effect of CBP on the expression of ATP-binding cassette transporter A1 (ABCA1) in macrophages, and to investigate the interventional effects of CBP on serum cytokine in allo-HSCT patients with severe infection. METHODS A total of 26 allo-HSCT patients with severe infection were included in this study. Before CBP and after CBP, blood samples were collected to observe hepatic and renal function, and the serum levels of TNF-α, IL-1, IL-6, and IL-10 were detected via ELISA. The THP-1 macrophages were exposed to serum samples obtained from patients at specific time points during CBP to test the changes of ABCA1 in macrophages by real-timePCR and Western blotting. RESULTS Serum creatinine, alanine aminotransferase, and C reaction protein (CRP) levels decreased significantly after CBP. Moreover, TNF-α, IL-1, and IL-6 serum levels decreased significantly, but IL-10 level increased significantly after CBP (P<.05). After CBP, ABCA1 expression levels were higher than those before CBP, and ABCA1 expression was significantly increased with the supplementation of CBP (P<.05). CONCLUSIONS CBP improved the condition of allo-HSCT patients with severe infection. CBP may be a potent up-regulator of the ABCA1 levels in macrophages of allo-HSCT patients with severe infection.
Collapse
|
36
|
Liu W, Yin Y, Zhou Z, He M, Dai Y. OxLDL-induced IL-1 beta secretion promoting foam cells formation was mainly via CD36 mediated ROS production leading to NLRP3 inflammasome activation. Inflamm Res 2014; 63:33-43. [PMID: 24121974 DOI: 10.1007/s00011-013-0667-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 09/13/2013] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE IL-1β is a master switch of inflammation and plays an important role in the pathogenesis of vascular disease. During early atherosclerosis development, it is not clearly understood how oxidized low density lipoprotein (oxLDL)induced signaling pathways control NLRP3 inflammasome activation and produce IL-1β and promote foam cells formation. METHODS The study used THP-1 macrophage as cell model. Western blot quantified the oxLDL-induced NLRP3 inflammasome related proteins. The FACS detected the expression of SR-A and CD36 receptors on the cells, and caspase-1 activation in the cells. The DCFH-DA assayed the reactive oxygen species (ROS). Oil red O staining techniques examined the intracellular lipid droplet. RESULTS The OxLDL remarkably increased not only IL-1β mRNA transcription and pro-IL-1β protein synthesis but also IL-1β secretion in human macrophages. The activation of the NLRP3 inflammasome depended on oxLDL-induced generation of ROS, potassium efflux and cathepsin B activity. The OxLDL-induced ROS production that mediates IL-1β maturation mainly depended on the scavenger receptor of CD36 but not SR-A. The secreted IL-1β served as an autocrine function for promoting macrophage foam cells formation. CONCLUSIONS These findings suggest that oxLDL-induced NLRP3 inflammasome activation mainly depends on CD36 involved in the progression of atherosclerosis by promoting oxLDL-mediated inflammation and foam cell formation.
Collapse
|
37
|
Abstract
Reactive oxygen species (ROS) are deadly weapons used by phagocytes and other cell types, such as lung epithelial cells, against pathogens. ROS can kill pathogens directly by causing oxidative damage to biocompounds or indirectly by stimulating pathogen elimination by various nonoxidative mechanisms, including pattern recognition receptors signaling, autophagy, neutrophil extracellular trap formation, and T-lymphocyte responses. Thus, one should expect that the inhibition of ROS production promote infection. Increasing evidences support that in certain particular infections, antioxidants decrease and prooxidants increase pathogen burden. In this study, we review the classic infections that are controlled by ROS and the cases in which ROS appear as promoters of infection, challenging the paradigm. We discuss the possible mechanisms by which ROS could promote particular infections. These mechanisms are still not completely clear but include the metabolic effects of ROS on pathogen physiology, ROS-induced damage to the immune system, and ROS-induced activation of immune defense mechanisms that are subsequently hijacked by particular pathogens to act against more effective microbicidal mechanisms of the immune system. The effective use of antioxidants as therapeutic agents against certain infections is a realistic possibility that is beginning to be applied against viruses.
Collapse
Affiliation(s)
- Claudia N Paiva
- Departamento de Imunologia, Instituto de Microbiologia , CCS Bloco D, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | |
Collapse
|
38
|
Kumar P, Raghavan S, Shanmugam G, Shanmugam N. Ligation of RAGE with ligand S100B attenuates ABCA1 expression in monocytes. Metabolism 2013; 62:1149-58. [PMID: 23523156 DOI: 10.1016/j.metabol.2013.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 02/04/2013] [Accepted: 02/19/2013] [Indexed: 01/17/2023]
Abstract
HYPOTHESIS ATP Binding Cassette Transporter (ABC) A1 is one of the key regulators of HDL synthesis and reverse cholesterol transport. Activation of Receptors for Advanced Glycation End products (RAGE) is involved in the pathogenesis of diabetes, and its complications. The aim of the present study is to examine the effect of RAGE ligand S100B on ABCA1 expression. METHODS S100B mediated regulation of LXR target genes like ABCA1, ABCG1, ABCG8, LXR-α and LXR-β in THP-1 cells was analyzed by real-time PCR, RT-PCR and western blots. ABCA1 mRNA expression in monocytes from diabetic patients was studied. Effect of LXR ligand on S100B induced changes in LXR target genes was also studied. Luciferase reporter assay was used for S100B induced ABCA1 promoter regulation. RESULTS S100B treatment resulted in a significant 2-3 fold reduction (p<0.01) in ABCA1 and ABCG1 mRNA in dose and time dependent manner in THP1 cells. ABCA1 protein level was also significantly (p<0.01) reduced. S100B-induced reduction on ABCA1 mRNA expression was blocked by treating THP-1 cell with anti-RAGE antibody. Reduced ABCA1 mRNA levels seen in peripheral blood monocytes from diabetes patients showed the in-vivo relevance of our in-vitro results. Effect of S100B on ABCA1 and ABCG1 expression was reversed by LXR ligand treatment. S100B treatment showed significant 2 fold (p<0.01) decrease in T1317 induced ABCA1 promoter activation. CONCLUSIONS These results show for the first time that ligation of RAGE with S100B can attenuate the expression of ABCA1 and ABCG1 through the LXRs. This could reduce ApoA-I-mediated cholesterol efflux from monocytes.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Diabetes and Cardiovascular Research Laboratory, Department of Biomedical Science, Bharathidasan University, Tiruchirappalli 620 024, Tamilnadu, India
| | | | | | | |
Collapse
|
39
|
Chang YC, Sheu WHH, Chien YS, Tseng PC, Lee WJ, Chiang AN. Hyperglycemia accelerates ATP-binding cassette transporter A1 degradation via an ERK-dependent pathway in macrophages. J Cell Biochem 2013; 114:1364-73. [DOI: 10.1002/jcb.24478] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 12/05/2012] [Indexed: 11/11/2022]
|
40
|
Straliotto MR, de Oliveira J, Mancini G, Bainy AC, Latini A, Deobald AM, Rocha JB, de Bem AF. Disubstituted diaryl diselenides as potential atheroprotective compounds: Involvement of TrxR and GPx-like systems. Eur J Pharm Sci 2013; 48:717-25. [DOI: 10.1016/j.ejps.2013.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 01/03/2013] [Indexed: 02/08/2023]
|
41
|
Okuda LS, Castilho G, Rocco DD, Nakandakare ER, Catanozi S, Passarelli M. Advanced glycated albumin impairs HDL anti-inflammatory activity and primes macrophages for inflammatory response that reduces reverse cholesterol transport. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:1485-92. [DOI: 10.1016/j.bbalip.2012.08.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 08/03/2012] [Accepted: 08/15/2012] [Indexed: 12/12/2022]
|
42
|
Alba G, Reyes ME, Santa-María C, Ramírez R, Geniz I, Jiménez J, Martín-Nieto J, Pintado E, Sobrino F. Transcription of liver X receptor is down-regulated by 15-deoxy-Δ(12,14)-prostaglandin J(2) through oxidative stress in human neutrophils. PLoS One 2012; 7:e42195. [PMID: 23115616 PMCID: PMC3480349 DOI: 10.1371/journal.pone.0042195] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 07/04/2012] [Indexed: 01/04/2023] Open
Abstract
Liver X receptors (LXRs) are ligand-activated transcription factors of the nuclear receptor superfamily. They play important roles in controlling cholesterol homeostasis and as regulators of inflammatory gene expression and innate immunity, by blunting the induction of classical pro-inflammatory genes. However, opposite data have also been reported on the consequences of LXR activation by oxysterols, resulting in the specific production of potent pro-inflammatory cytokines and reactive oxygen species (ROS). The effect of the inflammatory state on the expression of LXRs has not been studied in human cells, and constitutes the main aim of the present work. Our data show that when human neutrophils are triggered with synthetic ligands, the synthesis of LXRα mRNA became activated together with transcription of the LXR target genes ABCA1, ABCG1 and SREBP1c. An inflammatory mediator, 15-deoxy-Δ12,14-prostaglandin J2 (15dPGJ2), hindered T0901317-promoted induction of LXRα mRNA expression together with transcription of its target genes in both neutrophils and human macrophages. This down-regulatory effect was dependent on the release of reactive oxygen species elicited by 15dPGJ2, since it was enhanced by pro-oxidant treatment and reversed by antioxidants, and was also mediated by ERK1/2 activation. Present data also support that the 15dPGJ2-induced serine phosphorylation of the LXRα molecule is mediated by ERK1/2. These results allow to postulate that down-regulation of LXR cellular levels by pro-inflammatory stimuli might be involved in the development of different vascular diseases, such as atherosclerosis.
Collapse
Affiliation(s)
- Gonzalo Alba
- Departamento de Bioquímica Médica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - María Edith Reyes
- Departamento de Bioquímica Médica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - Consuelo Santa-María
- Departamento de Bioquímica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - Remedios Ramírez
- Departamento de Bioquímica Médica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - Isabel Geniz
- Distrito Sanitario Sevilla Norte, Servicio Andaluz de Salud, Sevilla, Spain
| | - Juan Jiménez
- Departamento de Bioquímica Médica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - José Martín-Nieto
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Alicante, Spain
| | - Elízabeth Pintado
- Departamento de Bioquímica Médica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - Francisco Sobrino
- Departamento de Bioquímica Médica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
- * E-mail:
| |
Collapse
|
43
|
Song D, Fang G, Mao SZ, Ye X, Liu G, Gong Y, Liu SF. Chronic intermittent hypoxia induces atherosclerosis by NF-κB-dependent mechanisms. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1650-9. [PMID: 22846605 DOI: 10.1016/j.bbadis.2012.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 07/16/2012] [Accepted: 07/23/2012] [Indexed: 02/06/2023]
Abstract
Chronic intermittent hypoxia (CIH) causes atherosclerosis in mice fed a high cholesterol diet (HCD). The mechanisms by which CIH promotes atherosclerosis are incompletely understood. This study defined the mechanistic role of NF-κB pathway in CIH+HCD induced atherosclerosis. Wild type (WT) and mice deficient in the p50 subunit of NF-κB (p50-KO) were fed normal chow diet (ND) or HCD, and exposed to sham or CIH. Atherosclerotic lesions on the en face aortic preparation and cross-sections of aortic root were examined. In WT mice, neither CIH nor HCD exposure alone caused, but CIH+HCD caused evident atherosclerotic lesions on both preparations after 20weeks of exposure. WT mice on ND and exposed to CIH for 35.6weeks did not develop atherosclerotic lesions. P50 gene deletion diminished CIH+HCD induced NF-κB activation and abolished CIH+HCD induced atherosclerosis. P50 gene deletion inhibited vascular wall inflammation, reduced hepatic TNF-α level, attenuated the elevation in serum cholesterol level and diminished macrophage foam cell formation induced by CIH+HCD exposure. These results demonstrate that inhibition of NF-κB activation abrogates the activation of three major atherogenic mechanisms associated with an abolition of CIH+HCD induced atherosclerosis. NF-κB may be a central common pathway through which CIH+HCD exposure activates multiple atherogenic mechanisms, leading to atherosclerosis.
Collapse
Affiliation(s)
- D Song
- Centers for Heart and Lung Research and Pulmonary, Critical Care and Sleep Medicine, the Feinstein Institute for Medical Research, Manhasset, NY, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
ten Freyhaus H, Calay ES, Yalcin A, Vallerie SN, Yang L, Calay ZZ, Saatcioglu F, Hotamisligil GS. Stamp2 controls macrophage inflammation through nicotinamide adenine dinucleotide phosphate homeostasis and protects against atherosclerosis. Cell Metab 2012; 16:81-9. [PMID: 22704678 PMCID: PMC4163924 DOI: 10.1016/j.cmet.2012.05.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/06/2012] [Accepted: 05/11/2012] [Indexed: 01/21/2023]
Abstract
The six-transmembrane protein Stamp2 plays an important role in metabolically triggered inflammation and insulin action. We report that Stamp2 is expressed in human and mouse macrophages, is regulated upon differentiation or activation, acts as an anti-inflammatory protein, and regulates foam cell formation. Absence of Stamp2 results in significant increases in cellular NADPH levels, and both NADPH homeostasis and the exaggerated inflammatory response of Stamp2(-/-) macrophages are rescued by exogenous wild-type but not by a reductase-deficient Stamp2 molecule. Chemical and genetic suppression of NADPH production in Stamp2(-/-) macrophages reverts the heightened inflammatory response. Stamp2 is detected in mouse and human atherosclerotic plaques, and its deficiency promotes atherosclerosis in mice. Furthermore, bone marrow transplantation experiments demonstrated that Stamp2 in myeloid cells is sufficient to protect against atherosclerosis. Our data reveal a role of Stamp2 in controlling intermediary metabolites to regulate inflammatory responses in macrophages and in progression of atherosclerosis.
Collapse
Affiliation(s)
- Henrik ten Freyhaus
- Department of Genetics, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
PAPP-A negatively regulates ABCA1, ABCG1 and SR-B1 expression by inhibiting LXRα through the IGF-I-mediated signaling pathway. Atherosclerosis 2012; 222:344-54. [DOI: 10.1016/j.atherosclerosis.2012.03.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 02/16/2012] [Accepted: 03/06/2012] [Indexed: 01/14/2023]
|
46
|
Gu MX, Fu Y, Sun XL, Ding YZ, Li CH, Pang W, Pan S, Zhu Y. Proteomic analysis of endothelial lipid rafts reveals a novel role of statins in antioxidation. J Proteome Res 2012; 11:2365-73. [PMID: 22428589 DOI: 10.1021/pr300098f] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
As inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, statins have pleiotropic vascular-protective effects, such as anti-inflammatory and antioxidative effects. We investigated the short-term beneficial effects of statins on modulating the translocation of lipid-raft-related proteins in endothelial cells (ECs). Human umbilical vein ECs were treated with atorvastatin for 30 min or 2 h; lipid-raft proteins were isolated and examined by quantitative proteome assay. Functional classification of identified proteins in lipid rafts revealed upregulated antioxidative proteins; downregulated proteins were associated with inflammation and cell adhesion. Among proteins verified by Western blot analysis, endoplasmic reticulum protein 46 (ERp46) showed increased level in lipid rafts with atorvastatin. Further, atorvastatin inhibited the activation of membrane-bound NADPH oxidase in both untreated and angiotensin II-treated ECs, as shown by reduced reactive oxygen species production. Co-immunoprecipitation and immunofluorescence experiments revealed that atorvastatin increased the association of ERp46 and Nox2, an NADPH oxidase isoform, in lipid rafts, thereby inhibiting Nox2 assembly with its regulatory subunits, such as p47phox and p67phox. Our results reveal a novel antioxidative role of atorvastatin by promoting the membrane translocation of ERp46 and its binding with Nox2 to inhibit Nox2 activity in ECs, which may offer another insight into the pleiotropic functions of statins.
Collapse
Affiliation(s)
- Ming-Xia Gu
- Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Sciences of Education Ministry, Peking University Health Science Center, Beijing, 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ota M, Fujii T, Nemoto K, Tatsumi M, Moriguchi Y, Hashimoto R, Sato N, Iwata N, Kunugi H. A polymorphism of the ABCA1 gene confers susceptibility to schizophrenia and related brain changes. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1877-83. [PMID: 21839797 DOI: 10.1016/j.pnpbp.2011.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/14/2011] [Accepted: 07/26/2011] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The ATP-binding cassette transporter A1 (ABCA1) mediates cellular cholesterol efflux through the transfer of cholesterol from the inner to the outer layer of the cell membrane and regulates extracellular cholesterol levels in the central nervous system. Several lines of evidence have indicated lipid and myelin abnormalities in schizophrenia. METHOD Initially, we examined the possible association of the polymorphisms of the ABCA1 gene (ABCA1) with susceptibility to schizophrenia in 506 patients with schizophrenia (DSM-IV) and 941 controls. The observed association was then subject to a replication analysis in an independent sample of 511 patients and 539 controls. We further examined the possible effect of the risk allele on gray matter volume assessed with magnetic resonance imaging (MRI) in 86 patients with schizophrenia (49 males) and 139 healthy controls (47 males). RESULTS In the initial association study, the 1587 K allele (rs2230808) was significantly more common in male patients with schizophrenia than in male controls. Although such a significant difference was not observed in the second sample alone, the increased frequency of the 1587 K allele in male patients remained to be significant in the combined male sample of 556 patients and 594 controls. Male schizophrenia patients carrying the 1587 K allele had a smaller amount of gray matter volume than those who did not carry the allele. CONCLUSION Our data suggest a male-specific association of the 1587 K allele of ABCA1 with susceptibility to schizophrenia and smaller gray matter volume in schizophrenia.
Collapse
Affiliation(s)
- Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Curcumin promotes cholesterol efflux from adipocytes related to PPARgamma-LXRalpha-ABCA1 passway. Mol Cell Biochem 2011; 358:281-5. [PMID: 21748336 DOI: 10.1007/s11010-011-0978-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/29/2011] [Indexed: 12/20/2022]
Abstract
Curcumin affects the functions of adipocytes. But it is not known whether curcumin has some effect on the cholesterol efflux process of adipocytes. Rabbit subcutaneous adipocytes were incubated with 5, 10 and 20 μg/ml curcumin for 24 h. The cholesterol efflux onto apoAI was assessed, and the peroxisome proliferators-activated receptor (PPAR) γ, liver X receptor (LXR) α and ATP-binding cassette transporter A1 (ABCA1) mRNA expression in adipocytes were quantified by reverse-transcription polymerase chain reaction (RT-PCR). Curcumin increased the cholesterol efflux from adipocytes in dose-dependent manner. The increased expression of PPARγ, LXRα and ABCA1 caused by curcumin were parallel. When the adipocytes were pre-treated by GW9662, the increased expression of PPARγ induced by curcumin was partially prevented, subsequent to the down-regulation of LXRα and ABCA1. Curcumin can affect the cholesterol efflux from adipocytes by regulating the PPARγ-LXR-ABCA1 passway.
Collapse
|
49
|
Fan D, Li L, Wang C, Cui XB, Zhou Y, Wu LL. Adiponectin induces interleukin-6 production and its underlying mechanism in adult rat cardiac fibroblasts. J Cell Physiol 2011; 226:1793-802. [PMID: 21069809 DOI: 10.1002/jcp.22512] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It has been reported that adiponectin enhances interleukin-6 (IL-6) production in cardiac fibroblasts derived from neonatal rats and adult mice, but the mechanisms involved remain unknown. In the present study, we explored the effect and mechanisms of adiponectin on IL-6 production in adult rat cardiac fibroblasts. Globular adiponectin (gAd) increased IL-6 mRNA expression and protein secretion in cultured adult rat cardiac fibroblasts. gAd-induced IL-6 release was attenuated after RNA interference inhibition of adiponectin receptor 1 (AdipoR1), but not AdipoR2 or an adaptor protein APPL1. gAd increased the phosphorylation of AMP-activated protein kinase (AMPK), p38 mitogen-activated protein kinase (p38MAPK), extracellular signal-regulated kinase 1/2 (ERK1/2), and c-Jun-N-terminal kinase (JNK). Inhibitors of AMPK (araA), p38MAPK (SB202190), and ERK1/2 (PD98059 and U0126) but not JNK (SP600125) suppressed gAd-induced IL-6 production. In transient transfection assays of IL-6 promoter/luciferase reporter plasmids, gAd increased the transcriptional activity of the full-length IL-6 promoter. Deletion analysis of the IL-6 promoter indicated that activator protein-1 (AP-1), nuclear factor for IL-6 (NF-IL-6) and nuclear factor κB (NF-κB) binding sites were important for gAd-induced IL-6 transcription. Our data suggest that gAd enhances IL-6 synthesis and release in adult rat cardiac fibroblasts through AdipoR1. Activation of AMPK, p38MAPK, and ERK1/2 mediates the intracellular signal transduction. AP-1, NF-IL-6, and NF-κB cis-elements are required for gAd-induced IL-6 transcription.
Collapse
Affiliation(s)
- Dong Fan
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | | | | | | | | | | |
Collapse
|
50
|
McLaren JE, Michael DR, Ashlin TG, Ramji DP. Cytokines, macrophage lipid metabolism and foam cells: implications for cardiovascular disease therapy. Prog Lipid Res 2011; 50:331-47. [PMID: 21601592 DOI: 10.1016/j.plipres.2011.04.002] [Citation(s) in RCA: 278] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 04/14/2011] [Accepted: 04/14/2011] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease is the biggest killer globally and the principal contributing factor to the pathology is atherosclerosis; a chronic, inflammatory disorder characterized by lipid and cholesterol accumulation and the development of fibrotic plaques within the walls of large and medium arteries. Macrophages are fundamental to the immune response directed to the site of inflammation and their normal, protective function is harnessed, detrimentally, in atherosclerosis. Macrophages contribute to plaque development by internalizing native and modified lipoproteins to convert them into cholesterol-rich foam cells. Foam cells not only help to bridge the innate and adaptive immune response to atherosclerosis but also accumulate to create fatty streaks, which help shape the architecture of advanced plaques. Foam cell formation involves the disruption of normal macrophage cholesterol metabolism, which is governed by a homeostatic mechanism that controls the uptake, intracellular metabolism, and efflux of cholesterol. It has emerged over the last 20 years that an array of cytokines, including interferon-γ, transforming growth factor-β1, interleukin-1β, and interleukin-10, are able to manipulate these processes. Foam cell targeting, anti-inflammatory therapies, such as agonists of nuclear receptors and statins, are known to regulate the actions of pro- and anti-atherogenic cytokines indirectly of their primary pharmacological function. A clear understanding of macrophage foam cell biology will hopefully enable novel foam cell targeting therapies to be developed for use in the clinical intervention of atherosclerosis.
Collapse
Affiliation(s)
- James E McLaren
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | | | | | | |
Collapse
|