1
|
Bartoloni E, Cacciapaglia F, Erre GL, Gremese E, Manfredi A, Piga M, Sakellariou G, Spinelli FR, Viapiana O, Atzeni F. Immunomodulation for accelerated atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus. Autoimmun Rev 2025; 24:103760. [PMID: 39894242 DOI: 10.1016/j.autrev.2025.103760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
In the last decades, consisting evidence supported a close relationship between both innate and adaptive immune systems and the accelerated cardiovascular (CV) disease characterizing autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Indeed, several cell lines involved in the pathogenesis of these autoimmune diseases, such as macrophages and dendritic cells, as well as different T and B lymphocyte subsets, and inflammatory cytokines, have been demonstrated to be directly involved in the mechanisms underlying early atherosclerotic arterial wall damage. Traditional CV risk factors play a concomitant role but do not sufficiently account for the increased prevalence of CV disease in these patients. Indeed, the pathophysiological link between RA and SLE and atherosclerosis is based on complex inflammatory pathways that interconnect these conditions and may explain the significant morbidity and mortality rates demonstrated in these patients, with consequent significant negative effects on quality of life and long-term survival. Consequently, it is intriguing to hypothesize that immunosuppressive drugs commonly used in the treatment of these pathologies may also exert an immunomodulatory and anti-inflammatory effect in mitigating the atherosclerotic damage that has been demonstrated to occur early in the initial stages of the disease. Recognizing risk factors, predicting occurrences and early intervention to prevent CV disease development have emerged as critical objectives in RA and SLE treatment. In this review, we aimed to provide an updated overview of the atherogenic effects exerted by the immune and inflammatory pathways involved in the pathogenesis of RA and SLE. Moreover, we examined the available evidence which may support the potential effects of immunosuppressive therapies in reducing CV damage and, consequently, CV disease risk in these patients.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Italy
| | - Fabio Cacciapaglia
- Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DePReMeI), University of Bari, Bari, Italy; Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro" Casamassima & Rheumatology Service "Miulli" General Hospital Acquaviva delle Fonti, Bari, Italy
| | - Gian Luca Erre
- Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Italy
| | - Elisa Gremese
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andreina Manfredi
- University of Modena and Reggio Emilia,AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Matteo Piga
- Rheumatology Unit, AOU Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Italy
| | - Garifallia Sakellariou
- Department of Internal Medicine and Therapeutics, University of Pavia, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Francesca Romana Spinelli
- Reumatology Unit, Department of Internal, Anesthesiological, and Cardiovascular Clinical Sciences, Sapienza University of Rome, Rome, Italy
| | - Ombretta Viapiana
- Rheumatology Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Italy.
| |
Collapse
|
2
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
3
|
Boosani CS, Burela L. The Exacerbating Effects of the Tumor Necrosis Factor in Cardiovascular Stenosis: Intimal Hyperplasia. Cancers (Basel) 2024; 16:1435. [PMID: 38611112 PMCID: PMC11010976 DOI: 10.3390/cancers16071435] [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: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
TNF-α functions as a master regulator of inflammation, and it plays a prominent role in several immunological diseases. By promoting important cellular mechanisms, such as cell proliferation, migration, and phenotype switch, TNF-α induces its exacerbating effects, which are the underlying cause of many proliferative diseases such as cancer and cardiovascular disease. TNF-α primarily alters the immune component of the disease, which subsequently affects normal functioning of the cells. Monoclonal antibodies and synthetic drugs that can target TNF-α and impair its effects have been developed and are currently used in the treatment of a few select human diseases. Vascular restenosis is a proliferative disorder that is initiated by immunological mechanisms. In this review, the role of TNF-α in exacerbating restenosis resulting from neointimal hyperplasia, as well as molecular mechanisms and cellular processes affected or induced by TNF-α, are discussed. As TNF-α-targeting drugs are currently not approved for the treatment of restenosis, the summation of the topics discussed here is anticipated to provide information that can emphasize on the use of TNF-α-targeting drug candidates to prevent vascular restenosis.
Collapse
Affiliation(s)
- Chandra Shekhar Boosani
- Somatic Cell and Genome Editing Center, Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
- MU HealthCare, University of Missouri, Columbia, MO 65211, USA
- Technology and Platform Development, Soma Life Science Solutions, Winston-Salem, NC 27103, USA
| | | |
Collapse
|
4
|
Karpouzas GA, Papotti B, Ormseth SR, Palumbo M, Hernandez E, Adorni MP, Zimetti F, Budoff MJ, Ronda N. Inflammation and immunomodulatory therapies influence the relationship between ATP-binding cassette A1 membrane transporter-mediated cholesterol efflux capacity and coronary atherosclerosis in rheumatoid arthritis. J Transl Autoimmun 2023; 7:100209. [PMID: 37520890 PMCID: PMC10371792 DOI: 10.1016/j.jtauto.2023.100209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023] Open
Abstract
Objectives High-density lipoprotein (HDL) removes cholesterol from cells in atherosclerotic lesions, a function known as cholesterol efflux capacity (CEC). ATP-binding-cassette A1 (ABCA1) membrane transporter starts cholesterol transfer from macrophages to HDL particles. In rheumatoid arthritis (RA), methotrexate and biologic disease modifying drugs (bDMARDs) are atheroprotective whereas corticosteroids and C-reactive protein (CRP) are proatherogenic. We evaluated the influence of these factors on the relationship of ABCA1-CEC with atherosclerosis and cardiovascular events. Methods Atherosclerosis was evaluated with computed tomography angiography in 140 patients with RA and repeated in 99 after 6.9 ± 0.3 years. Events including acute coronary syndromes, stroke, cardiovascular death, claudication, revascularization, and heart failure were recorded. ABCA1-CEC was quantified in J774A.1 murine macrophages and reported as percentage of effluxed over intracellular cholesterol. Results Higher ABCA1-CEC associated with (i) more calcified plaques at baseline only in patients with CRP>7 mg/L (median) (p-interaction = 0.001) and methotrexate nonusers (p-interaction = 0.037), and more partially-calcified plaques only in bDMARD nonusers (p-interaction = 0.029); (ii) fewer new calcified plaques in patients with below-median but not higher time-averaged CRP (p-interaction = 0.028); (iii) fewer new total and calcified plaques in prednisone unexposed but not patients exposed to prednisone during follow-up (p-interaction = 0.034 and 0.004) and (iv) more new plaques in baseline bDMARD nonusers and fewer in bDMARD users (p-interaction ≤ 0.001). Also, ABCA1-CEC associated with greater cardiovascular risk only in baseline prednisone users (p-interaction = 0.027). Conclusion ABCA1-CEC associated with decreased atherosclerosis in patients with below-median baseline and time-averaged CRP and bDMARD use. Conversely, ABCA1-CEC associated with increased plaque in those with higher CRP, corticosteroid users, methotrexate nonusers, and bDMARD nonusers. While in well-treated and controlled disease ABCA1-CEC appears atheroprotective, in uncontrolled RA its action may be masked or fail to counteract the inflammation-driven proatherogenic state.
Collapse
Affiliation(s)
- George A. Karpouzas
- Division of Rheumatology, Harbor-UCLA and The Lundquist Institute, Torrance, CA, USA
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Sarah R. Ormseth
- Division of Rheumatology, Harbor-UCLA and The Lundquist Institute, Torrance, CA, USA
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | | | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Matthew J. Budoff
- Division of Cardiology, Harbor-UCLA and The Lundquist Institute, Torrance, CA, USA
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| |
Collapse
|
5
|
Bhardwaj A, Singh A, Midha V, Sood A, Wander GS, Mohan B, Batta A. Cardiovascular implications of inflammatory bowel disease: An updated review. World J Cardiol 2023; 15:553-570. [PMID: 38058397 PMCID: PMC10696203 DOI: 10.4330/wjc.v15.i11.553] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/22/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
Emerging data highlights the heightened risk of atherosclerotic cardiovascular diseases (ASCVD) in patients with chronic inflammatory disorders, particularly those afflicted with inflammatory bowel disease (IBD). This review delves into the epidemiological connections between IBD and ASCVD, elucidating potential underlying mechanisms. Furthermore, it discusses the impact of current IBD treatments on cardiovascular risk. Additionally, the cardiovascular adverse effects of novel small molecule drugs used in moderate-to-severe IBD are investigated, drawing parallels with observations in patients with rheumatoid arthritis. This article aims to comprehensively evaluate the existing evidence supporting these associations. To achieve this, we conducted a meticulous search of PubMed, spanning from inception to August 2023, using a carefully selected set of keywords. The search encompassed topics related to IBD, such as Crohn's disease and ulcerative colitis, as well as ASCVD, including coronary artery disease, cardiovascular disease, atrial fibrillation, heart failure, conduction abnormalities, heart blocks, and premature coronary artery disease. This review encompasses various types of literature, including retrospective and prospective cohort studies, clinical trials, meta-analyses, and relevant guidelines, with the objective of providing a comprehensive overview of this critical intersection of inflammatory bowel disease and cardiovascular health.
Collapse
Affiliation(s)
- Arshia Bhardwaj
- Department of Gastroenterology, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India
| | - Arshdeep Singh
- Department of Gastroenterology, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India
| | - Vandana Midha
- Department of Internal Medicine, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India
| | - Ajit Sood
- Department of Gastroenterology, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India
| | - Gurpreet Singh Wander
- Department of Cardiology, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India
| | - Bishav Mohan
- Department of Cardiology, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India
| | - Akash Batta
- Department of Cardiology, Dayanand Medical College and Hospital, Punjab, Ludhiana 141001, India.
| |
Collapse
|
6
|
Ma L, Du Y, Ma C, Liu M. Association of HMGCR inhibition with rheumatoid arthritis: a Mendelian randomization and colocalization study. Front Endocrinol (Lausanne) 2023; 14:1272167. [PMID: 38047111 PMCID: PMC10691537 DOI: 10.3389/fendo.2023.1272167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/25/2023] [Indexed: 12/05/2023] Open
Abstract
Objective The objective of this study was to investigate the association between hydroxymethylglutaryl coenzyme A reductase (HMGCR) inhibition and rheumatoid arthritis (RA) using drug-target Mendelian randomization (MR) and genetic colocalization analyses. Methods Two sets of genetic instruments were employed to proxy HMGCR inhibitors: expression quantitative trait loci (eQTLs) of target genes from the eQTLGen Consortium and genetic variants associated with low-density lipoprotein cholesterol (LDL-C) levels with HMGCR locus from open genome-wide association studies (GWAS). Positive control analyses were conducted on type 2 diabetes and coronary heart disease, and multiple sensitivity analyses were performed. Results Genetically proxied expression of eQTL was associated with a lower risk of RA (OR=0.996, 95% CI =0.992-0.999, p= 0.032). Similarly, hydroxymethylglutaryl coenzyme A reductase (HMGCR)-mediated low-density lipoprotein cholesterol was negatively associated with risk of RA (OR=0.995, 95% CI =0.991-0.998, p= 0.007) in the inverse variance weighted (IVW) method. Colocalization analysis suggested a 74.6% posterior probability of sharing a causal variant within the SNPs locus (PH4 = 74.6%). A causal relationship also existed between HMGCR-mediated LDL and RA risk factors. The results were also confirmed by multiple sensitivity analyses. The results in positive control were consistent with the previous study. Conclusion Our study suggested that HMGCR inhibition was associated with an increased risk of RA while also highlighting an increased risk of current smoking and obesity. These findings contribute to a growing body of evidence regarding the adverse effects of HMGCR inhibition on RA risk, calling for further research on alternative approaches using HMGCR inhibitors in RA management.
Collapse
Affiliation(s)
- Li Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Department of General Practice, Heze Municiple Hospital, Heze, Shandong, China
| | - Yufei Du
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Ma
- Department of Urology, Heze Municiple Hospital, Heze, Shandong, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
7
|
Mirza Z, Al-Saedi DA, Saddeek S, Almowallad S, AlMassabi RF, Huwait E. Atheroprotective Effect of Fucoidan in THP-1 Macrophages by Potential Upregulation of ABCA1. Biomedicines 2023; 11:2929. [PMID: 38001931 PMCID: PMC10669811 DOI: 10.3390/biomedicines11112929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Targeting foam cells reduces the risk and pathophysiology of atherosclerosis, of which they are one of its early hallmarks. The precise mechanism of action of fucoidan, a potential anti-atherogenic drug, is still unknown. Our objective was to assess the ability of fucoidan to regulate expression of ATP-binding cassette transporter A1 (ABCA1) in ox-LDL-induced THP-1 macrophages. Molecular docking was used to predict how fucoidan interacts with anti-foam cell markers, and further in vitro experiments were performed to evaluate the protective effect of fucoidan on modulating uptake and efflux of lipids. THP-1 macrophages were protected by 50 µg/mL of fucoidan and were then induced to form foam cells with 25 µg/mL of ox-LDL. Expression levels were assessed using RT-qPCR, and an Oil Red O stain was used to observe lipid accumulation in THP-1 macrophages. In addition, ABCA1 protein was examined by Western blot, and cellular cholesterol efflux was determined using fluorescently labeled cholesterol. Under a light microscope, decreased lipid accumulation in ox-LDL-induced-THP-1 macrophages pre-treated with fucoidan showed a significant effect, although it did not affect the expression of scavenger receptors (SR-AI and CD36). It is interesting to note that fucoidan dramatically increased the gene and protein expression of ABCA1, perhaps via the liver X receptor-α (LXR-α). Moreover, fucoidan's ability to increase and control the efflux of cholesterol from ox-LDL-induced THP-1 macrophages revealed how it may alter ABCA1's conformation and have a major effect on how it interacts with apolipoprotein A (ApoA1). In vitro results support a rationale for predicting fucoidan and its interaction with its receptor targets' predicted data, hence validating its anti-atherogenic properties and suggesting that fucoidan could be promising as an atheroprotective.
Collapse
Affiliation(s)
- Zeenat Mirza
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Dalal A. Al-Saedi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Cell Culture Lab, Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Salma Saddeek
- Department of Chemistry, Faculty of Sciences, University of Hafr Al Batin, Hafr Al Batin 39511, Saudi Arabia;
| | - Sanaa Almowallad
- Department of Biochemistry, Faculty of Sciences, University of Tabuk, Tabuk 48322, Saudi Arabia (R.F.A.)
| | - Rehab F. AlMassabi
- Department of Biochemistry, Faculty of Sciences, University of Tabuk, Tabuk 48322, Saudi Arabia (R.F.A.)
| | - Etimad Huwait
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Cell Culture Lab, Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
8
|
Gabbiadini R, Dal Buono A, Mastrorocco E, Solitano V, Repici A, Spinelli A, Condorelli G, Armuzzi A. Atherosclerotic cardiovascular diseases in inflammatory bowel diseases: to the heart of the issue. Front Cardiovasc Med 2023; 10:1143293. [PMID: 37260950 PMCID: PMC10227624 DOI: 10.3389/fcvm.2023.1143293] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Atherosclerotic cardiovascular disease and stroke are the leading causes of morbidity and mortality worldwide. Along to the traditional risk factors for these diseases, chronic inflammation is known to be an important player in accelerating the process of atherosclerosis, which can result in an increased incidence of arterial thromboembolic events. As in other chronic inflammatory diseases, in the past few years, several studies suggested that subjects affected by inflammatory bowel diseases (IBD) may also be at an incremented risk of atherosclerotic disease, especially during the periods of disease's flare. Therefore, IBD treatment may assume an important role for achieving both disease remission and the control of the atherosclerotic risk. In this article we aimed to perform a comprehensive review on evidence on the increased risk of arterial thromboembolic events in patients affected by IBD and discuss the potential role of IBD therapy in reducing this risk.
Collapse
Affiliation(s)
- Roberto Gabbiadini
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Arianna Dal Buono
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Elisabetta Mastrorocco
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Virginia Solitano
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Alessandro Repici
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Division of Colon and Rectal Surgery, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Cardiovascular Medicine, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alessandro Armuzzi
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
9
|
Ronda N, Zimetti F, Adorni MP, Palumbo M, Karpouzas GA, Bernini F. Role of Lipoprotein Levels and Function in Atherosclerosis Associated with Autoimmune Rheumatic Diseases. Rheum Dis Clin North Am 2023; 49:151-163. [PMID: 36424022 DOI: 10.1016/j.rdc.2022.07.006] [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
Immune and inflammatory mediators in autoimmune rheumatic diseases induce modification in the activity of enzymes pivotal for lipid metabolism and promote a proatherogenic serum lipid profile. However, disturbances in low- and high-density lipoprotein composition and increased lipid oxidation also occur. Therefore, lipoprotein dysfunction causes intracellular cholesterol accumulation in macrophages, smooth muscle cells, and platelets. Overall, both plaque progression and acute cardiovascular events are promoted. Single rheumatic diseases may present a particular pattern of lipid disturbances so that standard methods to evaluate cardiovascular risk may not be accurate enough. In general, antirheumatic drugs positively affect lipid metabolism in these patients.
Collapse
Affiliation(s)
- Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy.
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, Via Volturno 39/F, Parma 43125, Italy
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy
| | - George A Karpouzas
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute, Torrance, CA, USA
| | - Franco Bernini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy
| |
Collapse
|
10
|
Qian Y, Mao M, Nian F. The Effect of TNF- α on CHD and the Relationship between TNF- α Antagonist and CHD in Rheumatoid Arthritis: A Systematic Review. Cardiol Res Pract 2022; 2022:6192053. [PMID: 36060429 PMCID: PMC9433296 DOI: 10.1155/2022/6192053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) plays an important role in coronary heart disease (CHD), a chronic inflammatory process. Meanwhile, this pro-inflammatory factor is also involved in the pathogenesis of autoimmune diseases such as rheumatoid arthritis (RA). Patients with RA correspond to a higher risk of CHD. TNF-α antagonist, one of the main treatments for RA, may reduce the risk of CHD in patients with RA. This review summarizes the pathogenesis of TNF-α in CHD and discusses the relationship between TNF-α antagonist and CHD in patients with RA.
Collapse
Affiliation(s)
- Yezhou Qian
- Department of Cardiology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Menghui Mao
- Department of Cardiology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Feige Nian
- Department of Rheumatology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
11
|
Singh S, Changkija S, Mudgal R, Ravichandiran V. Bioactive components to inhibit foam cell formation in atherosclerosis. Mol Biol Rep 2022; 49:2487-2501. [PMID: 35013861 DOI: 10.1007/s11033-021-07039-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/30/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND The production of lipid-laden cells in macrophages after significant ingestion of oxidized low-density lipoprotein is considered the most critical phase in the creation of atherosclerotic lesions, which is known as foam cell formation. Targeting foam cell development to find a potential therapeutic strategy for the management of atherosclerosis has yielded numerous promising outcomes. Multiple variables influence foam cell growth, including scavenger receptor expression, cholesterol transporter expression acyl CoA: cholesterol acyltransferase activity, and neutral cholesteryl ester hydrolase activity. Plants used during herbal therapy have been shown to assist with a variety of ailments. RESULT In this study, we found medicinal plants and their bioactive components suppress foam cell formation in a variety of ways; some inhibit cholesterol transporter and lectin-like oxidized low-density lipoprotein receptor-1 upregulation, while others inhibit the function of acyl CoA: cholesterol acyltransferase activity, and neutral cholesteryl ester hydrolase activity. CONCLUSION Recent study findings related to the synthesis of the new active component from plant sources by focusing on the typical process involved in the generation of foam cells. We're also looking at using a cellular target-based therapeutic approach to generate novel plant-based medications for the cure of atherosclerosis.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Vaishali District, Hajipur, Bihar, 844102, India.
| | - Senti Changkija
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Vaishali District, Hajipur, Bihar, 844102, India
| | - Rajat Mudgal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Vaishali District, Hajipur, Bihar, 844102, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Vaishali District, Hajipur, Bihar, 844102, India
| |
Collapse
|
12
|
Luo Y, Ren X, Weng S, Yan C, Mao Q, Peng D. Improvements in High-Density Lipoprotein Quantity and Quality Contribute to the Cardiovascular Benefits by Anti-tumor Necrosis Factor Therapies in Rheumatoid Arthritis: A Systemic Review and Meta-Analysis. Front Cardiovasc Med 2021; 8:765749. [PMID: 34778416 PMCID: PMC8585789 DOI: 10.3389/fcvm.2021.765749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/30/2021] [Indexed: 12/29/2022] Open
Abstract
Objective: Inflammation plays important role in atherosclerotic cardiovascular diseases (CVDs), but the interaction between the inflammation and lipid profile is largely unrevealed in humans. Patients with rheumatoid arthritis (RA) suffer from a higher risk of CVDs. Decreased total cholesterol (TC) and high-density lipoprotein (HDL) were prevalent in patients with RA. Anti-tumor necrosis factor (TNF) therapies relieve disease activity and decrease CVDs risk in RA, but their comprehensive effects on the lipid profile are unclear. This study aims to investigate the changes in blood lipid profile along time in the patients with RA accepting anti-TNF therapies by meta-analysis. Methods: The MEDLINE, the Embase, and the Cochrane Central Register of Controlled Trials (CENTRAL) were searched for eligible literature. Data of lipids were classified into short-, mid-, and long-term according to treatment duration. Meta-analyses were performed to compare the lipid levels before and after treatments. Results: A total of 44 records and 3,935 patients were included in the meta-analyses. Anti-TNF therapies were associated with significant increase in TC [mean difference (MD): +0.14, +0.23, and +0.26 mmol/l, respectively] and HDL (MD): +0.11, +0.12, and +0.11 mmol/l, respectively) in the short-, mid-, and long-term; anti-TNF therapies were associated with increased low-density lipoprotein (LDL) (MD: +0.06 mmol/l) and apolipoprotein A1 (ApoA1) (MD: +0.07 g/l) in the short-term, but not in the mid-term and long-term; triglyceride (TG) and apolipoprotein B (ApoB) do not change significantly in all the periods; proatherosclerotic indexes (TC/HDL, ApoB/ApoA1, and LDL/HDL) tend to decrease in the short- and mid-term, but return to baseline in the long-term after TNF inhibition. Conclusion: Anti-TNF therapies were related to a long-term raised HDL level, which, together with evidence of improved HDL function, may contribute partially to the decreased CVDs risk by TNF inhibition.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaolei Ren
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuwei Weng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunhui Yan
- Department of Cardiovascular Medicine, Brain Hospital of Hunan Province, Changsha, China
| | - Qiaoxia Mao
- Department of Cardiovascular Medicine, Loudi Central Hospital, Loudi, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Abstract
Tumour necrosis factor (TNF) is a classical, pleiotropic pro-inflammatory cytokine. It is also the first 'adipokine' described to be produced from adipose tissue, regulated in obesity and proposed to contribute to obesity-associated metabolic disease. In this review, we provide an overview of TNF in the context of metabolic inflammation or metaflammation, its discovery as a metabolic messenger, its sites and mechanisms of action and some critical considerations for future research. Although we focus on TNF and the studies that elucidated its immunometabolic actions, we highlight a conceptual framework, generated by these studies, that is equally applicable to the complex network of pro-inflammatory signals, their biological activity and their integration with metabolic regulation, and to the field of immunometabolism more broadly.
Collapse
Affiliation(s)
- Jaswinder K Sethi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard-MIT Broad Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
- The Joslin Diabetes Center, Boston, MA, USA.
| |
Collapse
|
14
|
Identifying Infliximab- (IFX-) Responsive Blood Signatures for the Treatment of Rheumatoid Arthritis. BIOMED RESEARCH INTERNATIONAL 2021. [DOI: 10.1155/2021/5556784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Rheumatoid arthritis (RA) is a severe chronic pathogenic inflammatory abnormality that damages small joints. Comprehensive diagnosis and treatment procedures for RA have been established because of its severe symptoms and relatively high morbidity. Medication and surgery are the two major therapeutic approaches. Infliximab (IFX) is a novel biological agent applied for the treatment of RA. IFX improves physical functions and benefits the achievement of clinical remission even under discontinuous medication. However, not all patients react to IFX, and distinguishing IFX-sensitive and IFX-resistant patients is quite difficult. Thus, how to predict the therapeutic effects of IFX on patients with RA is one of the urgent translational medicine problems in the clinical treatment of RA. In this study, we present a novel computational method for the identification of the applicable and substantial blood gene signatures of IFX sensitivity by liquid biopsy, which may assist in the establishment of a clinical drug sensitivity test standard for RA and contribute to the revelation of unique IFX-associated pharmacological mechanisms.
Collapse
|
15
|
Serhal L, Lwin MN, Holroyd C, Edwards CJ. Rheumatoid arthritis in the elderly: Characteristics and treatment considerations. Autoimmun Rev 2020; 19:102528. [DOI: 10.1016/j.autrev.2020.102528] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/23/2022]
|
16
|
Carbone F, Bonaventura A, Liberale L, Paolino S, Torre F, Dallegri F, Montecucco F, Cutolo M. Atherosclerosis in Rheumatoid Arthritis: Promoters and Opponents. Clin Rev Allergy Immunol 2020; 58:1-14. [PMID: 30259381 DOI: 10.1007/s12016-018-8714-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Substantial epidemiological data identified cardiovascular (CV) diseases as a main cause of mortality in patients with rheumatoid arthritis (RA). In light of this, RA patients may benefit from additional CV risk screening and more intensive prevention strategies. Nevertheless, current algorithms for CV risk stratification still remain tailored on general population and are burdened by a significant underestimation of CV risk in RA patients. Acute CV events in patients with RA are largely related to an accelerated atherosclerosis. As pathophysiological features of atherosclerosis overlap those occurring in the inflamed RA synovium, the understanding of those common pathways represents an urgent need and a leading challenge for CV prevention in patients with RA. Genetic background, metabolic status, gut microbiome, and systemic inflammation have been also suggested as additional key pro-atherosclerotic factors. The aim of this narrative review is to update the current knowledge about pathophysiology of atherogenesis in RA patients and potential anti-atherosclerotic effects of disease-modifying anti-rheumatic drugs.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, 12 Wagistrasse, 8952, Schlieren, Switzerland
| | - Sabrina Paolino
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, San Martino Polyclinic Hospital, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132, Genoa, Italy
| | - Francesco Torre
- IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132, Genoa, Italy
- Clinic of Emergency Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Maurizio Cutolo
- IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132, Genoa, Italy.
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, San Martino Polyclinic Hospital, Genoa, Italy.
| |
Collapse
|
17
|
Frambach SJCM, de Haas R, Smeitink JAM, Rongen GA, Russel FGM, Schirris TJJ. Brothers in Arms: ABCA1- and ABCG1-Mediated Cholesterol Efflux as Promising Targets in Cardiovascular Disease Treatment. Pharmacol Rev 2020; 72:152-190. [PMID: 31831519 DOI: 10.1124/pr.119.017897] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular disease worldwide, and hypercholesterolemia is a major risk factor. Preventive treatments mainly focus on the effective reduction of low-density lipoprotein cholesterol, but their therapeutic value is limited by the inability to completely normalize atherosclerotic risk, probably due to the disease complexity and multifactorial pathogenesis. Consequently, high-density lipoprotein cholesterol gained much interest, as it appeared to be cardioprotective due to its major role in reverse cholesterol transport (RCT). RCT facilitates removal of cholesterol from peripheral tissues, including atherosclerotic plaques, and its subsequent hepatic clearance into bile. Therefore, RCT is expected to limit plaque formation and progression. Cellular cholesterol efflux is initiated and propagated by the ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. Their expression and function are expected to be rate-limiting for cholesterol efflux, which makes them interesting targets to stimulate RCT and lower atherosclerotic risk. This systematic review discusses the molecular mechanisms relevant for RCT and ABCA1 and ABCG1 function, followed by a critical overview of potential pharmacological strategies with small molecules to enhance cellular cholesterol efflux and RCT. These strategies include regulation of ABCA1 and ABCG1 expression, degradation, and mRNA stability. Various small molecules have been demonstrated to increase RCT, but the underlying mechanisms are often not completely understood and are rather unspecific, potentially causing adverse effects. Better understanding of these mechanisms could enable the development of safer drugs to increase RCT and provide more insight into its relation with atherosclerotic risk. SIGNIFICANCE STATEMENT: Hypercholesterolemia is an important risk factor of atherosclerosis, which is a leading pathological mechanism underlying cardiovascular disease. Cholesterol is removed from atherosclerotic plaques and subsequently cleared by the liver into bile. This transport is mediated by high-density lipoprotein particles, to which cholesterol is transferred via ATP-binding cassette transporters ABCA1 and ABCG1. Small-molecule pharmacological strategies stimulating these transporters may provide promising options for cardiovascular disease treatment.
Collapse
Affiliation(s)
- Sanne J C M Frambach
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ria de Haas
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan A M Smeitink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerard A Rongen
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
19
|
Li SS, Cao H, Shen DZ, Chen C, Xing SL, Dou FF, Jia QL. Effect of Quercetin on Atherosclerosis Based on Expressions of ABCA1, LXR-α and PCSK9 in ApoE -/- Mice. Chin J Integr Med 2019; 26:114-121. [PMID: 31144159 DOI: 10.1007/s11655-019-2942-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2018] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate the effect of quercetin on ATP binding cassette transporter A1 (ABCA1), liver X receptor (LXR), and proprotein convertase subtilisin/kexin type 9 (PCSK9) expressions in apoE-knockout (ApoE-/-) mice. METHODS The high-fat diet-induced atherosclerosis (AS) in ApoE-/- mice was established. Thirty-six mice were divided into 3 groups using random number table method: model group (n=12), quercetin group (n=12), and atorvastatin group (n=12), with C57BL/6J mice of the same strain and age as the control group (n=12). Quercetin group and atorvastatin group were administrated with quercetin and atorvastatin by oral gavage, with doses of 12.5 and 4 mg/(kg•d), respectively. Animals in the control and model groups were given an equal volume of distilled water by oral gavage once per day for a total of 12 weeks. Western blot and immunohistochemical methods were employed to determine the aortic ABCA1, LXR-α and PCSK9 protein expression. Enzyme linked immunosorbent assay method was used to detect the expression of serum total cholesterol (TC), triglyceride (TG), high density lipoprotein-cholesterol (HDL-C), low density lipoprotein-cholesterol (LDL-C), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-10, combined with tissue pathological examination. RESULTS ApoE-/- mice fed with a high-fat diet had notable atherosclerosis lesions, with reduced ABCA1, LXR-α and IL-10 levels (all P<0.01), elevated PCSK9, TNF-α and IL-6 expression, and increased TC and LDL-C contents (all P<0.01). After quercetin intervention, the areas of AS plaques and the expressions of PCSK9, TNF-α and IL-6 were significantly reduced (all P<0.01), while the expressions of ABCA1 and LXR-α were increased significantly (all P<0.01). CONCLUSION Quercetin effectively interfered with AS development by regulating the expressions of ABCA1, LXR- α and PCSK9 in ApoE-/- mice.
Collapse
Affiliation(s)
- Shan-Shan Li
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - Hui Cao
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - Ding-Zhu Shen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China.
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - San-Li Xing
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - Fang-Fang Dou
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - Qing-Ling Jia
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| |
Collapse
|
20
|
Li S, Cao H, Shen D, Jia Q, Chen C, Xing SL. Quercetin protects against ox‑LDL‑induced injury via regulation of ABCAl, LXR‑α and PCSK9 in RAW264.7 macrophages. Mol Med Rep 2018; 18:799-806. [PMID: 29845234 PMCID: PMC6059709 DOI: 10.3892/mmr.2018.9048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/04/2018] [Indexed: 02/01/2023] Open
Abstract
Quercetin is a flavonoid that has anti‑inflammatory, anti‑oxidant and lipid metabolic effects. It has also been reported to reduce the risk of cardiovascular disease. The present study measured the effects of quercetin on the expression of ATP‑binding cassette transporter 1 (ABCAl), ATP‑binding cassette sub‑family G member 1 (ABCG1), liver X receptor‑α (LXR‑α), proprotein convertase subtilisin/kexin type 9 (PCSK9), p53, p21 and p16 induced by oxidized low density lipoprotein (ox‑LDL). RAW264.7 macrophages were exposed to ox‑LDL with or without 20 µmol/l quercetin and cell proliferation and senescence were quantified using β‑gal staining. Superoxide dismutase (SOD), malondialdehyde (MDA) and lipid droplets were measured in the cytoplasm using oil red staining, while intracellular and total cholesterol (TC) were measured using filipin staining and a TC kit. Immunofluorescent studies and western blot analysis were performed to quantify the expression of ABCAl, ABCG1, LXR‑α, PCSK9, p53, p21 and p16. Quercetin increased RAW264.7 cell viability and reduced lipid accumulation, senescence, lipid droplets, intracellular cholesterol and TC. It was concluded that quercetin inhibits ox‑LDL‑induced lipid droplets in RAW264.7 cells by upregulation of ABCAl, ABCG1, LXR‑α and downregulation of PCSK9, p53, p21 and p16.
Collapse
Affiliation(s)
- Shanshan Li
- Shanghai Geriatrics Institute of Chinese Medicine, Shanghai 200031, P.R. China
| | - Hui Cao
- Shanghai Geriatrics Institute of Chinese Medicine, Shanghai 200031, P.R. China
| | - Dingzhu Shen
- Shanghai Geriatrics Institute of Chinese Medicine, Shanghai 200031, P.R. China
| | - Qingling Jia
- Shanghai Geriatrics Institute of Chinese Medicine, Shanghai 200031, P.R. China
| | - Chuan Chen
- Shanghai Geriatrics Institute of Chinese Medicine, Shanghai 200031, P.R. China
| | - San Li Xing
- Shanghai Geriatrics Institute of Chinese Medicine, Shanghai 200031, P.R. China
| |
Collapse
|
21
|
Talbot CP, Plat J, Ritsch A, Mensink RP. Determinants of cholesterol efflux capacity in humans. Prog Lipid Res 2018; 69:21-32. [PMID: 29269048 DOI: 10.1016/j.plipres.2017.12.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/09/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
|
22
|
Deyab G, Hokstad I, Whist JE, Smastuen MC, Agewall S, Lyberg T, Ronda N, Mikkelsen K, Hjeltnes G, Hollan I. Methotrexate and anti-tumor necrosis factor treatment improves endothelial function in patients with inflammatory arthritis. Arthritis Res Ther 2017; 19:232. [PMID: 29041979 PMCID: PMC5646156 DOI: 10.1186/s13075-017-1439-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022] Open
Abstract
Background Inflammatory arthritis (IA), including rheumatoid arthritis (RA), ankylosing spondylitis (AS) and psoriatic arthritis (PsA), leads to increased cardiovascular disease occurrence probably due to atherosclerosis. One of the first stages in atherogenesis is endothelial dysfunction (ED). Therefore, we aimed to compare endothelial function (EF) in patients with IA, and to examine the effects of methotrexate (MTX) monotherapy and antitumor necrosis factor (anti-TNF) treatment with or without MTX comedication (anti-TNF ± MTX) on EF. Methods From the PSARA observational study, all patients with RA (n = 64), PsA (n = 29), and AS (n = 20) were evaluated for EF. In patients with ED at baseline (n = 40), we evaluated changes in the Reactive Hyperemic Index (RHI) after 6 weeks and 6 months of antirheumatic therapy. Results In IA patients with ED, RHI significantly improved after 6 weeks (p < 0.001) and 6 months (p < 0.001) of treatment, independent of changes in disease activity parameters. After 6 months, RHI had improved more in the MTX group than in the anti-TNF ± MTX group, and the difference remained statistically significant after adjustments for potential confounders. Among patients with active RA, AS, and PsA, those with AS appeared to have the worst endothelial function, although they were the youngest. Conclusion Treatment with MTX and anti-TNF ± MTX was associated with a relatively fast improvement of EF in IA patients with ED, independent of change in disease activity. Therefore, modes of action other than the anti-inflammatory effect may contribute to the EF improvement. After 6 months, the EF improvement was more pronounced in the MTX group than in the anti-TNF ± MTX group. Trial registration Clinicaltrials, NCT00902005. Registered on 13 May 2009.
Collapse
Affiliation(s)
- Gia Deyab
- Department of Medical Biochemistry, Innlandet Hospital Trust, Hamar, Norway.
| | - Ingrid Hokstad
- Lillehammer Hospital for Rheumatic Diseases, Lillehammer, Norway
| | - Jon Elling Whist
- Department of Medical Biochemistry, Innlandet Hospital Trust, Hamar, Norway.,Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| | - Milada Cvancarova Smastuen
- Institution of Health Care, Health Science PhD Program, Oslo and Akershus University College, Oslo, Norway
| | - Stefan Agewall
- Oslo University Hospital, Ullevål, Oslo, Norway.,Institute of Clinical Sciences, University of Oslo, Oslo, Norway
| | - Torstein Lyberg
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Knut Mikkelsen
- Lillehammer Hospital for Rheumatic Diseases, Lillehammer, Norway
| | | | - Ivana Hollan
- Lillehammer Hospital for Rheumatic Diseases, Lillehammer, Norway.,Department of Medicine, Brigham and Women's Hospital, Boston, USA.,Harvard Medical School, Boston, USA.,Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| |
Collapse
|
23
|
Rodríguez-Carrio J, Alperi-López M, López P, López-Mejías R, Alonso-Castro S, Abal F, Ballina-García FJ, González-Gay MÁ, Suárez A. High triglycerides and low high-density lipoprotein cholesterol lipid profile in rheumatoid arthritis: A potential link among inflammation, oxidative status, and dysfunctional high-density lipoprotein. J Clin Lipidol 2017; 11:1043-1054.e2. [DOI: 10.1016/j.jacl.2017.05.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 11/30/2022]
|
24
|
O'Neill F, Charakida M, Topham E, McLoughlin E, Patel N, Sutill E, Kay CWM, D'Aiuto F, Landmesser U, Taylor PC, Deanfield J. Anti-inflammatory treatment improves high-density lipoprotein function in rheumatoid arthritis. Heart 2016; 103:766-773. [PMID: 27852695 PMCID: PMC5529963 DOI: 10.1136/heartjnl-2015-308953] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 09/20/2016] [Accepted: 10/12/2016] [Indexed: 12/31/2022] Open
Abstract
Objective Patients with rheumatoid arthritis (RA) are at increased cardiovascular risk. Recent studies suggest that high-density lipoprotein (HDL) may lose its protective vascular phenotype in inflammatory conditions. However, the effects of common anti-inflammatory treatments on HDL function are not yet known. Methods We compared the function of HDL in 18 patients with RA and 18 matched healthy controls. Subsequently, patients were randomised to (methotrexate+infliximab (M+I) (5 mg/kg)) or methotrexate+placebo (M+P) infusions for 54 weeks. At week 54 and thereafter, all patients received infliximab therapy until completion of the trial (110 weeks), enabling assessment of the impact of 1 year of infliximab therapy in all patients. HDL functional properties were assessed at baseline, 54 weeks and 110 weeks by measuring the impact on endothelial nitric oxide (NO) bioavailability and superoxide production (SO), paraoxonase activity (PON-1) and cholesterol efflux. Results All HDL vascular assays were impaired in patients compared with controls. After 54 weeks, NO in response to HDL was significantly greater in patients who received M+I compared with those who received M+P. Endothelial SO in response to HDL was reduced in both groups, but PON-1 and cholesterol efflux remained unchanged. All vascular measures improved compared with baseline after ≥1 infliximab therapy in the analysis at 110 weeks. No significant trend was noted for cholesterol efflux. Conclusions HDL function can be improved with anti-inflammatory treatment in patients with RA. The M+I combination was superior to the M+P alone, suggesting that the tumour necrosis factor-α pathway may have a role in HDL vascular properties.
Collapse
Affiliation(s)
- Francis O'Neill
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Marietta Charakida
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Eric Topham
- Institute of Structural & Molecular Biology and London Centre for Nanotechnology, University College London, London, UK
| | - Eve McLoughlin
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Neha Patel
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Emma Sutill
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Christopher W M Kay
- Institute of Structural & Molecular Biology and London Centre for Nanotechnology, University College London, London, UK
| | - Francesco D'Aiuto
- Periodontology Unit, Department of Clinical Research, University College London Eastman Dental Institute, London, UK
| | - Ulf Landmesser
- Department of Cardiology, Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Peter C Taylor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - John Deanfield
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK.,National Institute for Cardiovascular Outcomes Research, University College London, London, UK
| |
Collapse
|
25
|
Rheumatoid Arthritis Pharmacotherapies: Do They Have Anti-Atherosclerotic Activity? Curr Rheumatol Rep 2016; 18:27. [DOI: 10.1007/s11926-016-0578-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Fan A, Wang Q, Yuan Y, Cheng J, Chen L, Guo X, Li Q, Chen B, Huang X, Huang Q. Liver X receptor-α and miR-130a-3p regulate expression of sphingosine 1-phosphate receptor 2 in human umbilical vein endothelial cells. Am J Physiol Cell Physiol 2015; 310:C216-26. [PMID: 26669941 DOI: 10.1152/ajpcell.00102.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023]
Abstract
Recent studies have shown that activation of liver X receptors (LXRs) attenuates the development of atherosclerosis, not only by regulating lipid metabolism but also by suppressing inflammatory signaling. Sphingosine 1-phosphate receptor 2 (S1PR2), an important inflammatory gene product, plays a role in the development of various inflammatory diseases. It was proposed that S1PR2 might be regulated by LXR-α. In the present study, the effect of LXR-α on tumor necrosis factor-α (TNF-α)-induced S1PR2 expression in human umbilical vein endothelial cells (HUVECs) was investigated and the underlying mechanism was explored. The results demonstrated that TNF-α led to an increase in S1PR2 expression and triggered a downregulation of LXR-α expression in HUVECs as well. Downregulation of LXR-α with specific small interfering RNA (siRNA) remarkably enhanced the primary as well as TNF-α-induced expression of S1PR2 in HUVECs. Activation of LXR-α by agonist GW3965 inhibited both primary and TNF-α-induced S1PR2 expression. GW3965 also attenuated S1PR2-induced endothelial barrier dysfunction. The data further showed that TNF-α induced a significant decrease in miR-130a-3p expression. Overexpression of miR-130a-3p with mimic product reduced S1PR2 protein expression, and inhibition of miR-130a-3p by specific inhibitor resulted in an increase in S1PR2 protein expression. Furthermore, activation of LXRs with agonist enhanced the expression of miR-130a-3p, and knockdown of LXR-α by siRNA suppressed miR-130a-3p expression. These results suggest that LXR-α might downregulate S1PR2 expression via miR-130a-3p in quiescent HUVECs. Stimulation of TNF-α attenuates the activity of LXR-α and results in enhanced S1PR2 expression.
Collapse
Affiliation(s)
- Aihui Fan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China; Department of Physiology, Guangdong Medical College, Dongguan, People's Republic of China; and
| | - Qian Wang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Yongjun Yuan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Jilun Cheng
- Department of Pharmacology, Guangdong Medical College, Dongguan, People's Republic of China
| | - Lixian Chen
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaohua Guo
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Qiang Li
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Bo Chen
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Xuliang Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China;
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
27
|
Voloshyna I, Mucci T, Sher J, Fonacier LS, Littlefield MJ, Carsons S, Reiss AB. Plasma IL-33 in atopic patients correlates with pro-inflammatory cytokines and changes cholesterol transport protein expression: a surprising neutral overall impact on atherogenicity. Clin Exp Allergy 2015; 45:1554-65. [DOI: 10.1111/cea.12516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Indexed: 12/12/2022]
Affiliation(s)
- I. Voloshyna
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
- Winthrop Research Institute; Winthrop University Hospital; Mineola NY USA
| | - T. Mucci
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
| | - J. Sher
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
| | - L. S. Fonacier
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
| | - M. J. Littlefield
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
- Winthrop Research Institute; Winthrop University Hospital; Mineola NY USA
| | - S. Carsons
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
- Winthrop Research Institute; Winthrop University Hospital; Mineola NY USA
| | - A. B. Reiss
- Department of Medicine; Winthrop University Hospital; Mineola NY USA
- Winthrop Research Institute; Winthrop University Hospital; Mineola NY USA
| |
Collapse
|
28
|
Effects of tumor necrosis factor inhibitor on serum level of HLA-B27 and PDCD-1 in patients with ankylosing spondylitis. Cell Biochem Biophys 2015; 70:1453-7. [PMID: 25005770 DOI: 10.1007/s12013-014-0082-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The aim of this study was to evaluate the effect of tumor necrosis factor-alpha (TNF-α) inhibitor-infliximab on ankylosing spondylitis (AS) patients and detect the serum level of HLA-B27 and PDCD-1 before and after TNF inhibitor treatment. 138 patients at active stage of AS were treated with infliximab; serum was collected before and after TNF-α inhibitor treatment for analysis. Reverse transcription-polymerase chain reaction (RT-PCR), flow cytometry, and enzyme-linked immuno sorbent assay were applied to detect the levels of HLA-B27 and PDCD-1 at different time points, which were used for statistical analysis with clinical data including two AS indicators (erythrocyte sedimentation rate--ESR and C-reactive protein--CRP). After the treatment for 6 weeks, RT-PCR showed that the gene expressions of HLA-B27 and PDCD-1 were significantly downregulated compared with baseline before infliximab treatment (P < 0.05); flow cytometry showed that the HLA-B27 and PDCD-1 double-labeled cells were significantly downregulated (P < 0.05). After 2, 6, or 10 weeks of infliximab treatment, the levels of ESR, CRP, serum HLA-B27, and PDCD-1 of the AS patients were all significantly lower than the baseline levels (P < 0.05), and the serum HLA-B27 and PDCD-1 levels were all significantly correlated with ESR (P < 0.05). Infliximab, an anti-TNF-α inhibitor, decreases significantly not only ESR and CRP, but also the serum levels of HLA-B27 and PDCD-1 in patients with AS. HLA-B27 and PDCD-1 are involved in the pathogenesis, and disease activities of AS. HLA-B27 and PDCD-1 are potentially the useful markers of AS activity and useful parameters to evaluate the effectiveness of anti-TNF-α inhibitor in treating AS.
Collapse
|
29
|
Ronda N, Greco D, Adorni MP, Zimetti F, Favari E, Hjeltnes G, Mikkelsen K, Borghi MO, Favalli EG, Gatti R, Hollan I, Meroni PL, Bernini F. Newly Identified Antiatherosclerotic Activity of Methotrexate and Adalimumab: Complementary Effects on Lipoprotein Function and Macrophage Cholesterol Metabolism. Arthritis Rheumatol 2015; 67:1155-64. [DOI: 10.1002/art.39039] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 01/15/2015] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ivana Hollan
- Lillehammer Hospital for Rheumatic Diseases; Lillehammer Norway
| | - Pier Luigi Meroni
- University of Milan and IRCCS Istituto Auxologico Italiano; Milan Italy
| | | |
Collapse
|
30
|
Toussirot É. Effects of TNFα inhibitors on adiposity and other cardiovascular risk factors: implications for the cardiovascular prognosis in patients with rheumatoid arthritis. Expert Opin Drug Saf 2015; 14:525-32. [DOI: 10.1517/14740338.2015.1007041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|