151
|
Tong DC, Quinn S, Nasis A, Hiew C, Roberts-Thomson P, Adams H, Sriamareswaran R, Htun NM, Wilson W, Stub D, van Gaal W, Howes L, Collins N, Yong A, Bhindi R, Whitbourn R, Lee A, Hengel C, Asrress K, Freeman M, Amerena J, Wilson A, Layland J. Colchicine in Patients With Acute Coronary Syndrome. Circulation 2020; 142:1890-1900. [DOI: 10.1161/circulationaha.120.050771] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background:
Inflammation plays a crucial role in clinical manifestations and complications of acute coronary syndromes (ACS). Colchicine, a commonly used treatment for gout, has recently emerged as a novel therapeutic option in cardiovascular medicine owing to its anti-inflammatory properties. We sought to determine the potential usefulness of colchicine treatment in patients with ACS.
Methods:
This was a multicenter, randomized, double-blind, placebo-controlled trial involving 17 hospitals in Australia that provide acute cardiac care service. Eligible participants were adults (18–85 years) who presented with ACS and had evidence of coronary artery disease on coronary angiography managed with either percutaneous coronary intervention or medical therapy. Patients were assigned to receive either colchicine (0.5 mg twice daily for the first month, then 0.5 mg daily for 11 months) or placebo, in addition to standard secondary prevention pharmacotherapy, and were followed up for a minimum of 12 months. The primary outcome was a composite of all-cause mortality, ACS, ischemia-driven (unplanned) urgent revascularization, and noncardioembolic ischemic stroke in a time to event analysis.
Results:
A total of 795 patients were recruited between December 2015 and September 2018 (mean age, 59.8±10.3 years; 21% female), with 396 assigned to the colchicine group and 399 to the placebo group. Over the 12-month follow-up, there were 24 events in the colchicine group compared with 38 events in the placebo group (
P
=0.09, log-rank). There was a higher rate of total death (8 versus 1;
P
=0.017, log-rank) and, in particular, noncardiovascular death in the colchicine group (5 versus 0;
P
=0.024, log-rank). The rates of reported adverse effects were not different (colchicine 23.0% versus placebo 24.3%), and they were predominantly gastrointestinal symptoms (colchicine, 23.0% versus placebo, 20.8%).
Conclusions:
The addition of colchicine to standard medical therapy did not significantly affect cardiovascular outcomes at 12 months in patients with ACS and was associated with a higher rate of mortality.
Registration:
URL:
https://www.anzctr.org.au
; Unique identifier: ACTRN12615000861550.
Collapse
Affiliation(s)
- David C. Tong
- St Vincent’s Hospital Melbourne, Fitzroy, Victoria, Australia (D.C.T., R.W., A.W., J.L.)
- Cardiology, Department of Medicine, Peninsula Health, Frankston, Victoria, Australia (D.C.T., R.S., N.M.H., J.L.)
| | - Stephen Quinn
- Swinburne University of Technology, Department of Health Science and Biostatistics, Hawthorn, Victoria, Australia (S.Q.)
| | - Arthur Nasis
- MonashHeart, Monash Health, Clayton, Victoria, Australia (A.N.)
| | - Chin Hiew
- Barwon Health, University Hospital Geelong, Victoria, Australia (C.H., J.A.)
| | | | - Heath Adams
- Royal Hobart Hospital, Tasmania, Australia (P.R.-T., H.A.)
| | - Rumes Sriamareswaran
- Cardiology, Department of Medicine, Peninsula Health, Frankston, Victoria, Australia (D.C.T., R.S., N.M.H., J.L.)
| | - Nay M. Htun
- Cardiology, Department of Medicine, Peninsula Health, Frankston, Victoria, Australia (D.C.T., R.S., N.M.H., J.L.)
| | - William Wilson
- Royal Melbourne Hospital, Parkville, Victoria, Australia (W.W.)
| | - Dion Stub
- Western Health, St Albans, Victoria, Australia (D.S.)
| | | | - Laurie Howes
- Gold Coast University Hospital, Southport, Queensland, Australia (L.H.)
| | - Nicholas Collins
- John Hunter Hospital, New Lambton Heights, New South Wales, Australia (N.C.)
| | - Andy Yong
- Concord Repatriation General Hospital, New South Wales, Australia (A.Y.)
| | - Ravinay Bhindi
- Royal North Shore Hospital, St Leonards, New South Wales, Australia (R.B.)
| | - Robert Whitbourn
- St Vincent’s Hospital Melbourne, Fitzroy, Victoria, Australia (D.C.T., R.W., A.W., J.L.)
| | - Astin Lee
- Wollongong Hospital, New South Wales, Australia (A.L.)
| | - Chris Hengel
- Ballarat Health Services, Victoria, Australia (C.H.)
| | - Kaleab Asrress
- Bankstown-Lidcombe Hospital, Bankstown, New South Wales, Australia (K.A.)
| | | | - John Amerena
- Barwon Health, University Hospital Geelong, Victoria, Australia (C.H., J.A.)
| | - Andrew Wilson
- St Vincent’s Hospital Melbourne, Fitzroy, Victoria, Australia (D.C.T., R.W., A.W., J.L.)
| | - Jamie Layland
- St Vincent’s Hospital Melbourne, Fitzroy, Victoria, Australia (D.C.T., R.W., A.W., J.L.)
- Cardiology, Department of Medicine, Peninsula Health, Frankston, Victoria, Australia (D.C.T., R.S., N.M.H., J.L.)
- Peninsula Clinical School, Central Clinical School, Monash University, Victoria, Australia (J.L.)
| |
Collapse
|
152
|
Feuillet V, Canard B, Trautmann A. Combining Antivirals and Immunomodulators to Fight COVID-19. Trends Immunol 2020; 42:31-44. [PMID: 33281063 PMCID: PMC7664349 DOI: 10.1016/j.it.2020.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
The majority of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected individuals remain paucisymptomatic, contrasting with a minority of infected individuals in danger of death. Here, we speculate that the robust disease resistance of most individuals is due to a swift production of type I interferon (IFNα/β), presumably sufficient to lower the viremia. A minority of infected individuals with a preexisting chronic inflammatory state fail to mount this early efficient response, leading to a delayed harmful inflammatory response. To improve the epidemiological scenario, we propose combining: (i) the development of efficient antivirals administered early enough to assist in the production of endogenous IFNα/β; (ii) potentiating early IFN responses; (iii) administering anti-inflammatory treatments when needed, but not too early to interfere with endogenous antiviral responses. Although the coronavirus disease 2019 (COVID-19) pandemic is exceptional, lessons may be learned from previous outbreaks (coronavirus, dengue, influenza viruses), especially when considering drug design and cytokine storms. We propose that efficient treatments for COVID-19 patients should combine antivirals and immunomodulators. This combination and, especially the use of immunomodulators, might be adapted according to the disease stage. Among the repurposed antiviral drugs currently being tested against COVID-19, none shows high potency. We posit that the innate type 1 interferon (IFNα/β)-dependent antiviral immune response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection should be amplified. To this end, we propose two putative approaches: the inhibition of transforming growth factor (TGFβ) signaling, and perhaps, the administration of 1,8-cineole. We suggest that an early diagnosis during COVID-19 is essential when aiming to purposely combine antivirals with the use of an immunomodulator (e.g., a drug to potentiate IFNα/β), ideally early in the disease course to lower the risk of cytokine storm manifestation. When the disease becomes severe, the new combination should prioritize targeting of the cytokine storm.
Collapse
Affiliation(s)
- Vincent Feuillet
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| | - Bruno Canard
- CNRS UMR 7257, Aix-Marseille University, Marseille, France
| | - Alain Trautmann
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| |
Collapse
|
153
|
Yu Y, Sun H, Zhu L, Ji L, Liu H. Downregulating lncRNA PRNCR1 ameliorates LPS-induced pulmonary vascular endothelial cell injury by modulating miR-330-5p/TLR4 axis. J Biochem Mol Toxicol 2020; 35:e22644. [PMID: 33049095 DOI: 10.1002/jbt.22644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/08/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary vascular endothelial cell (PVEC) injury following acute lung injury or acute respiratory distress syndrome seriously affects disease development. Recently, accumulating evidence has suggested that long noncoding RNA (lncRNA) exerts significant effects in vascular endothelial cell injury. However, PRNCR1, a novel lncRNA, remains scarcely understood in terms of its functions in PVEC injury. Both in vivo and in vitro models of PVEC injury were constructed by lipopolysaccharide (LPS) administration. The relative expressions of PRNCR1, miR-330-5p, and TLR4 were detected by quantitative reverse transcription-polymerase chain reaction, Western blot, and immunohistochemistry. Besides, gain and loss assays of PRNCR1/miR-330-5p were conducted to verify their effects on LPS-induced PVEC injury. Cell Counting Kit-8 assay used to measure cell viability and flow cytometry was used to detect apoptosis. Besides, the protein levels of caspase 3, nuclear factor-κB (NF-κB), and inflammatory cytokines (including tumor necrosis factor-α, interleukin-1β [IL-1β], and IL-6) were evaluated via Western blot and enzyme-linked immunosorbent assay. Moreover, a dual-luciferase activity experiment and RNA immunoprecipitation were applied to confirm the targeting relationship between PRNCR1 and miR-330-5p, miR-330-5p, and TLR4. PRNCR1 and TLR4 levels were significantly upregulated in LPS-treated PVEC, both in vivo and in vitro, while miR-330-5p were downregulated. Inhibiting PRNCR1 or overexpressing miR-330-5p markedly attenuated LPS-induced PVEC injury, expressions of TLR4, NF-κB, and inflammatory cytokines. Mechanistically, PRNCR1 functioned as a competitive endogenous RNA by sponging miR-330-5p and then promoting TLR4 expression. PRNCR1 was upregulated in LPS-induced PVEC and aggravated its injury via modulating the miR-330-5p/TLR4 axis.
Collapse
Affiliation(s)
- Yingqing Yu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongzhi Sun
- Department of Intensive Medicine, Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Zhu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Lianfeng Ji
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Haibo Liu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
154
|
Mareev VY, Orlova YA, Pavlikova EP, Akopyan ZA, Matskeplishvili ST, Plisyk AG, Seredenina EM, Potapenko AV, Malakhov PS, Samokhodskaya LM, Mershina ЕА, Sinitsyn VE, Asratyan DA, Zhdanova EA, Mareev YV, Begrambekova YL, Shatochina EA, Kamalov АА. [Proactive anti-inflammatory and anticoagulant therapy in the treatment of advanced stages of novel coronavirus infection (COVID-19). Case Series and Study Design: COLchicine versus ruxolitinib and secukinumab in open prospective randomIzed trial (COLORIT)]. ACTA ACUST UNITED AC 2020; 60:4-21. [PMID: 33131470 DOI: 10.18087/cardio.2020.9.n1338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 01/08/2023]
Abstract
The article is devoted to the treatment of the new coronavirus infection (COVID-19) in the advanced stages of the disease. The types of response of the immune system to the viral load of SARS-CoV-2 with the start of the inflammation process are considered. The situation is analyzed in detail in which the growing autoimmune inflammation (up to the development of a "cytokine storm") affects not only the pulmonary parenchyma, but also the endothelium of the small vessels of the lungs. Simultaneous damage to the alveoli and microthrombosis of the pulmonary vessels are accompanied by a progressive impairment of gas exchange, the development of acute respiratory distress syndrome, the treatment of which, even with the use of invasive ventilation, is ineffective and does not really change the prognosis of patients with COVID-19. In order to interrupt the pathological process at the earliest stages of the disease, the necessity of proactive anti-inflammatory therapy in combination with active anticoagulation treatment is substantiated. The results of the first randomized studies on the use of inhibitors of pro-inflammatory cytokines and chemokines (interleukin-6 (tocilizumab), interleukin-17 (secukinumab), Janus kinase blockers, through which the signal is transmitted to cells (ruxolitinib)), which have potential in the early treatment of COVID- 19. The use of a well-known anti-inflammatory drug colchicine (which is used for gout treatment) in patients with COVID-19 is considered. The design of the original COLORIT comparative study on the use of colchicine, ruxolitinib and secukinumab in the treatment of COVID-19 is presented. Clinical series presented, illustrated early anti-inflammatory therapy together with anticoagulants in patients with COVID-19 and the dangers associated with refusing to initiate such therapy on time.
Collapse
Affiliation(s)
- V Yu Mareev
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - Ya A Orlova
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - E P Pavlikova
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - Z A Akopyan
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - S T Matskeplishvili
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia
| | - A G Plisyk
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - E M Seredenina
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - A V Potapenko
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - P S Malakhov
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia
| | - L M Samokhodskaya
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - Е А Mershina
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - V E Sinitsyn
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - D A Asratyan
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia
| | - E A Zhdanova
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - Yu V Mareev
- National Medical Research Centre for Therapy and Preventive Medicine, Moscow, Russia Robertson Centre for Biostatistics, Glasgow, Great Britain
| | - Yu L Begrambekova
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| | - E A Shatochina
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia
| | - А А Kamalov
- Medical Research and Educational Center of the M. V. Lomonosov Moscow State University, Moscow, Russia Faculty of Fundamental Medicine, Lomonosov Moscow State University, Russia
| |
Collapse
|
155
|
Jin J, Xie Y, Shi C, Ma J, Wang Y, Qiao L, Li K, Sun T. Lipoxin A4 Inhibits NLRP3 Inflammasome Activation in Rats With Non-compressive Disc Herniation Through the JNK1/Beclin-1/PI3KC3 Pathway. Front Neurosci 2020; 14:799. [PMID: 33071721 PMCID: PMC7539067 DOI: 10.3389/fnins.2020.00799] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/07/2020] [Indexed: 01/05/2023] Open
Abstract
Background Non-compressive disc herniation is induced by an inflammatory response from the nucleus pulposus tissue and nerve roots. Lipoxins (LXs) are important endogenous anti-inflammatory mediators in the body, helping to inhibit neutrophil recruitment and stimulate autophagy in monocytes and macrophages. Here, we investigated the molecular mechanisms underlying the effects of exogenous lipoxin administration on rats with non-compressive disc herniation. Method A non-compressive disc herniation model was established in rats. Fifty rats were randomly divided into: sham group, model group, PI3K inhibitor (LY294002) group, lipoxin A4 group (LXA4), and PI3K inhibitor and lipoxin A4 group (LY294002 + LXA4). Similar groupings were established for rat spinal neurons. Changes in the mechanical pain threshold and thermal pain threshold were monitored at different times. The expression of proinflammatory and anti-inflammatory mediators was assessed by ELISA, while immunohistochemistry was employed to measure the expression levels of NLRP3 and p-JNK1. The expression levels of autophagy-related proteins were measured by western blot. Results In vivo, the pain threshold was markedly decreased in the model group at each time point examined compared with that in sham group. LY294002 treatment further reduced the pain threshold. After LXA4 injection, the pain threshold was significantly increased, and the effect of LY294002 was significantly weakened (p < 0.05). The levels of proinflammatory cytokines were increased in rats with non-compressive disc herniation, and these levels were further increased by LY294002 treatment (p < 0.05). However, treatment with LXA4 significantly reduced the levels of these proinflammatory cytokines in the model group (p < 0.05). The opposite effect was observed for anti-inflammatory mediators. The expression of NLRP3 was largely increased in the model group compared with that in the sham group (p < 0.05). Treatment with LY294002 also increased the NLRP3 expression level, while the administration of LXA4 elicited the opposite effect. Furthermore, western blot analysis showed that the expression of autophagy-related proteins was greatly decreased in the model group, whereas it was significantly increased in the LXA4 group (p < 0.05). The in vitro results were consistent with the outcomes observed in vivo. Conclusions These data suggested that LXA4 inhibited NLRP3 activation in rats with non-compressive disc herniation by regulating the JNK1/beclin-1/PI3KC3 pathway.
Collapse
Affiliation(s)
- Jin Jin
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yonggang Xie
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Cunxian Shi
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jiahai Ma
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yihao Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao, China
| | - Leyan Qiao
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kezhong Li
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Tao Sun
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
156
|
Misra DP, Gasparyan AY, Zimba O. Benefits and adverse effects of hydroxychloroquine, methotrexate and colchicine: searching for repurposable drug candidates. Rheumatol Int 2020; 40:1741-1751. [PMID: 32880032 PMCID: PMC7467139 DOI: 10.1007/s00296-020-04694-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022]
Abstract
Repurposing of antirheumatic drugs has garnered global attention. The aim of this article is to overview available evidence on the use of widely used antirheumatic drugs hydroxychloroquine, methotrexate and colchicine for additional indications. Hydroxychloroquine has endothelial stabilizing and anti-thrombotic effects. Its use has been explored as an adjunctive therapy in refractory thrombosis in antiphospholipid syndrome. It may also prevent recurrent pregnancy losses in the absence of antiphospholipid antibodies. Hydroxychloroquine favourably modulates atherogenic lipid and glycaemic profiles. Methotrexate has been tried for modulation of cardiovascular events in non-rheumatic clinical conditions, although a large clinical trial failed to demonstrate a benefit. Colchicine has been shown to successfully reduce the risk of recurrent cardiovascular events in a large multicentric trial. Potential antifibrotic effects of colchicine require further exploration. Hydroxychloroquine, methotrexate and colchicine are also being tried at different stages of the ongoing Coronavirus Disease 19 (COVID-19) pandemic for prophylaxis and treatment. While the use of these agents is being diversified, their adverse effects should be timely diagnosed and prevented. Hydroxychloroquine can cause retinopathy and rarely cardiac and auditory toxicity, retinopathy being dose and time dependent. Methotrexate can cause transaminitis, cytopenias and renal failure, particularly in acute overdoses. Colchicine can rarely cause myopathies, cardiomyopathy, cytopenias and transaminitis. Strong evidence is warranted to keep balance between benefits of repurposing these old antirheumatic drugs and risk of their adverse effects.
Collapse
Affiliation(s)
- Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Rae Bareli Road, Lucknow, 226014 India
| | - Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands UK
| | - Olena Zimba
- Department of Internal Medicine #2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
157
|
Opstal TSJ, Hoogeveen RM, Fiolet ATL, Silvis MJM, The SHK, Bax WA, de Kleijn DPV, Mosterd A, Stroes ESG, Cornel JH. Colchicine Attenuates Inflammation Beyond the Inflammasome in Chronic Coronary Artery Disease: A LoDoCo2 Proteomic Substudy. Circulation 2020; 142:1996-1998. [PMID: 32864998 DOI: 10.1161/circulationaha.120.050560] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Tjerk S J Opstal
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands (T.S.J.O., J.H.C.).,Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands (T.S.J.O., J.H.C.)
| | - Renate M Hoogeveen
- Department of Vascular Medicine, Amsterdam University Medical Centers, the Netherlands (R.M.H., E.S.G.S.)
| | - Aernoud T L Fiolet
- Departments of Cardiology (A.T.L.F., M.J.M.S., D.P.V.d.K.), University Medical Center Utrecht, the Netherlands.,Dutch Network for Cardiovascular Research, Utrecht, the Netherlands (A.T.L.F., A.M., J.H.C.)
| | - Max J M Silvis
- Departments of Cardiology (A.T.L.F., M.J.M.S., D.P.V.d.K.), University Medical Center Utrecht, the Netherlands
| | - Salem H K The
- Department of Cardiology, Treant Medical Center, Emmen, the Netherlands (S.H.K.T.)
| | - Willem A Bax
- Department of Internal Medicine, Northwest Clinics, Alkmaar, the Netherlands (W.A.B.)
| | - Dominique P V de Kleijn
- Departments of Cardiology (A.T.L.F., M.J.M.S., D.P.V.d.K.), University Medical Center Utrecht, the Netherlands.,Vascular Surgery (D.P.V.d.K.), University Medical Center Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands (D.P.V.d.K.)
| | - Arend Mosterd
- Julius Center for Health Sciences and Primary Care (A.M.), University Medical Center Utrecht, the Netherlands.,Dutch Network for Cardiovascular Research, Utrecht, the Netherlands (A.T.L.F., A.M., J.H.C.).,Department of Cardiology, Meander Medical Center, Amersfoort, the Netherlands (A.M.)
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, the Netherlands (R.M.H., E.S.G.S.)
| | - Jan H Cornel
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands (T.S.J.O., J.H.C.).,Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands (T.S.J.O., J.H.C.).,Dutch Network for Cardiovascular Research, Utrecht, the Netherlands (A.T.L.F., A.M., J.H.C.)
| |
Collapse
|
158
|
Olcum M, Tastan B, Ercan I, Eltutan IB, Genc S. Inhibitory effects of phytochemicals on NLRP3 inflammasome activation: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 75:153238. [PMID: 32507349 DOI: 10.1016/j.phymed.2020.153238] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/12/2020] [Accepted: 04/27/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The NLRP3 inflammasome formation and following cytokine secretion is a crucial step in innate immune responses. Internal and external factors may trigger inflammasome activation and result in inflammatory cytokine secretion. Inflammasome formation and activity play critical roles in several disease pathologies such as cardiovascular, metabolic, renal, digestive, and CNS diseases. Underlying pathways are not yet clear, but phytochemicals as alternative therapies have been extensively used for suppression of inflammatory responses. PURPOSE In this review, we aimed to summarize in vivo and in vitro effects on NLRP3 inflammasome activation of selected phytochemicals. METHOD Three phytochemicals; Sulforaphane, Curcumin, and Resveratrol were selected, and studies were reviewed to clarify their intracellular signaling mechanism in NLRP3 inflammasome activity. PubMed and Scopus databases are used for the search. For sulforaphane, 8 articles, for curcumin, 25 articles, and for resveratrol, 41 articles were included in the review. CONCLUSION In vitro and in vivo studies pointed out that the selected phytochemicals have inhibitory properties on NLRP3 inflammasome activity. However, neither the mechanism is clear, nor the study designs and doses are standardized.
Collapse
Affiliation(s)
- Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Bora Tastan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ilkcan Ercan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Irem B Eltutan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Izmir, Turkey.
| |
Collapse
|
159
|
Parra-Medina R, Sarmiento-Monroy JC, Rojas-Villarraga A, Garavito E, Montealegre-Gómez G, Gómez-López A. Colchicine as a possible therapeutic option in COVID-19 infection. Clin Rheumatol 2020; 39:2485-2486. [PMID: 32556936 PMCID: PMC7301347 DOI: 10.1007/s10067-020-05247-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 05/25/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Rafael Parra-Medina
- Department of Pathology, Fundación Universitaria de Ciencias de la Salud, Institución, Cra. 19 N 8ª-32, Bogotá, Colombia.
- Research Institute, Fundación Universitaria de Ciencias de la Salud, Institución, Cra. 19 N 8ª-32, Bogotá, Colombia.
| | | | - Adriana Rojas-Villarraga
- Research Institute, Fundación Universitaria de Ciencias de la Salud, Institución, Cra. 19 N 8ª-32, Bogotá, Colombia
| | - Edgar Garavito
- Immunology Department, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia
- Universidad Nacional de Colombia, Bogotá, Colombia
- Fundación Universitaria Sanitas, Bogotá, Colombia
| | - Giovanni Montealegre-Gómez
- Department of Plastic and Reconstructive Surgery, Hospital de San José, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia
| | - Arley Gómez-López
- Research Institute, Fundación Universitaria de Ciencias de la Salud, Institución, Cra. 19 N 8ª-32, Bogotá, Colombia
| |
Collapse
|
160
|
Akodad M, Sicard P, Fauconnier J, Roubille F. Colchicine and myocardial infarction: A review. Arch Cardiovasc Dis 2020; 113:652-659. [PMID: 32712201 DOI: 10.1016/j.acvd.2020.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022]
Abstract
The inflammatory response is frequent after acute myocardial infarction, and may worsen ischaemia-reperfusion injuries, leading to increased infarct size and poor prognosis. Therefore, inflammation may be a promising therapeutic target, and anti-inflammatory drugs appear to be potential additional treatments in this context. Among these treatments, colchicine-a well-known drug that has been used for centuries in clinical practice for rheumatism-may represent the ideal candidate. Indeed, colchicine exerts direct anti-inflammatory and pleiotropic effects, with potential anti-arrhythmic, anti-fibrotic and anti-atherosclerotic effects, which are particularly interesting in this population of patients. The effects of colchicine in the context of acute myocardial infarction were first studied in preclinical models, with a decrease in inflammation demonstrated in several in vitro and in vivo models. Moreover, a decrease in infarct size and positive effects on haemodynamic variables were also recently demonstrated in a mouse model. Regarding clinical studies, the positive effect of colchicine in stable coronary disease and atherosclerosis was assessed initially. More recently, the value of colchicine in acute myocardial infarction has been studied, showing a positive effect on inflammation and infarct size reduction. Finally, a randomised trial (the COLCOT study) has shown a reduction in outcomes in patients with acute coronary syndrome treated with colchicine.
Collapse
Affiliation(s)
- Mariama Akodad
- Department of Cardiology, Montpellier University Hospital, 34295 Montpellier, France; PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France.
| | - Pierre Sicard
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France
| | - Jérémy Fauconnier
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France
| | - François Roubille
- Department of Cardiology, Montpellier University Hospital, 34295 Montpellier, France; PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France
| |
Collapse
|
161
|
Zhang R, Song B, Hong X, Shen Z, Sui L, Wang S. microRNA-9 Inhibits Vulnerable Plaque Formation and Vascular Remodeling via Suppression of the SDC2-Dependent FAK/ERK Signaling Pathway in Mice With Atherosclerosis. Front Physiol 2020; 11:804. [PMID: 32765295 PMCID: PMC7378740 DOI: 10.3389/fphys.2020.00804] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
microRNAs (miRNAs or miRs) play important roles in modulating the occurrence and progression of atherosclerosis and acute coronary syndrome (ACS). Herein, this study aimed to investigate the possible role of miR-9 in the development of atherosclerosis. Initially, the differentially expressed genes associated with ACS were screened and miRNAs that regulate syndecan-2 (SDC2) were predicted using microarray analysis. Furthermore, the biological functions of miR-9 and SDC2 on aortic plaque area, proliferation of collagen fibers, Mac-3-labeled macrophages, inflammatory response, and levels of the focal adhesion kinase/extracellular signal-regulated kinase (FAK/ERK) signaling pathway-related proteins in atherosclerosis were evaluated after ectopic miR-9 expression or SDC2 depletion in ACS mice using oil red O staining, Masson’s trichrome staining, immunohistochemistry, and Western blot analysis, respectively. SDC2 was highly-expressed, while miR-9 was poorly-expressed in atherosclerosis. Additionally, miR-9 targeted SDC2 and negatively-regulated its expression. Up-regulation of miR-9 reduced aortic plaque area, the proliferation of collagen fibers, Mac-3-labeled macrophages and levels of IL-6, IL-1β, and TNF-α by suppressing SDC2 and the FAK/ERK signaling pathway, thereby ameliorating atherosclerosis in ACS mice. In conclusion, the current study provides evidence that miR-9 retards atherosclerosis by repressing SDC2 and the FAK/ERK signaling pathway, highlighting a new theoretical basis for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Ruihong Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Beibei Song
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaojian Hong
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiyuan Shen
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Sui
- Department of Emergency, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Siyu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
162
|
McKnight AH, Katzenberger DR, Britnell SR. Colchicine in Acute Coronary Syndrome: A Systematic Review. Ann Pharmacother 2020; 55:187-197. [PMID: 32659104 DOI: 10.1177/1060028020942144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The purpose of this review is to evaluate the efficacy and safety of colchicine after acute coronary syndrome (ACS). DATA SOURCES English-language searches were made of MEDLINE and EMBASE from database inception through mid-June 2020. STUDY SELECTION AND DATA EXTRACTION Randomized trials characterizing the effects of colchicine in ACS were considered. Of 627 title and abstracts identified, nine trials were included. Two reviewers extracted data and rated study quality. DATA SYNTHESIS Four studies showed colchicine did not attenuate C-reactive protein production. Colchicine did modulate the NOD-like receptor family pyrin domain containing 3 inflammasome in 3 studies and reduced production of chemokine ligand 2 (CCL2), CCL5, and C-X3-C motif chemokine ligand 1 in 1 study. Major adverse cardiovascular events (MACE) were not significantly different at 30 days in 3 studies, administered as 1.8 mg preprocedurally or scheduled 1 mg daily. One study found a significant reduction in MACE with colchicine 0.5 mg daily over median 22.6 months (hazard ratio = 0.77; 95% CI = 0.61-0.96). Colchicine is associated with increased gastrointestinal adverse events but was generally well tolerated. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Colchicine is likely to reduce MACE in an ACS population if administered for greater than 30 days but does not improve MACE when administered only preprocedurally. CONCLUSIONS Adjunctive colchicine 0.5 mg daily for greater than 30 days is reasonable for an ACS population on guideline-directed medical therapy treated with PCI. Additional studies are needed to validate and determine the durability of these benefits.
Collapse
|
163
|
Prasinou M, Smith R, Vrettos A, Jayne DRW. The role of IL-18 in Behcet's disease; a potential therapeutic target. Autoimmun Rev 2020; 19:102613. [PMID: 32663625 DOI: 10.1016/j.autrev.2020.102613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 03/22/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Maria Prasinou
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom.
| | - Rona Smith
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom.
| | | | - David R W Jayne
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
164
|
NLPR3 Inflammasomes and Their Significance for Atherosclerosis. Biomedicines 2020; 8:biomedicines8070205. [PMID: 32664349 PMCID: PMC7399980 DOI: 10.3390/biomedicines8070205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a serious disorder, with numerous potential complications such as cardiovascular disease, ischemic stroke, and myocardial infarction. The origin of atherosclerosis is related to chronic inflammation, lipid metabolism alterations, and oxidative stress. Inflammasomes are the cytoplasmic multiprotein complex triggering the activation of inflammatory response. NLRP3 inflammasomes have a specific activation pathway that involves numerous stimuli, including a wide range of PAMPs and DAMPs. Recent studies of atherosclerotic pathology are focused on the mitochondria that appear to be a promising target for therapeutic approach development. Mitochondria are the main source of reactive oxygen species (ROS) associated with oxidative stress. It was previously shown that NLRP3 inflammasome activation results in mitochondrial damage, but the exact mechanisms of this need to be specified. In this review, we focused on the features of NLRP3 inflammasomes and their significance for atherosclerosis, especially concerning mitochondria.
Collapse
|
165
|
Imazio M, Brucato A, Lazaros G, Andreis A, Scarsi M, Klein A, De Ferrari GM, Adler Y. Anti-inflammatory therapies for pericardial diseases in the COVID-19 pandemic: safety and potentiality. J Cardiovasc Med (Hagerstown) 2020; 21:625-629. [DOI: 10.2459/jcm.0000000000001059] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
166
|
Waters DD. Inflammation in Cardiovascular Disease: From Basic Concepts to Clinical Application. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2020. [DOI: 10.36660/ijcs.20200048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
167
|
Imazio M, Andreis A, Brucato A, Adler Y, De Ferrari GM. Colchicine for acute and chronic coronary syndromes. Heart 2020; 106:1555-1560. [PMID: 32611559 DOI: 10.1136/heartjnl-2020-317108] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Colchicine is an ancient drug, traditionally used for the treatment and prevention of gouty attacks; it has become standard of treatment for pericarditis with a potential role in the treatment of coronary artery disease. Atherosclerotic plaque formation, progression, destabilisation and rupture are influenced by active proinflammatory cytokines interleukin (IL)-1β and IL-18 that are generated in the active forms by inflammasomes, which are cytosolic multiprotein oligomers of the innate immune system responsible for the activation of inflammatory responses. Colchicine has a unique anti-inflammatory mechanism: it is not only able to concentrate in leucocytes, especially neutrophils, and block tubulin polymerisation, affecting the microtubules assembly, but also inhibits (NOD)-like receptor protein 3 (NLRP3) inflammasome. On this basis, colchicine interferes with several functions of leucocytes and the assembly and activation of the inflammasome as well, reducing the production of interleukin 1β and interleukin 18. Long-term use of colchicine has been associated with a reduced rate of cardiovascular events both in chronic and acute coronary syndromes, with an overall good safety profile. This review will focus on the influence of colchicine on the pathophysiology of coronary artery disease, reviewing essential pharmacology and discussing the most important and recent clinical studies. On the basis of current literature, colchicine is emerging as a possible new valuable, safe and cheap agent for the treatment of acute and chronic coronary syndromes.
Collapse
Affiliation(s)
- Massimo Imazio
- University Cardiology, AOU Città della Salute e della Scienza di Torino, Torino, Italy .,Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | - Alessandro Andreis
- University Cardiology, Department of Medical Sciences, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Antonio Brucato
- Department of Biomedical and Clinical Sciences, Fatebenefratelli Hospital and University of Milano, Milano, Italy
| | - Yehuda Adler
- College of Law and Business, Sackler Faculty of Medicine, Tel Aviv University, Ramat Gan And Tel Aviv, Israel
| | - Gaetano Maria De Ferrari
- University Cardiology, Department of Medical Sciences, AOU Città della Salute e della Scienza di Torino and University of Torino, Torino, Italy
| |
Collapse
|
168
|
Potì F, Pozzoli C, Adami M, Poli E, Costa LG. Treatments for COVID-19: emerging drugs against the coronavirus. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:118-136. [PMID: 32420936 PMCID: PMC7569629 DOI: 10.23750/abm.v91i2.9639] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 01/08/2023]
Abstract
The Coronavirus disease 19 (COVID-19) outbreak has been recognized as a global threat to public health. It is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and no effective therapies currently exist against this novel viral agent. Along with extensive public health measures, an unprecedented global effort in identifying effective drugs for the treatment is being implemented. Potential drug targets are emerging as the result of a fast-evolving understanding of SARS-CoV-2 virology, host response to the infection, and clinical course of the disease. This brief review focuses on the latest and most promising pharmacological treatments against COVID-19 currently under investigation and discuss their potential use based on either documented efficacy in similar viral infections, or their activity against inflammatory syndromes. Ongoing clinical trials are also emphasized.
Collapse
Affiliation(s)
- Francesco Potì
- Department of Medicine and Surgery - Unit of Neurosciences, University of Parma, Parma, Italy.
| | - Cristina Pozzoli
- Department of Medicine and Surgery - Unit of Neurosciences, University of Parma, Parma, Italy.
| | - Maristella Adami
- Department of Medicine and Surgery - Unit of Neurosciences, University of Parma, Parma, Italy.
| | - Enzo Poli
- Department of Medicine and Surgery - Unit of Neurosciences, University of Parma, Parma, Italy.
| | - Lucio G Costa
- Department of Medicine and Surgery - Unit of Neurosciences, University of Parma, Parma, Italy.
| |
Collapse
|
169
|
Webb CA, Barry AR. Colchicine for Secondary Cardiovascular Prevention: A Systematic Review. Pharmacotherapy 2020; 40:575-583. [PMID: 32259308 DOI: 10.1002/phar.2401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Despite advancements in medical and interventional therapy, patients with cardiovascular disease (CVD) continue to have residual risk for recurrent cardiovascular events. Colchicine has a unique antiinflammatory mechanism that has generated interest in its potential use as a secondary cardiovascular preventive therapy. The objective of this systematic review was to evaluate the evidence for long-term (6 months or more) colchicine therapy in patients with established CVD. A search of Medline and Embase from inception to February 2020 was performed. Included were randomized controlled trials (RCTs) or propensity score-matched observational studies that compared colchicine (at any dose) with placebo or no treatment. Outcomes of interest included any major adverse cardiovascular event, cardiovascular hospitalization, coronary artery restenosis, cardiovascular death, or all-cause death. Five RCTs were included. The dose of colchicine ranged from 0.5 mg/day to 0.6 mg twice/day, and follow-up ranged from ~6-36 months. Two trials (one double blind and one single blind) showed a reduction in composite outcomes of major adverse cardiovascular events. One study failed to demonstrate a benefit with colchicine in restenosis or recurrent ischemia after angioplasty; however, it was conducted before the routine use of modern percutaneous coronary intervention and medical therapies. In contrast, a more recent trial found that colchicine reduced the rate of in-stent restenosis in patients who received a bare metal stent. Finally, one trial in patients with heart failure with reduced ejection fraction did not observe a benefit in death or heart failure hospitalization with colchicine despite a reduction in inflammatory markers. No trial demonstrated a reduction in cardiovascular or all-cause death, and most trials showed an increase in the rate of diarrhea with colchicine. Overall, colchicine has demonstrated promising results for the secondary prevention of CVD; however, further studies are required to confirm these findings before colchicine can be routinely recommended in practice.
Collapse
Affiliation(s)
- Carly A Webb
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Arden R Barry
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Chilliwack General Hospital, Lower Mainland Pharmacy Services, Chilliwack, British Columbia, Canada
| |
Collapse
|
170
|
Role of Colchicine in Stroke Prevention: An Updated Meta-Analysis. J Stroke Cerebrovasc Dis 2020; 29:104756. [DOI: 10.1016/j.jstrokecerebrovasdis.2020.104756] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/27/2020] [Accepted: 02/11/2020] [Indexed: 12/13/2022] Open
|
171
|
Alekberova ZS, Nasonov EL. PROSPECTS FOR USING COLCHICINE IN MEDICINE: NEW EVIDENCE. RHEUMATOLOGY SCIENCE AND PRACTICE 2020. [DOI: 10.14412/1995-4484-2020-183-190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology;
I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| |
Collapse
|
172
|
Cong L, Gao Z, Zheng Y, Ye T, Wang Z, Wang P, Li M, Dong B, Yang W, Li Q, Qiao S, Wang C, Shen Y, Li H, Tian W, Yang L. Electrical stimulation inhibits Val-boroPro-induced pyroptosis in THP-1 macrophages via sirtuin3 activation to promote autophagy and inhibit ROS generation. Aging (Albany NY) 2020; 12:6415-6435. [PMID: 32289749 PMCID: PMC7185124 DOI: 10.18632/aging.103038] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/04/2020] [Indexed: 12/19/2022]
Abstract
The incidence of atherosclerosis (AS), a major contributor to cardiovascular disease, is steadily rising along with an increasingly older population worldwide. Pyroptosis, a form of inflammatory programmed cell death, determines the release of pro-inflammatory mediators by endothelial cells, smooth muscle cells, and atheroma-associated macrophages and foam cells, thereby playing a critical role in AS progression. Canonical pyroptosis is mediated by inflammasome formation, activation of caspase-1, and maturation and release of proinflammatory cytokines. Electrical stimulation (ES) is a noninvasive, safe therapy that has been shown to alleviate symptoms in several health conditions. Here, we investigated the anti-inflammatory and anti-pyroptotic effects of ES in human THP-1 macrophages treated with the dipeptidyl peptidase inhibitor Val-boroPro (VbP). We found that ES downregulated NOD-like receptor family protein 3 (NLRP3) inflammasome, ASC, and caspase-1 expression and abrogated the release of Interleukin-1β (IL-1β) and Interleukin-18 (IL-18), indicating effective pyroptosis inhibition. These changes were paralleled by a reduction in reactive oxygen species (ROS) production, reversal of VbP-induced sirtuin3 (Sirt3) downregulation, deacetylation of ATG5, and induction of autophagy. These findings suggest that ES may be a viable strategy to counteract pyroptosis-mediated inflammation in AS by raising Sirt3 to promote autophagy and inhibit ROS generation.
Collapse
Affiliation(s)
- Lin Cong
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Ziyu Gao
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Yinghong Zheng
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Ting Ye
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Zitong Wang
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Pengyu Wang
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Manman Li
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Bowen Dong
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Wei Yang
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Quanfeng Li
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Shupei Qiao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150006, China
| | - Cao Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150006, China
| | - Yijun Shen
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150006, China
| | - Hong Li
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Weiming Tian
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150006, China
| | - Liming Yang
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing 100037, China
| |
Collapse
|
173
|
NLRP3 Inflammasome: A Potential Alternative Therapy Target for Atherosclerosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1561342. [PMID: 32328119 PMCID: PMC7150718 DOI: 10.1155/2020/1561342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/21/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis (AS) is a complex and chronic inflammatory disease that occurs in multiple systems of the human body. It is an important pathological basis for a variety of diseases and a serious threat to human health. So far, many theories have been formed to explain the pathogenesis of atherosclerosis, among which “inflammation theory” has gradually become a research focus. This theory presents that inflammatory response runs through the whole progress of AS, inflammatory cells play as the main executors of AS, and inflammatory mediators are the key molecules of AS. In the inflammatory process of atherosclerosis, the role of NLRP3 in the atherosclerosis has gradually got the attention of researchers. NLRP3 is a kind of signal-transductional pattern recognition receptors (PRRs). After recognizing and binding to the damage factors, NLRP3 inflammasome will be assembled to activate IL-1β and caspase-1 pathways, resulting in promoting the inflammation process of AS, reducing the stability of the plaques, and finally increasing the incidence of adverse cardiovascular events. Taken above, the article will review the potential benefits of drugs targeting the NLRP3 inflammasome in the therapy of AS.
Collapse
|
174
|
Afzali M, Mostafavi A, Shamspur T. Sensitive detection of colchicine at a glassy carbon electrode modified with magnetic ionic liquid/CuO nanoparticles/carbon nanofibers in pharmaceutical and plasma samples. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2020. [DOI: 10.1007/s13738-020-01894-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
175
|
Abstract
PURPOSE OF REVIEW Based on compelling data from animal and human studies, over the past few decades, the viewpoint of atherosclerosis as an exclusively lipid-driven disease, has been gradually replaced by the concept of a chronic low-grade inflammatory process of the arterial wall. This review presents a brief description on the role of inflammation in atherosclerosis, and examines selected anti-inflammatory interventions that have been tested in clinical trials designed to prevent adverse cardiovascular disease (CVD) events and excess CVD risk. RECENT FINDINGS The Canakinumab Anti-inflammatory Thrombosis Outcomes Study trial has provided convincing evidence that neutralization of the interleukin (IL)-1β inflammatory pathway by the selective antibody canakinumab reduces major CVD events and significantly lowers IL-1β, IL-6 and high-sensitivity C-reactive protein, without affecting low-density lipoprotein cholesterol levels. In contrast, in the latest Cardiovascular Inflammation Reduction Trial, low-dose methotrexate compared with placebo did not reduce CVD events, probably because there was no reduction in IL-1β, or in downstream inflammatory biomarker levels either. SUMMARY Notwithstanding the utilization of effective medical treatments including statins and proprotein convertase subtilisin/kexin type 9 inhibitors or precise revascularizations, the recurrence of CVD events remains unacceptably high. Canakinumab is, at present, the only anti-inflammatory agent that has been proven to reduce cardiovascular events in patients with elevated markers of inflammation without modifying cholesterol levels. Nevertheless, clinical application related to this new evidence and associated knowledge has not yet been implemented in daily practice.
Collapse
|
176
|
Effects of Colchicine on Atherosclerotic Plaque Stabilization: a Multimodality Imaging Study in an Animal Model. J Cardiovasc Transl Res 2020; 14:150-160. [PMID: 32140929 DOI: 10.1007/s12265-020-09974-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Colchicine demonstrated clinical benefits in the treatment of stable coronary artery disease. Our aim was to evaluate the effects of colchicine on atherosclerotic plaque stabilization. Atherosclerosis was induced in the abdominal aorta of 20 rabbits with high-cholesterol diet and balloon endothelial denudation. Rabbits were randomized to receive either colchicine or placebo. All animals underwent MRI, 18F-FDG PET/CT, optical coherence tomography (OCT), and histology. Similar progression of atherosclerotic burden was observed in the two groups as relative increase of normalized wall index (NWI). Maximum 18F-FDG standardized uptake value (meanSUVmax) decreased after colchicine treatment, while it increased in the placebo group with a trend toward significance. Animals with higher levels of cholesterol showed significant differences in favor to colchicine group, both as NWI at the end of the protocol and as relative increase in meanSUVmax. Colchicine may stabilize atherosclerotic plaque by reducing inflammatory activity and plaque burden, without altering macrophage infiltration or plaque typology.
Collapse
|
177
|
Dudam R, Dekate P. Old Drug, New Trick: Colchicine for Cardiovascular Diseases. J R Coll Physicians Edinb 2020; 50:49-50. [DOI: 10.4997/jrcpe.2020.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Rajkiran Dudam
- Consultant Rheumatologist and Director, Hyderabad Rheumatology Center, Begumpet, Hyderabad, India
| | - Prajakta Dekate
- Senior Resident, Hyderabad Rheumatology Center, Begumpet, Hyderabad, India
| |
Collapse
|
178
|
[Colchicine treatment after myocardial infarction : Colchicine Cardiovascular Outcomes Trial (COLCOT)]. Internist (Berl) 2020; 61:766-769. [PMID: 32103281 DOI: 10.1007/s00108-020-00768-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
179
|
Atherosclerosis: Insights into Vascular Pathobiology and Outlook to Novel Treatments. J Cardiovasc Transl Res 2020; 13:744-757. [PMID: 32072564 DOI: 10.1007/s12265-020-09961-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
The pathobiology of atherosclerosis and its current and potential future treatments are summarized, with a spotlight on three central cell types involved: (i) endothelial cells (ECs), (ii) macrophages, and (iii) vascular smooth muscle cells (VSMCs). (i) EC behaviour is regulated by the central transcription factors YAP/TAZ in reaction to biomechanical forces, such as hemodynamic shear stress. (ii) VSMC transdifferentiation (phenotype switching) to a macrophage-like phenotype contributes to the majority of cells positive for common cell surface macrophage markers in atherosclerotic plaques. (iii) Intra-plaque macrophages originate in a significant number from vascular resident macrophages. They can be activated via pattern recognition receptors on cell membrane (e.g. toll-like receptors) and inside cells (e.g. inflammasomes), requiring priming by neutrophil extracellular traps (NETs). ECs and macrophages can also be characterized by single-cell RNA sequencing. Adaptive immunity plays an important role in the inflammatory process. Future therapeutic options include vaccination, TRAF-STOPs, senolysis, or CD47 blockade. Graphical Abstract.
Collapse
|
180
|
Abstract
Inflammasomes are multiprotein innate immune complexes that regulate caspase-dependent inflammation and cell death. Pattern recognition receptors, such as nucleotide-binding oligomerization domain (NOD)-like receptors and absent in melanoma 2 (AIM2)-like receptors, sense danger signals or cellular events to activate canonical inflammasomes, resulting in caspase 1 activation, pyroptosis and the secretion of IL-1β and IL-18. Non-canonical inflammasomes can be activated by intracellular lipopolysaccharides, toxins and some cell signalling pathways. These inflammasomes regulate the activation of alternative caspases (caspase 4, caspase 5, caspase 11 and caspase 8) that lead to pyroptosis, apoptosis and the regulation of other cellular pathways. Many inflammasome-related genes and proteins have been implicated in animal models of kidney disease. In particular, the NLRP3 (NOD-, LRR- and pyrin domain-containing 3) inflammasome has been shown to contribute to a wide range of acute and chronic microbial and non-microbial kidney diseases via canonical and non-canonical mechanisms that regulate inflammation, pyroptosis, apoptosis and fibrosis. In patients with chronic kidney disease, immunomodulation therapies targeting IL-1β such as canakinumab have been shown to prevent cardiovascular events. Moreover, findings in experimental models of kidney disease suggest that small-molecule inhibitors targeting NLRP3 and other inflammasome components are promising therapeutic agents.
Collapse
Affiliation(s)
- Takanori Komada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Daniel A Muruve
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
181
|
Corrêa R, Silva LFF, Ribeiro DJS, Almeida RDN, Santos IDO, Corrêa LH, de Sant'Ana LP, Assunção LS, Bozza PT, Magalhães KG. Lysophosphatidylcholine Induces NLRP3 Inflammasome-Mediated Foam Cell Formation and Pyroptosis in Human Monocytes and Endothelial Cells. Front Immunol 2020; 10:2927. [PMID: 31998284 PMCID: PMC6962110 DOI: 10.3389/fimmu.2019.02927] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/28/2019] [Indexed: 12/04/2022] Open
Abstract
Foam cells are specialized lipid-loaded macrophages derived from monocytes and are a key pathological feature of atherosclerotic lesions. Lysophosphatidylcholine (LPC) is a major lipid component of the plasma membrane with a broad spectrum of proinflammatory activities and plays a key role in atherosclerosis. However, the role of LPC in lipid droplet (LD) biogenesis and the modulation of inflammasome activation is still poorly understood. In the present study, we investigated whether LPC can induce foam cell formation through an analysis of LD biogenesis and determined whether the cell signaling involved in this process is mediated by the inflammasome activation pathway in human endothelial cells and monocytes. Our results showed that LPC induced foam cell formation in both types of cells by increasing LD biogenesis via a NLRP3 inflammasome-dependent pathway. Furthermore, LPC induced pyroptosis in both cells and the activation of the inflammasome with IL-1β secretion, which was dependent on potassium efflux and lysosomal damage in human monocytes. The present study described the IL-1β secretion and foam cell formation triggered by LPC via an inflammasome-mediated pathway in human monocytes and endothelial cells. Our results will help improve our understanding of the relationships among LPC, LD biogenesis, and NLRP3 inflammasome activation in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Rafael Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Luís Felipe Fonseca Silva
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | | | - Raquel das Neves Almeida
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Igor de Oliveira Santos
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Lívia Pimentel de Sant'Ana
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | | | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Institute of Oswaldo Cruz-Fiocruz, Rio de Janeiro, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| |
Collapse
|
182
|
Colchicine-Binding Site Inhibitors from Chemistry to Clinic: A Review. Pharmaceuticals (Basel) 2020; 13:ph13010008. [PMID: 31947889 PMCID: PMC7168938 DOI: 10.3390/ph13010008] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
It is over 50 years since the discovery of microtubules, and they have become one of the most important drug targets for anti-cancer therapies. Microtubules are predominantly composed of the protein tubulin, which contains a number of different binding sites for small-molecule drugs. There is continued interest in drug development for compounds targeting the colchicine-binding site of tubulin, termed colchicine-binding site inhibitors (CBSIs). This review highlights CBSIs discovered through diverse sources: from natural compounds, rational design, serendipitously and via high-throughput screening. We provide an update on CBSIs reported in the past three years and discuss the clinical status of CBSIs. It is likely that efforts will continue to develop CBSIs for a diverse set of cancers, and this review provides a timely update on recent developments.
Collapse
|
183
|
Hamroun A, Frimat M, Beuscart JB, Buob D, Lionet A, Lebas C, Daroux M, Provôt F, Hazzan M, Boulanger É, Glowacki F. [Kidney disease care for the elderly]. Nephrol Ther 2019; 15:533-552. [PMID: 31711751 DOI: 10.1016/j.nephro.2019.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In our aging population, kidney disease management needs to take into account the frailty of the elderly. Standardized geriatric assessments can be proposed to help clinicians apprehend this dimension in their daily practice. These tools allow to better identify frail patients and offer them more personalized and harmless treatments. This article aims to focus on the kidney diseases commonly observed in elderly patients and analyze their specific nephrogeriatric care modalities. It should be noticed that all known kidney diseases can be also observed in the elderly, most often with a quite similar clinical presentation. This review is thus focused on the diseases most frequently and most specifically observed in elderly patients (except for monoclonal gammopathy associated nephropathies, out of the scope of this work), as well as the peculiarities of old age nephrological care.
Collapse
Affiliation(s)
- Aghilès Hamroun
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France
| | - Marie Frimat
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France
| | | | - David Buob
- Service d'anatomopathologie, Centre de biologie-pathologie, CHRU de Lille, 59037 Lille, France
| | - Arnaud Lionet
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France
| | - Céline Lebas
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France
| | - Maïté Daroux
- Service de néphrologie, hôpital Duchenne, allée Jacques Monod, 62200 Boulogne-sur-Mer, France
| | - François Provôt
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France
| | - Marc Hazzan
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France
| | - Éric Boulanger
- Service de gériatrie, CHRU de Lille, 59037 Lille, France
| | - François Glowacki
- Service de néphrologie, hôpital Huriez, CHRU de Lille, 59037 Lille, France.
| |
Collapse
|
184
|
ER Stress Activates the NLRP3 Inflammasome: A Novel Mechanism of Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3462530. [PMID: 31687078 PMCID: PMC6800950 DOI: 10.1155/2019/3462530] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/21/2019] [Accepted: 08/31/2019] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is an important organelle that regulates several fundamental cellular processes, and ER dysfunction has implications for many intracellular events. The nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is an intracellularly produced macromolecular complex that can trigger pyroptosis and inflammation, and its activation is induced by a variety of signals. ER stress has been found to affect NLRP3 inflammasome activation through multiple effects including the unfolded protein response (UPR), calcium or lipid metabolism, and reactive oxygen species (ROS) generation. Intriguingly, the role of ER stress in inflammasome activation has not attracted a great deal of attention. In addition, increasing evidence highlights that both ER stress and NLRP3 inflammasome activation contribute to atherosclerosis (AS). AS is a common cardiovascular disease with complex pathogenesis, and the precise mechanisms behind its pathogenesis remain to be determined. Both ER stress and the NLRP3 inflammasome have emerged as critical individual contributors of AS, and owing to the multiple associations between these two events, we speculate that they contribute to the mechanisms of pathogenesis in AS. In this review, we aim to summarize the molecular mechanisms of ER stress, NLRP3 inflammasome activation, and the cross talk between these two pathways in AS in the hopes of providing new pharmacological targets for AS treatment.
Collapse
|
185
|
Sokolova M, Ranheim T, Louwe MC, Halvorsen B, Yndestad A, Aukrust P. NLRP3 Inflammasome: A Novel Player in Metabolically Induced Inflammation-Potential Influence on the Myocardium. J Cardiovasc Pharmacol 2019; 74:276-284. [PMID: 31584530 DOI: 10.1097/fjc.0000000000000704] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolic and immune systems are among the most fundamental requirements for survival. Many metabolic and immune response pathways or nutrient- and pathogen-sensing systems are evolutionarily conserved throughout species. As a result, the immune response and metabolic regulation are highly integrated and the proper function of each is dependent on the other. This interaction between metabolic disturbances and the immune system has been most extensively studied in disorders related to obesity such as insulin resistance, type 2 diabetes, and nonalcoholic fatty liver disease. Metabolically induced inflammation seems also to play a role in the development and progression of atherosclerosis including its complications such as myocardial infarction (MI) and post-MI remodeling. There are several lines of evidence suggesting that NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is a sensor of metabolic stress linking metabolic disturbances to inflammation. Here, we will discuss the role of the NLRP3 inflammasome in the pathogenesis of obesity and diabetes, 2 important risk factors for atherosclerosis and MI. We will also discuss the role of NLRP3 inflammasome in the interaction between metabolic disturbances and myocardial inflammation during MI and during metabolically induced myocardial remodeling.
Collapse
Affiliation(s)
- Marina Sokolova
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, The Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mieke C Louwe
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, The Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
186
|
Liang L, Hou X, Bainey KR, Zhang Y, Tymchak W, Qi Z, Li W, Banh HL. The association between hyperuricemia and coronary artery calcification development: A systematic review and meta-analysis. Clin Cardiol 2019; 42:1079-1086. [PMID: 31571239 PMCID: PMC6837029 DOI: 10.1002/clc.23266] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
Hyperuricemia coincides with coronary artery calcification (CAC) development, but the role of serum uric acid (SUA) as a risk factor for CAC remains unclear. The objective of this study was to gain an insight into the association between SUA and CAC in adults by performing a meta-analysis. MEDLINE, EMBASE, the Cochrane Library, and EBSCO (CINAHL) were searched for relevant observational studies published until 2 June 2019. Studies were included only if they reported data on CAC presence (Agatston score > 0) or progression related to hyperuricemia in subclinical adult patients. The pooled estimates of crude and adjusted odds ratios (ORs) and 95% confidence interval (CI) were calculated to evaluate the association between CAC presence or progression and hyperuricemia. A total of 11 studies were identified involving 11 108 adults. The pooled OR based on the frequency of CAC presence showed that patients in the high SUA group had 1.806-fold risk for developing CAC (95% CI: 1.491-2.186) under the minimal threshold of hyperuricemia (more than 6 mg/dL or 357 μmoL/L). When SUA levels were analyzed as categorical variables, the pooled estimate of adjusted ORs was 1.48 (95% CI: 1.23-1.79) for CAC presence. Additionally, for each increase of 1 mg/dL of SUA level, the risk of CAC progression was increased by 31% (95% CI: 1.15-1.49) with an average follow-up duration ranged from 4.6 to 6.1 years. Hyperuricemia is closely associated with increased risk of CAC development and CAC progression in asymptomatic patients.
Collapse
Affiliation(s)
- Ling Liang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Department of Cardiology, First Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xianghua Hou
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Department of Nephrology, First Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Kevin R Bainey
- Division of Cardiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Yanlin Zhang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Department of Nephrology, First Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wayne Tymchak
- Division of Cardiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Zhongquan Qi
- Institute of Organ Transplantation, Xiamen University, Xiamen, China
| | - Weihua Li
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Department of Cardiology, First Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Hoan Linh Banh
- Department of Family Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
187
|
Bartlett B, Ludewick HP, Misra A, Lee S, Dwivedi G. Macrophages and T cells in atherosclerosis: a translational perspective. Am J Physiol Heart Circ Physiol 2019; 317:H375-H386. [DOI: 10.1152/ajpheart.00206.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is now considered a chronic maladaptive inflammatory disease. The hallmark feature in both human and murine disease is atherosclerotic plaques. Macrophages and various T-cell lineages play a crucial role in atherosclerotic plaque establishment and disease progression. Humans and mice share many of the same processes that occur within atherogenesis. The various similarities enable considerable insight into disease mechanisms and those which contribute to cardiovascular complications. The apolipoprotein E-null and low-density lipoprotein receptor-null mice have served as the foundation for further immunological pathway manipulation to identify pro- and antiatherogenic pathways in attempt to reveal more novel therapeutic targets. In this review, we provide a translational perspective and discuss the roles of macrophages and various T-cell lineages in contrasting proatherosclerotic and atheroprotective settings.
Collapse
Affiliation(s)
- Benjamin Bartlett
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Herbert P. Ludewick
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| |
Collapse
|
188
|
Ozkok A. Cholesterol-embolization syndrome: current perspectives. Vasc Health Risk Manag 2019; 15:209-220. [PMID: 31371977 PMCID: PMC6626893 DOI: 10.2147/vhrm.s175150] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Cholesterol-embolization syndrome (CES) is a multisystemic disease with various clinical manifestations. CES is caused by embolization of cholesterol crystals (CCs) from atherosclerotic plaques located in the major arteries, and is induced mostly iatrogenically by interventional and surgical procedures; however, it may also occur spontaneously. Embolized CCs lead to both ischemic and inflammatory damage to the target organ. Therefore, anti-inflammatory agents, such as corticosteroids and cyclophosphamide, have been investigated as treatment for CES in several studies, with conflicting results. Recent research has revealed that CES is actually a kind of autoinflammatory disease in which inflammasome pathways, such as NLRP3 and IL1, are induced by CCs. These recent findings may have clinical implications such that colchicine and IL1 inhibitors, namely canakinumab, may be beneficial in the early stages of CES.
Collapse
Affiliation(s)
- Abdullah Ozkok
- Department of Internal Medicine and Nephrology, Memorial Şişli Hospital, Istanbul, Turkey
| |
Collapse
|
189
|
Marchio P, Guerra-Ojeda S, Vila JM, Aldasoro M, Victor VM, Mauricio MD. Targeting Early Atherosclerosis: A Focus on Oxidative Stress and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8563845. [PMID: 31354915 PMCID: PMC6636482 DOI: 10.1155/2019/8563845] [Citation(s) in RCA: 369] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/10/2019] [Accepted: 05/19/2019] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic vascular inflammatory disease associated to oxidative stress and endothelial dysfunction. Oxidation of low-density lipoprotein (LDL) cholesterol is one of the key factors for the development of atherosclerosis. Nonoxidized LDL have a low affinity for macrophages, so they are not themselves a risk factor. However, lowering LDL levels is a common clinical practice to reduce oxidation and the risk of major events in patients with cardiovascular diseases (CVD). Atherosclerosis starts with dysfunctional changes in the endothelium induced by disturbed shear stress which can lead to endothelial and platelet activation, adhesion of monocytes on the activated endothelium, and differentiation into proinflammatory macrophages, which increase the uptake of oxidized LDL (oxLDL) and turn into foam cells, exacerbating the inflammatory signalling. The atherosclerotic process is accelerated by a myriad of factors, such as the release of inflammatory chemokines and cytokines, the generation of reactive oxygen species (ROS), growth factors, and the proliferation of vascular smooth muscle cells. Inflammation and immunity are key factors for the development and complications of atherosclerosis, and therefore, the whole atherosclerotic process is a target for diagnosis and treatment. In this review, we focus on early stages of the disease and we address both biomarkers and therapeutic approaches currently available and under research.
Collapse
Affiliation(s)
- Patricia Marchio
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - José M. Vila
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Martín Aldasoro
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Victor M. Victor
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Maria D. Mauricio
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| |
Collapse
|
190
|
Carrero JJ, Andersson Franko M, Obergfell A, Gabrielsen A, Jernberg T. hsCRP Level and the Risk of Death or Recurrent Cardiovascular Events in Patients With Myocardial Infarction: a Healthcare-Based Study. J Am Heart Assoc 2019; 8:e012638. [PMID: 31140334 PMCID: PMC6585357 DOI: 10.1161/jaha.119.012638] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background Beyond the controlled setting of trials, scarce information exists on the burden, predictors, and outcomes associated with elevated hsCRP (high‐sensitivity C‐reactive protein) in “real‐world” patients with myocardial infarction (MI). Methods and Results We included all‐coming MI survivors undergoing hsCRP testing >30 days after an MI during routine health care in Stockholm, Sweden (2006–2011). hsCRP tests measured during hospitalization/emergency department visits, followed by antibiotics or indicative of acute illness, were excluded, together with patients with ongoing/recent cancer, chronic infections, or immunosuppression. Inflammation was defined over a 3‐month baseline window and associated with subsequent death and major adverse cardiovascular events (composite of MI, ischemic stroke, or cardiovascular death). Included were 17 464 patients (63% men; mean age, 72.6 years) with a median hsCRP level of 2.2 (interquartile range, 1.0–6.0) mg/L and a median of 2.2 (interquartile range, 0.8–4.9) years since their MI. Most (66%) had hsCRP ≥2 mg/L, and 40% had hsCRP >3 mg/L. Lower hemoglobin, lower estimated glomerular filtration rate, and comorbidities (eg, heart failure, peripheral vascular disease, stroke, atrial fibrillation, diabetes mellitus, and rheumatoid diseases) were associated with higher odds of hsCRP ≥2 mg/L. Conversely, previous percutaneous coronary intervention, ongoing renin‐angiotensin blockade, and statins were associated with lower hsCRP ≥2 mg/L odds. Patients with hsCRP ≥2 mg/L were at higher risk of major adverse cardiovascular events (n=3900; adjusted hazard ratio, 1.28; 95% CI, 1.18–1.38) and death (n=4138; adjusted hazard ratio, 1.42; 95% CI, 1.31–1.53). Results were robust across subgroups of patients and after exclusion of events occurring during the first 6 to 12 months. On a continuous scale, the association between hsCRP and outcomes was linear until hsCRP >5 mg/L, plateauing thereafter. Conclusions Most patients with MI exhibit elevated hsCRP levels. Besides identifying populations at high‐inflammatory risk, this study extends the prognostic validity of this biomarker from trial evidence to real‐world healthcare settings.
Collapse
Affiliation(s)
- Juan Jesus Carrero
- 1 Department of Medical Epidemiology and Biostatistics Karolinska Institutet Stockholm Sweden
| | - Mikael Andersson Franko
- 1 Department of Medical Epidemiology and Biostatistics Karolinska Institutet Stockholm Sweden
| | | | - Anders Gabrielsen
- 3 Cardiovascular Medicine Unit Department of Medicine Solna Karolinska University Hospital Solna Karolinska Institutet Stockholm Sweden
| | - Tomas Jernberg
- 4 Department of Clinical Sciences Danderyd University Hospital Karolinska Institutet Stockholm Sweden
| |
Collapse
|
191
|
Yang Q, Luo J, Lv H, Wen T, Shi B, Liu X, Zeng N. Pulegone inhibits inflammation via suppression of NLRP3 inflammasome and reducing cytokine production in mice. Immunopharmacol Immunotoxicol 2019; 41:420-427. [PMID: 31134844 DOI: 10.1080/08923973.2019.1588292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Context: Pulegone, a key compound in Schizonepeta essential oil, has been identified as an anti-inflammatory. However, its underlying molecular mechanisms on NLR family pyrin domain containing 3 (NLRP3) inflammasome have not been elucidated. Objective: Here, the modulatory effects of pulegone on NLRP3 inflammasome were investigated. Materials and methods: The C57BL/6J mice were randomly divided into five groups: Normal, Lipopolysaccharides (LPS), Dexamethasone (DEX, 5 mg/kg), Pulegone (0.095 and 0.190 g/kg) groups. All mice were challenged by LPS except for the Normal group. Results: A reduced expression of Interleukin-18 (IL-18), Interleukin-1β (IL-1β), Interleukin-5 (IL-5), Tumor necrosis factor-α (TNF-α), Interferon-gamma (IFN-γ), Monocyte chemoattratctant protein-1 (MCP-1), Macrophage inflammatory protein-1β (MIP-1β), Monocyte colony stimulating factor (M-CSF) and Granulocyte-macrophage colony stimulating factor (GM-CSF) in serum were detected in the pulegone groups as compared to the LPS group. In addition, a reduced mRNA and protein expression production of ASC, NLRP3, and Caspase-1 were detected in lungs after pulegone administration. Histological analysis results indicated that the histological changes of lungs caused by LPS were ameliorated by pulegone. Immunohistochemical study showed a decreased positive cell numbers of P2X7R in Pulegone (0.095 and 0.190 g/kg) groups. Conclusion: Pulegone exerts anti-inflammatory effects on LPS-induced sepsis mice via inhibition of the NLRP3 expression.
Collapse
Affiliation(s)
- Qingxin Yang
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China.,b Sichuan College of Traditional Chinese Medicine , Mianyang , PR China
| | - Jie Luo
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China
| | - Hongjun Lv
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China
| | - Taoqun Wen
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China
| | - Boyu Shi
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China
| | - Xiaobo Liu
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China
| | - Nan Zeng
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , PR China
| |
Collapse
|
192
|
Cocchi E, Chiale F, Gianoglio B, Deorsola L, Pace Napoleone C, Fagioli F, Peruzzi L. Colchicine: An Impressive Effect on Posttransplant Capillary Leak Syndrome and Renal Failure. Pediatrics 2019; 143:peds.2018-2820. [PMID: 31000685 DOI: 10.1542/peds.2018-2820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2018] [Indexed: 11/24/2022] Open
Abstract
Capillary leak syndrome is a critical condition occasionally occurring posttransplant and is characterized by acute endothelial hyperpermeability leading to systemic protein-rich fluid extravasation and consequent hypovolemia, hypoperfusion, and acute kidney injury. Treatment is merely supportive and is based on osmotic drugs, diuretics, continuous renal replacement therapy, and surgical drainage. However, removal of the underlying inflammatory cause is mandatory to achieve stable resolution. Herein, we report the first successful treatment with colchicine in 2 life-threatening pediatric cases of capillary leak syndrome with renal failure occurring after transplant (heart and bone marrow) and unresponsive to any other line of therapy. Both cases were only palliated by supportive therapy and revealed an impressively rapid response to colchicine both in terms of diuresis and clinical condition recovery, allowing for the cessation of renal replacement therapy in a few hours. In both patients, colchicine was temporarily discontinued for transient leukopenia (attributed to an additive effect with mycophenolate mofetil), resulting in extravasation, and renal failure recurrence was restored only after colchicine reintroduction. Although the association of colchicine with an immunosuppressive drug was formerly contraindicated, no other adverse events were noted when using a minimized dose. Both patients are now maintaining a good renal function without recurrence of extravasation after 6 months of follow-up. In conclusion, this strikingly positive experience forces physicians to consider this old and cost-effective drug as a new, powerful rescue tool in such critical cases.
Collapse
Affiliation(s)
- Enrico Cocchi
- University of Turin, Turin, Italy; and.,Nephrology, Dialysis and Transplantation Unit
| | - Federica Chiale
- University of Turin, Turin, Italy; and.,Nephrology, Dialysis and Transplantation Unit
| | | | | | | | - Franca Fagioli
- Pediatric Onco-Hematology Unit, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy
| | - Licia Peruzzi
- University of Turin, Turin, Italy; and.,Nephrology, Dialysis and Transplantation Unit
| |
Collapse
|
193
|
Ridker PM. Anti-inflammatory therapy for atherosclerosis: interpreting divergent results from the CANTOS and CIRT clinical trials. J Intern Med 2019; 285:503-509. [PMID: 30472762 DOI: 10.1111/joim.12862] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- P M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
194
|
Khandkar C, Vaidya K, Patel S. Colchicine for Stroke Prevention: A Systematic Review and Meta-analysis. Clin Ther 2019; 41:582-590.e3. [DOI: 10.1016/j.clinthera.2019.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 12/17/2022]
|
195
|
Arbel Y, Abuzeid W, Rosenson RS, Weisman A, Farkouh ME. Old Drugs for New Indications in Cardiovascular Medicine. Cardiovasc Drugs Ther 2019; 32:223-232. [PMID: 29633048 DOI: 10.1007/s10557-018-6785-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Inflammation participates in the initiation and progression of atherosclerotic cardiovascular disease, and it is a critical inciting factor leading to acute ischemic events. Evidence has shown that certain anti-inflammatory medications used to treat non-atherosclerotic inflammatory diseases reduce cardiovascular events. This article reviews evidence that commonly used anti-inflammatory therapies (colchicine, allopurinol, methotrexate), reduce cardiovascular events. We discuss potential mechanisms of action, efficacy, and safety of these therapies and propose a clinical trials design to investigate their efficacy.
Collapse
Affiliation(s)
- Yaron Arbel
- Department of Cardiology, Tel Aviv Medical Center, Tel Aviv, affiliated to the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Wael Abuzeid
- Schulich Heart Centre, Division of Cardiology, Sunnybrook Health Sciences Centre, Ontario, Canada, affiliated with the University of Toronto, Toronto, Ontario, Canada
| | - Robert S Rosenson
- Mount Sinai Icahn School of Medicine, Mount Sinai Hospital, New York, USA
| | - Alanna Weisman
- Division of Endocrinology & Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael E Farkouh
- Peter Munk Centre, Heart & Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
196
|
The Role of Colchicine in Acute Coronary Syndromes. Clin Ther 2019; 41:11-20. [DOI: 10.1016/j.clinthera.2018.07.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 07/29/2018] [Indexed: 12/23/2022]
|
197
|
Nidorf SM, Thompson PL. Why Colchicine Should Be Considered for Secondary Prevention of Atherosclerosis: An Overview. Clin Ther 2018; 41:41-48. [PMID: 30591286 DOI: 10.1016/j.clinthera.2018.11.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Colchicine is a widely available, inexpensive drug with a range of antiinflammatory properties that may make it suitable for the secondary prevention of atherosclerosis. This review examines how past and contemporary approaches to antiinflammatory therapy for atherosclerosis have led to a better understanding of the nature of the disease and sets out the reasons why colchicine has the potential to become a cornerstone therapy in its management. METHODS We performed a literature search using PubMed, the Cochrane library, and clinical trial registries to identify completed and ongoing clinical studies on colchicine in coronary artery disease, and a PubMed search to identify publications on the mechanism of action of colchicine relevant to atherosclerosis. FINDINGS A large body of data confirms that inflammation plays a pivotal role in atherosclerosis. The translation of this extensive knowledge into improved clinical outcomes has until recently been elusive. Findings from statin trials support the possibility that targeting inflammation may be beneficial, but this evidence has been inconclusive. Direct inhibition of atherosclerotic inflammation is being explored in current clinical trials. Targeted inhibition of interleukin 1β with canakinumab provided the proof of principle that limiting inflammation can improve outcomes in atherosclerotic vascular disease, but long-term treatment with a monoclonal antibody is unlikely to have widespread uptake. Other approaches using agents with a wider set of targets are being explored. Findings from observational studies suggest that methotrexate may reduce cardiovascular risk in patients with rheumatoid arthritis, but CIRT (Cardiovascular Inflammation Reduction Trial) demonstrated that methotrexate provided no cardiovascular benefit in patients with atherosclerotic vascular disease. Recent demonstration that cholesterol crystals trigger the NLRP3 (nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3) inflammasome and the release of inflammatory cytokines that also drive uric acid crystal-induced inflammation indicates that the multiple actions of colchicine that make it effective in gout may be relevant to preventing inflammation and limiting inflammatory injury in atherosclerosis. The ongoing LoDoCo2 (Low Dose Colchicine2) and COLCOT (Colchicine Cardiovascular Outcomes Trial) trials and several other planned large-scale rigorous trials will determine the long-term tolerability and efficacy of low-dose colchicine for secondary prevention in patients with coronary disease. IMPLICATION Colchicine holds promise as an important, accessible drug that could be successfully repurposed for the secondary prevention of atherosclerotic cardiovascular disease should its tolerability and cardiovascular benefits be confirmed in ongoing clinical trials.
Collapse
Affiliation(s)
- Stefan Mark Nidorf
- GenesisCare, Perth, Western Australia, Australia; Heart Research Institute, Perth, Western Australia; Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.
| | - Peter Lindsay Thompson
- GenesisCare, Perth, Western Australia, Australia; Heart Research Institute, Perth, Western Australia; Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia; Sir Charles Gairdner Hospital, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
198
|
Dan GA, Dobrev D. Antiarrhythmic drugs for atrial fibrillation: Imminent impulses are emerging. IJC HEART & VASCULATURE 2018; 21:11-15. [PMID: 30225340 PMCID: PMC6138883 DOI: 10.1016/j.ijcha.2018.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/15/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022]
Abstract
Rhythm and rate strategies are considered equivalent for the management of atrial fibrillation (AF). Moreover, both strategies are intended for improving symptoms and quality of life. Despite the clinical availability of several antiarrhythmic drugs (AAD) the alternatives for the patient with comorbidities are significantly fewer because of the concern regarding many adverse effects, including proarrhythmias. The impetuous development of AF ablation gave rise to a false impression that AAD are a second line therapy. All these statements reflect, in fact, the weakness of the classical paradigm and classification regarding AAD and the gap between the current knowledge of AF mechanism and determinants and the "classical" AAD non-discriminatory action. A new paradigm in development of effective and safe AAD is based on modern knowledge of vulnerable parameters involved in the genesis and perpetuation of AF. New AAD will target specific triggers of AF and ion currents which are expressed preferentially in fibrillatory atrium. Such targets will include repolarizing currents and channels, as ultrarapid potassium current, two pore potassium current, the acetylcholine-gated potassium current, small-conductance calcium-dependent potassium channels, but, also, molecular targets involved in intracellular calcium kinetics, as Ca2+-calmodulin-dependent protein kinase, ryanodine receptors and non-coding miRNA. New mechanistic discoveries link AF to inflammation and modern anti-cytokine drugs. There is still a long way to win between basic research and clinical practice, but, without any doubt, antiarrhythmic drug therapy will remain and develop as a cornerstone therapy for AF not in conflict, but complementary and alternative to interventional therapy.
Collapse
Affiliation(s)
- Gheorghe-Andrei Dan
- Carol Davila Medicine University, Bucharest, Romania
- Colentina University Hospital, Bucharest, Romania
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
199
|
Scott L, Li N, Dobrev D. Role of inflammatory signaling in atrial fibrillation. Int J Cardiol 2018; 287:195-200. [PMID: 30316645 DOI: 10.1016/j.ijcard.2018.10.020] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 01/09/2023]
Abstract
Atrial fibrillation (AF), the most prevalent arrhythmia, is often associated with enhanced inflammatory response. Emerging evidence points to a causal role of inflammatory signaling pathways in the evolution of atrial electrical, calcium handling and structural remodeling, which create the substrate of AF development. In this review, we discuss the clinical evidence supporting the association between inflammatory indices and AF development, the molecular and cellular mechanisms of AF, which appear to involve multiple canonical inflammatory pathways, and the potential of anti-inflammatory therapeutic approaches in AF prevention/treatment.
Collapse
Affiliation(s)
- Larry Scott
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA; Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA; Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
200
|
Shao BZ, Cao Q, Liu C. Targeting NLRP3 Inflammasome in the Treatment of CNS Diseases. Front Mol Neurosci 2018; 11:320. [PMID: 30233319 PMCID: PMC6131647 DOI: 10.3389/fnmol.2018.00320] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) is one of the largest killers of people’s health all over the world. The overactivation of the immune and inflammatory responses is considered as an important factor, contributing to the pathogenesis and progression of CNS disorders. Among all kinds of immune and inflammatory reaction, the inflammasome, a complex of proteins, has been drawn increasingly attention to by researchers. The initiation and activation of the inflammasome is involved in the onset of various kinds of diseases. The NLRP3 inflammasome, the most studied member of the inflammasome, is closely associated with many kinds of CNS disorders. Here in this review, the roles of the NLRP3 inflammasome in the pathogenesis and progression of several well-known CNS diseases would be discussed, including cerebrovascular diseases, neurodegenerative diseases, multiple sclerosis, depression as well as other CNS disorders. In addition, several therapeutic strategies targeting on the NLRP3 inflammasome for the treatment of CNS disorders would be described in this review.
Collapse
Affiliation(s)
- Bo-Zong Shao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Qi Cao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Chong Liu
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| |
Collapse
|