1
|
Husaini ASA, Fathima A, Halawa D, Aakel N, Erre GL, Giordo R, Zayed H, Pintus G. Exploring endothelial dysfunction in major rheumatic diseases: current trends and future directions. J Mol Med (Berl) 2025:10.1007/s00109-025-02539-8. [PMID: 40229608 DOI: 10.1007/s00109-025-02539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The relationship between rheumatic diseases (RDs) and endothelial dysfunction (ED) is intricate and multifaceted, with chronic inflammation and immune system dysregulation playing key roles. RDs, including Osteoarthritis (OA), Rheumatoid arthritis (RA), Systemic Lupus erythematosus (SLE), Ankylosing spondylitis (AS), Psoriatic arthritis (PsA), Sjogren's syndrome (SS), Systemic sclerosis (SSc), Polymyalgia rheumatica (PMR) are characterized by chronic inflammation and immune dysregulation, leading to ED. ED is marked by reduced nitric oxide (NO) production, increased oxidative stress, and heightened pro-inflammatory and prothrombotic activities, which are crucial in the development of cardiovascular disease (CVD) and systemic inflammation. This association persists even in RD patients without conventional cardiovascular risk factors, suggesting a direct impact of RD-related inflammation on endothelial function. Studies also show that ED significantly contributes to atherosclerosis, thereby elevating cardiovascular risk in RD patients. This review synthesizes the molecular mechanisms connecting major RDs and ED, highlighting potential biomarkers and therapeutic targets. Ultimately, the review aims to enhance understanding of the complex interactions leading to ED in rheumatic patients and inform strategies to mitigate cardiovascular risks and improve patient outcomes.
Collapse
Affiliation(s)
- Arshiya S Anwar Husaini
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, 2713, Qatar
| | - Aseela Fathima
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, 2713, Qatar
| | - Dunia Halawa
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, 2713, Qatar
| | - Nada Aakel
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, 2713, Qatar
| | - Gian Luca Erre
- Rheumatology Unit, University Hospital (AOU) of Sassari, Sassari, Italy
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Roberta Giordo
- Department of Biomedical Sciences, University of Sassari, Sassari, 07100, Italy
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, 2713, Qatar.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari, 07100, Italy.
- Department of Medical Laboratory Sciences, College of Health Sciences, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
2
|
Nagy S, Ditchek J, Kesselman MM. Coronary Artery Calcification in Rheumatoid Arthritis Patients: A Systematic Review. Cureus 2024; 16:e70517. [PMID: 39479072 PMCID: PMC11524640 DOI: 10.7759/cureus.70517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Rheumatoid arthritis (RA) is one of the leading autoimmune causes of inflammatory arthropathy worldwide. The musculoskeletal impacts of RA are well described within the literature. More recently, research efforts have highlighted that inflammation associated with the condition is not solely isolated to the joint synovium. Specifically, data has demonstrated that the cardiovascular system is negatively impacted by inflammation tied to RA, with adverse cardiovascular outcomes considered the leading cause of mortality among patients with RA. One approach to determine the risk for cardiovascular disease (CVD) is computed tomography (CT) coronary angiography, a noninvasive imaging approach that analyzes the calcifications within the coronary vessels. This has increasingly been utilized to analyze plaque burden and vessel obstruction, which is measured using the coronary artery calcium (CAC) score. A total of 305 articles were analyzed, and 11 articles were selected for this review based on inclusion and exclusion criteria. The results indicated that nearly 60% of patients with RA experienced an elevated CAC score. As such, patients with RA likely carry a higher risk for adverse cardiovascular outcomes as compared to their healthy counterparts. Additional research is warranted based on these findings to determine whether the addition of CT coronary angiography and analysis of laboratory markers for CVD, including lipid markers in standard protocols for RA comorbid assessment, would help to reduce adverse cardiovascular complications.
Collapse
Affiliation(s)
- Stephanie Nagy
- Rheumatology, Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Jordan Ditchek
- Radiology, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, USA
| | - Marc M Kesselman
- Rheumatology, Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| |
Collapse
|
3
|
Araujo T, Spadella M, Carlos C, Tirapelli C, Chagas E, Pinheiro J, Chies A. Adjuvant-induced arthritis promotes vascular hyporesponsiveness to phenylephrine through a nitric oxide-related mechanism. Braz J Med Biol Res 2024; 57:e13304. [PMID: 38775546 PMCID: PMC11101166 DOI: 10.1590/1414-431x2024e13304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/04/2024] [Indexed: 05/25/2024] Open
Abstract
Arthritis has important cardiovascular repercussions. Phenylephrine-induced vasoconstriction is impaired in rat aortas in the early phase of the adjuvant-induced arthritis (AIA), around the 15th day post-induction. Therefore, the present study aimed to verify the effects of AIA on hyporesponsiveness to phenylephrine in rat aortas. AIA was induced by intradermal injection of Mycobacterium tuberculosis (3.8 mg/dL) in the right hind paw of male Wistar rats (n=27). Functional experiments in isolated aortas were carried out 15 days after AIA induction. Morphometric and stereological analyses of the aortas were also performed 36 days after the induction of AIA. AIA did not promote structural modifications in the aortas at any of the time points studied. AIA reduced phenylephrine-induced contraction in endothelium-intact aortas, but not in endothelium-denuded aortas. However, AIA did not change KCl-induced contraction in either endothelium-intact or denuded aortas. L-NAME (non-selective NOS inhibitor), 1400W (selective iNOS inhibitor), and ODQ (guanylyl cyclase inhibitor) reversed AIA-induced hyporesponsiveness to phenylephrine in intact aortas. 7-NI (selective nNOS inhibitor) increased the contraction induced by phenylephrine in aortas from AIA rats. In summary, the hyporesponsiveness to phenylephrine induced by AIA was endothelium-dependent and mediated by iNOS-derived NO through activation of the NO-guanylyl cyclase pathway.
Collapse
Affiliation(s)
- T.S. Araujo
- Laboratório de Farmacologia, Faculdade de Medicina de Marília,
Marília, SP, Brasil
| | - M.A. Spadella
- Laboratório de Embriologia Humana, Faculdade de Medicina de
Marília, Marília, SP, Brasil
| | - C.P. Carlos
- Laboratório de Pesquisa Experimental, Faculdade de Medicina
Faceres, São José do Rio Preto, SP, Brasil
- Disciplina de Fisiologia, Faculdade de Medicina de Marília,
Marília, SP, Brasil
| | - C.R. Tirapelli
- Laboratório de Farmacologia Cardiovascular, Escola de Enfermagem
de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - E.F.B. Chagas
- Centro Interdisciplinar de Diabetes, Universidade de Marília,
Marília, SP, Brasil
- Programa de Mestrado Interdisciplinar em Interações Estruturais
e Funcionais em Reabilitação, Universidade de Marília, Marília, SP, Brasil
- Programa de Mestrado em Saúde e Envelhecimento, Faculdade de
Medicina de Marília, Marília, SP, Brasil
| | - J.C.D. Pinheiro
- Laboratório de Farmacologia, Faculdade de Medicina de Marília,
Marília, SP, Brasil
| | - A.B. Chies
- Laboratório de Farmacologia, Faculdade de Medicina de Marília,
Marília, SP, Brasil
| |
Collapse
|
4
|
Okur S, Okumuş Z. Effects of low-level laser therapy and therapeutic ultrasound on Freund's complete adjuvant-induced knee arthritis model in rats. Arch Rheumatol 2023; 38:32-43. [PMID: 37235114 PMCID: PMC10208612 DOI: 10.46497/archrheumatol.2022.9409] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/05/2022] [Indexed: 08/08/2023] Open
Abstract
OBJECTIVES The aim of this study was to evaluate and monitor the effect of low-level laser therapy (LLLT) and therapeutic ultrasound (TU) alone, or combined with intra-articular prednisolone (P) in Freund's complete adjuvant (FCA)-induced knee arthritis model in rats. MATERIALS AND METHODS A total of 56 adult male Wistar rats were divided into seven groups: control (C), disease control (RA), P, TU, LLLT (L), P + TU (P+TU), P + LLLT (P+L) groups. The skin temperature, radiography, joint volume, serum rheumatoid factor (RF), interleukin (IL)-1β, serum tumor necrosis factor-alpha (TNF-α), and histopathological evaluation of joint were performed. RESULTS Thermal imaging and radiographic examination provided results consistent with the severity of the disease. The mean joint temperature (°C) was the highest in the RA (36.2±1.6) group on Day 28. The P+TU and P+L groups significantly decreased radiological scores at the end of the study. The rat serum TNF-α, IL-1β, and RF levels in all groups were significantly higher compared to the C group (p<0.05). Compared to the RA group, serum TNF-α, IL-1β, and RF levels were significantly lower in the treatment groups (p<0.05). The P+TU and P+L group was showed minimal chondrocyte degeneration and cartilage erosion and mild cartilage fibrillation and mononuclear cell infiltration of synovial membrane compared to the P, TU, and L group. CONCLUSION The LLLT and TU effectively reduced inflammation. In addition, a more effective result was obtained from the use of LLLT and TU combined with intra-articular P. This result may be due to insufficient dose of LLLT and TU, thus further studies should be focus on at higher dose ranges on FCA arthritis model in rats.
Collapse
Affiliation(s)
- Sıtkıcan Okur
- Department of Veterinary Surgery, Atatürk University Faculty of Veterinary Medicine, Erzurum, Türkiye
| | - Zafer Okumuş
- Department of Veterinary Surgery, Atatürk University Faculty of Veterinary Medicine, Erzurum, Türkiye
| |
Collapse
|
5
|
Manilall A, Mokotedi L, Gunter S, Le Roux R, Fourie S, Flanagan CA, Millen AME. Increased protein phosphatase 5 expression in inflammation-induced left ventricular dysfunction in rats. BMC Cardiovasc Disord 2022; 22:539. [PMID: 36494772 PMCID: PMC9732989 DOI: 10.1186/s12872-022-02977-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Titin phosphorylation contributes to left ventricular (LV) diastolic dysfunction. The independent effects of inflammation on the molecular pathways that regulate titin phosphorylation are unclear. METHODS We investigated the effects of collagen-induced inflammation and subsequent tumor necrosis factor-α (TNF-α) inhibition on mRNA expression of genes involved in regulating titin phosphorylation in 70 Sprague-Dawley rats. LV diastolic function was assessed with echocardiography. Circulating inflammatory markers were quantified by enzyme-linked immunosorbent assay and relative LV gene expression was assessed by Taqman® polymerase chain reaction. Differences in normally distributed variables between the groups were determined by two-way analysis of variance (ANOVA), followed by Tukey post-hoc tests. For non-normally distributed variables, group differences were determined by Kruskal-Wallis tests. RESULTS Collagen inoculation increased LV relative mRNA expression of vascular cell adhesion molecule 1 (VCAM1), pentraxin 3 (PTX3), and inducible nitric oxide synthase (iNOS) compared to controls, indicating local microvascular inflammation. Collagen inoculation decreased soluble guanylate cyclase alpha-2 (sGCα2) and soluble guanylate cyclase beta-2 (sGCβ2) expression, suggesting downregulation of nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling. Inhibiting TNF-α prevented collagen-induced changes in VCAM1, iNOS, sGCα2 and sGCβ2 expression. Collagen inoculation increased protein phosphatase 5 (PP5) expression. Like LV diastolic dysfunction, increased PP5 expression was not prevented by TNF-α inhibition. CONCLUSION Inflammation-induced LV diastolic dysfunction may be mediated by a TNF-α-independent increase in PP5 expression and dephosphorylation of the N2-Bus stretch element of titin, rather than by TNF-α-induced downregulation of NO-sGC-cGMP pathway-dependent titin phosphorylation. The steady rise in number of patients with inflammation-induced diastolic dysfunction, coupled with low success rates of current therapies warrants a better understanding of the systemic signals and molecular pathways responsible for decreased titin phosphorylation in development of LV diastolic dysfunction. The therapeutic potential of inhibiting PP5 upregulation in LV diastolic dysfunction requires investigation.
Collapse
Affiliation(s)
- Ashmeetha Manilall
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Lebogang Mokotedi
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Sulè Gunter
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Regina Le Roux
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Serena Fourie
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Colleen A. Flanagan
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Aletta M. E. Millen
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| |
Collapse
|
6
|
Peyronnel C, Totoson P, Petitcolin V, Bonnefoy F, Guillot X, Saas P, Verhoeven F, Martin H, Demougeot C. Effects of local cryotherapy on systemic endothelial activation, dysfunction, and vascular inflammation in adjuvant-induced arthritis (AIA) rats. Arthritis Res Ther 2022; 24:97. [PMID: 35488311 PMCID: PMC9052534 DOI: 10.1186/s13075-022-02774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Aim This study explored the systemic vascular effects of local cryotherapy with a focus on endothelial changes and arterial inflammation in the model of rat adjuvant-induced arthritis (AIA). Methods Cryotherapy was applied twice a day on hind paws of AIA rats from the onset of arthritis to the acute inflammatory phase. Endothelial activation was studied in the aorta by measuring the mRNA levels of chemokines (CXCL-1, MCP-1 (CCL-2), MIP-1α (CCL-3)) and adhesion molecules (ICAM-1, VCAM-1) by qRT-PCR. Endothelial dysfunction was measured in isolated aortic and mesenteric rings. Aortic inflammation was evaluated via the mRNA expression of pro-inflammatory cytokines (TNF-α, IL-6) by qRT-PCR and leucocyte infiltration analysis (flow cytometry). Plasma levels of TNF-α, IL-6, IL-1β, IL-17A, and osteoprotegerin (OPG) were measured using Multiplex/ELISA. Results AIA was associated with an increased aortic expression of CXCL-1 and ICAM-1 as well as an infiltration of leucocytes and increased mRNA expression of IL-6, IL-1β, and TNF-α. Local cryotherapy, which decreased arthritis score and structural damages, reduced aortic mRNA expression of CXCL-1, IL-6, IL-1β, and TNF-α, as well as aortic infiltration of leucocytes (T lymphocytes, monocytes/macrophages, neutrophils) and improved acetylcholine-induced vasorelaxation in the aorta and mesenteric arteries. Plasma levels of IL-17A and OPG were significantly reduced by cryotherapy, while the number of circulating leucocytes was not. IL-17A levels positively correlated with endothelial activation and dysfunction. Conclusion In the AIA model, local cryotherapy reduced systemic endothelial activation, immune cell infiltration, and endothelial dysfunction. Mechanistically, the reduction of circulating levels of IL-17A appears as the possible link between joint cooling and the remote vascular effects. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02774-1.
Collapse
Affiliation(s)
- C Peyronnel
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - P Totoson
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - V Petitcolin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - F Bonnefoy
- INSERM UMR 1098 RIGHT, EFS BFC, Univ. Bourgogne Franche-Comté, LabEX LipSTIC, F-25000, Besançon, France
| | - X Guillot
- Service de Rhumatologie, CHU Felix Guyon, Ile de la Réunion, Saint-Denis, France
| | - P Saas
- INSERM UMR 1098 RIGHT, EFS BFC, Univ. Bourgogne Franche-Comté, LabEX LipSTIC, F-25000, Besançon, France
| | - F Verhoeven
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.,Service de Rhumatologie, CHRU Besançon, F-25000, Besançon, France
| | - H Martin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.
| |
Collapse
|
7
|
Totoson P, Peyronnel C, Quirié A, Pédard M, Cefis M, Bermont L, Prigent-Tessier A, Prati C, Tournier M, Wendling D, Marie C, Demougeot C. Tofacitinib improved peripheral endothelial dysfunction and brain-derived neurotrophic factor levels in the rat adjuvant-induced arthritis model. Fundam Clin Pharmacol 2021; 36:363-374. [PMID: 34661311 DOI: 10.1111/fcp.12731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/12/2021] [Indexed: 01/10/2023]
Abstract
This study aimed to explore the effect of Tofacitinib on endothelial dysfunction and cerebral levels of brain-derived neurotrophic factor (BDNF) in the adjuvant-induced arthritis (AIA) rat model. Tofacitinib (10 mg/kg twice a day) or vehicle was administered from the first signs of inflammation. Arthritis scores were daily monitored while other parameters including endothelial function assessed from aortic rings, radiographic scores, blood pressure, heart rate, circulating levels of triglycerides, cholesterol, and interleukin (IL)-1β, tumor necrosis factor-α (TNF-α), IL-17A, and cerebral BDNF levels were determined after 3 weeks of treatment. A group of non-AIA rats served as controls. In AIA rats, as compared with vehicle, Tofacitinib significantly reduced arthritis and radiographic scores, decreased total cholesterol and low-density lipoprotein cholesterol (LDL-C), but changed neither blood pressure nor heart rate and proinflammatory cytokines levels. It also fully restored acetylcholine (Ach)-induced relaxation (p < 0.05) through increased nitric oxide (NO) synthase activity, reduced BH4 deficiency and O2 -° production, decreased cyclo-oxygenase-2 (COX-2)/arginase activities, and enhanced endothelium-derived hyperpolarizing factor (EDHF) production. These effects translated into a decrease in atherogenic index and an elevation of BDNF levels in the prefrontal cortex (p < 0.05) and hippocampus (p < 0.001). The present study identified Tofacitinib as an efficient therapeutic option to reduce cardiovascular risk and improve BDNF-dependent cognition in arthritis.
Collapse
Affiliation(s)
- Perle Totoson
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - Célian Peyronnel
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Sciences de Santé, Dijon, France
| | - Martin Pédard
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Sciences de Santé, Dijon, France
| | - Marina Cefis
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Sciences de Santé, Dijon, France
| | - Laurent Bermont
- Service de Biochimie médicale, CHRU Besançon, Besançon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Sciences de Santé, Dijon, France
| | - Clément Prati
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France.,Service de Rhumatologie, CHRU Besançon, Besançon, France
| | - Maude Tournier
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - Daniel Wendling
- Service de Rhumatologie, CHRU Besançon, Besançon, France.,EA 4266 "Agents Pathogènes et Inflammation", EPILAB, Université Bourgogne Franche-Comté, Besançon, France
| | - Christine Marie
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Sciences de Santé, Dijon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
8
|
Pedard M, Quirié A, Tessier A, Garnier P, Totoson P, Demougeot C, Marie C. A reconciling hypothesis centred on brain-derived neurotrophic factor to explain neuropsychiatric manifestations in rheumatoid arthritis. Rheumatology (Oxford) 2021; 60:1608-1619. [PMID: 33313832 DOI: 10.1093/rheumatology/keaa849] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/27/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease characterized by synovitis leading to joint destruction, pain and disability. Despite efficient antirheumatic drugs, neuropsychiatric troubles including depression and cognitive dysfunction are common in RA but the underlying mechanisms are unclear. However, converging evidence strongly suggests that deficit in brain-derived neurotrophic factor (BDNF) signalling contributes to impaired cognition and depression. Therefore, this review summarizes the current knowledge on BDNF in RA, proposes possible mechanisms linking RA and brain BDNF deficiency including neuroinflammation, cerebral endothelial dysfunction and sedentary behaviour, and discusses neuromuscular electrical stimulation as an attractive therapeutic option.
Collapse
Affiliation(s)
- Martin Pedard
- INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, F-21000, France
| | - Aurore Quirié
- INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, F-21000, France
| | - Anne Tessier
- INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, F-21000, France
| | - Philippe Garnier
- INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, F-21000, France
| | - Perle Totoson
- EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, F-25030, France
| | - Céline Demougeot
- EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, F-25030, France
| | - Christine Marie
- INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, F-21000, France
| |
Collapse
|
9
|
Zamora C, Cantó E, Vidal S. The Dual Role of Platelets in the Cardiovascular Risk of Chronic Inflammation. Front Immunol 2021; 12:625181. [PMID: 33868242 PMCID: PMC8046936 DOI: 10.3389/fimmu.2021.625181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/15/2021] [Indexed: 11/25/2022] Open
Abstract
Patients with chronic inflammatory diseases often exhibit cardiovascular risk. This risk is associated with the systemic inflammation that persists in these patients, causing a sustained endothelial activation. Different mechanisms have been considered responsible for this systemic inflammation, among which activated platelets have been regarded as a major player. However, in recent years, the role of platelets has become controversial. Not only can this subcellular component release pro- and anti-inflammatory mediators, but it can also bind to different subsets of circulating lymphocytes, monocytes and neutrophils modulating their function in either direction. How platelets exert this dual role is not yet fully understood.
Collapse
Affiliation(s)
| | | | - Sílvia Vidal
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| |
Collapse
|
10
|
Endothelial Dysfunction and Extra-Articular Neurological Manifestations in Rheumatoid Arthritis. Biomolecules 2021; 11:biom11010081. [PMID: 33435178 PMCID: PMC7827097 DOI: 10.3390/biom11010081] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/06/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, inflammatory autoimmune disease that affects about 1% of the global population, with a female–male ratio of 3:1. RA preferably affects the joints, with consequent joint swelling and deformities followed by ankylosis. However, evidence has accumulated showing that patients suffering from RA can also develop extra-articular manifestations, including cardiovascular disease states, neuropathies, and multiorgan dysfunction. In particular, peripheral nerve disorders showed a consistent impact in the course of the disease (prevalence about 20%) mostly associated to vasculitis of the nerve vessels leading to vascular ischemia, axonal degeneration, and neuronal demyelination. The pathophysiological basis of this RA-associated microvascular disease, which leads to impairment of assonal functionality, is still to be better clarified. However, endothelial dysfunction and alterations of the so-called brain-nerve barrier (BNB) seem to play a fundamental role. This review aims to assess the potential mechanisms underlying the impairment of endothelial cell functionality in the development of RA and to identify the role of dysfunctional endothelium as a causative mechanism of extra-articular manifestation of RA. On the other hand, the potential impact of lifestyle and nutritional interventions targeting the maintenance of endothelial cell integrity in patients with RA will be discussed as a potential option when approaching therapeutic solutions in the course of the disease.
Collapse
|
11
|
Hansildaar R, Vedder D, Baniaamam M, Tausche AK, Gerritsen M, Nurmohamed MT. Cardiovascular risk in inflammatory arthritis: rheumatoid arthritis and gout. THE LANCET. RHEUMATOLOGY 2021; 3:e58-e70. [PMID: 32904897 PMCID: PMC7462628 DOI: 10.1016/s2665-9913(20)30221-6] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The increased risk of cardiovascular morbidity and mortality in rheumatoid arthritis and gout has been increasingly acknowledged in past decades, with accumulating evidence that gout, just as with rheumatoid arthritis, is an independent cardiovascular risk factor. Although both diseases have a completely different pathogenesis, the underlying pathophysiological mechanisms in systemic inflammation overlap to some extent. Following the recognition that systemic inflammation has an important causative role in cardiovascular disease, anti-inflammatory therapy in both conditions and urate-lowering therapies in gout are expected to lower the cardiovascular burden of patients. Unfortunately, much of the existing data showing that urate-lowering therapy has consistent beneficial effects on cardiovascular outcomes in patients with gout are of low quality and contradictory. We will discuss the latest evidence in this respect. Cardiovascular disease risk management for patients with rheumatoid arthritis and gout is essential. Clinical guidelines and implementation of cardiovascular risk management in daily clinical practice, as well as unmet needs and areas for further investigation, will be discussed.
Collapse
Affiliation(s)
- Romy Hansildaar
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam and Reade, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Daisy Vedder
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam and Reade, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Milad Baniaamam
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam and Reade, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Anne-Kathrin Tausche
- Department of Rheumatology, University Clinic Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - Martijn Gerritsen
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam and Reade, Amsterdam, Netherlands
| | - Michael T Nurmohamed
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam and Reade, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| |
Collapse
|
12
|
Stucker S, Chen J, Watt FE, Kusumbe AP. Bone Angiogenesis and Vascular Niche Remodeling in Stress, Aging, and Diseases. Front Cell Dev Biol 2020; 8:602269. [PMID: 33324652 PMCID: PMC7726257 DOI: 10.3389/fcell.2020.602269] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/05/2020] [Indexed: 02/05/2023] Open
Abstract
The bone marrow (BM) vascular niche microenvironments harbor stem and progenitor cells of various lineages. Bone angiogenesis is distinct and involves tissue-specific signals. The nurturing vascular niches in the BM are complex and heterogenous consisting of distinct vascular and perivascular cell types that provide crucial signals for the maintenance of stem and progenitor cells. Growing evidence suggests that the BM niche is highly sensitive to stress. Aging, inflammation and other stress factors induce changes in BM niche cells and their crosstalk with tissue cells leading to perturbed hematopoiesis, bone angiogenesis and bone formation. Defining vascular niche remodeling under stress conditions will improve our understanding of the BM vascular niche and its role in homeostasis and disease. Therefore, this review provides an overview of the current understanding of the BM vascular niches for hematopoietic stem cells and their malfunction during aging, bone loss diseases, arthritis and metastasis.
Collapse
Affiliation(s)
- Sina Stucker
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fiona E. Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Adawi M, Sabbah F, Tzischinsky O, Blum N, Bragazzi NL, Yehuda I, Tamir S, Romanenko O, Blum A. Sleep disorders and vascular responsiveness in patients with rheumatoid arthritis. J Intern Med 2020; 288:439-445. [PMID: 32330326 DOI: 10.1111/joim.13087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is the most common systemic autoimmune disease characterized by chronic systemic inflammation. Half of the deaths of patients with RA are due to cardiovascular diseases (CVD), considered to be 1.5 to -2.0-fold that in the general population. Patients with RA also experience poor sleep, which by itself is associated with endothelial dysfunction, CVD events and sudden death. Our aim was to study the mechanistic pathways and the correlations between sleep efficiency and vascular reactivity of patients with RA. METHODS AND RESULTS A prospective study that evaluated quality of sleep using ACTi Graphs, vascular inflammation and endothelial function of 18 patients with RA. Inflammation was studied by levels of E-selectin, intercellular adhesion molecule 1 (ICAM-1) and NO in serum. Endothelial function was studied using the brachial artery plethysmography method. Eighteen RA patients (aged 57.56 ± 13.55 years; 16 women) with a long-standing active RA: Eight patients had impaired sleep efficiency and 10 had a good sleep efficiency. Those who had an impaired sleep had larger baseline diameters of the brachial artery (0.39 ± 0.08 cm vs. 0.32 ± 0.04 cm; P = 0.02). Negative correlations were found between baseline brachial artery diameter and sleep efficiency (P = 0.01), and with NO level (P = 0.04). Stepwise regression found that brachial artery diameter at baseline and NO level could predict sleep efficiency (r2 = 0.543, P = 0.01). CONCLUSION Vascular reactivity could predict quality of sleep in patients with RA. Quality of sleep may serve as an independent CVD risk factor in patients with RA.
Collapse
Affiliation(s)
- M Adawi
- From the, Department of Medicine, the Rheumatology Unit, Azrieli Faculty of Medicine, Baruch Padeh Medical Center, Lower Galilee, Israel
| | - F Sabbah
- From the, Department of Medicine, the Rheumatology Unit, Azrieli Faculty of Medicine, Baruch Padeh Medical Center, Lower Galilee, Israel
| | - O Tzischinsky
- Max Stern Academic College of Emek Yezreel, Emek Yezreel, Israel
| | - N Blum
- Max Stern Academic College of Emek Yezreel, Emek Yezreel, Israel
| | - N L Bragazzi
- Department of Health Sciences, School of Public Health, University of Genoa, Genoa, Italy
| | - I Yehuda
- Department of Nutrition Sciences & MIGAL, Galilee Technology Center, Tel-Hai Academic College, Kiryat Shmona, Israel
| | - S Tamir
- Department of Nutrition Sciences & MIGAL, Galilee Technology Center, Tel-Hai Academic College, Kiryat Shmona, Israel
| | - O Romanenko
- Department of Nutrition Sciences & MIGAL, Galilee Technology Center, Tel-Hai Academic College, Kiryat Shmona, Israel
| | - A Blum
- From the, Department of Medicine, the Rheumatology Unit, Azrieli Faculty of Medicine, Baruch Padeh Medical Center, Lower Galilee, Israel.,Azrieli Faculty of Medicine, Vascular Biology Center, Baruch Padeh Medical Center, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
14
|
Pita LM, Spadella MA, Montenote MC, Oliveira PB, Chies AB. Repercussions of adjuvant-induced arthritis on body composition, soleus muscle, and heart muscle of rats. ACTA ACUST UNITED AC 2020; 53:e8969. [PMID: 32130291 PMCID: PMC7057929 DOI: 10.1590/1414-431x20198969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/04/2019] [Indexed: 01/07/2023]
Abstract
This study investigated the repercussions of adjuvant-induced arthritis (AIA) on
body composition and the structural organization of the soleus and cardiac
muscles, including their vascularization, at different times of disease
manifestation. Male rats were submitted to AIA induction by intradermal
administration of 100 μL of Mycobacterium tuberculosis (50
mg/mL), in the right hind paw. Animals submitted to AIA were studied 4 (AIA4),
15 (AIA15), and 40 (AIA40) days after AIA induction as well as a control group
of animals not submitted to AIA. Unlike the control animals, AIA animals did not
gain body mass throughout the evolution of the disease. AIA reduced food
consumption, but only on the 40th day after induction. In the soleus muscle, AIA
reduced the wet mass in a time-dependent manner but increased the capillary
density by the 15th day and the fiber density by both 15 and 40 days after
induction. The diameter of the soleus fiber decreased from the 4th day after AIA
induction as well as the capillary/fiber ratio, which was most evident on the
40th day. Moreover, AIA induced slight histopathological changes in the cardiac
muscle that were more evident on the 15th day after induction. In conclusion,
AIA-induced changes in body composition as well as in the soleus muscle fibers
and vasculature have early onset but are more evident by the 15th day after
induction. Moreover, the heart may be a target organ of AIA, although less
sensitive than skeletal muscles.
Collapse
Affiliation(s)
- L M Pita
- Laboratório de Farmacologia, Faculdade de Medicina de Marília, Marília, SP, Brasil
| | - M A Spadella
- Laboratório de Embriologia Humana, Faculdade de Medicina de Marília, Marília, SP, Brasil
| | - M C Montenote
- Departamento de Farmacologia, Instituto de Biociências de Botucatu, Botucatu, SP, Brasil
| | - P B Oliveira
- Laboratório de Farmacologia, Faculdade de Medicina de Marília, Marília, SP, Brasil
| | - A B Chies
- Laboratório de Farmacologia, Faculdade de Medicina de Marília, Marília, SP, Brasil
| |
Collapse
|
15
|
Mangoni AA, Tommasi S, Zinellu A, Sotgia S, Bassu S, Piga M, Erre GL, Carru C. Methotrexate and Vasculoprotection: Mechanistic Insights and Potential Therapeutic Applications in Old Age. Curr Pharm Des 2019; 25:4175-4184. [DOI: 10.2174/1381612825666191112091700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Increasing age is a strong, independent risk factor for atherosclerosis and cardiovascular disease. Key
abnormalities driving cardiovascular risk in old age include endothelial dysfunction, increased arterial stiffness,
blood pressure, and the pro-atherosclerotic effects of chronic, low-grade, inflammation. The identification of
novel therapies that comprehensively target these alterations might lead to a major breakthrough in cardiovascular
risk management in the older population. Systematic reviews and meta-analyses of observational studies have
shown that methotrexate, a first-line synthetic disease-modifying anti-rheumatic drug, significantly reduces
cardiovascular morbidity and mortality in patients with rheumatoid arthritis, a human model of systemic
inflammation, premature atherosclerosis, and vascular aging. We reviewed in vitro and in vivo studies
investigating the effects of methotrexate on endothelial function, arterial stiffness, and blood pressure, and the
potential mechanisms of action involved. The available evidence suggests that methotrexate might have beneficial
effects on vascular homeostasis and blood pressure control by targeting specific inflammatory pathways,
adenosine metabolism, and 5' adenosine monophosphate-activated protein kinase. Such effects might be
biologically and clinically relevant not only in patients with rheumatoid arthritis but also in older adults with high
cardiovascular risk. Therefore, methotrexate has the potential to be repurposed for cardiovascular risk
management in old age because of its putative pharmacological effects on inflammation, vascular homeostasis,
and blood pressure. However, further study and confirmation of these effects are essential in order to adequately
design intervention studies of methotrexate in the older population.
Collapse
Affiliation(s)
- Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Stefania Bassu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Matteo Piga
- Rheumatology Unit, University Clinic and AOU of Cagliari, Cagliari, Italy
| | - Gian L. Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
16
|
Mokotedi L, Millen AM, Mogane C, Gomes M, Woodiwiss AJ, Norton GR, Michel FS. Associations of inflammatory markers and vascular cell adhesion molecule-1 with endothelial dysfunction in collagen-induced arthritis. Eur J Pharmacol 2019; 865:172786. [DOI: 10.1016/j.ejphar.2019.172786] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 01/01/2023]
|
17
|
Hsu T, Nguyen-Tran HH, Trojanowska M. Active roles of dysfunctional vascular endothelium in fibrosis and cancer. J Biomed Sci 2019; 26:86. [PMID: 31656195 PMCID: PMC6816223 DOI: 10.1186/s12929-019-0580-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation is the underlying pathological condition that results in fibrotic diseases. More recently, many forms of cancer have also been linked to chronic tissue inflammation. While stromal immune cells and myofibroblasts have been recognized as major contributors of cytokines and growth factors that foster the formation of fibrotic tissue, the endothelium has traditionally been regarded as a passive player in the pathogenic process, or even as a barrier since it provides a physical divide between the circulating immune cells and the inflamed tissues. Recent findings, however, have indicated that endothelial cells in fact play a crucial role in the inflammatory response. Endothelial cells can be activated by cytokine signaling and express inflammatory markers, which can sustain or exacerbate the inflammatory process. For example, the activated endothelium can recruit and activate leukocytes, thus perpetuating tissue inflammation, while sustained stimulation of endothelial cells may lead to endothelial-to-mesenchymal transition that contributes to fibrosis. Since chronic inflammation has now been recognized as a significant contributing factor to tumorigenesis, it has also emerged that activation of endothelium also occurs in the tumor microenvironment. This review summarizes recent findings characterizing the molecular and cellular changes in the vascular endothelium that contribute to tissue fibrosis, and potentially to cancer formation.
Collapse
Affiliation(s)
- Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, 300 Jhongda Rd, Taoyuan City, Taiwan, Republic of China. .,Center for Chronic Disease Research, National Central University, 300 Jhongda Rd, Taoyuan City, Taiwan, Republic of China.
| | - Hieu-Huy Nguyen-Tran
- Department of Biomedical Sciences and Engineering, National Central University, 300 Jhongda Rd, Taoyuan City, Taiwan, Republic of China
| | - Maria Trojanowska
- Arthritis Center, Boston University School of Medicine, 75 E. Newton St. Evans Building, Boston, MA, 02118, USA
| |
Collapse
|
18
|
Reiss AB, Silverman A, Khalfan M, Vernice NA, Kasselman LJ, Carsons SE, De Leon J. Accelerated Atherosclerosis in Rheumatoid Arthritis: Mechanisms and Treatment. Curr Pharm Des 2019; 25:969-986. [DOI: 10.2174/1381612825666190430113212] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/14/2019] [Indexed: 12/11/2022]
Abstract
Background:Rheumatoid arthritis (RA) is a chronic systemic autoimmune inflammatory disorder that increases the risk of developing cardiovascular disease. There is accumulating evidence that the RA disease state accelerates the formation of atherosclerotic plaques. Treatments for RA improve joint symptomatology and may reduce inflammation, but consideration of their effects on the cardiovascular system is generally low priority.Objective:Since cardiovascular disease is the leading cause of mortality in RA patients, the impact of RA therapies on atherosclerosis is an area in need of attention and the focus of this review.Results:The drugs used to treat RA may be analgesics, conventional disease-modifying anti-rheumatic drugs, and/or biologics, including antibodies against the cytokine tumor necrosis factor-α. Pain relievers such as nonselective non-steroidal anti-inflammatory drugs and cyclooxygenase inhibitors may adversely affect lipid metabolism and cyclooxygenase inhibitors have been associated with increased adverse cardiovascular events, such as myocardial infarction and stroke. Methotrexate, the anchor disease-modifying anti-rheumatic drug in RA treatment has multiple atheroprotective advantages and is often combined with other therapies. Biologic inhibitors of tumor necrosis factor-α may be beneficial in preventing cardiovascular disease because tumor necrosis factor-α promotes the initiation and progression of atherosclerosis. However, some studies show a worsening of the lipid profile in RA with blockade of this cytokine, leading to higher total cholesterol and triglycerides.Conclusion:Greater understanding of the pharmacologic activity of RA treatments on the atherosclerotic process may lead to improved care, addressing both damages to the joints and heart.
Collapse
Affiliation(s)
- Allison B. Reiss
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| | - Andrew Silverman
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| | - Muhammed Khalfan
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| | - Nicholas A. Vernice
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| | - Lora J. Kasselman
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| | - Steven E. Carsons
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| | - Joshua De Leon
- Winthrop Research Institute, Department of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY 11501, United States
| |
Collapse
|
19
|
Chouk M, Bordy R, Moretto J, Wendling D, Totoson P, Demougeot C. Pristane-induced arthritis in dark Agouti rat is a relevant model for mimicking vascular dysfunction and lipid paradox in rheumatoid arthritis. Joint Bone Spine 2019; 86:483-490. [DOI: 10.1016/j.jbspin.2018.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/19/2018] [Accepted: 12/05/2018] [Indexed: 12/25/2022]
|
20
|
Liang X, Chen Y, Wu L, Maharjan A, Regmi B, Zhang J, Gui S. In situ hexagonal liquid crystal for intra-articular delivery of sinomenine hydrochloride. Biomed Pharmacother 2019; 117:108993. [PMID: 31228805 DOI: 10.1016/j.biopha.2019.108993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate the release behaviors of sinomenine hydrochloride loaded via in situ hexagonal liquid crystal (ISH), and its potential to improve the local bioavailability in knee joints of sinomenine hydrochloride (SMH) after intra-articular administration. The ISH was prepared by a liquid precursor mixture containing phytantriol (PT), Vitamin E acetate (VEA), ethanol (ET), and water. The in vitro release profiles revealed a sustained release of SMH from the optimized ISH formula (PT/VEA/ET/water, 60.8:3.2:16.0:20.0, w/w/w/w), which was selected for the in vivo pharmacokinetics and preliminary pharmacodynamics studies. In both healthy and adjuvant-induced arthritis (AA) rats, the SMH loaded ISH showed significantly smaller SMH AUC0-∞ in plasma (P < 0.01), and higher SMH concentration in synoviums (2˜168 h) than that of SMH solution, indicating that the ISH significantly reduced the leakage of SMH into systemic circulation. The t1/2α of SMH loaded ISH in the knee joints of AA rats, was longer (13.42 h) than that of healthy rats (1.34 h) (P < 0.05), most likely that in vivo drug release behavior of SMH loaded ISH was affected by the physiological environment of the joint. It was found that the SMH loaded ISH could benefit AA-rats by suppressing the level of IL-1β in comparison to SMH solutions. The results of the histopathology of knee joints in AA rats displayed that the SMH loaded ISH might be suitable for the development of treatment strategies for rheumatoid arthritis diseases.
Collapse
Affiliation(s)
- Xiao Liang
- Anhui University of Chinese Medicine, No.1 Qianjiang Street, Xinzhan Distinct, 230012, Hefei, Anhui, China
| | - Yulin Chen
- Anhui University of Chinese Medicine, No.1 Qianjiang Street, Xinzhan Distinct, 230012, Hefei, Anhui, China
| | - Li Wu
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Abi Maharjan
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | - Jiwen Zhang
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shuangying Gui
- Anhui University of Chinese Medicine, No.1 Qianjiang Street, Xinzhan Distinct, 230012, Hefei, Anhui, China.; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province.
| |
Collapse
|
21
|
Amigues I, Tugcu A, Russo C, Giles JT, Morgenstein R, Zartoshti A, Schulze C, Flores R, Bokhari S, Bathon JM. Myocardial Inflammation, Measured Using 18-Fluorodeoxyglucose Positron Emission Tomography With Computed Tomography, Is Associated With Disease Activity in Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71:496-506. [PMID: 30407745 DOI: 10.1002/art.40771] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To determine the prevalence and correlates of subclinical myocardial inflammation in patients with rheumatoid arthritis (RA). METHODS RA patients (n = 119) without known cardiovascular disease underwent cardiac 18-fluorodeoxyglucose (FDG) positron emission tomography with computed tomography (PET-CT). Myocardial FDG uptake was assessed visually and measured quantitatively as the standardized uptake value (SUV). Multivariable linear regression was used to assess the associations of patient characteristics with myocardial SUVs. A subset of RA patients who had to escalate their disease-modifying antirheumatic drug (DMARD) therapy (n = 8) underwent a second FDG PET-CT scan after 6 months, to assess treatment-associated changes in myocardial FDG uptake. RESULTS Visually assessed FDG uptake was observed in 46 (39%) of the 119 RA patients, and 21 patients (18%) had abnormal quantitatively assessed myocardial FDG uptake (i.e., mean of the mean SUV [SUVmean ] ≥3.10 units; defined as 2 SD above the value in a reference group of 27 non-RA subjects). The SUVmean was 31% higher in patients with a Clinical Disease Activity Index (CDAI) score of ≥10 (moderate-to-high disease activity) as compared with those with lower CDAI scores (low disease activity or remission) (P = 0.005), after adjustment for potential confounders. The adjusted SUVmean was 26% lower among those treated with a non-tumor necrosis factor-targeted biologic agent compared with those treated with conventional (nonbiologic) DMARDs (P = 0.029). In the longitudinal substudy, the myocardial SUVmean decreased from 4.50 units to 2.30 units over 6 months, which paralleled the decrease in the mean CDAI from a score of 23 to a score of 12. CONCLUSION Subclinical myocardial inflammation is frequent in patients with RA, is associated with RA disease activity, and may decrease with RA therapy. Future longitudinal studies will be required to assess whether reduction in myocardial inflammation will reduce heart failure risk in RA.
Collapse
Affiliation(s)
- Isabelle Amigues
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Aylin Tugcu
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Cesare Russo
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Jon T Giles
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Rachelle Morgenstein
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Afshin Zartoshti
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Christian Schulze
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Raul Flores
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Sabahat Bokhari
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| | - Joan M Bathon
- Columbia University College of Physicians and Surgeons, and New York Presbyterian Hospital, New York, New York
| |
Collapse
|
22
|
Plasma from Patients with Rheumatoid Arthritis Reduces Nitric Oxide Synthesis and Induces Reactive Oxygen Species in A Cell-Based Biosensor. BIOSENSORS-BASEL 2019; 9:bios9010032. [PMID: 30818887 PMCID: PMC6468433 DOI: 10.3390/bios9010032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) has been associated with a higher risk of developing cardiovascular (CV) diseases. It has been proposed that systemic inflammation plays a key role in premature atherosclerosis development, and is therefore crucial to determine whether systemic components from RA patients promotes endothelial cell-oxidative stress by affecting reactive oxygen species (ROS) and nitric-oxide (NO) production. The aim of this study was to evaluate whether plasma from RA patients impair NO synthesis and ROS production by using the cell-line ECV-304 as a biosensor. NO synthesis and ROS production were measured in cells incubated with plasma from 73 RA patients and 52 healthy volunteers by fluorimetry. In addition, traditional CV risk factors, inflammatory molecules and disease activity parameters were measured. Cells incubated with plasma from RA patients exhibited reduced NO synthesis and increased ROS production compared to healthy volunteers. Furthermore, the imbalance between NO synthesis and ROS generation in RA patients was not associated with traditional CV risk factors. Our data suggest that ECV-304 cells can be used as a biosensor of systemic inflammation-induced endothelial cell-oxidative stress. We propose that both NO and ROS production are potential biomarkers aimed at improving the current assessment of CV risk in RA.
Collapse
|
23
|
Verhoeven F, Prati C, Totoson P, Bordy R, Wendling D, Demougeot C. Structural efficacy of NSAIDs, COX-2 inhibitor and glucocorticoid compared with TNFα blocker: a study in adjuvant-induced arthritis rats. Rheumatology (Oxford) 2019; 58:1099-1103. [DOI: 10.1093/rheumatology/key444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/03/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Frank Verhoeven
- EA 4267 ≪PEPITE≫, UFR Santé, Franche-Comté University, 19 rue Ambroise Paré, bâtiment S 25030 BESANCON cedex, France
- 2Department of Rheumatology, CHRU de Besançon, 3 boulevard Fleming 25000 BESANCON, France
| | - Clément Prati
- EA 4267 ≪PEPITE≫, UFR Santé, Franche-Comté University, 19 rue Ambroise Paré, bâtiment S 25030 BESANCON cedex, France
- 2Department of Rheumatology, CHRU de Besançon, 3 boulevard Fleming 25000 BESANCON, France
| | - Perle Totoson
- EA 4267 ≪PEPITE≫, UFR Santé, Franche-Comté University, 19 rue Ambroise Paré, bâtiment S 25030 BESANCON cedex, France
| | - Romain Bordy
- EA 4267 ≪PEPITE≫, UFR Santé, Franche-Comté University, 19 rue Ambroise Paré, bâtiment S 25030 BESANCON cedex, France
| | - Daniel Wendling
- 2Department of Rheumatology, CHRU de Besançon, 3 boulevard Fleming 25000 BESANCON, France
- EA 4266 ≪EPILAB≫, UFR Santé, Franche-Comté University, 19 rue Ambroise Paré, bâtiment S 25030 BESANCON cedex, France
| | - Céline Demougeot
- EA 4267 ≪PEPITE≫, UFR Santé, Franche-Comté University, 19 rue Ambroise Paré, bâtiment S 25030 BESANCON cedex, France
| |
Collapse
|
24
|
Nasriati F, Hidayat R, Budiman B, Rinaldi I. Correlation Between Tumor Necrosis Factor-α Levels, Free Fatty Acid Levels, and Soluble Vascular Cell Adhesion Molecule-1 Levels in Rheumatoid Arthritis Patients. Open Rheumatol J 2018; 12:86-93. [PMID: 30123370 PMCID: PMC6062898 DOI: 10.2174/1874312901812010086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/02/2018] [Accepted: 06/05/2018] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The mortality of Rheumatoid Arthritis (RA) is quite high, which is largely due to cardiovascular complications caused by endothelial dysfunction. One of the important inflammatory mediators that contribute to RA joints arthritis of TNF-α, also proven to play a role in endothelial dysfunction and play a role in increasing intracellular lipolysis, thus increasing circulating FFA levels. OBJECTIVES To determine the correlation between TNF-α levels with VCAM-1 levels, correlation of TNF-α levels with FFA levels, and correlation of FFA levels with VCAM-1 levels. METHODS Cross sectional and retrospective design studies of adult RA patients treated at Cipto Mangunkusumo Hospital (RSCM), without metabolic disturbances, acute infection, cardiovascular disorders, or other autoimmune diseases. The cross-sectional data was collected from October to November 2017, while retrospective samples were collected since August 2016. TNF-α, VCAM-1, and FFA levels were assessed by serum blood test by ELISA method. Correlation analysis is done by Pearson analysis when the data distribution is normal and with Spearman analysis when the data distribution is not normal. RESULTS A total of 35 subjects were enrolled in the study. Most (97.1%) were women with an average age of 45.29 years, median disease duration of 48 months, and most had moderate disease activity (65.7%). No significant correlation was found between TNF-α levels and VCAM-1 levels (p = 0.677; r = +0.073). as well betwen TNF-α levels and FFA levels (p = 0.227; r = -0.21). The correlation between FFA and VCAM-1 levels showed significant correlation with negative correlation and weak correlation (p = 0.036; r = -0.355). CONCLUSIONS (1) There was no correlation between TNF-α levels and VCAM-1 levels in RA patients; (2) There was no correlation between TNF-α levels and FFA levels in RA patients; (3) There was a negative correlation between FFA levels and VCAM-1 levels in RA patients.
Collapse
Affiliation(s)
- Fazria Nasriati
- Department of Internal Medicine, Faculty of Medicine Universitas Indonesia -s Cipto Mangunkusumo General Hospital, Jalan Diponegoro 71,Jakarta 10310,Indonesia
| | - Rudy Hidayat
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jalan Diponegoro 71,Jakarta,Indonesia
| | - Budiman Budiman
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jalan Diponegoro 71,Jakarta,Indonesia
| | - Ikhwan Rinaldi
- Clinical Epidemiology Unit, Department of Internal Medicine, Universitas Indonesia - Cipto Mangunkusumo General Hospital, Jalan Diponegoro 71,Jakarta,Indonesia
| |
Collapse
|
25
|
England BR, Thiele GM, Anderson DR, Mikuls TR. Increased cardiovascular risk in rheumatoid arthritis: mechanisms and implications. BMJ 2018; 361:k1036. [PMID: 29685876 PMCID: PMC6889899 DOI: 10.1136/bmj.k1036] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis is a systemic autoimmune disease characterized by excess morbidity and mortality from cardiovascular disease. Mechanisms linking rheumatoid arthritis and cardiovascular disease include shared inflammatory mediators, post-translational modifications of peptides/proteins and subsequent immune responses, alterations in the composition and function of lipoproteins, increased oxidative stress, and endothelial dysfunction. Despite a growing understanding of these mechanisms and their complex interplay with conventional cardiovascular risk factors, optimal approaches of risk stratification, prevention, and treatment in the context of rheumatoid arthritis remain unknown. A multifaceted approach to reduce the burden posed by cardiovascular disease requires optimal management of traditional risk factors in addition to those intrinsic to rheumatoid arthritis such as increased disease activity. Treatments for rheumatoid arthritis seem to exert differential effects on cardiovascular risk as well as the mechanisms linking these conditions. More research is needed to establish whether preferential rheumatoid arthritis therapies exist in terms of prevention of cardiovascular disease. Ultimately, understanding the unique mechanisms for cardiovascular disease in rheumatoid arthritis will aid in risk stratification and the identification of novel targets for meaningful reduction of cardiovascular risk in this patient population.
Collapse
Affiliation(s)
- Bryant R England
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Daniel R Anderson
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ted R Mikuls
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
26
|
Disease severity impacts the relationship of apelin with arterial function in patients with rheumatoid arthritis. Clin Rheumatol 2018; 37:1481-1491. [DOI: 10.1007/s10067-018-4013-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/26/2022]
|
27
|
Verhoeven F, Totoson P, Marie C, Prigent-Tessier A, Wendling D, Tournier-Nappey M, Prati C, Demougeot C. Diclofenac but not celecoxib improves endothelial function in rheumatoid arthritis: A study in adjuvant-induced arthritis. Atherosclerosis 2017; 266:136-144. [PMID: 29024866 DOI: 10.1016/j.atherosclerosis.2017.09.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/03/2017] [Accepted: 09/28/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND AIMS We aimed at investigating the effect of celecoxib (COX-2 selective inhibitor) and diclofenac (non-selective COX inhibitor) on endothelial function, and at identifying the underlying mechanisms in adjuvant-induced arthritis (AIA). METHODS At the first signs of AIA, diclofenac (5 mg/kg twice a day, i.p), celecoxib (3 mg/kg/day, i.p) or saline (Vehicle) was administered for 3 weeks. Endothelial function was studied in aortic rings relaxed with acetylcholine (Ach) with or without inhibitors of NOS, arginase, EDHF and superoxide anions (O2-°) production. Aortic expression of eNOS, Ser1177-phospho-eNOS, COX-2, arginase-2, p22phox and p47phox was evaluated by Western blotting analysis. Arthritis scores, blood pressure, glycaemia and serum ADMA levels were measured. RESULTS Diclofenac and celecoxib significantly reduced arthritis score to the same extent (p<0.05). As compared to vehicle-treated AIA, celecoxib did not change whereas diclofenac improved endothelial function (p<0.05) through increased EDHF production, decreased arginase activity and expression, decreased superoxide anions production and expression of p22phox and p47phox. Diclofenac but not celecoxib significantly enhanced blood pressure and serum ADMA levels. Glycaemia was unchanged by both treatments. CONCLUSIONS Our study reveals that the effect of NSAIDs on endothelial function cannot be extrapolated from their impact on arthritis severity and suggest that changes in blood pressure and plasma ADMA levels may not be useful to predict CV risk of NSAIDs in RA.
Collapse
Affiliation(s)
- Frank Verhoeven
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France; Service de Rhumatologie, CHRU Besançon, 25000 Besançon, France
| | - Perle Totoson
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France
| | - Christine Marie
- INSERM U1093, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | | | - Daniel Wendling
- Service de Rhumatologie, CHRU Besançon, 25000 Besançon, France; EA4266, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France
| | - Maude Tournier-Nappey
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France
| | - Clément Prati
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France; Service de Rhumatologie, CHRU Besançon, 25000 Besançon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France.
| |
Collapse
|
28
|
Arraki K, Totoson P, Decendit A, Badoc A, Zedet A, Jolibois J, Pudlo M, Demougeot C, Girard-Thernier C. Cyperaceae Species Are Potential Sources of Natural Mammalian Arginase Inhibitors with Positive Effects on Vascular Function. JOURNAL OF NATURAL PRODUCTS 2017; 80:2432-2438. [PMID: 28837342 DOI: 10.1021/acs.jnatprod.7b00197] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The inhibition of arginase is of substantial interest for the treatment of various diseases of public health interest including cardiovascular diseases. Using an ex vivo experiment on rat aortic rings and an in vitro assay with liver bovine purified arginase, it was demonstrated that several polyphenolic extracts from Cyperus and Carex species possess vasorelaxant properties and mammalian arginase inhibitory capacities. Phytochemical studies performed on these species led to the identification of eight compounds, including monomers, dimers, trimers, and tetramers of resveratrol. The potential of these stilbenes as inhibitors of mammalian arginase was assessed. Five compounds, scirpusin B (5), ε-viniferin (4), cyperusphenol B (6), carexinol A (7), and the new compound virgatanol (1), showed significant inhibition of arginase, with percentage inhibition ranging from 70% to 95% at 100 μg/mL and IC50 values between 12.2 and 182.1 μM, confirming that these stilbenes may be useful for the development of new pharmaceutical products.
Collapse
Affiliation(s)
- Kamel Arraki
- PEPITE EA4267, University of Bourgogne Franche-Comté , 25000 Besançon, France
| | - Perle Totoson
- PEPITE EA4267, University of Bourgogne Franche-Comté , 25000 Besançon, France
| | - Alain Decendit
- MIB-UR Œnologie, EA 4577, USC 1366 INRA, University of Bordeaux, ISVV , 33882 Villenave-d'Ornon, France
| | - Alain Badoc
- MIB-UR Œnologie, EA 4577, USC 1366 INRA, University of Bordeaux, ISVV , 33882 Villenave-d'Ornon, France
| | - Andy Zedet
- PEPITE EA4267, University of Bourgogne Franche-Comté , 25000 Besançon, France
| | - Julia Jolibois
- PEPITE EA4267, University of Bourgogne Franche-Comté , 25000 Besançon, France
| | - Marc Pudlo
- PEPITE EA4267, University of Bourgogne Franche-Comté , 25000 Besançon, France
| | - Céline Demougeot
- PEPITE EA4267, University of Bourgogne Franche-Comté , 25000 Besançon, France
| | | |
Collapse
|
29
|
Pirro M, Bianconi V, Paciullo F, Mannarino MR, Bagaglia F, Sahebkar A. Lipoprotein(a) and inflammation: A dangerous duet leading to endothelial loss of integrity. Pharmacol Res 2017; 119:178-187. [DOI: 10.1016/j.phrs.2017.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/08/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
|
30
|
Verhoeven F, Totoson P, Maguin-Gaté K, Prigent-Tessier A, Marie C, Wendling D, Moretto J, Prati C, Demougeot C. Glucocorticoids improve endothelial function in rheumatoid arthritis: a study in rats with adjuvant-induced arthritis. Clin Exp Immunol 2017; 188:208-218. [PMID: 28152574 DOI: 10.1111/cei.12938] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2017] [Indexed: 12/15/2022] Open
Abstract
To determine the effect of glucocorticoids (GCs) on endothelial dysfunction (ED) and on traditional cardiovascular (CV) risk factors in the adjuvant-induced arthritis (AIA) rat model. At the first signs of AIA, a high dose (HD) [10 mg/kg/day, intraperitoneally (i.p.), GC-HD] or low dose (LD) (1 mg/kg/day, i.p., GC-LD) of prednisolone was administered for 3 weeks. Endothelial function was studied in aortic rings relaxed with acetylcholine (Ach) with or without inhibitors of nitric oxide synthase (NOS), cyclooxygenase 2 (COX-2), arginase, endothelium derived hyperpolarizing factor (EDHF) and superoxide anions ( O2-°) production. Aortic expression of endothelial NOS (eNOS), Ser1177-phospho-eNOS, COX-2, arginase-2, p22phox and p47phox was evaluated by Western blotting analysis. Arthritis scores, blood pressure, heart rate and blood levels of cytokines, triglycerides, cholesterol and glucose were measured. GC-HD but not GC-LD reduced arthritis score significantly and improved Ach-induced relaxation (P < 0·05). The positive effect of GC-HD resulted from increased NOS activity and EDHF production and decreased COX-2/arginase activities and O2-° production. These functional effects relied upon increased phospho-eNOS expression and decreased COX-2, arginase-2 and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression. Despite the lack of effect of GC-LD on ED, it increased NOS and EDHF and down-regulated O2-° pathways but did not change arginase and COX-2 pathways. GC-HD increased triglycerides levels and blood pressure significantly (P < 0·05). Both doses of GCs decreased to the same extent as plasma interleukin (IL)-1β and tumour necrosis factor (TNF)-α levels (P < 0·05). Our data demonstrated that subchronic treatment with prednisolone improved endothelial function in AIA via pleiotropic effects on endothelial pathways. These effects occurred independently of the deleterious cardiometabolic effects and the impact of prednisolone on systemic inflammation.
Collapse
Affiliation(s)
- F Verhoeven
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France.,Service de Rhumatologie, CHRU Besançon, France
| | - P Totoson
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - K Maguin-Gaté
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | | | - C Marie
- INSERM U1093, Université Bourgogne Franche-Comté, Dijon, France
| | - D Wendling
- Service de Rhumatologie, CHRU Besançon, France.,EA 4266, Université Bourgogne Franche-Comté, Besançon, France
| | - J Moretto
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - C Prati
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France.,Service de Rhumatologie, CHRU Besançon, France
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
31
|
Moretto J, Guglielmetti AS, Tournier-Nappey M, Martin H, Prigent-Tessier A, Marie C, Demougeot C. Effects of a chronic l-arginine supplementation on the arginase pathway in aged rats. Exp Gerontol 2017; 90:52-60. [PMID: 28132871 DOI: 10.1016/j.exger.2017.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 02/05/2023]
Abstract
While ageing is frequently associated with l-arginine deficiency, clinical and experimental studies provided controversial data on the interest of a chronic l-arginine supplementation with beneficial, no or even deleterious effects. It was hypothesized that these discrepancies might relate to a deviation of l-arginine metabolism towards production of l-ornithine rather than nitric oxide as a result of age-induced increase in arginase activity. This study investigated the effect of ageing on arginase activity/expression in target tissues and determined whether l-arginine supplementation modulated the effect of ageing on arginase activity. Arginase activity and expression were measured in the heart, vessel, brain, lung, kidney and liver in young rats (3-months old) and aged Wistar rats (22-24-months-old) with or without l-arginine supplementation (2.25% in drinking water for 6weeks). Plasma levels of l-arginine and l-ornithine were quantified in order to calculate the plasma l-arginine/l-ornithine ratio, considered as a reflection of arginase activity. Cardiovascular parameters (blood pressure, heart rate) and aortic vascular reactivity were also studied. Ageing dramatically reduced plasma l-arginine and l-arginine/l-ornithine ratio, decreased liver and kidney arginase activities but did not change activities in other tissues. l-Arginine supplementation normalized plasma l-arginine and l-arginine/l-ornithine ratio, improved endothelial function and decreased systolic blood pressure. These effects were associated with decreased arginase activity in aorta along with no change in the other tissues except in the lung in which activity was increased. A strong mismatch was therefore observed between arginase activity and expression in analyzed tissues. The present study reveals that ageing selectively changes arginase activity in clearance tissues, but does not support a role of the arginase pathway in the potential deleterious effect of the l-arginine supplementation in aged patients. Moreover, our data argue against the use of the measurement of plasma l-arginine/l-ornithine ratio to estimate arginase activity in aged patients.
Collapse
Affiliation(s)
- Johnny Moretto
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | | | - Maude Tournier-Nappey
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Hélène Martin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France
| | | | - Christine Marie
- INSERM U1093, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| |
Collapse
|
32
|
Ikić Matijašević M, Flegar D, Kovačić N, Katavić V, Kelava T, Šućur A, Ivčević S, Cvija H, Lazić Mosler E, Kalajzić I, Marušić A, Grčević D. Increased chemotaxis and activity of circulatory myeloid progenitor cells may contribute to enhanced osteoclastogenesis and bone loss in the C57BL/6 mouse model of collagen-induced arthritis. Clin Exp Immunol 2016; 186:321-335. [PMID: 27612450 DOI: 10.1111/cei.12862] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2016] [Indexed: 01/01/2023] Open
Abstract
Our study aimed to determine the functional activity of different osteoclast progenitor (OCP) subpopulations and signals important for their migration to bone lesions, causing local and systemic bone resorption during the course of collagen-induced arthritis in C57BL/6 mice. Arthritis was induced with chicken type II collagen (CII), and assessed by clinical scoring and detection of anti-CII antibodies. We observed decreased trabecular bone volume of axial and appendicular skeleton by histomorphometry and micro-computed tomography as well as decreased bone formation and increased bone resorption rate in arthritic mice in vivo. In the affected joints, bone loss was accompanied with severe osteitis and bone marrow hypercellularity, coinciding with the areas of active osteoclasts and bone erosions. Flow cytometry analysis showed increased frequency of putative OCP cells (CD3- B220- NK1.1- CD11b-/lo CD117+ CD115+ for bone marrow and CD3- B220- NK1.1- CD11b+ CD115+ Gr-1+ for peripheral haematopoietic tissues), which exhibited enhanced differentiation potential in vitro. Moreover, the total CD11b+ population was expanded in arthritic mice as well as CD11b+ F4/80+ macrophage, CD11b+ NK1.1+ natural killer cell and CD11b+ CD11c+ myeloid dendritic cell populations in both bone marrow and peripheral blood. In addition, arthritic mice had increased expression of tumour necrosis factor-α, interleukin-6, CC chemokine ligand-2 (Ccl2) and Ccl5, with increased migration and differentiation of circulatory OCPs in response to CCL2 and, particularly, CCL5 signals. Our study characterized the frequency and functional properties of OCPs under inflammatory conditions associated with arthritis, which may help to clarify crucial molecular signals provided by immune cells to mediate systemically enhanced osteoresorption.
Collapse
Affiliation(s)
- M Ikić Matijašević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - D Flegar
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - N Kovačić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - V Katavić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - T Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - A Šućur
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - S Ivčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - H Cvija
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - E Lazić Mosler
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - I Kalajzić
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - A Marušić
- Department of Research in Biomedicine and Health, University of Split School of Medicine, Split, Croatia
| | - D Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
33
|
Correction: Endothelial Dysfunction in Rheumatoid Arthritis: Mechanistic Insights and Correlation with Circulating Markers of Systemic Inflammation. PLoS One 2016; 11:e0150874. [PMID: 26930507 PMCID: PMC4773246 DOI: 10.1371/journal.pone.0150874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
|