1
|
Liberale L, Torino C, Pizzini P, Mezzatesta S, D'Arrigo G, Gori M, Carbone F, Schiavetta E, Cugno V, Cabri M, Sgura C, Maioli E, Mbarga D, Rubini G, Tirandi A, Ramoni D, Mallamaci F, Tripepi G, Zoccali C, Montecucco F. Plasma levels of myeloperoxidase and resistin independently predict mortality in dialysis patients. Eur J Intern Med 2024; 129:87-92. [PMID: 39019736 DOI: 10.1016/j.ejim.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND In patients with kidney failure (KF) undergoing dialysis, neutrophils are dysfunctionally activated. Such chronic activation does not correspond to increased protection against infections and is thought to cause direct vascular damage accounting for the higher incidence of cardiovascular (CV) events. We hypothesized that circulating levels of neutrophil degranulation products (i.e. myeloperoxidase (MPO) and resistin) can predict overall and CV-specific mortality in dialysis patients. METHODS MPO and resistin levels were assessed in plasma samples from n = 1182 dialysis patients who were followed-up for median 2.9 years (IQR: 1.7-4.2). RESULTS Patients were 65 ± 14 (SD) years old and 36 % women. Median value of MPO and resistin were 78 ng/mL (IQR: 54 - 123) and 72 ng/mL (IQR: 46 - 110), respectively. MPO and resistin levels correlated with biomarkers of organ damage, nutritional status and inflammation. Both MPO and resistin levels predicted all-cause mortality even after adjustment for traditional risk factors and inflammation, nutritional and KF-related indexes (MPO, HRfor 1 ln unit increase: 1.26, 95 %CI 1.11 - 1.42, P < 0.001; Resistin, HRfor 1 ln unit increase: 1.25, 95 %CI 1.09 - 1.44, P = 0.001). Similarly, their predictive ability held true also for CV death (MPO, HRfor 1 ln unit increase: 1.19, 95 %CI 1.01 - 1.41, P = 0.04; Resistin, HRfor 1 ln unit increase: 1.29, 95 %CI 1.07 - 1.56, P = 0.007). CONCLUSION Plasma levels of MPO and resistin correlate with prospective overall and CV-specific mortality risk in KF patients undergoing dialysis and might be useful prognostic tools. Mediators of inflammation may be potential target to improve survival of those patients.
Collapse
Affiliation(s)
- Luca Liberale
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Claudia Torino
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Patrizia Pizzini
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Sabrina Mezzatesta
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Graziella D'Arrigo
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | | | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Elisa Schiavetta
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Valeria Cugno
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Mara Cabri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Cosimo Sgura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Elia Maioli
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Danielle Mbarga
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Gianluca Rubini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Davide Ramoni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Francesca Mallamaci
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy; Nephrology, Hypertension and Renal Transplantation Unit, Grande Ospedale Metropolitano, Reggio Cal. Italy
| | - Giovanni Tripepi
- Clinical Epidemiology of Renal Disease and Hypertension Unit. Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, USA; IPNET, c/o Nefrologia del Grande Ospedale Metropolitano, Reggio Cal. Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|
2
|
Bertolotto M, Verzola D, Contini P, de Totero D, Tirandi A, Ramoni D, Ministrini S, Giacobbe DR, Bonaventura A, Vecchié A, Castellani L, Mirabella M, Arboscello E, Liberale L, Viazzi F, Bassetti M, Montecucco F, Carbone F. Osteopontin is associated with neutrophil extracellular trap formation in elderly patients with severe sepsis. Eur J Clin Invest 2024; 54:e14159. [PMID: 38264915 DOI: 10.1111/eci.14159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/25/2024]
Affiliation(s)
- Maria Bertolotto
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Paola Contini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela de Totero
- Molecular Pathology Unit IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Davide Ramoni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Daniele Roberto Giacobbe
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Alessandra Vecchié
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | | | | | | | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Francesca Viazzi
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Clinic of Nephrology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Matteo Bassetti
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
3
|
Liberale L, Puspitasari YM, Ministrini S, Akhmedov A, Kraler S, Bonetti NR, Beer G, Vukolic A, Bongiovanni D, Han J, Kirmes K, Bernlochner I, Pelisek J, Beer JH, Jin ZG, Pedicino D, Liuzzo G, Stellos K, Montecucco F, Crea F, Lüscher TF, Camici GG. JCAD promotes arterial thrombosis through PI3K/Akt modulation: a translational study. Eur Heart J 2023; 44:1818-1833. [PMID: 36469488 PMCID: PMC10200023 DOI: 10.1093/eurheartj/ehac641] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 09/14/2022] [Accepted: 10/26/2022] [Indexed: 12/11/2022] Open
Abstract
AIMS Variants of the junctional cadherin 5 associated (JCAD) locus associate with acute coronary syndromes. JCAD promotes experimental atherosclerosis through the large tumor suppressor kinase 2 (LATS2)/Hippo pathway. This study investigates the role of JCAD in arterial thrombosis. METHODS AND RESULTS JCAD knockout (Jcad-/-) mice underwent photochemically induced endothelial injury to trigger arterial thrombosis. Primary human aortic endothelial cells (HAECs) treated with JCAD small interfering RNA (siJCAD), LATS2 small interfering RNA (siLATS2) or control siRNA (siSCR) were employed for in vitro assays. Plasma JCAD was measured in patients with chronic coronary syndrome or ST-elevation myocardial infarction (STEMI). Jcad-/- mice displayed reduced thrombogenicity as reflected by delayed time to carotid occlusion. Mechanisms include reduced activation of the coagulation cascade [reduced tissue factor (TF) expression and activity] and increased fibrinolysis [higher thrombus embolization episodes and D-dimer levels, reduced vascular plasminogen activator inhibitor (PAI)-1 expression]. In vitro, JCAD silencing inhibited TF and PAI-1 expression in HAECs. JCAD-silenced HAECs (siJCAD) displayed increased levels of LATS2 kinase. Yet, double JCAD and LATS2 silencing did not restore the control phenotype. si-JCAD HAECs showed increased levels of phosphoinositide 3-kinases (PI3K)/ proteinkinase B (Akt) activation, known to downregulate procoagulant expression. The PI3K/Akt pathway inhibitor-wortmannin-prevented the effect of JCAD silencing on TF and PAI-1, indicating a causative role. Also, co-immunoprecipitation unveiled a direct interaction between JCAD and Akt. Confirming in vitro findings, PI3K/Akt and P-yes-associated protein levels were higher in Jcad-/- animals. Lastly, as compared with chronic coronary syndrome, STEMI patients showed higher plasma JCAD, which notably correlated positively with both TF and PAI-1 levels. CONCLUSIONS JCAD promotes arterial thrombosis by modulating coagulation and fibrinolysis. Herein, reported translational data suggest JCAD as a potential therapeutic target for atherothrombosis.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine,
University of Genoa, 6 viale Benedetto XV, 16132
Genoa, Italy
| | - Yustina M Puspitasari
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Stefano Ministrini
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine
and Surgery, University of Perugia, piazzale Gambuli 1, 06124
Perugia, Italy
| | - Alexander Akhmedov
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Simon Kraler
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Nicole R Bonetti
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital
Zurich, Rämistrasse 100, 8092 Zurich, Switzerland
| | - Georgia Beer
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Ana Vukolic
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Dario Bongiovanni
- Division of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero
Cantonale (EOC), Lugano, Switzerland
- Department of Biomedical Sciences, Humanitas University, Pieve
Emanuele, Milan, Italy
- Department of Cardiovascular Medicine, IRCCS Humanitas Research
Hospital, Rozzano, Milan, Italy
- Department of Internal Medicine I, School of Medicine, University Hospital
rechts der Isar, Technical University of Munich,
Munich, Germany
| | - Jiaying Han
- Department of Internal Medicine I, School of Medicine, University Hospital
rechts der Isar, Technical University of Munich,
Munich, Germany
| | - Kilian Kirmes
- Department of Internal Medicine I, School of Medicine, University Hospital
rechts der Isar, Technical University of Munich,
Munich, Germany
| | - Isabell Bernlochner
- Department of Internal Medicine I, School of Medicine, University Hospital
rechts der Isar, Technical University of Munich,
Munich, Germany
| | - Jaroslav Pelisek
- Department of Vascular Surgery, University Hospital Zurich,
Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden,
Im Ergel 1, 5404 Baden, Switzerland
| | - Zheng-Gen Jin
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Daniela Pedicino
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario
A. Gemelli-IRCCS, Largo A. Gemelli 8, Rome 00168,
Italy
- Cardiovascular and Pulmonary Sciences, Catholic University,
Largo G. Vito, 1 - 00168 Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario
A. Gemelli-IRCCS, Largo A. Gemelli 8, Rome 00168,
Italy
- Cardiovascular and Pulmonary Sciences, Catholic University,
Largo G. Vito, 1 - 00168 Rome, Italy
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of
Medical Sciences, Newcastle University, Newcastle Upon
Tyne, UK
- Department of Cardiology, Freeman Hospital, Newcastle upon Tyne Hospitals
NHS Foundation Trust, Newcastle Upon Tyne,
UK
- Department of Cardiovascular Research, European Center for Angioscience
(ECAS), Medical Faculty Mannheim, Heidelberg University,
Mannheim, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für
Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site,
Mannheim, Germany
- Department of Cardiology, University Hospital Mannheim,
Mannheim, Germany
| | - Fabrizio Montecucco
- 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, L.go R. Benzi 10, 16132 Genoa, Italy
| | - Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario
A. Gemelli-IRCCS, Largo A. Gemelli 8, Rome 00168,
Italy
- Cardiovascular and Pulmonary Sciences, Catholic University,
Largo G. Vito, 1 - 00168 Rome, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals and Nationl Heart
and Lung Institute, Imperial College, London,
United Kingdom
| | - Giovanni G Camici
- Center for Molecular Cardiology, Schlieren Campus, University of
Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Research and Education, University Hospital
Zurich, Rämistrasse 100, 8092 Zurich, Switzerland
| |
Collapse
|
4
|
Liberale L, Kraler S, Puspitasari YM, Bonetti NR, Akhmedov A, Ministrini S, Montecucco F, Marx N, Lehrke M, Hartmann NUK, Beer JH, Wenzl FA, Paneni F, Lüscher TF, Camici GG. SGLT-2 inhibition by empagliflozin has no effect on experimental arterial thrombosis in a murine model of low-grade inflammation. Cardiovasc Res 2023; 119:843-856. [PMID: 35993135 DOI: 10.1093/cvr/cvac126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/04/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Low-grade inflammation couples dysmetabolic states to insulin resistance and atherosclerotic cardiovascular (CV) disease (ASCVD). Selective sodium-glucose co-transporter 2 (SGLT-2) inhibition by empagliflozin improves clinical outcomes in patients with ASCVD independently of its glucose lowering effects. Yet, its mechanism of action remains largely undetermined. Here, we aimed to test whether empagliflozin affects arterial thrombus formation in baseline (BSL) conditions or low-grade inflammatory states, a systemic milieu shared among patients with ASCVD. METHODS AND RESULTS Sixteen-week-old C57BL/6 mice were randomly assigned to acute administration of empagliflozin (25 mg/kg body weight) or vehicle, of which a subgroup was pre-treated biweekly over 4 weeks with super-low-dose lipopolysaccharide (LPS; 5 ng/kg body weight), before carotid thrombosis was induced by photochemical injury. The between-group difference in Doppler-flow probe detected time-to-occlusion remained within the predefined equivalence margin (Δ = |10.50|), irrespective of low-grade inflammation (95% confidence interval, -9.82 to 8.85 and -9.20 to 9.69), while glucose dropped by 1.64 and 4.84 mmoL/L, respectively. Ex vivo platelet aggregometry suggested similar activation status, corroborated by unchanged circulating platelet-factor 4 plasma levels. In concert, carotid PAI-1 expression and tissue factor (TF) activity remained unaltered upon SGLT-2 inhibition, and no difference in plasma D-dimer levels was detected, suggesting comparable coagulation cascade activation and fibrinolytic activity. In human aortic endothelial cells pre-treated with LPS, empagliflozin neither changed TF activity nor PAI-1 expression. Accordingly, among patients with established ASCVD or at high CV risk randomized to a daily dose of 10 mg empagliflozin signatures of thrombotic (i.e. TF) and fibrinolytic activity (i.e. PAI-1) remained unchanged, while plasma glucose declined significantly during 3 months of follow-up. CONCLUSION SGLT-2 inhibition by empagliflozin does not impact experimental arterial thrombus formation, neither under BSL conditions nor during sustained low-grade inflammation, and has no impact on proxies of thrombotic/fibrinolytic activity in patients with ASCVD. The beneficial pleiotropic effects of empagliflozin are likely independent of pathways mediating arterial thrombosis.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Yustina M Puspitasari
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Nicole R Bonetti
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy
| | - 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
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, RWTH, 52074 Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital Aachen, RWTH, 52074 Aachen, Germany
| | - Niels-Ulrik K Hartmann
- Department of Internal Medicine I, University Hospital Aachen, RWTH, 52074 Aachen, Germany
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, 5404 Baden, Switzerland
| | - Florian A Wenzl
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, SW3 6NP London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
5
|
Tirandi A, Sgura C, Carbone F, Montecucco F, Liberale L. Inflammatory biomarkers of ischemic stroke. Intern Emerg Med 2023; 18:723-732. [PMID: 36745280 PMCID: PMC10082112 DOI: 10.1007/s11739-023-03201-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/09/2023] [Indexed: 02/07/2023]
Abstract
Ischemic stroke remains the second leading cause of death and among the major causes of morbidity worldwide. Therapeutic options are currently limited to early reperfusion strategies, while pharmacological neuroprotective strategies despite showing promising results in the experimental setting constantly failed to enter the clinical arena. Inflammation plays an important role in the pathophysiology of ischemic stroke and mediators of inflammation have been longtime investigated as possible prognostic marker and therapeutic target for stroke patients. Here, we summarized available evidence on the role of cytokines, soluble adhesion molecules and adipokines in the pathophysiology, prognosis and therapy of ischemic stroke.
Collapse
Affiliation(s)
- Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Cosimo Sgura
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| |
Collapse
|
6
|
Pekkarinen PT, Carbone F, Minetti S, Ramoni D, Ristagno G, Latini R, Wihersaari L, Blennow K, Zetterberg H, Toppila J, Jakkula P, Reinikainen M, Montecucco F, Skrifvars MB. Markers of neutrophil mediated inflammation associate with disturbed continuous electroencephalogram after out of hospital cardiac arrest. Acta Anaesthesiol Scand 2023; 67:94-103. [PMID: 36053856 PMCID: PMC10087484 DOI: 10.1111/aas.14145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/06/2022] [Accepted: 08/17/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Achieving an acceptable neurological outcome in cardiac arrest survivors remains challenging. Ischemia-reperfusion injury induces inflammation, which may cause secondary neurological damage. We studied the association of ICU admission levels of inflammatory biomarkers with disturbed 48-hour continuous electroencephalogram (cEEG), and the association of the daily levels of these markers up to 72 h with poor 6-month neurological outcome. METHODS This is an observational, post hoc sub-study of the COMACARE trial. We measured serum concentrations of procalcitonin (PCT), high-sensitivity C-reactive protein (hsCRP), osteopontin (OPN), myeloperoxidase (MPO), resistin, and proprotein convertase subtilisin/kexin type 9 (PCSK9) in 112 unconscious, mechanically ventilated ICU-treated adult OHCA survivors with initial shockable rhythm. We used grading of 48-hour cEEG monitoring as a measure for the severity of the early neurological disturbance. We defined 6-month cerebral performance category (CPC) 1-2 as good and CPC 3-5 as poor long-term neurological outcome. We compared the prognostic value of biomarkers for 6-month neurological outcome to neurofilament light (NFL) measured at 48 h. RESULTS Higher OPN (p = .03), MPO (p < .01), and resistin (p = .01) concentrations at ICU admission were associated with poor grade 48-hour cEEG. Higher levels of ICU admission OPN (OR 3.18; 95% CI 1.25-8.11 per ln[ng/ml]) and MPO (OR 2.34; 95% CI 1.30-4.21) were independently associated with poor 48-hour cEEG in a multivariable logistic regression model. Poor 6-month neurological outcome was more common in the poor cEEG group (63% vs. 19% p < .001, respectively). We found a significant fixed effect of poor 6-month neurological outcome on concentrations of PCT (F = 7.7, p < .01), hsCRP (F = 4.0, p < .05), and OPN (F = 5.6, p < .05) measured daily from ICU admission to 72 h. However, the biomarkers did not have independent predictive value for poor 6-month outcome in a multivariable logistic regression model with 48-hour NFL. CONCLUSION Elevated ICU admission levels of OPN and MPO predicted disturbances in cEEG during the subsequent 48 h after cardiac arrest. Thus, they may provide early information about the risk of secondary neurological damage. However, the studied inflammatory markers had little value for long-term prognostication compared to 48-hour NFL.
Collapse
Affiliation(s)
- Pirkka T. Pekkarinen
- Division of Intensive Care, Department of Anaesthesiology, Intensive Care and Pain MedicineUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Federico Carbone
- Department of Internal MedicineIRCCS Ospedale Policlinico San Martino Genoa–Italian Cardiovascular NetworkGenoaItaly
- First Clinic of Internal Medicine, Department of Internal MedicineUniversity of GenoaGenoaItaly
| | - Silvia Minetti
- Department of Internal MedicineIRCCS Ospedale Policlinico San Martino Genoa–Italian Cardiovascular NetworkGenoaItaly
- First Clinic of Internal Medicine, Department of Internal MedicineUniversity of GenoaGenoaItaly
| | - Davide Ramoni
- Department of Internal MedicineIRCCS Ospedale Policlinico San Martino Genoa–Italian Cardiovascular NetworkGenoaItaly
- First Clinic of Internal Medicine, Department of Internal MedicineUniversity of GenoaGenoaItaly
| | - Giuseppe Ristagno
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Department of Anesthesiology, Intensive Care and EmergencyFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Roberto Latini
- Cardiovascular MedicineMario Negri Institute for Pharmacological Research IRCCSMilanItaly
| | - Lauri Wihersaari
- Department of Anaesthesiology and Intensive CareKuopio University Hospital and University of Eastern FinlandKuopioFinland
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Jussi Toppila
- Department of Clinical Neurophysiology, Medical Imaging Center, Helsinki University Central Hospital and Department of Clinical Neurosciences (Neurophysiology)University of HelsinkiHelsinkiFinland
| | - Pekka Jakkula
- Division of Intensive Care, Department of Anaesthesiology, Intensive Care and Pain MedicineUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Matti Reinikainen
- Department of Anaesthesiology and Intensive CareKuopio University Hospital and University of Eastern FinlandKuopioFinland
| | - Fabrizio Montecucco
- Department of Internal MedicineIRCCS Ospedale Policlinico San Martino Genoa–Italian Cardiovascular NetworkGenoaItaly
- First Clinic of Internal Medicine, Department of Internal MedicineUniversity of GenoaGenoaItaly
| | - Markus B. Skrifvars
- Department of Emergency Care and ServicesUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| |
Collapse
|
7
|
Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci 2022; 79:272. [PMID: 35503385 PMCID: PMC11073100 DOI: 10.1007/s00018-022-04286-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/11/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022]
Abstract
Characterized by a surplus of whole-body adiposity, obesity is strongly associated with the prognosis of atherosclerosis, a hallmark of coronary artery disease (CAD) and the major contributor to cardiovascular disease (CVD) mortality. Adipose tissue serves a primary role as a lipid-storage organ, secreting cytokines known as adipokines that affect whole-body metabolism, inflammation, and endocrine functions. Emerging evidence suggests that adipokines can play important roles in atherosclerosis development, progression, as well as regression. Here, we review the versatile functions of various adipokines in atherosclerosis and divide these respective functions into three major groups: protective, deteriorative, and undefined. The protective adipokines represented here are adiponectin, fibroblast growth factor 21 (FGF-21), C1q tumor necrosis factor-related protein 9 (CTRP9), and progranulin, while the deteriorative adipokines listed include leptin, chemerin, resistin, Interleukin- 6 (IL-6), and more, with additional adipokines that have unclear roles denoted as undefined adipokines. Comprehensively categorizing adipokines in the context of atherosclerosis can help elucidate the various pathways involved and potentially pave novel therapeutic approaches to treat CVDs.
Collapse
Affiliation(s)
- Longhua Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Zunhan Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiaohui Ji
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Wenqian Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jinwen Luan
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Tarik Zahr
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Li Qiang
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
8
|
Carbone F, Meessen J, Magnoni M, Andreini D, Maggioni AP, Latini R, Montecucco F. Osteopontin as Candidate Biomarker of Coronary Disease despite Low Cardiovascular Risk: Insights from CAPIRE Study. Cells 2022; 11:669. [PMID: 35203321 PMCID: PMC8870389 DOI: 10.3390/cells11040669] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
Stratification according high cardiovascular (CV) risk categories, still represents a clinical challenge. In this analysis of the CAPIRE study (NCT02157662), we investigate whether inflammation could fit between CV risk factors (RFs) and the presence of coronary artery disease (CAD). In total, 544 patients were included and categorized according with the presence of CAD and CV risk factor burden (low/multiple). The primary endpoint was to verify any independent association of neutrophil-related biomarkers with CAD across CV risk categories. The highest values of osteopontin (OPN) were detected in the low RF group and associated with CAD (23.2 vs. 19.4 ng/mL; p = 0.001), although no correlation with plaque extent and/or composition were observed. Conversely, myeloperoxidase (MPO) and resistin did not differ by CAD presence. Again, OPN was identified as independent variable associated with CAD but only in the low RF group (adjOR 8.42 [95% CI 8.42-46.83]; p-value = 0.015). As an ancillary finding, a correlation linked OPN with the neutrophil degranulation biomarker MPO (r = 0.085; p = 0.048) and resistin (r = 0.177; p = 3.4 × 10-5). In the present study, OPN further strengthens its role as biomarker of CAD, potentially bridging subclinical CV risk with development of atherosclerosis.
Collapse
Affiliation(s)
- 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
| | - Jennifer Meessen
- Department of Cardiovascular Research, IRCCS—Istituto di Ricerche Farmacologiche Mario Negri, 19 Via Giuseppe La Masa, 20156 Milan, Italy; (J.M.); (R.L.)
| | | | - Daniele Andreini
- Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy;
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, University of Milan, 20157 Milan, Italy
| | | | - Roberto Latini
- Department of Cardiovascular Research, IRCCS—Istituto di Ricerche Farmacologiche Mario Negri, 19 Via Giuseppe La Masa, 20156 Milan, Italy; (J.M.); (R.L.)
| | - Fabrizio Montecucco
- 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
| |
Collapse
|
9
|
Tirandi A, Carbone F, Montecucco F, Liberale L. The role of metabolic syndrome in sudden cardiac death risk: Recent evidence and future directions. Eur J Clin Invest 2022; 52:e13693. [PMID: 34714544 PMCID: PMC9286662 DOI: 10.1111/eci.13693] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/23/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a frequent condition whose deleterious effects on the cardiovascular system are often underestimated. MetS is nowadays considered a real pandemic with an estimated prevalence of 25% in general population. Individuals with MetS are at high risk of sudden cardiac death (SCD) as this condition accounts for 50% of all cardiac deaths in such a population. Of interest, recent studies demonstrated that individuals with MetS show 70% increased risk of SCD even without previous history of coronary heart disease (CHD). However, little is known about the interplay between the two conditions. MetS is a complex disease determined by genetic predisposition, unhealthy lifestyle and ageing with deleterious effects on different organs. MetS components trigger a systemic chronic low-grade pro-inflammatory state, associated with excess of sympathetic activity, cardiac hypertrophy, arrhythmias and atherosclerosis. Thus, MetS has an important burden on the cardiovascular system as demonstrated by both preclinical and clinical evidence. The aim of this review is to summarize recent evidence concerning the association between MetS and SCD, showing possible common aetiological processes, and to indicate prospective for future studies and therapeutic targets.
Collapse
Affiliation(s)
- Amedeo Tirandi
- First Clinic of Internal MedicineDepartment of Internal MedicineUniversity of GenoaGenoaItaly
| | - Federico Carbone
- First Clinic of Internal MedicineDepartment of Internal MedicineUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular NetworkGenoaItaly
| | - Fabrizio Montecucco
- First Clinic of Internal MedicineDepartment of Internal MedicineUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular NetworkGenoaItaly
| | - Luca Liberale
- First Clinic of Internal MedicineDepartment of Internal MedicineUniversity of GenoaGenoaItaly
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
| |
Collapse
|
10
|
Gene Differential Expression and Interaction Networks Illustrate the Biomarkers and Molecular Mechanisms of Atherosclerotic Cerebral Infarction. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3912697. [PMID: 35070236 PMCID: PMC8769835 DOI: 10.1155/2022/3912697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/18/2022]
Abstract
Atherosclerotic cerebral infarction (ACI) seriously threatens the health of the senile patients, and the strategies are urgent for the diagnosis and treatment of ACI. This study investigated the mRNA profiling of the patients with ischemic stroke and atherosclerosis via excavating the datasets in the GEO database and attempted to reveal the biomarkers and molecular mechanism of ACI. In this study, GES16561 and GES100927 were obtained from Gene Expression Omnibus (GEO) database, and the related differentially expressed genes (DEGs) were analyzed with R language. Furthermore, the DEGs were analyzed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Besides, the protein-protein interaction (PPI) network of DEGs was analyzed by STRING database and Cytoscape. The results showed that 133 downregulated DEGs and 234 upregulated DEGs were found in GES16561, 25 downregulated DEGs and 104 upregulated DEGs were found in GSE100927, and 6 common genes were found in GES16561 and GES100927. GO enrichment analysis showed that the functional models of the common genes were involved in neutrophil activation, neutrophil degranulation, neutrophil activation, and immune response. KEGG enrichment analysis showed that the DEGs in both GSE100927 and GSE16561 were connected with the pathways including Cell adhesion molecules (CAMs), Cytokine-cytokine receptor interaction, Phagosome, Antigen processing and presentation, and Staphylococcus aureus infection. The PPI network analysis showed that 9 common DEGs were found in GSE100927 and GSE16561, and a cluster with 6 nodes and 12 edges was also identified by PPI network analysis. In conclusion, this study suggested that FCGR3A and MAPK pathways were connected with ACI.
Collapse
|
11
|
Ministrini S, Andreozzi F, Montecucco F, Minetti S, Bertolotto M, Liberale L, Mannino GC, Succurro E, Cassano V, Miceli S, Perticone M, Sesti G, Sciacqua A, Carbone F. Neutrophil degranulation biomarkers characterize restrictive echocardiographic pattern with diastolic dysfunction in patients with diabetes. Eur J Clin Invest 2021; 51:e13640. [PMID: 34129696 PMCID: PMC9286613 DOI: 10.1111/eci.13640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/28/2021] [Accepted: 06/11/2021] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To investigate the potential association between neutrophil degranulation and patterns of myocardial dysfunction in a cohort of patients with type 2 diabetes mellitus (T2DM). BACKGROUND Two distinct phenotypes of diabetic cardiomyopathy have been described: a restrictive phenotype with diastolic dysfunction (restrictive/DD) and a dilative phenotype with systolic dysfunction (dilative/SD). However, the underlying determinants of these two patterns are not yet recognized. METHODS In this single-centre, observational, cross-sectional study, 492 patients were recruited. Ultrasonographic measurements were performed by two experienced sonographers, blinded to the clinical data of the participants. Serum biomarkers of neutrophil degranulation were measured by enzyme-linked immunosorbent sandwich assay (ELISA). RESULTS After adjustment for confounders, resistin, myeloperoxidase, matrix metalloproteinase 8 and matrix metalloproteinase 9/tissue inhibitor of metalloproteinases 1 complex were positively associated with the restrictive/DD pattern compared with the normal pattern. Similarly, MPO was positively associated with the dilative/SD pattern compared with the normal pattern, and resistin was negatively associated with the dilative/SD pattern compared with the restrictive/DD pattern. CONCLUSIONS Neutrophil degranulation is associated with the restrictive/DD echocardiographic pattern in patients with T2DM, but not with the normal pattern and dilative/SD patterns. Neutrophils could have a pivotal role in the pathogenesis of myocardial dysfunction, and particularly diastolic dysfunction, in patients with T2DM.
Collapse
MESH Headings
- Aged
- Biomarkers/metabolism
- Cardiomyopathy, Dilated/diagnostic imaging
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/physiopathology
- Cardiomyopathy, Restrictive/diagnostic imaging
- Cardiomyopathy, Restrictive/etiology
- Cardiomyopathy, Restrictive/metabolism
- Cardiomyopathy, Restrictive/physiopathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Cardiomyopathies/diagnostic imaging
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Echocardiography
- Female
- Heart Failure, Diastolic/diagnostic imaging
- Heart Failure, Diastolic/etiology
- Heart Failure, Diastolic/metabolism
- Heart Failure, Diastolic/physiopathology
- Heart Failure, Systolic/diagnostic imaging
- Heart Failure, Systolic/etiology
- Heart Failure, Systolic/metabolism
- Heart Failure, Systolic/physiopathology
- Humans
- Male
- Matrix Metalloproteinase 8/metabolism
- Matrix Metalloproteinase 9/metabolism
- Middle Aged
- Neutrophil Activation
- Peroxidase/metabolism
- Resistin/metabolism
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Tissue Inhibitor of Metalloproteinase-2/metabolism
Collapse
Affiliation(s)
- Stefano Ministrini
- Internal Medicine, Angiology and AtherosclerosisDepartment of Medicine and SurgeryUniversità degli Studi di PerugiaPerugiaItaly
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
| | - Francesco Andreozzi
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Fabrizio Montecucco
- Department of Internal MedicineFirst Clinic of internal MedicineUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoa – Italian Cardiovascular NetworkGenoaItaly
| | - Silvia Minetti
- Internal Medicine, Angiology and AtherosclerosisDepartment of Medicine and SurgeryUniversità degli Studi di PerugiaPerugiaItaly
| | - Maria Bertolotto
- Internal Medicine, Angiology and AtherosclerosisDepartment of Medicine and SurgeryUniversità degli Studi di PerugiaPerugiaItaly
| | - Luca Liberale
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
- Department of Internal MedicineFirst Clinic of internal MedicineUniversity of GenoaGenoaItaly
| | - Gaia Chiara Mannino
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Elena Succurro
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Velia Cassano
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Sofia Miceli
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Maria Perticone
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Giorgio Sesti
- Department of Clinical and Molecular MedicineUniversity of Rome‐SapienzaRomeItaly
| | - Angela Sciacqua
- Department of Medical and Surgical SciencesUniversity ‘Magna Græcia’ of CatanzaroCatanzaroItaly
| | - Federico Carbone
- Department of Internal MedicineFirst Clinic of internal MedicineUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoa – Italian Cardiovascular NetworkGenoaItaly
| |
Collapse
|
12
|
Liberale L, Akhmedov A, Vlachogiannis NI, Bonetti NR, Nageswaran V, Miranda MX, Puspitasari YM, Schwarz L, Costantino S, Paneni F, Beer JH, Ruschitzka F, Montecucco F, Lüscher TF, Stamatelopoulos K, Stellos K, Camici GG. Sirtuin 5 promotes arterial thrombosis by blunting the fibrinolytic system. Cardiovasc Res 2021; 117:2275-2288. [PMID: 32931562 DOI: 10.1093/cvr/cvaa268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/07/2020] [Accepted: 09/03/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Arterial thrombosis as a result of plaque rupture or erosion is a key event in acute cardiovascular events. Sirtuin 5 (SIRT5) belongs to the lifespan-regulating sirtuin superfamily and has been implicated in acute ischaemic stroke and cardiac hypertrophy. This project aims at investigating the role of SIRT5 in arterial thrombus formation. METHODS AND RESULTS Sirt5 transgenic (Sirt5Tg/0) and knock-out (Sirt5-/-) mice underwent photochemically induced carotid endothelial injury to trigger arterial thrombosis. Primary human aortic endothelial cells (HAECs) were treated with SIRT5 silencing-RNA (si-SIRT5) as well as peripheral blood mononuclear cells from acute coronary syndrome (ACS) patients and non-ACS controls (case-control study, total n = 171) were used to increase the translational relevance of our data. Compared to wild-type controls, Sirt5Tg/0 mice displayed accelerated arterial thrombus formation following endothelial-specific damage. Conversely, in Sirt5-/- mice, arterial thrombosis was blunted. Platelet function was unaltered, as assessed by ex vivo collagen-induced aggregometry. Similarly, activation of the coagulation cascade as assessed by vascular and plasma tissue factor (TF) and TF pathway inhibitor expression was unaltered. Increased thrombus embolization episodes and circulating D-dimer levels suggested augmented activation of the fibrinolytic system in Sirt5-/- mice. Accordingly, Sirt5-/- mice showed reduced plasma and vascular expression of the fibrinolysis inhibitor plasminogen activator inhibitor (PAI)-1. In HAECs, SIRT5-silencing inhibited PAI-1 gene and protein expression in response to TNF-α. This effect was mediated by increased AMPK activation and reduced phosphorylation of the MAP kinase ERK 1/2, but not JNK and p38 as shown both in vivo and in vitro. Lastly, both PAI-1 and SIRT5 gene expressions are increased in ACS patients compared to non-ACS controls after adjustment for cardiovascular risk factors, while PAI-1 expression increased across tertiles of SIRT5. CONCLUSION SIRT5 promotes arterial thrombosis by modulating fibrinolysis through endothelial PAI-1 expression. Hence, SIRT5 may be an interesting therapeutic target in the context of atherothrombotic events.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Alexander Akhmedov
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Nikolaos I Vlachogiannis
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Framlington Place, NE2 4HH Newcastle upon Tyne, UK
| | - Nicole R Bonetti
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Vanasa Nageswaran
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Melroy X Miranda
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Yustina M Puspitasari
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Lena Schwarz
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 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
| | - Jürg H Beer
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8092 Zurich, Switzerland
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, L.go R. 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, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, Dovehouse Street, London SW3 6LY, UK
| | - Kimon Stamatelopoulos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Framlington Place, NE2 4HH Newcastle upon Tyne, UK
- Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Lourou 4-2, 115 28 Athens, Greece
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Framlington Place, NE2 4HH Newcastle upon Tyne, UK
- Department of Cardiology, Newcastle Hospitals NHS Foundation Trust, Freeman Rd, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistrasse 12, 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
- Zurich Neuroscience Center, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
13
|
Lv M, Liu W. Hypoxia-Induced Mitogenic Factor: A Multifunctional Protein Involved in Health and Disease. Front Cell Dev Biol 2021; 9:691774. [PMID: 34336840 PMCID: PMC8319639 DOI: 10.3389/fcell.2021.691774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/23/2021] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-induced mitogenic factor (HIMF), also known as resistin-like molecule α (RELMα) or found in inflammatory zone 1 (FIZZ1) is a member of the RELM protein family expressed in mice. It is involved in a plethora of physiological processes, including mitogenesis, angiogenesis, inflammation, and vasoconstriction. HIMF expression can be stimulated under pathological conditions and this plays a critical role in pulmonary, cardiovascular and metabolic disorders. The present review summarizes the molecular characteristics, and the physiological and pathological roles of HIMF in normal and diseased conditions. The potential clinical significance of these findings for human is also discussed.
Collapse
Affiliation(s)
- Moyang Lv
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wenjuan Liu
- Department of Pathophysiology, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
14
|
Shen X, Li H, Zou WJ, Wu JM, Wang L, Wang W, Chen H, Zhou LL, Hu YH, Qin XH, Yang J. Network Pharmacology Analysis of the Therapeutic Mechanisms Underlying Beimu-Gualou Formula Activity against Bronchiectasis with In Silico Molecular Docking Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:3656272. [PMID: 33488758 PMCID: PMC7803403 DOI: 10.1155/2021/3656272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/14/2020] [Accepted: 12/19/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND The classical Chinese herbal prescription Beimu-Gualou formula (BMGLF) has been diffusely applied to the treatment of respiratory diseases, including bronchiectasis. Although concerning bronchiectasis the effects and mechanisms of action of the BMGLF constituents have been partially elucidated, it remains to be determined how the formula in its entirety exerts therapeutic effects. METHODS In this study, the multitarget mechanisms of BMGLF against bronchiectasis were predicted with network pharmacology analysis. Using prepared data, a drug-target interaction network was established and subsequently the core therapeutic targets of BMGLF were identified. Furthermore, the biological function and pathway enrichment of potential targets were analyzed to evaluate the therapeutic effects and pivotal signaling pathways of BMGLF. Finally, virtual molecular docking was performed to assess the affinities of compounds for the candidate targets. RESULTS The therapeutic action of BMGLF against bronchiectasis involves 18 core target proteins, including the aforementioned candidates (i.e., ALB, ICAM1, IL10, and MAPK1), which are assumed to be related to biological processes such as drug response, cellular response to lipopolysaccharide, immune response, and positive regulation of NF-κB activity in bronchiectasis. Among the top 20 signaling pathways identified, mechanisms of action appear to be primarily related to Chagas disease, allograft rejection, hepatitis B, and inflammatory bowel disease. CONCLUSION In summary, using a network pharmacology approach, we initially predicted the complex regulatory profile of BMGLF against bronchiectasis in which multilink suppression of immune/inflammatory responses plays an essential role. These results may provide a basis for novel pharmacotherapeutic approaches for bronchiectasis.
Collapse
Affiliation(s)
- Xin Shen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Hong Li
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wen-Jun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Jian-Ming Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wei Wang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Ling-Li Zhou
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yuan-Hui Hu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xu-Hua Qin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Jing Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
15
|
Christou KA, Christou GA, Karamoutsios A, Vartholomatos G, Gartzonika K, Tsatsoulis A, Tigas S. The regulation of serum resistin levels in metabolically healthy and unhealthy obese individuals. Hormones (Athens) 2020; 19:523-529. [PMID: 32328905 DOI: 10.1007/s42000-020-00201-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/07/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE We have recently demonstrated that absolute counts of circulating proinflammatory monocytes were lower in obese patients without metabolic syndrome (MS) (metabolically healthy obese, MHO) compared with those with MS (metabolically unhealthy obese, MUO), but higher compared with healthy lean controls (MHL). We hypothesized that circulating resistin, a cytokine secreted by white blood cells (WBC), is involved in obesity-related low-grade inflammation. The aim of this study was to (a) determine serum resistin levels among MUO and MHO subjects and (b) investigate the role of circulating WBC subsets as potential determinants of resistin. METHODS Study participants were 58 obese (33 MUO, 25 MHO) and 25 MHL individuals. Serum levels of resistin, high-sensitivity C-reactive protein (hsCRP), and absolute counts of circulating WBC subpopulations were determined. Comparisons were sex- and age-adjusted. RESULTS Serum resistin levels in MHL were lower compared with those of obese (p = 0.041), but similar to those of MHO (p = 0.856) individuals. Both resistin (p = 0.005) and absolute neutrophil count (NeuA) (p = 0.025) were higher in MUO compared with MHO. The difference in resistin levels between obese and MHL individuals disappeared after adjustment for NeuA. Resistin correlated positively with absolute total monocyte count (p = 0.037) in MHL and with body mass index (BMI) (p = 0.023), hsCRP (p = 0.022), and NeuA (p = 0.044) in obese subjects. Resistin association with ΒΜΙ disappeared after adjustment for hsCRP, while association with hsCRP disappeared after further adjustment for NeuA. CONCLUSION Circulating resistin was higher in MUO compared with MHO. The increased secretion of resistin by the greater number of neutrophils in the former may have contributed to this regulation.
Collapse
Affiliation(s)
| | - Georgios A Christou
- Department of Endocrinology, University of Ioannina, 45110, Ioannina, Greece
| | - Achilleas Karamoutsios
- Laboratory of Haematology, Molecular Biology Unit, Ioannina University Hospital, Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology, Molecular Biology Unit, Ioannina University Hospital, Ioannina, Greece
| | - Konstantina Gartzonika
- Laboratory of Immunology and Microbiology, School of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Stelios Tigas
- Department of Endocrinology, University of Ioannina, 45110, Ioannina, Greece.
| |
Collapse
|
16
|
Wijeratne T, Menon R, Sales C, Karimi L, Crewther S. Carotid artery stenosis and inflammatory biomarkers: the role of inflammation-induced immunological responses affecting the vascular systems. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1276. [PMID: 33178808 PMCID: PMC7607082 DOI: 10.21037/atm-20-4388] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The death, disability and economic cost of stroke are enormous. Indeed, among the 16 million people worldwide who suffer a stroke' annually, nearly six million die, and another five million are left permanently disabled making prevention of stroke one of the most important priorities in healthcare. Currently carotid artery stenosis (CS) or narrowing of the common carotid artery (CCA) or internal carotid artery (ICA) due to atherosclerotic plaque, accounts for 20-30% of all ischemic strokes. Atherosclerosis is now regarded as a chronic inflammatory disease in response to vascular compromise especially from hypertension. This has long been known to lead to inflammation and atherosclerotic plaque formation in the blood vessels. This mini-review aims to highlight the role of inflammation and neuro-immunological processes in carotid artery disease. Various cellular elements of inflammation and advanced imaging techniques have been identified as potential markers of plaque progression. Therapies related to decreasing and modulating immune-responsive inflammation in the carotid vessels have been shown to translate into decreased occurrence of acute neurologic events and improvement of clinical outcomes.
Collapse
Affiliation(s)
- Tissa Wijeratne
- Department of Neurology, AIMSS, WHCRE level three, Sunshine Hospital and Melbourne Medical School, St Albans, Victoria, Australia.,School of Public health and Psychology, La Trobe University, Bundoora, Victoria, Australia.,Department of Medicine, Faculty of Medicine, Rajarata University, Saliyapura, Anuradhapura, Sri Lanka
| | - Rohit Menon
- Department of Neurology and Stroke Service, Western Health, AIMSS, level 3, WHCRE, Sunshine Hospital, St Albans, Victoria, Australia
| | - Carmela Sales
- Department of Neurology and Stroke Service, Western Health, AIMSS, level 3, WHCRE, Sunshine Hospital, St Albans, Victoria, Australia
| | - Leila Karimi
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia.,Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Sheila Crewther
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Bonaventura A, Grossi F, Carbone F, Vecchié A, Minetti S, Bardi N, Elia E, Ansaldo AM, Ferrara D, Rijavec E, Dal Bello MG, Rossi G, Biello F, Tagliamento M, Alama A, Coco S, Spallarossa P, Dallegri F, Genova C, Montecucco F. Resistin is associated with overall survival in non-small cell lung cancer patients during nivolumab treatment. Clin Transl Oncol 2020; 22:1603-1610. [PMID: 32048158 DOI: 10.1007/s12094-020-02305-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Since the role of resistin was evaluated only in patients with non-small cell lung cancer (NSCLC) not treated with immunotherapy, we aimed to evaluate levels of resistin during immunotherapy (nivolumab) and its prognostic role with regard to OS. METHODS/PATIENTS From a cohort of 78 patients with advanced NSCLC enrolled in a prospective study at Ospedale Policlinico San Martino in Genoa (Italy), 43 patients have been considered for this sub-analysis because of the availability of samples. Before and during nivolumab administration, clinical information and blood samples were collected and resistin, matrix metalloproteinase (MMP)-8, MMP-9, and myeloperoxidase were evaluated by enzyme-linked immunosorbent assay (ELISA). RESULTS Median age was 71 with a prevalence of males and former smokers. Median resistin levels presented a peak at cycle 2 and then dropped down until the last cycle. Resistin correlated with all neutrophil degranulation products at cycle 1 (except for MMP-9) and at cycle 2 as well as with white blood cells and neutrophils. By a ROC curve analysis, a resistin value at cycle 2 of 19 ng/mL was tested as the best cut-off point for OS. Kaplan-Meier analysis demonstrated that patients above the resistin cut-off experienced a reduced OS (median OS 242.5 vs. 470 days, p = 0.0073), as confirmed by Cox proportional hazards regression analysis. CONCLUSIONS Resistin levels > 19 ng/mL at the time of the second cycle of nivolumab treatment independently predict a reduced OS in patients with advanced NSCLC.
Collapse
Affiliation(s)
- A Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy.
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA, 23298, USA.
| | - F Grossi
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Medical Oncology, Milan, Italy
| | - F Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, 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
| | - A Vecchié
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA, 23298, USA
| | - S Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy
| | - N Bardi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy
| | - E Elia
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy
| | - A M Ansaldo
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy
| | - D Ferrara
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132, Genoa, Italy
| | - E Rijavec
- IRCCS Ospedale Policlinico San Martino, UOS Tumori Polmonari, Largo R. Benzi 10, 16132, Genoa, Italy
| | - M G Dal Bello
- IRCCS Ospedale Policlinico San Martino, UOS Tumori Polmonari, Largo R. Benzi 10, 16132, Genoa, Italy
| | - G Rossi
- IRCCS Ospedale Policlinico San Martino, UOS Tumori Polmonari, Largo R. Benzi 10, 16132, Genoa, Italy
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - F Biello
- Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
| | - M Tagliamento
- IRCCS Ospedale Policlinico San Martino, UOS Tumori Polmonari, Largo R. Benzi 10, 16132, Genoa, Italy
| | - A Alama
- IRCCS Ospedale Policlinico San Martino, UOS Tumori Polmonari, Largo R. Benzi 10, 16132, Genoa, Italy
| | - S Coco
- IRCCS Ospedale Policlinico San Martino, UOS Tumori Polmonari, Largo R. Benzi 10, 16132, Genoa, Italy
| | - P Spallarossa
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino-Italian Cardiovascular Network, Genoa, Italy
| | - F Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, 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
| | - C Genova
- IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Largo R. Benzi 10, 16132, Genoa, Italy
| | - F Montecucco
- IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Largo R. 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, viale Benedetto XV 6, 16132, Genoa, Italy
| |
Collapse
|
20
|
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
|
21
|
Liberale L, Montecucco F. Adipocytokines and cardiovascular diseases: Putative role of Neuregulin 4. Eur J Clin Invest 2020; 50:e13306. [PMID: 32511758 DOI: 10.1111/eci.13306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/30/2020] [Indexed: 01/26/2023]
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Affiliation(s)
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany.
| |
Collapse
|