51
|
Rattis BAC, Ramos SG, Celes MRN. Curcumin as a Potential Treatment for COVID-19. Front Pharmacol 2021; 12:675287. [PMID: 34025433 PMCID: PMC8138567 DOI: 10.3389/fphar.2021.675287] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/21/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease that rapidly spread throughout the world leading to high mortality rates. Despite the knowledge of previous diseases caused by viruses of the same family, such as MERS and SARS-CoV, management and treatment of patients with COVID-19 is a challenge. One of the best strategies around the world to help combat the COVID-19 has been directed to drug repositioning; however, these drugs are not specific to this new virus. Additionally, the pathophysiology of COVID-19 is highly heterogeneous, and the way of SARS-CoV-2 modulates the different systems in the host remains unidentified, despite recent discoveries. This complex and multifactorial response requires a comprehensive therapeutic approach, enabling the integration and refinement of therapeutic responses of a given single compound that has several action potentials. In this context, natural compounds, such as Curcumin, have shown beneficial effects on the progression of inflammatory diseases due to its numerous action mechanisms: antiviral, anti-inflammatory, anticoagulant, antiplatelet, and cytoprotective. These and many other effects of curcumin make it a promising target in the adjuvant treatment of COVID-19. Hence, the purpose of this review is to specifically point out how curcumin could interfere at different times/points during the infection caused by SARS-CoV-2, providing a substantial contribution of curcumin as a new adjuvant therapy for the treatment of COVID-19.
Collapse
Affiliation(s)
- Bruna A. C. Rattis
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania, Brazil
| | - Simone G. Ramos
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Mara R. N. Celes
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania, Brazil
| |
Collapse
|
52
|
Bonaventura A, Vecchié A, Dagna L, Martinod K, Dixon DL, Van Tassell BW, Dentali F, Montecucco F, Massberg S, Levi M, Abbate A. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat Rev Immunol 2021; 21:319-329. [PMID: 33824483 PMCID: PMC8023349 DOI: 10.1038/s41577-021-00536-9] [Citation(s) in RCA: 619] [Impact Index Per Article: 154.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a clinical syndrome caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients with severe disease show hyperactivation of the immune system, which can affect multiple organs besides the lungs. Here, we propose that SARS-CoV-2 infection induces a process known as immunothrombosis, in which activated neutrophils and monocytes interact with platelets and the coagulation cascade, leading to intravascular clot formation in small and larger vessels. Microthrombotic complications may contribute to acute respiratory distress syndrome (ARDS) and other organ dysfunctions. Therapeutic strategies aimed at reducing immunothrombosis may therefore be useful. Several antithrombotic and immunomodulating drugs have been proposed as candidates to treat patients with SARS-CoV-2 infection. The growing understanding of SARS-CoV-2 infection pathogenesis and how it contributes to critical illness and its complications may help to improve risk stratification and develop targeted therapies to reduce the acute and long-term consequences of this disease.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.
- Department of Internal Medicine, ASST dei Sette Laghi, Varese, Italy.
| | - Alessandra Vecchié
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Internal Medicine, ASST dei Sette Laghi, Varese, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Dave L Dixon
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy and Outcome Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Benjamin W Van Tassell
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy and Outcome Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genova - Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Steffen Massberg
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
- Medizinische Klinik und Poliklinik I, Klinikum der Universität, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marcel Levi
- Department of Medicine and Cardio-metabolic Programme - NIHR UCLH/UCL BRC, University College London Hospitals NHS Foundation Trust, London, UK
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
53
|
Bonetti NR, Liberale L, Akhmedov A, Pasterk L, Gobbato S, Puspitasari YM, Vukolic A, Saeedi Saravi SS, Coester B, Horvath C, Osto E, Montecucco F, Lüscher TF, Beer JH, Camici GG. Long-term dietary supplementation with plant-derived omega-3 fatty acid improves outcome in experimental ischemic stroke. Atherosclerosis 2021; 325:89-98. [PMID: 33915355 DOI: 10.1016/j.atherosclerosis.2021.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/26/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Early revascularization -the gold standard therapy for ischemic stroke- is often withheld in the elderly population due to high risk of complications. Thus, safe and effective preventive and therapeutic options are needed. The plant-derived omega-3-fatty-acid alpha-linolenic-acid (ALA) has emerged as a novel cardiovascular-protective agent. As of yet, little is known about its potential therapeutic effects on stroke. We hereby aimed to investigate the impact of a clinically relevant long-term dietary intervention with ALA on stroke outcome. METHODS Six month-old C57BL/6 wildtype males were either fed an ALA-rich (high ALA) or a control diet (low ALA) for 12 months. At 18 months, brain ischemia/reperfusion was induced by transient middle cerebral artery occlusion (tMCAO). Stroke size and neurological function were assessed. Functional blood-brain-barrier-(BBB) permeability and protein expression were assessed by immunohistochemistry. Baseline inflammatory markers were measured at 18 months. RESULTS High ALA-fed animals displayed decreased circulating TNF-α levels and Neutrophil-to-Lymphocyte Ratios at 18 months. Stroke size and neurological dysfunction were significantly reduced in high ALA-fed animals. Coherently to the reduced stroke size, functional BBB integrity and occludin endothelial expression were maintained by high ALA supplementation. Additionally, ALA reduced endothelial activation and thus recruitment and activation of macrophages and resident microglia. Finally, high ALA diet reduced the expression of BBB-degrading and neurotoxic MMP-3 and MMP-9. CONCLUSIONS We demonstrate the beneficial effects of a clinically relevant and feasible dietary intervention with a safe and readily available compound in the setting of stroke. The protective effects observed with ALA supplementation may relate to blunting of inflammation and might pave the way for novel stroke treatments.
Collapse
Affiliation(s)
- Nicole R Bonetti
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Lisa Pasterk
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Sara Gobbato
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Ana Vukolic
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Seyed Soheil Saeedi Saravi
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Bernd Coester
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Carla Horvath
- Institute of Food, Nutrition and Health, Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Elena Osto
- University and University Hospital Zurich, Institute of Clinical Chemistry, Zurich, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
54
|
Liberale L, Ministrini S, Carbone F, Camici GG, Montecucco F. Cytokines as therapeutic targets for cardio- and cerebrovascular diseases. Basic Res Cardiol 2021; 116:23. [PMID: 33770265 PMCID: PMC7997823 DOI: 10.1007/s00395-021-00863-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Despite major advances in prevention and treatment, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. In this context, inflammation is involved in the chronic process leading atherosclerotic plaque formation and its complications, as well as in the maladaptive response to acute ischemic events. For this reason, modulation of inflammation is nowadays seen as a promising therapeutic strategy to counteract the burden of cardio- and cerebrovascular disease. Being produced and recognized by both inflammatory and vascular cells, the complex network of cytokines holds key functions in the crosstalk of these two systems and orchestrates the progression of atherothrombosis. By binding to membrane receptors, these soluble mediators trigger specific intracellular signaling pathways eventually leading to the activation of transcription factors and a deep modulation of cell function. Both stimulatory and inhibitory cytokines have been described and progressively reported as markers of disease or interesting therapeutic targets in the cardiovascular field. Nevertheless, cytokine inhibition is burdened by harmful side effects that will most likely prevent its chronic use in favor of acute administrations in well-selected subjects at high risk. Here, we summarize the current state of knowledge regarding the modulatory role of cytokines on atherosclerosis, myocardial infarction, and stroke. Then, we discuss evidence from clinical trials specifically targeting cytokines and the potential implication of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland.
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Stefano Ministrini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
55
|
Hasan A, Al-Ozairi E, Al-Baqsumi Z, Ahmad R, Al-Mulla F. Cellular and Humoral Immune Responses in Covid-19 and Immunotherapeutic Approaches. Immunotargets Ther 2021; 10:63-85. [PMID: 33728277 PMCID: PMC7955763 DOI: 10.2147/itt.s280706] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (Covid-19), caused by the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can range in severity from asymptomatic to severe/critical disease. SARS-CoV-2 uses angiotensin-converting enzyme 2 to infect cells leading to a strong inflammatory response, which is most profound in patients who progress to severe Covid-19. Recent studies have begun to unravel some of the differences in the innate and adaptive immune response to SARS-CoV-2 in patients with different degrees of disease severity. These studies have attributed the severe form of Covid-19 to a dysfunctional innate immune response, such as a delayed and/or deficient type I interferon response, coupled with an exaggerated and/or a dysfunctional adaptive immunity. Differences in T-cell (including CD4+ T-cells, CD8+ T-cells, T follicular helper cells, γδ-T-cells, and regulatory T-cells) and B-cell (transitional cells, double-negative 2 cells, antibody-secreting cells) responses have been identified in patients with severe disease compared to mild cases. Moreover, differences in the kinetic/titer of neutralizing antibody responses have been described in severe disease, which may be confounded by antibody-dependent enhancement. Importantly, the presence of preexisting autoantibodies against type I interferon has been described as a major cause of severe/critical disease. Additionally, priorVaccine and multiple vaccine exposure, trained innate immunity, cross-reactive immunity, and serological immune imprinting may all contribute towards disease severity and outcome. Several therapeutic and preventative approaches have been under intense investigations; these include vaccines (three of which have passed Phase 3 clinical trials), therapeutic antibodies, and immunosuppressants.
Collapse
Affiliation(s)
- Amal Hasan
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Ebaa Al-Ozairi
- Clinical Research Unit, Medical Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
- Department of Medicine, Faculty of Medicine, Jabriya, Kuwait City, Kuwait
| | - Zahraa Al-Baqsumi
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Functional Genomics, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| |
Collapse
|
56
|
Valois MV, de Oliveira C, Lapa AJ, Souccar C, Oliva MLV. Bauhinia Protease Inhibitors Attenuate Gastric Ulcer by Blocking Neutrophil Enzymes. PLANTA MEDICA 2021; 87:169-176. [PMID: 32663895 DOI: 10.1055/a-1202-4799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Proteases play a pivotal role in many signaling pathways; inhibitors of well-established proteases have shown a substantial therapeutic success. This study aimed to examine the in vivo effects of 3 protease inhibitors isolated from Bauhinia species: i) Bauhinia mollis elastase inhibitor, which blocks human neutrophil elastase (Kiapp 2.8 nM) and cathepsin G (Kiapp 1.0 nM) activities; ii) Bauhinia mollis trypsin inhibitor, a trypsin inhibitor (Kiapp 5.0 nM); and iii) Bauhinia bauhinioides cruzipain inhibitor, which inhibits elastase (Kiapp 2.6 nM), cathepsin G (Kiapp 160.0 nM), and the cysteine proteases cathepsin L (Kiapp 0.2 nM). Bauhinia bauhinioides cruzipain inhibitor, Bauhinia mollis elastase inhibitor, and Bauhinia mollis trypsin inhibitor were isolated using acetone and ammonium sulfate fractionations, DEAE-Sephadex, trypsin-Sepharose, and Resource-Q chromatographies. Mice and rats were treated intraperitoneally with 1 dose of inhibitor; gastric mucosal lesions were induced by cold-restraint stress. Oral pretreatment of mice with Bauhinia mollis elastase inhibitor or Bauhinia mollis trypsin inhibitor (1 - 10 mg/kg) did not show anti-ulcer effect, while Bauhinia bauhinioides cruzipain inhibitor (0.1 - 1.0 mg/kg) produced a similar reduction of the index of mucosal damage at all effective doses (30 to 33% < control). In rats at doses lower than those used in mice, Bauhinia mollis elastase inhibitor and Bauhinia bauhinioides cruzipain inhibitor reduced the index of mucosal damage by 66% and 54% of controls, respectively. The results indicate a protective effect against gastric mucosal lesions associated with elastase inhibition but not inhibition of trypsin activities. Moreover, the lack of Bauhinia mollis elastase inhibitor efficacy observed in mice may possibly be related to the reported structural differences of elastase in mice and rats.
Collapse
Affiliation(s)
- Mayara Vioto Valois
- Department of Biochemistry, Universidade Federal de São Paulo, Escola Paulista de Medicina, SP, Brazil
| | - Cleide de Oliveira
- Department of Biochemistry, Universidade Federal de São Paulo, Escola Paulista de Medicina, SP, Brazil
| | - Antonio José Lapa
- Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, SP, Brazil
- Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Caden Souccar
- Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, SP, Brazil
| | - Maria Luiza Vilela Oliva
- Department of Biochemistry, Universidade Federal de São Paulo, Escola Paulista de Medicina, SP, Brazil
| |
Collapse
|
57
|
Liberale L, Montecucco F, Schwarz L, Lüscher TF, Camici GG. Inflammation and cardiovascular diseases: lessons from seminal clinical trials. Cardiovasc Res 2021; 117:411-422. [PMID: 32666079 DOI: 10.1093/cvr/cvaa211] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammation has been long regarded as a key contributor to atherosclerosis. Inflammatory cells and soluble mediators play critical roles throughout arterial plaque development and accordingly, targeting inflammatory pathways effectively reduces atherosclerotic burden in animal models of cardiovascular (CV) diseases. Yet, clinical translation often led to inconclusive or even contradictory results. The Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) followed by the Colchicine Cardiovascular Outcomes Trial (COLCOT) were the first two randomized clinical trials to convincingly demonstrate the effectiveness of specific anti-inflammatory treatments in the field of CV prevention, while other phase III trials-including the Cardiovascular Inflammation Reduction Trial one using methotrexate-were futile. This manuscript reviews the main characteristics and findings of recent anti-inflammatory Phase III trials in cardiology and discusses their similarities and differences in order to get further insights into the contribution of specific inflammatory pathways on CV outcomes. CANTOS and COLCOT demonstrated efficacy of two anti-inflammatory drugs (canakinumab and colchicine, respectively) in the secondary prevention of major adverse CV events (MACE) thus providing the first confirmation of the involvement of a specific inflammatory pathway in human atherosclerotic CV disease (ASCVD). Also, they highlighted the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome-related pathway as an effective therapeutic target to blunt ASCVD. In contrast, other trials interfering with a number of inflammasome-independent pathways failed to provide benefit. Lastly, all anti-inflammatory trials underscored the importance of balancing the risk of impaired host defence with an increase in infections and the prevention of MACE in CV patients with residual inflammatory risk.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Lena Schwarz
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8092 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8092, Zurich, Switzerland
| |
Collapse
|
58
|
Ginsburg I, Fibach E. Polycations and polyanions in SARS-CoV-2 infection. Med Hypotheses 2021; 146:110470. [PMID: 33412501 PMCID: PMC7837050 DOI: 10.1016/j.mehy.2020.110470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/17/2020] [Indexed: 01/11/2023]
Abstract
We hypothesize that polycations, such as nuclear histones, released by neutrophils COVID-19 aggravate COVID-19 by multiple mechanisms: (A) Neutralization of the electrostatic repulsion between the virus particles and the cell membrane, thereby enhancing receptor-mediated entry. (B) Binding to the virus particles, thereby inducing opsonin-mediated endocytosis. (C) Adding to the cytotoxicity, in conjunction with oxidants, cytokines and other pro-inflammatory substances secreted by cells of the innate immunity system. These effects may be alleviated by the administration of negatively charged polyanions such as heparins and heparinoids.
Collapse
Affiliation(s)
- I Ginsburg
- The Hebrew University - Hadassah School of Medicine, The Faculty of Dental Medicine, The Ein-Kerm Campus, Jerusalem, Israel
| | - E Fibach
- The Hebrew University - Hadassah School of Medicine, Department of Hematology, The Ein-Kerm Campus, Jerusalem, Israel.
| |
Collapse
|
59
|
Wang LL, Song YP, Mi JH, Ding ML. Peptidyl arginine deiminase 4 and its potential role in Alzheimer's disease. Med Hypotheses 2020; 146:110466. [PMID: 33412502 DOI: 10.1016/j.mehy.2020.110466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) is the main cause of dementia, and its pathogenesis is still not clear. Peptidyl arginine deiminases 4(PAD4) as one of the important members of PAD family, is the only protein with nuclear transfer function, it can regulate the expression of many proteins through citrullinating histone. PAD4 can also interact with many transcription factors, involved in regulating gene expression. PAD4 expression is closely related to the inflammatory factors secreted, cell autophagy, tumorigenesis and other neurodegenerative diseases. More importantly, PAD4 and its citrullinated protein were found in cortical and hippocampal neurons of AD patients. To study the expression and regulatory pathway of PAD4 in vivo and in vitro experiments on AD may be of helpful to elucidate the pathogenesis of AD. Meanwhile, detection of anti-citrullinated antibody will have potential value as novel biomarkers of AD.
Collapse
Affiliation(s)
- Li-Ling Wang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 201100, China
| | - Ye-Ping Song
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 201100, China
| | - Jian-Hua Mi
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 201100, China
| | - Meng-Lei Ding
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tongji University, 200120, China.
| |
Collapse
|
60
|
Flohé L. Looking Back at the Early Stages of Redox Biology. Antioxidants (Basel) 2020; 9:E1254. [PMID: 33317108 PMCID: PMC7763103 DOI: 10.3390/antiox9121254] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/12/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The beginnings of redox biology are recalled with special emphasis on formation, metabolism and function of reactive oxygen and nitrogen species in mammalian systems. The review covers the early history of heme peroxidases and the metabolism of hydrogen peroxide, the discovery of selenium as integral part of glutathione peroxidases, which expanded the scope of the field to other hydroperoxides including lipid hydroperoxides, the discovery of superoxide dismutases and superoxide radicals in biological systems and their role in host defense, tissue damage, metabolic regulation and signaling, the identification of the endothelial-derived relaxing factor as the nitrogen monoxide radical (more commonly named nitric oxide) and its physiological and pathological implications. The article highlights the perception of hydrogen peroxide and other hydroperoxides as signaling molecules, which marks the beginning of the flourishing fields of redox regulation and redox signaling. Final comments describe the development of the redox language. In the 18th and 19th century, it was highly individualized and hard to translate into modern terminology. In the 20th century, the redox language co-developed with the chemical terminology and became clearer. More recently, the introduction and inflationary use of poorly defined terms has unfortunately impaired the understanding of redox events in biological systems.
Collapse
Affiliation(s)
- Leopold Flohé
- Dipartimento di Medicina Molecolare, Università degli Studi di Padova, v.le G. Colombo 3, 35121 Padova, Italy;
- Departamento de Bioquímica, Universidad de la República, Avda. General Flores 2125, 11800 Montevideo, Uruguay
| |
Collapse
|
61
|
Cao J, Su J, An M, Yang Y, Zhang Y, Zuo J, Zhang N, Zhao Y. Novel DEK-Targeting Aptamer Delivered by a Hydrogel Microneedle Attenuates Collagen-Induced Arthritis. Mol Pharm 2020; 18:305-316. [PMID: 33253580 DOI: 10.1021/acs.molpharmaceut.0c00954] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
DEK protein is critical to the formation of neutrophil extracellular traps (NETs) in rheumatoid arthritis (RA). Blocking DEK using the aptamer DTA via articular injection has been shown to have robust anti-inflammatory efficacy in a previous study. However, DTA is prone to nuclease degradation and renal clearance in vivo. RA is a systemic disease that involves multiple joints, and local injection is impractical in clinical settings. In this study, DTA was modified with methoxy groups on all deoxyribose sugar units and inverted deoxythymidine on the 3' end (DTA4) to enhance its stability against nuclease. DTA4 is stable for 72 h in 90% mouse serum and maintains a high binding affinity to DEK. DTA4 effectively inhibits the formation of NETs and the migration of HUVECs in vitro. DTA4 was then modified with cholesterol on its 5' end to form DTA6. DTA6 dramatically reduces DEK expression in inflammatory RAW264.7 cells. A hydrogel microneedle (hMN) was then fabricated for the transdermal delivery of DTA6. The hMN maintains morphological integrity after absorbing the aptamer solution, effectively pierces the skin, and rapidly releases DTA6 into the dermis. The DTA6-loaded hMN significantly attenuates inflammation and protects joints from cartilage/bone erosion in collagen-induced arthritis (CIA) mice.
Collapse
Affiliation(s)
- Jian Cao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Jingjing Su
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Mengchen An
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Yang Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Yi Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Jing Zuo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou 450001, Henan, P. R. China.,Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Henan Province, Zhengzhou 450001, Henan, P. R. China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, P. R. China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou 450001, Henan, P. R. China.,Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Henan Province, Zhengzhou 450001, Henan, P. R. China
| |
Collapse
|
62
|
Scandolara TB, da Silva JC, Malanowski J, de Oliveira JA, Rech D, Panis C. Anti-neutrophil antibodies (anti-MPO-ANCAs) are associated with poor prognosis in breast cancer patients. Immunobiology 2020; 225:152011. [PMID: 33130517 DOI: 10.1016/j.imbio.2020.152011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/20/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022]
Abstract
Anti-neutrophil antibodies are capable of activating neutrophils in sterile environments, releasing extracellular traps containing myeloperoxidase (MPO) and anti-MPO antibodies (MPO-ANCAs or anti-MPO-ANCAs), which have been implicated in the pathogenesis of several diseases. The present study evaluated systemic and tumor tissue levels of anti-MPO-ANCAs breast cancer patients, and its relation to clinicopathological characteristics. Anti-MPO-ANCAs were measured in serum and tissue samples of 150 patients by enzyme-linked immunoassay. Samples were pooled according to clinicopathological characteristics of patients. Higher anti-MPO-ANCAs levels were detected in groups presenting negative clinicopathological characteristics, such as high histological grade tumors and risk factors such as body mass index, menopausal status and early onset at diagnosis. The present data highlights anti-MPO-ANCAs as associated to poor prognosis in breast cancer, a role beyond its actually discussed role in autoimmunity and vasculitis.
Collapse
Affiliation(s)
- Thalita Basso Scandolara
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Federal University of Rio de Janeiro, Brazil; Post-graduation Program of Health-Applied Sciences, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil
| | - Janaína Carla da Silva
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Post-graduation Program of Health-Applied Sciences, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil
| | - Jéssica Malanowski
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil
| | - Janoário Athanázio de Oliveira
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Francisco Beltrão Cancer Hospital (Ceonc), Francisco Beltrão, Paraná, Brazil
| | - Daniel Rech
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Post-graduation Program of Health-Applied Sciences, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Francisco Beltrão Cancer Hospital (Ceonc), Francisco Beltrão, Paraná, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Post-graduation Program of Health-Applied Sciences, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil.
| |
Collapse
|
63
|
Age-related cerebral small vessel disease and inflammaging. Cell Death Dis 2020; 11:932. [PMID: 33127878 PMCID: PMC7603301 DOI: 10.1038/s41419-020-03137-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
The continued increase in global life expectancy predicts a rising prevalence of age-related cerebral small vessel diseases (CSVD), which requires a better understanding of the underlying molecular mechanisms. In recent years, the concept of "inflammaging" has attracted increasing attention. It refers to the chronic sterile low-grade inflammation in elderly organisms and is involved in the development of a variety of age-related chronic diseases. Inflammaging is a long-term result of chronic physiological stimulation of the immune system, and various cellular and molecular mechanisms (e.g., cellular senescence, immunosenescence, mitochondrial dysfunction, defective autophagy, metaflammation, gut microbiota dysbiosis) are involved. With the deepening understanding of the etiological basis of age-related CSVD, inflammaging is considered to play an important role in its occurrence and development. One of the most critical pathophysiological mechanisms of CSVD is endothelium dysfunction and subsequent blood-brain barrier (BBB) leakage, which gives a clue in the identification of the disease by detecting circulating biological markers of BBB disruption. The regional analysis showed blood markers of vascular inflammation are often associated with deep perforating arteriopathy (DPA), while blood markers of systemic inflammation appear to be associated with cerebral amyloid angiopathy (CAA). Here, we discuss recent findings in the pathophysiology of inflammaging and their effects on the development of age-related CSVD. Furthermore, we speculate the inflammaging as a potential target for future therapeutic interventions to delay or prevent the progression of the age-related CSVD.
Collapse
|
64
|
da Silva RF, Baptista D, Roth A, Miteva K, Burger F, Vuilleumier N, Carbone F, Montecucco F, Mach F, J. Brandt K. Anti-Apolipoprotein A-1 IgG Influences Neutrophil Extracellular Trap Content at Distinct Regions of Human Carotid Plaques. Int J Mol Sci 2020; 21:7721. [PMID: 33086507 PMCID: PMC7588926 DOI: 10.3390/ijms21207721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neutrophils accumulate in atherosclerotic plaques. Neutrophil extracellular traps (NET) were recently identified in experimental atherosclerosis and in complex human lesions. However, not much is known about the NET marker citrullinated histone-3 (H3Cit) expression and functionality in human carotid plaques. Moreover, the association between the proatherosclerotic autoantibody anti-apolipoprotein A-1 (anti-ApoA-1 IgG) and NET has never been investigated. METHODS Atherosclerotic plaques have been obtained from 36 patients with severe carotid stenosis that underwent carotid endarterectomy for severe carotid stenosis. Samples were sectioned into upstream and downstream regions from the same artery segment. Plaque composition and expression of NET markers neutrophil elastase (NE) and H3Cit were quantified by immunohistochemistry. H3Cit expression and function was evaluated by immunofluorescence and confocal analysis in a subset of patients. RESULTS Pathological features of vulnerable phenotypes were exacerbated in plaques developed at downstream regions, including higher accumulation of neutrophils and enhanced expression of NE and H3Cit, as compared to plaques from upstream regions. The H3Cit signal was also more intense in downstream regions, with significant extracellular distribution in spaces outside of neutrophils. The percentage of H3Cit colocalization with CD66b (neutrophils) was markedly lower in downstream portions of carotid plaques, confirming the extrusion of NET in this region. In agreement, the maximum distance of the H3Cit signal from neutrophils, extrapolated from vortex distance calculation in all possible directions, was also higher in downstream plaques. The serum anti-ApoA-1index positively correlated with the expression of H3Cit in downstream segments of plaques. Expression of the H3Cit signal outside of neutrophils and H3Cit maximal distance from CD66b-positive cells increased in plaques from serum positive anti-ApoA-1 patients compared with serum negative patients. CONCLUSION NET elements are differentially expressed in upstream versus downstream regions of human carotid plaques and may be influenced by circulating levels of anti-ApoA-1 IgG. These findings could warrant the investigation of NET elements as potential markers of vulnerability.
Collapse
Affiliation(s)
- Rafaela F. da Silva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Nicolas Vuilleumier
- Department of Diagnostics, Division of Laboratory Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland;
- Department of Medical Specialities, Division of Laboratory Medicine, Faculty of Medicine, 1211 Geneva, Switzerland
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV n6, 16132 Genoa, Italy; (F.C.); (F.M.)
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Largo Rosanna Benzi n10, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV n6, 16132 Genoa, Italy; (F.C.); (F.M.)
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Largo Rosanna Benzi n10, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Karim J. Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| |
Collapse
|
65
|
Zuo Y, Yalavarthi S, Gockman K, Madison JA, Gudjonsson JE, Kahlenberg JM, Joseph McCune W, Bockenstedt PL, Karp DR, Knight JS. Anti-Neutrophil Extracellular Trap Antibodies and Impaired Neutrophil Extracellular Trap Degradation in Antiphospholipid Syndrome. Arthritis Rheumatol 2020; 72:2130-2135. [PMID: 32729667 DOI: 10.1002/art.41460] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The release of neutrophil extracellular traps (NETs) by hyperactive neutrophils has recently been recognized to play an important role in antiphospholipid syndrome (APS). This study was undertaken to evaluate autoantibodies targeting NETs in patients with primary APS, and to determine their potential functions and clinical associations. METHODS We measured global anti-NET activity in 76 patients with primary APS, 23 patients with systemic lupus erythematosus without antiphospholipid antibodies (aPL), 11 patients with a history of unprovoked venous thrombosis without aPL, and 44 healthy controls. The ability of APS sera to degrade NETs was also assessed. RESULTS We found markedly elevated levels of anti-NET IgG and IgM in patients with primary APS compared with healthy controls (for IgG, mean ± SD optical density 0.55 ± 0.34 versus 0.33 ± 0.17; for IgM, mean ± SD optical density 0.76 ± 0.51 versus 0.26 ± 0.23). This anti-NET activity did not correlate with levels of traditional aPL and was relatively stable over time. Mechanistically, anti-NET antibodies (especially of the IgG isotype) impaired the ability of patient sera to degrade NETs (r = 0.4, P = 0.003). Levels of anti-NET IgM inversely correlated with complement C4 (r = 0.4, P = 0.019). Clinically, anti-NET antibodies associated with certain APS clinical manifestations, and in particular recurrent venous thrombosis (odds ratio 4.3; P = 0.002). Interestingly, anti-NET antibody levels also appeared to be associated with unprovoked venous thrombosis in the general population (for IgM, mean ± SD optical density 0.67 ± 0.34 versus 0.26 ± 0.23). CONCLUSION Our data indicate high levels of anti-NET antibodies in patients with primary APS, which may impair NET clearance and activate the complement cascade. These findings may ultimately enable more effective risk stratification.
Collapse
Affiliation(s)
- Yu Zuo
- University of Michigan, Ann Arbor
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Plana E, Oto J, Medina P, Fernández-Pardo Á, Miralles M. Novel contributions of neutrophils in the pathogenesis of abdominal aortic aneurysm, the role of neutrophil extracellular traps: A systematic review. Thromb Res 2020; 194:200-208. [PMID: 32788119 DOI: 10.1016/j.thromres.2020.07.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a vascular disorder caused by a dilatation of the aortic diameter that can be potentially lethal in case of rupture. Molecular mechanisms underlying the development of AAA are complex and not completely understood. However, inflammation plays a pivotal role in AAA development. Infiltration of inflammatory cells, especially macrophages, has been widely observed in lesion areas. Nevertheless, neutrophils have been gaining importance in the context of AAA. The release of neutrophil extracellular traps (NETs), extracellular structures formed by DNA, histones, granular and cytoplasmic proteins, is a recently discovered mechanism of neutrophil activation that can be triggered by endogenous inflammatory stimulus. The number of studies about the role of NETs in several vascular diseases like thrombosis and atherosclerosis has increased in last decade. However, its role in AAA has been scarcely analysed. The aim of this review is to deepen in the latest advances concerning the potential role neutrophils and especially NETs in AAA development.
Collapse
Affiliation(s)
- Emma Plana
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, Spain; Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain.
| | - Julia Oto
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Pilar Medina
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Álvaro Fernández-Pardo
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Manuel Miralles
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, Spain; Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain; Department of Surgery, University of Valencia, Valencia, Spain
| |
Collapse
|
67
|
Bonaventura A, Carbone F, Vecchié A, Meessen J, Ferraris S, Beck E, Keim R, Minetti S, Elia E, Ferrara D, Ansaldo AM, Novelli D, Caironi P, Latini R, Montecucco F. The role of resistin and myeloperoxidase in severe sepsis and septic shock: Results from the ALBIOS trial. Eur J Clin Invest 2020; 50:e13333. [PMID: 32585739 DOI: 10.1111/eci.13333] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory biomarkers are useful in detecting patients with sepsis. The prognostic role of resistin and myeloperoxidase (MPO) has been investigated in sepsis. MATERIALS AND METHODS Plasma resistin and MPO were measured on days 1, 2 and 7 in 957 patients enrolled in the Albumin Italian Outcome Sepsis (ALBIOS) trial. The association between resistin and MPO levels on day 1, 2 and 7 and 90-day mortality was assessed. RESULTS Plasma resistin and MPO concentrations were higher at day 1 and decreased until day 7. Both biomarkers were positively correlated with each other and with physiological parameters. Higher levels of resistin and MPO on day 1 were associated with the development of new organ failures. Patients experiencing death at 90 days showed higher levels of resistin and MPO compared with survivors. At day 1, only MPO in the 4th quartile (Q4), but not resistin, was found to predict 90-day death (adjusted hazard ratio [aHR] 1.55 vs Q1). At day 2, resistin in the Q3 and Q4 predicted a > 40% increase in mortality as also did MPO in the Q4. On day 7, Q4 resistin was able to predict 90-day mortality, while all quartiles of MPO were not. CONCLUSIONS High levels of MPO, but not of resistin, on day 1 were able to predict 90-day mortality. These findings may either suggest that early hyper-activation of neutrophils is detrimental in patients with sepsis or reflect the burden of the inflammatory process caused by sepsis. Further studies are warranted to deepen these aspects (ALBIOS ClinicalTrials.gov Identifier: NCT00707122).
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA, 23298, USA
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Genoa, Italy
| | - Alessandra Vecchié
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA, 23298, USA
| | - Jennifer Meessen
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | - Roberto Keim
- UOC Anestesia Rianimazione e Terapia Intensiva - ASST Bergamo Est - Ospedale Bolognini di Seriate, Seriate, Italy
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Edoardo Elia
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Daniele Ferrara
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Anna Maria Ansaldo
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Deborah Novelli
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Pietro Caironi
- SCDU Anestesia e Rianimazione, Azienda Ospedaliero-Universitaria S. Luigi Gonzaga, Orbassano, Italy
- Dipartimento di Oncologia, Università degli Studi di Torino, Turin, Italy
| | - Roberto Latini
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
68
|
Liberale L, Bertolotto M, Minetti S, Contini P, Verzola D, Ameri P, Ghigliotti G, Pende A, Camici GG, Carbone F, Montecucco F. Recombinant Tissue Plasminogen Activator (r-tPA) Induces In-Vitro Human Neutrophil Migration via Low Density Lipoprotein Receptor-Related Protein 1 (LRP-1). Int J Mol Sci 2020; 21:7014. [PMID: 32977685 PMCID: PMC7582901 DOI: 10.3390/ijms21197014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023] Open
Abstract
Thrombolysis is the gold standard treatment for acute ischemic stroke. Besides its fibrinolytic role, recombinant tissue plasminogen activator (r-tPA) holds several non-fibrinolytic functions. Here, we investigated the potential role of r-tPA on human primary neutrophil migration in vitro. By means of modified Boyden chamber migration assay and checkerboard analysis we showed a dose-dependent chemotactic effect of r-TPA with a maximum effect reached by 0.03 mg/mL (0.003-1 mg/mL). Pre-incubation with MAP kinases inhibitors allowed the identification of PI3K/Akt, but not ERK1/2 as the intracellular pathway mediating the observed effects. Furthermore, by means of real-time PCR, immunocytochemistry and cytofluorimetry we demonstrated that the r-tPA receptor low density lipoprotein receptor-related protein 1 (LRP-1) is synthetized and expressed by neutrophils in response to r-tPA and TNF-α. Inhibition of LRP-1 by receptor-associated protein (RAP), prevented r-tPA-mediated F-actin polymerization, migration and signal through Akt but not ERK1/2. Lastly, also neutrophil degranulation in response to r-tPA seems to be mediated by LRP-1 under adhesion conditions. In conclusion, we show that r-tPA induces neutrophil chemotaxis through LRP-1/Akt pathway. Blunting r-tPA-mediated neutrophil activation might be beneficial as an adjuvant therapy to thrombolysis in this setting.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; (L.L.); (G.G.C.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Paola Contini
- Clinical Immunology, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy;
| | - Daniela Verzola
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy;
| | - Pietro Ameri
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Laboratory of Cardiovascular Biology, IRCCS Ospedale Policlinico San Martino & Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Giorgio Ghigliotti
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Laboratory of Cardiovascular Biology, IRCCS Ospedale Policlinico San Martino & Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Aldo Pende
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Clinic of Emergency Medicine, Department of Emergency Medicine, University of Genoa, 16126 Genoa, Italy
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; (L.L.); (G.G.C.)
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
| |
Collapse
|
69
|
Nargiz Medical Center, Baku, Azerbaijan, Gahramanova M, ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Ukraine;, Khalilova I, Department of Life Sciences & Center for Cell Pathology Research, Khazar University, Baku, Azerbaijan;, Omarov A, Department of Life Sciences & Center for Cell Pathology Research, Khazar University, Baku, Azerbaijan;, Susak Y, Bogomolets National Medical University, Kyiv, Ukraine, Rudyk M, ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Ukraine;, Skivka L, ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Ukraine;. Anti-inflammatory and hepatoprotective effects of polyherbal composition in patients with chronic cholecystitis. UKRAINIAN BIOCHEMICAL JOURNAL 2020; 92:77-84. [DOI: 10.15407/ubj92.04.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
70
|
Middleton EA, He XY, Denorme F, Campbell RA, Ng D, Salvatore SP, Mostyka M, Baxter-Stoltzfus A, Borczuk AC, Loda M, Cody MJ, Manne BK, Portier I, Harris ES, Petrey AC, Beswick EJ, Caulin AF, Iovino A, Abegglen LM, Weyrich AS, Rondina MT, Egeblad M, Schiffman JD, Yost CC. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020; 136:1169-1179. [PMID: 32597954 PMCID: PMC7472714 DOI: 10.1182/blood.2020007008] [Citation(s) in RCA: 1075] [Impact Index Per Article: 215.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/25/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 affects millions of patients worldwide, with clinical presentation ranging from isolated thrombosis to acute respiratory distress syndrome (ARDS) requiring ventilator support. Neutrophil extracellular traps (NETs) originate from decondensed chromatin released to immobilize pathogens, and they can trigger immunothrombosis. We studied the connection between NETs and COVID-19 severity and progression. We conducted a prospective cohort study of COVID-19 patients (n = 33) and age- and sex-matched controls (n = 17). We measured plasma myeloperoxidase (MPO)-DNA complexes (NETs), platelet factor 4, RANTES, and selected cytokines. Three COVID-19 lung autopsies were examined for NETs and platelet involvement. We assessed NET formation ex vivo in COVID-19 neutrophils and in healthy neutrophils incubated with COVID-19 plasma. We also tested the ability of neonatal NET-inhibitory factor (nNIF) to block NET formation induced by COVID-19 plasma. Plasma MPO-DNA complexes increased in COVID-19, with intubation (P < .0001) and death (P < .0005) as outcome. Illness severity correlated directly with plasma MPO-DNA complexes (P = .0360), whereas Pao2/fraction of inspired oxygen correlated inversely (P = .0340). Soluble and cellular factors triggering NETs were significantly increased in COVID-19, and pulmonary autopsies confirmed NET-containing microthrombi with neutrophil-platelet infiltration. Finally, COVID-19 neutrophils ex vivo displayed excessive NETs at baseline, and COVID-19 plasma triggered NET formation, which was blocked by nNIF. Thus, NETs triggering immunothrombosis may, in part, explain the prothrombotic clinical presentations in COVID-19, and NETs may represent targets for therapeutic intervention.
Collapse
Affiliation(s)
- Elizabeth A Middleton
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY
| | | | - Robert A Campbell
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - David Ng
- Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY
| | - Steven P Salvatore
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Maria Mostyka
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
| | - Amelia Baxter-Stoltzfus
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
| | - Alain C Borczuk
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Mark J Cody
- Molecular Medicine Program and
- Department of Pediatrics and
| | | | | | - Estelle S Harris
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Aaron C Petrey
- Molecular Medicine Program and
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Ellen J Beswick
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | | | - Anthony Iovino
- Department of Pediatrics and
- PEEL Therapeutics, Inc, Salt Lake City, UT; and
| | - Lisa M Abegglen
- Department of Pediatrics and
- PEEL Therapeutics, Inc, Salt Lake City, UT; and
| | - Andrew S Weyrich
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
- Department of Internal Medicine and
- Geriatric Research, Education, and Clinical Center (GRECC), George E. Wahlen Veterans Affairs (VA) Medical Center, Salt Lake City, UT
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY
| | - Joshua D Schiffman
- Molecular Medicine Program and
- Department of Pediatrics and
- PEEL Therapeutics, Inc, Salt Lake City, UT; and
| | | |
Collapse
|
71
|
Liberale L, Montecucco F, Tardif JC, Libby P, Camici GG. Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease. Eur Heart J 2020; 41:2974-2982. [PMID: 32006431 PMCID: PMC7453832 DOI: 10.1093/eurheartj/ehz961] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/10/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
The ongoing worldwide increase in life expectancy portends a rising prevalence of age-related cardiovascular (CV) diseases in the coming decades that demands a deeper understanding of their molecular mechanisms. Inflammation has recently emerged as an important contributor for CV disease development. Indeed, a state of chronic sterile low-grade inflammation characterizes older organisms (also known as inflamm-ageing) and participates pivotally in the development of frailty, disability, and most chronic degenerative diseases including age-related CV and cerebrovascular afflictions. Due to chronic activation of inflammasomes and to reduced endogenous anti-inflammatory mechanisms, inflamm-ageing contributes to the activation of leucocytes, endothelial, and vascular smooth muscle cells, thus accelerating vascular ageing and atherosclerosis. Furthermore, inflamm-ageing promotes the development of catastrophic athero-thrombotic complications by enhancing platelet reactivity and predisposing to plaque rupture and erosion. Thus, inflamm-ageing and its contributors or molecular mediators might furnish targets for novel therapeutic strategies that could promote healthy ageing and conserve resources for health care systems worldwide. Here, we discuss recent findings in the pathophysiology of inflamm-ageing, the impact of these processes on the development of age-related CV diseases, results from clinical trials targeting its components and the potential implementation of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, v.le Benedetto XV 10, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, L.go Rosanna Benzi 10, 16132 Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, v.le Benedetto XV 10, 16132 Genoa, Italy
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montreal, Rue Bélanger 5000, Montreal, QC H1T 1C8, Canada
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Francis Street 75, Boston, MA 02115, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
72
|
Zhang S, Zhang Q, Wang F, Guo X, Liu T, Zhao Y, Gu B, Chen H, Li Y. Hydroxychloroquine inhibiting neutrophil extracellular trap formation alleviates hepatic ischemia/reperfusion injury by blocking TLR9 in mice. Clin Immunol 2020; 216:108461. [PMID: 32437924 DOI: 10.1016/j.clim.2020.108461] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 01/30/2023]
Abstract
Hepatic ischemia/reperfusion (I/R) injury may arise after partial hepatectomy and liver transplantation. Neutrophil extracellular traps (NETs) were involved in hepatic I/R injury. This study tested the hypothesis that blocking NETs formation could be a potential therapeutic target against hepatic I/R injury. NETs were excessively formed within liver and in serum of I/R mice models and were testified to be an independent contributor to hepatic I/R injury. Hydroxychloroquine (HCQ) alleviated hepatic I/R injury by inhibiting NETs formation in SCID and c57BL/6 mice models. In vitro, HCQ inhibited neutrophils to form NETs at a concentration of 100 μg/ml. CpG-ODN reversed the effect of HCQ inhibiting NETs formation. HCQ inhibited PAD4 and Rac2 expressions by blocking TLR9. NETs are essential contributors to hepatic I/R injury. HCQ blocking TLR9 protects against hepatic I/R injury by inhibiting NETs formation, which may suggest utility of HCQ or other TLR9 agonists for preventing hepatic I/R injury in clinical practices.
Collapse
Affiliation(s)
- Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Key Laboratory of Digestive System Tumors of Gansu Province, China
| | - Qiuyue Zhang
- the Second Clinical Medical College, Lanzhou University, China
| | - Furong Wang
- Department of Pathology, Lanzhou University Second Hospital, China
| | - Xuehui Guo
- Gansu Emergency Medical Aid Center, China
| | - Tao Liu
- Lanzhou University Second Hospital, Key Laboratory of Digestive System Tumors of Gansu Province, China
| | - Yang Zhao
- Lanzhou University Second Hospital, Key Laboratory of Digestive System Tumors of Gansu Province, China
| | - Baohong Gu
- the Second Clinical Medical College, Lanzhou University, China
| | - Hao Chen
- Lanzhou University Second Hospital, Key Laboratory of Digestive System Tumors of Gansu Province, China
| | - Yumin Li
- Lanzhou University Second Hospital, Key Laboratory of Digestive System Tumors of Gansu Province, China.
| |
Collapse
|
73
|
Arisan ED, Uysal-Onganer P, Lange S. Putative Roles for Peptidylarginine Deiminases in COVID-19. Int J Mol Sci 2020; 21:E4662. [PMID: 32629995 PMCID: PMC7370447 DOI: 10.3390/ijms21134662] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 02/06/2023] Open
Abstract
Peptidylarginine deiminases (PADs) are a family of calcium-regulated enzymes that are phylogenetically conserved and cause post-translational deimination/citrullination, contributing to protein moonlighting in health and disease. PADs are implicated in a range of inflammatory and autoimmune conditions, in the regulation of extracellular vesicle (EV) release, and their roles in infection and immunomodulation are known to some extent, including in viral infections. In the current study we describe putative roles for PADs in COVID-19, based on in silico analysis of BioProject transcriptome data (PRJNA615032 BioProject), including lung biopsies from healthy volunteers and SARS-CoV-2-infected patients, as well as SARS-CoV-2-infected, and mock human bronchial epithelial NHBE and adenocarcinoma alveolar basal epithelial A549 cell lines. In addition, BioProject Data PRJNA631753, analysing patients tissue biopsy data (n = 5), was utilised. We report a high individual variation observed for all PADI isozymes in the patients' tissue biopsies, including lung, in response to SARS-CoV-2 infection, while PADI2 and PADI4 mRNA showed most variability in lung tissue specifically. The other tissues assessed were heart, kidney, marrow, bowel, jejunum, skin and fat, which all varied with respect to mRNA levels for the different PADI isozymes. In vitro lung epithelial and adenocarcinoma alveolar cell models revealed that PADI1, PADI2 and PADI4 mRNA levels were elevated, but PADI3 and PADI6 mRNA levels were reduced in SARS-CoV-2-infected NHBE cells. In A549 cells, PADI2 mRNA was elevated, PADI3 and PADI6 mRNA was downregulated, and no effect was observed on the PADI4 or PADI6 mRNA levels in infected cells, compared with control mock cells. Our findings indicate a link between PADI expression changes, including modulation of PADI2 and PADI4, particularly in lung tissue, in response to SARS-CoV-2 infection. PADI isozyme 1-6 expression in other organ biopsies also reveals putative links to COVID-19 symptoms, including vascular, cardiac and cutaneous responses, kidney injury and stroke. KEGG and GO pathway analysis furthermore identified links between PADs and inflammatory pathways, in particular between PAD4 and viral infections, as well as identifying links for PADs with a range of comorbidities. The analysis presented here highlights roles for PADs in-host responses to SARS-CoV-2, and their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Elif Damla Arisan
- Gebze Technical University, Institute of Biotechnology, Gebze, 41400 Kocaeli, Turkey;
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK;
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK
| |
Collapse
|
74
|
Oto J, Plana E, Solmoirago MJ, Fernández-Pardo Á, Hervás D, Cana F, España F, Artoni A, Bucciarelli P, Carrabba G, Navarro S, Merati G, Medina P. microRNAs and Markers of Neutrophil Activation as Predictors of Early Incidental Post-Surgical Pulmonary Embolism in Patients with Intracranial Tumors. Cancers (Basel) 2020; 12:cancers12061536. [PMID: 32545233 PMCID: PMC7353032 DOI: 10.3390/cancers12061536] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism (VTE) is a common complication of cancer that severely increases morbidity and mortality. Patients with intracranial tumors are more likely to develop VTE than patients with cancers at other sites. Conversely, limited tools exist to identify patients with high thrombotic risk. Upon activation, neutrophils release their content through different mechanisms triggering thrombosis. We explored the ability of microRNAs (miRNAs) and plasma markers of neutrophil activation measured before surgery to predict the risk of early post-surgical pulmonary embolism (PE) in glioma and meningioma patients. We recruited and prospectively followed 50 patients with glioma and 50 with meningioma, 34% of whom in each group developed an early objectively-diagnosed post-surgical PE. We measured miRNA expression and neutrophil markers (cell-free DNA, nucleosomes, calprotectin and myeloperoxidase) before surgery. In glioma patients, we adjusted and validated a predictive model for post-surgical PE with 6 miRNAs: miR-363-3p, miR-93-3p, miR-22-5p, miR-451a, miR-222-3p and miR-140-3p (AUC = 0.78; 95% Confidence Interval (CI) [0.63, 0.94]) and another with cfDNA and myeloperoxidase as predictors (AUC = 0.71; 95% CI [0.52, 0.90]). Furthermore, we combined both types of markers and obtained a model with myeloperoxidase and miR-140-3p as predictors (AUC = 0.79; 95% CI [0.64, 0.94]). In meningioma patients we fitted and validated a predictive model with 6 miRNAs: miR-29a-3p, miR-660-5p, miR-331-3p, miR-126-5p, miR-23a-3p and miR-23b-3p (AUC = 0.69; 95% CI [0.52, 0.87]). All our models outperformed the Khorana score. This is the first study that analyzes the capability of plasma miRNAs and neutrophil activation markers to predict early post-surgical PE in glioma and meningioma patients. The estimation of the thrombotic risk before surgery may promote a tailored thromboprophylaxis in a selected group of high-risk patients, in order to minimize the incidence of PE and avoid bleedings.
Collapse
Affiliation(s)
- Julia Oto
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
| | - Emma Plana
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
| | - María José Solmoirago
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
| | - Álvaro Fernández-Pardo
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
| | - David Hervás
- Data Science, Biostatistics and Bioinformatics Unit, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain;
| | - Fernando Cana
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
| | - Francisco España
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
| | - Andrea Artoni
- A. Bianchi Bonomi Hemophilia and Thrombosis Centre, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (P.B.); (G.M.)
| | - Paolo Bucciarelli
- A. Bianchi Bonomi Hemophilia and Thrombosis Centre, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (P.B.); (G.M.)
| | - Giorgio Carrabba
- Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Silvia Navarro
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
| | - Giuliana Merati
- A. Bianchi Bonomi Hemophilia and Thrombosis Centre, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.A.); (P.B.); (G.M.)
| | - Pilar Medina
- Haemostasis, Thrombosis, Atherosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (J.O.); (E.P.); (M.J.S.); (Á.F.-P.); (F.C.); (F.E.); (S.N.)
- Correspondence:
| |
Collapse
|
75
|
Liberale L, Camici GG. The Role of Vascular Aging in Atherosclerotic Plaque Development and Vulnerability. Curr Pharm Des 2020; 25:3098-3111. [PMID: 31470777 DOI: 10.2174/1381612825666190830175424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/24/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The ongoing demographical shift is leading to an unprecedented aging of the population. As a consequence, the prevalence of age-related diseases, such as atherosclerosis and its thrombotic complications is set to increase in the near future. Endothelial dysfunction and vascular stiffening characterize arterial aging and set the stage for the development of cardiovascular diseases. Atherosclerotic plaques evolve over time, the extent to which these changes might affect their stability and predispose to sudden complications remains to be determined. Recent advances in imaging technology will allow for longitudinal prospective studies following the progression of plaque burden aimed at better characterizing changes over time associated with plaque stability or rupture. Oxidative stress and inflammation, firmly established driving forces of age-related CV dysfunction, also play an important role in atherosclerotic plaque destabilization and rupture. Several genes involved in lifespan determination are known regulator of redox cellular balance and pre-clinical evidence underlines their pathophysiological roles in age-related cardiovascular dysfunction and atherosclerosis. OBJECTIVE The aim of this narrative review is to examine the impact of aging on arterial function and atherosclerotic plaque development. Furthermore, we report how molecular mechanisms of vascular aging might regulate age-related plaque modifications and how this may help to identify novel therapeutic targets to attenuate the increased risk of CV disease in elderly people.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland.,University Heart Center, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
76
|
Hosur V, Skelly DA, Francis C, Low BE, Kohar V, Burzenski LM, Amiji MM, Shultz LD, Wiles MV. Improved mouse models and advanced genetic and genomic technologies for the study of neutrophils. Drug Discov Today 2020; 25:1013-1025. [PMID: 32387410 DOI: 10.1016/j.drudis.2020.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022]
Abstract
Mice have been excellent surrogates for studying neutrophil biology and, furthermore, murine models of human disease have provided fundamental insights into the roles of human neutrophils in innate immunity. The emergence of novel humanized mice and high-diversity mouse populations offers the research community innovative and powerful platforms for better understanding, respectively, the mechanisms by which human neutrophils drive pathogenicity, and how genetic differences underpin the variation in neutrophil biology observed among humans. Here, we review key examples of these new resources. Additionally, we provide an overview of advanced genetic engineering tools available to further improve such murine model systems, of sophisticated neutrophil-profiling technologies, and of multifunctional nanoparticle (NP)-based neutrophil-targeting strategies.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA.
| | - Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Christopher Francis
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Benjamin E Low
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Vivek Kohar
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Lisa M Burzenski
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Leonard D Shultz
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Michael V Wiles
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| |
Collapse
|
77
|
Liberale L, Carbone F, Montecucco F, Sahebkar A. Statins reduce vascular inflammation in atherogenesis: A review of underlying molecular mechanisms. Int J Biochem Cell Biol 2020; 122:105735. [PMID: 32126319 DOI: 10.1016/j.biocel.2020.105735] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 01/23/2020] [Accepted: 02/28/2020] [Indexed: 01/09/2023]
Abstract
Chronic inflammation enhances the detrimental role of dyslipidaemia during atherogenesis. Statins are among the most effective anti-atherosclerotic medications, being able to impact on both cardiovascular morbidity and mortality. Although these molecules have been first described as lipid-lowering medications, several lines of evidence suggest additional benefits through their "pleiotropic" anti-atherosclerotic activities. Specifically, statins can modulate vascular atherosclerotic inflammation by directly improving functions of endothelial cells, vascular smooth muscle cells, platelets, and immune cells. Here, we discuss basic and clinical evidence to provide an update on the molecular mechanisms underlying the protective anti-inflammatory role of statins in atherogenesis.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
78
|
Davies MJ, Hawkins CL. The Role of Myeloperoxidase in Biomolecule Modification, Chronic Inflammation, and Disease. Antioxid Redox Signal 2020; 32:957-981. [PMID: 31989833 DOI: 10.1089/ars.2020.8030] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Significance: The release of myeloperoxidase (MPO) by activated leukocytes is critical in innate immune responses. MPO produces hypochlorous acid (HOCl) and other strong oxidants, which kill bacteria and other invading pathogens. However, MPO also drives the development of numerous chronic inflammatory pathologies, including atherosclerosis, neurodegenerative disease, lung disease, arthritis, cancer, and kidney disease, which are globally responsible for significant patient mortality and morbidity. Recent Advances: The development of imaging approaches to precisely identify the localization of MPO and the molecular targets of HOCl in vivo is an important advance, as typically the involvement of MPO in inflammatory disease has been inferred by its presence, together with the detection of biomarkers of HOCl, in biological fluids or diseased tissues. This will provide valuable information in regard to the cell types responsible for releasing MPO in vivo, together with new insight into potential therapeutic opportunities. Critical Issues: Although there is little doubt as to the value of MPO inhibition as a protective strategy to mitigate tissue damage during chronic inflammation in experimental models, the impact of long-term inhibition of MPO as a therapeutic strategy for human disease remains uncertain, in light of the potential effects on innate immunity. Future Directions: The development of more targeted MPO inhibitors or a treatment regimen designed to reduce MPO-associated host tissue damage without compromising pathogen killing by the innate immune system is therefore an important future direction. Similarly, a partial MPO inhibition strategy may be sufficient to maintain adequate bacterial activity while decreasing the propagation of inflammatory pathologies.
Collapse
Affiliation(s)
- Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
79
|
Otandault A, Anker P, Al Amir Dache Z, Guillaumon V, Meddeb R, Pastor B, Pisareva E, Sanchez C, Tanos R, Tousch G, Schwarzenbach H, Thierry AR. Recent advances in circulating nucleic acids in oncology. Ann Oncol 2020; 30:374-384. [PMID: 30753271 DOI: 10.1093/annonc/mdz031] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) is one of the fastest growing and most exciting areas in oncology in recent years. Its potential clinical uses cover now each phase of cancer patient management care (predictive information, detection of the minimal residual disease, early detection of resistance, treatment monitoring, recurrence surveillance, and cancer early detection/screening). This review relates the recent advances in the application of circulating DNA or RNA in oncology building on unpublished or initial findings/work presented at the 10th international symposium on circulating nucleic acids in plasma and serum held in Montpellier from the 20th to the 22nd of September 2017. This year, presenters revealed their latest data and crucial observations notably in relation to (i) the circulating cell-free (cfDNA) structure and implications regarding their optimal detection; (ii) their role in the metastatic or immunological processes; (iii) evaluation of miRNA panels for cancer patient follow up; (iv) the detection of the minimal residual disease; (v) the evaluation of a screening tests for cancer using cfDNA analysis; and (vi) elements of preanalytical guidelines. This work reviews the recent progresses in the field brought to light in the meeting, as well as in the most important reports from the literature, past and present. It proposes a broader picture of the basic research and its potential, and of the implementation and current challenges in the use of circulating nucleic acids in oncology.
Collapse
Affiliation(s)
- A Otandault
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - P Anker
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - Z Al Amir Dache
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - V Guillaumon
- Regional Institute of Cancer of Montpellier, Montpellier; SIRIC, Integrated Cancer Research Site, Montpellier, France
| | - R Meddeb
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - B Pastor
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - E Pisareva
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - C Sanchez
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - R Tanos
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - G Tousch
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier
| | - H Schwarzenbach
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A R Thierry
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier; INSERM, U1194, Montpellier; Department of Oncology, Montpellier University, Montpellier; Regional Institute of Cancer of Montpellier, Montpellier.
| |
Collapse
|
80
|
Scandolara TB, Panis C. Neutrophil traps, anti-myeloperoxidase antibodies and cancer: Are they linked? Immunol Lett 2020; 221:33-38. [PMID: 32092357 DOI: 10.1016/j.imlet.2020.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/13/2020] [Accepted: 02/20/2020] [Indexed: 12/17/2022]
Abstract
Myeloperoxidase is an enzyme present in neutrophils and has been demonstrated to be an important molecule for neutrophil extracellular traps (NETs) formation and function. Yet, it is also a source of autoantigens for anti-neutrophil or anti-myeloperoxidase antibodies (ANCAs), which are capable of activating these immune cells and provoke tissue damage in a sterile microenvironment. The presence of these antibodies in cancer has been related by case reports, but a few studies addressed the significance of this finding beyond autoimmunity context. In this review, we discuss the evidences regarding ANCAs and cancer and its putative clinical meaning in the context of tumor immunology.
Collapse
Affiliation(s)
- Thalita Basso Scandolara
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Health-Applied Sciences Post Graduation Program, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Federal University of Rio de Janeiro, UFRJ, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil; Health-Applied Sciences Post Graduation Program, State University of West Paraná (Unioeste), Francisco Beltrão, Paraná, Brazil.
| |
Collapse
|
81
|
Carbone F, Bonaventura A, Montecucco F. Neutrophil-Related Oxidants Drive Heart and Brain Remodeling After Ischemia/Reperfusion Injury. Front Physiol 2020; 10:1587. [PMID: 32116732 PMCID: PMC7010855 DOI: 10.3389/fphys.2019.01587] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/18/2019] [Indexed: 12/22/2022] Open
Abstract
The inflammatory response associated with myocardial and brain ischemia/reperfusion injury (IRI) is a critical determinant of tissue necrosis, functional organ recovery, and long-term clinical outcomes. In the post-ischemic period, reactive oxygen species (ROS) are involved in tissue repair through the clearance of dead cells and cellular debris. Neutrophils play a critical role in redox signaling due to their early recruitment and the large variety of released ROS. Noteworthy, ROS generated during IRI have a relevant role in both myocardial healing and activation of neuroprotective pathways. Anatomical and functional differences contribute to the responses in the myocardial and brain tissue despite a significant gene overlap. The exaggerated activation of this signaling system can result in adverse consequences, such as cell apoptosis and extracellular matrix degradation. In light of that, blocking the ROS cascade might have a therapeutic implication for cardiomyocyte and neuronal loss after acute ischemic events. The translation of these findings from preclinical models to clinical trials has so far failed because of differences between humans and animals, difficulty of agents to penetrate into specific cellular organs, and specifically unravel oxidant and antioxidant pathways. Here, we update knowledge on ROS cascade in IRI, focusing on the role of neutrophils. We discuss evidence of ROS blockade as a therapeutic approach for myocardial infarction and ischemic stroke.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
82
|
Carbone F, Bonaventura A, Liberale L, Paolino S, Torre F, Dallegri F, Montecucco F, Cutolo M. Atherosclerosis in Rheumatoid Arthritis: Promoters and Opponents. Clin Rev Allergy Immunol 2020; 58:1-14. [PMID: 30259381 DOI: 10.1007/s12016-018-8714-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Substantial epidemiological data identified cardiovascular (CV) diseases as a main cause of mortality in patients with rheumatoid arthritis (RA). In light of this, RA patients may benefit from additional CV risk screening and more intensive prevention strategies. Nevertheless, current algorithms for CV risk stratification still remain tailored on general population and are burdened by a significant underestimation of CV risk in RA patients. Acute CV events in patients with RA are largely related to an accelerated atherosclerosis. As pathophysiological features of atherosclerosis overlap those occurring in the inflamed RA synovium, the understanding of those common pathways represents an urgent need and a leading challenge for CV prevention in patients with RA. Genetic background, metabolic status, gut microbiome, and systemic inflammation have been also suggested as additional key pro-atherosclerotic factors. The aim of this narrative review is to update the current knowledge about pathophysiology of atherogenesis in RA patients and potential anti-atherosclerotic effects of disease-modifying anti-rheumatic drugs.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, 12 Wagistrasse, 8952, Schlieren, Switzerland
| | - Sabrina Paolino
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, San Martino Polyclinic Hospital, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132, Genoa, Italy
| | - Francesco Torre
- IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132, Genoa, Italy
- Clinic of Emergency Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Maurizio Cutolo
- IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132, Genoa, Italy.
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, San Martino Polyclinic Hospital, Genoa, Italy.
| |
Collapse
|
83
|
MicroRNAs and Neutrophil Activation Markers Predict Venous Thrombosis in Pancreatic Ductal Adenocarcinoma and Distal Extrahepatic Cholangiocarcinoma. Int J Mol Sci 2020; 21:ijms21030840. [PMID: 32012923 PMCID: PMC7043221 DOI: 10.3390/ijms21030840] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer-associated venous thrombosis (VTE) increases mortality and morbidity. However, limited tools are available to identify high risk patients. Upon activation, neutrophils release their content through different mechanisms, thereby prompting thrombosis. We explored plasma microRNAs (miRNAs) and neutrophil activation markers to predict VTE in pancreatic ductal adenocarcinoma (PDAC) and distal extrahepatic cholangiocarcinoma (DECC). Twenty-six PDAC and 6 DECC patients recruited at cancer diagnosis, were examined for deep vein thrombosis and pulmonary embolisms, and were then followed-up with clinical examinations, blood collections, and biCUS. Ten patients developed VTE and were compared with 22 age- and sex-matched controls. miRNA expression levels were measured at diagnosis and right before VTE, and neutrophil activation markers (cell-free DNA, nucleosomes, calprotectin, and myeloperoxidase) were measured in every sample obtained during follow-up. We obtained a profile of 7 miRNAs able to estimate the risk of future VTE at diagnosis (AUC = 0.95; 95% Confidence Interval (CI) (0.987, 1)) with targets involved in the pancreatic cancer and complement and coagulation cascades pathways. Seven miRNAs were up- or down-regulated before VTE compared with diagnosis. We obtained a predictive model of VTE with calprotectin as predictor (AUC = 0.77; 95% CI (0.57, 0.95)). This is the first study that addresses the ability of plasma miRNAs and neutrophil activation markers to predict VTE in PDAC and DECC.
Collapse
|
84
|
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Diana A Gorog
- School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Hertfordshire, UK
- Department of Cardiology, East and North Hertfordshire NHS Trust, Hertfordshire, UK
- Faculty of Medicine, National Heart & Lung Institute, Imperial College, London, UK
| |
Collapse
|
85
|
Bonaventura A, Vecchié A, Abbate A, Montecucco F. Neutrophil Extracellular Traps and Cardiovascular Diseases: An Update. Cells 2020; 9:231. [PMID: 31963447 PMCID: PMC7016588 DOI: 10.3390/cells9010231] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are formed by decondensed chromatin, histones, and neutrophil granular proteins and have a role in entrapping microbial pathogens. NETs, however, have pro-thrombotic properties by stimulating fibrin deposition, and increased NET levels correlate with larger infarct size and predict major adverse cardiovascular (CV) events. NETs have been involved also in the pathogenesis of diabetes, as high glucose levels were found to induce NETosis. Accordingly, NETs have been described as drivers of diabetic complications, such as diabetic wound and diabetic retinopathy. Inflammasomes are macromolecular structures involved in the release of pro-inflammatory mediators, such as interleukin-1, which is a key mediator in CV diseases. A crosstalk between the inflammasome and NETs is known for some rheumatologic diseases, while this link is still under investigation and not completely understood in CV diseases. In this review, we summarized the most recent updates about the role of NETs in acute myocardial infarction and metabolic diseases and provided an overview on the relationship between NET and inflammasome activities in rheumatologic diseases, speculating a possible link between these two entities also in CV diseases.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA 23298, USA; (A.V.); (A.A.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132 Genoa, Italy
| | - Alessandra Vecchié
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA 23298, USA; (A.V.); (A.A.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132 Genoa, Italy
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA 23298, USA; (A.V.); (A.A.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, viale Benedetto XV 6, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino Genova—Italian Cardiovascular Network, Largo R. Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
86
|
Inflammatory Biomarkers for Cardiovascular Risk Stratification in Familial Hypercholesterolemia. Rev Physiol Biochem Pharmacol 2020; 177:25-52. [PMID: 32691159 DOI: 10.1007/112_2020_26] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Familial hypercholesterolemia (FH) is a frequent autosomal genetic disease characterized by elevated concentrations of low-density lipoprotein cholesterol (LDL) from birth with increased risk of premature atherosclerotic complications. Accumulating evidence has shown enhanced inflammation in patients with FH. In vessels, the deposition of modified cholesterol lipoproteins triggers local inflammation. Then, inflammation facilitates fatty streak formation by activating the endothelium to produce chemokines and adhesion molecules. This process eventually results in the uptake of vascular oxidized LDL (OxLDL) by scavenger receptors in monocyte-derived macrophages and formation of foam cells. Further leukocyte recruitment into the sub-endothelial space leads to plaque progression and activation of smooth muscle cells proliferation. Several inflammatory biomarkers have been reported in this setting which can be directly synthetized by activated inflammatory/vascular cells or can be indirectly produced by organs other than vessels, e.g., liver. Of note, inflammation is boosted in FH patients. Inflammatory biomarkers might improve the risk stratification for coronary heart disease and predict atherosclerotic events in FH patients. This review aims at summarizing the current knowledge about the role of inflammation in FH and the potential application of inflammatory biomarkers for cardiovascular risk estimation in these patients.
Collapse
|
87
|
Yang F, He Y, Zhai Z, Sun E. Programmed Cell Death Pathways in the Pathogenesis of Systemic Lupus Erythematosus. J Immunol Res 2019; 2019:3638562. [PMID: 31871956 PMCID: PMC6913273 DOI: 10.1155/2019/3638562] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/04/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease characterized by excessive inflammatory and immune responses and tissue damage. Increasing evidence has demonstrated the important role of programmed cell death in SLE pathogenesis. When apoptosis encounters with defective clearance, accumulated apoptotic cells lead to secondary necrosis. Different forms of lytic cell death, including secondary necrosis after apoptosis, NETosis, necroptosis, and pyroptosis, contribute to the release of damage-associated molecular patterns (DAMPs) and autoantigens, resulting in triggering immunity and tissue damage in SLE. However, the role of autophagy in SLE pathogenesis is in dispute. This review briefly discusses different forms of programmed cell death pathways and lay particular emphasis on inflammatory cell death pathways such as NETosis, pyroptosis, and necroptosis and their roles in the inflammatory and immune responses in SLE.
Collapse
Affiliation(s)
- Fangyuan Yang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Zeqing Zhai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
- Department of Rheumatology and Immunology, Shunde Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
88
|
Liberale L, Holy EW, Akhmedov A, Bonetti NR, Nietlispach F, Matter CM, Mach F, Montecucco F, Beer JH, Paneni F, Ruschitzka F, Libby P, Lüscher TF, Camici GG. Interleukin-1β Mediates Arterial Thrombus Formation via NET-Associated Tissue Factor. J Clin Med 2019; 8:2072. [PMID: 31779200 PMCID: PMC6947515 DOI: 10.3390/jcm8122072] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 01/31/2023] Open
Abstract
CANTOS reported reduced secondary atherothrombotic events in patients with residual inflammatory risk treated with the inhibitory anti-IL-1β antibody, Canakinumab. Yet, mechanisms that underlie this benefit remain elusive. Recent work has implicated formation of neutrophil extracellular traps (NETosis) in arterial thrombosis. Hence, the present study explored the potential link between IL-1β, NETs, and tissue factor (TF)-the key trigger of the coagulation cascade-in atherothrombosis. To this end, ST-elevation myocardial infarction (STEMI) patients from the Swiss multicenter trial SPUM-ACS were retrospectively and randomly selected based on their CRP levels. In particular, 33 patients with STEMI and high C-reactive protein (CRP) levels (≥ 10 mg/L) and, 33 with STEMI and low CRP levels (≤ 4 mg/L) were investigated. High CRP patients displayed elevated circulating IL-1β, NETosis, and NET-associated TF plasma levels compared with low CRP ones. Additionally, analysis of patients stratified by circulating IL-1β levels yielded similar results. Moreover, NETosis and NET-associated TF plasma levels correlated positively in the whole population. In addition to the above, translational research experiments provided mechanistic confirmation for the clinical data identifying IL-1β as the initial trigger for the release of the pro-coagulant, NET-associated TF. In conclusion, blunted TF presentation by activated neutrophils undergoing NETosis may provide a mechanistic explanation to reduced secondary atherothrombotic events as observed in canakinumab-treated patients in CANTOS.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy;
| | - Erik W. Holy
- Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland; (E.W.H.); (F.N.); (F.R.)
| | - Alexander Akhmedov
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
| | - Nicole R. Bonetti
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- Department of Internal Medicine, Cantonal Hospital of Baden, 5404 Baden, Switzerland
| | - Fabian Nietlispach
- Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland; (E.W.H.); (F.N.); (F.R.)
| | - Christian M. Matter
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland; (E.W.H.); (F.N.); (F.R.)
| | - François Mach
- Department of Cardiology, Hopital Universitaire de Geneve, 1206 Geneve, Switzerland;
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Jürg H. Beer
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- Department of Internal Medicine, Cantonal Hospital of Baden, 5404 Baden, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland; (E.W.H.); (F.N.); (F.R.)
- Department of Research and Education, University Hospital Zurich, 8001 Zurich, Switzerland
| | - Frank Ruschitzka
- Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland; (E.W.H.); (F.N.); (F.R.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115 Boston, MA, USA;
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- Royal Brompton and Harefield Hospitals and Imperial College, London SW3 6NP, UK
| | - Giovanni G. Camici
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8952 Schlieren, Switzerland; (L.L.); (A.A.); (N.R.B.); (C.M.M.); (F.P.); (T.F.L.)
- Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland; (E.W.H.); (F.N.); (F.R.)
- Department of Research and Education, University Hospital Zurich, 8001 Zurich, Switzerland
| |
Collapse
|
89
|
Zhang Z, Luo Y, Zhang Y, Guo K. Enhanced protective immune response to PCV2 adenovirus vaccine by fusion expression of Cap protein with InvC in pigs. J Vet Sci 2019; 20:e35. [PMID: 31364320 PMCID: PMC6669209 DOI: 10.4142/jvs.2019.20.e35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/19/2019] [Accepted: 05/20/2019] [Indexed: 01/21/2023] Open
Abstract
The major immunogenic protein capsid (Cap) of porcine circovirus type 2 (PCV2) is critical to induce neutralizing antibodies and protective immune response against PCV2 infection. This study was conducted to investigate the immune response of recombinant adenovirus expressing PCV2b Cap and C-terminal domain of Yersinia pseudotuberculosis invasin (Cap-InvC) fusion protein in pigs. The recombinant adenovirus rAd-Cap-InvC, rAd-Cap and rAd were generated and used to immunize pigs. The phosphate-buffered saline was used as negative control. The specific antibodies levels in rAd-Cap-InvC and ZJ/C-strain vaccine groups were higher than that of rAd-Cap group (p < 0.05), and the neutralization antibody titer in rAd-Cap-InvC group was significantly higher than those of other groups during 21–42 days post-immunization (DPI). Moreover, lymphocyte proliferative level, interferon-γ and interleukin-13 levels in rAd-Cap-InvC group were increased compared to rAd-Cap group (p < 0.05). After virulent challenge, viruses were not detected from the blood samples in rAd-Cap-InvC and ZJ/C-strain vaccine groups after 49 DPI. And the respiratory symptom, rectal temperature, lung lesion and lymph node lesion were minimal and similar in the ZJ/C-strain and rAd-Cap-InVC groups. In conclusion, our results demonstrated that rAd-Cap-InvC was more efficiently to stimulate the production of antibody and protect pigs from PCV2 infection. We inferred that InvC is a good candidate gene for further development and application of PCV2 genetic engineering vaccine.
Collapse
Affiliation(s)
- Zhencang Zhang
- Department of Animal Engineering, Yangling Vocational and Technical College, Yangling 712100, China.,College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yan Luo
- Department of Animal Engineering, Yangling Vocational and Technical College, Yangling 712100, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
90
|
Liberale L, Carbone F, Camici GG, Montecucco F. IL-1β and Statin Treatment in Patients with Myocardial Infarction and Diabetic Cardiomyopathy. J Clin Med 2019; 8:1764. [PMID: 31652822 PMCID: PMC6912287 DOI: 10.3390/jcm8111764] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
Statins are effective lipid-lowering drugs with a good safety profile that have become, over the years, the first-line therapy for patients with dyslipidemia and a real cornerstone of cardiovascular (CV) preventive therapy. Thanks to both cholesterol-related and "pleiotropic" effects, statins have a beneficial impact against CV diseases. In particular, by reducing lipids and inflammation statins, they can influence the pathogenesis of both myocardial infarction and diabetic cardiomyopathy. Among inflammatory mediators involved in these diseases, interleukin (IL)-1β is a pro-inflammatory cytokine that recently been shown to be an effective target in secondary prevention of CV events. Statins are largely prescribed to patients with myocardial infarction and diabetes, but their effects on IL-1β synthesis and release remain to be fully characterized. Of interest, preliminary studies even report IL-1β secretion to rise after treatment with statins, with a potential impact on the inflammatory microenvironment and glycemic control. Here, we will summarize evidence of the role of statins in the prevention and treatment of myocardial infarction and diabetic cardiomyopathy. In accordance with the dual lipid-lowering and anti-inflammatory effect of these drugs and in light of the important results achieved by IL-1β inhibition through canakinumab in CV secondary prevention, we will dissect the current evidence linking statins with IL-1β and outline the possible benefits of a potential double treatment with statins and canakinumab.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Schlieren, 8092, Switzerland.
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy.
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy.
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Schlieren, 8092, Switzerland.
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8001 Zurich, Switzerland.
- Department of Research and Education, University Hospital Zurich, 8001 Zurich, Switzerland.
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy.
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, University of Genoa, 16132 Genoa, Italy.
| |
Collapse
|
91
|
Yang F, Luo X, Luo G, Zhai Z, Zhuang J, He J, Han J, Zhang Y, Zhuang L, Sun E, He Y. Inhibition of NET formation by polydatin protects against collagen-induced arthritis. Int Immunopharmacol 2019; 77:105919. [PMID: 31655341 DOI: 10.1016/j.intimp.2019.105919] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/21/2019] [Accepted: 09/14/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systematic, inflammatory, autoimmune disease, associated with a high number of disabilities. Increasing evidence has demonstrated that neutrophil extracellular trap (NET) formation plays a significant role in the pathogenesis and progression of RA. In this study, we have aimed to investigate the effects of polydatin (PD) on NET formation and its effects on disease activity in a collagen-induced arthritis (CIA) mouse model. METHODS In the presence of PD or vehicle, neutrophils isolated from RA patients and mice were treated with phorbol 12-myristate 13-acetate (PMA) for 4 h, and NET formation investigated. For in vivo experiments, PD was administered intraperitoneally (45 mg/kg per day) to collagen-induced arthritis (CIA) mice. The incidence and severity of collagen-induced arthritis were assessed and NET deposition tested. RESULTS In vitro, PD significantly suppressed NET formation of neutrophils from RA patients. Consistently, decreased NETs were observed in PD treated bone marrow-derived neutrophils. In CIA mouse model, PD treatment delayed the onset of arthritis and attenuated arthritis severity. Compared with vehicle-treated CIA mice, the deposition of NETs in ankle joints was also reduced in PD-treated CIA mice. CONCLUSION In this study, we found that PD treatment markedly inhibited NET formation and protected CIA mice from the development of arthritis. These findings suggest that inhibition of NET formation by PD may serve as a novel mechanism for the treatment of RA.
Collapse
Affiliation(s)
- Fangyuan Yang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Xiaoqing Luo
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Guihu Luo
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Zeqing Zhai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Jian Zhuang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Juan He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Jiaochan Han
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Yanan Zhang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Lili Zhuang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China.
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Institute of Clinical Immunology, Academy of Orthopedics, Guangdong Province, Guangzhou, China.
| |
Collapse
|
92
|
Szucs B, Szucs C, Petrekanits M, Varga JT. Molecular Characteristics and Treatment of Endothelial Dysfunction in Patients with COPD: A Review Article. Int J Mol Sci 2019; 20:E4329. [PMID: 31487864 PMCID: PMC6770145 DOI: 10.3390/ijms20184329] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) show systemic consequences, such as chronic systemic inflammation leading to changes in the airway, airway penetrability, and endothelial function. Endothelial dysfunction is characterized by a list of alterations of endothelium towards reduced vasodilation, proinflammatory state, detachment and apoptosis of endothelial cells, and development of atherosclerosis. COPD-induced endothelial dysfunction is associated with elevated cardiovascular risk. The increment of physical activities such as pulmonary rehabilitation (PR) training have a significant effect on COPD, thus, PR can be an integrative part of COPD treatment. In this narrative review the focus is on the function of endothelial inflammatory mediators [cytokines, chemokines, and cellular proteases] and pulmonary endothelial cells and endothelial dysfunction in COPD as well as the effects of dysfunction of the endothelium may play in COPD-related pulmonary hypertension. The relationship between smoking and endothelial dysfunction is also discussed. The connection between different pulmonary rehabilitation programs, arterial stiffness and pulse wave velocity (PWV) is presented. Endothelial dysfunction is a significant prognostic factor of COPD, which can be characterized by PWV. We discuss future considerations, like training programs, as an important part of the treatment that has a favorable impact on the endothelial function.
Collapse
Affiliation(s)
- Botond Szucs
- PharmaFlight Research and Training Center, H-4030 Debrecen, Hungary
| | - Csilla Szucs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
| | - Mate Petrekanits
- Institute of Exercise Physiology and Sport Medicine, University of Physical Education, H-1123 Budapest, Hungary
| | - Janos T Varga
- Department of Pulmonary Rehabilitation, National Koranyi Institute for Pulmonology, H-1121 Budapest, Hungary.
| |
Collapse
|
93
|
Targeting neutrophil extracellular traps enhanced tPA fibrinolysis for experimental intracerebral hemorrhage. Transl Res 2019; 211:139-146. [PMID: 31103467 DOI: 10.1016/j.trsl.2019.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 01/06/2023]
Abstract
The minimally invasive surgery plus fibrinolysis has been identified as a promising treatment for spontaneous intracerebral hemorrhage (ICH). However, the fibrinolytic efficacy is not satisfactory. Neutrophil extracellular traps (NETs) have been demonstrated to impair fibrinolysis in sepsis and acute ischemic stroke. Therefore, we decided to explore the presence and potential effect of NETs in ICH fibrinolysis. Intracerebral hemorrhage was induced by autologous arterial blood injection into the basal ganglia in rats. First, at 0.5 hour, 1 hour, and 1.5 hours after blood injection, the brains were collected for NETs detection by immune-staining. Second, ICH rats were given intrahematoma fibrinolysis: rats were randomized to receive the equal amount of saline, DNAse 1, tissue-plasminogen activator (tPA), and tPA + DNAse 1 at 1 hour after hematoma placement. On day 3, animals were sacrificed for terminal deoxynucleotidyl transferase-mediated dUTP Nick-end labeling staining following MRI and behavioral tests. Third, on day 3 after ICH, the hematoma within brain were collected for ex vivo fibrinolysis assay to further evaluate the effect of NETs in ICH fibrinolysis. Co-staining of DAPI, H3, and MPO confirmed the presence of NETs in ICH. Disintegration of NETs using DNAse 1 enhanced tPA-induced hematoma fibrinolysis, relieved brain swelling, reduced cell death, and improved the functional outcome in ICH rats. Therefore, we concluded that NETs impaired the efficacy of tPA for ICH fibrinolysis in rats. Targeting NETs may be a new alternative to improve the fibrinolytic therapy following ICH.
Collapse
|
94
|
Salazar-Gonzalez H, Zepeda-Hernandez A, Melo Z, Saavedra-Mayorga DE, Echavarria R. Neutrophil Extracellular Traps in the Establishment and Progression of Renal Diseases. ACTA ACUST UNITED AC 2019; 55:medicina55080431. [PMID: 31382486 PMCID: PMC6722876 DOI: 10.3390/medicina55080431] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 01/27/2023]
Abstract
Uncontrolled inflammatory and immune responses are often involved in the development of acute and chronic forms of renal injury. Neutrophils are innate immune cells recruited early to sites of inflammation, where they produce pro-inflammatory cytokines and release mesh-like structures comprised of DNA and granular proteins known as neutrophil extracellular traps (NETs). NETs are potentially toxic, contribute to glomerular injury, activate autoimmune processes, induce vascular damage, and promote kidney fibrosis. Evidence from multiple studies suggests that an imbalance between production and clearance of NETs is detrimental for renal health. Hence strategies aimed at modulating NET-associated processes could have a therapeutic impact on a myriad of inflammatory diseases that target the kidney. Here, we summarize the role of NETs in the pathogenesis of renal diseases and their mechanisms of tissue damage.
Collapse
Affiliation(s)
- Hector Salazar-Gonzalez
- Decanato de Ciencia y Tecnología, Universidad Autónoma de Guadalajara, Zapopan 45129, Mexico
| | | | - Zesergio Melo
- CONACyT-Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800 Col. Independencia, Guadalajara 44340, Mexico
| | - Diego Eduardo Saavedra-Mayorga
- Facultad de Medicina, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Raquel Echavarria
- CONACyT-Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800 Col. Independencia, Guadalajara 44340, Mexico.
| |
Collapse
|
95
|
Cell-Free Nucleic Acids and their Emerging Role in the Pathogenesis and Clinical Management of Inflammatory Bowel Disease. Int J Mol Sci 2019; 20:ijms20153662. [PMID: 31357438 PMCID: PMC6696129 DOI: 10.3390/ijms20153662] [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: 06/30/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Cell-free nucleic acids (cfNAs) are defined as any nucleic acids that are present outside the cell. They represent valuable biomarkers in various diagnostic protocols such as prenatal diagnostics, the detection of cancer, and cardiovascular or autoimmune diseases. However, in the current literature, little is known about their implication in inflammatory bowel disease (IBD). IBD is a group of multifactorial, autoimmune, and debilitating diseases with increasing incidence worldwide. Despite extensive research, their etiology and exact pathogenesis is still unclear. Since cfNAs were observed in other autoimmune diseases and appear to be relevant in inflammatory processes, their role in the pathogenesis of IBD has also been suggested. This review provides a summary of knowledge from the available literature about cfDNA and cfRNA and the structures involving them such as exosomes and neutrophil extracellular traps and their association with IBD. Current studies showed the promise of cfNAs in the management of IBD not only as biomarkers distinguishing patients from healthy people and differentiating active from inactive disease state, but also as a potential therapeutic target. However, the detailed biological characteristics of cfNAs need to be fully elucidated in future experimental and clinical studies.
Collapse
|
96
|
An S, Raju I, Surenkhuu B, Kwon JE, Gulati S, Karaman M, Pradeep A, Sinha S, Mun C, Jain S. Neutrophil extracellular traps (NETs) contribute to pathological changes of ocular graft-vs.-host disease (oGVHD) dry eye: Implications for novel biomarkers and therapeutic strategies. Ocul Surf 2019; 17:589-614. [PMID: 30965123 PMCID: PMC6721977 DOI: 10.1016/j.jtos.2019.03.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE To investigate the role of neutrophil extracellular traps (NETs) and NET-associated proteins in the pathogenesis of oGVHD and whether dismantling of NETs with heparin reduces those changes. METHODS Ocular surface washings from oGVHD patients and healthy subjects were analyzed. Isolated peripheral blood human neutrophils were stimulated to generate NETs and heparinized NETs. We performed in vitro experiments using cell lines (corneal epithelial, conjunctival fibroblast, meibomian gland (MG) epithelial and T cells), and in vivo experiments using murine models, and compared the effects of NETs, heparinized NETs, NET-associated proteins and neutralizing antibodies to NET-associated proteins. RESULTS Neutrophils, exfoliated epithelial cells, NETs and NET-associated proteins (extracellular DNA, Neutrophil Elastase, Myeloperoxidase, Oncostatin M (OSM), Neutrophil gelatinase-associated lipocalin (NGAL) and LIGHT/TNFSF14) are present in ocular surface washings (OSW) and mucocellular aggregates (MCA). Eyes with high number of neutrophils in OSW have more severe signs and symptoms of oGVHD. NETs (and OSM) cause epitheliopathy in murine corneas. NETs (and LIGHT/TNFSF14) increase proliferation of T cells. NETs (and NGAL) inhibit proliferation and differentiation of MG epithelial cells. NETs enhance proliferation and myofibroblast transformation of conjunctival fibroblasts. Sub-anticoagulant dose Heparin (100 IU/mL) dismantles NETs and reduces epithelial, fibroblast, T cell and MG cell changes induced by NETs. CONCLUSION NETs and NET-associated proteins contribute to the pathological changes of oGVHD (corneal epitheliopathy, conjunctival cicatrization, ocular surface inflammation and meibomian gland disease). Our data points to the potential of NET-associated proteins (OSM or LIGHT/TNFSF14) to serve as biomarkers and NET-dismantling biologics (heparin eye drops) as treatment for oGVHD.
Collapse
Affiliation(s)
- Seungwon An
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ilangovan Raju
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bayasgalan Surenkhuu
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ji-Eun Kwon
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shilpa Gulati
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Muge Karaman
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Anubhav Pradeep
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | | | - Christine Mun
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sandeep Jain
- Cornea Translational Biology Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
97
|
Bonaventura A, Montecucco F, Dallegri F, Carbone F, Lüscher TF, Camici GG, Liberale L. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 2019; 115:1266-1285. [PMID: 30918936 DOI: 10.1093/cvr/cvz084] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/04/2019] [Accepted: 03/26/2019] [Indexed: 08/30/2023] Open
Abstract
Neutrophils are the most abundant circulating leucocytes in healthy humans. These cells are central players during acute inflammatory responses, although a growing body of evidence supports a crucial role in chronic inflammation and chemokines and cytokines related to it as well. Thus, both humoral and cellular components are involved in the development of plaque formation and atherosclerosis. Accordingly, CANTOS trial using an interleukin-1 beta antibody confirmed that inflammatory cytokines contribute to the occurrence of myocardial infarction and cardiac death independent of changes in lipids. Recent data revealed that neutrophils are a heterogeneous population with different subsets and functional characteristics (i.e. CD177+ cells, OLFM4+ neutrophils, proangiogenic neutrophils, neutrophils undergoing reverse migration, and aged neutrophils). Importantly, neutrophils are able to synthesize de novo proteins. Neutrophil extracellular trap generation and NETosis have been considered as very important weapons in sterile inflammation. Neutrophil-derived microvesicles represent another mechanism by which neutrophils amplify inflammatory processes, being found at high levels both at the site of injury and in the bloodstream. Finally, neutrophil aging can influence their functions also in relation with host age. These recent acquisitions in the field of neutrophil biology might pave the way for new therapeutic targets to prevent or even treat patients experiencing cardiovascular (CV) diseases. Here, we discuss novel findings in neutrophil biology, their impact on CV and cerebrovascular diseases, and the potential implementation of these notions into daily clinical practice.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- University Heart Center, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
- Department of Research and Education, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| |
Collapse
|
98
|
Bonaventura A, Montecucco F. Inflammation and pericarditis: Are neutrophils actors behind the scenes? J Cell Physiol 2019; 234:5390-5398. [PMID: 30417336 DOI: 10.1002/jcp.27436] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022]
Abstract
The morbidity of acute pericarditis is increasing over time impacting on patient quality of life. Recent clinical trials focused especially on clinical aspects, with a modest interest in pathophysiological mechanisms. This narrative review, based on papers in English language obtained via PubMed up to April 2018, aims at focusing on the role of the innate immunity in pericarditis and discussing future potential therapeutic strategies impacting on disease pathophysiology. In developed countries, most cases of pericarditis are referred to as idiopathic, although etiological causes have been described, with autoreactive/lymphocytic, malignant, and infectious ones as the most frequent causes. Apart the known impairment of the adaptive immunity, recently a large body evidence indicated the central role of the innate immune system in the pathogenesis of recurrent pericarditis, starting from similarities with autoinflammatory diseases. Accordingly, the "inflammasome" has been shown to behave as an important player in pericarditis development. Similarly, the beneficial effect of colchicine in recurrent pericarditis confirms that neutrophils are important effectors as colchicine, which can block neutrophil chemotaxis, interferes with neutrophil adhesion and recruitment to injured tissues and abrogate superoxide production. Anyway, the role of the adaptive immune system in pericarditis cannot be reduced to a black or white issue as mechanisms often overlap. Therefore, we believe that more efficient therapeutic strategies have to be investigated by targeting neutrophil-derived mediators (such as metalloproteinases) and disentangling the strict interplay between neutrophils and platelets. In this view, some progress has been done by using the recombinant human interleukin-1 receptor antagonist anakinra.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
99
|
Benincasa G, Costa D, Infante T, Lucchese R, Donatelli F, Napoli C. Interplay between genetics and epigenetics in modulating the risk of venous thromboembolism: A new challenge for personalized therapy. Thromb Res 2019; 177:145-153. [DOI: 10.1016/j.thromres.2019.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 02/09/2023]
|
100
|
Elia E, Montecucco F, Portincasa P, Sahebkar A, Mollazadeh H, Carbone F. Update on pathological platelet activation in coronary thrombosis. J Cell Physiol 2019; 234:2121-2133. [PMID: 30317596 DOI: 10.1002/jcp.27575] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022]
Abstract
Although coronary thrombosis (CT) is integral to cardiovascular outcomes, the underlying pathophysiological mechanisms remain unclear. CT may occur in case of atherosclerotic plaque erosion/rupture, or even after stenting implantation. Platelets (PLT) activation is the keystone of atherothrombosis and depends on many dysregulated elements, including endothelial dysfunction, oxidized lipoproteins, and immune response. Besides the classical view of PLT as an effector of hemostatic response, a new repertoire of PLT activities is emerging. PLT lipidome oxidation is a self-maintaining process which promotes PLT reactivity, coagulation cascade, and inflammatory cell activation. PLT-innate immune cell interaction is also sustained by neutrophil extracellular traps and NLRP3 inflammasome pathways. Other noteworthy emerging mechanisms are implicated in the crosstalk between PLT and surrounding cells. Especially, microvesicles (MVs) released from PLT may extend their signaling network far beyond the classical cell-cell interactions. Moreover, the recognition of noncoding RNA in PLT MVs introduce another layer of complexity in terms of intercellular signaling by a direct regulation of messenger RNA profile and gene expression in the recipient cells. The aim of this narrative review is to update the recent advance in CT and intracoronary stent thrombosis, including causal factors and potential translation of experimental evidence into the clinical setting.
Collapse
Affiliation(s)
- Edoardo Elia
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine, First Clinic of Internal Medicine, Ospedale Policlinico San Martino, 10 Largo Benzi, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri," University of Bari Medical School, Bari, Italy
| | - Amirhossein Sahebkar
- Department of Pharmaceutical Biotechnology, Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|