Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 1078891, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591269

RESUMO

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.


Assuntos
COVID-19 , Armadilhas Extracelulares , Humanos , Armadilhas Extracelulares/metabolismo , COVID-19/metabolismo , Neutrófilos/metabolismo , DNA/metabolismo , Ativação do Complemento
2.
Cancer Res ; 76(18): 5302-12, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27488527

RESUMO

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.


Assuntos
Selectina E/metabolismo , Monócitos/metabolismo , Invasividade Neoplásica/patologia , Migração Transendotelial e Transepitelial/fisiologia , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase , Junções Íntimas/metabolismo , Junções Íntimas/patologia
3.
Sci Rep ; 6: 22789, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27004454

RESUMO

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.


Assuntos
Dermatite Atópica/diagnóstico por imagem , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Pele/patologia , Tomografia Óptica/métodos , Algoritmos , Biópsia , Núcleo Celular/metabolismo , Células Cultivadas , Dermatite Atópica/metabolismo , Humanos , Queratinócitos/metabolismo , Mitocôndrias/metabolismo , Pele/citologia , Pele/metabolismo
4.
Thromb Haemost ; 113(5): 1095-108, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25608503

RESUMO

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.


Assuntos
Ligante de CD40/metabolismo , Células Endoteliais/metabolismo , Fator de von Willebrand/metabolismo , Animais , Artérias/metabolismo , Aterosclerose/metabolismo , Plaquetas/metabolismo , Cálcio/química , Artéria Carótida Primitiva/patologia , Adesão Celular , Eletrofisiologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Isquemia/patologia , Camundongos , Microscopia de Fluorescência , Monócitos/citologia , Monócitos/metabolismo , Selectina-P/metabolismo , Perfusão , Proteínas Recombinantes/metabolismo , Resistência ao Cisalhamento , Transdução de Sinais , Acidente Vascular Cerebral , Fosfolipases Tipo C/metabolismo , Corpos de Weibel-Palade/metabolismo
5.
Exp Dermatol ; 23(9): 670-6, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25041487

RESUMO

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.


Assuntos
Interleucina-1/metabolismo , Melanoma/metabolismo , NF-kappa B/metabolismo , Sinalização do Cálcio , Permeabilidade Capilar , Linhagem Celular Tumoral , Citocinas/metabolismo , Progressão da Doença , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Melanoma/irrigação sanguínea , Melanoma/patologia , Modelos Biológicos , Fenótipo , Tromboplastina/metabolismo , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/metabolismo , Fator de von Willebrand/metabolismo
6.
PLoS One ; 9(2): e89491, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558503

RESUMO

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.


Assuntos
Movimento Celular/fisiologia , Desmogleína 2/metabolismo , Regulação da Expressão Gênica/fisiologia , Melanoma/fisiopatologia , Bromodesoxiuridina , Linhagem Celular Tumoral , Movimento Celular/genética , Desmogleína 2/deficiência , Impedância Elétrica , Perfilação da Expressão Gênica , Humanos , Immunoblotting , Imuno-Histoquímica , Microscopia de Fluorescência , Interferência de RNA , Radioimunoensaio , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Secretogranina II/metabolismo
7.
Stem Cells Dev ; 22(5): 791-803, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23025550

RESUMO

The increasing use of mesenchymal stromal cells (MSC) in clinical cellular therapy requires a safe and controlled production process compliant with Good Manufacturing Practice guidelines. Pooled blood group AB human serum (HS) has been used to replace fetal bovine serum (FBS), critically rated by the regulatory agencies, since it can support the expansion of adipose tissue-derived mesenchymal stromal cells (ASC). However, it remains unknown whether the choice of serum affects application-relevant characteristics of ASC. A microarray-based screen has revealed differentially expressed adhesion and extracellular matrix-associated molecules in HS- and FBS-ASC. Since cell therapy relies on the cells' efficacy to home and engraft, HS- and FBS-ASC were compared by analyzing adhesion, migration, and transmigration as well as short-term homing in vivo. HS-cultivated ASC demonstrated a higher adhesion to plastic, but reduced adhesion to extracellular matrix molecules, that is, laminin, and to endothelial cells both under static and flow conditions. Migration and transmigration assays confirmed the attraction of ASC by the tumor conditioned medium irrespective of the supplement. Coinjecting differently labeled HS- and FBS-ASC into nonobese diabetic, severe combined immunodeficiency mice revealed reduced numbers of HS-ASC in lungs and liver. This has been interpreted as reduced capillary entrapment. Our data indicate that varying the serum supplement may alter application-relevant characteristics of ASC, such as adhesion, as well as lung entrapment after infusion. Appropriate injury models and further molecular analyses are required to provide mechanistic insight into the differential effects of HS versus FBS on ASC cultures.


Assuntos
Tecido Adiposo/citologia , Adesão Celular , Células-Tronco Mesenquimais/fisiologia , Soro/metabolismo , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Meios de Cultura , Endotélio/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
8.
Steroids ; 77(14): 1543-50, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23064006

RESUMO

Progesterone receptor membrane component type 2 (PGRMC2) is strongly homologous to PGRMC1 which is highly expressed in ovarian cancer and other cancer cells and was claimed to play an important role in chemotherapy resistance. Whereas PGRMC1 has been extensively characterized in in vitro studies, comparably little is known about PGRMC2. To determine PGRMC2's role in ovarian cancer cell proliferation and mobility PGRMC1- and 2-depleted and -overexpressing SKOV-3 cells were generated. In electric cell-substrate impedance sensing studies, PGRMC2 negatively affects SKOV-3 migration rate if overexpressed; oppositely, depletion was associated with an increased migration rate. PGRMC1 had no effect in this assay. These effects were not associated with f-actin regulation or actin cytoskeleton reorganization. Yet, these highly homologous proteins share many properties. Both PGRMC1 and 2 are localized to the endoplasmic reticulum. As PGRMC1 was reported to interact with cytochrome P450 proteins (CYP) binding of two different CYPs to PGRMC2 was tested; a stable interaction of PGRMC2 with CYP3A4 and CYP21A2 was found in human embryonic kidney cells. For both PGRMC types, cell viability assays revealed no significant differences of SKOV-3 survival in overexpressing and depleted cells. PGRMC2 also does not seem to have any influence on the apoptotic effect of cisplatin or the antiapoptotic effect of progesterone which had been reported for PGRMC1. In contrast to PGRMC1, protein levels of PGRMC2 in SKOV-3 cells are reduced by treatment with cisplatin (30-60µM). In conclusion, we show for the first time that PGRMC2 inhibits migration of SKOV-3 ovarian cancer cells in vitro.


Assuntos
Movimento Celular , Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Células HEK293 , Humanos , Proteínas de Membrana/antagonistas & inibidores , Neoplasias Ovarianas , Receptores de Progesterona/antagonistas & inibidores
9.
Am J Pathol ; 181(2): 693-705, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22659470

RESUMO

Tumor cell extravasation is a critical step in the metastatic cascade and requires interaction between the tumor cell and the endothelium. Although cancer progression depends on a complex network of mechanisms, including inflammation and coagulation, the involvement of tumor-induced endothelium activation and the subsequent release of procoagulatory factors in this process are not well understood. Using tissue sections from patients with malignant melanoma, immunofluorescence studies for the presence of von Willebrand factor (VWF) clearly demonstrated endothelium activation and the formation of ultra-large VWF fibers in these patients. In vitro analyses revealed that supernatants from highly invasive melanoma cells induced an acute endothelium activation measured by VWF, P-selectin, and angiopoietin-2 release. Proteome profiling identified vascular endothelial growth factor A (VEGF-A) as the main mediator of endothelium activation. Inhibition and knock-down of VEGF-A in melanoma cells led to a rigorous decrease in VWF exocytosis. Selective small-interfering RNA to matrix metalloproteinase-2 (MMP-2) inhibited endothelium activation, and this effect correlated with reduced VEGF-A content in the supernatants of melanoma cells. Further experiments showed that active MMP-2 regulates VEGF-A in melanoma cells on a transcriptional level via an integrin αvß5/phosphoinositide-3-kinase-dependent pathway. In conclusion, these results indicate an important role of VEGF-A in acute endothelium activation and provide clear evidence that MMP-2 plays a pivotal role in the autocrine regulation of VEGF-A expression in melanoma cells.


Assuntos
Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Metaloproteinase 2 da Matriz/metabolismo , Melanoma/enzimologia , Melanoma/patologia , Receptores de Vitronectina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Melanoma/irrigação sanguínea , Melanoma/genética , Camundongos , Modelos Biológicos , Invasividade Neoplásica , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator de von Willebrand/metabolismo
10.
Thromb Haemost ; 98(2): 283-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17721608

RESUMO

Neoplastic diseases are often associated with thromboembolic events, however the precise mechanism underlying this observation is a matter of ongoing investigation. It is known that matrixmetalloproteinase-1 (MMP-1) canonically activates the thrombin receptor (PAR-1) and we recently reported that highly metastatic tumor cells of melanoma and colon cancer are secreting matrixmetalloproteinase-1. This tumor-derived MMP1 was shown to be a major activator of endothelial PAR-1, thus leading to endothelial cell activation. As tumor-induced thrombosis is a characteristic of metastazing tumors, we investigated whether tumor-derived supernatant (TUSN) from melanoma and colon cancer may induce adhesion of circulating platelets, an initial step in thrombus formation. A time-course study revealed that TU-SN induces a rapid secretion of von Willebrand factor (VWF) within minutes. Using a novel microfluidic device we analyzed platelet-endothelial interactions in a closed circuit. Immunofluorescence imaging showed that TU-SN rapidly induces platelet-string formation via secreted VWF. We demonstrated that tumor-derived supernatant is a potent agonist inducing platelet adhesion under flow conditions.


Assuntos
Endotélio Vascular/patologia , Microfluídica/métodos , Neoplasias/complicações , Adesividade Plaquetária/efeitos dos fármacos , Tromboembolia/etiologia , Fator de von Willebrand/metabolismo , Células Cultivadas , Neoplasias do Colo/complicações , Neoplasias do Colo/metabolismo , Meios de Cultivo Condicionados/farmacologia , Humanos , Metaloproteinase 1 da Matriz , Melanoma/complicações , Melanoma/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Receptor PAR-1
11.
J Cell Physiol ; 211(2): 399-409, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17167769

RESUMO

Reperfusion after ischemic conditions induces massive endothelial cell (EC) activation, an initial step of reperfusion injury. Reperfusion is characterized by reoxygenation, realkalinization and a localized increase of inflammatory stimuli. In this study, we focused on the influence of extracellular realkalinization on human umbilical vein endothelial cell (HUVEC) activation. We examined intracellular pH (pH(in)) and intracellular free calcium concentration ([Ca(2+)](in)), a second messenger known to mediate von Willebrand factor (VWF) exocytosis in endothelium, upon realkalinization. Furthermore, we measured the agonist-stimulated exocytosis of VWF, Interleukin-8 and soluble P-selectin (sP-Selectin) as markers of EC activation. To verify a morphological correlate of EC activation, we finally observed platelet-endothelial adherence during realkalinization using shear flow. Realkalinization of HUVEC was simulated by switching from bicarbonate buffered Ringer solution of an acidotic pH(ex) of 6.4 to a physiologic pH(ex) of 7.4. Extracellular realkalinization was accompanied by pH(in) recovery from 6.5 to 7.2 within 10 min. Application of cariporide, an inhibitor of the Na(+)/H(+) exchanger subtype 1 (NHE), during extracellular realkalinization significantly delayed the early kinetics of intracellular realkalinization. Histamine stimulated [Ca(2+)](in) was significantly increased upon realkalinization compared to control cells. Also agonist-stimulated release of VWF, Interleukin-8 and sP-Selectin was massively enhanced during pH(in) recovery in comparison to control. Furthermore, we observed an increased platelet binding to endothelium. Interestingly, each of these realkalinization-induced effects were significantly reduced by early application of cariporide. Therefore, delay of acute NHE-dependent pH(in) recovery may represent a promising mechanism for inhibition of EC activation upon reperfusion.


Assuntos
Acidose/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Líquido Intracelular/metabolismo , Traumatismo por Reperfusão/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Acidose/sangue , Acidose/fisiopatologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Cálcio/metabolismo , Adesão Celular , Células Cultivadas , Antiportadores de Cloreto-Bicarbonato/antagonistas & inibidores , Antiportadores de Cloreto-Bicarbonato/metabolismo , AMP Cíclico/metabolismo , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Exocitose , Guanidinas/farmacologia , Histamina/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Interleucina-8/metabolismo , Líquido Intracelular/efeitos dos fármacos , Cinética , Modelos Lineares , Metacrilatos/farmacologia , Selectina-P/metabolismo , Adesividade Plaquetária , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/fisiopatologia , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Sulfonas/farmacologia , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo , Fator de von Willebrand/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA