Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 272: 116080, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38350215

RESUMEN

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.


Asunto(s)
Mercurio , Metales Pesados , Masculino , Humanos , Estados Unidos , Antígeno Prostático Específico , Cadmio , Encuestas Nutricionales , Zinc
2.
Dongwuxue Yanjiu ; 32(1): 56-61, 2011 Feb.
Artículo en Chino | MEDLINE | ID: mdl-21341385

RESUMEN

Similar to humans, most of non-human primates also contain the ABO blood group system. In this study, we sought to evaluate the ABO antibody (Ab) levels in monkeys using a modified flow cytometry method (FCM). The standard commercial human A or B-red blood cells (RBCs) were used as target cells. The binding of target cells and anti-A or B blood group Ab in sera of rhesus or cynomolgus monkeys was detected by flow cytometry after adding secondary specific fluorescence-labeled anti-human IgG or IgM Ab. Human healthy blood donor sera were used as controls. The results revealed that, using clear monkey sera, which were pre-absorbed on normal human type O RBCs to remove non-specific anti-human Abs, the modified FCM gave an accurate detection of ABO Ab levels in monkeys. When compared with the results of human sera, the distribution of ABO Ab levels in monkey sera were significantly lower (P<0.05). We concluded that the modified FCM could be used for the detection of monkey ABO Ab levels. The technique and data will be very valuable for the future establishment of ABO-incompatible organ transplant models in non-human primates, which could improve clinical applications.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Tipificación y Pruebas Cruzadas Sanguíneas/métodos , Citometría de Flujo/métodos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Macaca fascicularis/inmunología , Macaca mulatta/inmunología , Adulto , Animales , Eritrocitos/inmunología , Femenino , Humanos , Macaca fascicularis/sangre , Macaca mulatta/sangre , Masculino , Persona de Mediana Edad
3.
Toxicon ; 54(6): 763-71, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19523971

RESUMEN

A new L-amino acid oxidase (designated as DRS-LAAO) was purified from Daboia russellii siamensis venom by ion-exchange, gel filtration and affinity chromatographies. DRS-LAAO is a homodimeric enzyme with a molecular weight of 120.0 kDa as measured by size exclusion chromatography and the monomeric molecular weight of 58.0 kDa as measured by SDS-PAGE under both non-reducing and reducing conditions. The N-terminal amino acid sequence (ADDKNPLEECFREDD) of DRS-LAAO shares high identity with other snake venom L-amino acid oxidases, especially with those isolated from viperid venoms. The enzyme displayed high specificity towards hydrophobic L-amino acids. The best substrate of DRS-LAAO was L-Leu followed by L-Phe and L-Ile, while five substrates--L-Pro, L-Asn, L-Gly, L-Ser and L-Cys were not oxidized. Optimal pH of DRS-LAAO was 8.8. The enzyme showed no hemorrhagic activity even at a dosage of 55.0 microg. DRS-LAAO dose-dependently inhibited platelet aggregation induced by ADP (83.33 microM) and TMVA (55.0 nM) with an IC(50) value of 32.8 microg/ml and 32.3 microg/ml, respectively. The minimum inhibitory concentrations (MICs) of DRS-LAAO against Staphylococci aureus (ATCC 25923), Pseudomonas aeruginosa (ATCC 27853) and Escherichia coli (ATCC 25922) were 9.0, 144.0 and 288.0 microg/ml, respectively. The minimum bactericidal concentrations (MBCs) of the enzyme for these strains were twice of the MIC values. These results showed that DRS-LAAO had the strongest antimicrobial activity against S. aureus among these three international standard stains. Antibacterial-activities of DRS-LAAO against eight clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates were also tested. The MICs of DRS-LAAO against these isolates ranged from 4.5 to 36.0 microg/ml. And the MBCs of the enzyme against these isolates ranged from 9.0 to 72.0 microg/ml.


Asunto(s)
L-Aminoácido Oxidasa/aislamiento & purificación , Venenos de Víboras/enzimología , Secuencia de Aminoácidos , Cromatografía en Gel , Electroforesis en Gel de Poliacrilamida , Escherichia coli/efectos de los fármacos , Humanos , L-Aminoácido Oxidasa/química , L-Aminoácido Oxidasa/farmacología , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Agregación Plaquetaria/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Especificidad por Sustrato
4.
Toxicon ; 52(2): 277-84, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18590752

RESUMEN

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.


Asunto(s)
Venenos de Crotálidos/química , Venenos de Crotálidos/enzimología , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Trimeresurus/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Coagulación Sanguínea/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Fraccionamiento Químico , Venenos de Crotálidos/farmacología , Humanos , Datos de Secuencia Molecular , Filogenia , Agregación Plaquetaria/efectos de los fármacos , Alineación de Secuencia , Análisis de Secuencia de ADN , Serina Endopeptidasas/farmacología
5.
Zhonghua Yi Xue Za Zhi ; 86(21): 1460-3, 2006 Jun 06.
Artículo en Chino | MEDLINE | ID: mdl-16842697

RESUMEN

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.


Asunto(s)
Venenos Elapídicos/uso terapéutico , Rechazo de Injerto/prevención & control , Trasplante de Corazón/métodos , Acondicionamiento Pretrasplante/métodos , Animales , Inmunoglobulina G/inmunología , Distribución Aleatoria , Ratas , Ratas Endogámicas BN , Ratas Endogámicas Lew , Trasplante de Piel/métodos , Trasplante Homólogo
6.
Biochimie ; 88(10): 1331-42, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16793192

RESUMEN

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.


Asunto(s)
Venenos de Crotálidos/enzimología , Citocinas/metabolismo , Fosfolipasas A/química , Fosfolipasas A/farmacología , Secuencia de Aminoácidos , Animales , Antibacterianos/farmacología , Arginina , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Peso Molecular , Monocitos/efectos de los fármacos , Monocitos/inmunología , Fosfolipasas A/aislamiento & purificación , Ratas , Ratas Wistar , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
7.
Biochim Biophys Acta ; 1760(3): 462-71, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16413680

RESUMEN

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.


Asunto(s)
Venenos de Crotálidos/aislamiento & purificación , Fosfolipasas A/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Venenos de Crotálidos/química , Venenos de Crotálidos/farmacología , Fosfolipasas A2 Grupo II , Ratones , Datos de Secuencia Molecular , Peso Molecular , Músculo Esquelético/efectos de los fármacos , Fosfolipasas A/química , Fosfolipasas A/farmacología , Fosfolipasas A2 , Filogenia , Agregación Plaquetaria/efectos de los fármacos , Conejos , Proteínas de Reptiles , Alineación de Secuencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Trimeresurus
8.
Transplantation ; 81(2): 273-83, 2006 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-16436972

RESUMEN

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.


Asunto(s)
Anticuerpos Heterófilos/biosíntesis , Rechazo de Injerto/inmunología , Trasplante de Riñón/inmunología , Trisacáridos/inmunología , Enfermedad Aguda , Animales , Animales Modificados Genéticamente , Rechazo de Injerto/etiología , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/patología , Pruebas de Neutralización , Papio , Sus scrofa , Trasplante Heterólogo
9.
Int J Biochem Cell Biol ; 38(4): 510-20, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16310401

RESUMEN

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.


Asunto(s)
Venenos Elapídicos/química , Liberación de Histamina/efectos de los fármacos , Mastocitos/metabolismo , Metaloproteasas/farmacología , Proteínas de Reptiles/farmacología , Animales , Células Cultivadas , Venenos Elapídicos/farmacología , Fibrinógeno/metabolismo , Humanos , Mastocitos/citología , Metaloproteasas/química , Metaloproteasas/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Proteínas de Reptiles/química , Proteínas de Reptiles/aislamiento & purificación , Factores de Tiempo
10.
Toxicon ; 47(1): 104-12, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16330060

RESUMEN

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.


Asunto(s)
Lectinas Tipo C/antagonistas & inhibidores , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Venenos de Víboras/química , Viperidae , Adenosina Difosfato/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Concentración 50 Inhibidora , Lectinas Tipo C/genética , Lectinas Tipo C/aislamiento & purificación , Datos de Secuencia Molecular , Peso Molecular , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Ristocetina/farmacología , Venenos de Víboras/genética , Venenos de Víboras/farmacología
11.
Nat Med ; 11(12): 1295-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16311604

RESUMEN

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.


Asunto(s)
Animales Modificados Genéticamente , Rechazo de Injerto/inmunología , Tolerancia Inmunológica/inmunología , Trasplante de Riñón/inmunología , Trasplante Heterólogo/mortalidad , Trasplante Heterólogo/métodos , Animales , Anticuerpos/sangre , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Terapia de Inmunosupresión/métodos , Papio , Sus scrofa/genética , Linfocitos T/inmunología , Trasplante Heterólogo/inmunología
12.
J Biol Chem ; 280(49): 40714-22, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16215260

RESUMEN

Jerdostatin represents a novel RTS-containing short disintegrin cloned by reverse transcriptase-PCR from the venom gland mRNA of the Chinese Jerdons pit viper Trimeresurus jerdonii. The jerdostatins precursor cDNA contained a 333-bp open reading frame encoding a signal peptide, a pre-peptide, and a 43-amino acid disintegrin domain, whose amino acid sequence displayed 80% identity with that of the KTS-disintegrins obtustatin and viperistatin. The jerdostatin cDNA structure represents the first complete open reading frame of a short disintegrin and points to the emergence of jerdostatin from a short-coding gene. The different residues between jerdostatin and obtustatin/viperistatin are segregated within the integrin-recognition loop and the C-terminal tail. Native jerdostatin (r-jerdostatin-R21) and a R21K mutant (r-jerdostatin-K21) were produced in Escherichia coli. In each case, two conformers were isolated. One-dimensional (1)H NMR showed that conformers 1 and 2 of r-jerdostatin-R21 represent, respectively, well folded and unfolded proteins. The two conformers of the wild-type and the R21K mutant inhibited the adhesion of alpha(1)-K562 cells to collagen IV with IC(50) values of 180 and 703 nm, respectively. The IC(50) values of conformers 2 of r-jerdostatin-R21 and r-jerdostatin-K21 were, respectively, 5.95 and 12.5 microm. Neither r-jerdostatin-R21 nor r-jerdostatin-K21 showed inhibitory activity toward other integrins, including alpha(IIb)beta(3), alpha(v)beta(3), alpha(2)beta(1), alpha(5)beta(1), alpha(4)beta(1), alpha(6)beta(1), and alpha(9)beta(1) up to a concentration of 24 mum. Although the RTS motif appears to be more potent than KTS inhibiting the alpha(1)beta(1) integrin, r-jerdostatin-R21 is less active than the KTS-disintegrins, strongly suggesting that substitutions outside the integrin-binding motif and/or C-terminal proteolytic processing are responsible for the decreased inhibitory activity.


Asunto(s)
ADN Complementario/genética , Desintegrinas/genética , Integrina alfa1beta1/antagonistas & inhibidores , Trimeresurus/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Venenos de Crotálidos , Cisteína/análisis , Desintegrinas/química , Desintegrinas/farmacología , Disulfuros/análisis , Glándulas Exocrinas/química , Expresión Génica , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Sistemas de Lectura Abierta , Mapeo Peptídico , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes , Tripsina/metabolismo
13.
Toxicon ; 45(3): 353-60, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15683874

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Venenos de Crotálidos/enzimología , Fibrinógeno/química , Serina Endopeptidasas/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Datos de Secuencia Molecular , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serina Endopeptidasas/aislamiento & purificación , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato , Trimeresurus
14.
Toxicon ; 44(6): 649-56, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15501291

RESUMEN

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.


Asunto(s)
Venenos de Crotálidos/química , Activación Plaquetaria/efectos de los fármacos , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Trimeresurus , Venenos de Víboras/farmacología , Animales , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Metaloendopeptidasas/farmacología , Factor de Activación Plaquetaria/metabolismo , Factor de Activación Plaquetaria/farmacología , Venenos de Víboras/metabolismo
15.
Comp Biochem Physiol B Biochem Mol Biol ; 139(1): 117-22, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15364294

RESUMEN

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.


Asunto(s)
Desintegrinas/farmacología , Agregación Plaquetaria/efectos de los fármacos , Interacciones Espermatozoide-Óvulo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Venenos de Crotálidos/química , Venenos de Crotálidos/farmacología , ADN Complementario/genética , Desintegrinas/química , Desintegrinas/genética , Desintegrinas/aislamiento & purificación , Femenino , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Peso Molecular , Óvulo/efectos de los fármacos , Óvulo/metabolismo , Análisis de Secuencia , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo
16.
Toxicon ; 44(3): 281-7, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15302534

RESUMEN

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.


Asunto(s)
Venenos de Crotálidos/metabolismo , Metaloproteasas/metabolismo , Fragmentos de Péptidos/metabolismo , Protrombina/metabolismo , Trimeresurus , Secuencia de Aminoácidos , Animales , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Ácido Edético/metabolismo , Electroforesis en Gel de Poliacrilamida , Fibrinógeno/metabolismo , Hemorragia/inducido químicamente , Humanos , Insulina/metabolismo , Metaloproteasas/antagonistas & inhibidores , Ratones , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de Proteína
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 35(2): 194-7, 2004 Mar.
Artículo en Chino | MEDLINE | ID: mdl-15071914

RESUMEN

OBJECTIVE: To investigate the therapeutic effects of a nerve growth factor (NGF) isolated and purified from the venom of Naja naja atra on injured sciatic nerves in adult cat. METHODS: Model of sciatic nerve crush lesion in 20 cats was made. After the operation, in the therapeutic group, NGF(2 micrograms/kg) was injected intramuscularly into the cat's leg once a day for 10 d (n = 5) and for 30 d (n = 5) respectively; in the control group, NGF was not injected and the cats were allowed to survive 10 d (n = 5) and 30 d (n = 5) respectively. RESULTS: Ten days after the operation, the number of distal nerve fibers was significantly smaller in the control group than in the therapeutic group (P < 0.01); the reaction to planta stimulation appeared earlier and the leg action recovered faster in the therapeutic group. Thirty day after the operation, the number of distal nerve fibers in the therapeutic group was significantly greater than that of other groups (P < 0.01), but the histologic structure of the nerve fibers was in disorder and the axon and Ranvier node disappeared. About 16 days after injury and injection of NGF for consecutive days, the reaction to planta stimulation disappeared and leg paraplegia occurred in the operated side. CONCLUSION: The results showed that NGF could obviously reduce the degeneration of nerve fibers and enhance the peripheral nerve regeneration and functional recovery from ingury early, but daily injection of NGF in the injured region for a long period could significantly result in over-regeneration of nerve fibers, and the conductive function of the injured peripheral nerves would be lost.


Asunto(s)
Venenos Elapídicos/química , Factor de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Nervio Ciático/lesiones , Animales , Gatos , Compresión Nerviosa , Factor de Crecimiento Nervioso/aislamiento & purificación , Distribución Aleatoria , Nervio Ciático/patología , Nervio Ciático/fisiopatología
18.
Toxicon ; 43(1): 69-75, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15037031

RESUMEN

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.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Venenos de Crotálidos/química , Desintegrinas/aislamiento & purificación , Melanoma Experimental/tratamiento farmacológico , Oligopéptidos/aislamiento & purificación , Trimeresurus , Secuencia de Aminoácidos , Animales , Antineoplásicos/farmacología , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Clonación Molecular , Venenos de Crotálidos/genética , Venenos de Crotálidos/aislamiento & purificación , Venenos de Crotálidos/farmacología , ADN Complementario/genética , Desintegrinas/química , Desintegrinas/genética , Desintegrinas/farmacología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Oligopéptidos/genética , Oligopéptidos/farmacología , Homología de Secuencia , Tasa de Supervivencia
19.
Comp Biochem Physiol B Biochem Mol Biol ; 137(2): 219-24, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14990218

RESUMEN

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.


Asunto(s)
Venenos Elapídicos/química , Inhibidores de Tripsina/química , Inhibidores de Tripsina/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Cromatografía Liquida , Quimotripsina/química , Elapidae , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Xenotransplantation ; 11(2): 123-32, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14962274

RESUMEN

At present, acute vascular rejection (AVR) remains a primary obstacle inhibiting long-term graft survival in the pig-to-non-human primate transplant model. The present study was undertaken to determine whether repetitive injection of low dose Yunnan-cobra venom factor (Y-CVF), a potent complement inhibitor derived from the venom of Naja kaouthia can completely abrogate hemolytic complement activity and subsequently improve the results in a pig-to-rhesus monkey heterotopic heart transplant model. Nine adult rhesus monkeys received a heterotopic heart transplant from wild-type pigs and the recipients were allocated into two groups: group 1 (n = 4) received repetitive injection of low dose Y-CVF until the end of the study and group 2 (n = 5) did not receive Y-CVF. All recipients were treated with cyclosporine A (CsA), cyclophosphamide (CyP) and steroids. Repetitive Y-CVF treatment led to very dramatic fall in CH50 and serum C3 levels (CH50 < 3 units/C3 remained undetectable throughout the experiment) and successfully prevented hyperacute rejection (HAR), while three of five animals in group 2 underwent HAR. However, the continuous suppression of circulating complement did not prevent AVR and the grafts in group 1 survived from 8 to 13 days. Despite undetectable C3 in circulating blood, C3 deposition was present in these grafts. The venular thrombosis was the predominant histopathologic feature of AVR. We conclude that repetitive injection of low dose Y-CVF can be used to continuously suppress circulating complement in a very potent manner and successfully prevent HAR. However, this therapy did not inhibit complement deposition in the graft and failed to prevent AVR. These data suggest that using alternative pig donors [i.e. human decay accelerating factor (hDAF)-transgenic] in combination with the systemic use of complement inhibitors may be necessary to further control complement activation and improve survival in pig-to-non-human primate xenotransplant model.


Asunto(s)
Proteínas Inactivadoras de Complemento/metabolismo , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Trasplante Heterólogo/inmunología , Animales , Complemento C3/metabolismo , Proteínas Inactivadoras de Complemento/antagonistas & inhibidores , Proteínas del Sistema Complemento/análisis , Electrocardiografía , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Trasplante de Corazón/métodos , Trasplante de Corazón/patología , Trasplante de Corazón/fisiología , Terapia de Inmunosupresión/métodos , Macaca mulatta , Porcinos , Factores de Tiempo , Trasplante Heterólogo/métodos , Trasplante Heterólogo/patología , Trasplante Heterólogo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...