Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1426526, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39055717

RESUMEN

Introduction: Complement-mediated damage to the myocardium during acute myocardial infarction (AMI), particularly the late components of the terminal pathway (C5-convertase and C5b-9), have previously been characterized. Unfortunately, only few studies have reported a direct association between dysregulated complement activation and endothelial function. Hence, little attention has been paid to the role of the anaphylatoxin C5a. The endothelial glycocalyx (eGC) together with the cellular actin cortex provide a vasoprotective barrier against chronic vascular inflammation. Changes in their nanomechanical properties (stiffness and height) are recognized as hallmarks of endothelial dysfunction as they correlate with the bioavailability of vasoactive substances, such as nitric oxide (NO). Here, we determined how the C5a:C5aR1 axis affects the eGC and endothelial function in AMI. Methods: Samples of fifty-five patients with ST-elevation myocardial infarction (STEMI) vs. healthy controls were analyzed in this study. eGC components and C5a levels were determined via ELISA; NO levels were quantified chemiluminescence-based. Endothelial cells were stimulated with C5a or patient sera (with/without C5a-receptor1 antagonist "PMX53") and the nanomechanical properties of eGC quantified using the atomic force microscopy (AFM)-based nanoindentation technique. To measure actin cytoskeletal tension regulator activation (RhoA and Rac1) G-LISA assays were applied. Vascular inflammation was examined by quantifying monocyte-endothelium interaction via AFM-based single-cell-force spectroscopy. Results: Serum concentrations of eGC components and C5a were significantly increased during STEMI. Serum and solely C5a stimulation decreased eGC height and stiffness, indicating shedding of the eGC. C5a enhanced RhoA activation, resulting in increased cortical stiffness with subsequent reduction in NO concentrations. Monocyte adhesion to the endothelium was enhanced after both C5a and stimulation with STEMI serum. eGC degradation- and RhoA-induced cortical stiffening with subsequent endothelial dysfunction were attenuated after administering PMX53. Conclusion: This study demonstrates that dysregulated C5a activation during AMI results in eGC damage with subsequent endothelial dysfunction and reduced NO bioavailability, indicating progressively developing vascular inflammation. This could be prevented by antagonizing C5aR1, highlighting the role of the C5a:C5a-Receptor1 axis in vascular inflammation development and endothelial dysfunction in AMI, offering new therapeutic approaches for future investigations.


Asunto(s)
Activación de Complemento , Complemento C5a , Glicocálix , Infarto del Miocardio , Receptor de Anafilatoxina C5a , Humanos , Glicocálix/metabolismo , Glicocálix/patología , Complemento C5a/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Receptor de Anafilatoxina C5a/metabolismo , Masculino , Persona de Mediana Edad , Femenino , Anciano , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Transducción de Señal , Óxido Nítrico/metabolismo
3.
Blood Adv ; 7(21): 6411-6427, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37257194

RESUMEN

In ischemic tissue, platelets can modulate angiogenesis. The specific factors influencing this function, however, are poorly understood. Here, we characterized the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) expressed on platelets as a potent regulator of ischemia-driven revascularization. We assessed the relevance of the anaphylatoxin receptor C5aR1 on platelets in patients with coronary artery disease as well as those with peripheral artery disease and used genetic mouse models to characterize its significance for ischemia and growth factor-driven revascularization. The presence of C5aR1-expressing platelets was increased in the hindlimb ischemia model. Ischemia-driven angiogenesis was significantly improved in C5aR1-/- mice but not in C5-/- mice, suggesting a specific role of C5aR1. Experiments using the supernatant of C5a-stimulated platelets suggested a paracrine mechanism of angiogenesis inhibition by platelets by means of antiangiogenic CXC chemokine ligand 4 (CXCL4, PF4). Lineage-specific C5aR1 deletion verified that the secretion of CXCL4 depends on C5aR1 ligation on platelets. Using C5aR1-/-CXCL4-/- mice, we observed no additional effect in the revascularization response, underscoring a strong dependence of CXCL4 secretion on the C5a-C5aR1-axis. We identified a novel mechanism for inhibition of neovascularization via platelet C5aR1, which was mediated by the release of antiangiogenic CXCL4.


Asunto(s)
Anafilatoxinas , Péptidos y Proteínas de Señalización Intercelular , Humanos , Ratones , Animales , Isquemia/etiología , Receptor de Anafilatoxina C5a
4.
STAR Protoc ; 3(3): 101664, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36097382

RESUMEN

Different types of immune cells are involved in atherogenesis and may act atheroprotective or atheroprogressive. Here, we describe an in vitro approach to analyze CD11c+ cells and CD11c+-derived ApoE in atherosclerosis. The major steps include harvesting mouse bone marrow, plating cells in culture dishes, treating them with differentiation factors, and collecting cells after removal of undesirable populations. This protocol can be adapted for CD11c+ cells in different contexts, thus, serving as models for different diseases and to analyze cell-specific molecules. For complete details on the use and execution of this protocol, please refer to Sauter et al. (2021).


Asunto(s)
Médula Ósea , Células Dendríticas , Animales , Apolipoproteínas E , Células de la Médula Ósea , Antígeno CD11c , Ratones
5.
J Immunol ; 208(7): 1729-1741, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35277420

RESUMEN

Recent evidence suggests interaction of platelets with dendritic cells (DCs), while the molecular mechanisms mediating this heterotypic cell cross-talk are largely unknown. We evaluated the role of integrin Mac-1 (αMß2, CD11b/CD18) on DCs as a counterreceptor for platelet glycoprotein (GP) Ibα. In a dynamic coincubation model, we observed interaction of human platelets with monocyte-derived DCs, but also that platelet activation induced a sharp increase in heterotypic cell binding. Inhibition of CD11b or GPIbα led to significant reduction of DC adhesion to platelets in vitro independent of GPIIbIIIa, which we confirmed using platelets from Glanzmann thrombasthenia patients and transgenic mouse lines on C57BL/6 background (GPIbα-/-, IL4R-GPIbα-tg, and muMac1 mice). In vivo, inhibition or genetic deletion of CD11b and GPIbα induced a significant reduction of platelet-mediated DC adhesion to the injured arterial wall. Interestingly, only intravascular antiCD11b inhibited DC recruitment, suggesting a dynamic DC-platelet interaction. Indeed, we could show that activated platelets induced CD11b upregulation on Mg2+-preactivated DCs, which was related to protein kinase B (Akt) and dependent on P-selectin and P-selectin glycoprotein ligand 1. Importantly, specific pharmacological targeting of the GPIbα-Mac-1 interaction site blocked DC-platelet interaction in vitro and in vivo. These results demonstrate that cross-talk of platelets with DCs is mediated by GPIbα and Mac-1, which is upregulated on DCs by activated platelets in a P-selectin glycoprotein ligand 1-dependent manner.


Asunto(s)
Plaquetas , Antígenos CD18 , Animales , Plaquetas/fisiología , Antígenos CD18/metabolismo , Adhesión Celular , Comunicación Celular , Células Dendríticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo
6.
iScience ; 25(1): 103677, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35036868

RESUMEN

Atherosclerosis is studied in models with dysfunctional lipid homeostasis-predominantly the ApoE-/- mouse. The role of antigen-presenting cells (APCs) for lipid homeostasis is not clear. Using a LacZ reporter mouse, we showed that CD11c+ cells were enriched in aortae of ApoE-/- mice. Systemic long-term depletion of CD11c+ cells in ApoE-/- mice resulted in significantly increased plaque formation associated with reduced serum ApoE levels. In CD11ccre+ApoEfl/fl and Albumincre+ApoEfl/fl mice, we could show that ≈70% of ApoE is liver-derived and ≈25% originates from CD11c+ cells associated with significantly increased atherosclerotic plaque burden in both strains. Exposure to acLDL promoted cholesterol efflux from CD11c+ cells and cell-specific deletion of ApoE resulted in increased inflammation reflected by increased IL-1ß serum levels. Our results determined for the first time the level of ApoE originating from CD11c+ cells and demonstrated that CD11c+ cells ameliorate atherosclerosis by the secretion of ApoE.

7.
Life (Basel) ; 11(7)2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34357044

RESUMEN

Cardiovascular diseases (CVD) constitute the main cause of death worldwide. Both inflammation and oxidative stress have been reported to be involved in the progress of CVD. It is well known that generation of oxidative stress during the course of CVD is involved in tissue damage and inflammation, causing deleterious effects such as hypertension, dysfunctional metabolism, endothelial dysfunction, stroke, and myocardial infarction. Remarkably, natural antioxidant strategies have been increasingly discovered and are subject to current scientific investigations. Here, we addressed the activation of immune cells in the context of ROS production, as well as how their interaction with other cellular players and further (immune) mediators contribute to metabolic and cardiovascular disorders. We also highlight how a dysregulated complement system contributes to immune imbalance and tissue damage in the context of increases oxidative stress. Additionally, modulation of hypothalamic oxidative stress is discussed, which may offer novel treatment strategies for type-2 diabetes and obesity. Together, we provide new perspectives on therapy strategies for CVD caused by oxidative stress, with a focus on oxidative stress.

8.
Nat Commun ; 12(1): 3352, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099640

RESUMEN

Platelets contribute to the regulation of tissue neovascularization, although the specific factors underlying this function are unknown. Here, we identified the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) on platelets as a negative regulatory mechanism of vessel formation. We showed that platelets expressing C5aR1 exert an inhibitory effect on endothelial cell functions such as migration and 2D and 3D tube formation. Growth factor- and hypoxia-driven vascularization was markedly increased in C5ar1-/- mice. Platelet-specific deletion of C5aR1 resulted in a proangiogenic phenotype with increased collateralization, capillarization and improved pericyte coverage. Mechanistically, we found that C5a induced preferential release of CXC chemokine ligand 4 (CXCL4, PF4) from platelets as an important antiangiogenic paracrine effector molecule. Interfering with the C5aR1-CXCL4 axis reversed the antiangiogenic effect of platelets both in vitro and in vivo.In conclusion, we identified a mechanism for the control of tissue neovascularization through C5a/C5aR1 axis activation in platelets and subsequent induction of the antiangiogenic factor CXCL4.


Asunto(s)
Plaquetas/metabolismo , Factor Plaquetario 4/metabolismo , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Inductores de la Angiogénesis , Animales , Activación de Complemento , Complemento C5a , Inflamación , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptores CXCR3/genética , Transducción de Señal
9.
Br J Pharmacol ; 178(14): 2892-2904, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33817781

RESUMEN

A mutual relationship exists between immune activation and mechanisms of thrombus formation. In particular, elements of the innate immune response such as the complement system can modulate platelet activation and subsequently thrombus formation. Several components of the complement system including C3 or the membrane attack complex have been reported to be associated with platelets and become functionally active in the micromilieu of platelet activation. The exact mechanisms how this interplay is regulated and its consequences for tissue inflammation, damage or recovery remain to be defined. This review addresses the current state of knowledge on this topic and puts it into context with diseases featuring both thrombosis and complement activation. LINKED ARTICLES: This article is part of a themed issue on Canonical and non-canonical functions of the complement system in health and disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.14/issuetoc.


Asunto(s)
Trombosis , Plaquetas , Proteínas del Sistema Complemento , Humanos , Inflamación , Activación Plaquetaria
10.
Front Immunol ; 11: 548631, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123127

RESUMEN

Beyond platelets function in hemostasis, there is emerging evidence to suggest that platelets contribute crucially to inflammation and immune responses. Therefore, considering the detrimental role of inflammatory conditions in severe neurological disorders such as multiple sclerosis or stroke, this review outlines platelets involvement in neuroinflammation. For this, distinct mechanisms of platelet-mediated thrombosis and inflammation are portrayed, focusing on the interaction of platelet receptors with other immune cells as well as brain endothelial cells. Furthermore, we draw attention to the intimate interplay between platelets and the complement system as well as between platelets and plasmatic coagulation factors in the course of neuroinflammation. Following the thorough exposition of preclinical approaches which aim at ameliorating disease severity after inducing experimental autoimmune encephalomyelitis (a counterpart of multiple sclerosis in mice) or brain ischemia-reperfusion injury, the clinical relevance of platelet-mediated neuroinflammation is addressed. Thus, current as well as future propitious translational and clinical strategies for the treatment of neuro-inflammatory diseases by affecting platelet function are illustrated, emphasizing that targeting platelet-mediated neuroinflammation could become an efficient adjunct therapy to mitigate disease severity of multiple sclerosis or stroke associated brain injury.


Asunto(s)
Plaquetas/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/metabolismo , Trombosis/etiología , Trombosis/metabolismo , Inmunidad Adaptativa , Animales , Autoinmunidad , Coagulación Sanguínea , Plaquetas/inmunología , Susceptibilidad a Enfermedades , Humanos , Inmunidad Innata , Inflamación/diagnóstico , Enfermedades del Sistema Nervioso/diagnóstico , Activación Plaquetaria/genética , Activación Plaquetaria/inmunología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombosis/diagnóstico
11.
Atherosclerosis ; 307: 97-108, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32653088

RESUMEN

Cardiovascular disease remains the main cause of death worldwide. For this reason, strategies for the primary prevention of atherosclerosis and atherosclerosis-related pathologies like stroke or myocardial infarction are needed. Platelets are key players of atherosclerosis-related vascular thrombotic pathologies and their role as targets in secondary prevention of atherosclerosis-related complications is uncontested. However, platelets also play an important role in the initiation and progression of atherosclerosis. Currently, though, there is no generally valid recommendation for the use of antiplatelet therapy in primary prevention of cardiovascular disease. Recent clinical studies have shown that the benefit from antiplatelet therapy in primary prevention is counteracted by the entailed bleeding risk. This review addresses the important role platelets play in initiating and sustaining vascular inflammation, which drives atherosclerosis. Specifically, platelet-lipid interactions as well as platelet-endothelium interactions in the context of atherosclerosis are illustrated. We also depict how platelets help recruit immune cells like monocytes, neutrophils or dendritic cells to the subendothelial space. Finally, we portray the role of complement and platelets in atherosclerosis. Platelets appear to act as mediators of tissue homeostasis and may also modulate the microenvironment of the atherosclerotic plaque. Overall, this review addresses the role of platelets in atherosclerosis with particular focus on potential targets for pharmacological interventions into platelet functions distinct from aggregation. By eliminating the bleeding risk of antiplatelet therapy, platelets are likely to regain a role in primary prevention of cardiovascular disease.


Asunto(s)
Aterosclerosis , Trombosis , Aterosclerosis/prevención & control , Plaquetas , Hemostasis , Humanos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Pruebas de Función Plaquetaria
12.
Int J Cardiol Heart Vasc ; 24: 100413, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31508480

RESUMEN

INTRODUCTION: In this study, we evaluated right ventricular (RV) function before and after percutaneous mitral valve repair (PMVR) using conventional echocardiographic parameters and novel 3DE data sets acquired prior to and directly after the procedure. PATIENTS AND METHODS: Observational study on 45 patients undergoing PMVR at an university hospital. RESULTS: In the overall collective, the 3D RV-EF before and after PMVR showed no significant change (p = 0.16). While there was a significant increase of the fractional area change (FAC, from 23 [19-29] % to 28 [24-33] %, p = 0.001), no significant change of the tricuspid annular plane systolic excursion (TAPSE, from 17 ±â€¯6 mm to 18 ±â€¯5 mm (standard deviation), p = 0.33) was observed. Regarding patients with a reduced RV-EF (< 35%), a significant RV-EF improvement was observed (from 27 [23-34] % to 32.5 [30-39] % (p = 0.001). 71.4% of patients had an improved clinical outcome (improvement in 6-minute walk test and/or improvement in NYHA class of more than one grade), whereas clinical outcome did not improve in 28.6% of patients. Using univariate logistic regression analysis, the post-PMVR RV-EF (OR 1.15: 95% CI 1.02-1.29; p = 0.02) and the change in RV-EF (OR 1.13: 95% CI 1.02-1.25; p = 0.02) were significant predictors for improved clinical outcome at 6 months follow up. CONCLUSION: Thus, RV function may be an important non-invasive parameter to add to the predictive parameters indicating a potential clinical benefit from treatment of severe mitral regurgitation using PMVR.

13.
Front Immunol ; 10: 1731, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31402914

RESUMEN

Besides mediating hemostatic functions, platelets are increasingly recognized as important players of inflammation. Data from experiments in mice and men revealed various intersection points between thrombosis, hemostasis, and inflammation, which are addressed and discussed in this review in detail. One such example is the intrinsic coagulation cascade that is initiated after platelet activation thereby further propagating and re-enforcing wound healing or thrombus formation but also contributing to the pathophysiology of severe diseases. FXII of the intrinsic pathway connects platelet activation with the coagulation cascade during immune reactions. It can activate the contact system thereby either creating an inflammatory state or accelerating inflammation. Recent insights into platelet biology could show that platelets are equipped with complement receptors. Platelets are important for tissue remodeling after injury has been inflicted to the endothelial barrier and to the subendothelial tissue. Thus, platelets are increasingly recognized as more than just cells relevant for bleeding arrest. Future insights into platelet biology are to be expected. This research will potentially offer novel opportunities for therapeutic intervention in diseases featuring platelet abundance.


Asunto(s)
Plaquetas/inmunología , Plaquetas/metabolismo , Susceptibilidad a Enfermedades , Inmunidad , Animales , Médula Ósea , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Humanos , Inmunidad Innata , Inflamación/etiología , Inflamación/metabolismo , Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica , Transducción de Señal , Trombopoyesis , Trombosis/etiología , Trombosis/metabolismo
14.
J Am Heart Assoc ; 8(13): e011366, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31248323

RESUMEN

Background This study analyzed the effects on long-term outcome of residual mitral regurgitation ( MR ) and mean mitral valve pressure gradient ( MVPG ) after percutaneous edge-to-edge mitral valve repair using the MitraClip system. Methods and Results Two hundred fifty-five patients who underwent percutaneous edge-to-edge mitral valve repair were analyzed. Kaplan-Meier and Cox regression analyses were performed to evaluate the impact of residual MR and MVPG on clinical outcome. A combined clinical end point (all-cause mortality, MV surgery, redo procedure, implantation of a left ventricular assist device) was used. After percutaneous edge-to-edge mitral valve repair, mean MVPG increased from 1.6±1.0 to 3.1±1.5 mm Hg ( P<0.001). Reduction of MR severity to ≤2+ postintervention was achieved in 98.4% of all patients. In the overall patient cohort, residual MR was predictive of the combined end point while elevated MVPG >4.4 mm Hg was not according to Kaplan-Meier and Cox regression analyses. We then analyzed the cohort with degenerative and that with functional MR separately to account for these different entities. In the cohort with degenerative MR , elevated MVPG was associated with increased occurrence of the primary end point, whereas this was not observed in the cohort with functional MR . Conclusions MVPG >4.4 mm Hg after MitraClip implantation was predictive of clinical outcome in the patient cohort with degenerative MR . In the patient cohort with functional MR , MVPG >4.4 mm Hg was not associated with increased clinical events.


Asunto(s)
Cateterismo Cardíaco , Anuloplastia de la Válvula Mitral , Insuficiencia de la Válvula Mitral/cirugía , Presión , Anciano , Anciano de 80 o más Años , Ecocardiografía , Femenino , Corazón Auxiliar , Humanos , Estimación de Kaplan-Meier , Masculino , Insuficiencia de la Válvula Mitral/diagnóstico por imagen , Insuficiencia de la Válvula Mitral/etiología , Insuficiencia de la Válvula Mitral/fisiopatología , Mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Reoperación , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
16.
Circulation ; 138(16): 1720-1735, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29802205

RESUMEN

BACKGROUND: Platelets have distinct roles in the vascular system in that they are the major mediator of thrombosis, critical for restoration of tissue integrity, and players in vascular inflammatory conditions. In close spatiotemporal proximity, the complement system acts as the first line of defense against invading microorganisms and is a key mediator of inflammation. Whereas the fluid phase cross-talk between the complement and coagulation systems is well appreciated, the understanding of the pathophysiological implications of such interactions is still scant. METHODS: We analyzed coexpression of the anaphylatoxin receptor C3aR with activated glycoprotein IIb/IIIa on platelets of 501 patients with coronary artery disease using flow cytometry; detected C3aR expression in human or murine specimen by polymerase chain reaction, immunofluorescence, Western blotting, or flow cytometry; and examined the importance of platelet C3aR by various in vitro platelet function tests, in vivo bleeding time, and intravital microscopy. The pathophysiological relevance of C3aR was scrutinized with the use of disease models of myocardial infarction and stroke. To approach underlying molecular mechanisms, we identified the platelet small GTPase Rap1b using nanoscale liquid chromatography coupled to tandem mass spectrometry. RESULTS: We found a strong positive correlation of platelet complement C3aR expression with activated glycoprotein IIb/IIIa in patients with coronary artery disease and coexpression of C3aR with glycoprotein IIb/IIIa in thrombi obtained from patients with myocardial infarction. Our results demonstrate that the C3a/C3aR axis on platelets regulates distinct steps of thrombus formation such as platelet adhesion, spreading, and Ca2+ influx. Using C3aR-/- mice or C3-/- mice with reinjection of C3a, we uncovered that the complement activation fragment C3a regulates bleeding time after tail injury and thrombosis. Notably, C3aR-/- mice were less prone to experimental stroke and myocardial infarction. Furthermore, reconstitution of C3aR-/- mice with C3aR+/+ platelets and platelet depletion experiments demonstrated that the observed effects on thrombosis, myocardial infarction, and stroke were specifically caused by platelet C3aR. Mechanistically, C3aR-mediated signaling regulates the activation of Rap1b and thereby bleeding arrest after injury and in vivo thrombus formation. CONCLUSIONS: Overall, our findings uncover a novel function of the anaphylatoxin C3a for platelet function and thrombus formation, highlighting a detrimental role of imbalanced complement activation in cardiovascular diseases.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Inmunidad Innata , Infarto del Miocardio/sangre , Receptores de Complemento/sangre , Accidente Cerebrovascular/sangre , Trombosis/sangre , Animales , Plaquetas/inmunología , Señalización del Calcio , Activación de Complemento , Complemento C3/genética , Complemento C3/inmunología , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/inmunología , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Accidente Cerebrovascular/inmunología , Trombosis/inmunología
17.
Semin Immunol ; 37: 43-52, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29426568

RESUMEN

The complement system is a versatile part of our immune system. Various intersection points of complement with other cells and molecules of the immune response are well described. Platelets are classically conceived as cells of hemostasis. In recent years, however, several functions of platelets "beyond thrombosis" were discovered. This review depicts the crosstalk of platelets with components of the immune system in the context of thrombo-inflammation. In particular, the various ways, in which platelets interact with the complement system, are illustrated. Platelets cannot only aggravate vascular inflammation and cardiovascular diseases, but they also contribute to organ remodeling and tissue homeostasis. Here, we portray the role of complement factors associated with platelet activation in tissue remodeling. Importantly, the clinical relevance of this platelet-complement crosstalk is addressed. A focus lies on thrombo-inflammatory disorders, other diseases with thrombo-embolic mechanisms or complications, but also autoimmune diseases. Finally, we draw attention to the growing body of evidence on the role of complement-platelet crosstalk in cardiovascular diseases. For future clinical, translational and basic science approaches, this crosstalk may prove a fruitful area of research in order to procure novel therapeutic and diagnostic targets in cardiovascular medicine and previously less addressed diseases featuring a platelet-complement axis.


Asunto(s)
Plaquetas/inmunología , Proteínas del Sistema Complemento/metabolismo , Inmunidad Innata , Inflamación/inmunología , Enfermedades Vasculares/inmunología , Animales , Humanos , Activación Plaquetaria , Trombosis , Remodelación Vascular
19.
Platelets ; 27(6): 593-597, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27025272

RESUMEN

Both oxidized lipids as well as the complement system contribute to atherothrombosis. The expression of complement receptors correlates with the expression of platelet activation markers, and platelet bound oxidized low-density lipoprotein (oxLDL) modulates platelet function. In the present study, we investigated the relationship of markers of complement activation, the anaphylatoxins C5a and C3a, and oxidized low-density lipoprotein. Two hundred and seven patients with coronary artery disease (CAD) were analyzed in this study. Using enzyme-linked immunosorbent assays, plasma levels of oxLDL, C3a, and C5a were measured. Moreover, we assessed platelet bound oxLDL by flow cytometry. The overall level of C5a in the troponin negative group (stable angina (SA) and unstable angina (UA)) compared to the troponin positive group (non-ST-elevation myocardial infarction (NSTEMI) and ST-elevation myocardial infarction (STEMI)) did not differ significantly (62.7 ± 32.4 ng/ml versus 65.8 ± 40.3 ng/ml). While C5a and C3a showed a significant correlation with each other (r = 0.25, p < 0.001), there was no statistically significant relationship between C3a and platelet bound oxLDL (r = 0.06, p = 0.37). Furthermore, plasma oxLDL did not correlate with either C3a or C5a. However, we observed a moderate, yet significant negative correlation between plasma C5a and platelet bound oxLDL (r = -0.15, p = 0.04). Partial correlation analysis correcting for the presence of acute coronary syndrome (ACS), troponin status or the subgroups SA, UA, NSTEMI, or STEMI did not alter this correlation substantially. Interestingly, flow cytometric analysis of human platelets showed increased expression of C5aR and P-selectin after in vitro stimulation with oxLDL. In conclusion, the complement anaphylatoxin C5a shows an inverse correlation with platelet bound oxLDL. The relationship of oxidized lipids to particular complement components may add to the platelet-lipid interplay in atherogenesis and trigger future clinical and mechanistic studies.


Asunto(s)
Plaquetas/metabolismo , Complemento C5a/inmunología , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/inmunología , Lipoproteínas LDL/metabolismo , Humanos , Selectina-P/metabolismo , Activación Plaquetaria , Agregación Plaquetaria , Unión Proteica
20.
Front Immunol ; 6: 98, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25798138

RESUMEN

Platelets contribute to processes beyond thrombus formation and may play a so far underestimated role as an immune cell in various circumstances. This review outlines immune functions of platelets in host defense, but also how they may contribute to mechanisms of infectious diseases. A particular emphasis is placed on the interaction of platelets with other immune cells. Furthermore, this article outlines the features of atherosclerosis as an inflammatory vascular disease highlighting the role of platelet crosstalk with cellular and soluble factors involved in atheroprogression. Understanding, how platelets influence these processes of vascular remodeling will shed light on their role for tissue homeostasis beyond intravascular thrombosis. Finally, translational implications of platelet-mediated inflammation in atherosclerosis are discussed.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...