Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Eur J Haematol ; 112(4): 516-529, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37994576

RESUMO

OBJECTIVES: To describe real-world use/effectiveness of pegcetacoplan (PEG) in paroxysmal nocturnal haemoglobinuria (PNH). METHODS: Data were drawn from the Adelphi PNH Disease Specific Programme™, a cross-sectional survey conducted in France, Italy, Germany, Spain and the United States from January to November 2022. Patients had a confirmed PNH diagnosis and received PEG for ≥1 month. Physicians reported patient characteristics, treatment use/satisfaction and their perception of patients' fatigue and health-related quality of life (HRQoL). Patients reported treatment satisfaction and completed questionnaires assessing fatigue, HRQoL and productivity. Descriptive statistics were reported. RESULTS: Overall, 14 physicians provided data for 61 patients who had received 1080 mg/dose PEG for 1.3-14.8 months. At data collection compared to PEG initiation: haemoglobin was 2.5 g/dL higher on average; proportion of patients with lactate dehydrogenase (LDH) ≥1.5 × upper limit of normal was reduced by 27.4%; physician-perceived fatigue was lower and HRQoL better. Physician- and patient-reported treatment satisfaction was high for >90% of patients. Physicians and patients were more satisfied with PEG than previously prescribed C5 complement inhibitors. Mean work impairment and activity impairment in the 7 days prior to data collection were 32.9% and 22.4%, respectively. CONCLUSIONS: These real-world data support the effectiveness of PEG through positive effects on haemoglobin, LDH, fatigue and HRQoL.


Assuntos
Hemoglobinúria Paroxística , Peptídeos Cíclicos , Qualidade de Vida , Humanos , Estados Unidos , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/tratamento farmacológico , Estudos Transversais , Resultado do Tratamento , L-Lactato Desidrogenase , Hemoglobinas
2.
J Thromb Haemost ; 21(12): 3450-3462, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37678546

RESUMO

BACKGROUND: Severe hemophilia A (HA) negatively impacts health-related quality of life (HRQOL). OBJECTIVES: We aimed to analyze HRQOL in adult men with severe HA without inhibitors after valoctocogene roxaparvovec gene transfer in the phase 3 trial GENEr8-1. METHODS: Participant-reported outcomes were the hemophilia-specific quality of life questionnaire for adults (Haemo-QOL-A), the EQ-5D-5L instrument, the Hemophilia Activities List (HAL), and the Work Productivity and Activity Impairment Questionnaire: Hemophilia Specific (WPAI+CIQ:HS). Participants completed the questionnaires at baseline and through 104 weeks postinfusion with 6 × 1013 vg/kg of valoctocogene roxaparvovec. Scores were analyzed per participant characteristics and outcomes. RESULTS: For 132 HIV-negative participants, mean change from baseline in Haemo-QOL-A Total Score met the anchor-based clinically important difference (CID: 5.5) by week 12; the mean (SD) increase was 7.0 (12.6) at week 104. At week 104, improvement in Consequences of Bleeding, Treatment Concern, Worry, and Role Functioning domain scores exceeded the CID (6). EQ-5D-5L Utility Index scores improved above the CID at week 52, but not at week 104. EQ-5D-5L visual analog scale and HAL scores increased from baseline to week 104. Participants reported less activity and work impairment at week 104 than baseline. Participants with problem joints had lower mean baseline Haemo-QOL-A Total and domain scores than those without them, but improved over 104 weeks, except for 11 participants with ≥3 problem joints. Participants with 0 bleeds during the baseline prophylaxis period reported Haemo-QOL-A score improvements above the CID, including in the Consequences of Bleeding domain. CONCLUSION: Valoctocogene roxaparvovec provided clinically meaningful HRQOL improvement for men with severe HA.


Assuntos
Hemofilia A , Adulto , Masculino , Humanos , Hemofilia A/diagnóstico , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Qualidade de Vida , Hemorragia , Inquéritos e Questionários
4.
PLoS One ; 12(2): e0171067, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28152086

RESUMO

Invariant NKT (iNKT) cells can be activated to stimulate a broad inflammatory response. In murine models of sickle cell disease (SCD), interruption of iNKT cell activity prevents tissue injury from vaso-occlusion. NKTT120 is an anti-iNKT cell monoclonal antibody that has the potential to rapidly and specifically deplete iNKT cells and, potentially, prevent vaso-occlusion. We conducted an open-label, multi-center, single-ascending-dose study of NKTT120 to determine its pharmacokinetics, pharmacodynamics and safety in steady-state patients with SCD. Doses were escalated in a 3+3 study design over a range from 0.001 mg/kg to 1.0 mg/kg. Twenty-one adults with SCD were administered NKTT120 as part of 7 dose cohorts. Plasma levels of NKTT120 predictably increased with higher doses. Median half-life of NKTT120 was 263 hours. All subjects in the higher dose cohorts (0.1 mg/kg, 0.3 mg/kg, and 1 mg/kg) demonstrated decreased iNKT cells below the lower limit of quantification within 6 hours after infusion, the earliest time point at which they were measured. In those subjects who received the two highest doses of NKTT120 (0.3, 1 mg/kg), iNKT cells were not detectable in the peripheral blood for a range of 2 to 5 months. There were no serious adverse events in the study deemed to be related to NKTT120. In adults with SCD, NKTT120 produced rapid, specific and sustained iNKT cell depletion without any infusional toxicity or attributed serious adverse events. The next step is a trial to determine NKTT120's ability to decrease rate of vaso-occlusive pain episodes. TRIAL REGISTRATION: clinicaltrials.gov NCT01783691.


Assuntos
Anemia Falciforme/imunologia , Anemia Falciforme/terapia , Anticorpos Monoclonais Humanizados/uso terapêutico , Depleção Linfocítica/métodos , Células T Matadoras Naturais/imunologia , Adolescente , Adulto , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacocinética , Criança , Estudos de Coortes , Feminino , Meia-Vida , Humanos , Masculino , Adulto Jovem
5.
Blood Adv ; 1(20): 1645-1649, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-29296811

RESUMO

Adenosine A2A receptor (A2AR) agonists have been shown to decrease tissue inflammation induced by hypoxia/reoxygenation in mice with sickle cell disease (SCD). The key mediator of the A2AR agonist's anti-inflammatory effects is a minor lymphocyte subset, invariant natural killer T (iNKT) cells. We tested the hypothesis that administration of an A2AR agonist in patients with SCD would decrease iNKT cell activation and dampen the severity of vaso-occlusive (VO) crises. In a phase 2, randomized, placebo-controlled trial, we administered a 48-hour infusion of the A2AR agonist regadenoson (1.44 µg/kg per hour) to patients with SCD during VO crises to produce a plasma concentration of ∼5 nM, a concentration known from prior studies to suppress iNKT cell activation in SCD. The primary outcome measure was a >30% reduction in the percentage of activated iNKT cells. Ninety-two patients with SCD were randomized to receive a 48-hour infusion of regadenoson or placebo, in addition to standard-of-care treatment, during hospital admission for a VO crisis and had analyzable iNKT cell samples. The proportion of subjects who demonstrated a reduction of >30% in activated iNKT cells was not significantly different between the regadenoson and placebo arms (43% vs 23%; P = .07). There were also no differences between regadenoson and placebo groups in length of hospital stay, mean total opioid use, or pain scores. These data demonstrate that a low-dose infusion of regadenoson intended to reduce the activity of iNKT cells is not sufficient to produce a statistically significant reduction in such activation or in measures of clinical efficacy. This trial was registered at www.clinicaltrials.gov as #NCT01788631.

6.
Curr Biol ; 25(3): 286-295, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25544610

RESUMO

BACKGROUND: O-fucose is added to cysteine-rich domains called thrombospondin type 1 repeats (TSRs) by protein O-fucosyltransferase 2 (POFUT2) and is elongated with glucose by ß3-glucosyltransferase (B3GLCT). Mutations in B3GLCT result in Peters plus syndrome (PPS), an autosomal recessive disorder characterized by eye and other developmental defects. Although 49 putative targets are known, the function of the disaccharide and its role in PPS remain unexplored. RESULTS: Here we show that while POFUT2 is required for secretion of all targets tested, B3GLCT only affects the secretion of a subset, consistent with the observation that B3GLCT mutant phenotypes in PPS patients are less severe than embryonic lethal phenotypes of Pofut2-null mice. O-glycosylation occurs cotranslationally, as TSRs fold. Mass spectral analysis reveals that TSRs from mature, secreted protein are stoichiometrically modified with the disaccharide, whereas TSRs from protein still folding in the ER are partially modified, suggesting that O-glycosylation marks folded TSRs and promotes ER exit. In vitro unfolding assays demonstrate that fucose and glucose stabilize folded TSRs in an additive manner. In vitro refolding assays under redox conditions showed that POFUT2 recognizes, glycosylates, and stabilizes the folded form of TSRs, resulting in a net acceleration of folding. CONCLUSIONS: While known ER quality-control machinery rely on identifying and tagging unfolded proteins, we find that POFUT2 and B3GLCT mediate a noncanonical ER quality-control mechanism that recognizes folded TSRs and stabilizes them by glycosylation. Our findings provide a molecular basis for the defects observed in PPS and potential targets that contribute to the pathology.


Assuntos
Fenda Labial/genética , Córnea/anormalidades , Retículo Endoplasmático/genética , Fucosiltransferases/metabolismo , Galactosiltransferases/metabolismo , Glucosiltransferases/metabolismo , Transtornos do Crescimento/genética , Deformidades Congênitas dos Membros/genética , Western Blotting , Fenda Labial/metabolismo , Córnea/metabolismo , Retículo Endoplasmático/fisiologia , Fucose/metabolismo , Galactosiltransferases/genética , Glucose/metabolismo , Glucosiltransferases/genética , Transtornos do Crescimento/metabolismo , Células HEK293 , Humanos , Imunoprecipitação , Deformidades Congênitas dos Membros/metabolismo , Modelos Biológicos , Mutação/genética , Oxirredução , Dobramento de Proteína , RNA Interferente Pequeno/genética , Trombospondina 1/metabolismo
7.
Blood ; 121(17): 3329-34, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23377438

RESUMO

Adenosine A2A receptor (A2AR) agonists reduce invariant natural killer T (iNKT) cell activation and decrease inflammation in sickle cell disease (SCD) mice. We conducted a phase 1 trial of the A2AR agonist regadenoson in adults with SCD. The target dose was 1.44 µg/kg/h. iNKT cell activation was evaluated using antibodies targeting the p65 subunit of nuclear factor-κB (phospho-NF-κB p65), interferon-γ (IFN-γ), and A2AR. Regadenoson was administered to 27 adults with SCD. We examined 21 patients at steady state and 6 during painful vaso-occlusive crises (pVOC). iNKT cell activation was also measured in 14 African-American controls. During pVOC, the fraction of iNKT cells demonstrating increased phospho-NF-κB p65 and A2AR expression was significantly higher compared with controls (P < .01) and steady-state patients (P < .05). IFN-γ expression was also significantly higher compared with controls (P = .02). After a 24-hour infusion of regadenoson during pVOC, phospho-NF-κB p65 activation in iNKT cells decreased compared to baseline by a median of 48% (P = .03) to levels similar to controls and steady-state SCD. No toxicities were identified. Infusional regadenoson administered to adults with SCD at 1.44 µg/kg/h during pVOC decreases activation of iNKT cells without toxicity.


Assuntos
Agonistas do Receptor A2 de Adenosina/uso terapêutico , Anemia Falciforme/tratamento farmacológico , Células T Matadoras Naturais/metabolismo , Purinas/uso terapêutico , Pirazóis/uso terapêutico , Receptor A2A de Adenosina/química , Doenças Vasculares/tratamento farmacológico , Agonistas do Receptor A2 de Adenosina/farmacocinética , Adulto , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Estudos de Casos e Controles , Feminino , Citometria de Fluxo , Humanos , Infusões Intravenosas , Interferon gama/metabolismo , Masculino , Fosforilação , Prognóstico , Purinas/farmacocinética , Pirazóis/farmacocinética , Receptor A2A de Adenosina/metabolismo , Distribuição Tecidual , Fator de Transcrição RelA/metabolismo , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
8.
Trans Am Clin Climatol Assoc ; 123: 312-7; discussion 317-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23303999

RESUMO

A humanized murine sickle cell-disease (SCD) model (NY1DD) has been used to study ischemia/reperfusion injury (IRI) in sickle cell anemia, and iNKT cells (a very small subset of murine and human T cells) have been found to instigate such injury in this model. Furthermore, levels of activated iNKT cells are generally elevated in the circulation of patients with SCD. Because activated iNKT cells are rich in adenosine A2A receptors which, when agonized, down-regulate the inflammatory cytokine expression that characterizes the cells, we have conducted a phase 1 trial of a constant infusion of low-dose regadenoson (an adenosine analogue with high A2A receptor specificity) to determine its safety and the capacity of a safe dose to down-regulate circulating iNKT cells in patients with SCD. We have found two dose rates that are both safe and effective and now plan a controlled Phase 2B clinical trial to determine whether our highest dose, administered as a 48-hour constant infusion, will induce faster remission in both painful vaso-occlusive crisis (pVOC) and acute chest syndrome (ACS).


Assuntos
Agonistas do Receptor A2 de Adenosina/uso terapêutico , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/patologia , Células T Matadoras Naturais/patologia , Purinas/uso terapêutico , Pirazóis/uso terapêutico , Agonistas do Receptor A2 de Adenosina/administração & dosagem , Contagem de Células , Relação Dose-Resposta a Droga , Humanos , Infusões Intravenosas , Purinas/administração & dosagem , Pirazóis/administração & dosagem , Indução de Remissão , Fatores de Tempo , Resultado do Tratamento
9.
J Biol Chem ; 284(45): 30925-32, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19729451

RESUMO

ADAMTS13 is a plasma metalloprotease that cleaves ultralarge von Willebrand factor multimers to generate less thrombogenic fragments. Although this cleavage can occur at the surface of endothelial cells, it is currently unknown whether this process involves binding of the ADAMTS13 to the endothelial cell plasma membrane. Using different assay systems, we present evidence that ADAMTS13 binds to endothelial cells in a specific, reversible, and time-dependent manner with a K(d) of 58 nm. This binding requires the COOH-terminal thrombospondin type 1 repeats of the protease. Binding is inhibited in the presence of heparin and by trypsin treatment of the cells. ADAMTS13 that was prebound to endothelial cells exhibited increased proteolysis of VWF as compared with ADAMTS13 present only in solution. These data support the notion that cleavage of VWF occurs mainly at the endothelial cell surface.


Assuntos
Proteínas ADAM/metabolismo , Endotélio Vascular/metabolismo , Processamento de Proteína Pós-Traducional , Fator de von Willebrand/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS13 , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Ligação Proteica
10.
Haematologica ; 92(10): 1419-22, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17768109

RESUMO

Reportedly, complexes between factor XI and ADAMTS13 are detected with a commercial ADAMTS13/FXI ELISA kit in plasma and are decreased in thrombotic thrombocytopenic purpura (TTP). Using this kit, control and TTP patient plasma contained varying amounts of signal (25-670% of a reference plasma) but no signal was observed for mixtures of recombinant enzymes, suggesting little interaction. ADAMTS13/FXI complexes were undetectable by immunoprecipitation or gel filtration chromatography in control plasma or mixtures of recombinant proteins. These results suggest that ADAMTS13/FXI complexes are insignificant in plasma and unlikely to affect the function of either protein during normal hemostasis or in TTP.


Assuntos
Proteínas ADAM/sangue , Fator XI/metabolismo , Proteína ADAMTS13 , Cromatografia em Gel , Humanos , Ligação Proteica
11.
J Biol Chem ; 282(23): 17014-23, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17395589

RESUMO

ADAMTS13 is a plasma metalloproteinase that cleaves von Willebrand factor to smaller, less thrombogenic forms. Deficiency of ADAMTS13 activity in plasma leads to thrombotic thrombocytopenic purpura. ADAMTS13 contains eight thrombospondin type 1 repeats (TSR), seven of which contain a consensus sequence for the direct addition of fucose to the hydroxyl group of serine or threonine. Mass spectral analysis of tryptic peptides derived from human ADAMTS13 indicate that at least six of the TSRs are modified with an O-fucose disaccharide. Analysis of [(3)H]fucose metabolically incorporated into ADAMTS13 demonstrated that the disaccharide has the structure glucose-beta1,3-fucose. Mutation of the modified serine to alanine in TSR2, TSR5, TSR7, and TSR8 reduced the secretion of ADAMTS13. Mutation of more than one site dramatically reduced secretion regardless of the sites mutated. When the expression of protein O-fucosyltransferase 2 (POFUT2), the enzyme that transfers fucose to serines in TSRs, was reduced using siRNA, the secretion of ADAMTS13 decreased. A similar outcome was observed when ADAMTS13 was expressed in a cell line unable to synthesize the donor for fucose addition, GDP-fucose. Although overexpression of POFUT2 did not affect the secretion of wild-type ADAMTS13, it did increase the secretion of the ADAMTS13 TSR1,2 double mutant but not that of ADAMTS13 TSR1-8 mutant. Together these findings indicate that O-fucosylation is functionally significant for secretion of ADAMTS13.


Assuntos
Proteínas ADAM/metabolismo , Fucose/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS13 , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Primers do DNA , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , RNA Interferente Pequeno
12.
Proc Natl Acad Sci U S A ; 103(50): 19099-104, 2006 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-17146059

RESUMO

Von Willebrand factor (VWF) is a multimeric protein that mediates platelet adhesion at sites of vascular injury, and ADAMTS13 (a disintegrin and metalloprotease with thrombospondin)is a multidomain metalloprotease that limits platelet adhesion by a feedback mechanism in which fluid shear stress induces proteolysis of VWF and prevents disseminated microvascular thrombosis. Cleavage of the Tyr(1605)-Met(1606) scissile bond in the VWF A2 domain depends on a Glu(1660)-Arg(1668) segment in the same domain and on the noncatalytic spacer domain of ADAMTS13, suggesting that extensive enzyme-substrate interactions facilitate substrate recognition. Based on mutagenesis and kinetic analysis, we find that the ADAMTS13 spacer domain binds to an exosite near the C terminus of the VWF A2 domain. Deleting the spacer domain from ADAMTS13 or deleting the exosite from the VWF substrate reduced the rate of cleavage approximately 20-fold. A cleavage product containing the exosite was a hyperbolic mixed-type inhibitor of ADAMTS13 proteolysis of either VWF multimers or model peptide substrates but only if the ADAMTS13 enzyme contained the spacer domain. The specificity of this unique mechanism depends on tension-induced unfolding of the VWF A2 domain, which exposes the scissile bond and exosite for interaction with complementary sites on ADAMTS13.


Assuntos
Metaloendopeptidases/metabolismo , Trombose/metabolismo , Fator de von Willebrand/metabolismo , Linhagem Celular , Variação Genética/genética , Humanos , Cinética , Metaloendopeptidases/genética , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Trombose/genética , Fator de von Willebrand/genética
13.
J Biol Chem ; 280(23): 21773-8, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15824096

RESUMO

ADAMTS13, a metalloprotease, cleaves von Willebrand factor (VWF) in plasma to generate smaller, less thrombogenic fragments. The interaction of von Willebrand factor with specific ADAMTS13 domains was characterized with a binding assay employing von Willebrand factor immobilized on a plastic surface. ADAMTS13 binding was saturable and reversible. Equilibrium binding occurred within 2 h and the half-time for dissociation was approximately 4 h. Binding to von Willebrand factor was similar with either recombinant ADAMTS13 or normal plasma ADAMTS13; plasma from a patient who lacked ADAMTS13 activity showed no binding. The stoichiometry of binding was one ADAMTS13 per two von Willebrand factor monomers, and the K(d) was 14 nm. The ADAMTS13 metalloprotease and disintegrin domains did not bind VWF detectably. ADAMTS13 truncated after the first thrombospondin type 1 repeat bound VWF with a K(d) of 206 nm, whereas ADAMTS13 truncated after the spacer domain had a K(d) of 23 nm, which is comparable with that of full-length ADAMTS13. Truncation after the eighth thrombospondin type 1 repeat reduced the binding affinity by approximately 3-fold and truncation after the seventh thrombospondin type 1 repeat in addition to the CUB domains increased the affinity for von Willebrand factor by approximately 2-fold. Therefore, the spacer domain is required for ADAMTS13 binding to von Willebrand factor. The first thrombospondin repeat also affects binding, and the C-terminal thrombospondin type 1 and CUB domains of ADAMTS13 may modulate this interaction.


Assuntos
Metaloendopeptidases/metabolismo , Fator de von Willebrand/metabolismo , Proteínas ADAM , Proteína ADAMTS13 , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Imunoglobulina G/química , Cinética , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Trombospondinas/química , Fatores de Tempo
14.
J Biol Chem ; 278(47): 46643-8, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-12975358

RESUMO

ADAMTS13 belongs to the "a disintegrin and metalloprotease with thrombospondin repeats" family, and cleaves von Willebrand factor multimers into smaller forms. For several related proteases, normal folding and enzymatic latency depend on an NH2-terminal propeptide that is removed by proteolytic processing during biosynthesis. However, the ADAMTS13 propeptide is unusually short and poorly conserved, suggesting it may not perform these functions. ADAMTS13 was secreted from transfected HeLa cells with a half-time of 7 h and the rate-limiting step was exported from the endoplasmic reticulum. Deletion of the propeptide did not impair the secretion of active ADAMTS13, indicating that the propeptide is dispensable for folding. Furin was shown to be sufficient for ADAMTS13 propeptide processing in two ways. First, mutation of the furin consensus recognition site prevented propeptide cleavage in HeLa cells and resulted in secretion of pro-ADAMTS13. Second, furin-deficient LoVo cells secreted ADAMTS13 with the propeptide intact, and cotransfection with furin restored propeptide cleavage. In both cell lines, secreted pro-ADAMTS13 had normal proteolytic activity toward von Willebrand factor. In cells coexpressing both ADAMTS13 and von Willebrand factor, pro-ADAMTS13 cleaved pro-von Willebrand factor intracellularly. Therefore, the ADAMTS13 propeptide is not required for folding or secretion, and does not perform the common function of maintaining enzyme latency.


Assuntos
Metaloendopeptidases/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas ADAM , Proteína ADAMTS13 , Sequência de Aminoácidos , Furina/metabolismo , Células HeLa , Humanos , Cinética , Dobramento de Proteína , Alinhamento de Sequência , Deleção de Sequência , Transfecção , Fator de von Willebrand/metabolismo
15.
J Biol Chem ; 278(32): 30136-41, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12791682

RESUMO

ADAMTS13 consists of a reprolysin-type metalloprotease domain followed by a disintegrin domain, a thrombospondin type 1 motif (TSP1), Cys-rich and spacer domains, seven more TSP1 motifs, and two CUB domains. ADAMTS13 limits platelet accumulation in microvascular thrombi by cleaving the Tyr1605-Met1606 bond in von Willebrand factor, and ADAMTS13 deficiency causes a lethal syndrome, thrombotic thrombocytopenic purpura. ADAMTS13 domains required for substrate recognition were localized by the characterization of recombinant deletion mutants. Constructs with C-terminal His6 and V5 epitopes were expressed by transient transfection of COS-7 cells or in a baculovirus system. No association with extracellular matrix or cell surface was detected for any ADAMTS13 variant by immunofluorescence microscopy or chemical modification. Both plasma and recombinant full-length ADAMTS13 cleaved von Willebrand factor subunits into two fragments of 176 kDa and 140 kDa. Recombinant ADAMTS13 was divalent metal ion-dependent and was inhibited by IgG from a patient with idiopathic thrombotic thrombocytopenic purpura. ADAMTS13 that was truncated after the metalloprotease domain, the disintegrin domain, the first TSP1 repeat, or the Cys-rich domain was not able to cleave von Willebrand factor, whereas addition of the spacer region restored protease activity. Therefore, the spacer region is necessary for normal ADAMTS13 activity toward von Willebrand factor, and the more C-terminal TSP1 and CUB domains are dispensable in vitro.


Assuntos
Metaloendopeptidases/química , Fator de von Willebrand/metabolismo , Proteínas ADAM , Proteína ADAMTS13 , Motivos de Aminoácidos , Animais , Baculoviridae/metabolismo , Biotinilação , Células COS , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Cisteína/química , DNA Complementar/metabolismo , Eletroforese em Gel de Poliacrilamida , Epitopos , Matriz Extracelular/metabolismo , Deleção de Genes , Vetores Genéticos , Humanos , Insetos , Microscopia de Fluorescência , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transfecção , Tirosina/química , Fator de von Willebrand/química
16.
Curr Opin Hematol ; 9(5): 389-94, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12172456

RESUMO

Thrombotic thrombocytopenic purpura (TTP) has been a mysterious and deadly disease that often could be treated effectively by plasma exchange, but without real understanding of the underlying pathophysiology. Recent advances now suggest that deficiency of a specific von Willebrand factor (VWF) cleaving protease promotes tissue injury in TTP. VWF multimers participate in the formation of platelet thrombi. Proteolytic cleavage of VWF multimers normally limits platelet thrombus growth, and failure to cleave VWF appears to encourage microvascular thrombosis. The VWF cleaving protease proves to be a new member of the ADAMTS family of metalloproteases, designated ADAMTS13. Autoantibodies that inhibit ADAMTS13 cause sporadic TTP, and mutations in the ADAMTS13 gene cause an autosomal recessive form of chronic relapsing TTP. Further studies of ADAMTS13 seem likely to change our approach to the diagnosis and treatment of TTP and other thrombotic microangiopathies.


Assuntos
Metaloendopeptidases/fisiologia , Púrpura Trombocitopênica Trombótica/etiologia , Proteínas ADAM , Proteína ADAMTS13 , Humanos , Metaloendopeptidases/deficiência , Metaloendopeptidases/genética , Microcirculação , Mutação , Púrpura Trombocitopênica Trombótica/diagnóstico , Púrpura Trombocitopênica Trombótica/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...