51
|
Lee HR, Jo MK, Park KY, Jang YJ, Heo TH. Anti-TNF effect of combined pravastatin and cilostazol treatment in an in vivo mouse model. Immunopharmacol Immunotoxicol 2019; 41:179-184. [PMID: 30714456 DOI: 10.1080/08923973.2019.1569045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Objectives: Pravastatin and cilostazol are used as lipid-lowering and antiplatelet agents, respectively. Regarding their well-known anti-inflammatory effects, the additive effect of the two drugs on anti-TNF functions has not yet been investigated. In the present investigation, the beneficial effect of combined pravastatin and cilostazol on their anti-TNF activities was assessed using an in vivo mouse model. Methods: Mice were pretreated with pravastatin and/or cilostazol (40 mg/kg of each), orally once two hour prior to an LPS (5 mg/kg, i.p.) challenge. One hour post challenge, blood and descending aorta were collected for serum TNF levels and immune cell infiltration analyses. For survival analysis, pravastatin and/or cilostazol (40 mg/kg of each) were administered 30 minutes prior to d-galactosamine administration (700 mg/kg, i.p.) and TNF (10 µg/kg, i.p.) challenge and mice survival was monitored. We also examined the effect of either drug or the combination of drugs on TNF-mediated MAPK and NF-κB signaling, using Western blot analysis. Results: Combined treatment of pravastatin and cilostazol significantly decreased serum TNF release and immune cell infiltration in the descending aorta following LPS administration, compared to each single treatment. Additionally, the combined drugs significantly decreased TNF-mediated mouse mortality and downregulated TNF-induced MAPK and NF-κB activation. Conclusions: These findings suggest that combined pravastatin and cilostazol is more effective for reducing TNF-driven inflammation through their anti-TNF activity than monotherapy.
Collapse
Affiliation(s)
- Hae-Ri Lee
- a Laboratory of Pharmaco-Immunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 PLUS Team for Creative Leader Program for Pharmacomics-Based Future Pharmacy, College of Pharmacy , The Catholic University of Korea , Bucheon , Republic of Korea
| | - Min-Kyung Jo
- b College of Pharmacy , ILAb, Inc., NP513, The Catholic University of Korea , Bucheon , Republic of Korea
| | - Kyung-Yeon Park
- a Laboratory of Pharmaco-Immunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 PLUS Team for Creative Leader Program for Pharmacomics-Based Future Pharmacy, College of Pharmacy , The Catholic University of Korea , Bucheon , Republic of Korea
| | - You-Jin Jang
- a Laboratory of Pharmaco-Immunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 PLUS Team for Creative Leader Program for Pharmacomics-Based Future Pharmacy, College of Pharmacy , The Catholic University of Korea , Bucheon , Republic of Korea
| | - Tae-Hwe Heo
- a Laboratory of Pharmaco-Immunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 PLUS Team for Creative Leader Program for Pharmacomics-Based Future Pharmacy, College of Pharmacy , The Catholic University of Korea , Bucheon , Republic of Korea
| |
Collapse
|
52
|
Arterial Stiffness in Early Phases of Prehypertension. UPDATES IN HYPERTENSION AND CARDIOVASCULAR PROTECTION 2019. [DOI: 10.1007/978-3-319-75310-2_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
53
|
Bartoloni E, Alunno A, Valentini V, Luccioli F, Valentini E, La Paglia GMC, Leone MC, Cafaro G, Marcucci E, Gerli R. Targeting Inflammation to Prevent Cardiovascular Disease in Chronic Rheumatic Diseases: Myth or Reality? Front Cardiovasc Med 2018; 5:177. [PMID: 30619884 PMCID: PMC6297850 DOI: 10.3389/fcvm.2018.00177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/29/2018] [Indexed: 01/10/2023] Open
Abstract
Evidence for increased risk of cardiovascular morbidity and mortality in chronic inflammatory rheumatic diseases has accumulated during the last years. Traditional cardiovascular risk factors contribute in part to the excess of cardiovascular risk in these patients and several mechanisms, including precocious acceleration of subclinical atherosclerotic damage, inflammation, and immune system deregulation factors, have been demonstrated to strictly interplay in the induction and progression of atherosclerosis. In this setting, chronic inflammation is a cornerstone of rheumatic disease pathogenesis and exerts also a pivotal role in all stages of atherosclerotic damage. The strict link between inflammation and atherosclerosis suggests that cardiovascular risk may be reduced by rheumatic disease activity control. There are data to suggest that biologic therapies, in particular TNFα antagonists, may improve surrogate markers of cardiovascular disease and reduce CV adverse outcome. Thus, abrogation of inflammation is considered an important outcome for achieving not only control of rheumatic disease, but also reduction of cardiovascular risk. However, the actual effect of anti-rheumatic therapies on atherosclerosis progression and CV outcome in these patients is rather uncertain due to great literature inconsistency. In this paper, we will summarize some of the main mechanisms linking the inflammatory pathogenic background underlying rheumatic diseases and the vascular damage observed in these patients, with a particular emphasis on the pathways targeted by currently available therapies. Moreover, we will analyze current evidence on the potential atheroprotective effects of these treatments on cardiovascular outcome pointing out still unresolved questions.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Alessia Alunno
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Valentina Valentini
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Filippo Luccioli
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Eleonora Valentini
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | | | - Maria Comasia Leone
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Elisa Marcucci
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Roberto Gerli
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| |
Collapse
|
54
|
Barbati C, Vomero M, Colasanti T, Diociaiuti M, Ceccarelli F, Ferrigno S, Finucci A, Miranda F, Novelli L, Perricone C, Spinelli FR, Truglia S, Conti F, Valesini G, Alessandri C. TNFα expressed on the surface of microparticles modulates endothelial cell fate in rheumatoid arthritis. Arthritis Res Ther 2018; 20:273. [PMID: 30526655 PMCID: PMC6286582 DOI: 10.1186/s13075-018-1768-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/13/2018] [Indexed: 11/13/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is associated with a high prevalence of atherosclerosis. Recently increased levels of microparticles (MPs) have been reported in patients with RA. MPs could represent a link between autoimmunity and endothelial dysfunction by expressing tumor necrosis factor alpha (TNFα), a key cytokine involved in the pathogenesis of RA, altering endothelial apoptosis and autophagy. The aim of this study was to investigate TNFα expression on MPs and its relationship with endothelial cell fate. Methods MPs were purified from peripheral blood from 20 healthy controls (HC) and from 20 patients with RA, before (time (T)0) and after (T4) 4-month treatment with etanercept (ETA). Surface expression of TNFα was performed by flow cytometry analysis. EA.hy926 cells, an immortalized endothelial cell line, were treated with RA-MPs purified at T0 and at T4 and also, with RA-MPs in vitro treated with ETA. Apoptosis and autophagy were then evaluated. Results RA-MPs purified at T0 expressed TNFα on their surface and this expression significantly decreased at T4. Moreover, at T0 RA-MPs, significantly increased both apoptosis and autophagy levels on endothelial cells, in a dose-dependent manner. RA-MPs did not significantly change these parameters after 4 months of in vivo treatment with ETA. Conclusions Our data demonstrate that MPs isolated from patients with RA exert a pathological effect on endothelial cells by TNFα expressed on their surface. In vivo and in vitro treatment with ETA modulates this effect, suggesting anti-TNF therapy protects against endothelial damage in patients with RA. Electronic supplementary material The online version of this article (10.1186/s13075-018-1768-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cristiana Barbati
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy.
| | - Marta Vomero
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Tania Colasanti
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Marco Diociaiuti
- Technology and health Department, Istituto Superiore di Sanità, Rome, Italy
| | - Fulvia Ceccarelli
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Sara Ferrigno
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Annacarla Finucci
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Francesca Miranda
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Lucia Novelli
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Carlo Perricone
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Francesca Romana Spinelli
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Simona Truglia
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
55
|
Park KY, Heo TH. Combination therapy with cilostazol and pravastatin improves antiatherogenic effects in low-density lipoprotein receptor knockout mice. Cardiovasc Ther 2018; 36:e12476. [PMID: 30378752 DOI: 10.1111/1755-5922.12476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/29/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
AIMS Despite the therapeutic efficacy of statins and antiplatelet agents for atherosclerosis, monotherapy with each drug alone is often insufficient to achieve the patient's therapeutic goals. We previously showed that combined statin/antiplatelet agent/anti-tumor necrosis factor (TNF) agent therapy (pravastatin/sarpogrelate/etanercept) reduces atherosclerotic lesions by inhibiting TNF, an atherogenic cytokine that contributes to the progression of arteriosclerosis. In addition, our previous study showed that combined treatment with pravastatin and cilostazol is effective for reducing TNF-driven inflammation through anti-TNF activity. Therefore, in the present study, we evaluated the additive effects of combined pravastatin and cilostazol therapy on atherosclerotic progression using low-density lipoprotein receptor (LDLR) knockout (KO) mice. METHODS Ten-week-old LDLR KO mice were fed a high-fat, high-cholesterol diet and orally administered pravastatin and cilostazol alone or in combination. Body weight, plasma lipid levels, and the levels of intracellular adhesion molecules and inflammatory cytokines were analyzed. In addition, aortas and aortic roots were stained with Oil Red O, and atherosclerotic plaques were quantified. RESULTS The atherosclerotic plaques in the combined pravastatin and cilostazol treatment groups were significantly reduced compared to those in each drug monotherapy group. The combination therapy group also showed the downregulation of ICAM-1, MOMA-2, TNF, interleukin (IL)-6, triglyceride, total cholesterol, and low-density lipoprotein levels and the upregulation of high-density lipoprotein levels compared to those of the pravastatin- or cilostazol-treated groups. CONCLUSIONS Our results suggest that combination therapy with pravastatin and cilostazol exerts beneficial effects by decreasing atherosclerotic lesion progression and improving the pro-inflammatory state in the vascular endothelium. These effects are mediated by the reduction in adhesion molecule expression, immune cell infiltration, and cytokine levels and the antiatherosclerotic modulation of serum cholesterol levels. Therefore, we conclude that combined treatment with pravastatin and cilostazol may be a more effective antiatherosclerotic strategy than treatment with either agent alone.
Collapse
Affiliation(s)
- Kyung-Yeon Park
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Korea
| | - Tae-Hwe Heo
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Korea
| |
Collapse
|
56
|
Markelova EI, Novikova DS, Korotaeva TV, Loginova EY. Dynamics of Carotid Intima-Media Thickness, Parameters of Arterial Stiffness and Ambulatory Blood Pressure Monitoring during Therapy with Inhibitor of Tumor Necrosis Factor-Alpha in Patients with Early Psoriatic Arthritis. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2018. [DOI: 10.20996/1819-6446-2018-14-5-711-715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
57
|
Zardi EM, Pipita ME, Giorgi C, Lichinchi D, Zardi DM, Afeltra A. Differences in carotid atherosclerosis between patients with ankylosing spondylitis treated with tumor necrosis factor-α antagonists and healthy matched controls. Medicine (Baltimore) 2018; 97:e11250. [PMID: 29979389 PMCID: PMC6076036 DOI: 10.1097/md.0000000000011250] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
An increased vascular risk is present in patients with ankylosing spondylitis (AS). In this report, we evaluate the presence and grade of atherosclerosis in patients with AS, uninterruptedly treated with tumor necrosis factor-α (TNF-α) antagonists for 2 years, in comparison to that in a nontreated group of healthy controls.Fourteen patients with AS and 14 healthy controls underwent carotid sonography to measure intima-media thickness (IMT) and to evaluate the presence of plaque. Bath Ankylosing Spondylitis Disease Activity Index, Bath Ankylosing Spondylitis Metrology Index, Bath Ankylosing Spondylitis Functional Index scores, erythrocyte sedimentation rate, C-reactive protein, glycemia, total cholesterol, and triglyceride levels were also recorded.Patients with AS showed significantly lower values of mean and maximum IMT at the level of the common carotid (P = .02 and .04, respectively) and the carotid bulb (P = .0006 and .0005, respectively) compared to those of healthy controls. They also had a number of carotid plaques significantly lower than that of healthy controls (P = .02). No differences were found in IMT values at the level of internal carotid between the 2 populations.The significantly lower carotid atherosclerosis found in patients with AS treated with TNF antagonists than in healthy controls shows the important complementary role of this treatment in reducing vascular disease progression probably by decreasing inflammation.
Collapse
Affiliation(s)
| | | | - Chiara Giorgi
- Radiology Department, S Maria della Misericordia Hospital, Urbino
| | | | - Domenico Maria Zardi
- Division of Cardiology, Faculty of Medicine and Psychology, University of Rome “Sapienza,” Sant’Andrea Hospital
| | - Antonella Afeltra
- Immunorheumatology Unit, “Campus Bio-Medico” University, Rome, Italy
| |
Collapse
|
58
|
Park KY, Heo TH. Critical role of TNF inhibition in combination therapy for elderly mice with atherosclerosis. Cardiovasc Ther 2018. [PMID: 28643478 DOI: 10.1111/1755-5922.12280] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
AIMS We have previously shown that the combination of pravastatin and sarpogrelate is synergistically beneficial for atherosclerosis. In this study, we investigated whether the pravastatin-sarpogrelate combination was sufficient for treatment in an old mouse model of atherosclerosis or if additional intervention would be needed to address the newly included aging factor and its complex pathophysiological impact on the atherosclerogenic state. We added an anti-TNF biological to the combination treatment cocktail because of the known pathologic roles of TNF in the aging process. METHODS Sixty-week-old low-density lipoprotein receptor knockout mice were fed a high-fat, high-cholesterol diet and treated with the sarpogrelate and pravastatin combination, etanercept alone, or the triple combination. RESULTS Although, etanercept alone did not significantly reduce aortic root and atherosclerotic plaque areas, the pravastatin-sarpogrelate combination, and pravastatin-sarpogrelate-etanercept triple therapy significantly reduced the plaque areas. Surprisingly, TNF inhibition was critically required to reduce the plaque areas of aortic roots and the expression of ICAM-1, MOMA-2, and TNF. More importantly, a lipid-lowering effect by pravastatin was observed only in the triple therapy group and not in the pravastatin and sarpogrelate combination group. CONCLUSIONS These results suggest that TNF-inhibitory intervention should be added to the conventional therapy as a novel strategy for treating the elderly patients with atherosclerosis.
Collapse
Affiliation(s)
- Kyung-Yeon Park
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Korea.,ILAb Inc, Bucheon-si, Gyeonggi-do, Korea
| | - Tae-Hwe Heo
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Korea.,ILAb Inc, Bucheon-si, Gyeonggi-do, Korea
| |
Collapse
|
59
|
Systemic Inflammatory Response and Atherosclerosis: The Paradigm of Chronic Inflammatory Rheumatic Diseases. Int J Mol Sci 2018; 19:ijms19071890. [PMID: 29954107 PMCID: PMC6073407 DOI: 10.3390/ijms19071890] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/17/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
Patients with Chronic Inflammatory Rheumatic diseases (CIRD) are at increased risk of cardiovascular disease (CVD), ascribed not only to classical risk factors, but also to the presence of chronic systemic inflammatory response. Αtherosclerosis, the cornerstone of CVD, is known to be accelerated in CIRD; rheumatoid arthritis promotes atheromatosis and associates with preclinical atherosclerosis equivalent to Diabetes Mellitus, which also seems to apply for systemic lupus erythematosus. Data on ankylosing spondylitis and psoriatic arthritis, albeit more limited, also support an increased CV risk in these patients. The association between inflammation and atherosclerosis, has been thoroughly investigated in the last three decades and the role of inflammation in the pathogenesis and progression of atherogenesis has been well established. Endothelial dysfunction, oxidative stress in vascular endothelial cells and macrophage accumulation, toll-like receptor signaling, NLPR-3 formation and subsequent pro-inflammatory cytokine production, such as TNFa, IL-1β, IL-6, and TNF-like cytokine 1A, are few of the mechanisms implicated in the atherogenic process. Moreover, there is evidence that anti-inflammatory biologic drugs, such as anti-TNF and anti-IL1β agents, can decelerate the atherogenic process, thus setting new therapeutic targets for early and effective disease control and suppression of inflammation, in addition to aggressive management of classical CV risk factors.
Collapse
|
60
|
Li B, Li W, Li X, Zhou H. Inflammation: A Novel Therapeutic Target/Direction in Atherosclerosis. Curr Pharm Des 2018; 23:1216-1227. [PMID: 28034355 PMCID: PMC6302344 DOI: 10.2174/1381612822666161230142931] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/27/2016] [Indexed: 12/27/2022]
Abstract
Over the past two decades, the viewpoint of atherosclerosis has been replaced gradually by a lipid-driven, chronic, low-grade inflammatory disease of the arterial wall. Current treatment of atherosclerosis is focused on limiting its risk factors, such as hyperlipidemia or hypertension. However, treatment targeting the inflammatory nature of atherosclerosis is still very limited and deserves further attention to fight atherosclerosis successfully. Here, we review the current development of inflammation and atherosclerosis to discuss novel insights and potential targets in atherosclerosis, and to address drug discovery based on anti-inflammatory strategy in atherosclerotic disease.
Collapse
Affiliation(s)
- Bin Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing 400038. China
| | - Weihong Li
- Assisted Reproductive Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016. China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing 400038. China
| | - Hong Zhou
- Department of Pharmacology, College of Pharamacy, The Third Military Medical University, P.O. Box: 400038, Chongqing. China
| |
Collapse
|
61
|
Anti-Inflammatory Treatment. Coron Artery Dis 2018. [DOI: 10.1016/b978-0-12-811908-2.00013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
62
|
Chodara AM, Wattiaux A, Bartels CM. Managing Cardiovascular Disease Risk in Rheumatoid Arthritis: Clinical Updates and Three Strategic Approaches. Curr Rheumatol Rep 2017; 19:16. [PMID: 28361332 DOI: 10.1007/s11926-017-0643-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ᅟ: The increase in cardiovascular disease (CVD) risk in rheumatoid arthritis (RA) is well known; however, appropriate management of this elevated risk in rheumatology clinics is less clear. PURPOSE OF REVIEW By critically reviewing literature published within the past 5 years, we aim to clarify current knowledge and gaps regarding CVD risk management in RA. RECENT FINDINGS We examine recent guidelines, recommendations, and evidence and discuss three approaches: (1) RA-specific management including treat-to-target and medication management, (2) assessment of comprehensive individual risk, and (3) targeting traditional CVD risk factors (hypertension, smoking, hyperlipidemia, diabetes, obesity, and physical inactivity) at a population level. Considering that 75% of US RA visits occur in specialty clinics, further research is needed regarding evidence-based strategies to manage and reduce CVD risk in RA. This review highlights clinical updates including US cardiology and international professional society guidelines, successful evidence-based population approaches from primary care, and novel opportunities in rheumatology care to reduce CVD risk in RA.
Collapse
Affiliation(s)
- Ann M Chodara
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Aimée Wattiaux
- Rheumatology Division, Department of Medicine, University of Wisconsin (UW) School of Medicine and Public Health (SMPH), 1685 Highland Ave, Rm 4132, 53705-2281, Madison, WI, USA
| | - Christie M Bartels
- Rheumatology Division, Department of Medicine, University of Wisconsin (UW) School of Medicine and Public Health (SMPH), 1685 Highland Ave, Rm 4132, 53705-2281, Madison, WI, USA.
| |
Collapse
|
63
|
Pedersen BK. Anti-inflammatory effects of exercise: role in diabetes and cardiovascular disease. Eur J Clin Invest 2017; 47:600-611. [PMID: 28722106 DOI: 10.1111/eci.12781] [Citation(s) in RCA: 399] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Persistent inflammation is involved in the pathogenesis of chronic diseases such as type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). AIMS The aim of this review was to provide the reader with an update of the mechanisms whereby exercise-induced cytokines may impact cardiometabolic diseases. RESULTS Evidence exists that interleukin (IL)-1β is involved in pancreatic β-cell damage, whereas TNF-α is a key molecule in peripheral insulin resistance. In addition, TNF-α appears to be involved in the pathogenesis of atherosclerosis and heart failure. A marked increase in IL-6 and IL-10 is provoked by exercise and exerts direct anti-inflammatory effects by an inhibition of TNF-α and by stimulating IL-1ra, thereby limiting IL-1β signalling. Moreover, muscle-derived IL-6 appears to have direct anti-inflammatory effects and serves as a mechanism to improve glucose tolerance. In addition, indirect anti-inflammatory effects of long-term exercise are mediated via improvements in body composition. CONCLUSION Physical activity represents a natural, strong anti-inflammatory strategy with minor side effects and should be integrated in the management of patients with cardiometabolic diseases.
Collapse
Affiliation(s)
- Bente Klarlund Pedersen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
64
|
Arida A, Protogerou AD, Konstantonis G, Fragiadaki K, Kitas GD, Sfikakis PP. Atherosclerosis is not accelerated in rheumatoid arthritis of low activity or remission, regardless of antirheumatic treatment modalities. Rheumatology (Oxford) 2017; 56:934-939. [PMID: 28160488 DOI: 10.1093/rheumatology/kew506] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Indexed: 11/14/2022] Open
Abstract
Objectives RA associates with increased cardiovascular disease (CVD) morbidity and mortality due to accelerated atherosclerosis, attributed to both classical risk factors and chronic inflammation. The aim of this study was to test the hypothesis that effective disease control over 3 years modifies acceleration of atherosclerosis in RA. Methods Consecutive, non-diabetic RA patients previously examined by ultrasonography for subclinical atherosclerosis were re-evaluated after 3.2 (0.2) years, provided that they were in remission/low disease activity (DAS28 <3.2) for at least 75% of this period. Patients (n = 139) were demographically matched with 139 non-diabetic, non-RA control individuals studied in parallel. Results Patients and controls (mean age of 56 years at baseline) had a comparable burden of classical CVD risk factors. Patients' pulse wave velocity (reflecting arterial stiffness) changed by 0.07 m/s/year and left carotid intima-media thickness (reflecting wall hypertrophy) increased by 0.009 mm/year; formation of new atheromatic plaques in carotid and/or femoral arterial beds occurred in 22%. Multivariate analysis after correcting for all classical CVD risk factors and anti-hypertensive/lipid-lowering therapies demonstrated no significant differences between patients and controls in any of the subclinical atherosclerosis indices. Changes in all atherosclerosis indices from baseline to end of follow-up were comparable between those 56 patients treated with biologic DMARDs and their demographically matched patients treated with synthetic DMARDs. Conclusion Effective disease control may abrogate any RA-specific effect on the progression of atherosclerosis regardless of treatment. Whether early and sustained RA control translates to the CVD outcomes expected in the general population should be examined in prospective studies.
Collapse
Affiliation(s)
- Aikaterini Arida
- Rheumatology Unit, 1st Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian Univeristy of Athens.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Greece
| | - Athanasios D Protogerou
- Rheumatology Unit, 1st Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian Univeristy of Athens.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Greece
| | - George Konstantonis
- Rheumatology Unit, 1st Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian Univeristy of Athens.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Greece
| | - Kalliopi Fragiadaki
- Rheumatology Unit, 1st Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian Univeristy of Athens.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Greece
| | - George D Kitas
- Rheumatology Unit, 1st Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian Univeristy of Athens.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Greece
| | - Petros P Sfikakis
- Rheumatology Unit, 1st Department of Propaedeutic Internal Medicine, Laikon Hospital, National and Kapodistrian Univeristy of Athens.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Greece
| |
Collapse
|
65
|
Anti-TNF-alpha agents and endothelial function in rheumatoid arthritis: a systematic review and meta-analysis. Sci Rep 2017; 7:5346. [PMID: 28706194 PMCID: PMC5509678 DOI: 10.1038/s41598-017-05759-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/05/2017] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) has been associated with endothelial dysfunction, a pathophysiological feature of atherosclerosis. Our aim was to determine whether TNF-α blockade has a beneficial effect on endothelial function in RA. We performed a systematic review with meta-analysis to evaluate the effect of anti-TNF-α agents on endothelial function in RA patients. MedLine, Cochrane CENTRAL and SCOPUS were searched up to March 2016. Inclusion criteria were: 1) randomised controlled trial (RCT), quasi-RCT, before-after cohort study; 2) including RA patients; 3) treatment with anti-TNF-α medications; 4) evaluating the change from baseline in endothelial function. The search strategy retrieved 180 records, of which 20 studies were included in the systematic review. Pooled analysis using a random-effects model demonstrated a significant improvement in endothelial function following anti-TNF-α treatment (SDM 0.987, 95%CI [0.64–1.33], p < 0.0001). Generalisation of the results of the meta-analysis may be limited due to the presence of heterogeneity (I2 = 82.65%, p < 0.001) and evidence of possible publication bias. Meta-regression showed that endothelial function measurement technique was a significant contributor to heterogeneity. In conclusion, although limited by the methodological quality of the included studies, our meta-analysis suggests that anti-TNF-α treatment may improve endothelial function in RA patients.
Collapse
|
66
|
Agca R, Heslinga M, Kneepkens EL, van Dongen C, Nurmohamed MT. The Effects of 5-year Etanercept Therapy on Cardiovascular Risk Factors in Patients with Psoriatic Arthritis. J Rheumatol 2017; 44:1362-1368. [PMID: 28572461 DOI: 10.3899/jrheum.161418] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To investigate the effects of etanercept (ETN) on lipid metabolism and other known cardiovascular disease (CVD) risk factors in patients with psoriatic arthritis (PsA). METHODS In an observational cohort of 118 consecutive patients with PsA, CVD risk factors were assessed over 5 years. Mixed-model analyses were performed to investigate the effects of ETN therapy on CVD risk factors over time. RESULTS Disease Activity Score in 28 joints, C-reactive protein (CRP), and erythrocyte sedimentation rate decreased during therapy with ETN. There was an increase in total cholesterol (TC), high-density lipoprotein cholesterol (HDLc), and low-density lipoprotein cholesterol. The TC/HDLc ratio remained unaltered. The apolipoprotein B to apolipoprotein A-I (apoB/apoA-I) ratio decreased significantly. An increase in CRP was associated with an increase in the apoB/apoA-1 ratio. CONCLUSION Serum lipid concentrations showed small changes over a 5-year period of ETN therapy and were inversely associated with inflammatory markers. Other CVD risk factors remained stable. The apoB/apoA-1 ratio decreased over time and an increase in disease activity was associated with an increase in this ratio. However, this modest lipid modulation cannot explain the observed beneficial CV effects of ETN, and ETN likely exerts those effects through inflammation-related mechanisms.
Collapse
Affiliation(s)
- Rabia Agca
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center, Amsterdam, the Netherlands.,R. Agca, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; M. Heslinga, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; E.L. Kneepkens, MD, PhD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; C. van Dongen, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; M.T. Nurmohamed, MD, Professor, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center
| | - Maaike Heslinga
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center, Amsterdam, the Netherlands. .,R. Agca, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; M. Heslinga, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; E.L. Kneepkens, MD, PhD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; C. van Dongen, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; M.T. Nurmohamed, MD, Professor, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center.
| | - Eva L Kneepkens
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center, Amsterdam, the Netherlands.,R. Agca, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; M. Heslinga, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; E.L. Kneepkens, MD, PhD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; C. van Dongen, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; M.T. Nurmohamed, MD, Professor, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center
| | - Carlo van Dongen
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center, Amsterdam, the Netherlands.,R. Agca, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; M. Heslinga, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; E.L. Kneepkens, MD, PhD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; C. van Dongen, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; M.T. Nurmohamed, MD, Professor, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center
| | - Michael T Nurmohamed
- From the Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center, Amsterdam, the Netherlands.,R. Agca, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; M. Heslinga, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center; E.L. Kneepkens, MD, PhD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; C. van Dongen, MD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- Reade; M.T. Nurmohamed, MD, Professor, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center
- VU University Medical Center
| |
Collapse
|
67
|
Pascual J, González I, Corona D, Hispán P, Ramos J, Sánchez-Paya J, Jemec G. Assessment of subclinical atherosclerosis in hidradenitis suppurativa. J Eur Acad Dermatol Venereol 2017; 31:1229-1238. [DOI: 10.1111/jdv.14076] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/23/2016] [Indexed: 01/02/2023]
Affiliation(s)
- J.C. Pascual
- Department of Dermatology; Alicante University General Hospital; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation); Alicante Spain
| | - I. González
- Department of Dermatology; Alicante University General Hospital; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation); Alicante Spain
| | - D. Corona
- Department of Neurology; Alicante University General Hospital; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation); Alicante Spain
| | - P. Hispán
- Department of Dermatology; Alicante University General Hospital; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation); Alicante Spain
| | - J.M. Ramos
- Department of Internal Medicine; Alicante University General Hospital; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation); Alicante Spain
| | - J. Sánchez-Paya
- Epidemiology Unit; Alicante University General Hospital; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation); Alicante Spain
| | - G.B. Jemec
- Department of Dermatology; Zealand University Hospital Roskilde; Health Sciences Faculty; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
68
|
Dimitroulas T, Sandoo A, Skeoch S, O’Sullivan M, Yessirkepov M, Ayvazyan L, Gasparyan A, Metsios G, Kitas G. Rheumatoid Arthritis. THE HEART IN RHEUMATIC, AUTOIMMUNE AND INFLAMMATORY DISEASES 2017:129-165. [DOI: 10.1016/b978-0-12-803267-1.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
69
|
Dzieża-Grudnik A, Sulicka J, Strach M, Siga O, Klimek E, Korkosz M, Grodzicki T. Arterial stiffness is not increased in patients with short duration rheumatoid arthritis and ankylosing spondylitis. Blood Press 2016; 26:115-121. [DOI: 10.1080/08037051.2016.1232586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Anna Dzieża-Grudnik
- Department of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Sulicka
- Department of Rheumatology and Balneology, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Strach
- Department of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Olga Siga
- Department of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Ewa Klimek
- Department of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Balneology, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz Grodzicki
- Department of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
70
|
Lucke M, Messner W, Kim ESH, Husni ME. The impact of identifying carotid plaque on addressing cardiovascular risk in psoriatic arthritis. Arthritis Res Ther 2016; 18:178. [PMID: 27485213 PMCID: PMC4969674 DOI: 10.1186/s13075-016-1074-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/11/2016] [Indexed: 01/30/2023] Open
Abstract
Background Patients with psoriatic arthritis (PsA) are at an increased risk for cardiovascular (CV) disease. The aim of this study was to identify the frequency of carotid plaque in asymptomatic patients with psoriatic arthritis at baseline and follow-up screening, and to assess for the impact of demonstrating plaque on management of traditional cardiovascular risk factors. Methods Eighty-seven PsA patients underwent carotid duplex ultrasound screening. Repeat carotid duplex ultrasound was offered to all patients between 12 and 30 months. Preventive cardiology referrals were generated for all patients through the electronic health record. Traditional cardiovascular risk factors, medication use, and rates of utilization of preventive cardiology services were compared between patients with and without plaque. Results Carotid plaque was identified in 34/87 (39 %) of PsA patients. Age and triglyceride levels were predictors of plaque presence. Patients with plaque trended toward higher rates of smoking and diabetes, and higher low-density lipoprotein levels. Only 9/87 (10 %) patients completed at least one visit with preventive cardiology after enrollment despite referral. Low use of statin (21 %) and antiplatelet (27 %) medication was observed. Rates of biologic medication use for PsA were higher (75 %) than studies in similar cohorts of patients with carotid plaque. No association was seen between disease duration or activity and the presence of carotid plaque. Conclusion Despite demonstration of high cardiac risk by the presence of carotid plaque, implementation of preventive cardiovascular services and rates of statin and antiplatelet use remained low. Age and triglyceride levels were significant variables in predicting plaque presence. There is no evidence that demonstration of plaque resulted in further evaluation or changes in treatment regimens to address heightened cardiovascular risk.
Collapse
Affiliation(s)
- Michael Lucke
- Department of Rheumatology, Allegheny Health Network, Pittsburgh, PA, USA
| | - William Messner
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Esther S H Kim
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - M Elaine Husni
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, 9500 Euclid Ave, desk A50, Cleveland, OH, 44195, USA.
| |
Collapse
|
71
|
Yuan XL, Li Y, Pan XH, Zhou M, Gao QY, Li MC. Production of recombinant human interleukin-38 and its inhibitory effect on the expression of proinflammatory cytokines in THP-1 cells. Mol Biol 2016. [DOI: 10.1134/s0026893316030134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
72
|
Ikdahl E, Rollefstad S, Wibetoe G, Olsen IC, Berg IJ, Hisdal J, Uhlig T, Haugeberg G, Kvien TK, Provan SA, Semb AG. Predictive Value of Arterial Stiffness and Subclinical Carotid Atherosclerosis for Cardiovascular Disease in Patients with Rheumatoid Arthritis. J Rheumatol 2016; 43:1622-30. [DOI: 10.3899/jrheum.160053] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2016] [Indexed: 12/31/2022]
Abstract
Objective.We evaluated the predictive value of these vascular biomarkers for cardiovascular disease (CVD) events in patients with rheumatoid arthritis (RA): aortic pulse wave velocity (aPWV), augmentation index (AIx), carotid intima-media thickness (cIMT), and carotid plaques (CP). They are often used as risk markers for CVD.Methods.In 2007, 138 patients with RA underwent clinical examination, laboratory tests, blood pressure testing, and vascular biomarker measurements. Occurrence of CVD events was recorded in 2013. Predictive values were assessed in Kaplan-Meier plots, log-rank, and crude and adjusted Cox proportional hazard (PH) regression analyses.Results.Baseline median age and disease duration was 59.0 years and 17.0 years, respectively, and 76.1% were women. CVD events occurred in 10 patients (7.2%) during a mean followup of 5.4 years. Compared with patients with low aPWV, AIx, cIMT, and without CP, patients with high aPWV (p < 0.001), high AIx (p = 0.04), high cIMT (p = 0.01), and CP (p < 0.005) at baseline experienced more CVD events. In crude Cox PH regression analyses, aPWV (p < 0.001), cIMT (p < 0.001), age (p = 0.01), statin (p = 0.01), and corticosteroid use (p = 0.01) were predictive of CVD events, while AIx was nonsignificant (p = 0.19). The Cox PH regression estimates for vascular biomarkers were not significantly altered when adjusting individually for demographic variables, traditional CVD risk factors, RA disease-related variables, or medication. All patients who developed CVD had CP at baseline.Conclusion.CP, aPWV, and cIMT were predictive of CVD events in this cohort of patients with RA. Future studies are warranted to examine the additive value of arterial stiffness and carotid atherosclerosis markers in CVD risk algorithms. Regional Ethical Committee approval numbers 2009/1582 and 2009/1583.
Collapse
|
73
|
Tousoulis D, Oikonomou E, Economou EK, Crea F, Kaski JC. Inflammatory cytokines in atherosclerosis: current therapeutic approaches. Eur Heart J 2016; 37:1723-1732. [PMID: 26843277 DOI: 10.1093/eurheartj/ehv759] [Citation(s) in RCA: 343] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/24/2015] [Indexed: 12/20/2022] Open
Abstract
The notion of atherosclerosis as a chronic inflammatory disease has intensified research on the role of cytokines and the way these molecules act and interact to initiate and sustain inflammation in the microenvironment of an atherosclerotic plaque. Cytokines are expressed by all types of cells involved in the pathogenesis of atherosclerosis, act on a variety of targets exerting multiple effects, and are largely responsible for the crosstalk among endothelial, smooth muscle cells, leucocytes, and other vascular residing cells. It is now understood that widely used drugs such as statins, aspirin, methotrexate, and colchicine act in an immunomodulatory way that may beneficially affect atherogenesis and/or cardiovascular disease progression. Moreover, advancement in pharmaceutical design has enabled the production of highly specific antibodies against key molecules involved in the perpetuation of the inflammatory cascade, raising hope for advances in the treatment of atherosclerosis. This review describes the actions and effects of these agents, their potential clinical significance, and future prospects.
Collapse
Affiliation(s)
- Dimitris Tousoulis
- First Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Vasilissis Sofias 114, TK 115 28 Athens, Greece
| | - Evangelos Oikonomou
- First Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Vasilissis Sofias 114, TK 115 28 Athens, Greece
| | - Evangelos K Economou
- First Department of Cardiology, 'Hippokration' Hospital, University of Athens Medical School, Vasilissis Sofias 114, TK 115 28 Athens, Greece
| | - Filippo Crea
- Cardiovascular Sciences Department, Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Juan Carlos Kaski
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
74
|
Zhao JJ, Hu YW, Huang C, Ma X, Kang CM, Zhang Y, Guo FX, Lu JB, Xiu JC, Qiu YR, Sha YH, Gao JJ, Wang YC, Li P, Xu BM, Zheng L, Wang Q. Dihydrocapsaicin suppresses proinflammatory cytokines expression by enhancing nuclear factor IA in a NF-κB-dependent manner. Arch Biochem Biophys 2016; 604:27-35. [PMID: 27267730 DOI: 10.1016/j.abb.2016.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/09/2016] [Accepted: 06/02/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease and represents the leading cause of morbidity and mortality throughout the world. Accumulating evidences have showed that Dihydrocapsaicin (DHC) has been found to exert multiple pharmacological and physiological effects. Nevertheless, the effects and possible mechanism of DHC on proinflammatory response remain largely unexplained. METHODS AND RESULTS We found that DHC markedly upregulated NFIA and suppressed NF-κB expression in THP-1 macrophages. Up-regulation of proinflammatory cytokines induced by LPS including TNF-α, IL-1β and IL-6 were markedly suppressed by DHC treatment. We also observed that protein level of NFIA was significantly increased while NF-κB and proinflammatory cytokines were decreased by DHC treatment in apoE(-/-) mice. Lentivirus-mediated overexpression of NFIA suppressed NF-κB and proinflammatory cytokines expression both in THP-1 macrophages and plaque tissues of apoE-/- mice. Moreover, treatment with lentivirus-mediated overexpression of NFIA made the down-regulation of DHC on NF-κB and proinflammatory cytokines expression notably accentuated in THP-1 macrophages and apoE(-/-) mice. In addition, treatment with siRNA targeting NF-κB accentuated the suppression of proinflammatory cytokines by lentivirus-mediated overexpression of NFIA. CONCLUSION These observations demonstrated that DHC can significantly decrease proinflammatory cytokines through enhancing NFIA and inhibiting NF-κB expression and thus DHC may be a promising candidate as an anti-inflammatory drug for atherosclerosis as well as other disorders.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chuan Huang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xin Ma
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chun-Min Kang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuan Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Feng-Xia Guo
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jing-Bo Lu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jian-Cheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu-Rong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Hua Sha
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ji-Juan Gao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Chao Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Pan Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Bang-Ming Xu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
75
|
Bernelot Moens SJ, van der Valk FM, Strang AC, Kroon J, Smits LP, Kneepkens EL, Verberne HJ, van Buul JD, Nurmohamed MT, Stroes ESG. Unexpected arterial wall and cellular inflammation in patients with rheumatoid arthritis in remission using biological therapy: a cross-sectional study. Arthritis Res Ther 2016; 18:115. [PMID: 27209093 PMCID: PMC4875657 DOI: 10.1186/s13075-016-1008-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/28/2016] [Indexed: 12/11/2022] Open
Abstract
Background Increasing numbers of patients (up to 40 %) with rheumatoid arthritis (RA) achieve remission, yet it remains to be elucidated whether this also normalizes their cardiovascular risk. Short-term treatment with TNF inhibitors lowers arterial wall inflammation, but not to levels of healthy controls. We investigated whether RA patients in long-term remission are characterized by normalized inflammatory activity of the arterial wall and if this is dependent on type of medication used (TNF-inhibitor versus nonbiological disease-modifying antirheumatic drugs (DMARDs)). Methods Arterial wall inflammation, bone marrow and splenic activity (index of progenitor cell activity) was assessed with 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in RA patients in remission (disease activity score (DAS28) <2.6 for >6 months) and healthy controls. We performed ex vivo characterization of monocytes using flow cytometry and a transendothelial migration assay. Results Overall, arterial wall inflammation was comparable in RA patients (n = 23) in long-term remission and controls (n = 17). However, RA subjects using current anti-TNF therapy (n = 13, disease activity score 1.98[1.8–2.2]) have an almost 1.2-fold higher 18F-FDG uptake in the arterial wall compared to those using DMARDs (but with previous anti-TNF therapy) (n = 10, disease activity score 2.24[1.3–2.5]), which seemed to be predominantly explained by longer duration of their rheumatic disease in a multivariate linear regression analysis. This coincided with increased expression of pro-adhesive (CCR2) and migratory (CD11c, CD18) surface markers on monocytes and a concomitant increased migratory capacity. Finally, we found increased activity in bone marrow and spleen in RA patients using anti-TNF therapy compared to those with DMARDs and controls. Conclusions A subset of patients with RA in clinical remission have activated monocytes and increased inflammation in the arterial wall, despite the use of potent TNF blocking therapies. In these subjects, RA disease duration was the most important contributor to the level of arterial wall inflammation. This increased inflammatory state implies higher cardiovascular risk in these patients, who thus may require more stringent CV risk management. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1008-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sophie J Bernelot Moens
- Department of Vascular Medicine, Academic Medical Center, Room F4-211, PO Box 22660, Amsterdam, 1100 DD, The Netherlands.
| | - Fleur M van der Valk
- Department of Vascular Medicine, Academic Medical Center, Room F4-211, PO Box 22660, Amsterdam, 1100 DD, The Netherlands
| | - Aart C Strang
- Department of Vascular Medicine, Academic Medical Center, Room F4-211, PO Box 22660, Amsterdam, 1100 DD, The Netherlands
| | - Jeffrey Kroon
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, The Netherlands
| | - Loek P Smits
- Department of Vascular Medicine, Academic Medical Center, Room F4-211, PO Box 22660, Amsterdam, 1100 DD, The Netherlands
| | - Eva L Kneepkens
- Departments of Rheumatology Reade, Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Hein J Verberne
- Department of Nuclear Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, The Netherlands
| | - Michael T Nurmohamed
- Departments of Rheumatology Reade, Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Room F4-211, PO Box 22660, Amsterdam, 1100 DD, The Netherlands
| |
Collapse
|
76
|
Li J, McArdle S, Gholami A, Kimura T, Wolf D, Gerhardt T, Miller J, Weber C, Ley K. CCR5+T-bet+FoxP3+ Effector CD4 T Cells Drive Atherosclerosis. Circ Res 2016; 118:1540-52. [PMID: 27021296 PMCID: PMC4867125 DOI: 10.1161/circresaha.116.308648] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
RATIONALE CD4 T cells are involved in the pathogenesis of atherosclerosis, but atherosclerosis-specific CD4 T cells have not been described. Moreover, the chemokine(s) that regulates T-cell trafficking to the atherosclerotic lesions is also unknown. OBJECTIVE In Apoe(-/-) mice with mature atherosclerotic lesions (5 months of high fat diet), we find that most aortic T cells express CCR5 and interferon-γ with a unique combination of cell surface markers (CD4(+)CD25(-)CD44(hi)CD62L(lo)) and transcription factors (FoxP3(+)T-bet(+)). We call these cells CCR5Teff. We investigated the role of CCR5 in regulating T-cell homing to the atherosclerotic aorta and the functionality of the CCR5Teff cells. METHODS AND RESULTS CCR5Teff cells are exclusively found in the aorta and para-aortic lymph nodes of Apoe(-/-) mice. They do not suppress T-cell proliferation in vitro and are less potent than regulatory T cells at inhibiting cytokine secretion. Blocking or knocking out CCR5 or its ligand CCL5 significantly blocks T-cell homing to atherosclerotic aortas. Transcriptomic analysis shows that CCR5Teff cells are more similar to effector T cells than to regulatory T cells. They secrete interferon-γ, interleukin-2, interleukin-10, and tumor necrosis factor. Adoptive transfer of these CCR5Teff cells significantly increases atherosclerosis. CONCLUSIONS CCR5 is specifically needed for CD4 T-cell homing to the atherosclerotic plaques. CCR5(+)CD4 T cells express an unusual combination of transcription factors, FoxP3 and T-bet. Although CCR5Teff express FoxP3, we showed that they are not regulatory and adoptive transfer of these cells exacerbates atherosclerosis.
Collapse
Affiliation(s)
- Jie Li
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Sara McArdle
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Amin Gholami
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Takayuki Kimura
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Dennis Wolf
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Teresa Gerhardt
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Jacqueline Miller
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Christian Weber
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Klaus Ley
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.).
| |
Collapse
|
77
|
Pina T, Corrales A, Lopez-Mejias R, Armesto S, Gonzalez-Lopez MA, Gómez-Acebo I, Ubilla B, Remuzgo-Martínez S, Gonzalez-Vela MC, Blanco R, Hernández JL, Llorca J, Gonzalez-Gay MA. Anti-tumor necrosis factor-alpha therapy improves endothelial function and arterial stiffness in patients with moderate to severe psoriasis: A 6-month prospective study. J Dermatol 2016; 43:1267-1272. [DOI: 10.1111/1346-8138.13398] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/27/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Trinitario Pina
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Alfonso Corrales
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Raquel Lopez-Mejias
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Susana Armesto
- Dermatology Division; IDIVAL; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Marcos A. Gonzalez-Lopez
- Dermatology Division; IDIVAL; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Ines Gómez-Acebo
- Department of Epidemiology and Computational Biology; School of Medicine; IDIVAL; University of Cantabria and CIBER Epidemiología y Salud Pública (CIBERESP); Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Begoña Ubilla
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Sara Remuzgo-Martínez
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - M. Carmen Gonzalez-Vela
- Pathology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Ricardo Blanco
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Jose L. Hernández
- Department of Internal Medicine; IDIVAL; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Javier Llorca
- Dermatology Division; IDIVAL; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| | - Miguel A. Gonzalez-Gay
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases; Rheumatology Division; Hospital Universitario Marqués de Valdecilla; University of Cantabria; Santander Spain
| |
Collapse
|
78
|
Shen J, Shang Q, Tam LS. Targeting inflammation in the prevention of cardiovascular disease in patients with inflammatory arthritis. Transl Res 2016; 167:138-51. [PMID: 26051628 DOI: 10.1016/j.trsl.2015.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 12/15/2022]
Abstract
Patients with inflammatory arthritis have increased risk of cardiovascular diseases (CVDs) compared with the general population. Subclinical carotid atherosclerosis and increased arterial stiffness are also common in these patients, which may serve as surrogate end points for cardiovascular (CV) events in clinical trials. Although exact mechanisms are still unclear, persistent systemic inflammation in patients with inflammatory arthritis may contribute to the development of CVD. Dysregulated innate immunity pathways in these patients may also play a role in accelerating atherosclerosis. During the last decade, effective suppression of inflammation by biological disease-modifying antirheumatic drugs has improved the disease outcome dramatically in patients with inflammatory arthritis. Growing evidence suggests that antitumor necrosis factor (TNF) therapy may prevent CVD in patients with rheumatoid arthritis. Nonetheless, data on non-TNF biologics are limited. Whether anti-TNF therapy may prevent CVD in patients with spondyloarthritis also remained unclear. In this review, we summarized the effect of both anti-TNF and non-TNF biologics on the CV system, including traditional CVD risk factors, endothelial function, arterial stiffness, subclinical atherosclerosis, and clinical CVD in patients with inflammatory arthritis.
Collapse
Affiliation(s)
- Jiayun Shen
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Qing Shang
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
79
|
Boyd T, Kavanaugh A. Novel approaches to biological therapy for psoriatic arthritis. Expert Opin Biol Ther 2015; 16:173-86. [DOI: 10.1517/14712598.2016.1118045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
80
|
Schaarschmidt ML, Kromer C, Herr R, Schmieder A, Sonntag D, Goerdt S, Peitsch WK. Patient Preferences for Biologicals in Psoriasis: Top Priority of Safety for Cardiovascular Patients. PLoS One 2015; 10:e0144335. [PMID: 26633680 PMCID: PMC4669171 DOI: 10.1371/journal.pone.0144335] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/17/2015] [Indexed: 02/08/2023] Open
Abstract
Patients with psoriasis are often affected by comorbidities, which largely influence treatment decisions. Here we performed conjoint analysis to assess the impact of comorbidities on preferences of patients with moderate-to-severe psoriasis for outcome (probability of 50% and 90% improvement, time until response, sustainability of success, probability of mild and severe adverse events (AE), probability of ACR 20 response) and process attributes (treatment location, frequency, duration and delivery method) of biologicals. The influence of comorbidities on Relative Importance Scores (RIS) was determined with analysis of variance and multivariate regression. Among the 200 participants completing the study, 22.5% suffered from psoriatic arthritis, 31.5% from arterial hypertension, 15% from cardiovascular disease (myocardial infarction, stroke, coronary artery disease, and/or arterial occlusive disease), 14.5% from diabetes, 11% from hyperlipidemia, 26% from chronic bronchitis or asthma and 12.5% from depression. Participants with psoriatic arthritis attached greater importance to ACR 20 response (RIS = 10.3 vs. 5.0, p<0.001; β = 0.278, p<0.001) and sustainability (RIS = 5.8 vs. 5.0, p = 0.032) but less value to time until response (RIS = 3.4 vs. 4.8, p = 0.045) than those without arthritis. Participants with arterial hypertension were particularly interested in a low risk of mild AE (RIS 9.7 vs. 12.1; p = 0.033) and a short treatment duration (RIS = 8.0 vs. 9.6, p = 0.002). Those with cardiovascular disease worried more about mild AE (RIS = 12.8 vs. 10, p = 0.027; β = 0.170, p = 0.027) and severe AE (RIS = 23.2 vs. 16.2, p = 0.001; β = 0.203, p = 0.007) but cared less about time until response (β = -0.189, p = 0.013), treatment location (β = -0.153, p = 0.049), frequency (β = -0.20, p = 0.008) and delivery method (β = -0.175, p = 0.023) than others. Patients’ concerns should be addressed in-depth when prescribing biologicals to comorbid patients, keeping in mind that TNF antagonists may favourably influence cardiovascular risk.
Collapse
Affiliation(s)
- Marthe-Lisa Schaarschmidt
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian Kromer
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Raphael Herr
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Astrid Schmieder
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Diana Sonntag
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sergij Goerdt
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Wiebke K. Peitsch
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- * E-mail:
| |
Collapse
|
81
|
Ogdie A, Eder L. Improving cardiovascular health and metabolic comorbidities in patients with psoriatic arthritis. ACTA ACUST UNITED AC 2015; 10:451-459. [PMID: 27134682 DOI: 10.2217/ijr.15.45] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Numerous studies have suggested a link between psoriatic arthritis (PsA) and comorbidities, in particular cardiovascular disease and metabolic comorbidities such as diabetes. The co-existence of these comorbidities is likely the result of systemic inflammation. In order to improve the health of patients with PsA and provide optimal care, these comorbidities must be addressed. However, little is known about how to improve metabolic and cardiovascular health in patients with PsA. In this perspective, we describe the research needs in the area of improving cardiovascular disease and metabolic comorbidities among patients with PsA.
Collapse
Affiliation(s)
- Alexis Ogdie
- Perelman School of Medicine, University of Pennsylvania, White Building, Room 5024, 3400 Spruce St, Philadelphia, PA 19104, USA
| | - Lihi Eder
- Centre for Prognosis Studies in the Rheumatic Diseases, University of Toronto, Toronto Western Hospital, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| |
Collapse
|
82
|
|
83
|
Eder L, Gladman DD. Atherosclerosis in psoriatic disease: latest evidence and clinical implications. Ther Adv Musculoskelet Dis 2015; 7:187-95. [PMID: 26425147 DOI: 10.1177/1759720x15591801] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
It is widely accepted that atherosclerosis is caused by chronic low-grade inflammation that results from an interaction between immune mechanisms and metabolic abnormalities within the vessel wall. Population-based studies have found an increased cardiovascular risk in patients with psoriasis and psoriatic arthritis (PsA). This risk is higher in patients with severe disease phenotypes, such as those with severe psoriasis and with musculoskeletal inflammation. Higher levels of inflammatory biomarkers also predict the development of clinical cardiovascular events in these patients. The effect of medications used for PsA on cardiovascular risk is limited to observational studies. Antitumor necrosis factor agents and methotrexate have been associated with reduced cardiovascular risk. These data highlight the importance of screening for cardiovascular risk factors in these patients.
Collapse
Affiliation(s)
- Lihi Eder
- Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Dafna D Gladman
- Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital, 399 Bathurst St. 1E-410B, Toronto, Ontario, Canada M5T 2S8
| |
Collapse
|
84
|
Arida A, Protogerou AD, Konstantonis G, Konsta M, Delicha EM, Kitas GD, Sfikakis PP. Subclinical Atherosclerosis Is Not Accelerated in Patients with Ankylosing Spondylitis with Low Disease Activity: New Data and Metaanalysis of Published Studies. J Rheumatol 2015; 42:2098-105. [PMID: 26428207 DOI: 10.3899/jrheum.150316] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Chronic inflammatory rheumatic diseases are associated with accelerated atherosclerosis, but data in ankylosing spondylitis (AS) are limited and the relative contribution of inflammation versus classical cardiovascular (CV) risk factors remains a matter of controversy. We addressed this in an original study and a metaanalysis of previous studies. METHODS Atheromatic plaques in carotid and femoral arteries, carotid hypertrophy [intima-media thickness (IMT), cross-sectional area], and carotid stiffness by ultrasound, as well as aortic stiffness by pulse wave velocity, were examined in consecutive nondiabetic, CV disease (CVD)-free patients with AS. Healthy individuals carefully matched 1:1 with patients for age, sex, smoking habits, hyperlipidemia, and hypertension served as controls. A metaanalysis of original studies that examined subclinical atherosclerosis in patients with AS versus controls with comparable CVD risk factors was also performed. RESULTS Carotid and femoral atheromatic plaques were slightly less prevalent compared with controls in a contemporary cohort consisting of 67 patients with AS (82% men), aged 47.5 ± 12.5 years (mean ± SD), with a median disease duration of 12 years and a Bath AS Disease Activity Index (BASDAI) of 1.8 (interquartile range 0.4-3.6), of whom 66% were receiving anti-tumor necrosis factor (TNF) treatment. Carotid hypertrophy and stiffness, as well as aortic stiffness, were similar between patients and their matched controls. Metaanalysis of all published studies revealed a significantly increased carotid IMT, but not plaque burden, in AS versus controls. Notably, however, increased IMT was not evident in studies involving patients with low disease activity (mean BASDAI < 4) or in those studies that included > 50% of patients treated with anti-TNF. CONCLUSION Low AS disease activity is not associated with accelerated atherosclerosis.
Collapse
Affiliation(s)
- Aikaterini Arida
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School
| | - Athanasios D Protogerou
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School
| | - George Konstantonis
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School
| | - Maria Konsta
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School
| | - Evi M Delicha
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School
| | - George D Kitas
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School
| | - Petros P Sfikakis
- From the Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital; Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School, Athens, Greece.A. Arida, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; A.D. Protogerou, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G. Konstantonis, MSc, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; M. Konsta, MD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; E.M. Delicha, PhD, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; G.D. Kitas, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School; P.P. Sfikakis, Professor, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Laikon Hospital, and Joint Academic Rheumatology Programme, National Kapodistrian University of Athens Medical School.
| |
Collapse
|
85
|
González-Quintanilla V, Toriello M, Palacio E, González-Gay MA, Castillo J, Montes S, Martínez-Nieto R, Fernandez J, Rojo A, Gutiérrez S, Pons E, Oterino A. Systemic and cerebral endothelial dysfunction in chronic migraine. A case-control study with an active comparator. Cephalalgia 2015; 36:552-60. [PMID: 26395894 DOI: 10.1177/0333102415607857] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 08/30/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVE Unlike migraine and migraine with aura, little information exists regarding chronic migraine (CM) as a risk factor for cardiovascular disease. In this study we aim to determine whether an association between CM and endothelial dysfunction exists. METHODS Individuals 18 years and older diagnosed with episodic migraine (EM) and CM according to ICHD criteria were studied. After an overnight fast and abstinence from vasoactive drugs, ultrasound studies were performed and blood samples taken from patients and matched controls according to internationally agreed on protocols. RESULTS A total of 113 individuals were enrolled (35 CM, 37 EM, 41 controls). CM patients had a lower percentage of flow-mediated vasodilation (FMD; difference of means = 5.03%; p = 1.0E-6) and breath-holding index (BHI; difference of means 0.754; p = 2.0E-6), as well as increased carotid intima media thickness (cIMT; difference of means = 0.128 mm; p = 7.0E-5) than controls. The EM patients and controls comparison found similar, but less pronounced, differences: decreased BHI (p = 0.031), and increased cIMT (p = 0.028). Fibrinogen (r = 0.277; p = 0.006), C-reactive protein (r = 0.288; p = 0.003), and erythrocyte rate sedimentation (r = 0.298; p = 0.002) also correlated with cIMT, and inversely with BHImV and FMD. CONCLUSIONS Migraine is associated with systemic and cerebral endothelial dysfunction demonstrated by ultrasound studies and biological markers. The degree of these changes was strongly associated with the severity of migraine. Our data indicate that migraine may be a cerebral disorder with systemic endothelial damage.
Collapse
Affiliation(s)
| | - María Toriello
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Enrique Palacio
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Miguel A González-Gay
- Service of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Jesús Castillo
- Health Service of Camargo Costa, Servicio Cántabro de Salud, Spain
| | - Silvia Montes
- Health Service of Camargo Costa, Servicio Cántabro de Salud, Spain
| | | | - Jenifer Fernandez
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Alvaro Rojo
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Silvia Gutiérrez
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Enar Pons
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| | - Agustín Oterino
- Service of Neurology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Spain
| |
Collapse
|
86
|
Ríos-Navarro C, de Pablo C, Collado-Diaz V, Orden S, Blas-Garcia A, Martínez-Cuesta MÁ, Esplugues JV, Alvarez A. Differential effects of anti-TNF-α and anti-IL-12/23 agents on human leukocyte-endothelial cell interactions. Eur J Pharmacol 2015; 765:355-65. [PMID: 26344475 DOI: 10.1016/j.ejphar.2015.08.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 12/14/2022]
Abstract
Enhanced leukocyte recruitment is an inflammatory process that occurs during early phases of the vascular dysfunction that characterises atherosclerosis. We evaluated the impact of anti-TNF-α (adalimumab, infliximab and etanercept) and anti-IL-12/23 (ustekinumab) on interactions between human leukocytes and endothelial cells in a flow chamber that reproduced in vivo conditions. Clinical concentrations of anti-TNF-α were evaluated on the leukocyte recruitment induced by a variety of endothelial (TNF-α, interleukin-1β, lymphotoxin-α and angiotensin-II) and leukocyte (PAF, IL-12 and IL-23) stimuli related to inflammation and atherosclerosis. Treatment with anti-TNF-α, even before or after establishing the inflammatory situation induced by TNF-α, diminished leukocyte-endothelial cell interactions induced by this stimuli. Our results also implicated adhesion molecules (ICAM-1, VCAM-1 and E-selectin) in the actions of anti-TNF-α in terms of leukocyte adhesion to endothelium. However, anti-TNF-α drugs did not influence the actions of interleukin-1β, but prevented those of lymphotoxin-α and angiotensin-II. However, once established, inflammatory response elicited by the latter three stimuli could not be reversed. Pre-treatment with anti-TNF-α, also prevented leukocyte actions induced by IL-23 on PBMC rolling flux and rolling velocity and by IL-12 on PMN adhesion. Ustekinumab exhibited a more discreet profile, having no effect on leukocyte recruitment induced by any of the endothelial stimuli, while blocking the effects of IL-23 on leukocyte activation and those of IL-12 on PMN adhesion and PAF on PBMC rolling velocity. These findings endorse the idea that biological anti-inflammatory drugs, in particular anti-TNF-α, have the capacity to influence cardiovascular risk accompanying psoriasis and rheumatoid arthritis by ameliorating vascular inflammation.
Collapse
Affiliation(s)
- Cesar Ríos-Navarro
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO- Hospital Universitario Dr. Peset, Valencia, Spain.
| | - Carmen de Pablo
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.
| | - Víctor Collado-Diaz
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO- Hospital Universitario Dr. Peset, Valencia, Spain.
| | - Samuel Orden
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO- Hospital Universitario Dr. Peset, Valencia, Spain.
| | - Ana Blas-Garcia
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO- Hospital Universitario Dr. Peset, Valencia, Spain.
| | | | - Juan V Esplugues
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO- Hospital Universitario Dr. Peset, Valencia, Spain.
| | - Angeles Alvarez
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; Fundación General Universidad de Valencia, Valencia, Spain.
| |
Collapse
|
87
|
Le risque cardiovasculaire dans la spondyloarthrite axiale. Presse Med 2015; 44:907-11. [DOI: 10.1016/j.lpm.2015.03.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/16/2015] [Accepted: 03/25/2015] [Indexed: 12/28/2022] Open
|
88
|
Berg IJ, Semb AG, van der Heijde D, Kvien TK, Olsen IC, Dagfinrud H, Provan SA. CRP and ASDAS are associated with future elevated arterial stiffness, a risk marker of cardiovascular disease, in patients with ankylosing spondylitis: results after 5-year follow-up. Ann Rheum Dis 2015; 74:1562-6. [PMID: 25795906 DOI: 10.1136/annrheumdis-2014-206773] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 03/02/2015] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To identify factors associated with elevated arterial stiffness in a 5-year follow-up of patients with ankylosing spondylitis (AS). METHODS C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), Bath AS disease activity index (BASDAI) and AS disease activity score (ASDAS) were recorded in 2003, and arterial stiffness (Augmentation Index (AIx) and pulse wave velocity (PWV)) in 2008/2009. Patients were grouped into quartiles according to baseline CRP, ESR and BASDAI and four ASDAS groups. Trend analyses were performed using ANCOVA (AIx/PWV as dependent variable) with separate models for CRP, ESR, BASDAI and ASDAS (age and gender adjusted). Independent predictors of future AIx and PWV levels were identified in multivariate linear regression models. RESULTS In total, 85 patients participated. Increasing baseline values of CRP, ESR and ASDAS were associated with elevated AIx on follow-up (p(trend) 0.01, 0.05 and 0.04, respectively). Similar non-significant patterns were seen for PWV. In the multivariate analyses, baseline CRP and ASDAS were independently associated with future elevated AIx (p=0.03 and0.02, respectively). In the multivariate PWV model, results for CRP and ASDAS were non-significant. CONCLUSIONS Baseline CRP and ASDAS were associated with future elevated arterial stiffness measured as AIx, supporting that disease activity is related to future risk of cardiovascular disease in patients with AS.
Collapse
Affiliation(s)
- Inger Jorid Berg
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Anne Grete Semb
- Department of Rheumatology, Preventive Cardio-Rheuma clinic, Diakonhjemmet Hospital, Oslo, Norway
| | - Désirée van der Heijde
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tore K Kvien
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Inge C Olsen
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Hanne Dagfinrud
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Sella A Provan
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
89
|
Khan R, Spagnoli V, Tardif JC, L'Allier PL. Novel anti-inflammatory therapies for the treatment of atherosclerosis. Atherosclerosis 2015; 240:497-509. [DOI: 10.1016/j.atherosclerosis.2015.04.783] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 12/18/2022]
|
90
|
Chaabo K, Kirkham B. Rheumatoid Arthritis - Anti-TNF. Int Immunopharmacol 2015; 27:180-4. [PMID: 25962818 DOI: 10.1016/j.intimp.2015.04.051] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 01/01/2023]
Abstract
This review will focus on the recent information and strategies now informing best use of TNF inhibitor therapy in RA. These issues include the role of TNFi therapy in early RA management, anti-drug antibodies in TNFi therapy, updates on safety and optimal dosage regimens in long term management.
Collapse
Affiliation(s)
- Khaldoun Chaabo
- Rheumatology Department, Guy's & St Thomas' NHS Foundation Trust, London SE1 9RT, UK
| | - Bruce Kirkham
- Rheumatology Department, Guy's & St Thomas' NHS Foundation Trust, London SE1 9RT, UK.
| |
Collapse
|
91
|
Shen J, Shang Q, Li EK, Leung YY, Kun EW, Kwok LW, Li M, Li TK, Zhu TY, Yu CM, Tam LS. Cumulative inflammatory burden is independently associated with increased arterial stiffness in patients with psoriatic arthritis: a prospective study. Arthritis Res Ther 2015; 17:75. [PMID: 25890227 PMCID: PMC4384323 DOI: 10.1186/s13075-015-0570-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/20/2015] [Indexed: 11/22/2022] Open
Abstract
Introduction The aim of this study was to examine whether the cumulative inflammatory burden is associated with an increase in arterial stiffness in a prospective cohort of psoriatic arthritis (PsA) patients. Methods In total, 72 PsA patients were followed for a median of 6.5 years. Cumulative inflammatory burden was represented by the cumulative averages of repeated measures of erythrocyte sedimentation rate (ca-ESR) and C-reactive protein (ca-CRP). Brachial-ankle pulse wave velocity (PWV) was measured at the last visit. We also included 47 healthy controls for PWV assessment. Results PWV was significantly higher in PsA patients compared with healthy controls after adjustment for age, gender and body weight (1466 ± 29 cm/s versus 1323 ± 38 cm/s, P = 0.008). PsA patients were divided into two groups based on whether their PWV value is ≥1450 cm/s (High PWV group, N = 38) or <1450 cm/s (Low PWV group, N = 34). The High PWV group had a significantly higher ca-ESR (29 (19 to 44) versus 18 (10 to 32) mm/1st hour, P = 0.005) and ca-CRP (0.7 (0.3 to 1.4) versus 0.4 (0.2 to 0.7) mg/dl, P = 0.029). Using regression analysis, high ca-ESR (defined as ≥75th percentile: 37 mm/1st hour) was associated with a higher likelihood of being in the High PWV group (odds ratio (OR): 9.455 (1.939 to 46.093), P = 0.005, adjusted for baseline clinical and cardiovascular risk factors; and 9.111 (1.875 to 44.275), P = 0.006, adjusted for last visit parameters). Conclusions Cumulative inflammatory burden, as reflected by ca-ESR, was associated with increased arterial stiffness in PsA patients even after adjustment for cardiovascular risk factors, emphasizing the important role of chronic inflammation in accelerating the development of cardiovascular risks in PsA patients.
Collapse
Affiliation(s)
- Jiayun Shen
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Qing Shang
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Edmund K Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Ying-Ying Leung
- Department of Rheumatology and Immunology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore, Singapore.
| | - Emily W Kun
- Department of Medicine and Geriatrics, Taipo Hospital, Taipo, Hong Kong, China.
| | - Lai-Wa Kwok
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Martin Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Tena K Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Tracy Y Zhu
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Cheuk-Man Yu
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
92
|
Selmi C. Hot topics in autoimmune diseases: Perspectives from the 2013 Asian Congress of Autoimmunity. Autoimmun Rev 2014; 13:781-7. [DOI: 10.1016/j.autrev.2014.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 05/05/2014] [Indexed: 12/12/2022]
|
93
|
Amaya-Amaya J, Montoya-Sánchez L, Rojas-Villarraga A. Cardiovascular involvement in autoimmune diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:367359. [PMID: 25177690 PMCID: PMC4142566 DOI: 10.1155/2014/367359] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 05/01/2014] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases (AD) represent a broad spectrum of chronic conditions that may afflict specific target organs or multiple systems with a significant burden on quality of life. These conditions have common mechanisms including genetic and epigenetics factors, gender disparity, environmental triggers, pathophysiological abnormalities, and certain subphenotypes. Atherosclerosis (AT) was once considered to be a degenerative disease that was an inevitable consequence of aging. However, research in the last three decades has shown that AT is not degenerative or inevitable. It is an autoimmune-inflammatory disease associated with infectious and inflammatory factors characterized by lipoprotein metabolism alteration that leads to immune system activation with the consequent proliferation of smooth muscle cells, narrowing arteries, and atheroma formation. Both humoral and cellular immune mechanisms have been proposed to participate in the onset and progression of AT. Several risk factors, known as classic risk factors, have been described. Interestingly, the excessive cardiovascular events observed in patients with ADs are not fully explained by these factors. Several novel risk factors contribute to the development of premature vascular damage. In this review, we discuss our current understanding of how traditional and nontraditional risk factors contribute to pathogenesis of CVD in AD.
Collapse
Affiliation(s)
- Jenny Amaya-Amaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 11001000 Bogotá, Colombia
- Mederi, Hospital Universitario Mayor, Calle 24 No. 29-45, 11001000 Bogotá, Colombia
| | - Laura Montoya-Sánchez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 11001000 Bogotá, Colombia
- Mederi, Hospital Universitario Mayor, Calle 24 No. 29-45, 11001000 Bogotá, Colombia
| | - Adriana Rojas-Villarraga
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 11001000 Bogotá, Colombia
- Mederi, Hospital Universitario Mayor, Calle 24 No. 29-45, 11001000 Bogotá, Colombia
| |
Collapse
|