Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
J Intellect Disabil Res ; 66(12): 1023-1033, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35698301

RESUMO

BACKGROUND: The promotion of physical activity and the decrease of inactivity and sedentary behaviour are crucial for a healthy lifestyle and positive quality of life. People with intellectual disabilities are at increased risk of inactivity and sedentary behaviour. Therefore, it is important to increase their physical activity by implementing physical activity guidelines in their daily life. Professional direct care providers can play a decisive role in supporting people with intellectual disabilities to participate in physical activity, but the engagement of direct care providers with this role may be reflective of their own attitudes and beliefs towards physical activity. Therefore, the link between the implementation of current physical activity guidelines for people with intellectual disabilities and direct care providers' own beliefs and behaviour with regard to physical activity is investigated. METHOD: A total of 104 direct care providers completed self-reported questionnaires about their own physical activity behaviour (IPAQ-SF), recommendations for people with intellectual disabilities (adaption of EMIQ-HP) and questions regarding global physical activity guidelines. They were also asked about potential barriers and facilitators for the recommendation of physical activity in open-ended questions. RESULTS: Personal physical activity behaviour is related to the recommended physical activity for people with intellectual disabilities (moderate-to-vigorous physical activity: rs  = 0.408, P = 0.005). However, recommended physical activity behaviour for people with intellectual disabilities is significantly lower than direct care providers' own physical activity behaviour (P < 0.001). 47.1% of the respondents recommended people with intellectual disabilities to participate in less than the 150 min of moderate intensity physical activity per week for that is recommended in global physical activity guidelines. CONCLUSION: Direct care providers may hold stereotypical views and insecurities about the potential harms associated with people with intellectual disabilities participating in physical activity. Therefore, the dissemination of physical activity recommendations for people with intellectual disabilities should be a major target for health professionals, social workers and scientists to address direct care providers' concerns. Furthermore, we need to emphasise the benefits of regular physical activity to professional direct care providers and directly to people with intellectual disabilities.


Assuntos
Deficiência Intelectual , Humanos , Qualidade de Vida , Exercício Físico , Comportamento Sedentário , Atividade Motora
2.
BMC Public Health ; 20(1): 72, 2020 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-31948416

RESUMO

BACKGROUND: Combining the strengths of physical activity (PA) diaries and questionnaires may be needed to improve the unsatisfying measurement quality of existing PA questionnaires. This study investigated the construct validity of a short PA questionnaire (Physical Activity Questionnaire for 24 h [PAQ24]) with a recall period of one day. METHODS: In this cross-sectional study, participants completed the PAQ24 on seven consecutive days while wearing an accelerometer (GENEActiv). Thereafter, the Global Physical Activity Questionnaire (GPAQ) was completed. Spearman correlation coefficients and Bland-Altman analysis were used to assess construct validity. RESULTS: Overall, 50 active adults (11 women, mean age = 25.1 ± 2.5) participated. Relative agreements between Total PA of PAQ24 and accelerometer were 0.37 ≤ ρ ≤ 0.72 for each day with satisfying agreement on five out of seven days. Weekly relative agreement for Total PA was moderate (ρ = 0.44). Relative agreements between PAQ24 and GPAQ were ρ = 0.43 for Total PA. Daily and weekly absolute agreements were poor indicated by wide limits of agreement. CONCLUSIONS: In contrast to weekly Total PA, the majority of daily results of the PAQ24 showed satisfying construct validity. A short recall period may improve the measurement quality of PA questionnaires, but measurement errors and the costs of multiple administrations must be considered in future studies.


Assuntos
Exercício Físico , Rememoração Mental , Inquéritos e Questionários , Adulto , Estudos Transversais , Feminino , Humanos , Masculino , Reprodutibilidade dos Testes , Fatores de Tempo , Adulto Jovem
3.
Proc Natl Acad Sci U S A ; 106(46): 19497-502, 2009 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-19875693

RESUMO

The initiator of coagulation, full-length tissue factor (flTF), in complex with factor VIIa, influences angiogenesis through PAR-2. Recently, an alternatively spliced variant of TF (asTF) was discovered, in which part of the TF extracellular domain, the transmembrane, and cytoplasmic domains are replaced by a unique C terminus. Subcutaneous tumors produced by asTF-secreting cells revealed increased angiogenesis, but it remained unclear if and how angiogenesis is regulated by asTF. Here, we show that asTF enhances angiogenesis in matrigel plugs in mice, whereas a soluble form of flTF only modestly enhances angiogenesis. asTF dose-dependently upregulates angiogenesis ex vivo independent of either PAR-2 or VIIa. Rather, asTF was found to ligate integrins, resulting in downstream signaling. asTF-alphaVbeta3 integrin interaction induces endothelial cell migration, whereas asTF-dependent formation of capillaries in vitro is dependent on alpha6beta1 integrin. Finally, asTF-dependent aortic sprouting is sensitive to beta1 and beta3 integrin blockade and a TF-antibody that disrupts asTF-integrin interaction. We conclude that asTF, unlike flTF, does not affect angiogenesis via PAR-dependent pathways but relies on integrin ligation. These findings indicate that asTF may serve as a target to prevent pathological angiogenesis.


Assuntos
Processamento Alternativo , Integrina alfa6beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Neovascularização Patológica/genética , Tromboplastina/genética , Animais , Aorta/crescimento & desenvolvimento , Aorta/metabolismo , Capilares/crescimento & desenvolvimento , Capilares/metabolismo , Movimento Celular , Endotélio Vascular/metabolismo , Fator V/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor PAR-2/metabolismo
4.
Br J Cancer ; 101(12): 2023-9, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19904262

RESUMO

BACKGROUND: Thromboembolic events are a major complication in ovarian cancer patients. Tissue factor (TF) is frequently overexpressed in ovarian cancer tissue and correlates with intravascular thrombosis. TF binds to coagulation factor VII (fVII), changing it to its active form, fVIIa. This leads to activation of the extrinsic coagulation cascade. fVII is produced by the liver and believed to be supplied from blood plasma at the site of coagulation. However, we recently showed that ovarian cancer cells express fVII transcripts under normoxia and that this transcription is inducible under hypoxia. These findings led us to hypothesise that ovarian cancer cells are intrinsically associated with TF-fVIIa coagulation activity, which could result in thrombosis. METHODS: In this study, we examined whether ectopically expressed fVII could cause thrombosis by means of immunohistochemistry, RT-PCR, western blotting and flow cytometry. RESULTS: Ectopic fVII expression occurs frequently in ovarian cancers, particularly in clear cell carcinoma. We further showed that ovarian cancer cells express TF-fVIIa on the cell surface under normoxia and that this procoagulant activity is enhanced by hypoxic stimuli. Moreover, we showed that ovarian cancer cells secrete microparticles (MPs) with TF-fVIIa activity. Production of this procoagulant secretion is enhanced under hypoxia. CONCLUSION: These results raise the possibility that cancer cell-derived TF-fVIIa could cause thrombotic events in ovarian cancer patients.


Assuntos
Fator VII/metabolismo , Neoplasias Ovarianas/metabolismo , Tromboplastina/metabolismo , Tromboembolia Venosa/etiologia , Hipóxia Celular , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Feminino , Humanos , Neoplasias Epiteliais e Glandulares/química , Neoplasias Ovarianas/complicações , Neoplasias Ovarianas/patologia
5.
J Cell Biol ; 133(2): 293-304, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8609163

RESUMO

The ability to regulate proteolytic functions is critical to cell biology. We describe events that regulate the initiation of the coagulation cascade on endothelial cell surfaces. The transmembrane protease receptor tissue factor (TF) triggers coagulation by forming an enzymatic complex with the serine protease factor VIIa (VIIa) that activates substrate factor X to the protease factor Xa (Xa). Feedback inhibition of the TF-VIIa enzymatic complex is achieved by the formation of a quaternary complex of TF-VIIa, Xa, and the Kunitz-type inhibitor tissue factor pathway inhibitor (TFPI). Concomitant with the downregulation of TF-VIIa function on endothelial cells, we demonstrate by immunogold EM that TF redistributes to caveolae. Consistently, TF translocates from the Triton X-100-soluble membrane fractions to low-density, detergent-insoluble microdomains that inefficiently support TF-VIIa proteolytic function. Downregulation of TF-VIIa function is dependent on quaternary complex formation with TFPI that is detected predominantly in detergent-insoluble microdomains. Partitioning of TFPI into low-density fractions results from the association of the inhibitor with glycosyl phosphatidylinositol anchored binding sites on external membranes. Free Xa is not efficiently bound by cell-associated TFPI; hence, we propose that the transient ternary complex of TF-VIIa with Xa supports translocation and assembly with TFPI in glycosphingolipid-rich microdomains. The redistribution of TF provides evidence for an assembly-dependent translocation of the inhibited TF initiation complex into caveolae, thus implicating caveolae in the regulation of cell surface proteolytic activity.


Assuntos
Coagulação Sanguínea/fisiologia , Caveolinas , Endotélio Vascular/metabolismo , Fator VIIa/metabolismo , Inibidores do Fator Xa , Lipoproteínas/metabolismo , Tromboplastina/metabolismo , Caveolina 1 , Fracionamento Celular , Linhagem Celular Transformada , Membrana Celular/química , Membrana Celular/metabolismo , Vesículas Revestidas/metabolismo , Detergentes , Regulação para Baixo , Fator Xa/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Humanos , Ligantes , Proteínas de Membrana/análise , Octoxinol , Solubilidade , Tromboplastina/análise , Tromboplastina/química , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais
6.
J Cell Biol ; 109(1): 389-95, 1989 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2663880

RESUMO

Tissue factor (TF) is an integral membrane glycoprotein which, as the receptor and essential cofactor for coagulation factors VII and VIIa (FVII and FVIIa, respectively), is the primary cellular activator of the coagulation protease cascade. Previous studies on the procoagulant activity of a variety of cell types (either lysed or in the intact state) have variously been interpreted as showing that TF is either stored intracellularly or is present in a cryptic form in the surface membrane. Using mAbs to TF, we have directly investigated the subcellular localization and functional activity of TF in lipopolysaccharide-stimulated blood monocytes and J82 bladder carcinoma cells. Blocking of surface TF of viable cells with inhibitory anti-TF mAbs abolished greater than 90% of TF activity of the intact cells as well as of lysed cells. Furthermore, quantitative analysis of the binding of FVII and anti-TF mAb to J82 cells demonstrated that all surface-expressed TF molecules were capable of binding the ligand, FVII. By immunoelectron microscopy, TF was present only in the surface membrane of monocytes and J82 cells, although the latter also contained apparently inactive TF antigen in multivesicular bodies. On the intact cell surface the catalytic activity of the TF-FVIIa complex was investigated and found to be markedly less relative to cell lysates. Membrane alterations that affect the cofactor activity of TF may be a means of regulating the extent of initiation of the coagulation protease cascade in various cellular settings.


Assuntos
Carcinoma/metabolismo , Monócitos/metabolismo , Tromboplastina/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Anticorpos Monoclonais/imunologia , Compartimento Celular , Membrana Celular/metabolismo , Fator VII/metabolismo , Fator Xa , Humanos , Técnicas Imunoenzimáticas , Técnicas In Vitro , Lipopolissacarídeos/farmacologia , Microscopia Eletrônica , Serina Endopeptidases/metabolismo , Células Tumorais Cultivadas
7.
J Cell Biol ; 140(5): 1241-53, 1998 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-9490735

RESUMO

Tissue factor (TF), the protease receptor initiating the coagulation system, functions in vascular development, angiogenesis, and tumor cell metastasis by poorly defined molecular mechanisms. We demonstrate that immobilized ligands for TF specifically support cell adhesion, migration, spreading, and intracellular signaling, which are not inhibited by RGD peptides. Two-hybrid screening identified actin-binding protein 280 (ABP-280) as ligand for the TF cytoplasmic domain. Extracellular ligation of TF is necessary for ABP-280 binding. ABP-280 recruitment to TF adhesion contacts is associated with reorganization of actin filaments, but cytoskeletal adaptor molecules typically found in integrin-mediated focal contacts are not associated with TF. Chimeric molecules of the TF cytoplasmic domain and an unrelated extracellular domain support cell spreading and migration, demonstrating that the extracellular domain of TF is not involved in the recruitment of accessory molecules that influence adhesive functions. Replacement of TF's cytoplasmic Ser residues with Asp to mimic phosphorylation enhances the interaction with ABP-280, whereas Ala mutations abolish coprecipitation of ABP-280 with immobilized TF cytoplasmic domain, and severely reduce cell spreading. The specific interaction of the TF cytoplasmic domain with ABP-280 provides a molecular pathway by which TF supports tumor cell metastasis and vascular remodeling.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Tromboplastina/metabolismo , Sítios de Ligação , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Transformada , Precipitação Química , Citoplasma/metabolismo , Filaminas , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Ligantes , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Células Tumorais Cultivadas
8.
J Thromb Haemost ; 16(10): 1941-1952, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30030891

RESUMO

The tissue factor (TF) pathway plays a central role in hemostasis and thrombo-inflammatory diseases. Although structure-function relationships of the TF initiation complex are elucidated, new facets of the dynamic regulation of TF's activities in cells continue to emerge. Cellular pathways that render TF non-coagulant participate in signaling of distinct TF complexes with associated proteases through the protease-activated receptor (PAR) family of G protein-coupled receptors. Additional co-receptors, including the endothelial protein C receptor (EPCR) and integrins, confer signaling specificity by directing subcellular localization and trafficking. We here review how TF is switched between its role in coagulation and cell signaling through thiol-disulfide exchange reactions in the context of physiologically relevant lipid microdomains. Inflammatory mediators, including reactive oxygen species, activators of the inflammasome, and the complement cascade play pivotal roles in TF procoagulant activation on monocytes, macrophages and endothelial cells. We furthermore discuss how TF, intracellular ligands, co-receptors and associated proteases are integrated in PAR-dependent cell signaling pathways controlling innate immunity, cancer and metabolic inflammation. Knowledge of the precise interactions of TF in coagulation and cell signaling is important for understanding effects of new anticoagulants beyond thrombosis and identification of new applications of these drugs for potential additional therapeutic benefits.


Assuntos
Coagulação Sanguínea , Transdução de Sinais , Tromboplastina/metabolismo , Animais , Células Endoteliais/metabolismo , Fator VIIa/metabolismo , Fator Xa/metabolismo , Humanos , Inflamação/sangue , Células Mieloides/metabolismo , Neoplasias/sangue , Receptor PAR-2/sangue , Trombose/sangue
9.
Thromb Haemost ; 118(2): 229-250, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29378352

RESUMO

Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.


Assuntos
Tromboembolia/terapia , Trombose/sangue , Trombose/terapia , Anticoagulantes/uso terapêutico , Biomarcadores/sangue , Coagulação Sanguínea , Eritrócitos/metabolismo , Fator VIII/metabolismo , Fator XII/metabolismo , Fator XIII/metabolismo , Humanos , Macrófagos/metabolismo , Países Baixos , Fenótipo , Placa Aterosclerótica/sangue , Placa Aterosclerótica/diagnóstico , Placa Aterosclerótica/terapia , Polifosfatos/metabolismo , Fatores de Risco , Transdução de Sinais , Tromboembolia/sangue , Tromboembolia/diagnóstico , Trombose/diagnóstico
10.
J Clin Invest ; 101(7): 1372-8, 1998 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9525979

RESUMO

Tissue factor (TF), the initiating cell surface receptor of the coagulation cascade, plays important roles in embryogenesis, angiogenesis, and tumor cell metastasis. It is controversial whether proteolytic function of TF complexed with its serine protease ligand VIIa is required for metastatic tumor dissemination. We show here in a model for TF-dependent experimental hematogenous metastasis, that TF supports metastasis by both proteolytic activity of the TF-VIIa complex and currently undefined functions of the cytoplasmic domain. We demonstrate that ligand binding of VIIa to TF is required for metastasis. Antimetastatic properties of covalently inactivated VIIa provide evidence that ligand binding is insufficient per se to support metastasis, emphasizing that proteolytic activity is necessary for the metastatic process. Ala or Asp mutations of cytoplasmic serine residues were introduced to preclude or mimic phosphorylation. In vivo analysis of these mutants suggests that local protease generation on the tumor cell surface does not serve simply to activate the cytoplasmic domain of TF by serine phosphorylation. Thus, extracellular functions of the catalytically active TF-VIIa complex cooperate with specific functions of the TF cytoplasmic domain to support the complex process of hematogenous tumor cell dissemination.


Assuntos
Fator VIIa/metabolismo , Metástase Neoplásica , Tromboplastina/metabolismo , Animais , Células CHO , Cricetinae , Citoplasma/metabolismo , Feminino , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Fosforilação , Relação Estrutura-Atividade , Tromboplastina/química , Transfecção
11.
J Clin Invest ; 104(9): 1213-21, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10545520

RESUMO

Tissue factor (TF), the cell-surface receptor for coagulation factor VIIa, supports metastasis. Equally important for this process are (a) interactions of the TF cytoplasmic domain, which binds the mobility-enhancing actin-binding protein 280, and (b) the formation of a proteolytically active TF-VIIa complex on the tumor cell surface. In primary bladder carcinoma cells, we find that this complex localizes to the invasive edge, in proximity to tumor-infiltrating vessels that stain intensely for TF pathway inhibitor (TFPI-1), the major inhibitor of the protease activity of the complex. In culture, binding of VIIa to TF-expressing tumor cells is sufficient to allow cell adhesion, migration, and intracellular signaling on immobilized TFPI-1. Immobilized heparin, a mimic for extracellular matrix-associated proteoglycans, binds physiological concentrations of TFPI-1 in a conformation that supports TF-VIIa-dependent cell adhesion. Consistent with a functional role of TFPI-1 in complex extracellular matrices, we show that TF cooperates with integrin-mediated adhesion and migration on composite matrices that contain ligands for both integrins and the TF-VIIa complex. This study thus provides evidence for a novel mechanism of protease-supported migration that is independent of proteolytic matrix degradation but rather involves protease-dependent bridging of TF's extracellular domain to an ECM-associated inhibitor.


Assuntos
Carcinoma/metabolismo , Proteínas de Neoplasias , Tromboplastina/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Carcinoma/patologia , Adesão Celular , Movimento Celular , Cisteína Endopeptidases/metabolismo , Relação Dose-Resposta a Droga , Endopeptidases/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Fator VIIa/metabolismo , Fibronectinas/farmacologia , Glicoproteínas/metabolismo , Heparina/farmacologia , Humanos , Imuno-Histoquímica , Lipoproteínas/metabolismo , Lipoproteínas/farmacologia , Proteínas da Gravidez/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/patologia
12.
Cancer Res ; 55(8): 1629-32, 1995 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-7712465

RESUMO

The human melanoma cell line M24met expresses tissue factor, the cellular initiator of the blood coagulation cascade. Blocking of the coagulation pathways at the level of tissue factor, factor Xa, or thrombin inhibits hematogenous M24met metastasis in SCID mice, implicating a role for thrombin generation in this process. Dependent on cell surface tissue factor activity, M24met cells generate thrombin in vitro. Thrombin and the thrombin receptor agonist peptide TRP-14 activate a signaling pathway in M24met cells that involves an increase in intracellular calcium and induces cell proliferation. Immunofluorescence evidences expression of the signaling thrombin receptor on these cells. Thus, M24met melanoma cells express both the initiating cell surface receptor for the coagulation pathways and the central signaling receptor of the coagulation system, suggesting the in situ generation of proliferative signals which can contribute to the malignant phenotype.


Assuntos
Melanoma/metabolismo , Receptores de Trombina/metabolismo , Trombina/biossíntese , Tromboplastina/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Cálcio/metabolismo , Divisão Celular , Linhagem Celular , Citosol/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Cinética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma/patologia , Melanoma/secundário , Camundongos , Camundongos SCID , Fragmentos de Peptídeos/farmacologia , Receptores de Trombina/agonistas , Receptores de Trombina/biossíntese , Transdução de Sinais/efeitos dos fármacos , Trombina/farmacologia , Tromboplastina/imunologia , Transplante Heterólogo , Células Tumorais Cultivadas
13.
Thromb Res ; 140 Suppl 1: S172, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27161681

RESUMO

INTRODUCTION: Tissue Factor (TF) promotes apoptosis-resistance and facilitates haematogenous metastasis. Cancer cells release TF-bearing microparticles (TF-MP) which have pro-coagulant activity. TF-MPs from cancer cells induce increase in TF expression and procoagulant activity of recipient endothelial cells. Drug resistance microparticles transfer between cancer cells, resulting in induction of recipient cell drug resistance, however transfer of TF-MPs between cancer cells has not been demonstrated. AIM: To determine if microparticles isolated from TF expressing cancer cell lines can induced TF-mediated procoagulant activity in cell lines that do not express TF. MATERIALS AND METHODS: A7 cells (a melanoma cell line that does not express TF) were transformed to express TF with adenoviral transformation. In addition, the TF-expressing breast cancer cell line MDAMB231 was used. Media was collected from (1) wild-type A7 (A7-WT) cells, which acted as control (2) MDAMB231 cells, (3) A7 cells transformed with 'low' levels of TF-adenovirus and (4) A7 cells transformed with 'high' levels of TF-adenovirus. MPs were isolated from media by centrifugation using standard techniques. Isolated MPs were incubated with wild-type A7 (A7-WT) cells overnight. Pro-coagulant activity of A7-WT cells was assessed using a FXa generation assay. RESULTS: i) A7-WT cells incubated overnight with MDAMB231-derived MPs produced an almost 5 fold increase in procoagulant activity compared to control (Mean 3.5 mOD/min S.E 0.32 vs control 0.68 S.E. 0.05, p<0.01). ii) MPs released from A7-TF cells induced a dose-dependant increase in cell based procoagulant activity when incubated with A7-WTs compared to control:A7-WT cells incubated with control MPs (Control): mean mOD 0.83, S.E. 0.07A7-WT cells incubated with A7-TF derived MPs (low levels of TF-adenovirus): mean mOD 1.27, S.E. 0.06 (p=0.04 vs control)A7-WT cells incubated with A7-TF derived MPs (low levels of TF-adenovirus): mean mOD 2.77, S.E. 0.23 (p<0.01 vs control). CONCLUSIONS: These results demonstrate, for the first time, microparticle-mediated transfer of TF procoagulant activity between cancer cells. MPs shed by cancer cells in vivo have been implicated in cancer cell progression and hypercoagulability. The acquisition and spread of of MP-mediated TF activity has implications for cancer cell biology and cancer-induced hypercoagulability.

14.
Eur J Pain ; 20(2): 263-73, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25929250

RESUMO

BACKGROUND: Until now, nonivamide/nicoboxil ointment has not been tested in a randomized trial for the treatment of acute non-specific low back pain. METHODS: This phase III randomized, double-blind, active- and placebo-controlled, multi-centre trial investigated efficacy, safety and tolerability of topical nicoboxil 2.5%/nonivamide 0.4% for treatment of acute non-specific low back pain [primary endpoint: pain intensity (PI) difference between pre-dose baseline and 8 h after the first application]. RESULTS: Patients (n = 805), 18-74 years of age were treated for up to 4 days with nicoboxil 2.5%/nonivamide 0.4%, nicoboxil 2.5%, nonivamide 0.4% or placebo ointment. Pre-dose baseline pain intensity (6.6 on a 0- to 10-point numerical rating scale) was reduced by 1.049 points with placebo, by 1.428 points with nicoboxil, by 2.252 points with nonivamide and by 2.410 points with nicoboxil/nonivamide after 8 h (p < 0.0001 for nicoboxil/nonivamide vs. placebo, nicoboxil; p = 0.4171 for nicoboxil/nonivamide vs. nonivamide). At the end of treatment, the combination provided more pronounced PI reduction (3.540 points) compared with nicoboxil (2.371, p < 0.0001), nonivamide (3.074, p = 0.0259) and placebo (1.884, p < 0.0001). Low back mobility scores on Day 1 were better for the combination compared with all other treatments (p < 0.044); on Day 2-4, scores were better than for placebo and nicoboxil (p < 0.003). Patients assessed efficacy of the combination as greater than of the comparators (p ≤ 0.0129). All treatments were tolerated well. No treatment-related serious adverse events were reported. CONCLUSION: Nicoboxil/nonivamide ointment is an effective, well-tolerated medication for the treatment of acute non-specific low back pain.


Assuntos
Dor Aguda/tratamento farmacológico , Capsaicina/análogos & derivados , Dor Lombar/tratamento farmacológico , Ácidos Nicotínicos/uso terapêutico , Adolescente , Adulto , Idoso , Capsaicina/efeitos adversos , Capsaicina/uso terapêutico , Método Duplo-Cego , Combinação de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ácidos Nicotínicos/efeitos adversos , Resultado do Tratamento , Adulto Jovem
15.
Biochim Biophys Acta ; 965(2-3): 169-75, 1988 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-3365451

RESUMO

Thrombin preferentially cleaves fibrinopeptides A (FPA) from fibrinogen resulting in the formation of desAA-fibrin from which most of the fibrinopeptides B (FPB) are then released with an enhanced rate. Kinetics of fibrinopeptide release from normal and dysfunctional fibrinogens were investigated in order to further characterize the mechanism of accelerated FPB release during desAA-fibrin polymerization. Dysfunctional fibrinogens London I and Ashford, exhibiting primary polymerization abnormalities (i.e., an abnormality present when all fibrinopeptides have been cleaved), which in the case of fibrinogen London I is believed to be caused by a defect in the D-domain, were shown to exhibit a decreased rate of FPB release compared with normal fibrinogen. While Gly-Pro-Arg-Pro, an inhibitor of fibrin polymerization, was shown to decrease the rate of FPB release from normal fibrinogen by a factor of 5, normal fragment D1, although inhibiting clot formation of normal fibrinogen, did not influence the acceleration of FPB release. On the other hand, the presence of fragment D1 did not enhance FPB release from fibrinogen London I, suggesting that interaction of D-domains in functional isolation with desAA-fibrin E-domains is not sufficient to enhance FPB release. Although clot formation was inhibited by the concentrations of fragment D1 used, the formation of small desAA-fibrin oligomers was hardly affected. Thus, small fibrin polymers, but not desAA-fibrin monomers, act as optimal substrates for the release of FPB by thrombin.


Assuntos
Fibrina/antagonistas & inibidores , Fibrinogênio/metabolismo , Fibrinogênios Anormais , Fibrinopeptídeo B/metabolismo , Trombina/metabolismo , Transtornos da Coagulação Sanguínea , Fibrina/metabolismo , Humanos , Cinética , Nefelometria e Turbidimetria , Polímeros
16.
J Am Coll Cardiol ; 7(5): 1057-62, 1986 May.
Artigo em Inglês | MEDLINE | ID: mdl-3958361

RESUMO

The effects of aspirin on myocardial blood flow in an area of ischemia were studied in 12 baboons. In each, a diagonal branch of the left anterior descending coronary artery was ligated. Six of the baboons received aspirin (2 X 600 mg orally, 12 hours and 1 hour before ligation); the other six did not receive aspirin and served as a control group. The extent of myocardial ischemia was delineated with an electrode wire grid on the surface of the anterior left ventricular wall. The maximal area circumscribed by electrodes with 2 mV or more ST segment elevation was compared with the area of reduced myocardial blood flow. Myocardial blood flow was measured with the radioactive microspheres method using strontium-85-labeled carbonized spheres. Two areas of reduced myocardial blood flow were noted, one with severely reduced flow in the center of the myocardial infarct (0 to 49% of noninfarcted myocardium) and another with mild to moderately reduced myocardial blood flow at the border of the myocardial infarct (50 to 90% of noninfarcted myocardium). Myocardial blood flow in the border area (margins of ST elevation area) for the total wall was 85 +/- 8% of normal in the aspirin-treated animals and 40 +/- 4% in the control group (p less than 0.01); for the epicardium it was 67 +/- 10% of normal in noninfarcted myocardium after aspirin and 37 +/- 5% for the control group (p less than 0.05); and for the endocardium it was 78 +/- 8% of normal in noninfarcted myocardium after aspirin and 39 +/- 6% in the control group (p less than 0.01).(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Aspirina/farmacologia , Circulação Coronária/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Animais , Aspirina/uso terapêutico , Eletrocardiografia , Coração/fisiologia , Papio
17.
J Mol Biol ; 277(4): 959-71, 1998 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-9545384

RESUMO

The catalytic activity of the trypsin-like serine protease coagulation factor VIIa is allosterically regulated. In this work, we employed monoclonal antibodies as probes to analyze conformational changes in the VII protease domain that are induced by zymogen activation, cofactor tissue factor (TF) binding, and active site occupancy. The epitopes of three monoclonal antibodies were mapped using a panel of 57 individual alanine replacement mutants in the protease domain. Two of the antibodies had typical "hot spot" epitopes in a basic cluster above the active site cleft and antibody binding to these epitopes was not affected by zymogen activation, TF binding, or active site occupancy. In contrast, the binding kinetics of VII/VIIa to a monoclonal antibody that mapped to an extended epitope overlapping with the macromolecular substrate exosite was affected by each of the conformational transitions of the VIIa protease domain. The changes in antibody affinity are consistent with a transition from zymogen VII to the TF.VIIa complex, with free enzyme VIIa as an intermediate that retains some zymogen-like features responsible for its low catalytic activity. In contrast, active site occupancy resulted in effects that were qualitatively different from the effects of zymogen activation on the antibody epitope. This provides novel insight into the conformational interdependence between the active site, the region for macromolecular substrate recognition, and the cofactor binding exosite of this allosterically regulated serine protease.


Assuntos
Fator VIIa/química , Fator VIIa/metabolismo , Regulação Alostérica , Animais , Anticorpos Monoclonais , Sítios de Ligação , Técnicas Biossensoriais , Ativação Enzimática , Mapeamento de Epitopos , Fator VII/química , Fator VII/metabolismo , Fator VIIa/genética , Humanos , Técnicas In Vitro , Substâncias Macromoleculares , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tromboplastina/química , Tromboplastina/genética , Tromboplastina/metabolismo
18.
J Mol Biol ; 275(5): 873-94, 1998 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-9480775

RESUMO

The tissue factor (TF)-initiated blood coagulation protease cascade can be greatly inhibited in vivo by a potent anti-human-TF monoclonal antibody, 5G9. This antibody binds the carboxyl module of the extracellular domain of TF with a nanomolar binding constant and inhibits the formation of the TF.VIIa.X ternary initiation complex. We have determined the crystal structures of the extra-cellular modules of human TF, Fab 5G9, and their complex (TF.5G9) to 2.4 A, 2. 5 A, and 3.0 A, respectively, and measured the apparent inhibition constants of 5G9 on a panel of TF mutants. In our unliganded TF structure, a 7 degrees change in the relative orientation between the D1 and D2 modules was observed when compared with other published TF structures. Comparison of the free and bound Fab 5G9 indicates that small segmental and side chain variation of the antibody complementarity determining regions occurred on complexation with TF. The antibody-antigen recognition involves 18 TF antigen residues and 19 Fab residues from six CDR with one of the largest buried surface areas seen to date. A combination of structural and mutagenesis data indicate that Tyr156, Lys169, Arg200, and Lys201 play the major role in the antibody recognition. The TF. 5G9 structure provides insights into the mechanism by which the antibody 5G9 inhibits formation of the TF.VIIa.X ternary complex.


Assuntos
Complexo Antígeno-Anticorpo/química , Fragmentos Fab das Imunoglobulinas/química , Conformação Proteica , Tromboplastina/química , Animais , Coagulação Sanguínea/imunologia , Cricetinae , Cricetulus , Cristalografia por Raios X , Humanos , Modelos Moleculares , Mutagênese
19.
Trends Cardiovasc Med ; 8(8): 350-6, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14987549

RESUMO

The integration of structure and function analysis of the tissue factor-factor VIIa complex has provided a detailed view of the functional surface of the extrinsic activation complex. An incomplete zymogen to enzyme transition is responsible for the strict cofactor dependence of catalytic function of factor VIIa. The mutational analysis demonstrates that factor VIIa is allosterically regulated by specific conformational linkages that involve the cofactor binding site, the catalytic cleft, and the macromolecular substrate exosite. Regions of the flexible activation domain appear to play an important role in the allosteric regulation of this cofactor-dependent coagulation serine protease.

20.
Cardiovasc Res ; 14(7): 408-18, 1980 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7459927

RESUMO

Despite numerous studies directed at determining the ability of glucocorticoids to minimise myocardial ischaemic damage following acute coronary occlusion, there remains no clear consensus concerning their usefulness. Within an ischaemic region, glucocorticoids produce membrane stabilising effects which decrease the autolytic effects of marked cellular swelling and lysosomal membrane rupture. Their use has also been associated with a decreased cellular lysis due to infiltrating inflammatory cells and with an increase in collateral blood flow. The use of glucocorticoids has remained clinically attractive due to experimental observations regarding these local actions; however, the potential of these actions to enhance long-term viability of ischaemic myocardium has remained uncertain. A major problem in the comparison of the extent of infarction between treated and untreated animals has been the variability in infarct size that results from coronary artery ligation at any given anatomical site. In the experimental baboon model which we have employed, we have previously shown that the ultimate epicardial area of infarction, as well as the volume of infarction as assessed histologically at 7 days post occlusion, shows a good linear relationship to the area of ischaemic injury at 1-hour post occlusion, as assessed by high resolution epicardial ST segment mapping. In this way animals may serve as their own controls and as long as an intervention is initiated at 1 h or later post occlusion, then the epicardial area or transmural volume of histologically assessed infarction at 7 days can be compared with the predicted epicardial area or transmural volume of infarction and hence determine the effect of that intervention in altering infarct size. This experimental model does not rely on a comparison of the absolute magnitude of infarcts between treated and untreated animals and, therefore, avoids the error introduced by the inherent variability in infarct size between animals after coronary occlusion.


Assuntos
Metilprednisolona/uso terapêutico , Infarto do Miocárdio/prevenção & controle , Animais , Relação Dose-Resposta a Droga , Eletrocardiografia , Metilprednisolona/administração & dosagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Papio , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa