1
|
Laspa Z, Dicenta-Baunach V, Schaale D, Sigle M, Hochuli R, Castor T, Bayrak A, Harm T, Müller KAL, Pillaiyar T, Laufer S, Rohlfing AK, Gawaz MP. Hemin-induced platelet activation is regulated by the ACKR3 chemokine surface receptor and has implications for passivation of vulnerable atherosclerotic plaques. FEBS J 2024. [PMID: 39387619 DOI: 10.1111/febs.17294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/11/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
In vulnerable atherosclerotic plaques, intraplaque hemorrhages (IPH) result in hemolysis of red blood cells and release of hemoglobin and free hemin. Hemin activates platelets and leads to thrombosis. Agonism of the inhibitory platelet receptor ACKR3 inhibits hemin-dependent platelet activation and thrombus formation. To characterize the effect of hemin and ACKR3 agonism on isolated human platelets, multi-color flow cytometry and classical experimental setup such as light transmission aggregometry and a flow chamber assay were used. Hemin induces platelet aggregation and ex vivo platelet-dependent thrombus formation on immobilized collagen under a low shear rate of 500 s-1, indicating that free hemin is a strong activator of platelet-dependent thrombosis. Recently, we described that ACKR3 is a prominent inhibitory receptor of platelet activation. Specific ACKR3 agonists but not conventional antiplatelet compounds such as COX-1 inhibitor (indometacin), ADP-receptor blocker (cangrelor), or PAR1 inhibitor (ML161) inhibit both hemin-dependent aggregation and thrombus formation. To further characterize the effect of hemin on platelet subpopulations, we established a multi-color flow cytometry assay. We found that hemin induces procoagulant (CD42bpos/PAC-1neg/AnnexinVpos), aggregatory (CD42bpos/PAC-1pos/AnnexinVneg), and inflammatory (CD42bpos/CXCR4pos/ACKR3pos/AnnexinVpos) platelet subpopulations. Treatment with ACKR3 agonists significantly decreased the formation of procoagulant and ACKR3pos platelets in response to hemin. We conclude that hemin is a strong activator for the formation of procoagulant platelets and thrombus formation which is dependent on the function of ACKR3. Activation of ACKR3 using specific agonists may offer a therapeutic strategy to regulate the vulnerability of atherosclerotic plaques in areas of IPH.
Collapse
Affiliation(s)
- Zoi Laspa
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Valerie Dicenta-Baunach
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - David Schaale
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Ravi Hochuli
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Alp Bayrak
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University Tübingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Thanigaimalai Pillaiyar
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Germany
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Germany
- iFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| |
Collapse
|
2
|
Starke A, Kollikowski AM, Vogt V, Stoll G, Nieswandt B, Pham M, Stegner D, Schuhmann MK. Soluble Glycoprotein VI Levels Assessed Locally within the Extra- and Intracerebral Circulation in Hyper-Acute Thromboembolic Stroke: A Pilot Study. Biomedicines 2024; 12:2191. [PMID: 39457504 PMCID: PMC11504204 DOI: 10.3390/biomedicines12102191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Severe acute ischemic stroke (AIS) is mainly caused by thromboembolism originating from symptomatic carotid artery (ICA) stenosis or in the heart due to atrial fibrillation. Glycoprotein VI (GPVI), a principal platelet receptor, facilitates platelet adherence and thrombus formation at sites of vascular injury such as symptomatic ICA stenosis. The shedding of GPVI from the platelet surface releases soluble GPVI (sGPVI) into the circulation. Here, we aimed to determine whether sGPVI can serve as a local biomarker to differentiate between local atherosclerotic and systemic cardiac thromboembolism in AIS. Methods: We conducted a cohort study involving 105 patients undergoing emergency endovascular thrombectomy (EVT) for anterior circulation stroke. First, sGPVI concentrations were measured in systemic arterial plasma samples collected at the ipsilateral ICA level, including groups with significantly (≥50%) stenotic and non-stenotic arteries. A second sample, taken from the intracerebral pial circulation, was used to assess GPVI shedding locally within the ischemic brain. Results: Our analysis revealed no significant increase in systemic sGPVI levels in patients with symptomatic ≥ 50% ICA stenosis (3.2 [95% CI 1.5-5.0] ng/mL; n = 33) compared with stroke patients without significant ICA stenosis (3.2 [95% CI 2.3-4.2] ng/mL; n = 72). Additionally, pial blood samples, reflecting intravascular molecular conditions during collateral flow, showed similar sGPVI levels when compared to the systemic ICA samples in both groups. Conclusions: Our findings indicate that GPVI is not locally cleaved and shed into the bloodstream in significant amounts during hyper-acute ischemic stroke, neither at the level of symptomatic ICA nor intracranially during collateral blood supply. Therefore, sGPVI does not appear to be suitable as a local stroke biomarker despite strong evidence of a major role for GPVI-signaling in stroke pathophysiology.
Collapse
Affiliation(s)
- Andreas Starke
- Rudolf Virchow Center for Integrative and Translational Imaging, Julius-Maximilians-Universität Würzburg (JMU), 97080 Würzburg, Germany; (A.S.); (B.N.); (D.S.)
| | - Alexander M. Kollikowski
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.M.K.); (M.P.)
| | - Vivian Vogt
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Guido Stoll
- Institute for Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Bernhard Nieswandt
- Rudolf Virchow Center for Integrative and Translational Imaging, Julius-Maximilians-Universität Würzburg (JMU), 97080 Würzburg, Germany; (A.S.); (B.N.); (D.S.)
- Institute for Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Mirko Pham
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.M.K.); (M.P.)
| | - David Stegner
- Rudolf Virchow Center for Integrative and Translational Imaging, Julius-Maximilians-Universität Würzburg (JMU), 97080 Würzburg, Germany; (A.S.); (B.N.); (D.S.)
- Institute for Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany;
| | | |
Collapse
|
3
|
Navarro S, Talucci I, Göb V, Hartmann S, Beck S, Orth V, Stoll G, Maric HM, Stegner D, Nieswandt B. The humanized platelet glycoprotein VI Fab inhibitor EMA601 protects from arterial thrombosis and ischaemic stroke in mice. Eur Heart J 2024:ehae482. [PMID: 39150906 DOI: 10.1093/eurheartj/ehae482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/12/2024] [Accepted: 07/12/2024] [Indexed: 08/18/2024] Open
Abstract
BACKGROUND AND AIMS Glycoprotein VI (GPVI) is a platelet collagen/fibrin(ogen) receptor and an emerging pharmacological target for the treatment of thrombotic and thrombo-inflammatory diseases, notably ischaemic stroke. A first anti-human GPVI (hGPVI) antibody Fab-fragment (ACT017/glenzocimab, KD: 4.1 nM) recently passed a clinical phase 1b/2a study in patients with acute ischaemic stroke and was found to be well tolerated, safe, and potentially beneficial. In this study, a novel humanized anti-GPVI antibody Fab-fragment (EMA601; KD: 0.195 nM) was developed that inhibits hGPVI function with very high potency in vitro and in vivo. METHODS Fab-fragments of the mouse anti-hGPVI IgG Emf6.1 were tested for functional GPVI inhibition in human platelets and in hGPVI expressing (hGP6tg/tg) mouse platelets. The in vivo effect of Emf6.1Fab was assessed in a tail bleeding assay, an arterial thrombosis model and the transient middle cerebral artery occlusion (tMCAO) model of ischaemic stroke. Using complementary-determining region grafting, a humanized version of Emf6.1Fab (EMA601) was generated. Emf6.1Fab/EMA601 interaction with hGPVI was mapped in array format and kinetics and quantified by bio-layer interferometry. RESULTS Emf6.1Fab (KD: 0.427 nM) blocked GPVI function in human and hGP6tg/tg mouse platelets in multiple assays in vitro at concentrations ≥5 µg/mL. Emf6.1Fab (4 mg/kg)-treated hGP6tg/tg mice showed potent hGPVI inhibition ex vivo and were profoundly protected from arterial thrombosis as well as from cerebral infarct growth after tMCAO, whereas tail-bleeding times remained unaffected. Emf6.1Fab binds to a so far undescribed membrane proximal epitope in GPVI. The humanized variant EMA601 displayed further increased affinity for hGPVI (KD: 0.195 nM) and fully inhibited the receptor at 0.5 µg/mL, corresponding to a >50-fold potency compared with ACT017. CONCLUSIONS EMA601 is a conceptually novel and promising anti-platelet agent to efficiently prevent or treat arterial thrombosis and thrombo-inflammatory pathologies in humans at risk.
Collapse
Affiliation(s)
- Stefano Navarro
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ivan Talucci
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Vanessa Göb
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Stefanie Hartmann
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| | - Sarah Beck
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | | | - Guido Stoll
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| | - Hans M Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - David Stegner
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- EMFRET Analytics GmbH, Eibelstadt, Germany
| |
Collapse
|
4
|
Scheldeman L, Sinnaeve P, Albers GW, Lemmens R, Van de Werf F. Acute myocardial infarction and ischaemic stroke: differences and similarities in reperfusion therapies-a review. Eur Heart J 2024; 45:2735-2747. [PMID: 38941344 DOI: 10.1093/eurheartj/ehae371] [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: 10/31/2023] [Revised: 04/16/2024] [Accepted: 05/28/2024] [Indexed: 06/30/2024] Open
Abstract
Acute ST-elevation myocardial infarction (STEMI) and acute ischaemic stroke (AIS) share a number of similarities. However, important differences in pathophysiology demand a disease-tailored approach. In both conditions, fast treatment plays a crucial role as ischaemia and eventually infarction develop rapidly. Furthermore, in both fields, the introduction of fibrinolytic treatments historically preceded the implementation of endovascular techniques. However, in contrast to STEMI, only a minority of AIS patients will eventually be considered eligible for reperfusion treatment. Non-invasive cerebral imaging always precedes cerebral angiography and thrombectomy, whereas coronary angiography is not routinely preceded by non-invasive cardiac imaging in patients with STEMI. In the late or unknown time window, the presence of specific patterns on brain imaging may help identify AIS patients who benefit most from reperfusion treatment. For STEMI, a uniform time window for reperfusion up to 12 h after symptom onset, based on old placebo-controlled trials, is still recommended in guidelines and generally applied. Bridging fibrinolysis preceding endovascular treatment still remains the mainstay of reperfusion treatment in AIS, while primary percutaneous coronary intervention is the strategy of choice in STEMI. Shortening ischaemic times by fine-tuning collaboration networks between ambulances, community hospitals, and tertiary care hospitals, optimizing bridging fibrinolysis, and reducing ischaemia-reperfusion injury are important topics for further research. The aim of this review is to provide insights into the common as well as diverging pathophysiology behind current reperfusion strategies and to explore new ways to enhance their clinical benefit.
Collapse
Affiliation(s)
- Lauranne Scheldeman
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology KU Leuven - University of Leuven, Leuven, Belgium
| | - Peter Sinnaeve
- Department of Cardiovascular Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, University of Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Gregory W Albers
- Department of Neurology, Stanford University Medical Center, Palo Alto, USA
| | - Robin Lemmens
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology KU Leuven - University of Leuven, Leuven, Belgium
| | - Frans Van de Werf
- Department of Cardiovascular Sciences, KU Leuven, University of Leuven, Herestraat 49, B-3000 Leuven, Belgium
| |
Collapse
|
5
|
Galanti K, Di Marino M, Mansour D, Testa S, Rossi D, Scollo C, Magnano R, Pezzi L, D'Alleva A, Forlani D, Vitulli P, Paloscia L, Ricci F, Renda G, Gallina S, Di Marco M. Current Antithrombotic Treatments for Cardiovascular Diseases: A Comprehensive Review. Rev Cardiovasc Med 2024; 25:281. [PMID: 39228474 PMCID: PMC11366999 DOI: 10.31083/j.rcm2508281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 09/05/2024] Open
Abstract
Antithrombotic therapies (ATT) play a pivotal role in the management of cardiovascular diseases, aiming to prevent ischemic events while maintaining a delicate balance with the patient's bleeding risk. Typically, ATT can be classified into antiplatelet and anticoagulant therapies. Their application spans a broad spectrum of cardiovascular conditions, ranging from ischemic heart disease to atrial fibrillation, encompassing venous thromboembolisms and innovative structural interventional cardiology procedures. The global burden of cardiovascular diseases is steadily increasing, often giving rise to overlapping clinical presentations. Accordingly, the adoption of combined pharmacological approaches becomes imperative, potentially disrupting the delicate equilibrium between ischemic and bleeding risk, thus leading to nuanced pharmacotherapeutic pathways. In this context, contemporary investigations strive to identify a convergence point that optimizes the duration of medical therapy while addressing the need for antithrombotic effects, especially in the context of ischemic heart disease. This review aims to comprehensively revisit the main antithrombotic strategies in cardiovascular diseases, with the intention of enhancing a systematic approach which is key for the effective clinical management of these patients. Also, the review will examine the most impactful studies that have established the groundwork for current scientific evidence, with acknowledgement of special populations. Finally, we will cast a gaze into the future of this dynamic and evolving research field, exploring forthcoming perspectives and advancements.
Collapse
Affiliation(s)
- Kristian Galanti
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Mario Di Marino
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Davide Mansour
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Sabrina Testa
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Davide Rossi
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Claudio Scollo
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Roberta Magnano
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| | - Laura Pezzi
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| | - Alberto D'Alleva
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| | - Daniele Forlani
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| | - Piergiusto Vitulli
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| | - Leonardo Paloscia
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| | - Fabrizio Ricci
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
- Department of Clinical Sciences, Lund University, 21428 Malmö, Sweden
| | - Giulia Renda
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
- University Cardiology Division, Heart Department, “SS Annunziata” Polyclinic University Hospital, 66100 Chieti, Italy
| | - Massimo Di Marco
- Cardiology and ICCU Department, Santo Spirito Hospital, 65124 Pescara, Italy
| |
Collapse
|
6
|
Corken A, Wahl EC, Sikes JD, Thakali KM. Western Diet Modifies Platelet Activation Profiles in Male Mice. Int J Mol Sci 2024; 25:8019. [PMID: 39125586 PMCID: PMC11311362 DOI: 10.3390/ijms25158019] [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: 06/07/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The correlation between obesity and cardiovascular disease has long been understood, yet scant investigations endeavored to determine the impact of an obesogenic diet on platelet activation or function. As platelets drive clot formation, the terminus of cardiovascular events, we aimed to elucidate the longitudinal effect of an obesogenic diet on platelet phenotype by assessing markers of platelet activation using flow cytometry. Male, weanling mice were fed either a Western diet (30% kcal sucrose, 40% kcal fat, 8.0% sodium) or Control diet (7% kcal sucrose, 10% kcal fat, 0.24% sodium). At 12, 16 and 20 weeks on diets, platelets were collected and stained to visualize glycoprotein Ibα (GPIbα), P-selectin and the conformationally active state of αIIbβ3 (a platelet specific integrin) after collagen stimulation. At all time points, a Western diet reduced GPIbα and αIIbβ3 expression in platelets broadly while P-selectin levels were unaffected. However, P-selectin was diminished by a Western diet in the GPIbα- subpopulation. Thus, a Western diet persistently primed platelets towards a blunted activation response as indicated by reduced active αIIbβ3 and P-selectin surface expression. This study provides a first look at the influence of diet on platelet activation and revealed that platelet activation is susceptible to dietary intervention.
Collapse
Affiliation(s)
- Adam Corken
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
- Arkansas Children’s Nutrition Center, Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Elizabeth C. Wahl
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
| | - James D. Sikes
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
| | - Keshari M. Thakali
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
- Arkansas Children’s Nutrition Center, Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| |
Collapse
|
7
|
Patrono C. Low-dose aspirin for the prevention of atherosclerotic cardiovascular disease. Eur Heart J 2024; 45:2362-2376. [PMID: 38839268 PMCID: PMC11242460 DOI: 10.1093/eurheartj/ehae324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/22/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024] Open
Abstract
During the past 30 years, several developments have occurred in the antiplatelet field, including the role of aspirin in primary prevention of atherosclerotic cardiovascular disease. There have been several attempts to develop antiplatelet drugs more effective and safer than aspirin and a shift in emphasis from efficacy to safety, advocating aspirin-free antiplatelet regimens after percutaneous coronary intervention. Evidence supporting a chemopreventive effect of low-dose aspirin against colorectal (and other digestive tract) cancer has also strengthened. The aim of this article is to revisit the role of aspirin in the prevention of atherothrombosis across the cardiovascular risk continuum, in view of developments in the antiplatelet field. The review will offer a clinical perspective on aspirin's mechanism of action, pharmacokinetics, and pharmacodynamics. This will be followed by a detailed discussion of its clinical efficacy and safety.
Collapse
Affiliation(s)
- Carlo Patrono
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo Francesco Vito 1, 00168 Rome, Italy
- Center of Excellence on Ageing, University of Chieti, Via Luigi Polacchi 11, 66100 Chieti, Italy
| |
Collapse
|
8
|
Velarde-Acosta K, Moscoso Ramirez JY, Rojas P, Susanibar L, Reusche LDQ, Cachicatari A, Baltodano-Arellano R. Shaggy aorta: ideal substrate for disaster. Updated review. ARCHIVOS PERUANOS DE CARDIOLOGIA Y CIRUGIA CARDIOVASCULAR 2024; 5:143-152. [PMID: 39411013 PMCID: PMC11473078 DOI: 10.47487/apcyccv.v5i3.410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024]
Abstract
Shaggy aorta (SA) is characterized by a critical and extensive atheromatous disease of the thoracic and abdominal aorta. This degenerative and dangerous pathology is the result of the confluence of multiple modifiable and non-modifiable risk factors. The clinical importance of this pathology relies on the various syndromes that can develop from its etiopathogenesis, which generates great morbidity and mortality in the affected patients. In this document, we present an updated and detailed review of this entity, developing aspects of its pathophysiology, diagnosis, including the importance of multimodal imaging, and its therapeutic approach. Finally, we present the clinical settings of patients with SA in different aortic scenarios (aortic dissection, ulcerated plaques, and thrombosed aneurysms) that denote the nature of this disease and its high mortality.
Collapse
Affiliation(s)
- Kevin Velarde-Acosta
- Clinical Cardiology Department; Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúClinical Cardiology DepartmentHospital Guillermo Almenara Irigoyen - EsSaludLimaPerú
| | - Josh Yefry Moscoso Ramirez
- Clinical Cardiology Department; Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúClinical Cardiology DepartmentHospital Guillermo Almenara Irigoyen - EsSaludLimaPerú
| | - Paol Rojas
- Clinical Cardiology Department; Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúClinical Cardiology DepartmentHospital Guillermo Almenara Irigoyen - EsSaludLimaPerú
- Interventional Cardiology Department; Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúInterventional Cardiology DepartmentHospital Guillermo Almenara Irigoyen - EsSaludLimaPerú
| | - Lucy Susanibar
- Clinical Cardiology Department; Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúClinical Cardiology DepartmentHospital Guillermo Almenara Irigoyen - EsSaludLimaPerú
| | - Lady Diana Quintana Reusche
- Clinical Cardiology Department; Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúClinical Cardiology DepartmentHospital Guillermo Almenara Irigoyen - EsSaludLimaPerú
| | - Angela Cachicatari
- Cardiac imaging area of Cardiology Department, Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúLimaPerú
| | - Roberto Baltodano-Arellano
- Cardiac imaging area of Cardiology Department, Hospital Guillermo Almenara Irigoyen - EsSalud, Lima, PerúLimaPerú
- School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, PerúLimaPerú
| |
Collapse
|
9
|
Wang S, He H, Mao Y, Zhang Y, Gu N. Advances in Atherosclerosis Theranostics Harnessing Iron Oxide-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308298. [PMID: 38368274 PMCID: PMC11077671 DOI: 10.1002/advs.202308298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/06/2024] [Indexed: 02/19/2024]
Abstract
Atherosclerosis, a multifaceted chronic inflammatory disease, has a profound impact on cardiovascular health. However, the critical limitations of atherosclerosis management include the delayed detection of advanced stages, the intricate assessment of plaque stability, and the absence of efficacious therapeutic strategies. Nanotheranostic based on nanotechnology offers a novel paradigm for addressing these challenges by amalgamating advanced imaging capabilities with targeted therapeutic interventions. Meanwhile, iron oxide nanoparticles have emerged as compelling candidates for theranostic applications in atherosclerosis due to their magnetic resonance imaging capability and biosafety. This review delineates the current state and prospects of iron oxide nanoparticle-based nanotheranostics in the realm of atherosclerosis, including pivotal aspects of atherosclerosis development, the pertinent targeting strategies involved in disease pathogenesis, and the diagnostic and therapeutic roles of iron oxide nanoparticles. Furthermore, this review provides a comprehensive overview of theranostic nanomedicine approaches employing iron oxide nanoparticles, encompassing chemical therapy, physical stimulation therapy, and biological therapy. Finally, this review proposes and discusses the challenges and prospects associated with translating these innovative strategies into clinically viable anti-atherosclerosis interventions. In conclusion, this review offers new insights into the future of atherosclerosis theranostic, showcasing the remarkable potential of iron oxide-based nanoparticles as versatile tools in the battle against atherosclerosis.
Collapse
Affiliation(s)
- Shi Wang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Hongliang He
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Yu Mao
- School of MedicineNanjing UniversityNanjing210093P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Ning Gu
- School of MedicineNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
10
|
Friebel J, Wegner M, Blöbaum L, Schencke PA, Jakobs K, Puccini M, Ghanbari E, Lammel S, Thevathasan T, Moos V, Witkowski M, Landmesser U, Rauch-Kröhnert U. Characterization of Biomarkers of Thrombo-Inflammation in Patients with First-Diagnosed Atrial Fibrillation. Int J Mol Sci 2024; 25:4109. [PMID: 38612918 PMCID: PMC11012942 DOI: 10.3390/ijms25074109] [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: 01/26/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Patients with first-diagnosed atrial fibrillation (FDAF) exhibit major adverse cardiovascular events (MACEs) during follow-up. Preclinical models have demonstrated that thrombo-inflammation mediates adverse cardiac remodeling and atherothrombotic events. We have hypothesized that thrombin activity (FIIa) links coagulation with inflammation and cardiac fibrosis/dysfunction. Surrogate markers of the thrombo-inflammatory response in plasma have not been characterized in FDAF. In this prospective longitudinal study, patients presenting with FDAF (n = 80), and 20 matched controls, were included. FIIa generation and activity in plasma were increased in the patients with early AF compared to the patients with chronic cardiovascular disease without AF (controls; p < 0.0001). This increase was accompanied by elevated biomarkers (ELISA) of platelet and endothelial activation in plasma. Pro-inflammatory peripheral immune cells (TNF-α+ or IL-6+) that expressed FIIa-activated protease-activated receptor 1 (PAR1) (flow cytometry) circulated more frequently in patients with FDAF compared to the controls (p < 0.0001). FIIa activity correlated with cardiac fibrosis (collagen turnover) and cardiac dysfunction (NT-pro ANP/NT-pro BNP) surrogate markers. FIIa activity in plasma was higher in patients with FDAF who experienced MACE. Signaling via FIIa might be a presumed link between the coagulation system (tissue factor-FXa/FIIa-PAR1 axis), inflammation, and pro-fibrotic pathways (thrombo-inflammation) in FDAF.
Collapse
Affiliation(s)
- Julian Friebel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Max Wegner
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Leon Blöbaum
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Philipp-Alexander Schencke
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Kai Jakobs
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Marianna Puccini
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Emily Ghanbari
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Stella Lammel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
| | - Tharusan Thevathasan
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Verena Moos
- Medical Department I, Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Marco Witkowski
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- Friede Springer Cardiovascular Prevention Center at Charité, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ursula Rauch-Kröhnert
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany; (J.F.); (P.-A.S.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| |
Collapse
|
11
|
Lemmens TP, Bröker V, Rijpkema M, Hughes CCW, Schurgers LJ, Cosemans JMEM. Fundamental considerations for designing endothelialized in vitro models of thrombosis. Thromb Res 2024; 236:179-190. [PMID: 38460307 DOI: 10.1016/j.thromres.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Endothelialized in vitro models for cardiovascular disease have contributed greatly to our current understanding of the complex molecular mechanisms underlying thrombosis. To further elucidate these mechanisms, it is important to consider which fundamental aspects to incorporate into an in vitro model. In this review, we will focus on the design of in vitro endothelialized models of thrombosis. Expanding our understanding of the relation and interplay between the different pathways involved will rely in part on complex models that incorporate endothelial cells, blood, the extracellular matrix, and flow. Importantly, the use of tissue-specific endothelial cells will help in understanding the heterogeneity in thrombotic responses between different vascular beds. The dynamic and complex responses of endothelial cells to different shear rates underlines the importance of incorporating appropriate shear in in vitro models. Alterations in vascular extracellular matrix composition, availability of bioactive molecules, and gradients in concentration and composition of these molecules can all regulate the function of both endothelial cells and perivascular cells. Factors modulating these elements in in vitro models should therefore be considered carefully depending on the research question at hand. As the complexity of in vitro models increases, so can the variability. A bottom-up approach to designing such models will remain an important tool for researchers studying thrombosis. As new techniques are continuously being developed and new pathways are brought to light, research question-dependent considerations will have to be made regarding what aspects of thrombosis to include in in vitro models.
Collapse
Affiliation(s)
- Titus P Lemmens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Vanessa Bröker
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Minke Rijpkema
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, and Department of Biomedical Engineering, University of California, Irvine, USA
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Tsai IT, Sun CK. Stem Cell Therapy against Ischemic Heart Disease. Int J Mol Sci 2024; 25:3778. [PMID: 38612587 PMCID: PMC11011361 DOI: 10.3390/ijms25073778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Ischemic heart disease, which is one of the top killers worldwide, encompasses a series of heart problems stemming from a compromised coronary blood supply to the myocardium. The severity of the disease ranges from an unstable manifestation of ischemic symptoms, such as unstable angina, to myocardial death, that is, the immediate life-threatening condition of myocardial infarction. Even though patients may survive myocardial infarction, the resulting ischemia-reperfusion injury triggers a cascade of inflammatory reactions and oxidative stress that poses a significant threat to myocardial function following successful revascularization. Moreover, despite evidence suggesting the presence of cardiac stem cells, the fact that cardiomyocytes are terminally differentiated and cannot significantly regenerate after injury accounts for the subsequent progression to ischemic cardiomyopathy and ischemic heart failure, despite the current advancements in cardiac medicine. In the last two decades, researchers have realized the possibility of utilizing stem cell plasticity for therapeutic purposes. Indeed, stem cells of different origin, such as bone-marrow- and adipose-derived mesenchymal stem cells, circulation-derived progenitor cells, and induced pluripotent stem cells, have all been shown to play therapeutic roles in ischemic heart disease. In addition, the discovery of stem-cell-associated paracrine effects has triggered intense investigations into the actions of exosomes. Notwithstanding the seemingly promising outcomes from both experimental and clinical studies regarding the therapeutic use of stem cells against ischemic heart disease, positive results from fraud or false data interpretation need to be taken into consideration. The current review is aimed at overviewing the therapeutic application of stem cells in different categories of ischemic heart disease, including relevant experimental and clinical outcomes, as well as the proposed mechanisms underpinning such observations.
Collapse
Affiliation(s)
- I-Ting Tsai
- Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung City 82445, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Cheuk-Kwan Sun
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
- Department of Emergency Medicine, E-Da Dachang Hospital, I-Shou University, Kaohsiung City 80794, Taiwan
| |
Collapse
|
13
|
Loutati R, Bruoha S, Taha L, Karmi M, Perel N, Maller T, Amsalem I, Hitter R, Levi N, Zacks N, Shrem M, Amro M, Shuvy M, Glikson M, Asher E. The Effect of War on STEMI Incidence: Insights from Intensive Cardiovascular Care Unit Admissions. J Clin Med 2024; 13:1356. [PMID: 38592151 PMCID: PMC10931653 DOI: 10.3390/jcm13051356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 04/10/2024] Open
Abstract
(1) Background: The impact of armed conflicts on public health is undeniable, with psychological stress emerging as a significant risk factor for cardiovascular disease (CVD). Nevertheless, contemporary data regarding the influence of war on CVD, and especially on acute coronary syndrome (ACS), are scarce. Hence, the aim of the current study was to assess the repercussions of war on the admission and prognosis of patients admitted to a tertiary care center intensive cardiovascular care unit (ICCU). (2) Methods: All patients admitted to the ICCU during the first three months of the Israel-Hamas war (2023) were included and compared with all patients admitted during the same period in 2022. The primary outcome was in-hospital mortality. (3) Results: A total of 556 patients (184 females [33.1%]) with a median age of 70 (IQR 59-80) were included. Of them, 295 (53%) were admitted to the ICCU during the first three months of the war. Fewer Arab patients and more patients with ST-segment elevation myocardial infraction (STEMI) were admitted during the war period (21.8% vs. 13.2%, p < 0.001, and 31.9% vs. 24.1%, p = 0.04, respectively), whereas non-STEMI (NSTEMI) patients were admitted more frequently in the pre-war year (19.3% vs. 25.7%, p = 0.09). In-hospital mortality was similar in both groups (4.4% vs. 3.4%, p = 0.71; HR 1.42; 95% CI 0.6-3.32, p = 0.4). (4) Conclusions: During the first three months of the war, fewer Arab patients and more STEMI patients were admitted to the ICCU. Nevertheless, in-hospital mortality was similar in both groups.
Collapse
Affiliation(s)
- Ranel Loutati
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Sharon Bruoha
- Department of Cardiology, Barzilai Medical Center, and The Ben-Gurion University of the Negev, Ashkelon 7830604, Israel;
| | - Louay Taha
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Mohammad Karmi
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Nimrod Perel
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Tomer Maller
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Itshak Amsalem
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Rafael Hitter
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Nir Levi
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Netanel Zacks
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Maayan Shrem
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Motaz Amro
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Mony Shuvy
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Michael Glikson
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| | - Elad Asher
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel; (R.L.); (L.T.); (M.K.); (N.P.); (T.M.); (I.A.); (R.H.); (N.L.); (N.Z.); (M.A.); (M.S.); (M.G.)
| |
Collapse
|
14
|
Drysdale A, Blanco-Lopez M, White SJ, Unsworth AJ, Jones S. Differential Proteoglycan Expression in Atherosclerosis Alters Platelet Adhesion and Activation. Int J Mol Sci 2024; 25:950. [PMID: 38256024 PMCID: PMC10815981 DOI: 10.3390/ijms25020950] [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: 12/07/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Proteoglycans are differentially expressed in different atherosclerotic plaque phenotypes, with biglycan and decorin characteristic of ruptured plaques and versican and hyaluronan more prominent in eroded plaques. Following plaque disruption, the exposure of extracellular matrix (ECM) proteins triggers platelet adhesion and thrombus formation. In this study, the impact of differential plaque composition on platelet function and thrombus formation was investigated. Platelet adhesion, activation and thrombus formation under different shear stress conditions were assessed in response to individual proteoglycans and composites representing different plaque phenotypes. The results demonstrated that all the proteoglycans tested mediated platelet adhesion but not platelet activation, and the extent of adhesion observed was significantly lower than that observed with type I and type III collagens. Thrombus formation upon the rupture and erosion ECM composites was significantly reduced (p < 0.05) compared to relevant collagen alone, indicating that proteoglycans negatively regulate platelet collagen responses. This was supported by results demonstrating that the addition of soluble biglycan or decorin to whole blood markedly reduced thrombus formation on type I collagen (p < 0.05). Interestingly, thrombus formation upon the erosion composite displayed aspirin sensitivity, whereas the rupture composite was intensive to aspirin, having implications for current antiplatelet therapy regimes. In conclusion, differential platelet responses and antiplatelet efficacy are observed on ECM composites phenotypic of plaque rupture and erosion. Proteoglycans inhibit thrombus formation and may offer a novel plaque-specific approach to limit arterial thrombosis.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Maria Blanco-Lopez
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Stephen J. White
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK;
| | - Amanda J. Unsworth
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| |
Collapse
|
15
|
Crea F. The key role of thrombosis: focus on acute coronary syndrome, venous thrombo-embolism, and atrial fibrillation. Eur Heart J 2024; 45:1-4. [PMID: 38160711 DOI: 10.1093/eurheartj/ehad835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Affiliation(s)
- Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Gemelli Isola Hospital, Rome, Italy
| |
Collapse
|