Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Nat Commun ; 7: 13107, 2016 10 06.
Article En | MEDLINE | ID: mdl-27708285

Snakes have numerous features distinctive from other tetrapods and a rich history of genome evolution that is still obscure. Here, we report the high-quality genome of the five-pacer viper, Deinagkistrodon acutus, and comparative analyses with other representative snake and lizard genomes. We map the evolutionary trajectories of transposable elements (TEs), developmental genes and sex chromosomes onto the snake phylogeny. TEs exhibit dynamic lineage-specific expansion, and many viper TEs show brain-specific gene expression along with their nearby genes. We detect signatures of adaptive evolution in olfactory, venom and thermal-sensing genes and also functional degeneration of genes associated with vision and hearing. Lineage-specific relaxation of functional constraints on respective Hox and Tbx limb-patterning genes supports fossil evidence for a successive loss of forelimbs then hindlimbs during snake evolution. Finally, we infer that the ZW sex chromosome pair had undergone at least three recombination suppression events in the ancestor of advanced snakes. These results altogether forge a framework for our deep understanding into snakes' history of molecular evolution.


Biological Evolution , DNA Transposable Elements , Snakes/anatomy & histology , Snakes/genetics , Animals , Cell Lineage , Evolution, Molecular , Female , Forelimb , Gene Expression Profiling , Gene Expression Regulation , Genome , Hindlimb , Lizards/genetics , Male , Phylogeny , Recombination, Genetic , Sex Chromosomes , Transcriptome
2.
CNS Neurosci Ther ; 21(6): 486-95, 2015 Jun.
Article En | MEDLINE | ID: mdl-25678034

BACKGROUND: Neuroactive steroids represent promising candidates for the treatment of neurological disorders. Our previous studies identified an endogenous steroid cholestane-3ß, 5α, 6ß-triol (Triol) as a novel neuroprotectant. AIM: We aimed to identify a potent candidate for stroke treatment through a screening of Triol analogs. METHODS: Hypoxia- and glutamate-induced neuronal injury models in vitro, middle cerebral artery occlusion (MCAO)-induced cerebral ischemia model in vivo, fluorescein diacetate (FDA) for alive and propidium iodide (PI) for dead staining, LDH assay, and calcium imaging techniques were used. RESULTS: 24-keto-cholest-5-en-3ß, 19-diol (Diol) showed the most potent neuroprotective effect among the screened structurally related compounds. FDA and PI staining showed that Diol concentration dependently increased the survival rate of cerebellar granule neurons (CGNs) challenged with glutamate or hypoxia, with an effective threshold concentration of 2.5 µM. Consistently, the quantitative LDH release assay showed the same concentration-dependent protection in both models. Diol, at 10 µM, potently decreased glutamate- and hypoxia-induced LDH release from 51.6 to 18.2% and 62.1 to 21.7%, respectively, which values are close to the normal LDH release (~16-18%). Moreover, we found Diol effectively decreased MCAO-induced infarction volume in mice from ~23% to 7%, at a dose of 6 mg/kg. We further explored the underlying mechanism and found that Diol attenuated NMDA-induced intracellular calcium ([Ca(2+) ]i ) increase in cortical neurons, suggesting a negative modulatory effect on NMDA receptor. CONCLUSION: Taken together, we identified Diol as a potent neuroprotectant. It may represent a novel and promising neuroprotectant for stroke intervention.


Cholestanols/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cell Hypoxia/drug effects , Cells, Cultured , Cerebellum/cytology , Cholestanols/chemistry , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/toxicity , L-Lactate Dehydrogenase/metabolism , N-Methylaspartate/toxicity , Oxygen/administration & dosage , Rats , Rats, Sprague-Dawley
3.
Biochem Pharmacol ; 85(6): 772-9, 2013 Mar 15.
Article En | MEDLINE | ID: mdl-23178656

Hyperacute rejection (HAR) is a main barrier in xenotransplantation, which is mediated by the combination of natural antibody to the xenograft and complement activation. Current therapies have focus on the inhibition of complement by development of complement inhibitor and transgenic animal organ. Here, we investigated the effects of rFII, a recombinant fibrinogenase from Agkistrodon acutus venom, on complement and HAR. The degradation effect of rFII on complement was tested by SDS-PAGE, CH50 examination, ELISA Kit and cofocal immunofluorescence microscopy in vitro and in vivo. An ex-vivo rat-to-human perfusion model and a vivo guinea-pig-to-rat heat HAR model were used to determine the protection of rFII against HAR. Our investigation indicated that rFII could significantly degrade human C5, C6, and C9, decrease the activity of complement, and inhibit the MAC deposition on HUVECs membrane in vitro. In addition, serum levels of C1q, C3 and C4 in rat were gradually reduced after infusion of rFII. Importantly, in an ex vivo rat-to-human perfusion model, the survival of rat hearts perfused with human serum treated with rFII (83.36 ± 16.63 min) were significantly longer than that of hearts perfused with fresh human serum(15.94 ± 4.75 min). At the time of 15 minutes after perfusion, functions of hearts added with 50 ug/ml rFII sustained well with heart rates at 283 ± 65.32 beats/minute and LVDP at 13.70 ± 5.45 Kpa, while that of hearts perfused with fresh human serum were severely damaged by HAR with heart rates at 107.77 ± 40.31 beats/minute and LVDP at 1.01 ± 0.83 Kpa. We also found that rFII significantly decreased the levels of C1q, C3 and C4 in human fresh serum perfusate. In a vivo guinea-pig-to-rat heat HAR model, the survival of rat hearts treated with rFII were significantly longer than that of hearts perfused with normal saline; and relieved heart damage by complete activation. Our finding demonstrates the anti-complement property of rFII and its protection against HAR, indicating that rFII might be as a potential therapeutic agent for xenotransplantation.


Complement System Proteins/metabolism , Crotalid Venoms/enzymology , Crotalid Venoms/pharmacology , Graft Rejection/prevention & control , Metalloendopeptidases/pharmacology , Animals , Guinea Pigs , Heart Transplantation , Hemolysis , Immunohistochemistry , Microscopy, Fluorescence , Proteolysis , Rats , Recombinant Proteins/pharmacology
4.
Biochem Pharmacol ; 84(7): 905-13, 2012 Oct 01.
Article En | MEDLINE | ID: mdl-22728069

The incidence of disseminated intravascular coagulation (DIC), which leads to multiple organ dysfunction and high mortality, has remained constant in recent years. At present, treatments of DIC have focused on preventing cytokine induction, inhibiting coagulation processes and promoting fibrinolysis. Recent clinical trials have supported the use of antithrombin and activated protein C supplementation in DIC. To better understand the mechanism of treatment on DIC, we here report a novel fibrinogenase from Agkistrodon acutus (FIIa) that effectively protected against LPS-induced DIC in a rabbit model, and detected the tissue factors expression in HUVE cells after using FIIa. In vivo, administration of FIIa reduced hepatic and renal damage, increased the concentration of fibrinogen, the activities of protein C, the platelet count, APTT, PT, FDP, the level of AT-III and t-PA, decreased the level of PAI-1, and increased survival rate in LPS-induced DIC rabbits. In vitro experiments, we further confirmed that FIIa up-regulated the expression of t-PA and u-PA, down-regulated the expression of PAI-1, and directly activated protein C. Our findings suggest that FIIa could effectively protect against DIC via direct degradation of microthrombi and activation of protein C as well as provide a novel strategy to develop a single proteinase molecule for targeting the main pathological processes of this disease.


Agkistrodon/physiology , Crotalid Venoms/enzymology , Disseminated Intravascular Coagulation/drug therapy , Protein C/metabolism , Serine Endopeptidases/metabolism , Thrombosis/metabolism , Animals , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Disseminated Intravascular Coagulation/chemically induced , Disseminated Intravascular Coagulation/prevention & control , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Kidney Diseases/prevention & control , Lipopolysaccharides/toxicity , Male , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein C/genetics , Rabbits , Serine Endopeptidases/pharmacology , Serine Endopeptidases/therapeutic use , Thrombosis/drug therapy , Tissue Plasminogen Activator/drug effects , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/drug effects , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
5.
Acta Pharmacol Sin ; 32(2): 239-44, 2011 Feb.
Article En | MEDLINE | ID: mdl-21293476

AIM: To evaluate the effects of the fibrinolytic enzyme FII(a) from Agkistrodon acutus venom on acute pulmonary thromboembolism (APT) in animal models. METHODS: Both rabbit and dog APT models were used. For the rabbit APT model, the thrombi weight before and after administration was measured. Central venous pressure (CVP) and mean arterial pressure (MAP) were measured before and 15, 30, 60, and 120 min after the injection of the blood clot. Partial thromboplastin time (APTT), prothrombin time (PT), platelet count, and fibrinogen concentration were measured using auto analyzers. Plasminogen activity was measured based on chromogenic substrates. In the dog APT model, pulmonary blood flow was recorded using pulmonary angiography. RESULTS: Intravenous administration of FIIa (0.1-5.0 mg/kg) improved the APT-induced hemodynamic derangements and reduced thrombi weight. The angiography evidence also showed that the pulmonary emboli had almost disappeared after FII(a) infusion. FII(a) (0.1, 0.5, or 1.0 mg/kg) did not impair the coagulation pathways, although very high doses of FII(a) (5.0 mg/kg) could stimulate the production of plasminogen and result in impairment of the pathways. CONCLUSION: FII(a) could effectively protect against APT via degradation of thrombi with less activation of plasminogen, and may provide a novel fibrinolytic enzyme for targeting the main pathological processes of the disease.


Agkistrodon , Crotalid Venoms/pharmacology , Fibrinolytic Agents/pharmacology , Pulmonary Embolism/drug therapy , Acute Disease , Angiography/methods , Animals , Crotalid Venoms/isolation & purification , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Fibrinolytic Agents/isolation & purification , Male , Metalloendopeptidases , Plasminogen/drug effects , Plasminogen/metabolism , Pulmonary Embolism/physiopathology , Rabbits , Time Factors
6.
Zhonghua Wei Chang Wai Ke Za Zhi ; 14(1): 27-30, 2011 Jan.
Article Zh | MEDLINE | ID: mdl-21271375

OBJECTIVE: To study the feasibility of MRI of human colon adenocarcinoma cell line (Lovo) labeled with superparamagnetic iron oxide(SPIO) nanoparticles in vitro. METHODS: Lovo cells (5 × 10(5) and 1 × 10(6)) were cultured in medium containing different SPIO nanoparticles (50 microl and 500 microl). Transmission electron microscopy was used to observe cellular ultrastructure and to determine the uptake and distribution of particles in Lovo cells at 1-, 3-, 6-hours. MRI of Lovo cells was performed with T1WI, T2WI sequences. Unlabeled cells were used as controls. RESULTS: Uptake of SPIO nanoparticles occurred within 6 hours. On T1 weighted imaging, there was no significant difference in signal intensity between the experimental groups and the control group. On T2 weighted imaging, there was no significant difference in signal intensity between the experimental groups and the control group after culture of 1 h. Signal intensity began to decrease in 1 × 10(6) Lovo cells labeled with 500 microl SPIO nanoparticle after 3 hours culture. Signal intensity decreased in all the experimental groups after 6 hours culture. CONCLUSION: Human colon adenocarcinoma cell line (Lovo) can be labeled with SPIO nanoparticles, and the labeled cells can be imaged with MRI equipment.


Magnetic Resonance Imaging/methods , Nanoparticles , Staining and Labeling/methods , Adenocarcinoma/pathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Humans , Iron , Magnetics , Oxides , Pilot Projects
7.
Acta Pharmacol Sin ; 31(1): 73-80, 2010 Jan.
Article En | MEDLINE | ID: mdl-19966835

AIM: To investigate whether aspirin is able to augment gemcitabine-induced cytotoxicity in human pancreatic cancer cells. METHODS: Two gemcitabine-insensitive human pancreatic cancer cell lines, PANC-1 and Capan-1, were used. Cells were treated with either aspirin or gemcitabine alone or both of them. Cell growth and apoptosis were determined by MTT assay, Annexin V or Hoechest 33258 staining. Cell cycle distribution was examined by flow cytometry. Western blot with specific phosphorylated protein antibodies was used to detect the activation of protein kinase. RT-PCR and Western blot were applied to assess the transcription and protein level for cyclin D1 and Bcl-2. RESULTS: Aspirin alone significantly inhibits the proliferation of PANC-1 cells by causing cell cycle arrest at G(1) phase. Aspirin potentiates the anti-survival effect of gemcitabine as well as its pro-apoptotic effect in PANC-1 cells, although aspirin per se does not trigger apoptosis. Aspirin inhibits GSK-3beta activation and suppresses the expression of its downstream gene products (cyclin D1 and Bcl-2), which are implicated in proliferation, survival and chemoresistance of pancreatic cancer. The effects of aspirin on Capan-1, were similar to that on PANC-1. CONCLUSION: Our results suggest that aspirin inhibits the proliferation of gemcitabine-resistant pancreatic cancer cells and augments the antisurvival effect of gemcitabine, probably by suppressing the activity of GSK-3beta and its downstream gene products.


Antimetabolites, Antineoplastic/pharmacology , Aspirin/pharmacology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/drug effects , Cyclin D1/genetics , Deoxycytidine/pharmacology , Drug Synergism , Flow Cytometry , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Pancreatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Gemcitabine
8.
Exp Toxicol Pathol ; 61(2): 137-43, 2009 Mar.
Article En | MEDLINE | ID: mdl-18703324

As a model of the reperfusion injury found in stroke, we treated cerebellar granule neurons (CGNs) with hypoxia followed by reoxygenation. Hypoxia for 3h followed by 24h reoxygenation (H/R) induced a typical apoptosis of CGNs. CGNs exposed to H/R responded by activating JNK, increasing the expression of p38 and ultimately caused CGNs dying. Furthermore, apoptosis of CGNs induced by H/R was inhibited by pre-treatment with SB203580 or SP600125, and the inhibitory effect of SB203580 was greater than that of SP600125. Additionally, we also found that H/R temporally activated Akt and inactivated glycogen synthesis kinase-3beta (GSK-3beta), two proteins the functions of which were important in cell survival and energy metabolism. These findings demonstrated that H/R-induced apoptosis in CGNs by enhancing JNK and p38 activity, which contributed at least in part to H/R-induced apoptosis of CGNs.


Apoptosis , Cerebellum/pathology , JNK Mitogen-Activated Protein Kinases/physiology , Neurons/pathology , Oxygen/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Blotting, Western , Cell Hypoxia/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/enzymology , Cerebellum/metabolism , Chromatin/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Toxicon ; 52(5): 638-46, 2008 Oct.
Article En | MEDLINE | ID: mdl-18765245

Snake venoms have demonstrated antinociceptive activity, and certain isolated neurotoxins have demonstrated significant analgesia in animal models. Here we report a novel analgesic toxin which was isolated from Naja naja atra and was given the name 'najanalgesin'. The LD(50) of the crude venom and najanalgesin were 0.89mg/kg and 2.69mg/kg, respectively. We used the writhing test and hot plate test to evaluate the antinociceptive properties of the crude venom and najanalgesin after intraperitoneal (ip) administration. The analgesic mechanism of najanalgesin was also studied. The response latency time was significantly prolonged in the hot plate test after ip administration of the crude venom of Naja naja atra (0.111-0.445mg/kg) in a dose-dependent manner. Najanalgesin (1mg/kg) elicited almost the same antinociceptive effect as that of the crude venom of Naja naja atra at the dose of 0.445mg/kg and remained for 6h after intraperitoneal injection, shown by hot plate test. The percentage of increase in the latency time for the venom and the najanalgesin 3h after drug administration was 96.2% and 112%, respectively. The number of writhes decreased to almost 1/3, 1/6, and 1/12 of the NS (physiological saline) group after intraperitoneal administration of najanalgesin at 0.25, 0.5, and 1.0mg/kg, respectively. Pretreatment with atropine (1mg/kg) or naloxone (3mg/kg) blocked the antinociception of najanalgesin in the hot plate test. Based on the sequence information, najanalgesin is found to be highly homologous with the conventional CTXs (cardiotoxins). To our knowledge, no study had previously reported that a toxin which was homologous with CTXs possessed the antinociceptive activity. Thus, this is the first report that the antinociceptive effect induced by najanalgesin is mediated by cholinergic and opioidergic mechanisms.


Analgesics/pharmacology , Cobra Neurotoxin Proteins/pharmacology , Neurotoxins/pharmacology , Analgesics/chemistry , Analgesics/isolation & purification , Animals , Atropine/pharmacology , Chromatography, High Pressure Liquid , Cobra Neurotoxin Proteins/chemistry , Cobra Neurotoxin Proteins/isolation & purification , Elapid Venoms/chemistry , Elapid Venoms/pharmacology , Lethal Dose 50 , Mice , Mice, Inbred Strains , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurotoxins/chemistry , Neurotoxins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Transl Res ; 150(5): 295-302, 2007 Nov.
Article En | MEDLINE | ID: mdl-17964518

A novel fibrinolytic enzyme, FII(a), was isolated from Agkistrodon acutus venom, which can degrade fibrin/fibrinogen and dissolve thrombus without activating plasminogen or influencing the activities of tissue plasminogen activator (t-PA) and plasminogen activator inhibitor type-1 (PAI-1). In this study, we evaluated the effect of FII(a) on lipopolysaccharide (LPS)-induced experimental disseminated intravascular coagulation (DIC) in rabbits, through the continuous infusion of 100-microg/kg/h LPS for a period of 6 h. Seven groups were established: LPS control, FII(a) (0.1, 0.3, and 0.6 mg/kg/h, respectively), heparin control (100 IU/kg/h), heparin + FII(a) (heparin 100 IU/kg/h associated with FII(a) 0.3 mg/kg/h), and a saline control group. A continuous injection of LPS induced a gradual impairment in hemostatic parameters, kidney fibrin deposition, and a high mortality rate. The intravenous administration of FII(a) improved the concentration of fibrinogen, the activities of protein C, plasminogen, t-PA, antithrombin III (ATIII), and PAI-1. Kidney fibrin deposition and the mortality also decreased. In the in vitro experiments, FII(a) can degrade fibrin/fibrinogen and high-dose FII(a) enhanced the activity of protein C. These findings suggest that the effects of FII(a) on LPS-induced DIC were from fibrinogen degradation and enhanced protein C activity. The simultaneous administration of FII(a) and heparin further improved all the hemostatic parameters, including decreased kidney fibrin deposition, and none of the rabbits died within 24 h, which indicates that the effects were mediated by degradation of fibrin/fibrinogen together with thrombin inhibition. We conclude that FII(a) may be useful in the treatment of DIC.


Crotalid Venoms/enzymology , Crotalid Venoms/therapeutic use , Disseminated Intravascular Coagulation/drug therapy , Fibrinolytic Agents/therapeutic use , Thrombosis/drug therapy , Animals , Antithrombin III/analysis , Disease Models, Animal , Disseminated Intravascular Coagulation/blood , Disseminated Intravascular Coagulation/etiology , Dose-Response Relationship, Drug , Fibrin/metabolism , Heparin/pharmacology , Injections, Intravenous , Kidney/blood supply , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/toxicity , Longevity/drug effects , Male , Metalloendopeptidases , Microcirculation/drug effects , Microcirculation/pathology , Plasminogen Activator Inhibitor 1/blood , Protein C/analysis , Rabbits , Tissue Plasminogen Activator/blood
11.
Acta Pharmacol Sin ; 28(10): 1580-4, 2007 Oct.
Article En | MEDLINE | ID: mdl-17883943

AIM: To purify and characterize the coagulant protein FIa from Daboia russelli siamensis (Myanmar) venom. METHODS: FIa was purified from Daboia russelli siamensis (Myanmar) venom by ion-exchange chromatography on CM-Sephadex C-50, and gel filtration on Sephadex G-75 and a Superdex 75 column. The hemostatic activity of FIa was determined by the method of Williams and Esnouf. The specific chromogenic substrates were used respectively to determine the activation of factor X and prothrombin. The fibrinogen-clotting activity of FIa was determined by the method of Gao et al. Normal saline was used as a negative control while factor Xa and thrombin were used as positive controls, respectively. RESULTS: FIa, a coagulant protein, was achieved by ion-exchange chromatography and gel filtration with a molecular weight of 34,479 and an isoelectric point of 7.2. FIa was shown to have strong hemostatic activity. The hemostatic activity of 0.5 mg FIa was equal to that of 1.5625 u thrombin. FIa primarily activated factor X, however, had no influence on prothrombin, nor did it cleave or clot fibrinogen. CONCLUSION: FIa is a factor X-activating enzyme, which could activate factor X to factor Xa, but has no effect on prothrombin and fibrinogen.


Metalloendopeptidases/isolation & purification , Viper Venoms/chemistry , Viperidae , Animals , Blood Coagulation/drug effects , Chromatography, Gel , Chromatography, Ion Exchange , Coagulants/chemistry , Coagulants/isolation & purification , Coagulants/pharmacology , Factor X/metabolism , Humans , Isoelectric Point , Molecular Weight , Prothrombin/metabolism , Viper Venoms/isolation & purification
12.
Acta Pharmacol Sin ; 28(2): 173-9, 2007 Feb.
Article En | MEDLINE | ID: mdl-17241518

AIM: This study was designed to investigate whether the activation of the phosphatidylinositol 3-kinase (PI3-K)/Akt pathway is required for thermal preconditioning to protect rat cerebellar granule neurons (CGN) against apoptosis induced by low potassium, and to explore the possibility of a link between the upregulated heat shock protein (HSP)70 expression and Akt activation in the acquisition of neuroprotection induced by thermal preconditioning. METHODS: CGN cultured for 8 d in vitro were switched to 5K medium for 24 h after thermal preconditioning (TP; 43.5 degree for 90 min, then 37 degree for 1 h). To study the role of the PI3-K/Akt pathway, a PI3-K inhibitor, LY294002 (20 micromol/L) was added into the cultures 1 h before TP. 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide (MTT) assay and fluorescein diacetate staining were used to determine cell viability. Hoechst 33258 staining and agar gel electrophoresis were used to test the morphological and biological characters of CGN. Western blot analysis was employed to detect the levels of phospho-Akt, phospho-glycogen synthase kinase 3beta (GSK3beta) Akt, GSK3beta, and HSP70. RESULTS: TP protected CGN against apoptosis induced by low potassium. LY294002 inhibited the neuroprotective effect on CGN induced by TP. TP induced a robust activation of Akt and the inactivation of GSK3beta via PI3-K. Furthermore, the activation of the PI3-K/Akt pathway by TP persisted for 24 h in the 5K cultures. LY294002 (20 micromol/L) failed to inhibit the upregulated HSP70 expression induced by TP. CONCLUSION: The activation of the PI3-K/Akt pathway is required for TP to protect CGN against apoptosis induced by low potassium, but the neuroprotective effect by Akt activation is not mediated through the downstream induction of HSP70 expression.


Apoptosis , Cerebellum/pathology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Chromones/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HSP70 Heat-Shock Proteins/biosynthesis , Hot Temperature , Morpholines/pharmacology , Phosphorylation , Potassium , Rats , Rats, Sprague-Dawley
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 22(2): 205-7, 2006 Mar.
Article Zh | MEDLINE | ID: mdl-16507260

AIM: To analyze YZ-2, a novel protein associated with cerebral ischemia and prepares its polyclonal antibody. METHODS: According to the bioinformatics analysis and prediction of the possible high structure, hydrophilicity and antigenicity of YZ-2, a 22-amino acid residue partial peptide of YZ-2 was synthesized. The synthesized peptide was then used to immunize. And the properties of anti-YZ2 were analyzed by ELISA and Western blot. RESULTS: The hydrophilicity and antigenicity were predicted by methods of bioinformatics. The polyclonal antibody of YZ-2 was successfully obtained and its specificity and sensitivity were conformed by ELISA and Western blot. CONCLUSION: By the bioinformatics analysis and prediction, the hydrophilicity and antigenicity of YZ2 were analyzed. The antibody of YZ-2 was successfully obtained.


Antibodies/immunology , Antibody Specificity , Computational Biology , Proteins/immunology , Animals , Blotting, Western , Clinical Laboratory Techniques , Enzyme-Linked Immunosorbent Assay , Male , Rabbits , Rats , Rats, Sprague-Dawley
14.
Toxicon ; 47(2): 241-8, 2006 Feb.
Article En | MEDLINE | ID: mdl-16386281

A novel metalloproteinase, recombinant fibrinogenase IV (rFIV(a)), was expressed and purified from Deinakistrodon acutus venom. It was a single chain protein with an apparent molecular weight 27 kDa and an isoeletric point of pH 7.1. RFIV(a) cleaved preferentially the Aalpha-chain and also cleaved Bbeta, gamma-chains of fibrinogen when the incubation time was prolonged. The proteolytic activity was inhibited by EDTA, l-cysteine, and DTT, indicating rFIV(a) was a metalloproteinase requiring disulfide bonds for its activity. It kept above 85% of the initial activity from pH 4.5-11, showed an equal maximum activity at the temperature range from 30 to 50 degrees C, and was inactivated by Zn2+, Cu2+ and Cd2+. Homology modeling of rFIV(a) showed that two highly conserved disulfide bonds (Cys159-Cys164 and Cys117-Cys197) was maintained from its structure, and it exhibited the characteristic conserved motif H142E143XXH146XXGXXH152, whose three histidine residues were involved in binding of the catalytically essential zinc ion. This work demonstrates the expression, purification and characterization of recombinant fibrinogenase IV, which belongs to class P-I metalloproteinase from D. acutus venom.


Fibrinogen/metabolism , Metalloproteases/chemistry , Metalloproteases/isolation & purification , Models, Molecular , Snake Venoms/enzymology , Snakes , Amino Acid Sequence , Animals , Enzyme Stability , Gene Expression , Hydrogen-Ion Concentration , Metalloproteases/metabolism , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Temperature
15.
Appl Microbiol Biotechnol ; 72(1): 72-76, 2006 Aug.
Article En | MEDLINE | ID: mdl-16429282

A novel metalloproteinase, recombinant fibrinogenase IV (rFIVa), was expressed and purified from Agkistrodon acutus venom. It is a single-chain protein with an apparent molecular weight of 27 kDa. Western blot showed that it had a good immunological reaction against anti-FIVa rabbit serum. The kinetic parameters Km and Kcat of rFIVa on the substrate T6140 were 7.471 x 10(-4) mol/l and 5.103 x 10(-5) s(-1). RFIVa cleaved preferentially the alpha-chain, and the beta- and gamma-chains of fibrinogen were also cleaved when the incubation time was prolonged. The administration of rFIVa (1.8 and 5.4 mg/kg) to animals with acute blood-stasis model produced a decrease in fibrinogen to control values. To our knowledge, this is the first report of the expression, purification, and evaluation of recombinant fibrinogenase IV, which belongs to class P-I metalloproteinase from A. acutus venom.


Agkistrodon , Crotalid Venoms/enzymology , Serine Endopeptidases/biosynthesis , Animals , Blotting, Western , Cloning, Molecular , Fibrinogen/metabolism , Kinetics , Models, Animal , Molecular Weight , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Yeasts/genetics , Yeasts/metabolism
16.
Acta Pharmacol Sin ; 26(12): 1474-8, 2005 Dec.
Article En | MEDLINE | ID: mdl-16297346

AIM: To study the enzymological characterization of a fibrinolytic enzyme (FII(a)) from Agkistrodon acutus venom. METHODS: The fibrinogenolytic effect and the influences of several protease inhibitors, chelating agents, and metal ions on fibrinogenolytic activity were visualized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The metal content of FII(a) was determined by atomic absorption spectroscopy. RESULTS: After incubation with FII(a) (0.25 g/L), Aalpha-, Bbeta- and gamma-chains of fibrinogen disappeared within 5 min, 30 min, and 8 h , respectively. The molecular weights of major degradation products were 45,000 and 41,000, which were different from those bands produced by plasmin. The fibrinogenolytic activity of FIIa was strongly inhibited by ethylenediamine tetraacetic acid (EDTA), ethyleneglycol tetraacetic acid (EGTA), dithiothreitol and cysteine, but not by phenylmethyl-sulfonyl fluoride and soybean trypsin inhibitor. Zinc (3171+/-25 mg/kg), potassium (489+/-17 mg/kg) and calcium (319+/-13 mg/kg) were found in FIIa. Zn2+, Ca2+ and Mg2+ could recover the fibrinogenolytic activity of FIIa, which was inhibited by EDTA. Only Ca2+ could recover the fibrinogenolytic activity inhibited by EGTA. CONCLUSION: FIIa can degrade the Aalpha-, Bbeta- and gamma-chains of fibrinogen. FII(a) is a metalloproteinase, and Zn2+, Ca2+, and disulfide bonds are necessary for its fibrinogenolytic activity.


Agkistrodon , Crotalid Venoms/metabolism , Fibrinogen/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/metabolism , Animals , Calcium/analysis , Calcium Chloride/pharmacology , Chlorides/pharmacology , Crotalid Venoms/antagonists & inhibitors , Crotalid Venoms/chemistry , Crotalid Venoms/isolation & purification , Cysteine/pharmacology , Dithiothreitol/pharmacology , Edetic Acid/pharmacology , Egtazic Acid/pharmacology , Fibrinolytic Agents/isolation & purification , Magnesium Chloride/pharmacology , Metalloendopeptidases , Metalloproteases/antagonists & inhibitors , Metalloproteases/isolation & purification , Metalloproteases/metabolism , Potassium/analysis , Zinc/analysis , Zinc Compounds/pharmacology
17.
Acta Pharmacol Sin ; 26(6): 691-5, 2005 Jun.
Article En | MEDLINE | ID: mdl-15916735

AIM: To investigate the fibrin(ogen)olytic character of FIIa isolated from Agkistrodon acutus venom in vitro and in vivo. METHODS: 125I-labeled human plasma clot lysis was measured in vitro and rabbit carotid artery thrombosis was as an in vivo model. RESULTS: In vitro, urokinase (UK) at 25, 35, 40, 45, 60 kU/L and FIIa at 0.08, 0.23, 0.4, 0.5, and 0.7 g/L resulted an equivalent clot lysis (20%, 40%, 50%, 60%, and 80%). UK at 25 to approximately 60 kU/L induced 27.3%+/-3.6%, 35.2%+/-2.3%, 39.3%+/-2.4%, 44.2%+/-4.6%, and 51.1%+/-1.2% fibrinogen degradation. But FIIa at 0.08 approximately -0.7 g/L induced 95.4%+/-0.3%, >95.6%, >95.6%, >95.6%, >95.6% fibrinogen degradation respectively. In vivo, UK 40 kU/kg and FIIa 1.0 mg/kg reduced the weight of residual thrombus to 9.0+/-2.5 mg and 7.8+/-3.5 mg compared with negative control group (30.0+/-5.4 mg). But the fibrinogen degradation rate after UK 40 kU/kg and FIIa 1.0 mg/kg treatment was 24.4%+/-6.2% and 4.1%+/-7.8%, respectively (P<0.05, n=6). The order of the lysis speed after UK 125 kU/L treatment was platelet poor plasma (PPP) clots>the whole blood clots>platelet rich plasma (PRP) clots. The sequence for FIIa 0.4 g/L was PRP >PPP >whole blood clots. CONCLUSION: At the same percentage of clot lysis, FIIa degraded more fibrinogen than UK did in vitro but less fibrinogen than UK did in vivo. The order of the lysis speed was PPP>whole blood clots>PRP clots for UK and PRP>PPP>whole blood clots for FIIa.


Agkistrodon , Carotid Artery Thrombosis/physiopathology , Crotalid Venoms/pharmacology , Fibrinolysis/drug effects , Animals , Crotalid Venoms/administration & dosage , Crotalid Venoms/chemistry , Crotalid Venoms/isolation & purification , Dose-Response Relationship, Drug , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/isolation & purification , Fibrinolytic Agents/pharmacology , Male , Metalloendopeptidases , Rabbits , Urokinase-Type Plasminogen Activator/pharmacology
18.
Acta Pharmacol Sin ; 25(4): 458-61, 2004 Apr.
Article En | MEDLINE | ID: mdl-15066213

AIM: To explore the possibility that expressions of different heat shock proteins (HSPs) are specifically involved in the protection of thermal preconditioning against apoptosis of cerebellar granule neurons (CGNs) induced by repolarization. METHODS: Western blotting was used to detect expressions of HSP27, HSP70, and HSP90 induced by thermal preconditioning (TP) in CGNs; reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the expression level of HSP70 mRNA; apoptosis of CGNs was induced by switching culture medium containing KCl 25 mmol/L to one containing KCl 5 mmol/L. RESULTS: No expression of HSP27 in cerebellar granule neurons was observed with TP at 44 degree. Expression of HSP90 was obvious in CGNs both without and with TP at 44 degree for different periods. Expression of HSP70 protein in CGNs was lower with TP at 44 degree for 5 min, but it increased gradually after the period was prolonged to 30, 60, or 90 min. HSP70 mRNA was detected after TP 44 degree for 30, 60, and 90 min and increased gradually with time. HSP70 antisense oligodeoxynucleotides 10 micromol/L for 72 h inhibited the protective effects of TP at 44 degree on apoptosis of CGNs induced by repolarization. CONCLUSION: HSP70 is involved in protective effects of thermal preconditioning on apoptosis in cerebellar granule neurons induced by repolarization.


Apoptosis , Cerebellum/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , Hot Temperature , Oligodeoxyribonucleotides, Antisense/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebellum/cytology , Female , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Male , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
19.
Acta Pharmacol Sin ; 25(4): 514-21, 2004 Apr.
Article En | MEDLINE | ID: mdl-15066223

AIM: To study the local hemorrhagic activity of a fibrinolytic enzyme (FII(a)) from Agkistrodon acutus venom and its mechanism. METHODS: The local hemorrhagic activity was determined by subcutaneous injection on the back of mouse. The effects of FIIa on factor X, prothrombin, gelatin, and collagen were visualized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Platelet aggregation assays were performed in rat platelet-rich plasma (PRP). Human umbilical vein endothelial cells (HUVEC) were cultured and passaged in complete M199 medium. Cell viability and nuclear morphology change were determined by fluorescein diacetate (FDA) staining and Hoechst 33258 staining, respectively. RESULTS: The minimum hemorrhagic dose (MHD) of FIIa was 89 microg. In vitro, FII(a) (0.25 g/L) degraded factor X, prothrombin, collagen, and gelatin, and dose-dependently (0.25, 0.50, 0.75, and 1.00 g/L) inhibited the platelet aggregation induced by ADP in rat PRP. When HUVEC in culture treated with FII(a), HUVEC showed detachment in a dose-dependent manner, but no apoptosis sign was observed. CONCLUSION: FIIa had local hemorrhagic activity, and the mechanism was related to the degradation of factor X, prothrombin, gelatin, and collagen, the inhibition of ADP-induced platelet aggregation, and inducement of HUVEC detachment.


Agkistrodon , Crotalid Venoms/chemistry , Crotalid Venoms/pharmacology , Endothelial Cells/drug effects , Fibrinolytic Agents/pharmacology , Animals , Cells, Cultured , Collagen/metabolism , Crotalid Venoms/isolation & purification , Endothelial Cells/cytology , Factor X/metabolism , Female , Humans , Male , Metalloendopeptidases , Mice , Platelet Aggregation/drug effects , Prothrombin/metabolism , Rats , Rats, Sprague-Dawley , Umbilical Veins/cytology
20.
Acta Pharmacol Sin ; 24(6): 539-48, 2003 Jun.
Article En | MEDLINE | ID: mdl-12791180

AIM: To investigate possible intracellular signal molecules involved in diphenylhydantoin (DPH)-mediated apoptosis of cerebellar granule neurons (CGN) and explore possible molecular mechanisms of neurotoxicity of DPH. METHODS: Fluorescein diacetate (FDA) stain, hochest 33258 stain, and agar gel electrophoresis were used to test morphological and biological characters of primary CGN and cortical neurons (CN) in the presence or absence of 100 micromol/L DPH; Western blot and RT-PCR were employed to further investigate apoptotic/survival signal moleculars involved in the neuronal apoptotic signal transdution. RESULTS: DPH 100 micromol/L induced a typical apoptosis of CGN but had no toxicity on CN. Cerebellar granule neural apoptosis induced by 100 micromol/L DPH was significantly inhibited by pre-treatment with SB203580 (10 micromol/L) or CEP-11004 (1 micromol/L) for 1 h. DPH markedly upregulated the levels of phospho-c-Jun (active c-Jun), total c-Jun protein and c-jun mRNA in CGN. The levels of phospho-c-Jun dramatically elevated by DPH at 8 h were significantly inhibited by SB203580 (10 micromol/L) or CEP-11004 (1 micromol/L). Moreover, the activities of p44/42 (ERK1/ERK2), other members of MAP kinases and generally believed to be important survival effetors in CGN, were markedly suppressed. However, the activities of both JNK and p38 were little affected in the process of apoptosis of CGN induced by 100 micromol/L DPH. CONCLUSION: The selective toxicity of DPH on CGN is likely due to its ability to induce apoptosis of CGN, it is a process involved activation of c-Jun and suppression of the activity of p44/42.


Apoptosis , Mitogen-Activated Protein Kinase 1/metabolism , Neurons/drug effects , Phenytoin/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Animals , Anticonvulsants/pharmacology , Cell Survival , Cells, Cultured , Cerebellar Cortex/cytology , Cytoplasmic Granules/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
...