Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biochem ; 174(1): 47-58, 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-36805939

RESUMO

The lipopolysaccharide (LPS)-triggered horseshoe crab coagulation cascade is composed of three protease zymogens, prochelicerase C (proC), prochelicerase B (proB) and the proclotting enzyme (proCE). In this study, we found that Ca 2+ ions increase the production of the clotting enzyme as a result of a cascade reaction reconstituted by recombinant proteins of wild-type (WT) proC, WT proB and WT proCE. We divided the cascade into three stages: autocatalytic activation of WT proC on the surface of LPS into WT α-chelicerase C (Stage 1); activation of WT proB on the surface of LPS into WT chelicerase B by WT α-chelicerase C (Stage 2) and activation of WT proce into WT CE by chelicerase B (Stage 3). Ca2+ ions enhanced the proteolytic activation in Stage 2, but not those in Stages 1 and 3. Moreover, we performed isothermal titration calorimetry to clarify the interaction of LPS or the recombinant zymogens with Ca2+ ions. LPS interacted with Ca2+ ions at an association constant of Ka = 4.7 × 104 M-1, but not with any of the recombinant zymogens. We concluded that LPS bound with Ca2+ ions facilitates the chain reaction of the cascade as a more efficient scaffold than LPS itself.


Assuntos
Caranguejos Ferradura , Lipopolissacarídeos , Animais , Lipopolissacarídeos/metabolismo , Cálcio/metabolismo , Coagulação Sanguínea , Precursores Enzimáticos/metabolismo
2.
Dev Comp Immunol ; 135: 104491, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35850280

RESUMO

The concept of a chain reaction of proteolytic activation of multiple protease zymogens was first proposed to explain the blood clotting system in mammals as an enzyme cascade. In multicellular organisms, similar enzyme cascades are widely present in signal transduction and amplification systems. The initiation step of the blood coagulation cascade often consists of autocatalytic activation of the corresponding zymogens located on the surfaces of host- or foreign-derived substances at injured sites. However, the molecular mechanism underlying the concept of autocatalytic activation remains speculative. In this review, we will focus on the autocatalytic activation of prochelicerase C on the surface of lipopolysaccharide as a potential initiator of hemolymph coagulation in horseshoe crabs. Prochelicerase C is presumed to have evolved from a common complement factor in Chelicerata; thus, evolutionary insights into the hemolymph coagulation and complement systems in horseshoe crabs will also be discussed.


Assuntos
Precursores Enzimáticos , Caranguejos Ferradura , Sequência de Aminoácidos , Animais , Precursores Enzimáticos/metabolismo , Hemolinfa/metabolismo , Lipopolissacarídeos , Mamíferos , Peptídeo Hidrolases , Serina Endopeptidases/metabolismo
3.
J Biochem ; 170(4): 489-500, 2021 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-34037771

RESUMO

The lipopolysaccharide (LPS)-triggered coagulation cascade in horseshoe crabs is composed of three zymogens belonging to the trypsinogen family: prochelicerase C, prochelicerase B (proB) and the proclotting enzyme (proCE). Trypsinogen-family members contain three conserved disulphides located around the active site. While it is known that proB evolutionarily lost one of the disulphides, the His-loop disulphide, the roles of the missing His-loop disulphide in proB remain unknown. Here, we prepared a proB mutant, named proB-murasame, equipped with a regenerated His-loop disulphide. The activation rate by upstream α-chelicerase C for proB-murasame was indistinguishable from that for wild-type (WT) proB. The resulting protease chelicerase B-murasame exhibited an 8-fold higher kcat value for downstream proCE than WT chelicerase B, whereas the Km value of chelicerase B-murasame was equivalent to that of WT chelicerase B. WT serpins-1, -2 and -3, identified as scavengers for the cascade, had no reactivity against WT chelicerase B, whereas chelicerase B-murasame was inhibited by WT serpin-2, suggesting that WT chelicerae B may trigger as-yet-unsolved phenomena after performing its duty in the cascade. The reconstituted LPS-triggered cascade containing proB-murasame exhibited ∼5-fold higher CE production than that containing WT proB. ProB-murasame might be used as a high value-adding reagent for LPS detection.


Assuntos
Proteínas de Artrópodes/metabolismo , Coagulação Sanguínea , Dissulfetos/metabolismo , Precursores Enzimáticos/metabolismo , Caranguejos Ferradura/enzimologia , Serina Proteases/metabolismo , Animais , Domínio Catalítico , Endopeptidases/metabolismo , Ativação Enzimática , Histonas/metabolismo , Lipopolissacarídeos/metabolismo , Serpinas/metabolismo , Tripsinogênio/metabolismo
4.
J Biol Chem ; 295(26): 8857-8866, 2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32409575

RESUMO

The lipopolysaccharide (LPS)-triggered coagulation cascade in horseshoe crabs comprises three protease zymogens: prochelicerase C (proC), prochelicerase B (proB), and the proclotting enzyme (proCE). The presence of LPS results in autocatalytic activation of proC to α-chelicerase C, which, in turn, activates proB to chelicerase B, converting proCE to the clotting enzyme (CE). ProB and proCE contain an N-terminal clip domain, but the roles of these domains in this coagulation cascade remain unknown. Here, using recombinant proteins and kinetics and binding assays, we found that five basic residues in the clip domain of proB are required to maintain its LPS-binding activity and activation by α-chelicerase C. Moreover, an amino acid substitution at a potential hydrophobic cavity in proB's clip domain (V55A-proB) reduced both its LPS-binding activity and activation rate. WT proCE exhibited no LPS-binding activity, and the WT chelicerase B-mediated activation of a proCE variant with a substitution at a potential hydrophobic cavity (V53A-proCE) was ∼4-fold slower than that of WT proCE. The kcat/Km value of the interaction of WT chelicerase B with V53A-proCE was 7-fold lower than that of the WT chelicerase B-WT proCE interaction. The enzymatic activities of V55A-chelicerase B and V53A-CE against specific peptide substrates were indistinguishable from those of the corresponding WT proteases. In conclusion, the clip domain of proB recruits it to a reaction center composed of α-chelicerase C and LPS, where α-chelicerase C is ready to activate proB, leading to chelicerase B-mediated activation of proCE via its clip domain.


Assuntos
Proteínas de Artrópodes/metabolismo , Caranguejos Ferradura/fisiologia , Peptídeo Hidrolases/metabolismo , Animais , Proteínas de Artrópodes/química , Coagulação Sanguínea , Endopeptidases/química , Endopeptidases/metabolismo , Ativação Enzimática , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Lipopolissacarídeos , Modelos Moleculares , Peptídeo Hidrolases/química , Domínios Proteicos
5.
Methods Mol Biol ; 2132: 277-283, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32306335

RESUMO

Tachylectin-5, a 41-kDa protein with a common fold of the C-terminal globular domain of the γ-chain of fibrinogen, is purified from horseshoe crab hemolymph plasma by affinity column chromatography, using acetyl-group-immobilized resin. Two types of isolectins, tachylectin-5A and tachylectin-5B, are obtained by stepwise elution with GlcNAc at 25 and 250 mM, respectively. Tachylectins-5A and -5B exhibit extraordinarily strong hemagglutinating activity against all types of human erythrocytes (the minimum agglutinating concentration of 0.004-0.008 µg/mL for tachylectin-5A and 0.077-0.27 µg/mL for tachylectin-5B). Their hemagglutinating activities are inhibited by acetyl group-containing sugars and noncarbohydrates such as sodium acetate, acetylcholine, and acetyl CoA (the minimum inhibitory concentrations of 1.3-1.6 mM), indicating that the acetyl group is required and sufficient for recognition by tachylectins-5A and -5B. EDTA inhibits their hemagglutinating activity, whereas the inhibition is overcome by adding an excess amount of Ca2+. Tachylectins-5A and -5B also exhibit bacterial agglutinating activity against both Gram-negative bacteria (the minimum agglutinating concentrations of 0.04-0.08 µg/mL for tachylectin-5A and 0.05-0.11 µg/mL for tachylectin-5B) and Gram-positive bacteria (the minimum agglutinating concentrations of 0.3-2.4 µg/mL for tachylectin-5A and 15.1-26.8 µg/mL for tachylectin-5B). Interestingly, tachylectins-5A and -5B enhance the antimicrobial activity of a hemocyte-derived peptide, big defensin.


Assuntos
Proteínas Sanguíneas/isolamento & purificação , Proteínas Sanguíneas/farmacologia , Caranguejos Ferradura/metabolismo , Lectinas/isolamento & purificação , Lectinas/farmacologia , Acetilglucosamina/metabolismo , Animais , Proteínas Sanguíneas/efeitos dos fármacos , Cromatografia de Afinidade , Ácido Edético/efeitos adversos , Eritrócitos/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Testes de Hemaglutinação , Hemolinfa/metabolismo , Humanos , Lectinas/efeitos dos fármacos
6.
Methods Mol Biol ; 2132: 309-316, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32306338

RESUMO

Tachylectin-2, a 27-kDa protein consisting of a five-bladed ß-propeller structure, is purified by three steps of chromatography, including dextran sulfate-Sepharose CL-6B, CM-Sepharose CL-6B, and Mono S. Three isolectins of tachylectin-2 including tachylectin-2a, -2b, and -2c are purified. These isolectins exhibit hemagglutinating activity against human A-type erythrocytes in a Ca2+-independent manner with tachylectin-2b showing the highest activity. Tachylectin-2b specifically agglutinates Staphylococcus saprophyticus KD. The tachylectin-2b-mediated hemagglutination is inhibited in the presence of GlcNAc and GalNAc. The association constants for GlcNAc and GalNAc are Ka = 1.95 × 104 M-1 and Ka = 1.11 × 103 M-1, respectively. Ultracentrifugation analysis shows that tachylectin-2b is present in monomer form in solution.


Assuntos
Caranguejos Ferradura/metabolismo , Lectinas/isolamento & purificação , Lectinas/farmacologia , Acetilgalactosamina/farmacologia , Acetilglucosamina/farmacologia , Testes de Aglutinação , Animais , Cálcio/metabolismo , Cromatografia , Eritrócitos/efeitos dos fármacos , Hemaglutinação/efeitos dos fármacos , Caranguejos Ferradura/química , Humanos , Lectinas/química , Isoformas de Proteínas/química , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/farmacologia , Multimerização Proteica , Staphylococcus saprophyticus/efeitos dos fármacos
7.
Methods Mol Biol ; 2132: 317-323, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32306339

RESUMO

An antimicrobial peptide tachycitin (73 amino acids) is purified by steps of chromatography, including Sephadex G-50 and S Sepharose FF, from the acid extract of hemocyte debris of horseshoe crabs. Tachycitin is present in monomer form in solution, revealed by ultracentrifugation analysis. Tachycitin exhibits bacterial agglutination activity and inhibits the growth of both Gram-negative bacteria, Gram-positive bacteria, and fungus Candida albicans. Interestingly, tachycitin shows synergistic antimicrobial activity in corporation with another antimicrobial peptide, big defensin. Tachycitin shows a specific binding activity to chitin but not to cellulose, mannan, xylan, and laminarin. Tachycitin is composed of the N-terminal three-stranded ß-sheet and the C-terminal two-stranded ß-sheet following a short helical turn, and the C-terminal structural motif shares a significant structural similarity with the chitin-binding domain derived from a plant chitin-binding protein, hevein.


Assuntos
Proteínas Sanguíneas/isolamento & purificação , Proteínas Sanguíneas/farmacologia , Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/farmacologia , Quitina/metabolismo , Caranguejos Ferradura/metabolismo , Testes de Aglutinação , Animais , Sítios de Ligação , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Candida albicans/efeitos dos fármacos , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Cromatografia , Defensinas/farmacologia , Dextranos/química , Sinergismo Farmacológico , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Modelos Moleculares , Estrutura Secundária de Proteína , Sefarose/química , Especificidade por Substrato
8.
J Biol Chem ; 293(29): 11589-11599, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29866883

RESUMO

Horseshoe crab hemolymph coagulation is believed to be triggered by the autocatalytic activation of serine protease zymogen factor C to the active form, α-factor C, belonging to the trypsin family, through an active transition state of factor C responding to bacterial lipopolysaccharide (LPS), designated factor C*. However, the existence of factor C* is only speculative, and its proteolytic activity has not been validated. In addition, it remains unclear whether the proteolytic cleavage of the Phe737-Ile738 bond (Phe737 site) of factor C required for the conversion to α-factor C occurs intramolecularly or intermolecularly between the factor C molecules. Here we show that the Phe737 site of a catalytic Ser-deficient mutant of factor C is LPS-dependently hydrolyzed by a Phe737 site-uncleavable mutant, clearly indicating the existence of the active transition state of factor C without cleavage of the Phe737 site. Moreover, we found the following facts using several mutants of factor C: the autocatalytic cleavage of factor C occurs intermolecularly between factor C* molecules on the LPS surface; factor C* does not exhibit intrinsic chymotryptic activity against the Phe737 site, but it may recognize a three-dimensional structure around the cleavage site; and LPS is required not only to complete the substrate-binding site and oxyanion hole of factor C* by interacting with the N-terminal region but also to allow the Phe737 site to be cleaved by inducing a conformational change around the Phe737 site or by acting as a scaffold to induce specific protein-protein interactions between factor C* molecules.


Assuntos
Proteínas de Artrópodes/metabolismo , Precursores Enzimáticos/metabolismo , Caranguejos Ferradura/enzimologia , Lipopolissacarídeos/metabolismo , Serina Endopeptidases/metabolismo , Animais , Proteínas de Artrópodes/química , Domínio Catalítico , Ativação Enzimática , Precursores Enzimáticos/química , Células HEK293 , Caranguejos Ferradura/química , Caranguejos Ferradura/metabolismo , Humanos , Conformação Proteica , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/química
9.
J Biol Chem ; 292(15): 6369-6380, 2017 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-28258224

RESUMO

In Drosophila, the final immune deficiency (IMD) pathway-dependent signal is transmitted through proteolytic conversion of the nuclear factor-κB (NF-κB)-like transcription factor Relish to the active N-terminal fragment Relish-N. Relish-N is then translocated from the cytosol into the nucleus for the expression of IMD-controlled genes. We previously demonstrated that transglutaminase (TG) suppresses the IMD pathway by polymerizing Relish-N to inhibit its nuclear translocation. Conversely, we also demonstrated that orally ingested synthetic amines, such as monodansylcadaverine (DCA) and biotin-labeled pentylamine, are TG-dependently incorporated into Relish-N, causing the nuclear translocation of modified Relish-N in gut epithelial cells. It remains unclear, however, whether polyamine-containing Relish-N retains transcriptional activity. Here, we used mass spectrometry analysis of a recombinant Relish-N modified with DCA by TG activity after proteolytic digestion and show that the DCA-modified Gln residues are located in the DNA-binding region of Relish-N. TG-catalyzed DCA incorporation inhibited binding of Relish-N to the Rel-responsive element in the NF-κB-binding DNA sequence. Subcellular fractionation of TG-expressing Drosophila S2 cells indicated that TG was localized in both the cytosol and nucleus. Of note, natural polyamines, including spermidine and spermine, competitively inhibited TG-dependent DCA incorporation into Relish-N. Moreover, in vivo experiments demonstrated that Relish-N was modified by spermine and that this modification reduced transcription of IMD pathway-controlled cecropin A1 and diptericin genes. These findings suggest that intracellular TG regulates Relish-N-mediated transcriptional activity by incorporating polyamines into Relish-N and via protein-protein cross-linking.


Assuntos
Cadaverina/análogos & derivados , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Fatores de Transcrição/metabolismo , Transglutaminases/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Cadaverina/metabolismo , Linhagem Celular , Núcleo Celular/genética , Citosol/metabolismo , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Drosophila melanogaster , Células Epiteliais/citologia , Intestinos/citologia , Domínios Proteicos , Fatores de Transcrição/genética , Transglutaminases/genética
10.
J Biol Chem ; 290(31): 19379-86, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26109069

RESUMO

Factor B is a serine-protease zymogen in the horseshoe crab coagulation cascade, and it is the primary substrate for activated factor C, the LPS-responsive initiator of the cascade. Factor C is autocatalytically activated to α-factor C on LPS and is artificially converted to ß-factor C, another activated form, by chymotrypsin. It is not known, however, whether LPS is required for the activation of factor B. Here we found that wild-type factor B expressed in HEK293S cells is activated by α-factor C, but not by ß-factor C, in an LPS-dependent manner and that ß-factor C loses the LPS binding activity of factor C through additional cleavage by chymotrypsin within the N-terminal LPS-binding region. Surface plasmon resonance and quartz crystal microbalance analyses revealed that wild-type factor B binds to LPS with high affinity comparable with that of factor C, demonstrating that factor B is the second LPS-binding zymogen in the cascade. An LPS-binding site of wild-type factor B was found in the N-terminal clip domain, and the activation rate of a clip domain deletion mutant was considerably slower than that of wild-type factor B. Moreover, in the presence of LPS, Triton X-100 inhibited the activation of wild-type factor B by α-factor C. We conclude that the clip domain of factor B has an important role in localizing factor B to the surface of Gram-negative bacteria or LPS released from bacteria to initiate effective proteolytic activation by α-factor C.


Assuntos
Proteínas de Artrópodes/química , Fator B do Complemento/química , Precursores Enzimáticos/química , Caranguejos Ferradura/enzimologia , Lipopolissacarídeos/química , Animais , Sítios de Ligação , Células HEK293 , Humanos , Ligação Proteica , Proteólise
11.
J Biol Chem ; 289(37): 25987-95, 2014 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-25077965

RESUMO

Factor C, a serine protease zymogen involved in innate immune responses in horseshoe crabs, is known to be autocatalytically activated on the surface of bacterial lipopolysaccharides, but the molecular mechanism of this activation remains unknown. In this study, we show that wild-type factor C expressed in HEK293S cells exhibits a lipopolysaccharide-induced activity equivalent to that of native factor C. Analysis of the N-terminal addition, deletion, or substitution mutants shows that the N-terminal Arg residue and the distance between the N terminus and the tripartite of lipopolysaccharide-binding site are essential factors for autocatalytic activation, and that the positive charge of the N terminus may interact with an acidic amino acid(s) of the molecule to convert the zymogen into an active form. Chemical cross-linking experiments indicate that the N terminus is required to form a complex of the factor C molecules in a sufficiently close vicinity to be chemically cross-linked on the surface of lipopolysaccharides. We propose a molecular mechanism of the autocatalytic activation of the protease zymogen on lipopolysaccharides functioning as a platform to induce specific protein-protein interaction between the factor C molecules.


Assuntos
Proteínas de Artrópodes/metabolismo , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Caranguejos Ferradura/enzimologia , Imunidade Inata/genética , Serina Proteases/genética , Serina Proteases/metabolismo , Sequência de Aminoácidos , Animais , Precursores Enzimáticos/biossíntese , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Lipopolissacarídeos/toxicidade , Serina Proteases/biossíntese
12.
PLoS One ; 7(5): e36783, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22611464

RESUMO

Complement C3 plays an essential role in the opsonization of pathogens in the mammalian complement system, whereas the molecular mechanism underlying C3 activation in invertebrates remains unknown. To understand the molecular mechanism of C3b deposition on microbes, we characterized two types of C2/factor B homologs (designated TtC2/Bf-1 and TtC2/Bf-2) identified from the horseshoe crab Tachypleus tridentatus. Although the domain architectures of TtC2/Bf-1 and TtC2/Bf-2 were identical to those of mammalian homologs, they contained five-repeated and seven-repeated complement control protein domains at their N-terminal regions, respectively. TtC2/Bf-1 and TtC2/Bf-2 were synthesized and glycosylated in hemocytes and secreted to hemolymph plasma, which existed in a complex with C3 (TtC3), and their activation by microbes was absolutely Mg(2+)-dependent. Flow cytometric analysis revealed that TtC3b deposition was Mg(2+)-dependent on Gram-positive bacteria or fungi, but not on Gram-negative bacteria. Moreover, this analysis demonstrated that Ca(2+)-dependent lectins (C-reactive protein-1 and tachylectin-5A) were required for TtC3b deposition on Gram-positive bacteria, and that a Ca(2+)-independent lectin (Tachypleus plasma lectin-1) was definitely indispensable for TtC3b deposition on fungi. In contrast, a horseshoe crab lipopolysaccharide-sensitive protease factor C was necessary and sufficient to deposit TtC3b on Gram-negative bacteria. We conclude that plasma lectins and factor C play key roles in microbe-specific TtC3b deposition in a C2/factor B-dependent or -independent manner.


Assuntos
Complemento C2/imunologia , Complemento C3b/imunologia , Fator B do Complemento/imunologia , Caranguejos Ferradura/imunologia , Animais , Clonagem Molecular , Ativação do Complemento/imunologia , Complemento C2/genética , Complemento C2/metabolismo , Complemento C3b/metabolismo , Fator B do Complemento/genética , Fator B do Complemento/metabolismo , DNA Complementar , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Fungos/imunologia , Bactérias Gram-Positivas/imunologia , Caranguejos Ferradura/microbiologia , Magnésio/metabolismo , Modelos Biológicos , Ligação Proteica/imunologia , Estrutura Terciária de Proteína
13.
J Immunol ; 183(6): 3810-8, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19710471

RESUMO

In the horseshoe crab, the recognition of beta-1,3-D-glucans by factor G triggers hemolymph coagulation. Factor G contains a domain of two tandem xylanase Z-like modules (Z1-Z2), each of which recognizes beta-1,3-D-glucans. To gain an insight into the recognition of beta-1,3-D-glucans from a structural view point, recombinants of Z1-Z2, the C-terminal module Z2, Z2 with a Cys to Ala substitution (Z2A), and its tandem repeat Z2A-Z2A were characterized. Z2 and Z1-Z2, but not Z2A and Z2A-Z2A, formed insoluble aggregates at higher concentrations more than approximately 30 and 3 microM, respectively. Z1-Z2 and Z2A-Z2A bound more strongly to an insoluble beta-1,3-D-glucan (curdlan) than Z2A. The affinity of Z2A for a soluble beta-1,3-D-glucan (laminarin) was equivalent to those of Z1-Z2, Z2A-Z2A, and native factor G, suggesting that the binding of a single xylanase Z-like module prevents the subsequent binding of another module to laminarin. Interestingly, Z2A as well as intact factor G exhibited fungal agglutinating activity, and fungi were specifically detected with fluorescently tagged Z2A by microscopy. The chemical shift perturbation of Z2A induced by the interaction with laminaripentaose was analyzed by nuclear magnetic resonance spectroscopy. The ligand-binding site of Z2A was located in a cleft on a beta-sheet in a predicted beta-sandwich structure, which was superimposed onto cleft B in a cellulose-binding module of endoglucanase 5A from the soil bacterium Cellvibrio mixtus. We conclude that the pattern recognition for beta-1,3-D-glucans by factor G is accomplished via a carbohydrate-binding cleft that is evolutionally conserved between horseshoe crab and bacteria.


Assuntos
Cellvibrio/química , Evolução Molecular , Caranguejos Ferradura/química , Lectinas/genética , beta-Glucanas/metabolismo , Animais , Sítios de Ligação , Celulase/química , Cellvibrio/enzimologia , Sequência Conservada , Endo-1,4-beta-Xilanases , Glucanos , Polissacarídeos/metabolismo , Proteoglicanas
14.
J Immunol ; 181(11): 7994-8001, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19017991

RESUMO

The complement system in vertebrates plays an important role in host defense against and clearance of invading microbes, in which complement component C3 plays an essential role in the opsonization of pathogens, whereas the molecular mechanism underlying C3 activation in invertebrates remains unknown. In an effort to understand the molecular activation mechanism of invertebrate C3, we isolated and characterized an ortholog of C3 (designated TtC3) from the horseshoe crab Tachypleus tridentatus. Flow cytometric analysis using an Ab against TtC3 revealed that the horseshoe crab complement system opsonizes both Gram-negative and Gram-positive bacteria. Evaluation of the ability of various pathogen-associated molecular patterns to promote the proteolytic conversion of TtC3 to TtC3b in hemocyanin-depleted plasma indicated that LPS, but not zymosan, peptidoglycan, or laminarin, strongly induces this conversion, highlighting the selective response of the complement system to LPS stimulation. Although originally characterized as an LPS-sensitive initiator of hemolymph coagulation stored within hemocytes, we identified factor C in hemolymph plasma. An anti-factor C Ab inhibited various LPS-induced phenomena, including plasma amidase activity, the proteolytic activation of TtC3, and the deposition of TtC3b on the surface of Gram-negative bacteria. Moreover, activated factor C present on the surface of Gram-negative bacteria directly catalyzed the proteolytic conversion of the purified TtC3, thereby promoting TtC3b deposition. We conclude that factor C acts as an LPS-responsive C3 convertase on the surface of invading Gram-negative bacteria in the initial phase of horseshoe crab complement activation.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Convertases de Complemento C3-C5/imunologia , Precursores Enzimáticos/imunologia , Caranguejos Ferradura/imunologia , Lipopolissacarídeos/farmacologia , Serina Endopeptidases/imunologia , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Positivas/imunologia , Hemócitos/enzimologia , Hemócitos/imunologia , Hemolinfa/enzimologia , Hemolinfa/imunologia , Caranguejos Ferradura/enzimologia , Caranguejos Ferradura/genética , Dados de Sequência Molecular , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Especificidade por Substrato/genética , Especificidade por Substrato/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA