Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Sci Rep ; 13(1): 5764, 2023 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-37031266

RESUMO

Defective regulation of the alternative complement pathway (AP) causes excessive activation and promotes the inflammation and renal injury observed in atypical hemolytic-uremic syndrome (aHUS). The usefulness of heat-inactivated Factor B (HFB) in reducing AP activation was evaluated in: fluid-phase reactions, using purified complement proteins and Factor H (FH)-depleted serum; and in surface-activated reactions using human endothelial cells (ECs). C3a and Ba levels, measured by quantitative Western blots, determined the extent of fluid-phase activation. In reactions using C3, FB, and Factor D proteins, HFB addition (2.5-fold FB levels), reduced C3a levels by 60% and Ba levels by 45%. In reactions using FH-depleted serum (supplemented with FH at 12.5% normal levels), Ba levels were reduced by 40% with HFB added at 3.5-fold FB levels. The effectiveness of HFB in limiting AP convertase formation on activated surfaces was evaluated using stimulated ECs. Fluorescent microscopy was used to quantify endogenously released C3, FB, and C5 attached to EC-secreted ultra-large VWF strings. HFB addition reduced attachment of C3b by 2.7-fold, FB by 1.5-fold and C5 by fourfold. Our data indicate that HFB may be of therapeutic value in preventing AP-mediated generation of C3a and C5a, and the associated inflammation caused by an overactive AP.


Assuntos
Fator B do Complemento , Fator de von Willebrand , Humanos , Fator B do Complemento/metabolismo , Fator de von Willebrand/metabolismo , Células Endoteliais/metabolismo , Temperatura Alta , Fator H do Complemento/metabolismo , Inflamação/metabolismo , Ativação do Complemento , Complemento C3b/metabolismo
2.
Sci Rep ; 11(1): 21852, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750441

RESUMO

In a previous study, we reported that human endothelial cells (ECs) express and produce their own coagulation factors (F) that can activate cell surface FX without the additions of external proteins or phospholipids. We now describe experiments that detail the expression and production in ECs and fibroblasts of the clotting proteins necessary for formation of active prothrombinase (FV-FX) complexes to produce thrombin on EC and fibroblast surfaces. EC and fibroblast thrombin generation was identified by measuring: thrombin activity; thrombin-antithrombin complexes; and the prothrombin fragment 1.2 (PF1.2), which is produced by the prothrombinase cleavage of prothrombin (FII) to thrombin. In ECs, the prothrombinase complex uses surface-attached FV and γ-carboxyl-glutamate residues of FX and FII to attach to EC surfaces. FV is also on fibroblast surfaces; however, lower fibroblast expression of the gene for γ-glutamyl carboxylase (GGCX) results in production of vitamin K-dependent coagulation proteins (FII and FX) with reduced surface binding. This is evident by the minimal surface binding of PF1.2, following FII activation, of fibroblasts compared to ECs. We conclude that human ECs and fibroblasts both generate thrombin without exogenous addition of coagulation proteins or phospholipids. The two cell types assemble distinct forms of prothrombinase to generate thrombin.


Assuntos
Fatores de Coagulação Sanguínea/biossíntese , Fatores de Coagulação Sanguínea/genética , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Trombina/biossíntese , Antitrombina III/genética , Antitrombina III/metabolismo , Carboidratos Epimerases/biossíntese , Carboxiliases/genética , Linhagem Celular , Fator V/genética , Fator V/metabolismo , Fator Xa/metabolismo , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Cetona Oxirredutases/biossíntese , Modelos Biológicos , Fragmentos de Peptídeos/metabolismo , Proteólise , Protrombina/biossíntese , Protrombina/genética , Protrombina/metabolismo , Trombomodulina/genética , Trombomodulina/metabolismo , Vitamina K Epóxido Redutases/genética
3.
Sci Rep ; 10(1): 2005, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029851

RESUMO

Human endothelial cells (ECs) synthesize, store, and secrete von Willebrand factor multimeric strings and coagulation factor (F) VIII. It is not currently known if ECs produce other coagulation factors for active participation in coagulation. We found that 3 different types of human ECs in primary culture produce clotting factors necessary for FX activation via the intrinsic (FVIII-FIX) and extrinsic (tissue factor [TF]-FVII) coagulation pathways, as well as prothrombin. Human dermal fibroblasts were used as comparator cells. TF, FVII, FIX, FX, and prothrombin were detected in ECs, and TF, FVII, FIX, and FX were detected in fibroblasts. In addition, FVII, FIX, FX, and prothrombin were detected by fluorescent microscopy in EC cytoplasm (associated with endoplasmic reticulum and Golgi proteins). FX activation occurred on human umbilical vein EC surfaces without the addition of external coagulation proteins, proteolytic enzymes, or phospholipids. Tumour necrosis factor, which suppresses the generation of activated protein C and increases TF, augmented FX activation. Fibroblasts also produced TF, but (in contrast to ECs) were incapable of activating FX without the exogenous addition of FX and had a marked increase in FX activation following the addition of both FX and FVII. We conclude that human ECs produce their own coagulation factors that can activate cell surface FX without the addition of exogenous proteins or phospholipids.


Assuntos
Coagulação Sanguínea , Fator X/metabolismo , Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Linhagem Celular , Citoplasma/metabolismo , Fator IX/análise , Fator IX/metabolismo , Fator VII/análise , Fator VII/metabolismo , Fator VIII/análise , Fator VIII/metabolismo , Fibroblastos/citologia , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Microscopia de Fluorescência , Cultura Primária de Células , Protrombina/análise , Protrombina/metabolismo , Pele/citologia , Tromboplastina/análise , Tromboplastina/metabolismo
4.
J Phys Chem B ; 122(47): 10653-10658, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30351116

RESUMO

Activation of proteins often involves conformational transitions, and these switches are often difficult to characterize in multidomain proteins. Full-length factor H (FH), consisting of 20 small consensus repeat domains (150 kD), is a complement control protein that regulates the activity of the alternative complement pathway. Different preparations of FH can also reduce the disulfide bonds linking large Von Willebrand factor (VWF) multimers into smaller, less adhesive forms. In contrast, commercially available purified FH (pFH) has little or no VWF reductase activity unless the pFH is chemically modified by either ethylenediaminetetraacetic acid (EDTA) or urea. We used atomic force microscopy single molecule force measurements to investigate different forms of FH, including recombinant FH and pFH, in the presence or absence of EDTA and urea, and to correlate the conformational changes to its activities. We found that the FH conformation depends on the method used for sample preparation, which affects the VWF reductase activity of FH.


Assuntos
Oxirredutases/química , Fator de von Willebrand/química , Catálise , Fator H do Complemento/química , Detergentes/química , Células HEK293 , Humanos , Microscopia de Força Atômica/métodos , Octoxinol/química , Organofosfatos/química , Oxirredução , Conformação Proteica , Domínios Proteicos , Proteínas Recombinantes/química , Termodinâmica
5.
J Biol Chem ; 293(19): 7195-7208, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29555686

RESUMO

Atypical hemolytic uremic syndrome (aHUS) and bone marrow transplantation-associated thrombotic microangiopathy (TA-TMA) are associated with excessive activation of the alternative complement pathway (AP) and with severe renal, but rarely cerebral, microvascular damage. Here, we compared AP activation and regulation in human glomerular and brain microvascular endothelial cells (GMVECs and BMVECs, respectively) unstimulated or stimulated by the proinflammatory cytokine, tumor necrosis factor (TNF). Compared with GMVECs and under both experimental conditions, BMVECs had increased gene expression of the AP-related genes C3, CFB, and C5 and decreased expression of CFD This was associated with increased expression in BMVECs (relative to GMVECs) of the genes for surface and soluble regulatory molecules (CD46, THBD, CD55, CFI, and CFH) suppressing formation of the AP C3 and C5 convertases. Of note, unlike GMVECs, BMVECs generated extremely low levels of C3a and C5a and displayed decreased activation of the AP (as measured by a lower percentage of Ba generation than GMVECs). Moreover, BMVECs exhibited increased function of CD141, mediating activation of the natural anticoagulant protein C, compared with GMVECs. We also found that the C3a receptor (C3aR) is present on both cell types and that TNF greatly increases C3AR1 expression in GMVECs, but only slightly in BMVECs. Higher AP activation and C3a generation in GMVECs than in BMVECs, coupled with an increase in C3aR production in TNF-stimulated GMVECs, provides a possible explanation for the predominance of renal damage, and the absence of cerebral injury, in individuals with episodes of aHUS and TA-TMA.


Assuntos
Encéfalo/irrigação sanguínea , Ativação do Complemento , Via Alternativa do Complemento/fisiologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Glomérulos Renais/metabolismo , Microvasos/metabolismo , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Via Alternativa do Complemento/genética , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Expressão Gênica , Humanos , Inflamação/metabolismo , Glomérulos Renais/citologia , Microvasos/citologia , Microvasos/efeitos dos fármacos , Modelos Biológicos , Proteínas Musculares/genética , Receptor da Anafilatoxina C5a/genética , Receptores de Complemento/genética , Microangiopatias Trombóticas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de von Willebrand/metabolismo
6.
TH Open ; 2(3): e338-e345, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31080944

RESUMO

von Willebrand factor (VWF) is a multimeric protein composed of monomeric subunits (~280 kD) linked by disulfide bonds. During hemostasis and thrombosis, ultralarge (UL) VWF (ULVWF) multimers initiate platelet adhesion. In vitro, human C3 binds to ULVWF multimeric strings secreted by and anchored to human endothelial cell to promote the assembly and activation of C3 convertase (C3bBb) and C5 convertase (C3bBbC3b) of the alternative complement pathway (AP). The purified and soluble C3 avidly binds to recombinant human VWF A1A2A3, as well as the recombinant isolated human VWF A3 domain. Notably, the binding of soluble human ULVWF multimers to purified human C3 was blocked by addition of a monovalent Fab fragment antibody to the VWF A3 domain. We conclude that the A3 domain in VWF/ULVWF contains a docking site for C3. In contrast, purified human C4, an essential component of the classical and lectin complement pathways, binds to soluble, isolated A1, but not to ULVWF strings secreted by and anchored to endothelial cells. Our findings should facilitate the design of new therapeutic agents to suppress the initiation of the AP on ULVWF multimeric strings during thrombotic and inflammatory disorders.

7.
Blood ; 130(3): 243-244, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28729338
8.
ASAIO J ; 63(6): 849-853, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28682993

RESUMO

Patients supported by left ventricular assist devices (LVADs) often present with the loss of large von Willebrand factor (VWF) multimers. This VWF deficiency is believed to contribute to the bleeding diathesis of patients on LVAD support and is caused by excessive VWF cleavage by the metalloprotease ADAMTS-13 under high shear stress. However, only a small percentage of patients who have suffered the loss of large VWF multimers bleed. The actual rates of VWF cleavage in these patients have not been reported, primarily because of the lack of reliable detection methods. We have developed and validated a selected reaction monitoring (SRM) mass spectrometry method to quantify VWF cleavage as the ratio of the ADAMTS-13-cleaved peptide MVTGNPASDEIK to the ILAGPAGDSNVVK peptide. The rate of VWF cleavage was found to be 1.26% ± 0.36% in normal plasma. It varied significantly in patient samples, ranging from 0.23% to 2.5% of total VWF antigen, even though all patients had the loss of large VWF multimers. Von Willebrand factor cleavage was greater in post-LVAD samples from patients in whom bleeding had developed, but was mostly reduced in patients in whom thrombosis had developed. This SRM method is reliable to quantify the rate of VWF cleavage in patients on LVAD support.


Assuntos
Proteína ADAMTS13/metabolismo , Coração Auxiliar/efeitos adversos , Fator de von Willebrand/metabolismo , Adulto , Idoso , Feminino , Hemorragia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Doenças de von Willebrand/etiologia
9.
Nat Rev Dis Primers ; 3: 17020, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28382967

RESUMO

Thrombotic thrombocytopenic purpura (TTP; also known as Moschcowitz disease) is characterized by the concomitant occurrence of often severe thrombocytopenia, microangiopathic haemolytic anaemia and a variable degree of ischaemic organ damage, particularly affecting the brain, heart and kidneys. Acute TTP was almost universally fatal until the introduction of plasma therapy, which improved survival from <10% to 80-90%. However, patients who survive an acute episode are at high risk of relapse and of long-term morbidity. A timely diagnosis is vital but challenging, as TTP shares symptoms and clinical presentation with numerous conditions, including, for example, haemolytic uraemic syndrome and other thrombotic microangiopathies. The underlying pathophysiology is a severe deficiency of the activity of a disintegrin and metalloproteinase with thrombospondin motifs 13 (ADAMTS13), the protease that cleaves von Willebrand factor (vWF) multimeric strings. Ultra-large vWF strings remain uncleaved after endothelial cell secretion and anchorage, bind to platelets and form microthrombi, leading to the clinical manifestations of TTP. Congenital TTP (Upshaw-Schulman syndrome) is the result of homozygous or compound heterozygous mutations in ADAMTS13, whereas acquired TTP is an autoimmune disorder caused by circulating anti-ADAMTS13 autoantibodies, which inhibit the enzyme or increase its clearance. Consequently, immunosuppressive drugs, such as corticosteroids and often rituximab, supplement plasma exchange therapy in patients with acquired TTP.


Assuntos
Púrpura Trombocitopênica Trombótica/complicações , Púrpura Trombocitopênica Trombótica/diagnóstico , Proteína ADAMTS13/análise , Proteína ADAMTS13/sangue , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Feminino , Glucocorticoides/farmacologia , Glucocorticoides/uso terapêutico , Infecções por HIV/etiologia , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Imunomodulação , Gravidez , Complicações na Gravidez/etiologia , Púrpura Trombocitopênica Trombótica/fisiopatologia , Rituximab/farmacologia , Rituximab/uso terapêutico , Toxinas Shiga/efeitos adversos , Esplenectomia/métodos
11.
Blood ; 127(25): 3133-41, 2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27143258

RESUMO

Left ventricular assist devices (LVAD) provide cardiac support for patients with end-stage heart disease as either bridge or destination therapy, and have significantly improved the survival of these patients. Whereas earlier models were designed to mimic the human heart by producing a pulsatile flow in parallel with the patient's heart, newer devices, which are smaller and more durable, provide continuous blood flow along an axial path using an internal rotor in the blood. However, device-related hemostatic complications remain common and have negatively affected patients' recovery and quality of life. In most patients, the von Willebrand factor (VWF) rapidly loses large multimers and binds poorly to platelets and subendothelial collagen upon LVAD implantation, leading to the term acquired von Willebrand syndrome (AVWS). These changes in VWF structure and adhesive activity recover quickly upon LVAD explantation and are not observed in patients with heart transplant. The VWF defects are believed to be caused by excessive cleavage of large VWF multimers by the metalloprotease ADAMTS-13 in an LVAD-driven circulation. However, evidence that this mechanism could be the primary cause for the loss of large VWF multimers and LVAD-associated bleeding remains circumstantial. This review discusses changes in VWF reactivity found in patients on LVAD support. It specifically focuses on impacts of LVAD-related mechanical stress on VWF structural stability and adhesive reactivity in exploring multiple causes of AVWS and LVAD-associated hemostatic complications.


Assuntos
Coração Auxiliar/efeitos adversos , Disfunção Ventricular Esquerda/cirurgia , Doenças de von Willebrand/etiologia , Insuficiência Cardíaca/cirurgia , Transplante de Coração/efeitos adversos , Transplante de Coração/instrumentação , Hemostasia/fisiologia , Humanos , Trombose/etiologia , Disfunção Ventricular Esquerda/fisiopatologia , Fator de von Willebrand/fisiologia
12.
Phys Rev E ; 93(1): 012410, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26871104

RESUMO

Von Willebrand factor (VWF) multimers are large adhesive proteins that are essential to the initiation of hemostatic plugs at sites of vascular injury. The binding of VWF multimers to platelets, as well as VWF proteolysis, is regulated by shear stresses that alter VWF multimeric conformation. We used single molecule manipulation with atomic force microscopy (AFM) to investigate the effect of high fluid shear stress on soluble dimeric and multimeric forms of VWF. VWF dimers are the smallest unit that polymerizes to construct large VWF multimers. The resistance to mechanical unfolding with or without exposure to shear stress was used to evaluate VWF conformational forms. Our data indicate that, unlike recombinant VWF multimers (RVWF), recombinant dimeric VWF (RDVWF) unfolding force is not altered by high shear stress (100dynes/cm^{2} for 3 min at 37^{∘}C). We conclude that under the shear conditions used (100dynes/cm^{2} for 3 min at 37^{∘}C), VWF dimers do not self-associate into a conformation analogous to that attained by sheared large VWF multimers.


Assuntos
Desdobramento de Proteína , Fator de von Willebrand/química , Células HEK293 , Humanos , Microscopia de Força Atômica , Modelos Moleculares , Polimerização , Multimerização Proteica , Proteínas Recombinantes/química , Estresse Mecânico
13.
J Immunol ; 196(2): 832-45, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26673143

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy with severe renal injury secondary to an overactive alternative complement pathway (AP). aHUS episodes are often initiated or recur during inflammation. We investigated gene expression of the surface complement regulatory proteins (CD55, CD59, CD46, and CD141 [thrombomodulin]) and AP components in human glomerular microvascular endothelial cells (GMVECs) and in HUVECs, a frequently used investigational model of endothelial cells. Surface complement regulatory proteins were also quantified by flow cytometry. All experiments were done with and without exposure to IL-1ß or TNF. Without cytokine stimulation, we found that GMVECs had greater AP activation than did HUVECs. With TNF stimulation, THBD gene expression and corresponding CD141 surface presence in HUVECs and GMVECs were reduced, and gene expression of complement components C3 (C3) and factor B (CFB) was increased. Consequently, AP activation, measured by Ba production, was increased, and conversion of protein C (PC) to activated PC by CD141-bound thrombin was decreased, in GMVECs and HUVECs exposed to TNF. IL-1ß had similar, albeit lesser, effects on HUVEC gene expression, and it only slightly affected GMVEC gene expression. To our knowledge, this is the first detailed study of the expression/display of AP components and surface regulatory proteins in GMVECs with and without cytokine stimulation. In aHUS patients with an underlying overactive AP, additional stimulation of the AP and inhibition of activated PC-mediated anticoagulation in GMVECs by the inflammatory cytokine TNF are likely to provoke episodes of renal failure.


Assuntos
Via Alternativa do Complemento/imunologia , Células Endoteliais/imunologia , Glomérulos Renais/imunologia , Proteína C/metabolismo , Antígenos CD/análise , Antígenos CD/biossíntese , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Células Endoteliais/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Imunoensaio , Glomérulos Renais/metabolismo , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
14.
PLoS One ; 10(10): e0140740, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26473492

RESUMO

The cellular synthesis site and ensuing storage location for human factor VIII (FVIII), the coagulation protein deficient in hemophilia A, has been elusive. FVIII stability and half-life is dependent on non-covalent complex formation with von Willebrand factor (VWF) to avoid proteolysis and clearance. VWF is synthesized in megakaryocytes and endothelial cells, and is stored and secreted from platelet alpha granules and Weibel-Palade bodies of endothelial cells. In this paper we provide direct evidence for FVIII synthesis in 2 types of primary human endothelial cells: glomerular microvascular endothelial cells (GMVECs) and umbilical vein endothelial cells (HUVECs). Gene expression quantified by real time PCR revealed that levels of F8 and VWF are similar in GMVECs and HUVECs. Previous clinical studies have shown that stimulation of vasopressin V2 receptors causes parallel secretion of both proteins. In this study, we found that both endothelial cell types express AVPR2 (vasopressin V2 receptor gene) and that AVPR2 mRNA levels are 5-fold higher in GMVECs than HUVECs. FVIII and VWF proteins were detected by fluorescent microscopy in Weibel-Palade bodies within GMVECs and HUVECs using antibodies proven to be target specific. Visual presence of FVIII and VWF in Weibel-Palade bodies was confirmed by correlation measurements. The high extent of correlation was compared with negative correlation values obtained from FVIII detection with cytoplasmic proteins, ß-actin and Factor H. FVIII activity was positive in GMVEC and HUVEC cell lysates. Stimulated GMVECs and HUVECs were found to secrete cell-anchored ultra-large VWF strings covered with bound FVIII.


Assuntos
Fator VIII/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Complexos Multiproteicos/metabolismo , Corpos de Weibel-Palade/metabolismo , Fator de von Willebrand/metabolismo , Regulação da Expressão Gênica/fisiologia , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Complexos Multiproteicos/ultraestrutura , Receptores de Vasopressinas/biossíntese , Corpos de Weibel-Palade/ultraestrutura
15.
PLoS One ; 10(6): e0130749, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26090873

RESUMO

Valve endothelial cells (VEC) have unique phenotypic responses relative to other types of vascular endothelial cells and have highly sensitive hemostatic functions affected by changes in valve tissues. Furthermore, effects of environmental factors on VEC hemostatic function has not been characterized. This work used a poly(ethylene glycol) diacrylate (PEGDA) hydrogel platform to evaluate the effects of substrate stiffness and cell adhesive ligands on VEC phenotype and expression of hemostatic genes. Hydrogels of molecular weights (MWs) 3.4, 8, and 20 kDa were polymerized into platforms of different rigidities and thiol-modified cell adhesive peptides were covalently bound to acrylate groups on the hydrogel surfaces. The peptide RKRLQVQLSIRT (RKR) is a syndecan-1 binding ligand derived from laminin, a trimeric protein and a basement membrane matrix component. Conversely, RGDS is an integrin binding peptide found in many extracellular matrix (ECM) proteins including fibronectin, fibrinogen, and von Willebrand factor (VWF). VECs adhered to and formed a stable monolayer on all RKR-coated hydrogel-MW combinations. RGDS-coated platforms supported VEC adhesion and growth on RGDS-3.4 kDa and RGDS-8 kDa hydrogels. VECs cultured on the softer RKR-8 kDa and RKR-20 kDa hydrogel platforms had significantly higher gene expression for all anti-thrombotic (ADAMTS-13, tissue factor pathway inhibitor, and tissue plasminogen activator) and thrombotic (VWF, tissue factor, and P-selectin) proteins than VECs cultured on RGDS-coated hydrogels and tissue culture polystyrene controls. Stimulated VECs promoted greater platelet adhesion than non-stimulated VECs on their respective culture condition; yet stimulated VECs on RGDS-3.4 kDa gels were not as responsive to stimulation relative to the RKR-gel groups. Thus, the syndecan binding, laminin-derived peptide promoted stable VEC adhesion on the softer hydrogels and maintained VEC phenotype and natural hemostatic function. In conclusion, utilization of non-integrin adhesive peptide sequences derived from basement membrane ECM may recapitulate balanced VEC function and may benefit endothelialization of valve implants.


Assuntos
Células Endoteliais/metabolismo , Hidrogéis/química , Peptídeos/química , Sequência de Aminoácidos , Animais , Valva Aórtica/citologia , Plaquetas/citologia , Plaquetas/metabolismo , Adesão Celular , Proliferação de Células , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Histamina/farmacologia , Humanos , Laminina/química , Laminina/metabolismo , Microscopia de Fluorescência , Peptídeos/metabolismo , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Suínos , Sindecana-1/química , Sindecana-1/metabolismo , Fator de von Willebrand/análise , Fator de von Willebrand/metabolismo
16.
PLoS One ; 10(3): e0121994, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25803806

RESUMO

It was recently reported that factor H, a regulatory component of the alternative complement pathway, is stored with von Willebrand factor (VWF) in the Weibel-Palade bodies of endothelial cells. If this were to be the case, it would have therapeutic importance for patients with the atypical hemolytic-uremic syndrome that can be caused either by a heterozygous defect in the factor H gene or by the presence of an autoantibody against factor H. The in vivo Weibel-Palade body secretagogue, des-amino-D-arginine vasopressin (DDAVP), would be expected to increase transiently the circulating factor H levels, in addition to increasing the circulating levels of VWF. We describe experiments demonstrating that factor H is released from endothelial cell cytoplasm without a secondary storage site. These experiments showed that factor H is not stored with VWF in endothelial cell Weibel-Palade bodies, and is not secreted in response in vitro in response to the Weibel-Palade body secretagogue, histamine. Furthermore, the in vivo Weibel-Palade body secretagogue, DDAVP does not increase the circulating factor H levels concomitantly with DDAVP-induced increased VWF. Factor I, a regulatory component of the alternative complement pathway that is functionally related to factor H, is also located in endothelial cell cytoplasm, and is also not present in endothelial cell Weibel-Palade bodies. Our data demonstrate that the factor H and factor I regulatory proteins of the alternative complement pathway are not stored in Weibel-Palade bodies. DDAVP induces the secretion into human plasma of VWF--but not factor H.


Assuntos
Fator H do Complemento/metabolismo , Via Alternativa do Complemento/imunologia , Citoplasma/metabolismo , Células Endoteliais/imunologia , Fibrinogênio/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoensaio , Microscopia de Fluorescência , Corpos de Weibel-Palade/metabolismo
17.
Transfusion ; 54(5): 1221-4, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24117464

RESUMO

BACKGROUND: Thrombotic thrombocytopenic purpura (TTP) is a life-threatening disease resulting in systemic microvascular thrombosis. The disease is caused by excessive platelet (PLT) adhesion to ultra-large (UL) von Willebrand factor (VWF) multimers inadequately cleaved by the processing enzyme ADAMTS-13. While many cases respond to plasma exchange performed with or without concurrent corticosteroids, treatment of the 10% to 20% of patients with refractory disease is difficult. Experimental studies demonstrating that N-acetylcysteine (NAC) inhibits PLT binding to endothelial cell-secreted and anchored UL VWF multimers suggest that NAC may be useful in the treatment of TTP. CASE REPORT: A 44-year-old woman presented with malaise, confusion, chest and abdominal pain, and transient visual loss. Laboratory results and peripheral blood smear were consistent with TTP. The patient was begun on plasma exchange and corticosteroid treatment, but after 10 days the PLT count was still less than 10.0 × 10(9) /L and she developed a fever. Rituximab was initiated, but the patient's condition worsened and she became comatose. Antibiotics were initiated, but cultures remained sterile. After 3 days of coma and further clinical deterioration, treatment with NAC was begun. The patient received a loading dose of 150 mg/kg NAC intravenously (IV) over 1 hour. Within 18 hours the patient awakened abruptly and began communicating with medical personnel. Plasma exchange, corticosteroids, rituximab, and NAC infusion (150 mg/kg IV over 17 hr daily × 10 days) were continued and by Day 17 the PLT count was more than 50 × 10(9) /L. The patient fully recovered and was discharged on Day 31. CONCLUSION: This is the first complete report of a TTP patient treated with NAC. NAC was a safe and effective supplementary treatment for refractory TTP in this patient.


Assuntos
Acetilcisteína/uso terapêutico , Púrpura Trombocitopênica Trombótica/tratamento farmacológico , Proteínas ADAM/sangue , Proteína ADAMTS13 , Adulto , Feminino , Humanos , Contagem de Plaquetas , Púrpura Trombocitopênica Trombótica/sangue
18.
Phys Rev Lett ; 110(10): 108102, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23521301

RESUMO

The mechanical force-induced activation of the adhesive protein von Willebrand factor (VWF), which experiences high hydrodynamic forces, is essential in initiating platelet adhesion. The importance of the mechanical force-induced functional change is manifested in the multimeric VWF's crucial role in blood coagulation, when high fluid shear stress activates plasma VWF (PVWF) multimers to bind platelets. Here, we showed that a pathological level of high shear stress exposure of PVWF multimers results in domain conformational changes, and the subsequent shifts in the unfolding force allow us to use force as a marker to track the dynamic states of the multimeric VWF. We found that shear-activated PVWF multimers are more resistant to mechanical unfolding than nonsheared PVWF multimers, as indicated in the higher peak unfolding force. These results provide insight into the mechanism of shear-induced activation of PVWF multimers.


Assuntos
Modelos Biológicos , Fator de von Willebrand/química , Agregação Plaquetária , Conformação Proteica , Estrutura Terciária de Proteína , Desdobramento de Proteína , Termodinâmica
19.
J Clin Invest ; 121(2): 593-603, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266777

RESUMO

Thrombotic thrombocytopenic purpura (TTP) is a life-threatening disease characterized by systemic microvascular thrombosis caused by adhesion of platelets to ultra-large vWF (ULVWF) multimers. These multimers accumulate because of a deficiency of the processing enzyme ADAMTS13. vWF protein forms long multimers from homodimers that first form through C-terminal disulfide bonds and then join through their N termini by further disulfide bonding. N-acetylcysteine (NAC) is an FDA-approved drug that has long been used to treat chronic obstructive lung disease and acetaminophen toxicity and is known to function in the former disorder by reducing mucin multimers. Here, we examined whether NAC could reduce vWF multimers, which polymerize in a manner similar to mucins. In vitro, NAC reduced soluble plasma-type vWF multimers in a concentration-dependent manner and rapidly degraded ULVWF multimer strings extruded from activated ECs. The effect was preceded by reduction of the intrachain disulfide bond encompassing the platelet-binding A1 domain. NAC also inhibited vWF-dependent platelet aggregation and collagen binding. Injection of NAC into ADAMTS13-deficient mice led to the rapid resolution of thrombi produced by ionophore treatment of the mesenteric venules and reduced plasma vWF multimers. These results suggest that NAC may be a rapid and effective treatment for patients with TTP.


Assuntos
Acetilcisteína/metabolismo , Plasma/metabolismo , Multimerização Proteica , Púrpura Trombocitopênica Trombótica/sangue , Fator de von Willebrand/química , Fator de von Willebrand/metabolismo , Proteína ADAMTS13 , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Animais , Antibacterianos/farmacologia , Células Endoteliais/metabolismo , Humanos , Masculino , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agregação Plaquetária/efeitos dos fármacos , Púrpura Trombocitopênica Trombótica/tratamento farmacológico , Ristocetina/farmacologia , Trombose/metabolismo , Trombose/patologia , Fator de von Willebrand/genética
20.
Blood ; 114(24): 5102-11, 2009 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19822897

RESUMO

Human umbilical vein endothelial cell (HUVEC)-released ADAMTS-13 (a disintegrin and metalloprotease with thrombospondin repeats) and HUVEC-secreted von Willebrand factor (VWF) strings were investigated under static conditions that allow the accumulation and analysis of ADAMTS-13. The latter was released constitutively from HUVECs and cleaved the secreted and cell-anchored VWF strings progressively during 15 minutes in Ca(2+)/Zn(2+)-containing buffer. HUVEC ADAMTS13 mRNA expression was approximately 1:100 of VWF monomeric subunit expression. In contrast to multimeric VWF stored within Weibel-Palade bodies and secreted rapidly in response to cell stimulation, ADAMTS-13 was released directly from the Golgi to the cell exterior without an organelle storage site. The constitutive release of ADAMTS-13 continued at the same slow rate regardless of the presence or absence of histamine stimulation of HUVECs. Consequently, the percentage of VWF strings cleaved by ADAMTS-13 at VWF Y(1605)-M(1606) decreased as the rate of VWF string secretion was increased by cell stimulation. Blockade of HUVEC ADAMTS-13 activity by antibodies to different ADAMTS-13 domains made it possible to detect the attachment of ADAMTS-13 all along the lengths of HUVEC-secreted VWF strings. Constitutive ADAMTS-13 released from endothelial cells may contribute to the maintenance of cell surfaces free of hyperadhesive VWF multimeric strings.


Assuntos
Proteínas ADAM/metabolismo , Células Endoteliais/metabolismo , Fator de von Willebrand/metabolismo , Proteína ADAMTS13 , Humanos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...