Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Ecotoxicol Environ Saf ; 272: 116080, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38350215

RESUMO

BACKGROUND: Serum prostate-specific antigen (PSA) is a primary metric for diagnosis and prognosis of prostate cancer (PCa). Exposure to heavy metals, such as lead, cadmium, mercury, and zinc can impact PSA levels in PCa patients. However, it is unclear whether this effect also occurs in men without PCa, which may lead to the overdiagnosis of PCa. METHOD: Data on a total of 5089 American men who had never been diagnosed with PCa were obtained from the National Health and Nutrition Examination Survey performed from 2003-2010. The relationship between serum PSA levels (dependent variable) and concentrations of lead (µmol/L), cadmium (nmol/L), and mercury (µmol/L) were investigated with dietary zinc intake being used as a potential modifier or covariate in a weighted linear regression model and a generalized additive model. A series of bootstrapping analyses were performed to evaluate sensitivity and specificity using these models. RESULTS: Regression analyses suggested that, in general, lead, cadmium, or mercury did not show an association with PSA levels, which was consistent with the results of the bootstrapping analyses. However, in a subgroup of participants with a high level of dietary zinc intake (≥14.12 mg/day), a significant positive association between cadmium and serum PSA was identified (1.06, 95% CI, P = 0.0268, P for interaction=0.0249). CONCLUSIONS: With high-level zinc intake, serum PSA levels may rise in PCa-free men as the exposure to cadmium increases, leading to a potential risk of an overdiagnosis of PCa and unnecessary treatment. Therefore, environmental variables should be factored in the current diagnostic model for PCa that is solely based on PSA measurements. Different criteria for PSA screening are necessary based on geographical variables. Further investigations are needed to uncover the biological and biochemical relationship between zinc, cadmium, and serum PSA levels to more precisely diagnose PCa.


Assuntos
Mercúrio , Metais Pesados , Masculino , Humanos , Estados Unidos , Antígeno Prostático Específico , Cádmio , Inquéritos Nutricionais , Zinco
2.
Nat Med ; 11(12): 1295-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16311604

RESUMO

We transplanted kidneys from alpha1,3-galactosyltransferase knockout (GalT-KO) pigs into six baboons using two different immunosuppressive regimens, but most of the baboons died from severe acute humoral xenograft rejection. Circulating induced antibodies to non-Gal antigens were markedly elevated at rejection, which mediated strong complement-dependent cytotoxicity against GalT-KO porcine target cells. These data suggest that antibodies to non-Gal antigens will present an additional barrier to transplantation of organs from GalT-KO pigs to humans.


Assuntos
Animais Geneticamente Modificados , Rejeição de Enxerto/imunologia , Tolerância Imunológica/imunologia , Transplante de Rim/imunologia , Transplante Heterólogo/mortalidade , Transplante Heterólogo/métodos , Animais , Anticorpos/sangue , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Terapia de Imunossupressão/métodos , Papio , Sus scrofa/genética , Linfócitos T/imunologia , Transplante Heterólogo/imunologia
3.
Toxicon ; 52(2): 277-84, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18590752

RESUMO

A number of inactive serine protease homologues (SPHs), which have poorly understood functions, have been identified in invertebrates and vertebrates. Recently, several SPH transcripts have been reported from snake venom glands, which provide potential new tools for the study of the functions of SPHs. Herein we report for the first time a snake venom serine protease homologue (svSPH) protein, designated as TjsvSPH, isolated from the venom of Trimeresurus jerdonii. Despite its high sequence similarity to snake venom serine proteases (SVSPs), TjsvSPH is devoid of arginine esterase and proteolytic activity. This is probably due to the replacement of Arg-43 by His-43 in the catalytic triad. TjsvSPH did not influence the coagulation time of human plasma, induce human platelet aggregation, inhibit adenosine diphosphate/thrombin-induced human platelet aggregation or increase capillary permeability. Phylogenetic analysis showed that svSPHs were separated from SVSPs and formed an independent group. Structural analysis revealed that the structures of svSPHs are quite different from those of SPHs previously reported. These results indicate that snake venoms contain a unique group of svSPH proteins.


Assuntos
Venenos de Crotalídeos/química , Venenos de Crotalídeos/enzimologia , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Trimeresurus/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Coagulação Sanguínea/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Fracionamento Químico , Venenos de Crotalídeos/farmacologia , Humanos , Dados de Sequência Molecular , Filogenia , Agregação Plaquetária/efeitos dos fármacos , Alinhamento de Sequência , Análise de Sequência de DNA , Serina Endopeptidases/farmacologia
4.
Biochim Biophys Acta ; 1760(3): 462-71, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16413680

RESUMO

A novel phospholipase A2 (PLA2) with Asn at its site 49 was purified from the snake venom of Protobothrops mucrosquamatus by using SP-Sephadex C25, Superdex 75, Heparin-Sepharose (FF) and HPLC reverse-phage C18 chromatography and designated as TM-N49. It showed a molecular mass of 13.875 kDa on MALDI-TOF. TM-N49 does not possess enzymatic, hemolytic and hemorrhagic activities. It fails to induce platelet aggregation by itself, and does not inhibit the platelet aggregation induced by ADP. However, it exhibits potent myotoxic activity causing inflammatory cell infiltration, severe myoedema, myonecrosis and myolysis in the gastrocnemius muscles of BALB/c mice. Phylogenetic analysis found that that TM-N49 combined with two phospholipase A2s from Trimeresurus stejnegeri, TsR6 and CTs-R6 cluster into one group. Structural and functional analysis indicated that these phospholipase A2s are distinct from the other subgroups (D49 PLA2, S49 PLA2 and K49 PLA2) and represent a unique subgroup of snake venom group II PLA2, named N49 PLA2 subgroup.


Assuntos
Venenos de Crotalídeos/isolamento & purificação , Fosfolipases A/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Venenos de Crotalídeos/química , Venenos de Crotalídeos/farmacologia , Fosfolipases A2 do Grupo II , Camundongos , Dados de Sequência Molecular , Peso Molecular , Músculo Esquelético/efeitos dos fármacos , Fosfolipases A/química , Fosfolipases A/farmacologia , Fosfolipases A2 , Filogenia , Agregação Plaquetária/efeitos dos fármacos , Coelhos , Proteínas de Répteis , Alinhamento de Sequência , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Trimeresurus
5.
Int J Biochem Cell Biol ; 38(4): 510-20, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16310401

RESUMO

Poisonous snakebite wound is a popular disease worldwide. However, the pathogenesis remains unclear. In the present study, a novel metalloproteinase atrahagin in Chinese cobra (Naja atra) snake venom was purified, using heparin-sepharose followed by Superdex 75 gel filtration chromatography. Apart from its alpha-fibrinogenase activity, atrahagin potently activated human colon, lung and tonsil mast cells with the net histamine release being 25.9+/-4.4, 17.0+/-1.9, 13.2+/-3.6%, respectively. Time course studies revealed that the peak histamine release induced by atrahagin occurred at 12, 12 and 8 min following incubation of the enzyme with colon, lung and tonsil mast cells, respectively. The response of mast cells to atrahagin was abolished by preincubation of the cells with metabolic inhibitors or pertussis toxin, and by removal of Ca2+ and Mg2+ from the challenge buffer. In conclusion, activation of human mast cells by atrahagin indicated that the enzyme might contribute to the pathogenesis of snakebite wound.


Assuntos
Venenos Elapídicos/química , Liberação de Histamina/efeitos dos fármacos , Mastócitos/metabolismo , Metaloproteases/farmacologia , Proteínas de Répteis/farmacologia , Animais , Células Cultivadas , Venenos Elapídicos/farmacologia , Fibrinogênio/metabolismo , Humanos , Mastócitos/citologia , Metaloproteases/química , Metaloproteases/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Répteis/química , Proteínas de Répteis/isolamento & purificação , Fatores de Tempo
6.
Biochimie ; 88(10): 1331-42, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16793192

RESUMO

Group IIA phospholipase A(2) (PLA(2)) are major components in Viperidae/Crotalidae venom. In the present study, a novel PLA(2) named promutoxin with Arg at the site 49 has been purified from the venom of Protobothrops mucrosquamatus by chromatography. It consists of 122 amino acid residues with a molecular mass of 13,656 Da assessed by MALDI-TOF. It has the structural features of snake venom group IIA PLA(2)s, but has no PLA(2) enzymatic activity. Promutoxin shows higher amino acid sequence identity to the K49 PLA(2)s (72-95%) than to D49 PLA(2)s (52-58%). Promutoxin exhibits potent myotoxicity in the animal model with as little as 1 microg of promutoxin causing myonecrosis and myoedema in the gastrocnemius muscle of mice. Promutoxin is also able to stimulate the release of IL-12, TNFalpha, IL-6 and IL-1beta from human monocytes, and induce IL-2, TNFalpha and IL-6 release from T cells, indicating that this snake venom group IIA PLA(2) is actively involved in the inflammatory process in man caused by snake venom poisoning.


Assuntos
Venenos de Crotalídeos/enzimologia , Citocinas/metabolismo , Fosfolipases A/química , Fosfolipases A/farmacologia , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Arginina , Sequência de Bases , Humanos , Dados de Sequência Molecular , Peso Molecular , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Fosfolipases A/isolamento & purificação , Ratos , Ratos Wistar , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
7.
Transplantation ; 81(2): 273-83, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16436972

RESUMO

BACKGROUND: The present study was undertaken to determine the role of preformed and induced anti-non-Gal antibodies in the rejection of hDAF pig-to-baboon kidney xenotransplants after anti-Gal antibody neutralization therapy. METHODS: Seven baboons received life-supporting kidney transplants from hDAF transgenic pigs. Anti-Gal antibodies were neutralized by GAS914 or TPC (a Gal PEG glycoconjugate polymer). Group 1 (n=5) underwent a conventional immunosuppressive therapy with FK506, rabbit anti-thymocyte serum/immunoglobulin, mycophenolate mofetil, and steroids. Group 2 (n=2) received an anti-humoral immunity regimen with LF15-0195, Rituxan and cobra venom factor in addition to ATG, FK506 and steroids. Levels of anti-non-Gal antibodies and their mediated complement-dependent cytotoxic activities (CDC) were detected by flow cytometry using Gal knockout (k/o) pig lymphocytes (LC) or endothelial cells (EC) as targets. RESULTS: Continuous infusion of GAS914/TPC significantly reduced anti-Gal antibodies. In Group 1, four of five baboons developed severe acute humoral xenograft rejection (AHXR) and the rejection was associated with either a high level of preformed anti-non-Gal IgG or a marked elevation in induced anti-non-Gal IgG and IgM. Sera collected at the time of AHXR had a high level of CDC to porcine LC/EC from Gal k/o animals. The intensive anti-humoral therapy in Group 2 completely inhibited both anti-Gal and non-Gal antibody production and prevented AHXR. However, this therapy was not well tolerated by the baboons. CONCLUSION: In a pig-to-baboon kidney transplant model, both preformed and induced anti-non-Gal antibodies are strongly associated with the pathogenesis of AHXR when anti-Gal antibodies are neutralized.


Assuntos
Anticorpos Heterófilos/biossíntese , Rejeição de Enxerto/imunologia , Transplante de Rim/imunologia , Trissacarídeos/imunologia , Doença Aguda , Animais , Animais Geneticamente Modificados , Rejeição de Enxerto/etiologia , Transplante de Rim/efeitos adversos , Transplante de Rim/patologia , Testes de Neutralização , Papio , Sus scrofa , Transplante Heterólogo
8.
Toxicon ; 47(1): 104-12, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16330060

RESUMO

A novel C-type lectin-like protein, dabocetin, was purified from Daboia russellii siamensis venom. On SDS-polyacrylamide gel electrophoresis, it showed a single band with an apparent molecular weight of 28 kDa and two distinct bands with the apparent molecular weights of 15.0 kDa and 14.5 kDa under non-reducing and reducing conditions, respectively. cDNA clones containing the coding sequences for dabocetin alpha and beta subunits were isolated and sequenced. The deduced protein sequences of both subunits were confirmed by N-terminal amino acid sequencing and trypsin-digested peptide mass fingerprinting. Dabocetin did not induce platelet aggregation in platelet-rich plasma. It also had little effect on the platelet aggregation induced by ADP, TMVA or stejnulxin. Whereas, dabocetin inhibited ristocetin-induced platelet agglutination in platelet-rich plasma in a dose-dependent manner with an IC50 value of 0.35 microM. Flow cytometry analysis showed that dabocetin significantly inhibited mAb SZ2 binding to platelet membrane glycoprotein Ib alpha, indicating that platelet membrane glycoprotein Ib is involved in the inhibitory effect of dabocetin on ristocetin-induced platelet agglutination.


Assuntos
Lectinas Tipo C/antagonistas & inibidores , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Venenos de Víboras/química , Viperidae , Difosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Sequência de Bases , Clonagem Molecular , DNA Complementar/genética , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Concentração Inibidora 50 , Lectinas Tipo C/genética , Lectinas Tipo C/isolamento & purificação , Dados de Sequência Molecular , Peso Molecular , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Ristocetina/farmacologia , Venenos de Víboras/genética , Venenos de Víboras/farmacologia
9.
Zhonghua Yi Xue Za Zhi ; 86(21): 1460-3, 2006 Jun 06.
Artigo em Chinês | MEDLINE | ID: mdl-16842697

RESUMO

OBJECTIVE: To investigate the effect of Yunnan-cobra venom factor (Y-CVF) in overcoming acute humoral rejection after allograft cardiac transplantation in presensitized recipients. METHODS: Fifteen Lewis rats received the transplantation of full-thickness skin graft of BN rats three times so as to be presensitized. Fifteen pairs of Lewis rat, as recipients of heart, and BN rat, as heart donors, were randomly divided into 2 groups: experimental group (n = 8), and control group (n = 7). The Lewis rats in the experimental group received heart transplantation of the heart of the BN rat 7 approximately 10 days after the third skin transplantation, and were injected with Y-CVF 80 microg/kg 24 hours before the heart transplantation. The Lewis rat in the control group received only the heart transplantation without Y-CVF injection. Blood samples were collected from all rats before pre-sensitization and 7 days after the third skin transplantation so as to determine the titer of anti-BN rat lymphocyte antibody. 0 and 24 hours, and 6 and 8 days after Y-CVF injection blood samples were collected from the Lewis rats to determine the total complement activity with the complement activity before Y-CVF injection defined as 100%. The survival time of the transplanted heart was observed. After the transplanted hearts stopped to beat, they were resected and underwent HE staining and microscopy. Immunohistochemistry was used to examine the deposition of IgG and complement 3 (C3). RESULTS: The titer of anti-BN rat lymphocyte antibody was 0 before the pre-sensitization, and increased to 1:1028 - 1:2056 7 days after the third skin pre-sensitization. The serum total complement activity of the Lewis rats decreased to 0 twenty-four hours after the Y-CVF injection, recovered to 2.01% - 15.41% 6 days after, and returned to the normal level (89.61% - 109.46%) 8 days after. The mean survival time of the transplanted hearts of the control group was 12.71 +/- 13.94 hours (with a range of 1.5 - 15 hours), significantly shorter than that of the experimental group (99.50 +/- 38.72 hours, with a range of 23 - 153 hours, P < 0.01). Pathological examination showed that the acute humoral rejection in the control group was mainly complement-dependent antibody-mediated humoral rejection characterized by intravascular thrombosis, and that in the experimental group was mainly cellular rejection, characterized by extensive infiltration of mononuclear cells. Immunohistochemistry showed that remarkable IgG deposition was seen in the cardiac muscle cells and vascular endothelial cells in both groups; however, C3 deposition in vascular endothelial cells could be seen only in the control group. CONCLUSION: Administration of CVF is effective in overcoming the acute humoral rejection after allograft cardiac transplantation in presensitized recipients.


Assuntos
Venenos Elapídicos/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/métodos , Condicionamento Pré-Transplante/métodos , Animais , Imunoglobulina G/imunologia , Distribuição Aleatória , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Transplante de Pele/métodos , Transplante Homólogo
10.
Toxicon ; 45(3): 353-60, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15683874

RESUMO

A fibrinogen-clotting enzyme designed as jerdonobin-II was isolated from the venom of Trimeresurus jerdonii. It differed in molecular weight and N-terminal sequence with the previously isolated jerdonobin, a thrombin-like enzyme from the same venom. The enzyme consists of a single polypeptide chain with molecular weights of 30,000 and 32,000 under non-reducing and reducing conditions, respectively. Jerdonobin-II showed weak fibrinogen clotting activity and its activity unit on fibrinogen was calculated to be less than one unit using human thrombin as standard. The precursor protein sequence of jerodonobin-II was deduced from cloned cDNA sequence. The sequence shows high similarity (identity=89%) to TSV-PA, a specific plasminogen activator from venom of T. stejnegeri. Despite of the sequence similarity, jerdonobin-II was found devoid of plasminogen activating effect. Sequence alignment analysis suggested that the replacement of Lys239 in TSV-PA to Gln239 in jerdonobin-II might play an important role on their plasminogen activating activity difference.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Venenos de Crotalídeos/enzimologia , Fibrinogênio/química , Serina Endopeptidases/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/isolamento & purificação , Serina Endopeptidases/metabolismo , Especificidade por Substrato , Trimeresurus
11.
Toxicon ; 42(3): 257-62, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14559076

RESUMO

A highly active cobra venom factor (CVF) was isolated from the venom of Naja kaouthia by sequential column chromatography. It displays strong anticomplementary activity, and has 1515 U of anticomplementary activity per mg protein. A single dose of 0.1 mg/kg CVF given i.v. to rats completely abrogated complement activity for nearly 5 days. Given 0.02 mg/kg of CVF, the complement activity of rats was reduced by more than 96.5% in 6 h. In guinea pig-to-rat heart transplant model, rats treated with a single dose of 0.05 mg/kg CVF had significantly prolonged xenograft survival (56.12+/-6.27 h in CVF-treated rats vs. 0.19+/-0.07 h in control rats, P<0.001).


Assuntos
Proteínas Inativadoras do Complemento/farmacologia , Venenos Elapídicos/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Coração/fisiologia , Transplante Heterólogo/fisiologia , Animais , Fatores Biológicos/isolamento & purificação , Fatores Biológicos/farmacologia , Proteínas Inativadoras do Complemento/isolamento & purificação , Venenos Elapídicos/química , Rejeição de Enxerto/patologia , Cobaias , Masculino , Miocárdio/patologia , Ratos , Ratos Wistar
12.
Toxicon ; 42(5): 539-47, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14529736

RESUMO

Three 26 kDa proteins, named as TJ-CRVP, NA-CRVP1 and NA-CRVP2, were isolated from the venoms of Trimeresurus jerdonii and Naja atra, respectively. The N-terminal sequences of TJ-CRVP and NA-CRVPs were determined. These components were devoid of the enzymatic activities tested, such as phospholipase A(2), arginine esterase, proteolysis, L-amino acid oxidase, 5'nucleotidase, acetylcholinesterase. Furthermore, these three components did not have the following biological activities: coagulant and anticoagulant activities, lethal activity, myotoxicity, hemorrhagic activity, platelet aggregation and platelet aggregation-inhibiting activities. These proteins are named as cysteine-rich venom protein (CRVP) because their sequences showed high level of similarity with mammalian cysteine-rich secretory protein (CRISP) family. Recently, some CRISP-like proteins were also isolated from several different snake venoms, including Agkistrodon blomhoffi, Trimeresurus flavoviridis, Lanticauda semifascita and king cobra. We presumed that CRVP might be a common component in snake venoms. Of particular interest, phylogenetic analysis and sequence alignment showed that NA-CRVP1 and ophanin, both from elapid snakes, share higher similarity with CRVPs from Viperidae snakes.


Assuntos
Venenos Elapídicos/química , Venenos Elapídicos/isolamento & purificação , Elapidae , Proteínas/química , Trimeresurus , Sequência de Aminoácidos , Animais , Clonagem Molecular/métodos , Cisteína/química , Edema/induzido quimicamente , Venenos Elapídicos/genética , Venenos Elapídicos/toxicidade , Cobaias , Hemólise/efeitos dos fármacos , Íleo/fisiologia , Dados de Sequência Molecular , Contração Muscular/efeitos dos fármacos , Músculo Liso/fisiologia , Filogenia , Homologia de Sequência de Aminoácidos
13.
Toxicon ; 44(6): 649-56, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15501291

RESUMO

TMVA is a C-type lectin-like protein with potent platelet activating activity from Trimeresurus mucrosquamatus venom. In the absence of von Willebrand factor (vWF), TMVA dose-dependently induced aggregation of washed platelets. Anti-GP Ib monoclonal antibodies (mAbs), HIP1, specifically inhibited TMVA-induced aggregation in a dose-dependent manner. The aggregation was also inhibited by mAb P2 (an anti-GP IIb mAb). Flow cytometric analysis revealed that FITC-TMVA bound to human formalin-fixed platelets in a saturable manner, and its binding was specifically blocked by HIP1 in a dose-dependent manner. Flow cytometric analysis showed that TMVA did not bind to platelet GPIX, GPIIb, GPIIIa, GPIa, GPIIa and GPIV. Moreover, the platelet aggregation induced by TMVA was partially inhibited when platelet was pretreated with mocarhagin, a snake venom protease that specifically cleaves human GPIb. These results suggest that TMVA is a strong platelet agonist via GPIb and it might have multiple functional binding-sites on GPIb molecule or on other unknown receptor.


Assuntos
Venenos de Crotalídeos/química , Ativação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Trimeresurus , Venenos de Víboras/farmacologia , Animais , Relação Dose-Resposta a Droga , Citometria de Fluxo , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Metaloendopeptidases/farmacologia , Fator de Ativação de Plaquetas/metabolismo , Fator de Ativação de Plaquetas/farmacologia , Venenos de Víboras/metabolismo
14.
Toxicon ; 43(1): 69-75, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15037031

RESUMO

A novel disintegrin, jerdonin, was purified from the Trimeresurus jerdonii venom by means of gel filtration and reverse phase high pressure liquid chromatography. Its coding cDNA was also isolated from the venom gland. The jerdonin coding cDNA is part of a precursor composed of proprotein, metalloproteinase, and disintegrin domains. From the deduced amino acid sequence, jerdonin is composed of 71 amino acid residues including 12 cysteines and the tripeptide sequence Arg-Gly-Asp (RGD), a well-known characteristic of the disintegrin family. Molecular mass of jerdonin was determined to be 7483Da by matrix-assisted laser desorption ionization time of flight mass spectrometry. Jerdonin inhibited ADP- and collagen-induced human platelet aggregation with IC(50) of 220 and 240 nM, respectively. In vivo, jerdonin inhibited the growth of subcutaneously inoculated B16 solid tumor in C57BL/6 mice and improved the survival time of the tumor-bearing mice.


Assuntos
Antineoplásicos/isolamento & purificação , Venenos de Crotalídeos/química , Desintegrinas/isolamento & purificação , Melanoma Experimental/tratamento farmacológico , Oligopeptídeos/isolamento & purificação , Trimeresurus , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacologia , Sequência de Bases , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , Venenos de Crotalídeos/genética , Venenos de Crotalídeos/isolamento & purificação , Venenos de Crotalídeos/farmacologia , DNA Complementar/genética , Desintegrinas/química , Desintegrinas/genética , Desintegrinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Oligopeptídeos/genética , Oligopeptídeos/farmacologia , Homologia de Sequência , Taxa de Sobrevida
15.
Toxicon ; 44(3): 281-7, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15302534

RESUMO

A hemorrhagic proteinase, jerdohagin, was purified from Trimeresurus jerdonii venom by gel filtration and ion-exchange chromatographies. It was a single chain polypeptide with an apparent molecular weight of 96 kDa as estimated by SDS-PAGE under the non-reducing and reducing conditions. Internal peptide sequencing indicated that it consisted of metalloproteinase, disintegrin-like and cysteine-rich domains and belonged to the class III snake venom metalloproteinases (class P-III SVMPs). Like other typical metalloproteinases, hemorrhagic activities of jerdohagin were completely inhibited by EDTA, but not by PMSF. Jerdohagin preferentially degraded alpha-chain of human fibrinogen. Interestingly, jerdohagin did not activate human prothrombin, whereas it cleaved human prothrombin and fragment F1 of activated human prothrombin.


Assuntos
Venenos de Crotalídeos/metabolismo , Metaloproteases/metabolismo , Fragmentos de Peptídeos/metabolismo , Protrombina/metabolismo , Trimeresurus , Sequência de Aminoácidos , Animais , Cromatografia em Gel , Cromatografia por Troca Iônica , Ácido Edético/metabolismo , Eletroforese em Gel de Poliacrilamida , Fibrinogênio/metabolismo , Hemorragia/induzido quimicamente , Humanos , Insulina/metabolismo , Metaloproteases/antagonistas & inibidores , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de Proteína
16.
Toxicon ; 40(9): 1313-9, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12220717

RESUMO

A phospholipase A(2) (PLA(2)), called jerdoxin, was isolated from Trimeresurus jerdonni snake venom and partially characterized. The protein was purified by three chromatographic steps. SDS-polyacrylamide gel electrophoresis in the presence or absence of dithiothreitol showed that it had a molecular mass of 15 kDa. Jerdoxin had an enzymatic activity of 39.4 micro mol/min/mg towards egg yolk phosphatidyl choline (PC). It induced edema in the footpads of mice. In addition, jerdoxin exhibited indirect hemolytic activity. About 97% hemolysis was observed when 2 micro g/ml enzyme was incubated for 90 min in the presence of PC and Ca(2+). No detectable hemolysis was noticed when PC was not added. Ca(2+) was necessary for jerdoxin to exert its hemolytic activity, since only 52% hemolysis was seen when Ca(2+) was absent in the reaction mixture. Furthermore, jerdoxin inhibited ADP induced rabbit platelet aggregation and the inhibition was dose dependent with an IC(50) of 1.0 micro M. The complete amino acid sequence of jerdoxin deduced from cDNA sequence shared high homology with other snake venom PLA(2)s, especially the D 49 PLA(2)s. Also, the residues concerned to Ca(2+) binding were conserved. This is the first report of cDNA sequence of T. jerdonii venom PLA(2).


Assuntos
Venenos de Crotalídeos/enzimologia , Fosfolipases A/genética , Fosfolipases A/toxicidade , Trimeresurus , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar/genética , Ditiotreitol/farmacologia , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Inibidores Enzimáticos/farmacologia , Hemólise/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Fosfatidilcolinas/metabolismo , Fosfolipases A/isolamento & purificação , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/isolamento & purificação , Inibidores da Agregação Plaquetária/toxicidade , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
17.
Toxicon ; 40(9): 1331-8, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12220719

RESUMO

TMVA, a novel C-type lectin-like protein that induces platelet aggregation in a dose-dependent manner, was purified from the venom of Trimeresurus mucrosquamatus. It consists of two subunits, alpha (15536 Da) and beta (14873 Da). The mature amino acid sequences of the alpha (135 amino acids) and beta subunits (123 amino acids) were deduced from cloned cDNAs. Both of the sequences show great similarity to C-type lectin-like venom proteins, including a carbohydrate recognition domain. The cysteine residues of TMVA are conserved at positions corresponding to those of flavocetin-A and convulxin, including the additional Cys135 in the alpha subunit and Cys3 in the beta subunit. SDS-PAGE, mass spectrometry analysis and amino acid sequence showed that native TMVA exists as two convertible multimers of (alpha beta)(2) and (alpha beta)(4) with molecular weights of 63680 and 128518 Da, respectively. The (alpha beta)(2) complex is stabilized by an interchain disulfide bridge between the two alpha beta-heterodimers, whereas the stabilization of the (alpha beta)(4) complex seems to involve non-covalent interactions between the (alpha beta)(2) complexes.


Assuntos
Venenos de Crotalídeos/química , Lectinas Tipo C/genética , Lectinas Tipo C/isolamento & purificação , Agregação Plaquetária , Trimeresurus , Venenos de Víboras/genética , Venenos de Víboras/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Plaquetas/efeitos dos fármacos , Clonagem Molecular , Venenos de Crotalídeos/genética , DNA Complementar/genética , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Espectrometria de Massas , Dados de Sequência Molecular , Análise de Sequência de Proteína , Venenos de Víboras/farmacologia
18.
Comp Biochem Physiol B Biochem Mol Biol ; 132(3): 529-34, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12091097

RESUMO

Jerdonobin and jerdofibrase are two serine proteases purified from the venom of Trimeresurus jerdonii. The Michaelis constant K(m) and the catalytic rate constant K(cat) of jerdonobin or jerdofibrase on three chromogenic substrates, H-D-Pro-Phe-Arg-pNA (S2302), H-D-Phe-pipecolyl-Arg-pNA (S2238), and H-D-Val-Leu-Lys-pNA (S2251) were obtained from lineweaver-Burk plots. Jerdofibrase could hydrolyze all three substrates, but jerdonobin had no detectable activity on S2251, suggesting a relatively broader substrate specificity for jerdofibrase than jerdonobin. By SDS-PAGE, jerdofibrase preferentially degraded Bbeta-chain of fibrinogen. It also degraded Aalpha-chain of fibrinogen with relatively slow activity, but did not act on the gamma-chain. In contrast, jerdonobin did not degrade fibrinogen within 12 h. Fibrinopeptides liberation test, identified by HPLC, showed jerdonobin released fibrinopeptide A and a small amount of fibrinopeptide B. Unlike jerdonobin, jerdofibrase mainly released fibrinopeptide B. These results indicate that the two enzymes differ in their ability to hydrolyze chromogenic substrates and in their actions on fibrinogen.


Assuntos
Compostos Cromogênicos/metabolismo , Venenos de Crotalídeos/enzimologia , Venenos de Crotalídeos/metabolismo , Fibrinogênio/metabolismo , Serina Endopeptidases/metabolismo , Animais , Coagulação Sanguínea , Bovinos , Venenos de Crotalídeos/isolamento & purificação , Eletroforese em Gel de Poliacrilamida , Fibrina/metabolismo , Humanos , Serina Endopeptidases/isolamento & purificação , Espectrofotometria , Trimeresurus
19.
Comp Biochem Physiol B Biochem Mol Biol ; 139(1): 117-22, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15364294

RESUMO

A novel disintegrin, jerdonatin, was purified to homogeneity from Trimeresurus jerdonii venom by gel filtration and reversed-phase high-pressure liquid chromatography. We isolated the cDNA encoding jerdonatin from the snake venom gland. Jerdonatin cDNA precursor encoded pre-peptide, metalloprotease and disintegrin domain. Jerdonatin is composed of 72 amino acid residues including 12 cysteines and the tripeptide sequence Arg-Gly-Asp (RGD), a well-known characteristic of the disintegrin family. Molecular mass of jerdonatin was determined to be 8011 Da by matrix-assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF-MS). Jerdonatin inhibited ADP- and collagen-induced human platelet aggregation with IC50 of 123 and 135 nM, respectively. We also investigated the effect of jerdonatin on the binding of B6D2F1 hybrid mice spermatozoa to mice zona-free eggs and their subsequent fusion. Jerdonatin significantly inhibited sperm-egg binding in a concentration-dependent manner, but had no effect on the fusion of sperm-egg. These results indicate that integrins on the egg play a role in mammalian fertilization.


Assuntos
Desintegrinas/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Interações Espermatozoide-Óvulo/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Venenos de Crotalídeos/química , Venenos de Crotalídeos/farmacologia , DNA Complementar/genética , Desintegrinas/química , Desintegrinas/genética , Desintegrinas/isolamento & purificação , Feminino , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Peso Molecular , Óvulo/efeitos dos fármacos , Óvulo/metabolismo , Análise de Sequência , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo
20.
Comp Biochem Physiol B Biochem Mol Biol ; 137(2): 219-24, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14990218

RESUMO

A chymotrypsin inhibitor, designated NA-CI, was isolated from the venom of the Chinese cobra Naja atra by three-step chromatography. It inhibited bovine alpha-chymotrypsin with a Ki of 25 nM. The molecular mass of NA-CI was determined to be 6403.8 Da by matrix-assisted laser-desorption ionization time-of-flight (MALDI-TOF) analysis. The complete amino acid sequence was determined after digestion of S-carboxymethylated inhibitor with Staphylococcus aureus V8 protease and porcine trypsin. NA-CI was a single polypeptide chain composed of 57 amino acid residues. The main contact site with the protease (P1) has a Phe, showing the specificity of the inhibitor. NA-CI shared great similarity with the chymotrypsin inhibitor from Naja naja venom (identities=89.5%) and other snake venom protease inhibitors.


Assuntos
Venenos Elapídicos/química , Inibidores da Tripsina/química , Inibidores da Tripsina/isolamento & purificação , Sequência de Aminoácidos , Animais , Cromatografia Líquida , Quimotripsina/química , Elapidae , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de Proteína , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA