Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
Pestic Biochem Physiol ; 198: 105744, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38225087

RESUMO

Cry2Ab is one of the important alternative Bt proteins that can be used to manage insect pests resistant to Cry1A toxins and to expand the insecticidal spectrum of pyramided Bt crops. Previous studies have showed that vacuolar H+-ATPase subunits A and B (V-ATPase A and B) may be involved in Bt insecticidal activities. The present study investigated the role of V-ATPases subunit E in the toxicity of Cry2Ab in Helicoverpa amigera. RT-PCR analysis revealed that oral exposure of H. amigera larvae to Cry2Ab led to a significant reduction in the expression of H. armigera V-ATPase E (HaV-ATPase E). Ligand blot, homologous and heterologous competition experiments confirmed that HaV-ATPases E physically and specifically bound to activated Cry2Ab toxin. Heterologous expressing of HaV-ATPase E in Sf9 cells made the cell line more susceptible to Cry2Ab, whereas knockdown of the endogenous V-ATPase E in H. zea midgut cells decreased Cry2Ab's cytotoxicity against this cell line. Further in vivo bioassay showed that H. armigera larvae fed a diet overlaid with both Cry2Ab and E. coli-expressed HaV-ATPase E protein suffered significantly higher mortality than those fed Cry2Ab alone. These results support that V-ATPases E is a putative receptor of Cry2Ab and can be used to improve Cry2Ab toxicity and manage Cry2Ab resistance at least in H. armigera.


Assuntos
Bacillus thuringiensis , Inseticidas , Mariposas , Animais , Helicoverpa armigera , Endotoxinas/toxicidade , Endotoxinas/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Escherichia coli , Toxinas de Bacillus thuringiensis/metabolismo , Mariposas/genética , Mariposas/metabolismo , Larva/metabolismo , Inseticidas/toxicidade , Inseticidas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/toxicidade , Proteínas Hemolisinas/metabolismo , Bacillus thuringiensis/metabolismo , Resistência a Inseticidas
2.
Aquat Toxicol ; 266: 106810, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38134819

RESUMO

There are few studies on the effects of nanoplastics on growth and hemolysin production of harmful algal bloom species at present. In this study, Karlodinium veneficum was exposed to different concentrations (0, 5, 25, 50, 75 mg/L) of polystyrene nanoplastics (PS-NPs, 100 nm) for 96 h. The effects of PS-NPs on growth of K. veneficum were investigated by measuring algal cell abundance, growth inhibition rate (IR), total protein (TP), malondialdehyde (MDA), glutathione reductase (GSH), superoxide dismutase (SOD), ATPase activity (Na+/K+ ATPase and Ca2+/Mg2+ ATPase). Scanning electron microscope and transmission electron microscope (SEM and TEM) images of microalgae with or without nanoplastics were also observed. The effects of PS-NPs on hemolysin production of K. veneficum were studied by measuring the changes of hemolytic toxin production of K. veneficum exposed to PS-NPs on 1, 3, 5 and 7 days. High concentrations (50 and 75 mg/L) of PS-NPs seriously affected the growth of K. veneficum and different degrees of damage to cell morphology and ultrastructure were found. Excessive free radicals and other oxidants were produced in the cells, which disrupted the intracellular redox balance state and caused oxidative damage to the cells, and the basic activities such as photosynthesis and energy metabolism were weakened. The athletic ability of K. veneficum was decreased, but the ability to produce hemolysin was enhanced. It was suggested that the presence of nanoplastics in seawater may strengthen the threat of harmful algal bloom species to aquatic ecosystems and human health.


Assuntos
Dinoflagellida , Microalgas , Poluentes Químicos da Água , Humanos , Poliestirenos/toxicidade , Microplásticos , Proteínas Hemolisinas/toxicidade , Ecossistema , Poluentes Químicos da Água/toxicidade , Adenosina Trifosfatases
3.
Toxicon ; 238: 107588, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38147939

RESUMO

Pest insects pose a heavy burden on global agricultural industries with small molecule insecticides being predominantly used for their control. Unwanted side effects and resistance development plagues most small molecule insecticides such as the neonicotinoids, which have been reported to be harmful to honeybees. Bioinsecticides like Bacillus thuringiensis (Bt) toxins can be used as environmentally-friendly alternatives. Arachnid venoms comprise another promising source of bioinsecticides, containing a multitude of selective and potent insecticidal toxins. Unfortunately, no standardised insect models are currently available to assess the suitability of insecticidal agents under laboratory conditions. Thus, we aimed to develop a laboratory model that closely mimics field conditions by employing a leaf disk assay (LDA) for oral application of insecticidal agents in a bioassay tray format. Neonate larvae of the cotton bollworm (Helicoverpa armigera) were fed with soybean (Glycine max) leaves that were treated with different insecticidal agents. We observed dose-dependent insecticidal effects for Bt toxin and the neonicotinoid insecticide imidacloprid, with imidacloprid exhibiting a faster response. Furthermore, we identified several insecticidal arachnid venoms that were active when co-applied with sub-lethal doses of Bt toxin. We propose the H. armigera LDA as a suitable tool for assessing the insecticidal effects of insecticidal agents against lepidopterans.


Assuntos
Venenos de Artrópodes , Bacillus thuringiensis , Inseticidas , Mariposas , Neonicotinoides , Nitrocompostos , Toxinas Biológicas , Humanos , Recém-Nascido , Animais , Inseticidas/toxicidade , Glycine max , Helicoverpa armigera , Toxinas de Bacillus thuringiensis/farmacologia , Larva , Insetos , Toxinas Biológicas/farmacologia , Venenos de Artrópodes/farmacologia , Bioensaio , Folhas de Planta , Proteínas de Bactérias/farmacologia , Proteínas Hemolisinas/toxicidade , Endotoxinas , Controle Biológico de Vetores , Resistência a Inseticidas
4.
Ecotoxicol Environ Saf ; 239: 113631, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35598445

RESUMO

Cadmium (Cd) pollution poses a serious threat to agricultural production and paddy field fauna. Crystalline proteins (e.g., Cry1Ab and Cry1Ac) are secreted by Bacillus thuringiensis, which can manage pests via a complicated toxic mechanism and have been widely used for pest control due to the commercialization of transgenic crops (e.g., cotton and rice) that expresses Bt insecticidal proteins. Nonetheless, studies on the effects of combined stress of Cd and Cry1Ab protein on field indicator species are limited. In the present study, we showed that spiders, Pirata subpiraticus, fed with Cd-containing flies+Cry1Ab had dramatically higher Cd accumulation than that in the spiders fed with Cd-containing flies (p < 0.05). In addition, the enrichment of Cd led to the activation of the protective mechanism by elevating the concentrations of glutathione peroxidase, glutathione S-transferase, and metallothionein in the spiders (p < 0.05). An in-depth transcriptome analysis revealed that the activities of ion metal binding proteins, transporters, and channels might play essential roles in the Cd accumulation process. More importantly, the higher Cd concentration in the combined Cd+Cry1Ab exposure prolonged developmental duration of P. subpiraticus, due to the down-regulated cuticle proteins (CPs) encoding genes involved in the molting process, which was regulated by a series of putative transcriptional factors such as ZBTB and zf-C2H2. Collectively, this integrated analysis illustrates that the combined Cd+Cry1Ab exposure increases the adverse effects of Cd stress on the growth, antioxidase, and CPs encoding genes of P. subpiraticus, thus providing a research basis and prospect for the rationality of transgenic Cry1Ab crops in the cultivation of heavy metal contaminated soil.


Assuntos
Bacillus thuringiensis , Aranhas , Animais , Bacillus thuringiensis/genética , Bacillus thuringiensis/metabolismo , Toxinas de Bacillus thuringiensis , Proteínas de Bactérias/metabolismo , Cádmio/metabolismo , Cádmio/toxicidade , Produtos Agrícolas/metabolismo , Endotoxinas/análise , Endotoxinas/genética , Endotoxinas/toxicidade , Proteínas Hemolisinas/análise , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/toxicidade , Plantas Geneticamente Modificadas/metabolismo , Aranhas/metabolismo , Transcriptoma
5.
FASEB J ; 36(3): e22197, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35147989

RESUMO

Neonatal meningitis-associated Escherichia coli (NMEC) is among the leading causes of bacterial meningitis and sepsis in newborn infants. Several virulence factors have been identified as common among NMEC, and have been shown to play an important role in the development of bacteremia and/or meningitis. However, there is significant variability in virulence factor expression between NMEC isolates, and relatively little research has been done to assess the impact of variable virulence factor expression on immune cell activation and the outcome of infection. Here, we investigated the role of NMEC strain-dependent P2X receptor (P2XR) signaling on the outcome of infection in neonatal mice. We found that alpha-hemolysin (HlyA)-expressing NMEC (HlyA+ ) induced robust P2XR-dependent macrophage cell death in vitro, while HlyA- NMEC did not. P2XR-dependent cell death was inflammasome independent, suggesting an uncoupling of P2XR and inflammasome activation in the context of NMEC infection. In vivo inhibition of P2XRs was associated with increased mortality in neonatal mice infected with HlyA+ NMEC, but had no effect on the survival of neonatal mice infected with HlyA- NMEC. Furthermore, we found that P2XR-dependent protection against HlyA+ NMEC in vivo required macrophages, but not neutrophils or NLRP3. Taken together, these data suggest that HlyA+ NMEC activates P2XRs which in turn confers macrophage-dependent protection against infection in neonates. In addition, our findings indicate that strain-dependent virulence factor expression should be taken into account when studying the immune response to NMEC.


Assuntos
Proteínas de Escherichia coli/toxicidade , Proteínas Hemolisinas/toxicidade , Inflamassomos/metabolismo , Meningite devida a Escherichia coli/metabolismo , Sepse Neonatal/metabolismo , Receptores Purinérgicos P2X/metabolismo , Animais , Células Cultivadas , Escherichia coli K12 , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Macrófagos/metabolismo , Meningite devida a Escherichia coli/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Sepse Neonatal/microbiologia , Receptores Purinérgicos P2X/genética
6.
Front Immunol ; 13: 815775, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154132

RESUMO

Many species of bacteria produce toxins such as cholesterol-dependent cytolysins that form pores in cell membranes. Membrane pores facilitate infection by releasing nutrients, delivering virulence factors, and causing lytic cell damage - cytolysis. Oxysterols are oxidized forms of cholesterol that regulate cellular cholesterol and alter immune responses to bacteria. Whether oxysterols also influence the protection of cells against pore-forming toxins is unresolved. Here we tested the hypothesis that oxysterols stimulate the intrinsic protection of epithelial cells against damage caused by cholesterol-dependent cytolysins. We treated epithelial cells with oxysterols and then challenged them with the cholesterol-dependent cytolysin, pyolysin. Treating HeLa cells with 27-hydroxycholesterol, 25-hydroxycholesterol, 7α-hydroxycholesterol, or 7ß-hydroxycholesterol reduced pyolysin-induced leakage of lactate dehydrogenase and reduced pyolysin-induced cytolysis. Specifically, treatment with 10 ng/ml 27-hydroxycholesterol for 24 h reduced pyolysin-induced lactate dehydrogenase leakage by 88%, and reduced cytolysis from 74% to 1%. Treating HeLa cells with 27-hydroxycholesterol also reduced pyolysin-induced leakage of potassium ions, prevented mitogen-activated protein kinase cell stress responses, and limited alterations in the cytoskeleton. Furthermore, 27-hydroxycholesterol reduced pyolysin-induced damage in lung and liver epithelial cells, and protected against the cytolysins streptolysin O and Staphylococcus aureus α-hemolysin. Although oxysterols regulate cellular cholesterol by activating liver X receptors, cytoprotection did not depend on liver X receptors or changes in total cellular cholesterol. However, oxysterol cytoprotection was partially dependent on acyl-CoA:cholesterol acyltransferase (ACAT) reducing accessible cholesterol in cell membranes. Collectively, these findings imply that oxysterols stimulate the intrinsic protection of epithelial cells against pore-forming toxins and may help protect tissues against pathogenic bacteria.


Assuntos
Bactérias/química , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/toxicidade , Oxisteróis/farmacologia , Fatores de Virulência/toxicidade , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Células Epiteliais/metabolismo , Células HeLa , Proteínas Hemolisinas/química , Humanos , Fatores de Virulência/química
7.
FASEB J ; 35(10): e21889, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34569656

RESUMO

Many species of pathogenic bacteria secrete toxins that form pores in mammalian cell membranes. These membrane pores enable the delivery of virulence factors into cells, result in the leakage of molecules that bacteria can use as nutrients, and facilitate pathogen invasion. Inflammatory responses to bacteria are regulated by the side-chain-hydroxycholesterols 27-hydroxycholesterol and 25-hydroxycholesterol, but their effect on the intrinsic protection of cells against pore-forming toxins is unclear. Here, we tested the hypothesis that 27-hydroxycholesterol and 25-hydroxycholesterol help protect cells against pore-forming toxins. We treated bovine endometrial epithelial and stromal cells with 27-hydroxycholesterol or 25-hydroxycholesterol, and then challenged the cells with pyolysin, which is a cholesterol-dependent cytolysin from Trueperella pyogenes that targets these endometrial cells. We found that treatment with 27-hydroxycholesterol or 25-hydroxycholesterol protected both epithelial and stomal cells against pore formation and the damage caused by pyolysin. The oxysterols limited pyolysin-induced leakage of potassium and lactate dehydrogenase from cells, and reduced cytoskeletal changes and cytolysis. This oxysterol cytoprotection against pyolysin was partially dependent on reducing cytolysin-accessible cholesterol in the cell membrane and on activating liver X receptors. Treatment with 27-hydroxycholesterol also protected the endometrial cells against Staphylococcus aureus α-hemolysin. Using mass spectrometry, we found 27-hydroxycholesterol and 25-hydroxycholesterol in uterine and follicular fluid. Furthermore, epithelial cells released additional 25-hydroxycholesterol in response to pyolysin. In conclusion, both 27-hydroxycholesterol and 25-hydroxycholesterol increased the intrinsic protection of bovine endometrial cells against pore-forming toxins. Our findings imply that side-chain-hydroxycholesterols may help defend the endometrium against pathogenic bacteria.


Assuntos
Bactérias/química , Proteínas de Bactérias/toxicidade , Endométrio/metabolismo , Proteínas Hemolisinas/toxicidade , Hidroxicolesteróis/farmacologia , Fatores de Virulência/toxicidade , Animais , Proteínas de Bactérias/química , Bovinos , Feminino , Proteínas Hemolisinas/química , Células Estromais/metabolismo , Fatores de Virulência/química
8.
Toxins (Basel) ; 13(8)2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34437391

RESUMO

Escherichia coli (E. coli) of the B2 phylotype reside in human and animal intestines. The bacteria possess pathogenicity factors such as α-hemolysin (HlyA) that can induce intestinal epithelial leaks. We addressed the questions which host cell processes were dysregulated by E. coli HlyA that can potentiate intestinal diseases. The colon carcinoma cell line Caco-2 was infected by HlyA+ E. coli. Cell polarity regulation was analyzed by live cell imaging for the phosphatidylinositol-4,5-bisphosphate (PIP2) abundance. In Caco-2 monolayers, transepithelial electrical resistance was measured for characterization of barrier function. Cell proliferation and separation were assessed microscopically. Epithelial regulation and cell signaling were analyzed by RNA-Seq and Ingenuity Pathway Analysis (IPA). Our main findings from E. coli HlyA toxinogenicity in the colon carcinoma cell line are that (i) PIP2 at the membrane decrease, (ii) PTEN (phosphatase and tensin homolog) inhibition leads to cell polarity changes, (iii) epithelial leakiness follows these polarity changes by disruption of cell junctions and (iv) epithelial cell detachment increases. HlyA affected pathways, e.g., the PTEN and metastasis signaling, were identified by RNA-Seq bioinformatics calculations in IPA. In conclusion, HlyA affects cell polarity, thereby inducing epithelial barrier dysfunction due to defective tight junctions and focal leak induction as an exemplary mechanism for leaky gut.


Assuntos
Proteínas de Escherichia coli/toxicidade , Proteínas Hemolisinas/toxicidade , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Células CACO-2 , Polaridade Celular , Proliferação de Células , Neoplasias do Colo/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Infecções por Escherichia coli/metabolismo , Humanos , Junções Intercelulares , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo
9.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33805017

RESUMO

Identification of novel agents for bladder cancer treatment is highly desirable due to the high incidence of tumor recurrence and the risk of progression to muscle-invasive disease. The key feature of the cholesterol-dependent toxin listeriolysin O mutant (LLO Y406A) is its preferential activity at pH 5.7, which could be exploited either directly for selective targeting of cancer cells or the release of accumulated therapeutics from acidic endosomes. Therefore, our goal was to compare the cytotoxic effect of LLO Y406A on cancer cells (RT4) and normal urothelial cells (NPU), and to identify which cell membranes are the primary target of LLO Y406A by viability assays, life-cell imaging, fluorescence, and electron microscopy. LLO Y406A decreased viability, altered cell morphology, provoked membrane blebbing, and induced apoptosis in RT4 cells, while it did not affect NPU cells. LLO Y406A did not cause endosomal escape in RT4 cells, while the plasma membrane of RT4 cells was revealed as the primary target of LLO Y406A. It has been concluded that LLO Y406A has the ability to selectively eliminate cancer urothelial cells through pore-forming activity at the plasma membrane, without cytotoxic effects on normal urothelial cells. This promising selective activity merits further testing as an anti-cancer agent.


Assuntos
Antineoplásicos/toxicidade , Toxinas Bacterianas/toxicidade , Membrana Celular/efeitos dos fármacos , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Neoplasias da Bexiga Urinária/metabolismo , Urotélio/efeitos dos fármacos , Animais , Toxinas Bacterianas/genética , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas/genética , Humanos , Mutação , Suínos , Urotélio/metabolismo
10.
In Vitro Cell Dev Biol Anim ; 57(1): 42-52, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33415662

RESUMO

The bioinsecticidal Cry1Ac proteins (protoxin and toxin) are potent immunogens that can activate macrophages by inducing upregulation of costimulatory molecules, pro-inflammatory cytokines, and mitogen-activated protein kinase (MAPK) signaling pathways. Besides, by the oral route, Cry1Ac toxin is mildly allergenic and induces intestinal lymphoid hyperplasia in mice. Given the potential utility of Cry1Ac protoxin as an adjuvant, as well as the human consumption of Cry1Ac toxin in transgenic crops, it is necessary to more deeply evaluate the toxicological potential of these proteins in mammalian immune cells. Here, were used in vitro evaluations in leukocyte and macrophage cell lines to test the potential toxicity of various doses of Cry1Ac proteins, by means of Alamar Blue, MTT, Annexin V, and JC1 assays. Our results indicated that neither Cry1Ac protoxin nor toxin elicited acute toxic effects, after monitoring the cell activity for 4, 8, 10, and 24 h of exposure. By flow cytometry and confocal microscopy analysis, it was observed that neither Cry1Ac toxin nor protoxin generated mitochondrial damage or depolarization or induced apoptosis or necrosis. In conclusion, despite their immunostimulatory effects, it was demonstrated that Cry1Ac proteins did not have cytotoxic effects, even at high concentrations, in primary leukocytes or macrophages or cell lines.


Assuntos
Toxinas de Bacillus thuringiensis/toxicidade , Toxinas Bacterianas/toxicidade , Endotoxinas/toxicidade , Proteínas Hemolisinas/toxicidade , Leucócitos/patologia , Macrófagos/patologia , Precursores de Proteínas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Leucócitos/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Necrose , Células RAW 264.7 , Baço/patologia , Células THP-1 , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
11.
Appl Environ Microbiol ; 87(2)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33127814

RESUMO

Helicoverpa armigera is a major insect pest of several crops worldwide. This insect is susceptible to some Bacillus thuringiensis (Bt) Cry insecticidal proteins expressed in transgenic crops or used in biopesticides. Previously, we identified H. armigera prohibitin (HaPHB) as a Cry1Ac-binding protein. Here, we further analyzed the potential role of PHB as a Cry toxin receptor in comparison to cadherin (CAD), well recognized as a Cry1Ac receptor. HaPHB-2 midgut protein and HaCAD toxin-binding region (TBR) fragment from H. armigera were expressed in Escherichia coli cells, and binding assays with different Cry1 toxins were performed. We demonstrated that Cry1Ab, Cry1Ac, and Cry1Fa toxins bound to HaPHB-2 in a manner similar to that seen with HaCAD-TBR. Different Cry1Ab mutant toxins located in domain II (Cry1AbF371A and Cry1AbG439D) or domain III (Cry1AbL511A and Cry1AbN514A), which were previously characterized and found to be affected in receptor binding, were analyzed regarding their binding interaction with HaPHB-2 and toxicity against H. armigera One ß-16 mutant (Cry1AbN514A) showed increased binding to HaPHB-2 that correlated with 6-fold-higher toxicity against H. armigera, whereas the other ß-16 mutant (Cry1AbL511A) was affected in binding to HaPHB-2 and lost toxicity against H. armigera Our data indicate that ß-16 from domain III of Cry1Ab is involved in interactions with HaPHB-2 and in toxicity. This report identifies a region of Cry1Ab involved in binding to HaPHB-2 from a Lepidoptera insect, suggesting that this protein may participate as a novel receptor in the mechanism of action of the Cry1 toxins in H. armigeraIMPORTANCEHelicoverpa armigera is a polyphagous pest that feeds on important crops worldwide. This insect pest is sensitive to different Cry1 toxins from Bacillus thuringiensis In this study, we analyzed the potential role of PHB-2 as a Cry1 toxin receptor in comparison to CAD. We show that different Cry1 toxins bound to HaPHB-2 and HaCAD-TBR similarly and identify ß-16 from domain III of Cry1Ab as a binding region involved in the interaction with HaPHB-2 and in toxicity. This report characterized HaPHB-Cry1 binding interaction, providing novel insights into potential target sites for improving Cry1 toxicity against H. armigera.


Assuntos
Toxinas de Bacillus thuringiensis/toxicidade , Endotoxinas/toxicidade , Proteínas Hemolisinas/toxicidade , Proteínas de Insetos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Toxinas de Bacillus thuringiensis/genética , Sítios de Ligação , Endotoxinas/genética , Proteínas Hemolisinas/genética , Larva , Mariposas , Proibitinas , Domínios Proteicos
12.
Toxins (Basel) ; 12(11)2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33233557

RESUMO

Staphylococcus aureus is a major human pathogen, inducing several infections ranging from the benign to the life-threatening, such as necrotising pneumonia. S. aureus is capable of producing a great variety of virulence factors, such as bicomponent pore-forming leucocidin, which take part in the physiopathology of staphylococcal infection. In necrotising pneumonia, Panton-Valentine leucocidin (PVL) induces not only lung injury and necrosis, but also leukopenia, regarded as a major factor of a poor prognosis. The aim of the present study was to evaluate the effect of bicomponent pore-forming leucocidin, PVL and gamma haemolysin on bone marrow leucocytes, to better understand the origin of leukopenia. Using multi-parameter cytometry, the expression of leucocidin receptors (C5aR, CXCR1, CXCR2, and CCR2) was assessed and toxin-induced lysis was measured for each bone marrow leucocyte population. We observed that PVL resulted in myeloid-derived cells lysis according to their maturation and their C5aR expression; it also induced monocytes lysis according to host susceptibility. Haemolysin gamma A, B, and C (HlgABC) displayed cytotoxicity to monocytes and natural killer cells, hypothetically through CXCR2 and CXCR1 receptors, respectively. Taken together, the data suggest that PVL and HlgABC can lyse bone marrow leucocytes. Nevertheless, the origin of leukopenia in severe staphylococcal infection is predominantly peripheral, since immature cells stay insensitive to leucocidins.


Assuntos
Toxinas Bacterianas/toxicidade , Exotoxinas/toxicidade , Proteínas Hemolisinas/toxicidade , Leucocidinas/toxicidade , Leucócitos/efeitos dos fármacos , Staphylococcus aureus , Células da Medula Óssea/citologia , Sobrevivência Celular/efeitos dos fármacos , Humanos
13.
Biochemistry ; 59(44): 4213-4224, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33108867

RESUMO

α-Hemolysin (HlyA) is an extracellular protein toxin secreted by uropathogenic strains of Escherichia coli that inserts into membranes of eukaryotic cells. The main goal of this work was to investigate the involvement of tryptophan (W) residues in the hemolytic activity of HlyA. We investigated the hemolytic activity of six single-point mutant proteins, in which one of the four Ws was replaced by cysteine (C) or leucine (L). We also analyzed the photoinactivation of HlyA with pterin (Ptr), an endogenous photosensitizer, as a method of unspecific oxidation of W and tyrosine (Y) residues. HlyA photoinactivation was analyzed by ultraviolet-visible spectrophotometry, hemolytic activity measurement, fluorescence spectroscopy, and electrophoretic analysis. The results indicate that Ws are important in the hemolytic process. Specifically, the chemical structure of the amino acid at position 578 is important for the acylation of HlyA at residue K563. Furthermore, the exposure of HlyA to ultraviolet radiation, with energy similar to that experienced under sun exposure, in the presence of Ptr induces the inactivation of the toxin, causing chemical changes in, at least, W and Y, the rate of damage to W residues being faster than that observed for Y residues. This work not only deepens our understanding of the structure-function relationship of the toxin but also introduces the possibility of using photoinactivation of HlyA for potential applications such as obtaining innocuous molecules for vaccine production and the elimination of the toxin from contaminated surfaces and drinking water.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidade , Proteínas Hemolisinas/química , Proteínas Hemolisinas/toxicidade , Luz , Triptofano , Substituição de Aminoácidos , Proteínas de Escherichia coli/genética , Proteínas Hemolisinas/genética , Hemólise/efeitos dos fármacos , Hemólise/efeitos da radiação
14.
PLoS One ; 15(5): e0233854, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32470006

RESUMO

Alpha-toxin (Hla) is a major virulence factor of Staphylococcus aureus (S. aureus) and plays an important role in S. aureus-induced pneumonia. It binds as a monomer to the cell surface of eukaryotic host cells and forms heptameric transmembrane pores. Sensitivities toward the toxin of various types of potential host cells have been shown to vary substantially, and the reasons for these differences are unclear. We used three human model airway epithelial cell lines (16HBE14o-, S9, A549) to correlate cell sensitivity (measured as rate of paracellular gap formation in the cell layers) with Hla monomer binding, presence of the potential Hla receptors ADAM10 or α5ß1 integrin, presence of the toxin-stabilizing factor caveolin-1 as well as plasma membrane lipid composition (phosphatidylserine/choline, sphingomyelin). The abundance of ADAM10 correlated best with gap formation or cell sensitivities, respectively, when the three cell types were compared. Caveolin-1 or α5ß1 integrin did not correlate with toxin sensitivity. The relative abundance of sphingomyelin in plasma membranes may also be used as a proxi for cellular sensitivity against alpha-toxin as sphingomyelin abundances correlated well with the intensities of alpha-toxin mediated gap formation in the cell layers.


Assuntos
Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidade , Interações Hospedeiro-Patógeno , Sistema Respiratório/patologia , Células A549 , Caveolina 1/metabolismo , Membrana Celular/efeitos dos fármacos , Tamanho Celular , Células Epiteliais/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Modelos Biológicos , Fosfolipídeos/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo
15.
Insect Biochem Mol Biol ; 119: 103317, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31978588

RESUMO

Bacillus thuringiensis (Bt) Cyt1Aa toxin shows toxicity to mosquitoes, to certain coleopteran pests and also to red blood cells (RBC). However, its mode of action in the different target cells is not well defined. This protein is a single α-ß domain pore-forming toxin, where a ß sheet is wrapped by two α-helices layers. The Cyt1Aa α-helix hairpin in the N-terminal has been proposed to be involved in initial membrane binding and oligomerization, while the ß sheet inserts into the membrane to form a pore that lyze the cells. To determine the role of the N-terminal α-helix hairpin region of Cyt1Aa in its mode of action, we characterized different single point mutations located in helices α-1 and α-2. Eight cysteine substitutions in different residues were produced in Bt, and we found that three of them: Cyt1AaA65C, Cyt1AaL85C and Cyt1AaN89C, lost insecticidal toxicity against Aedes aegypti larvae but retained similar or increased hemolytic activity towards rabbit RBC. Analysis of toxin binding and oligomerization using Ae. aegypti midgut brush border membrane vesicles showed that the three Cyt1Aa mutants non-toxic to Ae. aegypti were affected in oligomerization. However, these mutants were still hemolytic. Our data shows that oligomerization of Cyt1Aa toxin is essential for its toxicity to Ae. aegypti but not for its toxicity against RBC indicating that the mode of action of Cyt1Aa is different in these distinct target membranes.


Assuntos
Aedes/efeitos dos fármacos , Proteínas de Bactérias/química , Endotoxinas/química , Eritrócitos/efeitos dos fármacos , Proteínas Hemolisinas/química , Inseticidas/química , Aedes/crescimento & desenvolvimento , Animais , Bacillus thuringiensis/química , Toxinas de Bacillus thuringiensis , Proteínas de Bactérias/toxicidade , Endotoxinas/toxicidade , Proteínas Hemolisinas/toxicidade , Inseticidas/toxicidade , Larva/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Polimerização , Coelhos
16.
Plant Cell Rep ; 39(3): 317-323, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31797051

RESUMO

KEY MESSAGE: The Bacillus thuringiensis (Bt) cry3Bb gene was successfully introduced into poplar plastid genome, leading to transplastomic poplar with high mortality to Plagiodera versicolora. Poplar (Populus L.) is one of the main resource of woody industry, but being damaged by insect pests. The feasibility and efficiency of plastid transformation technology for controlling two lepidopteran caterpillars have been demonstrated previously. Here, we introduced B. thuringiensis (Bt) cry3Bb into poplar plastid genome by biolistic bombardment for controlling P. versicolora, a widely distributed forest pest. Chimeric cry3Bb gene is controlled by the tobacco plastid rRNA operon promoter combined with the 5'UTR from gene10 of bacteriophage T7 (NtPrrn:T7g10) and the 3'UTR from the E. coli ribosomal RNA operon rrnB (TrrnB). The integration of transgene and homoplasmy of transplastomic poplar plants was confirmed by Southern blot analysis. Northern blot analysis indicated that cry3Bb was transcribed to both read through and shorter length transcripts in plastid. The transplastomic poplar expressing Cry3Bb insecticidal protein showed the highest accumulation level in young leaves, which reach up to 16.8 µg/g fresh weight, and comparatively low levels in mature and old leaves. Feeding the young leaves from Bt-Cry3Bb plastid lines to P. versicolora caused 100% mortality in the first-instar larvae after only 1 day, in the second-instar larvae after 2 days, and in the third-instar larvae for 3 days. Thus, we report a successful extension of plastid engineering poplar against the chrysomelid beetle.


Assuntos
Bacillus thuringiensis/metabolismo , Proteínas de Bactérias/metabolismo , Besouros/fisiologia , Endotoxinas/metabolismo , Comportamento Alimentar , Proteínas Hemolisinas/metabolismo , Folhas de Planta/parasitologia , Plastídeos/metabolismo , Populus/genética , Populus/parasitologia , Animais , Toxinas de Bacillus thuringiensis , Proteínas de Bactérias/toxicidade , Besouros/efeitos dos fármacos , Endotoxinas/toxicidade , Comportamento Alimentar/efeitos dos fármacos , Vetores Genéticos/metabolismo , Genomas de Plastídeos , Proteínas Hemolisinas/toxicidade , Larva/efeitos dos fármacos , Larva/fisiologia , Fenótipo , Plantas Geneticamente Modificadas , Transformação Genética
17.
Biomaterials ; 217: 119286, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31284125

RESUMO

Antimicrobial peptides (AMPs) have recently attracted great attention due to their rapid action, broad spectrum of activity, and low propensity of resistance development. The successful application of AMPs in the treatment of intracellular infections, however, remains a challenge because of their low penetration efficiency into the pathogen's intracellular niche. Herein, we report that sub-micrometer-sized crystals of the protein Cry3Aa formed within Bacillus thuringiensis are readily and specifically taken up by macrophages. We demonstrate that these protein crystals efficiently encapsulate a known antileishmanial peptide, dermaseptin S1 (DS1), and thereby promote improved cellular uptake of DS1 and its lysosomal accumulation in macrophages. Notably, this targeted delivery of DS1 results in enhanced in vitro and in vivo antileishmanial activity, as well as reduced toxicity to the host macrophages. These findings suggest that the Cry3Aa crystal can be an effective delivery platform for AMPs to treat intramacrophage infections.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/química , Sistemas de Liberação de Medicamentos , Endotoxinas/química , Proteínas Hemolisinas/química , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/parasitologia , Proteínas de Anfíbios/farmacologia , Animais , Toxinas de Bacillus thuringiensis , Proteínas de Bactérias/toxicidade , Proteínas de Bactérias/ultraestrutura , Linhagem Celular Tumoral , Endotoxinas/toxicidade , Feminino , Proteínas Hemolisinas/toxicidade , Proteínas Hemolisinas/ultraestrutura , Hemólise/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Leishmania/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos Endogâmicos BALB C
18.
Toxins (Basel) ; 11(6)2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31212877

RESUMO

Cytolytic leukotoxins of the repeat in toxin (RTX) family are large proteins excreted by gram-negative bacterial pathogens through the type 1 secretion system (T1SS). Due to low yields and poor stability in cultures of the original pathogens, it is useful to purify recombinant fatty-acylated RTX cytolysins from inclusion bodies produced in E. coli. Such preparations are, however, typically contaminated by high amounts of E. coli lipopolysaccharide (LPS or endotoxin). We report a simple procedure for purification of large amounts of biologically active and endotoxin-free RTX toxins. It is based on the common feature of RTX cytolysins that are T1SS-excreted as unfolded polypeptides and fold into a biologically active toxin only upon binding of calcium ions outside of the bacterial cell. Mimicking this process, the RTX proteins are solubilized from inclusion bodies with buffered 8 M urea, bound onto a suitable chromatographic medium under denaturing conditions and the contaminating LPS is removed through extensive on-column washes with buffers containing 6 to 8 M urea and 1% Triton X-100 or Triton X-114. Extensive on-column rinsing with 8 M urea buffer removes residual detergent and the eluted highly active RTX protein preparations then contain only trace amounts of LPS. The procedure is exemplified using four prototypic RTX cytolysins, the Bordetella pertussis CyaA and the hemolysins of Escherichia coli (HlyA), Kingella kingae (RtxA), and Actinobacillus pleuropneumoniae (ApxIA).


Assuntos
Proteínas de Bactérias/isolamento & purificação , Citotoxinas/isolamento & purificação , Proteínas Hemolisinas/isolamento & purificação , Animais , Proteínas de Bactérias/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citotoxinas/toxicidade , Detergentes/química , Eritrócitos/efeitos dos fármacos , Escherichia coli/metabolismo , Proteínas Hemolisinas/toxicidade , Hemólise , Humanos , Lipopolissacarídeos/análise , Octoxinol/química , Ovinos , Células THP-1 , Ureia/química
19.
Sci Rep ; 9(1): 8507, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31186483

RESUMO

Transgenic rice lines expressing Bacillus thuringiensis (Bt) toxins have been successfully developed for the control of Chilo suppressalis. However, the evolution of insect resistance is a major threat to Bt rice durability. Bt toxins function by binding specific receptors in the midgut of target insects; specifically, cadherin proteins have been identified as Cry toxin receptors in diverse lepidopteran species. Here, we report the functional roles of cadherin CsCad in the midgut of C. suppressalis in Cry1Ab and Cry1C toxicity. We expressed a recombinant truncated CsCad peptide (CsCad-CR11-MPED) in Escherichia coli that included the eleventh cadherin repeat and MPED region. Based on ligand blotting and ELISA binding assays, the CsCad-CR11-MPED peptide specifically bound Cry1Ab with high affinity but weakly bound Cry1C. The CsCad-CR11-MPED peptide significantly enhanced the susceptibility of C. suppressalis larvae to Cry1Ab but not Cry1C. Furthermore, the knockdown of endogenous CsCad with Stealth siRNA reduced C. suppressalis larval susceptibility to Cry1Ab but not Cry1C, suggesting that CsCad plays differential functional roles in Cry1Ab and Cry1C intoxication in C. suppressalis. This information directly enhances our understanding of the potential resistance mechanisms of C. suppressalis against Bt toxins and may assist in the development of effective strategies for delaying insect resistance.


Assuntos
Proteínas de Bactérias/toxicidade , Caderinas/metabolismo , Endotoxinas/toxicidade , Proteínas Hemolisinas/toxicidade , Proteínas de Insetos/metabolismo , Mariposas/metabolismo , Sequência de Aminoácidos , Animais , Toxinas de Bacillus thuringiensis , Caderinas/química , Caderinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Insetos/química , Proteínas de Insetos/genética , Cinética , Larva/efeitos dos fármacos , Larva/metabolismo , Ligantes , Mariposas/efeitos dos fármacos , Mariposas/genética , Peptídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Interferência de RNA
20.
Cell Microbiol ; 21(9): e13045, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31099073

RESUMO

Serratia marcescens is an opportunistic pathogen with increasing incidence in clinical settings. This is mainly attributed to the timely expression of a wide diversity of virulence factors and intrinsic and acquired resistance to antibiotics, including ß-lactams, aminoglycosides, quinolones, and polypeptides. For these reasons, S. marcescens has been recently categorised by the World Health Organization as one priority to strengthen efforts directed to develop new antibacterial agents. Therefore, it becomes critical to understand the underlying mechanisms that allow Serratia to succeed within the host. S. marcescens ShlA pore-forming toxin mediates phenotypes that alter homeostatic and signal transduction pathways of host cells. It has been previously demonstrated that ShlA provokes cytotoxicity, haemolysis and autophagy and also directs Serratia egress and dissemination from invaded nonphagocytic cells. However, molecular details of ShlA mechanism of action are still not fully elucidated. In this work, we demonstrate that Ni2+ selectively and reversibly blocks ShlA action, turning wild-type S. marcescens into a shlA mutant strain phenocopy. Combined use of Ni2+ and calcium chelators allow to discern ShlA-triggered phenotypes that require intracellular calcium mobilisation and reveal ShlA function as a calcium channel, providing new insights into ShlA mode of action on target cells.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Canais de Cálcio/metabolismo , Proteínas Hemolisinas/antagonistas & inibidores , Níquel/farmacologia , Serratia marcescens/efeitos dos fármacos , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/metabolismo , Células CHO , Cálcio/metabolismo , Cricetulus , Eritrócitos/microbiologia , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Fenótipo , Serratia marcescens/metabolismo , Serratia marcescens/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA