Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 11(1): 6435, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33353951

RESUMO

Human ß-tryptase, a tetrameric trypsin-like serine protease, is an important mediator of allergic inflammatory responses in asthma. Antibodies generally inhibit proteases by blocking substrate access by binding to active sites or exosites or by allosteric modulation. The bivalency of IgG antibodies can increase potency via avidity, but has never been described as essential for activity. Here we report an inhibitory anti-tryptase IgG antibody with a bivalency-driven mechanism of action. Using biochemical and structural data, we determine that four Fabs simultaneously occupy four exosites on the ß-tryptase tetramer, inducing allosteric changes at the small interface. In the presence of heparin, the monovalent Fab shows essentially no inhibition, whereas the bivalent IgG fully inhibits ß-tryptase activity in a hinge-dependent manner. Our results suggest a model where the bivalent IgG acts akin to molecular pliers, pulling the tetramer apart into inactive ß-tryptase monomers, and may provide an alternative strategy for antibody engineering.


Assuntos
Anticorpos Monoclonais/metabolismo , Imunoglobulina G/metabolismo , Triptases/metabolismo , Regulação Alostérica/efeitos dos fármacos , Sequência de Aminoácidos , Heparina/farmacologia , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Imunoglobulina G/química , Modelos Moleculares , Proteínas Mutantes/química , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica , Triptases/química
3.
Cell ; 179(2): 417-431.e19, 2019 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-31585081

RESUMO

Severe asthma patients with low type 2 inflammation derive less clinical benefit from therapies targeting type 2 cytokines and represent an unmet need. We show that mast cell tryptase is elevated in severe asthma patients independent of type 2 biomarker status. Active ß-tryptase allele count correlates with blood tryptase levels, and asthma patients carrying more active alleles benefit less from anti-IgE treatment. We generated a noncompetitive inhibitory antibody against human ß-tryptase, which dissociates active tetramers into inactive monomers. A 2.15 Å crystal structure of a ß-tryptase/antibody complex coupled with biochemical studies reveal the molecular basis for allosteric destabilization of small and large interfaces required for tetramerization. This anti-tryptase antibody potently blocks tryptase enzymatic activity in a humanized mouse model, reducing IgE-mediated systemic anaphylaxis, and inhibits airway tryptase in Ascaris-sensitized cynomolgus monkeys with favorable pharmacokinetics. These data provide a foundation for developing anti-tryptase as a clinical therapy for severe asthma.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Asma/terapia , Mastócitos/enzimologia , Mastócitos/imunologia , Triptases/antagonistas & inibidores , Triptases/imunologia , Adolescente , Regulação Alostérica/imunologia , Animais , Linhagem Celular , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Coelhos
4.
Biochem J ; 472(2): 169-81, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26385991

RESUMO

High temperature requirement A1 (HtrA1) is a trypsin-fold serine protease implicated in the progression of age-related macular degeneration (AMD). Our interest in an antibody therapy to neutralize HtrA1 faces the complication that the target adopts a trimeric arrangement, with three active sites in close proximity. In the present study, we describe antibody 94, obtained from a human antibody phage display library, which forms a distinct macromolecular complex with HtrA1 and inhibits the enzymatic activity of recombinant and native HtrA1 forms. Using biochemical methods and negative-staining EM we were able to elucidate the molecular composition of the IgG94 and Fab94 complexes and the associated inhibition mechanism. The 246-kDa complex between the HtrA1 catalytic domain trimer (HtrA1_Cat) and Fab94 had a propeller-like organization with one Fab bound peripherally to each protomer. Low-resolution EM structures and epitope mapping indicated that the antibody binds to the surface-exposed loops B and C of the catalytic domain, suggesting an allosteric inhibition mechanism. The HtrA1_Cat-IgG94 complex (636 kDa) is a cage-like structure with three centrally located IgG94 molecules co-ordinating two HtrA1_Cat trimers and the six active sites pointing into the cavity of the cage. In both complexes, all antigen-recognition regions (paratopes) are found to bind one HtrA1 protomer and all protomers are bound by a paratope, consistent with the complete inhibition of enzyme activity. Therefore, in addition to its potential therapeutic usefulness, antibody 94 establishes a new paradigm of multimeric serine protease inhibition.


Assuntos
Anticorpos Neutralizantes/farmacologia , Complexo Antígeno-Anticorpo/química , Antineoplásicos/farmacologia , Melanoma/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Serina Endopeptidases/metabolismo , Regulação Alostérica , Substituição de Aminoácidos , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/metabolismo , Especificidade de Anticorpos , Antineoplásicos/química , Antineoplásicos/metabolismo , Sítios de Ligação de Anticorpos , Domínio Catalítico , Linhagem Celular Tumoral , Mapeamento de Epitopos , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fab das Imunoglobulinas/farmacologia , Imunoglobulina G/química , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Imunoglobulina G/farmacologia , Melanoma/enzimologia , Melanoma/metabolismo , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/farmacologia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Serina Endopeptidases/química , Serina Endopeptidases/genética
5.
J Biol Chem ; 287(52): 43482-91, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23135270

RESUMO

Proprotein convertase subtilisin/kexin 9 (PCSK9) regulates plasma LDL cholesterol levels by regulating the degradation of LDL receptors. Another proprotein convertase, furin, cleaves PCSK9 at Arg(218)-Gln(219) in the surface-exposed "218 loop." This cleaved form circulates in blood along with the intact form, albeit at lower concentrations. To gain a better understanding of how cleavage affects PCSK9 function, we produced recombinant furin-cleaved PCSK9 using antibody Ab-3D5, which binds the intact but not the cleaved 218 loop. Using Ab-3D5, we also produced highly purified hepsin-cleaved PCSK9. Hepsin cleaves PCSK9 at Arg(218)-Gln(219) more efficiently than furin but also cleaves at Arg(215)-Phe(216). Further analysis by size exclusion chromatography and mass spectrometry indicated that furin and hepsin produced an internal cleavage in the 218 loop without the loss of the N-terminal segment (Ser(153)-Arg(218)), which remained attached to the catalytic domain. Both furin- and hepsin-cleaved PCSK9 bound to LDL receptor with only 2-fold reduced affinity compared with intact PCSK9. Moreover, they reduced LDL receptor levels in HepG2 cells and in mouse liver with only moderately lower activity than intact PCSK9, consistent with the binding data. Single injection into mice of furin-cleaved PCSK9 resulted in significantly increased serum cholesterol levels, approaching the increase by intact PCSK9. These findings indicate that circulating furin-cleaved PCSK9 is able to regulate LDL receptor and serum cholesterol levels, although somewhat less efficiently than intact PCSK9. Therapeutic anti-PCSK9 approaches that neutralize both forms should be the most effective in preserving LDL receptors and in lowering plasma LDL cholesterol.


Assuntos
Colesterol/sangue , Furina/metabolismo , Pró-Proteína Convertases/metabolismo , Proteólise , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Animais , Anticorpos Monoclonais Murinos/química , Colesterol/genética , Furina/genética , Células Hep G2 , Humanos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/genética , Estrutura Secundária de Proteína , Receptores de LDL/genética , Serina Endopeptidases/genética
6.
Structure ; 20(6): 1040-50, 2012 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-22578544

RESUMO

The homotrimeric human serine protease HtrA1 is homologous to bacterial HtrA proteases regarding the trypsin-like catalytic and PDZ domains but differs by the presence of an N-terminal domain with IGFBP and Kazal homology. The crystal structures and SAXS analysis presented herein reveal the rare tandem of IGFBP- and Kazal-like modules, a protease active site that adopts a competent conformation in the absence of substrate or inhibitor and a model for the intact protein in solution. Highly sensitive enzymatic assays and binding studies demonstrate that the N-terminal tandem has no apparent effect on protease activity, and in accordance with the structure-based predictions, neither the IGFBP- nor Kazal-like module retains the function of their prototype proteins. Our structures of the unliganded HtrA1 active site suggest two-state equilibrium and a "conformational selection" model, in which substrate binds to the active conformer.


Assuntos
Serina Endopeptidases/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Apoproteínas/química , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Ativação Enzimática , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Ligação de Hidrogênio , Isoflurofato/química , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Espalhamento a Baixo Ângulo , Serina Endopeptidases/genética , Inibidores de Serina Proteinase/química , Especificidade por Substrato , Propriedades de Superfície
7.
Structure ; 17(12): 1614-1624, 2009 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-20004165

RESUMO

Recent structural studies have outlined the mechanism of protease inhibition by active site-directed antibodies. However, the molecular basis of allosteric inhibition by antibodies has been elusive. Here we report the 2.35 A resolution structure of the trypsin-like serine protease hepatocyte growth factor activator (HGFA) in complex with the allosteric antibody Ab40, a potent inhibitor of HGFA catalytic activity. The antibody binds at the periphery of the substrate binding cleft and imposes a conformational change on the entire 99-loop (chymotrypsinogen numbering). The altered conformation of the 99-loop is incompatible with substrate binding due to the partial collapse of subsite S2 and the reorganization of subsite S4. Remarkably, a single residue deletion of Ab40 abolished inhibition of HGFA activity, commensurate with the reversal of the 99-loop conformation to its "competent" state. The results define an "allosteric switch" mechanism as the basis of protease inhibition by an allosteric antibody.


Assuntos
Anticorpos/farmacologia , Serina Proteases/imunologia , Inibidores de Serina Proteinase/farmacologia , Regulação Alostérica , Sequência de Aminoácidos , Anticorpos/química , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Serina Proteases/metabolismo
8.
Proc Natl Acad Sci U S A ; 104(50): 19784-9, 2007 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-18077410

RESUMO

To better understand how the relatively flat antigen-combining sites of antibodies interact with the concave shaped substrate-binding clefts of proteases, we determined the structures of two antibodies in complex with the trypsin-like hepatocyte growth-factor activator (HGFA). The two inhibitory antibodies, Ab58 and Ab75, were generated from a human Fab phage display library with synthetic diversity in the three complementarity determining regions (H1, H2, and H3) of the heavy chain, mimicking the natural diversity of the human Ig repertoire. Biochemical studies and the structures of the Fab58:HGFA (3.5-A resolution) and the Fab75:HGFA (2.2-A resolution) complexes revealed that Ab58 obstructed substrate access to the active site, whereas Ab75 allosterically inhibited substrate hydrolysis. In both cases, the antibodies interacted with the same protruding element (99-loop), which forms part of the substrate-binding cleft. Ab58 inserted its H1 and H2 loops in the cleft to occupy important substrate interaction sites (S3 and S2). In contrast, Ab75 bound at the backside of the cleft to a region corresponding to thrombin exosite II, which is known to interact with allosteric effector molecules. In agreement with the structural analysis, binding assays with active site inhibitors and enzymatic assays showed that Ab58 is a competitive inhibitor, and Ab75 is a partial competitive inhibitor. These results provide structural insight into antibody-mediated protease inhibition. They suggest that unlike canonical inhibitors, antibodies may preferentially target protruding loops at the rim of the substrate-binding cleft to interfere with the catalytic machinery of proteases without requiring long insertion loops.


Assuntos
Anticorpos/química , Anticorpos/metabolismo , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Serina Endopeptidases/imunologia , Animais , Anticorpos/farmacologia , Sítios de Ligação de Anticorpos , Ligação Competitiva/imunologia , Catálise , Humanos , Camundongos , Inibidores de Proteases/farmacologia , Coelhos , Serina Endopeptidases/metabolismo
9.
Proc Natl Acad Sci U S A ; 104(13): 5306-11, 2007 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-17372204

RESUMO

Hepatocyte growth factor (HGF), the ligand for the receptor tyrosine kinase Met, is secreted as single chain pro-HGF that lacks signaling activity. Pro-HGF acquires functional competence upon cleavage between R494 and V495, generating a disulfide-linked alpha/beta-heterodimer, where the beta-chain of HGF (HGF beta) has a serine protease fold that lacks enzymatic activity. We show that, like serine proteases, insertion of the newly formed N terminus in the beta-chain is critical for activity, here by allosterically stabilizing interactions with Met. The HGF beta crystal structure shows that V495 inserts into the "activation pocket" near the Met binding site where the positively charged N terminus forms a salt bridge with the negatively charged D672, and the V495 side chain has hydrophobic interactions with main- and side-chain residues. Full-length two-chain HGF mutants designed to interrupt these interactions (D672N, V495G, V495A, G498I, and G498V) displayed <10% activity in Met receptor phosphorylation, cell migration, and proliferation assays. Impaired signaling of full-length mutants correlated with >50-fold decreases in Met binding of the low-affinity HGF beta domain alone bearing the same mutations and further correlated with impaired N-terminal insertion. Because high-affinity binding resides in the HGF alpha-chain, full-length mutants maintained normal Met binding and efficiently inhibited HGF-mediated Met activation. Conversion of HGF from agonist to antagonist was achieved by as little as removal of two methyl groups (V495A) or a single charge (D672N). Thus, although serine proteases and HGF have quite distinct functions in proteolysis and Met signal transduction, respectively, they share a similar activation mechanism.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Engenharia de Proteínas/métodos , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Serina Endopeptidases/metabolismo , Sítio Alostérico , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Cristalografia por Raios X , Humanos , Neoplasias/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-met/metabolismo , Serina Endopeptidases/química , Transdução de Sinais
10.
FEBS Lett ; 579(9): 1945-50, 2005 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-15792801

RESUMO

Hepsin, a type II transmembrane serine protease, is highly upregulated in prostate cancer and promotes tumor progression and metastasis. We generated a soluble form of hepsin comprising the entire extracellular domain to show that it efficiently converts single-chain hepatocyte growth factor (pro-HGF) into biologically active two-chain HGF. Hepsin activity was potently inhibited by soluble forms of the bi-Kunitz domain inhibitors HAI-1B (IC(50) 21.1+/-2.7 nM) and HAI-2 (IC(50) 1.3+/-0.3 nM). Enzymatic assays with HAI-1B Kunitz domain mutants (R260A and K401A) further demonstrated that inhibition was due to Kunitz domain-1. The results suggest a functional link between hepsin and the HGF/Met pathway, which may contribute to tumor progression.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Glicoproteínas de Membrana/fisiologia , Neoplasias da Próstata/metabolismo , Precursores de Proteínas/metabolismo , Serina Endopeptidases/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Masculino , Glicoproteínas de Membrana/farmacologia , Precursores de Proteínas/efeitos dos fármacos , Proteínas Secretadas Inibidoras de Proteinases , Serina Endopeptidases/farmacologia , Inibidor da Tripsina de Soja de Kunitz/farmacologia
11.
J Biol Chem ; 279(38): 39915-24, 2004 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-15218027

RESUMO

Hepatocyte growth factor (HGF), a plasminogen-related growth factor, is the ligand for Met, a receptor tyrosine kinase implicated in development, tissue regeneration, and invasive tumor growth. HGF acquires signaling activity only upon proteolytic cleavage of single-chain HGF into its alpha/beta heterodimer, similar to zymogen activation of structurally related serine proteases. Although both chains are required for activation, only the alpha-chain binds Met with high affinity. Recently, we reported that the protease-like HGF beta-chain binds to Met with low affinity (Stamos, J., Lazarus, R. A., Yao, X., Kirchhofer, D., and Wiesmann, C. (2004) EMBO J. 23, 2325-2335). Here we demonstrate that the zymogen-like form of HGF beta also binds Met, albeit with 14-fold lower affinity than the protease-like form, suggesting optimal interactions result from conformational changes upon cleavage of the single-chain form. Extensive mutagenesis of the HGF beta region corresponding to the active site and activation domain of serine proteases showed that 17 of the 38 purified two-chain HGF mutants resulted in impaired cell migration or Met phosphorylation but no loss in Met binding. However, reduced biological activities were well correlated with reduced Met binding of corresponding mutants of HGF beta itself in assays eliminating dominant alpha-chain binding contributions. Moreover, the crystal structure of HGF beta determined at 2.53 A resolution provides a structural context for the mutagenesis data. The functional Met binding site is centered on the "active site region" including "triad" residues Gln(534) [c57], Asp(578) [c102], and Tyr(673) [c195] and neighboring "activation domain" residues Val(692), Pro(693), Gly(694), Arg(695), and Gly(696) [c214-c219]. Together they define a region that bears remarkable resemblance to substrate processing regions of serine proteases. Models of HGF-dependent Met receptor activation are discussed.


Assuntos
Fator de Crescimento de Hepatócito/química , Fator de Crescimento de Hepatócito/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Neoplasias da Mama , Células CHO , Linhagem Celular Tumoral/citologia , Movimento Celular/fisiologia , Cricetinae , Cristalografia , Dimerização , Fator de Crescimento de Hepatócito/genética , Humanos , Insetos , Dados de Sequência Molecular , Mutagênese , Fosforilação , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-met/química , Serina Endopeptidases/metabolismo
12.
Biochemistry ; 43(5): 1223-9, 2004 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-14756558

RESUMO

Tissue factor (TF) binds the zymogen (VII) and activated (VIIa) forms of coagulation factor VII with high affinity. The structure determined for the sTF-VIIa complex [Banner, D. W., et al. (1996) Nature 380, 41-46] shows that all four domains of VIIa (Gla, EGF-1, EGF-2, and protease) are in contact with TF. Although a structure is not available for the TF-VII complex, the structure determined for free VII [Eigenbrot, C., et al. (2001) Structure 9, 675-682] suggests a significant conformational change for the zymogen to enzyme transition. In particular, the region of the protease domain that must contact TF has a conformation that is altered from that of VIIa, suggesting that the VII protease domain interacts with TF in a manner different from that of VIIa. To test this hypothesis, a panel of 12 single-site sTF mutants, having substitutions of residues observed to contact the proteolytic domain of VIIa, have been evaluated for binding to both zymogen VII and VIIa. Affinities were determined by surface plasmon resonance measurements using a noninterfering anti-TF monoclonal antibody to capture TF on the sensor chip surface. Dissociation constants (K(D)) measured for binding to wild-type sTF are 7.5 +/- 2.4 nM for VII and 5.1 +/- 2.3 nM for VIIa. All of the sTF mutants except S39A and E95A exhibited a significant decrease (>2-fold) in affinity for VIIa. The changes in affinity measured for VII or VIIa binding with substitution in sTF were comparable in magnitude. We conclude that the proteolytic domain of both VII and VIIa interacts with this region of sTF in a nearly identical fashion. Therefore, zymogen VII can readily adopt a VIIa-like conformation required for binding to TF.


Assuntos
Fator VII/química , Fator VIIa/química , Tromboplastina/química , Regulação Alostérica/genética , Substituição de Aminoácidos/genética , Sítios de Ligação/genética , Linhagem Celular , Endopeptidases/química , Endopeptidases/metabolismo , Ativação Enzimática , Precursores Enzimáticos/isolamento & purificação , Precursores Enzimáticos/metabolismo , Fator VII/isolamento & purificação , Fator VII/metabolismo , Fator VIIa/metabolismo , Fator X/química , Fator X/metabolismo , Humanos , Hidrólise , Mutagênese Sítio-Dirigida , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidade , Ressonância de Plasmônio de Superfície , Tromboplastina/genética , Tromboplastina/metabolismo
13.
J Mol Biol ; 331(2): 433-46, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12888350

RESUMO

6A6 is a murine monoclonal antibody raised against the humanized anti-tissue factor antibody D3H44. 6A6 is able to completely neutralize the anticoagulant activity of D3H44 in tissue factor-dependent functional assays, such as endotoxin-induced whole blood clotting, prothrombin time, as well as factor X and factor IX activation. ELISA-type assays further showed that 6A6 binds to an epitope with critical determinants on the V(L) domain of D3H44. The possibility that the anti-idiotypic 6A6 might carry an "internal image" of the original antigen (tissue factor) was examined using the X-ray structure of the 6A6-Fab/D3H44-Fab complex determined at 2.5A resolution. We find that 6A6 structurally mimics tissue factor only so far as it combines with the antigen recognition surface of D3H44. While 6A6 contacts both V(L) and V(H) domains of D3H44, as does tissue factor, there is more contact with the D3H44 V(L) domain and less with the D3H44 V(H) domain relative to the tissue factor contacts on D3H44. Additionally, there is an almost total lack of correspondence between 6A6 and tissue factor at the level of amino acid side-chain functional groups. Despite the fact that both tissue factor and 6A6 are composed largely of beta-sheets, they present fundamentally different elements of secondary structure to D3H44; tissue factor presents beta-sheets edge-on, while 6A6 uses mostly loops. Finally, the finding that 6A6 competes with tissue factor for D3H44 binding raises the possibility of using 6A6 as an antidote for D3H44 anticoagulant therapy. To this end, we constructed a chimeric murine/human 6A6-Fab, which effectively neutralized D3H44 and fully restored tissue factor function in enzymatic assays.


Assuntos
Anticorpos Monoclonais/química , Tromboplastina/imunologia , Animais , Antígenos/química , Cristalografia por Raios X , Relação Dose-Resposta Imunológica , Endotoxinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Epitopos , Fator IX/metabolismo , Fator X/metabolismo , Humanos , Fragmentos de Imunoglobulinas/química , Camundongos , Modelos Moleculares , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Tempo de Protrombina , Proteínas Recombinantes de Fusão/química , Tromboplastina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA