Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Exp Dermatol ; 32(12): 2131-2137, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37846872

RESUMEN

Laser-assisted drug delivery (LADD) is a treatment method to enhance the penetration of pharmaceuticals through the skin. The aim of the present study is to track hyaluronic acid (HA) and analyse its effect on human skin in vivo after ablative fractional laser (AFL) treatment. Healthy male and female subjects were recruited. Four areas were marked on their forearms of each volunteer, and each area was assigned to one of the following treatment options: AFL + HA, AFL only, HA only or untreated control. A carbon dioxide laser was used for the AFL treatment. Follow-up measurements were scheduled 30 min and 30 days after treatment using multiphoton tomography equipped with fluorescence lifetime imaging (MPT-FLIM). A total of 11 subjects completed the study. By detecting fluorescence lifetimes, the HA and the anaesthetic ointment were clearly distinguishable from surrounding tissue. After AFL treatment, HA could be visualized in all epidermal and upper dermal layers. In contrast, HA in intact skin was only detected in the superficial layers at distinctly lower levels. The applied HA gel seemed to have beneficial properties for the wound healing process after laser treatment. LADD has proven to be a fast and effective method to increase HA uptake into the skin, allowing for improved hydration and skin rejuvenation over time. Furthermore, LADD could be a beneficial treatment option in laser resurfacing. MPT-FLIM proved to be an appropriate diagnostic tool for drug delivery tracking and monitoring of treatment response for individualized therapy adjustment.


Asunto(s)
Ácido Hialurónico , Láseres de Gas , Humanos , Masculino , Femenino , Ácido Hialurónico/farmacología , Fluorescencia , Piel/diagnóstico por imagen , Cicatrización de Heridas , Láseres de Gas/uso terapéutico
2.
BMJ Open ; 12(7): e060811, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35798519

RESUMEN

INTRODUCTION: Chronic pruritus (CP) is a symptom of dermatologic, neurologic, systemic and psychosomatic diseases. CP has a prevalence of ~20% in the general population and is therefore a significant burden on society, but the transition from acute pruritus to CP is not well understood. It probably involves interactions between biological and psychosocial factors and pruritus-specific risk factors as well as mechanisms shared with other persistent somatic symptoms addressed in other projects of the SOMACROSS Research Unit (RU). Here we aim to identify psychosocial and biological factors and their interactions which might be associated with the persistence of CP with and without immunologic/inflammatory origin, that is, atopic dermatitis and pruritus on non-inflamed skin. We expect that psychosocial factors relevant to the persistence of symptoms such as fatigue and pain may also show associations to CP. METHODS AND ANALYSIS: In this prospective, exploratory observational study situated in Germany, three cohorts of 40 patients each with acute exacerbation of atopic dermatitis and chronic atopic dermatitis and 40 CP patients with unaffected skin will be recruited for a comprehensive translational investigation including pruritus-specific and the shared psychosocial assessments of the RU SOMACROSS. Pruritus-specific measures will include questionnaires, quantitative sensory testing, cutaneous nerve fibre morphology, skin barrier morphology, epidermal metabolism and pruritogen blood levels. Within 1 year, patients and 80 age-matched and sex-matched healthy controls will be examined at three time points, allowing cross-sectional comparison and a longitudinal investigation of predictive outcome factors in patients under treatment according to existing guidelines. ETHICS AND DISSEMINATION: The study has been approved by the ethics committees of Hamburg (2020-10200-BO-ff) and Münster (2020-676 f-S), Germany. All participants are required to provide written informed consent. Findings will be disseminated through peer-reviewed publications, scientific conferences and involvement of relevant stakeholders, patients and the lay public. TRIAL REGISTRATION NUMBER: DRKS00026646.


Asunto(s)
Dermatitis Atópica , Estudios Transversales , Dermatitis Atópica/complicaciones , Alemania/epidemiología , Humanos , Estudios Observacionales como Asunto , Estudios Prospectivos , Prurito/epidemiología
3.
Exp Dermatol ; 31(11): 1712-1719, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35837813

RESUMEN

With increased popularity of decorative tattoos, awareness of tattoo-based dermatological complications has been raised. Health issues include a broad spectrum dominated by allergies and infections. To examine the etiopathology and prognose the outcome of an appropriate therapy, a non-invasive intravital diagnostic approach is indicated. The present pilot study introduces multiphoton tomography equipped with fluorescence lifetime imaging as a diagnostic technique to examine the morphological and metabolic status of tattooed human skin at patient's bedside. The distributing course of tattoo particles can be visualised over time. By providing optical biopsies, inflammation-based alterations in freshly tattooed skin and tattoo complications can be analysed. The study concludes that multiphoton tomography combined with fluorescence lifetime imaging is a suitable technique for in vivo visualisation of tattoo pigments as well as for the assessment of quantitative and qualitative skin changes after injection of tattoo ink into human skin.


Asunto(s)
Tatuaje , Humanos , Proyectos Piloto , Colorantes , Color , Tinta , Tomografía
4.
Thromb Haemost ; 122(2): 226-239, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33385180

RESUMEN

The multimeric plasma glycoprotein (GP) von Willebrand factor (VWF) is best known for recruiting platelets to sites of injury during primary hemostasis. Generally, mutations in the VWF gene lead to loss of hemostatic activity and thus the bleeding disorder von Willebrand disease. By employing cone and platelet aggregometry and microfluidic assays, we uncovered a platelet GPIIb/IIIa-dependent prothrombotic gain of function (GOF) for variant p.Pro2555Arg, located in the C4 domain, leading to an increase in platelet aggregate size. We performed complementary biophysical and structural investigations using circular dichroism spectra, small-angle X-ray scattering, nuclear magnetic resonance spectroscopy, molecular dynamics simulations on the single C4 domain, and dimeric wild-type and p.Pro2555Arg constructs. C4-p.Pro2555Arg retained the overall structural conformation with minor populations of alternative conformations exhibiting increased hinge flexibility and slow conformational exchange. The dimeric protein becomes disordered and more flexible. Our data suggest that the GOF does not affect the binding affinity of the C4 domain for GPIIb/IIIa. Instead, the increased VWF dimer flexibility enhances temporal accessibility of platelet-binding sites. Using an interdisciplinary approach, we revealed that p.Pro2555Arg is the first VWF variant, which increases platelet aggregate size and shows a shear-dependent function of the VWF stem region, which can become hyperactive through mutations. Prothrombotic GOF variants of VWF are a novel concept of a VWF-associated pathomechanism of thromboembolic events, which is of general interest to vascular health but not yet considered in diagnostics. Thus, awareness should be raised for the risk they pose. Furthermore, our data implicate the C4 domain as a novel antithrombotic drug target.


Asunto(s)
Mutación con Ganancia de Función , Variación Genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Factor de von Willebrand/genética , Mutación con Ganancia de Función/genética , Hemostasis , Humanos , Agregación Plaquetaria , Dominios Proteicos/genética , Enfermedades de von Willebrand/sangre , Factor de von Willebrand/metabolismo
5.
Front Immunol ; 13: 1078891, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591269

RESUMEN

Introduction: The intravascular formation of neutrophil extracellular traps (NETs) is a trigger for coagulation and blood vessel occlusion. NETs are released from neutrophils as a response to strong inflammatory signals in the course of different diseases such as COVID-19, cancer or antiphospholipid syndrome. NETs are composed of large, chromosomal DNA fibers decorated with a variety of proteins such as histones. Previous research suggested a close mechanistic crosstalk between NETs and the coagulation system involving the coagulation factor XII (FXII), von Willebrand factor (VWF) and tissue factor. However, the direct impact of NET-related DNA fibers on blood flow and blood aggregation independent of the coagulation cascade has remained elusive. Methods: In the present study, we used different microfluidic setups in combination with fluorescence microscopy to investigate the influence of neutrophil-derived extracellular DNA fibers on blood rheology, intravascular occlusion and activation of the complement system. Results: We found that extended DNA fiber networks decelerate blood flow and promote intravascular occlusion of blood vessels independent of the plasmatic coagulation. Associated with the DNA dependent occlusion of the flow channel was the strong activation of the complement system characterized by the production of complement component 5a (C5a). Vice versa, we detected that the local activation of the complement system at the vascular wall was a trigger for NET release. Discussion: In conclusion, we found that DNA fibers as the principal component of NETs are sufficient to induce blood aggregation even in the absence of the coagulation system. Moreover, we discovered that complement activation at the endothelial surface promoted NET formation. Our data envisions DNA degradation and complement inhibition as potential therapeutic strategies in NET-induced coagulopathies.


Asunto(s)
COVID-19 , Trampas Extracelulares , Humanos , Trampas Extracelulares/metabolismo , COVID-19/metabolismo , Neutrófilos/metabolismo , ADN/metabolismo , Activación de Complemento
7.
BMC Mol Cell Biol ; 21(1): 64, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917131

RESUMEN

BACKGROUND: It has been demonstrated that von Willebrand factor (VWF) mediated platelet-endothelium and platelet-platelet interactions are shear dependent. The VWF's mobility under dynamic conditions (e.g. flow) is pivotal to platelet adhesion and VWF-mediated aggregate formation in the cascade of VWF-platelet interactions in haemostasis. RESULTS: Combining microfluidic tools with fluorescence and reflection interference contrast microscopy (RICM), here we show, that specific deletions in the A-domains of the biopolymer VWF affect both, adhesion and aggregation properties independently. Intuitively, the deletion of the A1-domain led to a significant decrease in both adhesion and aggregate formation of platelets. Nevertheless, the deletion of the A2-domain revealed a completely different picture, with a significant increase in formation of rolling aggregates (gain of function). We predict that the A2-domain effectively 'masks' the potential between the platelet glycoprotein (GP) Ib and the VWF A1-domain. Furthermore, the deletion of the A3-domain led to no significant variation in either of the two functional characteristics. CONCLUSIONS: These data demonstrate that the macroscopic functional properties i.e. adhesion and aggregate formation cannot simply be assigned to the properties of one particular domain, but have to be explained by cooperative phenomena. The absence or presence of molecular entities likewise affects the properties (thermodynamic phenomenology) of its neighbours, therefore altering the macromolecular function.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/fisiología , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Factor de von Willebrand/metabolismo , Biopolímeros/metabolismo , Línea Celular , Fluorescencia , Células HEK293 , Hemostasis/fisiología , Humanos , Microfluídica/métodos , Microscopía/métodos , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo
8.
Pharmaceutics ; 12(7)2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32698388

RESUMEN

Although, drugs are required in the various skin compartments such as viable epidermis, dermis, or hair follicles, to efficiently treat skin diseases, drug delivery into and across the skin is still challenging. An improved understanding of skin barrier physiology is mandatory to optimize drug penetration and permeation. The various barriers of the skin have to be known in detail, which means methods are needed to measure their functionality and outside-in or inside-out passage of molecules through the various barriers. In this review, we summarize our current knowledge about mechanical barriers, i.e., stratum corneum and tight junctions, in interfollicular epidermis, hair follicles and glands. Furthermore, we discuss the barrier properties of the basement membrane and dermal blood vessels. Barrier alterations found in skin of patients with atopic dermatitis are described. Finally, we critically compare the up-to-date applicability of several physical, biochemical and microscopic methods such as transepidermal water loss, impedance spectroscopy, Raman spectroscopy, immunohistochemical stainings, optical coherence microscopy and multiphoton microscopy to distinctly address the different barriers and to measure permeation through these barriers in vitro and in vivo.

9.
Nucleic Acids Res ; 48(13): 7333-7344, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32496552

RESUMEN

Neutrophils release their intracellular content, DNA included, into the bloodstream to form neutrophil extracellular traps (NETs) that confine and kill circulating pathogens. The mechanosensitive adhesive blood protein, von Willebrand Factor (vWF), interacts with the extracellular DNA of NETs to potentially immobilize them during inflammatory and coagulatory conditions. Here, we elucidate the previously unknown molecular mechanism governing the DNA-vWF interaction by integrating atomistic, coarse-grained, and Brownian dynamics simulations, with thermophoresis, gel electrophoresis, fluorescence correlation spectroscopy (FCS), and microfluidic experiments. We demonstrate that, independently of its nucleotide sequence, double-stranded DNA binds to a specific helix of the vWF A1 domain, via three arginines. This interaction is attenuated by increasing the ionic strength. Our FCS and microfluidic measurements also highlight the key role shear-stress has in enabling this interaction. Our simulations attribute the previously-observed platelet-recruitment reduction and heparin-size modulation, upon establishment of DNA-vWF interactions, to indirect steric hindrance and partial overlap of the binding sites, respectively. Overall, we suggest electrostatics-guiding DNA to a specific protein binding site-as the main driving force defining DNA-vWF recognition. The molecular picture of a key shear-mediated DNA-protein interaction is provided here and it constitutes the basis for understanding NETs-mediated immune and hemostatic responses.


Asunto(s)
ADN/química , Simulación del Acoplamiento Molecular , Factor de von Willebrand/química , Sitios de Unión , ADN/metabolismo , Humanos , Simulación de Dinámica Molecular , Concentración Osmolar , Unión Proteica , Electricidad Estática , Factor de von Willebrand/metabolismo
10.
Blood ; 133(4): 356-365, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30366922

RESUMEN

The frequent von Willebrand factor (VWF) variant p.Phe2561Tyr is located within the C4 domain, which also harbors the platelet GPIIb/IIIa-binding RGD sequence. To investigate its potential effect on hemostasis, we genotyped 865 patients with coronary artery disease (CAD), 915 with myocardial infarction (MI), and 417 control patients (Ludwigshafen Risk and Cardiovascular Health Study) and performed functional studies of this variant. A univariate analysis of male and female carriers of the Tyr2561 allele aged 55 years or younger revealed an elevated risk for repeated MI (odds ratio, 2.53; 95% confidence interval [CI], 1.07-5.98). The odds ratio was even higher in females aged 55 years or younger, at a value of 5.93 (95% CI, 1.12-31.24). Cone and plate aggregometry showed that compared with Phe2561, Tyr2561 was associated with increased platelet aggregate size both in probands' blood and with the recombinant variants. Microfluidic assays revealed that the critical shear rate for inducing aggregate formation was decreased to 50% by Tyr2561 compared with Phe2561. Differences in C-domain circular dichroism spectra resulting from Tyr2561 suggest an increased shear sensitivity of VWF as a result of altered association of the C domains that disrupts the normal dimer interface. In summary, our data emphasize the functional effect of the VWF C4 domain for VWF-mediated platelet aggregation in a shear-dependent manner and provide the first evidence that a functional variant of VWF plays a role in arterial thromboembolism.


Asunto(s)
Alelos , Mutación con Ganancia de Función/genética , Predisposición Genética a la Enfermedad , Infarto del Miocardio/genética , Tirosina/genética , Factor de von Willebrand/genética , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Conformación Proteica , Factores de Riesgo , Factor de von Willebrand/química
11.
Sci Rep ; 8(1): 10945, 2018 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-30026593

RESUMEN

Microangiopathy with subsequent organ damage represents a major complication in several diseases. The mechanisms leading to microvascular occlusion include von Willebrand factor (VWF), notably the formation of ultra-large von Willebrand factor fibers (ULVWFs) and platelet aggregation. To date, the contribution of erythrocytes to vascular occlusion is incompletely clarified. We investigated the platelet-independent interaction between stressed erythrocytes and ULVWFs and its consequences for microcirculation and organ function under dynamic conditions. In response to shear stress, erythrocytes interacted strongly with VWF to initiate the formation of ULVWF/erythrocyte aggregates via the binding of Annexin V to the VWF A1 domain. VWF-erythrocyte adhesion was attenuated by heparin and the VWF-specific protease ADAMTS13. In an in vivo model of renal ischemia/reperfusion injury, erythrocytes adhered to capillaries of wild-type but not VWF-deficient mice and later resulted in less renal damage. In vivo imaging in mice confirmed the adhesion of stressed erythrocytes to the vessel wall. Moreover, enhanced eryptosis rates and increased VWF binding were detected in blood samples from patients with chronic renal failure. Our study demonstrates that stressed erythrocytes have a pronounced binding affinity to ULVWFs. The discovered mechanisms suggest that erythrocytes are essential for the pathogenesis of microangiopathies and renal damage by actively binding to ULVWFs.


Asunto(s)
Eritrocitos/citología , Insuficiencia Renal Crónica/metabolismo , Enfermedades Vasculares/metabolismo , Factor de von Willebrand/metabolismo , Proteína ADAMTS13/metabolismo , Animales , Adhesión Celular , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Humanos , Ratones , Dominios Proteicos , Resistencia al Corte , Estrés Mecánico , Factor de von Willebrand/química
12.
Sci Rep ; 7(1): 14278, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-29079767

RESUMEN

The protein von Willebrand factor (VWF) is essential in primary hemostasis, as it mediates platelet adhesion to vessel walls. VWF retains its compact (globule-like) shape in equilibrium due to internal molecular associations, but is able to stretch when a high enough shear stress is applied. Even though the shear-flow sensitivity of VWF conformation is well accepted, the behavior of VWF under realistic blood flow conditions remains poorly understood. We perform mesoscopic numerical simulations together with microfluidic experiments in order to characterize VWF behavior in blood flow for a wide range of flow-rate and hematocrit conditions. In particular, our results demonstrate that the compact shape of VWF is important for its migration (or margination) toward vessel walls and that VWF stretches primarily in a near-wall region in blood flow making its adhesion possible. Our results show that VWF is a highly optimized protein in terms of its size and internal associations which are necessary to achieve its vital function. A better understanding of the relevant mechanisms for VWF behavior in microcirculation provides a further step toward the elucidation of the role of mutations in various VWF-related diseases.


Asunto(s)
Células Sanguíneas/citología , Adhesión Celular , Fenómenos Mecánicos , Movimiento , Factor de von Willebrand/metabolismo , Fenómenos Biomecánicos , Células HEK293 , Humanos , Dispositivos Laboratorio en un Chip , Modelos Biológicos , Estrés Mecánico
13.
Cancer Res ; 76(18): 5302-12, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27488527

RESUMEN

Tumor cells interact with blood constituents and these interactions promote metastasis. Selectins are vascular receptors facilitating interactions of tumor cells with platelets, leukocytes, and endothelium, but the role of endothelial E-selectin remains unclear. Here we show that E-selectin is a major receptor for monocyte recruitment to tumor cell-activated endothelium. Experimental and spontaneous lung metastasis using murine tumor cells, without E-selectin ligands, were attenuated in E-selectin-deficient mice. Tumor cell-derived CCL2 promoted endothelial activation, resulting in enhanced endothelial E-selectin expression. The recruitment of inflammatory monocytes to metastasizing tumor cells was dependent on the local endothelial activation and the presence of E-selectin. Monocytes promoted transendothelial migration of tumor cells through the induction of E-selectin-dependent endothelial retractions and a subsequent modulation of tight junctions through dephosphorylation of VE-cadherin. Thus, endothelial E-selectin shapes the tumor microenvironment through the recruitment, adhesion, and activation of monocytes that facilitate tumor cell extravasation and thereby metastasis. These findings provide evidence that endothelial E-selectin is a novel factor contributing to endothelial retraction required for efficient lung metastasis. Cancer Res; 76(18); 5302-12. ©2016 AACR.


Asunto(s)
Selectina E/metabolismo , Monocitos/metabolismo , Invasividad Neoplásica/patología , Migración Transendotelial y Transepitelial/fisiología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Citometría de Flujo , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Uniones Estrechas/metabolismo , Uniones Estrechas/patología
14.
Sci Rep ; 6: 22789, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27004454

RESUMEN

The application of multiphoton microscopy in the field of biomedical research and advanced diagnostics promises unique insights into the pathophysiology of inflammatory skin diseases. In the present study, we combined multiphoton-based intravital tomography (MPT) and fluorescence lifetime imaging (MPT-FLIM) within the scope of a clinical trial of atopic dermatitis with the aim of providing personalised data on the aetiopathology of inflammation in a non-invasive manner at patients' bedsides. These 'optical biopsies' generated via MPT were morphologically analysed and aligned with classical skin histology. Because of its subcellular resolution, MPT provided evidence of a redistribution of mitochondria in keratinocytes, indicating an altered cellular metabolism. Two independent morphometric algorithms reliably showed an even distribution in healthy skin and a perinuclear accumulation in inflamed skin. Moreover, using MPT-FLIM, detection of the onset and progression of inflammatory processes could be achieved. In conclusion, the change in the distribution of mitochondria upon inflammation and the verification of an altered cellular metabolism facilitate a better understanding of inflammatory skin diseases and may permit early diagnosis and therapy.


Asunto(s)
Dermatitis Atópica/diagnóstico por imagen , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Piel/patología , Tomografía Óptica/métodos , Algoritmos , Biopsia , Núcleo Celular/metabolismo , Células Cultivadas , Dermatitis Atópica/metabolismo , Humanos , Queratinocitos/metabolismo , Mitocondrias/metabolismo , Piel/citología , Piel/metabolismo
15.
Biophys J ; 108(9): 2312-21, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25954888

RESUMEN

Von Willebrand factor (VWF) plays a central role in hemostasis. Triggered by shear-stress, it adheres to platelets at sites of vascular injury. Inactivation of VWF has been associated to the shielding of its adhesion sites and proteolytic cleavage. However, the molecular nature of this shielding and its coupling to cleavage under shear-forces in flowing blood remain unknown. In this study, we describe, to our knowledge, a new force-sensory mechanism for VWF-platelet binding, which addresses these questions, based on a combination of molecular dynamics (MD) simulations, atomic force microscopy (AFM), and microfluidic experiments. Our MD simulations demonstrate that the VWF A2 domain targets a specific region at the VWF A1 domain, corresponding to the binding site of the platelet glycoprotein Ibα (GPIbα) receptor, thereby causing its blockage. This implies autoinhibition of the VWF for the binding of platelets mediated by the A1-A2 protein-protein interaction. During force-probe MD simulations, a stretching force dissociated the A1A2 complex, thereby unblocking the GPIbα binding site. Dissociation was found to be coupled to the unfolding of the A2 domain, with dissociation predominantly occurring before exposure of the cleavage site in A2, an observation that is supported by our AFM experiments. This suggests that the A2 domain prevents platelet binding in a force-dependent manner, ensuring that VWF initiates hemostasis before inactivation by proteolytic cleavage. Microfluidic experiments with an A2-deletion VWF mutant resulted in increased platelet binding, corroborating the key autoinhibitory role of the A2 domain within VWF multimers. Overall, autoinhibition of VWF mediated by force-dependent interdomain interactions offers the molecular basis for the shear-sensitive growth of VWF-platelet aggregates, and might be similarly involved in shear-induced VWF self-aggregation and other force-sensing functions in hemostasis.


Asunto(s)
Simulación de Dinámica Molecular , Factor de von Willebrand/química , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Datos de Secuencia Molecular , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Factor de von Willebrand/metabolismo
16.
Thromb Haemost ; 113(5): 1095-108, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25608503

RESUMEN

Co-stimulation via CD154 binding to CD40, pivotal for both innate and adaptive immunity, may also link haemostasis to vascular remodelling. Here we demonstrate that human platelet-bound or recombinant soluble CD154 (sCD154) elicit the release from and tethering of ultra-large (UL) von Willebrand factor (vWF) multimers to the surface of human cultured endothelial cells (ECs) exposed to shear stress. This CD40-mediated ULVWF multimer release from the Weibel-Palade bodies was triggered by consecutive activation of TRAF6, the tyrosine kinase c-Src and phospholipase Cγ1 followed by inositol-1,4,5 trisphosphate-mediated calcium mobilisation. Subsequent exposure to human washed platelets caused ULVWF multimer-platelet string formation on the EC surface in a shear stress-dependent manner. Platelets tethered to these ULVWF multimers exhibited P-selectin on their surface and captured labelled monocytes from the superfusate. When exposed to shear stress and sCD154, native ECs from wild-type but not CD40 or vWF-deficient mice revealed a comparable release of ULVWF multimers to which murine washed platelets rapidly adhered, turning P-selectin-positive and subsequently capturing monocytes from the perfusate. This novel CD154-provoked ULVWF multimer-platelet string formation at normal to fast flow may contribute to vascular remodelling processes requiring the perivascular or intravascular accumulation of pro-inflammatory macrophages such as arteriogenesis or atherosclerosis.


Asunto(s)
Ligando de CD40/metabolismo , Células Endoteliales/metabolismo , Factor de von Willebrand/metabolismo , Animales , Arterias/metabolismo , Aterosclerosis/metabolismo , Plaquetas/metabolismo , Calcio/química , Arteria Carótida Común/patología , Adhesión Celular , Electrofisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Isquemia/patología , Ratones , Microscopía Fluorescente , Monocitos/citología , Monocitos/metabolismo , Selectina-P/metabolismo , Perfusión , Proteínas Recombinantes/metabolismo , Resistencia al Corte , Transducción de Señal , Accidente Cerebrovascular , Fosfolipasas de Tipo C/metabolismo , Cuerpos de Weibel-Palade/metabolismo
17.
Exp Dermatol ; 23(9): 670-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25041487

RESUMEN

Spreading of melanoma is associated with efficient extravasation of circulating tumor cells from the vascular system into distant target organs. This process is accompanied and supported by proinflammatory and procoagulatory conditions. In this study, we analysed the ability of human melanoma cell lines to activate endothelial cells (ECs) in vitro. Some melanoma cells, that is, MV3, were shown to trigger an prompt calcium-flux-dependent, procoagulatory endothelial response that was accompanied by luminal release of ultra-large von Willebrand factor (ULVWF) fibres that were immobilized to the endothelial surface layer. In contrast to MV3-derived supernatant, prolonged treatment of ECs with WM9-derived supernatant mediated a pronounced activation of nuclear factor kappa B (NFκB). NFκB activation in ECs was dependent on both IL-1α and IL-1ß secreted from melanoma cells. Melanoma-derived IL-1 mediated an upregulation of proinflammatory cytokines IL-6 and IL-8, the intercellular adhesion molecule-1 (ICAM-1), the vascular cell adhesion molecule-1 (VCAM-1) and the procoagulatory tissue factor (TF) in ECs. Our data show that melanoma cells activate ECs either directly and within seconds or by an IL-1-mediated NFκB activation. Both pathways of EC activation convert the regular repressive function of ECs on inflammation and coagulation to a proinflammatory and procoagulatory surface that supports tumor progression.


Asunto(s)
Interleucina-1/metabolismo , Melanoma/metabolismo , FN-kappa B/metabolismo , Señalización del Calcio , Permeabilidad Capilar , Línea Celular Tumoral , Citocinas/metabolismo , Progresión de la Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Melanoma/irrigación sanguínea , Melanoma/patología , Modelos Biológicos , Fenotipo , Tromboplastina/metabolismo , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factor de von Willebrand/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 34(7): 1382-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24790143

RESUMEN

OBJECTIVE: Inflammatory conditions provoke essential processes in the human vascular system. It leads to the formation of ultralarge von Willebrand factor (VWF) fibers, which are immobilized on the endothelial cell surface and transform to highly adhesive strings under shear conditions. Furthermore, leukocytes release a meshwork of DNA (neutrophil extracellular traps) during the process of the recently discovered cell death program NETosis. In the present study, we characterized the interaction between VWF and DNA and possible binding sites to underline the role of VWF in thrombosis and inflammation besides its function in platelet adhesion. APPROACH AND RESULTS: Both functionalized surfaces and intact cell layers of human umbilical vein endothelial cells were perfused with isolated, protein-free DNA or leukocytes from whole blood at distinct shear rates. DNA-VWF interaction was monitored using fluorescence microscopy, ELISA-based assays, molecular dynamics simulations, and electrostatic potential calculations. Isolated DNA, as well as DNA released by stimulated leukocytes, was able to bind to shear-activated, but not inactivated, VWF. However, DNA-VWF binding does not alter VWF degradation by a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13. Moreover, DNA-VWF interaction can be blocked using unfractionated and low-molecular-weight heparin, and DNA-VWF complexes attenuate platelet binding to VWF. These findings were supported using molecular dynamics simulations and electrostatic calculations of the A1- and A2-domains. CONCLUSIONS: Our findings suggest that VWF directly binds and immobilizes extracellular DNA released from leukocytes. Therefore, we hypothesize that VWF might act as a linker for leukocyte adhesion to endothelial cells, supporting leukocyte extravasation and inflammation.


Asunto(s)
Adhesión Celular , ADN/metabolismo , Neutrófilos/metabolismo , Factor de von Willebrand/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Sitios de Unión , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Heparina de Bajo-Peso-Molecular/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Modelos Moleculares , Neutrófilos/efectos de los fármacos , Adhesividad Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Proteolisis , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Tiempo , Factor de von Willebrand/química
19.
PLoS One ; 9(2): e89491, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24558503

RESUMEN

During development and progression of malignant melanoma, an important role has been attributed to alterations of cell-cell adhesions, in particular, to a "cadherin switch" from E- to N-cadherin. We have previously shown that a subtype of melanoma cells express the desmosomal cadherin desmoglein 2 as non-junction-bound cell surface protein in addition to classical cadherins. To study the role of desmoglein 2 in melanoma cells, melanoma lines containing high endogenous amounts of desmoglein 2 were depleted of the protein by RNA interference. Transwell migration and scratch wounding assays showed markedly increased migration upon desmoglein 2 suppression whereas proliferation and viability remained unaltered. In gene expression profiles, desmoglein 2 depletion was associated with overexpression of migration-related genes. Strongest overexpression was found for secretogranin II which has not been reported in melanoma cells before. The bioactive peptide derived from secretogranin II, secretoneurin, is known to exert chemoattractive functions and was demonstrated here to stimulate melanoma cell migration. In summary, we show that desmoglein 2 expression attenuates migration of melanoma cells. The mechanism of desmoglein 2 impaired cell migration is mediated by downregulation of secretogranin II. Loss of desmoglein 2 increases expression of secretogranin II, followed by an enhanced migratory activity of melanoma cells. Our data add a new pathway of regulating melanoma cell migration related to a desmoglein 2-secretogranin II axis.


Asunto(s)
Movimiento Celular/fisiología , Desmogleína 2/metabolismo , Regulación de la Expresión Génica/fisiología , Melanoma/fisiopatología , Bromodesoxiuridina , Línea Celular Tumoral , Movimiento Celular/genética , Desmogleína 2/deficiencia , Impedancia Eléctrica , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Microscopía Fluorescente , Interferencia de ARN , Radioinmunoensayo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Secretogranina II/metabolismo
20.
Thromb Haemost ; 111(4): 598-609, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24573248

RESUMEN

The specific interactions of von Willebrand factor (VWF) with the vessel wall, platelets or other interfaces strongly depend on (a shear-induced) VWF activation. Shear flow has been shown to induce a conformational transition of VWF, but is modulated by its thermodynamic state (state-function relationship). The state in turn is determined by physical (e.g. vessel geometry), physico-chemical (e.g. pH) and molecular-biological (e.g. mutants, binding) factors. Combining established results with recent insights, we reconstruct VWF biology and its state-function relationship from endothelial cell release to final degradation in the human vasculature. After VWF secretion, endothelial-anchored and shear activated VWF multimers can rapidly interact with surrounding colloids, typically with platelets. Simultaneously, this VWF activation enables ADAMTS13 to cleave VWF multimers thereby limiting VWF binding capacity. The subsequent cell-surface dissociation leads to a VWF recoiling to a globular conformation, shielding from further degradation by ADAMTS13. High local concentrations of these soluble VWF multimers, transported to the downstream vasculature, are capable for an immediate reactivation and re-polymerisation initiating colloid-binding or VWF-colloid aggregation at the site of inflamed endothelium, vessel injuries or pathological high-shear areas. Focusing on these functional steps in the lifecycle of VWF, its qualitative and quantitative deficiencies in the different VWD types will facilitate more precise diagnostics and reliable risk stratification for prophylactic therapies. The underlying biophysical principles are of general character, which broadens prospective studies on the physiological and pathophysiological impact of VWF and VWF-associated diseases and beares hope for a more universal understanding of an entire class of phenomena.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Endotelio Vascular/metabolismo , Factor de von Willebrand/fisiología , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Regulación Alostérica , Animales , Trastornos de la Coagulación Sanguínea/genética , Trastornos de la Coagulación Sanguínea/fisiopatología , Humanos , Mutación/genética , Activación Plaquetaria , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Proteolisis , Relación Estructura-Actividad , Termodinámica , Factor de von Willebrand/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...