首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Bioassays of insecticidal proteins from Bacillus thuringiensis subsp. israelensis with larvae of the malaria vector mosquito Anophelesalbimanus showed that the cytolytic protein Cyt1Aa was not toxic alone, but it increased the toxicity of the crystalline proteins Cry4Ba and Cry11Aa. Synergism also occurred between Cry4Ba and Cry11Aa toxins. Whereas many previous analyses of synergism have been based on a series of toxin concentrations leading to comparisons between expected and observed values for the concentration killing 50% of insects tested (LC50), we describe and apply a method here that enables testing for synergism based on single concentrations of toxins.  相似文献   

2.
Bacillus thuringiensis ssp. israelensis (Bti) has been used worldwide for the control of dipteran insect pests. This bacterium produces several Cry and Cyt toxins that individually show activity against mosquitoes but together show synergistic effect. Previous work demonstrated that Cyt1Aa synergizes the toxic activity of Cry11Aa by functioning as a membrane-bound receptor. In the case of Cry toxins active against lepidopteran insects, receptor interaction triggers the formation of a pre-pore oligomer that is responsible for pore formation and toxicity. In this work we report that binding of Cry11Aa to Cyt1Aa facilitates the formation of a Cry11Aa pre-pore oligomeric structure that is capable of forming pores in membrane vesicles. Cry11Aa and Cyt1A point mutants affected in binding and in synergism had a correlative effect on the formation of Cry11Aa pre-pore oligomer and on pore-formation activity of Cry11Aa. These data further support that Cyt1Aa interacts with Cry11Aa and demonstrate the molecular mechanism by which Cyt1Aa synergizes or suppresses resistance to Cry11Aa, by providing a binding site for Cry11Aa that will result in an efficient formation of Cry11Aa pre-pore that inserts into membranes and forms ionic pores.  相似文献   

3.
Bacillus thuringiensis Cry4Aa toxin was previously shown to be much more toxic to Culex mosquito-larvae than its closely related toxin – Cry4Ba, conceivably due to their sequence differences within the β10-β11 receptor-binding loop. Here, single-Ala substitutions of five residues (Pro510, Thr512, Tyr513, Lys514 and Thr515) within the Cry4Aa β10-β11 loop revealed that only Lys514 corresponding to the relative position of Cry4Ba-Asp454 is crucial for toxicity against Culex quinquefasciatus larvae. Interestingly, charge-reversal mutations at Cry4Ba-Asp454 (D454R and D454K) revealed a marked increase in toxicity against such less-susceptible larvae. In situ binding analyses revealed that both Cry4Ba-D454R and D454K mutants exhibited a significant increase in binding to apical microvilli of Culex larval midguts, albeit at lower-binding activity when compared with Cry4Aa. Altogether, our present data suggest that a positively charged side-chain near the tip of the β10-β11 loop plays a critical role in determining target specificity of Cry4Aa against Culex spp., and hence a great increase in the Culex larval toxicity of Cry4Ba was obtained toward an opposite-charge conversion of the corresponding Asp454.  相似文献   

4.
Bacillus thuringiensis subs. israelensis produces at least three Cry toxins (Cry4Aa, Cry4Ba, and Cry11Aa) that are active against Aedes aegypti larvae. Previous work characterized a GPI-anchored alkaline phosphatase (ALP1) as a Cry11Aa binding molecule from the gut of A. aegypti larvae. We show here that Cry4Ba binds ALP1, and that the binding and toxicity of Cry4Ba mutants located in loop 2 of domain II is correlated. Also, we analyzed the contribution of ALP1 toward the toxicity of Cry4Ba and Cry11Aa toxins by silencing the expression of this protein though RNAi. Efficient silencing of ALP1 was demonstrated by real-time quantitative PCR (qPCR) and Western blot. ALP1 silenced larvae showed tolerance to both Cry4Ba and Cry11Aa although the silenced larvae were more tolerant to Cry11Aa in comparison to Cry4Ba. Our results demonstrate that ALP1 is a functional receptor that plays an important role in the toxicity of the Cry4Ba and Cry11Aa proteins.  相似文献   

5.
本研究测定了分别表达苏云金芽孢杆菌Cry4Aa、Cry4Ba、Cry11Aa、Cyt1Aa和球形芽孢杆菌二元毒素Bin的转化菌株Bt B60 1、Bt B611、Bt B640、Bt U 30和Bt CW 3全发酵培养物两两或两两以上不同组合对抗性库蚊的毒力 ,分析了杀蚊毒素间的协同作用。结果表明 ,Bin和Cry4Aa、Bin和Cry 4Ba间有明显的协同作用 ,此外 ,Cry4Aa和Cry4Ba、Cry4Aa和Cry11Aa、Cyt1Aa和Cry4Aa之间也有明显的协同作用  相似文献   

6.
Three members of the δ-endotoxin group of toxins expressed by Bacillus thuringiensis subsp. israelensis, Cyt2Ba, Cry4Aa and Cry11A, were individually expressed in recombinant acrystalliferous B. thuringiensis strains for in vitro evaluation of their toxic activities against insect and mammalian cell lines. Both Cry4Aa and Cry11A toxins, activated with either trypsin or Spodoptera frugiperda gastric juice (GJ), resulted in different cleavage patterns for the activated toxins as seen by SDS-PAGE. The GJ-processed proteins were not cytotoxic to insect cell cultures. On the other hand, the combination of the trypsin-activated Cry4Aa and Cry11A toxins yielded the highest levels of cytotoxicity to all insect cells tested. The combination of activated Cyt2Ba and Cry11A also showed higher toxic activity than that of toxins activated individually. When activated Cry4Aa, Cry11A and Cyt2Ba were used simultaneously in the same assay a decrease in toxic activity was observed in all insect cells tested. No toxic effect was observed for the trypsin-activated Cry toxins in mammalian cells, but activated Cyt2Ba was toxic to human breast cancer cells (MCF-7) when tested at 20 µg/mL.  相似文献   

7.
Bacillus thuringiensis serovar israelensis (B. thuringiensis subsp. israelensis) produces four insecticidal crystal proteins (ICPs) (Cry4A, Cry4B, Cry11A, and Cyt1A). Toxicity of recombinant B. thuringiensis subsp. israelensis strains expressing only one of the toxins was determined with first instars of Tipula paludosa (Diptera: Nematocera). Cyt1A was the most toxic protein, whereas Cry4A, Cry4B, and Cry11A were virtually nontoxic. Synergistic effects were recorded when Cry4A and/or Cry4B was combined with Cyt1A but not with Cry11A. The binding and pore formation are key steps in the mode of action of B. thuringiensis subsp. israelensis ICPs. Binding and pore-forming activity of Cry11Aa, which is the most toxic protein against mosquitoes, and Cyt1Aa to brush border membrane vesicles (BBMVs) of T. paludosa were analyzed. Solubilization of Cry11Aa resulted in two fragments, with apparent molecular masses of 32 and 36 kDa. No binding of the 36-kDa fragment to T. paludosa BBMVs was detected, whereas the 32-kDa fragment bound to T. paludosa BBMVs. Only a partial reduction of binding of this fragment was observed in competition experiments, indicating a low specificity of the binding. In contrast to results for mosquitoes, the Cyt1Aa protein bound specifically to the BBMVs of T. paludosa, suggesting an insecticidal mechanism based on a receptor-mediated action, as described for Cry proteins. Cry11Aa and Cyt1Aa toxins were both able to produce pores in T. paludosa BBMVs. Protease treatment with trypsin and proteinase K, previously reported to activate Cry11Aa and Cyt1Aa toxins, respectively, had the opposite effect. A higher efficiency in pore formation was observed when Cyt1A was proteinase K treated, while the activity of trypsin-treated Cry11Aa was reduced. Results on binding and pore formation are consistent with results on ICP toxicity and synergistic effect with Cyt1Aa in T. paludosa.  相似文献   

8.
The insecticidal Cry11Aa and Cyt1Aa proteins are produced by Bacillus thuringiensis as crystal inclusions. They work synergistically inducing high toxicity against mosquito larvae. It was proposed that these crystal inclusions are rapidly solubilized and activated in the gut lumen, followed by pore formation in midgut cells killing the larvae. In addition, Cyt1Aa functions as a Cry11Aa binding receptor, inducing Cry11Aa oligomerization and membrane insertion. Here, we used fluorescent labeled crystals, protoxins or activated toxins for in vivo localization at nano-scale resolution. We show that after larvae were fed solubilized proteins, these proteins were not accumulated inside the gut and larvae were not killed. In contrast, if larvae were fed soluble non-toxic mutant proteins, these proteins were found inside the gut bound to gut-microvilli. Only feeding with crystal inclusions resulted in high larval mortality, suggesting that they have a role for an optimal intoxication process. At the macroscopic level, Cry11Aa completely degraded the gastric caeca structure and, in the presence of Cyt1Aa, this effect was observed at lower toxin-concentrations and at shorter periods. The labeled Cry11Aa crystal protein, after midgut processing, binds to the gastric caeca and posterior midgut regions, and also to anterior and medium regions where it is internalized in ordered “net like” structures, leading finally to cell break down. During synergism both Cry11Aa and Cyt1Aa toxins showed a dynamic layered array at the surface of apical microvilli, where Cry11Aa is localized in the lower layer closer to the cell cytoplasm, and Cyt1Aa is layered over Cry11Aa. This array depends on the pore formation activity of Cry11Aa, since the non-toxic mutant Cry11Aa-E97A, which is unable to oligomerize, inverted this array. Internalization of Cry11Aa was also observed during synergism. These data indicate that the mechanism of action of Cry11Aa is more complex than previously anticipated, and may involve additional steps besides pore-formation activity.  相似文献   

9.
Bacillus thuringiensis serovar israelensis (B. thuringiensis subsp. israelensis) produces four insecticidal crystal proteins (ICPs) (Cry4A, Cry4B, Cry11A, and Cyt1A). Toxicity of recombinant B. thuringiensis subsp. israelensis strains expressing only one of the toxins was determined with first instars of Tipula paludosa (Diptera: Nematocera). Cyt1A was the most toxic protein, whereas Cry4A, Cry4B, and Cry11A were virtually nontoxic. Synergistic effects were recorded when Cry4A and/or Cry4B was combined with Cyt1A but not with Cry11A. The binding and pore formation are key steps in the mode of action of B. thuringiensis subsp. israelensis ICPs. Binding and pore-forming activity of Cry11Aa, which is the most toxic protein against mosquitoes, and Cyt1Aa to brush border membrane vesicles (BBMVs) of T. paludosa were analyzed. Solubilization of Cry11Aa resulted in two fragments, with apparent molecular masses of 32 and 36 kDa. No binding of the 36-kDa fragment to T. paludosa BBMVs was detected, whereas the 32-kDa fragment bound to T. paludosa BBMVs. Only a partial reduction of binding of this fragment was observed in competition experiments, indicating a low specificity of the binding. In contrast to results for mosquitoes, the Cyt1Aa protein bound specifically to the BBMVs of T. paludosa, suggesting an insecticidal mechanism based on a receptor-mediated action, as described for Cry proteins. Cry11Aa and Cyt1Aa toxins were both able to produce pores in T. paludosa BBMVs. Protease treatment with trypsin and proteinase K, previously reported to activate Cry11Aa and Cyt1Aa toxins, respectively, had the opposite effect. A higher efficiency in pore formation was observed when Cyt1A was proteinase K treated, while the activity of trypsin-treated Cry11Aa was reduced. Results on binding and pore formation are consistent with results on ICP toxicity and synergistic effect with Cyt1Aa in T. paludosa.  相似文献   

10.
Bacillus thuringiensis ssp. israelensis (Bti) produces four Cry toxins (Cry4Aa, Cry4Ba, Cry10Aa and Cry11Aa), and two Cyt proteins (Cyt1Aa and Cyt2Ba), toxic to mosquito‐larvae of the genus Aedes, Anopheles and Culex, important human disease vectors that transmit dengue virus, malaria and filarial parasites respectively. Previous work showed that Bti is highly toxic to Anopheles albimanus, the main vector for transmission of malaria in Mexico. In this work, we analysed the toxicity of isolated Cry proteins of Bti and identified an An. albimanus midgut protein as a putative Cry4Ba and Cry11Aa receptor molecule. Biossays showed that Cry4Ba and Cry11Aa of Bti are toxic to An. albimanus larvae. Ligand blot assays indicated that a 70 kDa glycosylphosphatidylinositol‐anchored protein present in midgut brush border membrane vesicles of An. albimanus interacts with Cry4Ba and Cry11Aa toxins. This protein was identified as an α‐amylase by mass spectrometry and enzymatic activity assays. The cDNA that codes for the α‐amylase was cloned by means of 5′‐ and 3′‐RACE experiments. Recombinant α‐amylase expressed in Escherichia coli specifically binds Cry4Ba and Cry11Aa toxins.  相似文献   

11.
Cry11Aa and Cyt1Aa of Bacillus thuringiensis are active against mosquitoes and show synergism. Cyt1Aa functions as a membrane receptor inducing Cry11Aa oligomerization. Here we characterized Cry11Aa helix α-3 mutants impaired in oligomerization and toxicity against Aedes aegypti, indicating that oligomerization of Cry11Aa is important for toxin action. Cyt1Aa did not recover the insecticidal activity of Cry11Aa mutants.Bacillus thuringiensis subsp. israelensis has been used worldwide for the control of different mosquitoes that are vectors of several human diseases (10, 11). This bacterium produces different toxins that individually show activity against mosquitoes, i.e., Cry4Aa, Cry4Ba, Cry11Aa, and Cyt1Aa (2). The toxicity of Cry11Aa and Cry4 toxins against Aedes aegypti is greatly increased in the presence of sublethal concentrations of Cyt1Aa (14). Also, Cyt1Aa overcomes the resistance of the Culex quinquefasciatus population to Cry11Aa (12, 13). Cyt1Aa synergizes the toxic activity of Cry11Aa by functioning as a Cry11Aa receptor, facilitating the oligomerization of Cry11Aa and its pore formation activity (7, 8). Oligomerization is a complex event that involves interaction with a toxin receptor and further proteolysis of helix α-1 (3). In the case of the Cry1Ab toxin, helix α-3 of domain I contains coiled-coil structures that are important for oligomerization (4). Some point mutations in helix α-3 do not affect interaction with receptors but severely affected oligomerization, influencing pore formation and toxicity against Manduca sexta larvae (4).Since binding with Cyt1Aa facilitates Cry11Aa oligomerization, we hypothesize that Cry11Aa mutants unable to oligomerize would be affected in synergism with Cyt1Aa and in toxicity. In this report, we analyzed the effect of point mutations in helix α-3 of Cry11Aa on oligomerization, synergism with Cyt1Aa, and toxicity against A. aegypti larvae.Helix α-3 of Cry11Aa potentially forms coiled-coil structures, as determined by the program COILS, which calculates the probability that a sequence will adopt a coiled-coil conformation (6). The coiled-coil structures are characterized by heptads of residues (abcdefg), where positions a and d are occupied mostly by apolar residues and g and e by charged residues. Here we mutagenized some residues located at positions g and a of the predicted coiled-coil (Fig. (Fig.1).1). Substitutions R90E, E97A, Y98E, V104E, and S105E were produced by site-directed mutagenesis (Quick Change; Stratagene, La Jolla, CA) using the pCG6 plasmid (1) as a template and appropriate mutagenic oligonucleotides. Point mutations were verified by automated DNA sequencing at Instituto de Biotecnología-UNAM and transformed into the acrystalliferous B. thuringiensis 407 strain. B. thuringiensis strains were grown in solid nutrient broth sporulation medium supplemented with 10 μg/ml erythromycin (5). Crystal inclusions were purified as described previously (8) and solubilized in 100 mM NaOH for 1 h at 4°C. After solubilization, the Cry11Aa protoxins were dialyzed for 12 h against 50 mM Na2CO3, pH 10.5. The pH was equilibrated at pH 8.6 with equal volumes of 1 M Tris-HCl, pH 8, and protoxins were activated with trypsin (1:50, wt/wt) for 2 h at 25°C. All mutants, with the exception of the V104E mutant, which was not analyzed further, produced crystal inclusions similar to those for the wild-type toxin, composed of a 70-kDa protoxin (Fig. (Fig.2A).2A). After trypsin activation, all mutants produced two polypeptides of 32 and 36 kDa, similarly to the Cry11Aa toxin, suggesting that these mutations did not cause a major structural disturbance (Fig. (Fig.2B).2B). The Cry11Aa and mutant activated toxins were analyzed by circular dichroism spectroscopy (Fig. (Fig.2C).2C). The activated toxins were dialyzed against 10 mM Na2HPO4, 50 mM NaF, pH 9, and then purified by anion-exchange chromatography with HiTrap Q-Sepharose (Pharmacia LKB Biotechnology) in the same buffer, using a linear NaF gradient from 50 to 400 mM. The similarities among the curves indicate that the mutant toxins have a structure similar to that of the wild-type toxin.Open in a separate windowFIG. 1.Schematic representation of the coiled-coil structures of the α-3 helices of Cry1Ab and Cry11Aa toxins. The positions of residues a, b, c, d, e, f, and g of the heptads are presented. The mutated residues in both toxins that affected oligomerization and toxicity are shown in boldface type (reference 4 and this work).Open in a separate windowFIG. 2.SDS-PAGE analysis and circular dichroism spectra of Cry11Aa mutant toxins. (A) The Cry11Aa protoxins were solubilized at pH 10.5 and analyzed by SDS-PAGE (15% acrylamide). (B) SDS-PAGE analysis (15% acrylamide) of the activated toxins with trypsin. Both SDS-polyacrylamide gels were stained with Coomassie blue. Lanes 1, Cry11Aa; lanes 2, E97A mutant; lanes 3, Y98E mutant; lanes 4, R90E mutant; lanes 5, S105E mutant. (C) Analysis of the secondary-structure compositions of the mutants and Cry11Aa activated toxins. Circular dichroism spectra were recorded with a Jasco model J-715 spectropolarimeter equipped with a Peltier temperature control supplied by Jasco. Spectra were collected from 190 to 250 nm. Eight replicate spectra were collected for each sample to improve the signal-to-noise ratios. The final purified-protein concentration was 0.3 mg/ml, and spectra were collected in a 0.1-cm-pathlength cell. The secondary-structure prediction was performed using the CDSSTR algorithm (1a, 11a). Solid black line, Cry11Aa; dotted black line, E97A mutant; dashed black line, Y98E mutant; solid gray line, R90E mutant; dotted gray line, S105E mutant; MRE, mean residue ellipticity; [θ], ellipticity.The toxicity of spore/crystal suspensions of Cry11Aa or the individual mutants (75 to 10,000 ng/ml) was analyzed with bioassays against 10 fourth-instar A. aegypti larvae reared at 28°C, 87% humidity, and 12:12 light-dark conditions in 100 ml dechlorinated water, and mortality was scored after 24 h (four independent assays). The Cry11Aa toxin showed a mean lethal concentration of 355 ng/ml, with 95% confidence limits of 265 to 446 (Probit analysis using Polo-PC LeOra Software). In contrast, the R90E, E97A, Y98E, and S105E mutants were severely affected in toxicity against A. aegypti larvae, since no mortality was observed at the highest concentration used (10,000 ng/ml).We then analyzed the oligomerization of Cry11Aa toxins as previously described (8). Small unilamelar vesicles (SUV), composed of egg yolk phosphatidyl choline, cholesterol (Avanti Polar Lipids, Alabaster, AL), and stearylamine (Sigma, St. Louis, MO) at a 10:3:1 proportion, respectively, were used (8). Cyt1Aa was purified from the 4Q7/pWF45 strain (14) grown as described above. Cyt1Aa inclusions were purified by sucrose gradients, solubilized in 50 mM Na2CO3, 10 mM dithiothreitol, pH 10.5 (2 h at 30°C), and activated with 1:100 proteinase K (Sigma-Aldrich Co.), wt/wt, for 20 min at 30°C.For oligomerization assays, 2.5 μg soluble Cry11Aa or mutant protoxin was incubated for 2 h at 37°C in a 100-μl final volume of 50 mM Na2CO3, pH 10.5, with 200 μM SUV, 1:50 trypsin (wt/wt), and 0.5 μg Cyt1Aa activated toxin. After 2 h of incubation, 1 mM phenylmethylsulfonyl fluoride was added to stop the reaction, and the membrane fraction was separated by centrifugation (1 h at 100,000 × g). The pellet was suspended in the same buffer solution. Oligomeric structures of Cry toxins are highly stable after boiling as well as after urea denaturation (9). The suspension was boiled for 4 min, analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) (8% acrylamide), and electrotransferred onto polyvinylidene difluoride membranes (Millipore, Bedford, MA). The oligomeric and monomeric structures of Cry11Aa were detected using polyclonal anti-Cry11Aa antibody (1/15,000; 1 h) and a secondary antibody coupled with horseradish peroxidase (Sigma, St. Louis, MO) (1/5,000; 1 h) followed by luminol (ECL; Amersham Pharmacia Biotech) as described by the manufacturers. Figure Figure33 shows that only the Cry11Aa wild-type toxin was able to oligomerize, while the mutants were severely impaired in oligomerization.Open in a separate windowFIG. 3.Analysis of Cry11Aa oligomer formation. Soluble Cry11Aa protoxin was activated with trypsin for 2 h at 37°C in the presence of SUV and Cyt1Aa activated toxin. The membrane fraction was separated by ultracentrifugation, and the Cry11Aa protein was analyzed by Western blotting of the membrane pellet with polyclonal anti-Cry11A antibody. The sizes of the proteins were estimated from a molecular prestained plus standard, all blue (Bio-Rad). Lane 1, Cry11Aa; lane 2, R90E mutant; lane 3, Y98E mutant; lane 4, E97A mutant; lane 5, S105E mutant.Finally, the synergistic activity between Cyt1Aa and Cry11Aa was analyzed. A concentration of Cyt1Aa that produced 10% mortality was assayed in the presence of a protein concentration of wild-type Cry11A that produced 20% mortality. Larvae were examined 24 h after treatment, in three repetitions. This particular protein mixture produced a synergism factor of 8. Under these conditions, mortality was more than 80%, due to the synergistic activities of both toxins. Similar experiments were performed with the mutant toxins, using the same concentration of Cyt1Aa toxin and different concentrations (up to 6,000 ng/ml) of the mutant toxins. Cyt1A did not increase the toxicity of the Cry11Aa mutants, since only 10% mortality was observed, even at the highest concentration of the mutant toxins.Previously, helix α-3 of a lepidopteran-specific toxin (Cry1Ab) was subjected to mutagenesis. The R99E and Y107E mutants of the Cry1Ab toxin were severely impaired in oligomerization and toxicity, showing that oligomer formation is a necessary step to kill the larvae (4). The data presented here indicate that oligomer formation is also an essential step in the mechanism of toxicity of the mosquitocidal Cry11Aa toxin and that helix α-3 is involved in this process.  相似文献   

12.
A 2,175-bp modified gene (cry11Ba-S1) encoding Cry11Ba from Bacillus thuringiensis subsp. jegathesan was designed and the recombinant protein was expressed as a fusion protein with glutathione S-transferase in Escherichia coli. The recombinant Cry11Ba was highly toxic against Culex pipiens mosquito larvae, being nine and 17 times more toxic than mosquitocidal Cry4Aa and Cry11Aa from Bacillus thuringiensis subsp. israelensis, respectively. Interestingly, a further increase in the toxicity of the recombinant Cry11Ba was achieved by mixing with Cry4Aa, but not with Cry11Aa. These findings suggested that Cry11Ba worked synergistically with Cry4Aa, but not with Cry11Aa, in exhibiting toxicity against C. pipiens larvae. On the other hand, the amount of Cry toxin bound to brush border membrane vesicles (BBMVs) did not significantly change between individual toxins and the toxin mixtures, suggesting that the increase in toxins binding to BBMVs was not a reason for the observed synergistic effect. It is generally accepted that synergism of toxins is a potentially powerful tool for enhancing insecticidal activity and managing Cry toxin resistance in mosquitoes. The mixture of Cry4Aa and Cry11Ba in order to increase toxicity would be very valuable in terms of mosquito control.  相似文献   

13.
The bioinsecticide Bacillus thuringiensis var. israelensis (Bti) is increasingly used worldwide for mosquito control. Although no established resistance to Bti has been described in the field so far, a resistant Aedes aegypti strain (LiTOX strain) was selected in the laboratory using field‐collected leaf litter containing Bti toxins. This selected strain exhibits a moderate resistance level to Bti, but a high resistance level to individual Cry toxins. As Bti contains four different toxins, generalist resistance mechanisms affecting mosquito tolerance to different toxins were expected in the resistant strain. In the present work, we show that the resistant strain exhibits an increase of various gut proteolytic activities including trypsins, leucine‐aminopeptidases, and carboxypeptidase A activities. These elevated proteolytic activities resulted in a faster activation of Cry4Aa protoxins while Cry4Ba or Cry11Aa were not affected. These results suggest that changes in proteolytic activities may contribute to Bti resistance in mosquitoes together with other mechanisms.  相似文献   

14.
在蚊幼虫生活水域里的离中不粘柄菌(Asticcacaulis excentricus,Ae)中已成功表达苏云金芽孢杆菌以色列亚种(Bacillus thuringiensis subsp.israelensis,Bti)杀蚊蛋白基因cry11Aa的基础上,将另一Bti杀蚊蛋白基因cyt1Aa转化入Ae中表达。构建并转化了分别单独含有cyt1Aa基因、及同时含有cry11Aa基因的表达质粒pSODCyt20和pSODCryCyt20,蛋白免疫杂交检测相应的Ae重组子分别表达产生了Cyt1Aa和Cry11Aa蛋白。为了探究Ae(pSODCryCyt20)重组子不能表达cyt1Aa的原因,提取了重组子总RNA、并与同是革兰氏染色阴性的大肠杆菌的总RNA比较,结果显示两者RNA系统显著不同,推测Ae中多个外源基因的表达,可能要求每个基因必需一个启动子。  相似文献   

15.
Transgenic rice to control stem borer damage is under development in China. To assess the potential of Bacillus thuringiensis (Bt) transgenes in stem borer control, the toxicity of five Bt protoxins (Cry1Aa, Cry1Ab, Cry1Ac, Cry1Ba and Cry1Ca) against two rice stem borers, Sesamia inferens (pink stem borer) and Chilo suppressalis (striped stem borer), was evaluated in the laboratory by feeding neonate larvae on artificial diets containing Bt protoxins. The results indicated that Cry1Ca exhibited the highest level of toxicity to both stem borers, with an LC50 of 0.24 and 0.30 μg/g for C. suppressalis and S. inferens, respectively. However, S. inferens was 4-fold lower in susceptibility to Cry1Aa, and 6- and 47-fold less susceptible to Cry1Ab and Cry1Ba, respectively, compared to C. suppressalis. To evaluate interactions among Bt protoxins in stem borer larvae, toxicity assays were performed with mixtures of Cry1Aa/Cry1Ab, Cry1Aa/Cry1Ca, Cry1Ac/Cry1Ca, Cry1Ac/Cry1Ba, Cry1Ab/Cry1Ac, Cry1Ab/Cry1Ba, and Cry1Ab/Cry1Ca at 1:1 (w/w) ratios. All protoxin mixtures demonstrated significant synergistic toxicity activity against C. suppressalis, with values of 1.6- to 11-fold higher toxicity than the theoretical additive effect. Surprisingly, all but one of the Bt protoxin mixtures were antagonistic in toxicity to S. inferens. In mortality-time response experiments, S. inferens demonstrated increased tolerance to Cry1Ab and Cry1Ac compared to C. suppressalis when treated with low or high protoxin concentrations. The data indicate the utility of Cry1Ca protoxin and a Cry1Ac/Cry1Ca mixture to control both stem borer populations.  相似文献   

16.
Quality control of Bacillus thuringiensis ssp. israelensis (Bti) products is currently based on international toxic units (ITUs). The potency of products is related to the activity of a standard (IPS-82, Institute Pasteur, Paris) assessed in bioassays using Aedes aegypti as a target host. The procedure is time consuming and costly, often producing variable results. The activity of Bti is based on four different insecticidal crystal proteins (ICPs): Cry4Aa, Cry4Ba, Cry11Aa and Cyt1Aa. Monoclonal antibodies were produced using IPS-82 for immunisation and an enzyme-linked immunosorbent assay (ELISA) was developed. Antibodies were selected with specificity against Cry11A and Cyt1A. Cry4 specific antibodies could not distinguish between Cry4A and Cry4B. Within five replicate assessments of the three ICPs (in µg mg-1 ICP protein), an error between 3 and 8% was recorded, whereas a 14% error was obtained comparing seven samples of the same production batch for ITUs mg-1. The toxicity against A. aegypti expressed in ITUs correlated well with the results of the ELISA (correlation coefficient r Cyt 1=0.79; Cry 11=0.87; Cry 4=0.91) also when related to the sum of all ICPs (r=0.87). The ELISA can reduce efforts to determine Bti quality compared with the labour-intensive and variable ITU bioassay.  相似文献   

17.
18.
Bacillus thuringiensis subs israelensis produces Cry toxins active against mosquitoes. Receptor binding is a key determinant for specificity of Cry toxins composed of three domains. We found that exposed loop alpha-8 of Cry11Aa toxin, located in domain II, is an important epitope involved in receptor interaction. Synthetic peptides corresponding to exposed regions in domain II (loop alpha-8, beta-4 and loop 3) competed binding of Cry11Aa to membrane vesicles from Aedes aegypti midgut microvilli. The role of loop alpha-8 of Cry11A in receptor interaction was demonstrated by phage display and site-directed mutagenesis. We isolated a peptide-displaying phage (P5.tox), that recognizes loop alpha-8 in Cry11Aa, interferes interaction with the midgut receptor and attenuates toxicity in bioassay. Loop alpha-8 mutants affected in toxicity and receptor binding were characterized.  相似文献   

19.
利用穿梭载体pBU4,将苏云金杆菌以色列亚种(Bti)的cry4Aa、cry4Ba和cry11Aa基因分别转入Bti无晶体突变株4Q7中,获得了转化菌株Bt-B601、Bt-B611和Bt-B640。SDS-PAGE结果显示:cry4Aa、cry4Ba和cry11Aa蛋白均分别获得了表达。透射电镜下观察,转化菌 有产生球形或菱形伴胞晶体。转化菌株对敏感和抗性致倦库蚊及白纹伊蚊幼虫的生物测定结果显示:cry4Aa、cry4Ba和cry11Aa蛋白对库蚊和伊蚊的毒力较低,二元毒素抗性库蚊幼虫对Bti杀蚊毒素蛋白无明显的交叉抗性。  相似文献   

20.
Vector control can be an effective strategy to interrupt disease transmission and biolarvicides based on the entomopathogenic bacteria Bacillus sphaericus, and Bacillus thuringiensis serovar israelensis (Bti) have been successfully used to control species of public health relevance from the genera Aedes, Culex, Anopheles and Simulium. The most important feature of these agents is their ability to produce insecticidal proteins with selective action on the larval midgut. These protoxins are produced as crystals that, once ingested by larvae, are processed into active toxins, interact with receptors in the midgut epithelium and trigger cytopathological effects leading to larval death. B. sphaericus and Bti toxins share the initial steps of the mode of action; however, they interact with different midgut molecules. B. sphaericus presents a single larvicidal factor, the binary (Bin) toxin, whose action relies on the binding to one class of midgut receptors, while Bti crystals contain four protoxins (Cry4Aa, Cry4Ba, Cry11Aa and Cyt1Aa), which display interactions with multiple midgut receptors. The mode of action of B. sphaericus displays a greater potential for resistance selection, compared to Bti, and, to date, there is no record of insect resistance to the latter, contrarily to B. sphaericus. The set of mosquitocidal toxins and their interaction with midgut target sites are described in this review, as well as the implications for the potential to select resistance amongst exposed populations. These biolarvicides have specific mode of action that rely on unique interactions and make them the most selective agents to control Diptera insects actually available.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号