Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
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
2.
Food Chem ; 344: 128571, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33221106

RESUMO

Extract of E. camaldulensis was partitioned into aqueous and ethanol fractions by a precipitation and sedimentation-based technique and profiled for phytochemical components. Antimicrobial evaluation yielded inhibitory concentrations of 16-64 and 158-316 µg/mL, and bactericidal concentrations of 32-64 and 316->2528 µg/mL for ethanol and aqueous fractions, respectively. Antioxidant activities evaluated using 2,2-diphenyl-1-picrylhydrazyl and 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic) acid assays showed IC50 values of 7.07 and 65.67 µg/mL, and 17.96 and 201.3 µg/mL for aqueous and ethanol fractions. Total phenolic content of 9.04 ± 0.26 and 3.58 ± 0.04 GAE/mg fraction, and flavonoid content of 2.07 ± 0.02 and 3.37 ± 0.05 QE/mg fraction were recorded for aqueous and ethanol fractions. At subinhibitory concentrations fractions significantly reduced listeriolysin O-induced haemolysis (p < 0.05), and ameliorated H2O2-induced toxicity by 8-23 and 15-83%. Nitrite production reduced by 4-17 and 3-14 µM following fractions treatment. The fractions showed bioactive properties, with oxidative stress amelioratory effects, and could be a potentials source of preservatives and functional food additives.


Assuntos
Toxinas Bacterianas/toxicidade , Colo/embriologia , Eucalyptus/química , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Antibacterianos/farmacologia , Antioxidantes/farmacologia , Colo/citologia , Microbiologia de Alimentos , Humanos
3.
Cell Rep ; 30(4): 1129-1140.e5, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31995754

RESUMO

Plasma membrane damage and cell death during processes such as necroptosis and apoptosis result from cues originating intracellularly. However, death caused by pore-forming agents, like bacterial toxins or complement, is due to direct external injury to the plasma membrane. To prevent death, the plasma membrane has an intrinsic repair ability. Here, we found that repair triggered by pore-forming agents involved TMEM16F, a calcium-activated lipid scramblase also mutated in Scott's syndrome. Upon pore formation and the subsequent influx of intracellular calcium, TMEM16F induced rapid "lipid scrambling" in the plasma membrane. This response was accompanied by membrane blebbing, extracellular vesicle release, preserved membrane integrity, and increased cell viability. TMEM16F-deficient mice exhibited compromised control of infection by Listeria monocytogenes associated with a greater sensitivity of neutrophils to the pore-forming Listeria toxin listeriolysin O (LLO). Thus, the lipid scramblase TMEM16F is critical for plasma membrane repair after injury by pore-forming agents.


Assuntos
Anoctaminas/metabolismo , Toxinas Bacterianas/toxicidade , Membrana Celular/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Timócitos/metabolismo , Animais , Anoctaminas/genética , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Membrana Celular/efeitos dos fármacos , Vesículas Extracelulares/efeitos dos fármacos , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Fígado/citologia , Fígado/metabolismo , Fígado/microbiologia , Fígado/patologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Neutrófilos/patologia , Proteínas de Transferência de Fosfolipídeos/genética , Baço/citologia , Baço/metabolismo , Baço/microbiologia , Baço/patologia , Timócitos/efeitos dos fármacos , Timócitos/ultraestrutura
4.
Nat Microbiol ; 3(12): 1472-1485, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30478389

RESUMO

Plasma membrane integrity is essential for the viability of eukaryotic cells. In response to bacterial pore-forming toxins, disrupted regions of the membrane are rapidly repaired. However, the pathways that mediate plasma membrane repair are unclear. Here we show that autophagy-related (ATG) protein ATG16L1 and its binding partners ATG5 and ATG12 are required for plasma membrane repair through a pathway independent of macroautophagy. ATG16L1 is required for lysosome fusion with the plasma membrane and blebbing responses that promote membrane repair. ATG16L1 deficiency causes accumulation of cholesterol in lysosomes that contributes to defective membrane repair. Cell-to-cell spread by Listeria monocytogenes requires membrane damage by the bacterial toxin listeriolysin O, which is restricted by ATG16L1-dependent membrane repair. Cells harbouring the ATG16L1 T300A allele associated with inflammatory bowel disease were also found to accumulate cholesterol and be defective in repair, linking a common inflammatory disease to plasma membrane integrity. Thus, plasma membrane repair could be an important therapeutic target for the treatment of bacterial infections and inflammatory disorders.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Listeria monocytogenes/efeitos dos fármacos , Animais , Autofagia , Proteína 12 Relacionada à Autofagia/metabolismo , Proteína 5 Relacionada à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/genética , Toxinas Bacterianas/toxicidade , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Colesterol/metabolismo , Modelos Animais de Doenças , Exocitose , Células HeLa , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Humanos , Listeria monocytogenes/metabolismo , Lisossomos , Masculino , Camundongos
5.
J Biol Chem ; 293(38): 14758-14774, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30093405

RESUMO

Amyloid and amyloid-like protein aggregations are hallmarks of multiple, varied neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. We previously reported that spinocerebellar ataxia type 14 (SCA14), a dominant-inherited neurodegenerative disease that affects cerebellar Purkinje cells, is characterized by the intracellular formation of neurotoxic amyloid-like aggregates of genetic variants of protein kinase Cγ (PKCγ). A number of protein chaperones, including heat shock protein 70 (Hsp70), promote the degradation and/or refolding of misfolded proteins and thereby prevent their aggregation. Here, we report that, in various SCA14-associated, aggregating PKCγ variants, endogenous Hsp70 is incorporated into aggregates and that expression of these PKCγ mutants up-regulates Hsp70 expression. We observed that PKCγ binds Hsp70 and that this interaction is enhanced in the SCA14-associated variants, mediated by the kinase domain that is involved in amyloid-like fibril formation as well as the C2 domain of PKCγ. Pharmacological up-regulation of Hsp70 by the Hsp90 inhibitors celastrol and herbimycin A attenuated the aggregation of mutant PKCγ in primary cultured Purkinje cells. Up-regulation of Hsp70 diminished net PKCγ aggregation by preventing aggregate formation, resulting in decreased levels of apoptotic cell death among primary cultured Purkinje cells expressing the PKCγ variant. Of note, herbimycin A also ameliorated abnormal dendritic development. Extending our in vitro observations, administration of celastrol to mice up-regulated cerebellar Hsp70. Our findings identify heat shock proteins as important endogenous regulators of pathophysiological PKCγ aggregation and point to Hsp90 inhibition as a potential therapeutic strategy in the treatment of SCA14.


Assuntos
Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/toxicidade , Mutação , Proteína Quinase C/genética , Proteína Quinase C/toxicidade , Ataxias Espinocerebelares/enzimologia , Animais , Linhagem Celular , Cerebelo/metabolismo , Detergentes/química , Humanos , Rifabutina/análogos & derivados , Rifabutina/farmacologia , Solubilidade , Ataxias Espinocerebelares/genética , Regulação para Cima
6.
Mol Cell Proteomics ; 17(8): 1627-1636, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29752379

RESUMO

Bacterial pathogens use various strategies to interfere with host cell functions. Among these strategies, bacteria modulate host gene transcription, thereby modifying the set of proteins synthetized by the infected cell. Bacteria can also target pre-existing host proteins and modulate their post-translational modifications or trigger their degradation. Analysis of protein levels variations in host cells during infection allows to integrate both transcriptional and post-transcriptional regulations induced by pathogens. Here, we focused on host proteome alterations induced by the toxin Listeriolysin O (LLO), secreted by the bacterial pathogen Listeria monocytogenes. We showed that a short-term treatment with LLO remodels the host cell proteome by specifically decreasing the abundance of 149 proteins. The same decrease in host protein levels was observed in different epithelial cell lines but not in macrophages. We show in particular that this proteome remodeling affects several ubiquitin and ubiquitin-like ligases and that LLO leads to major changes in the host ubiquitylome. Strikingly, this toxin-induced proteome remodeling involves only post-transcriptional regulations, as no modification in the transcription levels of the corresponding genes was observed. In addition, we could show that Perfringolysin O, another bacterial pore-forming toxin similar to LLO, also induces host proteome changes. Taken together, our data reveal that different bacterial pore-forming toxins induce important host proteome remodeling, that may impair epithelial cell functions.


Assuntos
Toxinas Bacterianas/toxicidade , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Interações Hospedeiro-Patógeno , Proteoma/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células HeLa , Células Hep G2 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Células RAW 264.7 , Ubiquitinação/efeitos dos fármacos
7.
Toxins (Basel) ; 10(2)2018 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-29439494

RESUMO

Pulmonary permeability edema is characterized by reduced alveolar Na⁺ uptake capacity and capillary barrier dysfunction and is a potentially lethal complication of listeriosis. Apical Na⁺ uptake is mainly mediated by the epithelial sodium channel (ENaC) and initiates alveolar liquid clearance. Here we examine how listeriolysin O (LLO), the pore-forming toxin of Listeria monocytogenes, impairs the expression and activity of ENaC. To that purpose, we studied how sub-lytic concentrations of LLO affect negative and positive regulators of ENaC expression in the H441 airway epithelial cell line. LLO reduced expression of the crucial ENaC-α subunit in H441 cells within 2 h and this was preceded by activation of PKC-α, a negative regulator of the channel's expression. At later time points, LLO caused a significant reduction in the phosphorylation of Sgk-1 at residue T256 and of Akt-1 at residue S473, both of which are required for full activation of ENaC. The TNF-derived TIP peptide prevented LLO-mediated PKC-α activation and restored phospho-Sgk-1-T256. The TIP peptide also counteracted the observed LLO-induced decrease in amiloride-sensitive Na⁺ current and ENaC-α expression in H441 cells. Intratracheally instilled LLO caused profound pulmonary edema formation in mice, an effect that was prevented by the TIP peptide; thus indicating the therapeutic potential of the peptide for the treatment of pore-forming toxin-associated permeability edema.


Assuntos
Toxinas Bacterianas/toxicidade , Canais Epiteliais de Sódio/fisiologia , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Edema Pulmonar/tratamento farmacológico , Animais , Brônquios/citologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Humanos , Proteínas Imediatamente Precoces/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Proteína Quinase C-alfa/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-28770170

RESUMO

The plasma membrane of mammalian cells is susceptible to disruption by mechanical and biochemical damages that frequently occur within tissues. Therefore, efficient and rapid repair of the plasma membrane is essential for maintaining cellular homeostasis and survival. Excessive damage of the plasma membrane and defects in its repair are associated with pathological conditions such as infections, muscular dystrophy, heart failure, diabetes, and lung and neurodegenerative diseases. The molecular events that remodel the plasma membrane during its repair remain poorly understood. In the present work, we report the development of a quantitative high-throughput assay that monitors the efficiency of the plasma membrane repair in real time using a sensitive microplate reader. In this assay, the plasma membrane of living cells is perforated by the bacterial pore-forming toxin listeriolysin O and the integrity and recovery of the membrane are monitored at 37°C by measuring the fluorescence intensity of the membrane impermeant dye propidium iodide. We demonstrate that listeriolysin O causes dose-dependent plasma membrane wounding and activation of the cell repair machinery. This assay was successfully applied to cell types from different origins including epithelial and muscle cells. In conclusion, this high-throughput assay provides a novel opportunity for the discovery of membrane repair effectors and the development of new therapeutic compounds that could target membrane repair in various pathological processes, from degenerative to infectious diseases.


Assuntos
Membrana Celular/fisiologia , Técnicas Citológicas/métodos , Ensaios de Triagem em Larga Escala/métodos , Animais , Toxinas Bacterianas/toxicidade , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Fluorometria/métodos , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Humanos , Células Musculares/efeitos dos fármacos , Células Musculares/fisiologia , Permeabilidade/efeitos dos fármacos , Propídio/análise , Coloração e Rotulagem/métodos , Temperatura
9.
Toxins (Basel) ; 9(1)2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28098781

RESUMO

Cholesterol-dependent cytolysins (CDCs) are protein toxins that originate from Gram-positive bacteria and contribute substantially to their pathogenicity. CDCs bind membrane cholesterol and build prepores and lytic pores. Some effects of the toxins are observed in non-lytic concentrations. Two pathogens, Streptococcus pneumoniae and Listeria monocytogenes, cause fatal bacterial meningitis, and both produce toxins of the CDC family-pneumolysin and listeriolysin O, respectively. It has been demonstrated that pneumolysin produces dendritic varicosities (dendrite swellings) and dendritic spine collapse in the mouse neocortex, followed by synaptic loss and astrocyte cell shape remodeling without elevated cell death. We utilized primary glial cultures and acute mouse brain slices to examine the neuropathological effects of listeriolysin O and to compare it to pneumolysin with identical hemolytic activity. In cultures, listeriolysin O permeabilized cells slower than pneumolysin did but still initiated non-lytic astrocytic cell shape changes, just as pneumolysin did. In an acute brain slice culture system, listeriolysin O produced dendritic varicosities in an NMDA-dependent manner but failed to cause dendritic spine collapse and cortical astrocyte reorganization. Thus, listeriolysin O demonstrated slower cell permeabilization and milder glial cell remodeling ability than did pneumolysin and lacked dendritic spine collapse capacity but exhibited equivalent dendritic pathology.


Assuntos
Astrócitos/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Encéfalo/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/metabolismo , Neurotoxinas/toxicidade , Animais , Animais Recém-Nascidos , Astrócitos/patologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/genética , Encéfalo/patologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Espinhas Dendríticas/patologia , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas/genética , Camundongos Endogâmicos C57BL , Neurotoxinas/genética , Cultura Primária de Células , Proteínas Recombinantes , Estreptolisinas/genética , Estreptolisinas/toxicidade
10.
J Vis Exp ; (93): e52089, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25407485

RESUMO

Many protein-misfolding disorders can be modeled in the budding yeast Saccharomyces cerevisiae. Proteins such as TDP-43 and FUS, implicated in amyotrophic lateral sclerosis, and α-synuclein, implicated in Parkinson's disease, are toxic and form cytoplasmic aggregates in yeast. These features recapitulate protein pathologies observed in patients with these disorders. Thus, yeast are an ideal platform for isolating toxicity suppressors from libraries of protein variants. We are interested in applying protein disaggregases to eliminate misfolded toxic protein conformers. Specifically, we are engineering Hsp104, a hexameric AAA+ protein from yeast that is uniquely capable of solubilizing both disordered aggregates and amyloid and returning the proteins to their native conformations. While Hsp104 is highly conserved in eukaryotes and eubacteria, it has no known metazoan homologue. Hsp104 has only limited ability to eliminate disordered aggregates and amyloid fibers implicated in human disease. Thus, we aim to engineer Hsp104 variants to reverse the protein misfolding implicated in neurodegenerative disorders. We have developed methods to screen large libraries of Hsp104 variants for suppression of proteotoxicity in yeast. As yeast are prone to spontaneous nonspecific suppression of toxicity, a two-step screening process has been developed to eliminate false positives. Using these methods, we have identified a series of potentiated Hsp104 variants that potently suppress the toxicity and aggregation of TDP-43, FUS, and α-synuclein. Here, we describe this optimized protocol, which could be adapted to screen libraries constructed using any protein backbone for suppression of toxicity of any protein that is toxic in yeast.


Assuntos
Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/toxicidade , Biblioteca de Peptídeos , Saccharomyces cerevisiae/metabolismo , Proteínas de Ligação a DNA/metabolismo , Doença de Parkinson/metabolismo , Conformação Proteica , Proteína FUS de Ligação a RNA/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , alfa-Sinucleína/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(34): 12432-7, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114211

RESUMO

SUMOylation is an essential ubiquitin-like modification involved in important biological processes in eukaryotic cells. Identification of small ubiquitin-related modifier (SUMO)-conjugated residues in proteins is critical for understanding the role of SUMOylation but remains experimentally challenging. We have set up a powerful and high-throughput method combining quantitative proteomics and peptide immunocapture to map SUMOylation sites and have analyzed changes in SUMOylation in response to stimuli. With this technique we identified 295 SUMO1 and 167 SUMO2 sites on endogenous substrates of human cells. We further used this strategy to characterize changes in SUMOylation induced by listeriolysin O, a bacterial toxin that impairs the host cell SUMOylation machinery, and identified several classes of host proteins specifically deSUMOylated in response to this toxin. Our approach constitutes an unprecedented tool, broadly applicable to various SUMO-regulated cellular processes in health and disease.


Assuntos
Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Sequência de Aminoácidos , Toxinas Bacterianas/toxicidade , Sítios de Ligação , Células HeLa , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Humanos , Dados de Sequência Molecular , Mapeamento de Peptídeos/métodos , Proteômica/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SUMO-1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação/efeitos dos fármacos
12.
Proc Natl Acad Sci U S A ; 110(40): 16003-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043775

RESUMO

We recently showed that infection by Listeria monocytogenes causes mitochondrial network fragmentation through the secreted pore-forming toxin listeriolysin O (LLO). Here, we examine factors involved in canonical fusion and fission. Strikingly, LLO-induced mitochondrial fragmentation does not require the traditional fission machinery, as Drp1 oligomers are absent from fragmented mitochondria following Listeria infection or LLO treatment, as the dynamin-like protein 1 (Drp1) receptor Mff is rapidly degraded, and as fragmentation proceeds efficiently in cells with impaired Drp1 function. LLO does not cause processing of the fusion protein optic atrophy protein 1 (Opa1), despite inducing a decrease in the mitochondrial membrane potential, suggesting a unique Drp1- and Opa1-independent fission mechanism distinct from that triggered by uncouplers or the apoptosis inducer staurosporine. We show that the ER marks LLO-induced mitochondrial fragmentation sites even in the absence of functional Drp1, demonstrating that the ER activity in regulating mitochondrial fission can be induced by exogenous agents and that the ER appears to regulate fission by a mechanism independent of the canonical mitochondrial fission machinery.


Assuntos
Toxinas Bacterianas/toxicidade , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/química , Listeriose/fisiopatologia , Dinâmica Mitocondrial/fisiologia , Toxinas Bacterianas/análise , Western Blotting , Dinaminas , Imunofluorescência , GTP Fosfo-Hidrolases/metabolismo , Células HeLa , Proteínas de Choque Térmico/análise , Proteínas Hemolisinas/análise , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Dinâmica Mitocondrial/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
13.
PLoS One ; 7(3): e32310, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22403645

RESUMO

The presentation of microbial protein antigens by Major Histocompatibility Complex (MHC) molecules is essential for the development of acquired immunity to infections. However, most biochemical studies of antigen processing and presentation deal with a few relatively inert non-microbial model antigens. The bacterial pore-forming toxin listeriolysin O (LLO) is paradoxical in that it is cytotoxic at nanomolar concentrations as well as being the source of dominant CD4 and CD8 T cell epitopes following infection with Listeria monocytogenes. Here, we examined the relationship of LLO toxicity to its antigenicity and immunogenicity. LLO offered to antigen presenting cells (APC) as a soluble protein, was presented to CD4 T cells at picomolar to femtomolar concentrations- doses 3000-7000-fold lower than free peptide. This presentation required a dose of LLO below the cytotoxic level. Mutations of two key tryptophan residues reduced LLO toxicity by 10-100-fold but had no effect on its presentation to CD4 T cells. Thus there was a clear dissociation between the cytotoxic properties of LLO and its very high antigenicity. Presentation of LLO to CD8 T cells was not as robust as that seen in CD4 T cells, but still occurred in the nanomolar range. APC rapidly bound and internalized LLO, then disrupted endosomal compartments within 4 hours of treatment, allowing endosomal contents to access the cytosol. LLO was also immunogenic after in vivo administration into mice. Our results demonstrate the strength of LLO as an immunogen to both CD4 and CD8 T cells.


Assuntos
Toxinas Bacterianas/imunologia , Toxinas Bacterianas/toxicidade , Proteínas de Choque Térmico/imunologia , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/imunologia , Proteínas Hemolisinas/toxicidade , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/efeitos dos fármacos , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Imunização , Camundongos , Transporte Proteico
14.
Int J Mol Sci ; 13(1): 1173-1185, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22312311

RESUMO

microRNAs represent a family of very small non-coding RNAs that control several physiologic and pathologic processes, including host immune response and cancer by antagonizing a number of target mRNAs. There is limited knowledge about cell expression and the regulatory role of microRNAs following bacterial infections. We investigated whether infection with a Gram-positive bacterium leads to altered expression of microRNAs involved in the host cell response in epithelial cells. Caco-2 cells were infected with Listeria monocytogenes EGD-e, a mutant strain (ΔinlAB or Δhly) or incubated with purified listeriolysin (LLO). Total RNA was isolated and microRNA and target gene expression was compared to the expression in non-infected cells using microRNA microarrays and qRT-PCR. We identified and validated five microRNAs (miR- 146b, miR-16, let-7a1, miR-145 and miR-155) that were significantly deregulated following listerial infection. We show that expression patterns of particular microRNAs strongly depend on pathogen localization and the presence of bacterial effector proteins. Strikingly, miR-155 which was shown to have an important role in inflammatory responses during infection was induced by wild-type bacteria, by LLO-deficient bacteria and following incubation with purified LLO. It was downregulated following ΔinlAB infection indicating a new potent role for internalins in listerial pathogenicity and miRNA regulation. Concurrently, we observed differences in target transcript expression of the investigated miRNAs. We provide first evidence that L. monocytogenes infection leads to deregulation of a set of microRNAs with important roles in host response. Distinct microRNA expression depends on both LLO and pathogen localization.


Assuntos
Listeria monocytogenes/patogenicidade , MicroRNAs/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Células CACO-2 , Citocinas/genética , Citocinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidade , Humanos , Listeria monocytogenes/metabolismo , Listeriose/genética , Listeriose/microbiologia , Listeriose/patologia , Reação em Cadeia da Polimerase em Tempo Real
15.
J Microbiol Biotechnol ; 21(11): 1193-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22127132

RESUMO

Encapsulation of biological material in the permiselective membrane allows to construct a system separating cells from their products, which may find biotechnological as well as biomedical applications in biological processes regulation. Application of a permiselective membrane allows avoiding an attack of the implanted microorganisms on the host. Our aim was to evaluate the performance of Bacillus subtilis encapsulated in an elaborate membrane system producing listeriolysin O, a cytolysin from Listeria monocytogenes, with chosen eukaryotic cells for future application in anticancer treatment. The system of encapsulating in membrane live Bacillus subtilis BR1-S secreting listeriolysin O was proven to exert the effective cytotoxic activity on eukaryotic cells. Interestingly, listeriolysin O showed selective cytotoxic activity on eukaryotic cells: more human leukemia Jurkat T cells were killed than human chronic lymphocytic B cells leukemia at similar conditions in vitro. This system of encapsulated B. subtilis, continuously releasing bacterial products, may affect selectively different types of cells and may have future application in local anticancer treatment.


Assuntos
Bacillus subtilis/metabolismo , Bacillus subtilis/patogenicidade , Toxinas Bacterianas/toxicidade , Composição de Medicamentos/métodos , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Linfócitos T/efeitos dos fármacos , Bacillus subtilis/genética , Toxinas Bacterianas/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Células Jurkat , Listeria monocytogenes/genética
16.
Vascul Pharmacol ; 52(5-6): 207-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20074664

RESUMO

Listeriosis can lead to potentially lethal pulmonary complications in newborns and immune compromised patients, characterized by extensive permeability edema. Listeriolysin (LLO), the main virulence factor of Listeria monocytogenes, induces a dose-dependent hyperpermeability in monolayers of human lung microvascular endothelial cells in vitro. The permeability increasing activity of LLO, which is accompanied by an increased reactive oxygen species (ROS) generation, RhoA activation and myosin light chain (MLC) phosphorylation, can be completely inhibited by the protein kinase C (PKC) alpha/beta inhibitor GO6976, indicating a crucial role for PKC in the induction of barrier dysfunction. The TNF-derived TIP peptide, which mimics the lectin-like domain of the cytokine, blunts LLO-induced hyperpermeability in vitro, upon inhibiting LLO-induced protein kinase C-alpha activation, ROS generation and MLC phosphorylation and upon restoring the RhoA/Rac 1 balance. These results indicate that the lectin-like domain of TNF has a potential therapeutic value in protecting from LLO-induced pulmonary endothelial hyperpermeability.


Assuntos
Toxinas Bacterianas/toxicidade , Endotélio Vascular/metabolismo , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/patogenicidade , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bovinos , Células Cultivadas , Endotélio Vascular/microbiologia , Humanos , Pulmão/citologia , Pulmão/metabolismo , Pulmão/microbiologia , Cadeias Leves de Miosina/metabolismo , Peptídeos/farmacologia , Permeabilidade , Fosforilação , Proteína Quinase C-alfa/antagonistas & inibidores , Artéria Pulmonar/metabolismo , Artéria Pulmonar/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Ovinos , Fator de Necrose Tumoral alfa/química , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Can J Microbiol ; 55(10): 1153-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19935887

RESUMO

Listeriolysin (LLO) is the key virulence factor critical for Listeria monocytogenes pathogenesis. Listerial cytolysin belongs to the family of cholesterol-dependent cytolysins (CDCs), a group of pore-forming toxins produced by related gram-positive bacteria. Most CDCs contain a cysteine residue in the conserved undecapeptide - a sequence that is highly preserved among this group of proteins. Substitutions of cysteine do not always lead to loss of hemolytic activity, questioning the purpose of such strong conservation of this amino acid in the sequence of CDC. The properties of 3 L. monocytogenes strains, a wild type and 2 mutants expressing modified LLO within the cysteine residue, were analyzed in this work. The first of these mutants producing a toxin with cysteine to alanine substitution showed similar features to the wild type except that a thiol-reducing agent was not necessary for hemolytic activity. Another strain secreting LLO containing serine instead of cysteine exhibited strikingly different properties than the wild type. Modified toxin is independent of the reducing reagents, less stable, and shows accelerated kinetics of cytolysis in comparison with the unchanged protein. However, both mutant strains are less invasive in the cell culture model showing the important role of cysteine in L. monocytogenes virulence.


Assuntos
Toxinas Bacterianas/química , Proteínas de Choque Térmico/química , Proteínas Hemolisinas/química , Listeria monocytogenes/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidade , Bovinos , Linhagem Celular , Cisteína/química , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Técnicas In Vitro , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Modelos Moleculares , Estabilidade Proteica , Ratos , Ovinos , Virulência
18.
Infect Immun ; 77(9): 4028-40, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19581399

RESUMO

Many pathogenic gram-positive bacteria release exotoxins that belong to the family of cholesterol-dependent cytolysins. Here, we report that human alpha-defensins HNP-1 to HNP-3 acted in a concentration-dependent manner to protect human red blood cells from the lytic effects of three of these exotoxins: anthrolysin O (ALO), listeriolysin O, and pneumolysin. HD-5 was very effective against listeriolysin O but less effective against the other toxins. Human alpha-defensins HNP-4 and HD-6 and human beta-defensin-1, -2, and -3 lacked protective ability. HNP-1 required intact disulfide bonds to prevent toxin-mediated hemolysis. A fully linearized analog, in which all six cysteines were replaced by aminobutyric acid (Abu) residues, showed greatly reduced binding and protection. A partially unfolded HNP-1 analog, in which only cysteines 9 and 29 were replaced by Abu residues, showed intact ALO binding but was 10-fold less potent in preventing hemolysis. Surface plasmon resonance assays revealed that HNP-1 to HNP-3 bound all three toxins at multiple sites and also that solution-phase HNP molecules could bind immobilized HNP molecules. Defensin concentrations that inhibited hemolysis by ALO and listeriolysin did not prevent these toxins from binding either to red blood cells or to cholesterol. Others have shown that HNP-1 to HNP-3 inhibit lethal toxin of Bacillus anthracis, toxin B of Clostridium difficile, diphtheria toxin, and exotoxin A of Pseudomonas aeruginosa; however, this is the first time these defensins have been shown to inhibit pore-forming toxins. An "ABCDE mechanism" that can account for the ability of HNP-1 to HNP-3 to inhibit so many different exotoxins is proposed.


Assuntos
Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Colesterol/farmacologia , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Glicoproteínas de Membrana/toxicidade , Estreptolisinas/toxicidade , alfa-Defensinas/farmacologia , Sequência de Aminoácidos , Animais , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Concentração de Íons de Hidrogênio , Dados de Sequência Molecular , Coelhos , Soro/fisiologia , alfa-Defensinas/química , alfa-Defensinas/metabolismo
19.
Cell Microbiol ; 11(9): 1382-98, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19500109

RESUMO

Listeria monocytogenes is a bacterial pathogen that replicates within the cytosol of infected host cells. The ability to rapidly escape the phagocytic vacuole is essential for efficient intracellular replication. In the murine model of infection, the pore-forming cytolysin listeriolysin O (LLO) is absolutely required for vacuolar dissolution, as LLO-deficient (DeltaLLO) mutants remain trapped within vacuoles. In contrast, in many human cell types DeltaLLO L. monocytogenes are capable of vacuolar escape at moderate to high frequencies. To better characterize the mechanism of LLO-independent vacuolar escape in human cells, we conducted an RNA interference screen to identify vesicular trafficking factors that play a role in altering vacuolar escape efficiency of DeltaLLO L. monocytogenes. RNA interference knockdown of 18 vesicular trafficking factors resulted in increased LLO-independent vacuolar escape. Our results suggest that knockdown of one factor, RABEP1 (rabaptin-5), decreased the maturation of vacuoles containing DeltaLLO L. monocytogenes. Thus, we provide evidence that increased vacuolar escape of DeltaLLO L. monocytogenes in human cells correlates with slower vacuolar maturation. We also determined that increased LLO-independent dissolution of vacuoles during RABEP1 knockdown required the bacterial broad-range phospholipase C (PC-PLC). We hypothesize that slowing the kinetics of vacuolar maturation generates an environment conducive for vacuolar escape mediated by the bacterial phospholipases.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Vacúolos/microbiologia , Vacúolos/fisiologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Interferência de RNA , Proteínas de Transporte Vesicular/fisiologia
20.
J Infect Dis ; 199(1): 124-33, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19032107

RESUMO

BACKGROUND: Toll-like receptor (TLR) 2 is the principal recognition receptor for gram-positive microbes. However, in some gram-positive bacterial infections, TLR2 is dispensable. One of the outstanding questions regarding host-bacteria interactions is why TLR2 is essential in some infections but dispensable in others. METHODS: We used a combination of bacterial plating, flow cytometry, enzyme-linked immunosorbent assay, and reverse-transcriptase polymerase chain reaction to analyze the inflammatory responses induced by Listeria monocytogenes and its toxin listeriolysin O (LLO) in vitro and in vivo. We analyzed wild-type, TLR2(-/-)-, TLR4(-/-)-, MyD88(-/-)-, interleukin (IL)-1beta(-/-)-, and IL-18(-/-)-deficient mice and the bone marrow-derived mast cells obtained from these respective groups. RESULTS: TLR2(-/-) mice had unaltered L. monocytogenes clearance and did not experience impairment of cytokine/chemokine induction and neutrophil mobilization by L. monocytogenes or purified LLO, but they were unresponsive to the LLO-deficient mutant L. monocytogenes (LmDeltahly). We show that L. monocytogenes and LLO mediate such responses in part via interleukin (IL)-1beta and IL-18-MyD88 pathways. CONCLUSIONS: The results illustrate that signals triggered by LLO contribute to TLR2 redundancy in recognition of L. monocytogenes. Under normal conditions, multiple and, sometimes, redundant pathways cooperate to induce a rapid antimicrobial defense. When one signaling pathway-in this case, TLR2-is removed from the system, the other pathways are still capable of mounting a sufficient response to ensure survival of the host.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/isolamento & purificação , Listeriose/fisiopatologia , Transdução de Sinais/fisiologia , Receptor 2 Toll-Like/deficiência , Receptor 4 Toll-Like/deficiência , Animais , Medula Óssea/fisiologia , Primers do DNA , Inflamação/microbiologia , Interleucina-18/deficiência , Interleucina-18/genética , Interleucina-1beta/deficiência , Interleucina-1beta/genética , Listeriose/genética , Mastócitos/fisiologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA