Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 294
Filtrar
1.
Microbiol Spectr ; 10(4): e0181022, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35856678

RESUMO

Listeria monocytogenes remains a nonnegligible cause of foodborne infection, posing a critical threat to public health. Under the global antibiotic crisis, novel alternative approaches are urgently needed. The indispensable role of listeriolysin O (LLO) in the intracellular life cycle, barrier penetration, colonization, and systemic dissemination of L. monocytogenes renders it a potent drug target, which means curbing L. monocytogenes via interfering with LLO-associated pathogenic mechanisms. Here, we identified kaempferol, a natural small molecule compound, as an effective LLO inhibitor that engaged the residues Glu437, Ile468, and Tyr469 of LLO, thereby suppressing LLO-mediated membrane perforation and barrier disruption. Moreover, we found that kaempferol also suppressed host-derived inflammation in a distinct way independent of LLO inhibition. The in vivo study revealed that kaempferol treatment significantly reduced bacterial burden and cytokine burst in target organs, thereby effectively protecting mice from systemic L. monocytogenes infection. Our findings present kaempferol as a potential therapeutic application for L. monocytogenes infection, which is less likely to induce drug resistance than antibiotics because of its superiority of interfering with the pathogenesis process rather than exerting pressure on bacterial viability. IMPORTANCE Currently, we are facing a global crisis of antibiotic resistance, and novel alternative approaches are urgently needed to curb L. monocytogenes infection. Our study demonstrated that kaempferol alleviated L. monocytogenes infection via suppressing LLO pore formation and inflammation response, which might represent a novel antimicrobial-independent strategy to curb listeriosis.


Assuntos
Listeria monocytogenes , Listeriose , Animais , Toxinas Bacterianas , Proteínas de Choque Térmico , Proteínas Hemolisinas/fisiologia , Inflamação/tratamento farmacológico , Quempferóis/farmacologia , Quempferóis/uso terapêutico , Listeria monocytogenes/fisiologia , Listeriose/tratamento farmacológico , Listeriose/microbiologia , Camundongos
2.
Toxins (Basel) ; 13(11)2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34822531

RESUMO

The insect integument (exoskeleton) is an effective physiochemical barrier that limits disease-causing agents to a few portals of entry, including the gastrointestinal and reproductive tracts. The bacterial biopesticide Bacillus thuringiensis (Bt) enters the insect host via the mouth and must thwart gut-based defences to make its way into the body cavity (haemocoel) and establish infection. We sought to uncover the main antibacterial defences of the midgut and the pathophysiological features of Bt in a notable insect pest, the Colorado potato beetle Leptinotarsa decemlineata (CPB). Exposing the beetles to both Bt spores and their Cry3A toxins (crystalline δ-endotoxins) via oral inoculation led to higher mortality levels when compared to either spores or Cry3A toxins alone. Within 12 h post-exposure, Cry3A toxins caused a 1.5-fold increase in the levels of reactive oxygen species (ROS) and malondialdehyde (lipid peroxidation) within the midgut - key indicators of tissue damage. When Cry3A toxins are combined with spores, gross redox imbalance and 'oxidation stress' is apparent in beetle larvae. The insect detoxification system is activated when Bt spores and Cry3A toxins are administered alone or in combination to mitigate toxicosis, in addition to elevated mRNA levels of candidate defence genes (pattern-recognition receptor, stress-regulation, serine proteases, and prosaposin-like protein). The presence of bacterial spores and/or Cry3A toxins coincides with subtle changes in microbial community composition of the midgut, such as decreased Pseudomonas abundance at 48 h post inoculation. Both Bt spores and Cry3A toxins have negative impacts on larval health, and when combined, likely cause metabolic derangement, due to multiple tissue targets being compromised.


Assuntos
Toxinas de Bacillus thuringiensis/fisiologia , Bacillus thuringiensis/fisiologia , Besouros/microbiologia , Endotoxinas/fisiologia , Proteínas Hemolisinas/fisiologia , Controle de Insetos , Controle Biológico de Vetores , Esporos Bacterianos/fisiologia , Animais , Besouros/crescimento & desenvolvimento , Larva/crescimento & desenvolvimento , Larva/microbiologia , Longevidade
3.
Cell Host Microbe ; 28(3): 402-410.e5, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32544461

RESUMO

Bacteria and their toxins are associated with significant human morbidity and mortality. While a few bacterial toxins are well characterized, the mechanism of action for most toxins has not been elucidated, thereby limiting therapeutic advances. One such example is the highly potent pore-forming toxin, hemolysin BL (HBL), produced by the gram-positive pathogen Bacillus cereus. However, how HBL exerts its effects and whether it requires any host factors is unknown. Here, we describe an unbiased genome-wide CRISPR-Cas9 knockout screen that identified LPS-induced TNF-α factor (LITAF) as the HBL receptor. Using LITAF-deficient cells, a second, subsequent whole-genome CRISPR-Cas9 screen identified the LITAF-like protein CDIP1 as a second, alternative receptor. We generated LITAF-deficient mice, which exhibit marked resistance to lethal HBL challenges. This work outlines and validates an approach to use iterative genome-wide CRISPR-Cas9 screens to identify the complement of host factors exploited by bacterial toxins to exert their myriad biological effects.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Bacillus cereus/patogenicidade , Proteínas de Bactérias/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Hemolisinas/fisiologia , Receptores de Enterotoxina/fisiologia , Fatores de Transcrição/fisiologia , Animais , Proteínas Reguladoras de Apoptose/genética , Células CHO , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Cricetulus , Proteínas de Ligação a DNA/genética , Células Endoteliais , Feminino , Técnicas de Silenciamento de Genes , Estudo de Associação Genômica Ampla , Interações Hospedeiro-Patógeno , Humanos , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Enterotoxina/genética , Fatores de Transcrição/genética , Fatores de Virulência
4.
J Neurogenet ; 34(3-4): 430-439, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32362197

RESUMO

Across animal phyla, sleep is associated with increased cellular repair, suggesting that cellular damage may be a core component of sleep pressure. In support of this notion, sleep in the nematode Caenorhabditis elegans can be triggered by damaging conditions, including noxious heat, high salt, and ultraviolet light exposure. It is not clear, however, whether this stress-induced sleep (SIS) is a direct consequence of cellular damage, or of a resulting energy deficit, or whether it is triggered simply by the sensation of noxious conditions. Here, we show that thermosensation is dispensable for heat-induced sleep, that osmosensation is dispensable for salt-induced sleep, and that wounding is also a sleep trigger, together indicating that SIS is not triggered by sensation of noxious environments. We present evidence that genetic variation in cellular repair pathways impacts sleep amount, and that SIS involves systemic monitoring of cellular damage. We show that the low-energy sensor AMP-activated protein kinase (AMPK) is not required for SIS, suggesting that energy deficit is not the primary sleep trigger. Instead, AMPK-deficient animals display enhanced SIS responses, and pharmacological activation of AMPK reduces SIS, suggesting that ATP-dependent repair of cellular damage mitigates sleep pressure.


Assuntos
Caenorhabditis elegans/fisiologia , Sono/fisiologia , Cicatrização/fisiologia , Adenilato Quinase/fisiologia , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Toxinas de Bacillus thuringiensis/fisiologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , Endotoxinas/fisiologia , Ativação Enzimática , Proteínas Hemolisinas/fisiologia , Temperatura Alta , Noxas , Pressão Osmótica/fisiologia , Ribonucleotídeos/farmacologia , Sono/genética , Cloreto de Sódio/farmacologia , Estresse Fisiológico/genética , Estresse Fisiológico/fisiologia , Raios Ultravioleta , Ferimentos e Lesões/fisiopatologia
5.
Biochemistry ; 59(4): 605-614, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31808340

RESUMO

Thermostable direct hemolysin (TDH) of Vibrio parahemolyticus is a membrane-damaging pore-forming toxin with potent cytolytic/cytotoxic activity. TDH exists as a tetramer consisting of protomers with a core ß-sandwich domain, flanked by an 11-amino acid long N-terminal region (NTR). This NTR could not be modeled in the previously determined crystal structure of TDH. Moreover, the functional implication of NTR for the membrane-damaging action of TDH remains unknown. In the present study, we have explored the implications of NTR for the structure-function mechanism of TDH. Our data show that the presence of NTR modulates the physicochemical property of TDH in terms of augmenting the amyloidogenic propensity of the protein. Deletion of NTR compromises the binding of TDH toward target cell membranes and drastically affects the membrane-damaging cytolytic/cytotoxic activity of the toxin. Mutations of aromatic/hydrophobic residues within NTR also confer compromised cell-killing activity. Moreover, covalent trapping of NTR, via an engineered disulfide bond, against the core ß-sandwich domain also abrogates the cytolytic/cytotoxic activity of TDH. This observation suggests that an unrestrained configuration of NTR is crucial for the membrane-damaging action of TDH. On the basis of our study, we propose a model explaining the role of NTR in the membrane-damaging function of TDH.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/ultraestrutura , Proteínas de Bactérias/química , Toxinas Bacterianas/metabolismo , Fenômenos Bioquímicos/genética , Transporte Biológico/genética , Proteínas Hemolisinas/química , Proteínas Hemolisinas/fisiologia , Hemólise , Humanos , Mutação/genética , Subunidades Proteicas/metabolismo , Vibrio parahaemolyticus/química , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/metabolismo
6.
Adv Exp Med Biol ; 1183: 35-51, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31376138

RESUMO

Bordetella pertussis produces several toxins that affect host-pathogen interactions. Of these, the major toxins that contribute to pertussis infection and disease are pertussis toxin, adenylate cyclase toxin-hemolysin and tracheal cytotoxin. Pertussis toxin is a multi-subunit protein toxin that inhibits host G protein-coupled receptor signaling, causing a wide array of effects on the host. Adenylate cyclase toxin-hemolysin is a single polypeptide, containing an adenylate cyclase enzymatic domain coupled to a hemolysin domain, that primarily targets phagocytic cells to inhibit their antibacterial activities. Tracheal cytotoxin is a fragment of peptidoglycan released by B. pertussis that elicits damaging inflammatory responses in host cells. This chapter describes these three virulence factors of B. pertussis, summarizing background information and focusing on the role of each toxin in infection and disease pathogenesis, as well as their role in pertussis vaccination.


Assuntos
Toxina Adenilato Ciclase/toxicidade , Bordetella pertussis/patogenicidade , Toxina Pertussis/toxicidade , Fatores de Virulência de Bordetella/toxicidade , Adenilil Ciclases/fisiologia , Toxinas Bacterianas , Bordetella pertussis/enzimologia , Bordetella pertussis/genética , Proteínas Hemolisinas/fisiologia , Humanos , Fatores de Virulência , Coqueluche/microbiologia , Coqueluche/prevenção & controle
7.
Mol Microbiol ; 112(4): 1253-1269, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31376198

RESUMO

Proteins of the aegerolysin family have a high abundance in Fungi. Due to their specific binding to membrane lipids, and their membrane-permeabilization potential in concert with protein partner(s) belonging to a membrane-attack-complex/perforin (MACPF) superfamily, they were proposed as useful tools in different biotechnological and biomedical applications. In this work, we performed functional studies on expression of the genes encoding aegerolysin and MACPF-like proteins in Aspergillus niger. Our results suggest the sporulation process being crucial for strong induction of the expression of all these genes. However, deletion of either of the aegerolysin genes did not influence the growth, development, sporulation efficiency and phenotype of the mutants, indicating that aegerolysins are not key factors in the sporulation process. In all our expression studies we noticed a strong correlation in the expression of one aegerolysin and MACPF-like gene. Aegerolysins were confirmed to be secreted from the fungus. We also showed the specific interaction of a recombinant A. niger aegerolysin with an invertebrate-specific membrane sphingolipid. Moreover, using this protein labelled with mCherry we successfully stained insect cells membranes containing this particular sphingolipid. Our combined results suggest, that aegerolysins in this species, and probably also in other aspergilli, could be involved in defence against predators.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Hemolisinas/metabolismo , Perforina/metabolismo , Aspergillus niger/genética , Aspergillus niger/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/genética , Proteínas Fúngicas/fisiologia , Regulação Fúngica da Expressão Gênica/genética , Proteínas Hemolisinas/fisiologia , Proteínas de Membrana/metabolismo , Perforina/genética , Esfingolipídeos/metabolismo , Esporos Fúngicos/genética , Esporos Fúngicos/metabolismo
8.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(2): 152-156, 2019 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-31106531

RESUMO

OBJECTIVE: To study the primary function of ivanolysin O (ILO) and Listeriolysin O (LLO) and compare the effects of these two hemolysins in helping bacteria adhere, invade cell and intracellularly multiply. METHODS: The targeting plasmids carrying the upstream and downstream sequences of i-hly and lacZ gene sequence or hly gene sequence were constructed. Then two recombinant strains, the ILO deletion strain LIΔi-hly::lacZ and LLO compensative expressing strain LIΔi-hly::hly, were constructed by plasmid targeting recombinant technique. The adhesive and invasive ability of LIΔi-hly::hly, LI and LIΔi-hly::lacZ were evaluated in HepG2 cells, and their intracellular multiplication abilities were evaluated in RAW264.7 macrophages. RESULTS: Genome sequences of the recombinant strains were as expected. The adhesive rate of LIΔi-hly::i-hly LI and LIΔi-hly::lacZ were (3.43±0.82)%, (3.43±1.59)% and (3.41±1.12)% respectively, and the invasive rate were (1.74±0.46)%, (1.22±0.75)% and (1.39±0.46)% respectively. Difference in adhesive and invasive rates showed no significance. Among three strains, LIΔi-hly::lacZ showed the lowest intracellular proliferation rate, and LIΔi-hly::hly possessed the highest intracellular proliferation rate in RAW264.7 macrophages. CONCLUSION: The intracellular multiplication ability of LI is related to ILO. Deletion of ILO induces a distinct decrease in intracellular multiplication for LI. Compared with ILO, LLO shows a stronger ability in helping the bacteria escape from the phagosome into the host cell cytosol.


Assuntos
Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Listeria/citologia , Animais , Toxinas Bacterianas , Camundongos , Células RAW 264.7
9.
Infect Immun ; 87(8)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31138613

RESUMO

Streptococcus suis is an important zoonotic pathogen which can infect humans and pigs worldwide, posing a potential risk to global public health. Suilysin, a pore-forming cholesterol-dependent cytolysin, is considered to play an important role in the pathogenesis of S. suis infections. It is known that infection with influenza A viruses may favor susceptibility to secondary bacterial infection, resulting in more severe disease and increased mortality. However, the molecular mechanisms underlying these coinfections are incompletely understood. Applying highly differentiated primary porcine respiratory epithelial cells grown under air-liquid interface (ALI) conditions, we analyzed the contribution of swine influenza viruses (SIV) to the virulence of S. suis, with a special focus on its cytolytic toxin, suilysin. We found that during secondary bacterial infection, suilysin of S. suis contributed to the damage of well-differentiated respiratory epithelial cells in the early stage of infection, whereas the cytotoxic effects induced by SIV became prominent at later stages of infection. Prior infection by SIV enhanced the adherence to and colonization of porcine airway epithelial cells by a wild-type (wt) S. suis strain and a suilysin-negative S. suis mutant in a sialic acid-dependent manner. A striking difference was observed with respect to bacterial invasion. After bacterial monoinfection, only the wt S. suis strain showed an invasive phenotype, whereas the mutant remained adherent. When the epithelial cells were preinfected with SIV, the suilysin-negative mutant also showed an invasion capacity. Therefore, we propose that coinfection with SIV may compensate for the lack of suilysin in the adherence and invasion process of suilysin-negative S. suis.


Assuntos
Aderência Bacteriana/fisiologia , Coinfecção/microbiologia , Proteínas Hemolisinas/fisiologia , Pulmão/microbiologia , Infecções por Orthomyxoviridae/microbiologia , Streptococcus suis/patogenicidade , Animais , Células Cultivadas , Cães , Células Epiteliais/microbiologia , Suínos
10.
Exp Eye Res ; 181: 263-270, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30822400

RESUMO

Colonization by Staphylococcus aureus (S. aureus) has been implicated in many infectious and wound healing disorders. This study was performed to characterize the pathogenic role of S. aureus alpha-hemolysin (alpha-toxin) in corneal epithelial wound healing and infectious keratitis in the setting of a corneal wound. The effect of wild-type and isogenic Hla mutant (α-hemolysin gene deleted) S. aureus bacteria and conditioned media on corneal epithelial wound healing was tested in vitro using a scratch assay and in vivo using a murine epithelial debridement model. The invasiveness of wild-type and Hla mutant S. aureus was evaluated in vitro in human corneal epithelial cells and in vivo in a murine model of infectious keratitis following total epithelial debridement. S. aureus and its conditioned media significantly delayed epithelial wound closure both in vitro (P < 0.05) and in vivo (P < 0.05). The effect of S. aureus on wound healing was significantly diminished with the Hla mutant strain (P < 0.05). Likewise, compared to the wild-type strain, the Hla mutant strain demonstrated significantly reduced ability to invade corneal epithelial cells in vitro (P < 0.05) and infect murine corneas following total epithelial debridement in vivo (P < 0.05). In conclusion, S. aureus alpha-hemolysin plays a major role in the pathologic modulation of corneal epithelial wound healing and the intracellular invasion of the bacteria. Limiting colonization by S. aureus and/or blocking alpha-hemolysin may provide a therapeutic approach for corneal wound healing and infectious disorders.


Assuntos
Doenças da Córnea/microbiologia , Epitélio Corneano/lesões , Proteínas Hemolisinas/fisiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Cicatrização/fisiologia , Animais , Doenças da Córnea/patologia , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Epitélio Corneano/microbiologia , Humanos , Ceratite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/patologia
11.
Toxins (Basel) ; 10(9)2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231498

RESUMO

Cytolytic pore-forming toxins including alpha hemolysin (Hla) and bicomponent leukotoxins play an important role in the pathogenesis of Staphylococcus aureus. These toxins kill the polymorphonuclear phagocytes (PMNs), disrupt epithelial and endothelial barriers, and lyse erythrocytes to provide iron for bacterial growth. The expression of these toxins is regulated by the two-component sensing systems Sae and Agr. Here, we report that a point mutation (L18P) in SaeS, the histidine kinase sensor of the Sae system, renders the S. aureus Newman hemolytic activity fully independent of Hla and drastically increases the PMN lytic activity. Furthermore, this Hla-independent activity, unlike Hla itself, can lyse human erythrocytes. The Hla-independent activity towards human erythrocytes was also evident in USA300, however, under strict agr control. Gene knockout studies revealed that this Hla-independent Sae-regulated activity was entirely dependent on gamma hemolysin A subunit (HlgA). In contrast, hemolytic activity of Newman towards human erythrocytes from HlgAB resistant donors was completely dependent on agr. The culture supernatant from Newman S. aureus could be neutralized by antisera against two vaccine candidates based on LukS and LukF subunits of Panton-Valentine leukocidin but not by an anti-Hla neutralizing antibody. These findings display the complex involvement of Sae and Agr systems in regulating the virulence of S. aureus and have important implications for vaccine and immunotherapeutics development for S. aureus disease in humans.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas Hemolisinas/fisiologia , Proteínas Quinases/fisiologia , Staphylococcus aureus/patogenicidade , Animais , Toxinas Bacterianas , Eritrócitos/fisiologia , Hemólise , Humanos , Camundongos , Pneumonia , Virulência
12.
Microb Pathog ; 123: 242-245, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30031890

RESUMO

Vibrio parahaemolyticus is a leading cause of seafood-associated bacterial gastroenteritis. The pathogen produces the thermostable direct hemolysin (TDH), which is the sole cause of the Kanagawa phenomenon (KP), a special ß-type haemolysis in the Wagatsuma agar. TDH also exerts several other biological activities, the major includes lethal toxicity, cytotoxicity, and enterotoxicity. The structure and roles of TDH and the transcriptional regulation of tdh genes, are summarized in this review, which will give a better understanding of the pathogenesis of V. parahaemolyticus.


Assuntos
Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/fisiologia , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Gastroenterite/microbiologia , Gastroenterite/veterinária , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos/genética , Modelos Moleculares , Pandemias , Conformação Proteica , Alimentos Marinhos/microbiologia , Vibrioses/microbiologia , Vibrioses/veterinária , Vibrio parahaemolyticus/patogenicidade , Fatores de Virulência/genética
13.
Toxins (Basel) ; 10(4)2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29659477

RESUMO

Biofilms complicate treatment of Staphylococcus aureus (SA) wound infections. Previously, we determined alpha-toxin (AT)-promoted SA biofilm formation on mucosal tissue. Therefore, we evaluated SA wound isolates for AT production and biofilm formation on epithelium and assessed the role of AT in biofilm formation. Thirty-eight wound isolates were molecularly typed by pulsed-field gel electrophoresis (PFGE), multilocus sequence typing (ST), and spa typing. We measured biofilm formation of these SA isolates in vitro and ex vivo and quantified ex vivo AT production. We also investigated the effect of an anti-AT monoclonal antibody (MEDI4893*) on ex vivo biofilm formation by methicillin-resistant SA (USA 300 LAC) and tested whether purified AT rescued the biofilm defect of hla mutant SA strains. The predominant PFGE/ST combinations were USA100/ST5 (50%) and USA300/ST8 (33%) for methicillin-resistant SA (MRSA, n = 18), and USA200/ST30 (20%) for methicillin-susceptible SA (MSSA, n = 20). Ex vivo AT production correlated significantly with ex vivo SA wound isolate biofilm formation. Anti-alpha-toxin monoclonal antibody (MEDI4893*) prevented ex vivo biofilm formation by MRSA USA300 strain LAC. Wild-type AT rescued the ex vivo biofilm defect of non-AT producing SA strains. These findings provide evidence that AT plays a role in SA biofilm formation on epithelial surfaces and suggest that neutralization of AT may be useful in preventing and treating SA infections.


Assuntos
Proteínas Hemolisinas/fisiologia , Staphylococcus aureus/fisiologia , Animais , Toxinas Bacterianas , Biofilmes , Feminino , Genótipo , Humanos , Mucosa , Suínos , Vagina , Ferimentos e Lesões
14.
Mol Biol Cell ; 29(3): 270-284, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29187576

RESUMO

Host cell invasion is an indispensable step for a successful infection by intracellular pathogens. Recent studies identified pathogen-induced host cell plasma membrane perforation as a novel mechanism used by diverse pathogens (Trypanosoma cruzi, Listeria monocytogenes, and adenovirus) to promote their internalization into target cells. It was concluded that T. cruzi and adenovirus damage the host cell plasma membrane to hijack the endocytic-dependent membrane resealing machinery, thereby invading the host cell. We studied L. monocytogenes and its secreted pore-forming toxin listeriolysin O (LLO) to identify key signaling events activated upon plasma membrane perforation that lead to bacterial internalization. Using various approaches, including fluorescence resonance energy transfer imaging, we found that the influx of extracellular Ca2+ subsequent to LLO-mediated plasma membrane perforation is required for the activation of a conventional protein kinase C (cPKC). cPKC is positioned upstream of Rac1 and the Arp2/3 complex, which activation leads to F-actin--dependent bacterial internalization. Inhibition of this pathway did not prevent membrane resealing, revealing that perforation-dependent L. monocytogenes endocytosis is distinct from the resealing machinery. These studies identified the LLO-dependent endocytic pathway of L. monocytogenes and support a novel model for pathogen uptake promoted by plasma membrane injury that is independent of membrane resealing.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/fisiologia , Listeriose/fisiopatologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/metabolismo , Proteínas de Bactérias , Membrana Celular/metabolismo , Citoplasma/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Células Hep G2 , Humanos , Listeria monocytogenes/patogenicidade , Listeria monocytogenes/fisiologia , Membranas/metabolismo , Proteína Quinase C/metabolismo , Proteólise , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo
15.
Rev. argent. microbiol ; 49(3): 273-281, set. 2017. graf, tab
Artigo em Inglês | LILACS | ID: biblio-958008

RESUMO

A total of 268 Bacillus thuringiensis strains obtained from different sources of Argentina were analyzed to determine the diversity and distribution of the cryl, cry2, cry8, cry9 and vip3A genes encoding for lepidopteran-specific insecticidal proteins. Twin strains were excluded. Ten different profiles were detected among the 80 selected B. thuringiensis strains. Two of these profiles (cry1Aa, cry1Ac, cry1Ia, cry2Aa, cry2Ab and vip3Aa (35/80), and cry1Aa, cry1Ab, cry1Ac, cry1Ia, cry2Aa, cry2Ab and vip3Aa (25/80)) pooled 75% of the strains. The existence of this low diversity is rare, since in most of the studied collections a great diversity of insecticidal toxin gene profiles has been described. In addition, the most frequently detected profile was also most frequently derived from soil (70%), stored product dust (59%) and spider webs (50%). In contrast, the cry1Aa, cry1Ab, cry1Ac, cry1Ia, cry2Aa, cry2Ab and vip3Aa profiles were mainly detected in strains isolated from leaves (40%) and dead insect larvae (50%). Six of the identified insecticidal toxin gene profiles were discovered in strains isolated from stored product dust and leaves indicating higher diversity of profiles in these kinds of sources than in others. Some strains with high insecticidal activity against Epinotia aporema (Lepidoptera) larvae were identified, which is important to explore future microbial strategies for the control of this crop pest in the region.


Se analizaron 268 cepas de Bacillus thuringiensis obtenidas de diferentes fuentes de Argentina con el objeto de determinar la diversidad y distribución de genes cryl, cry2, cry8, cry9 y vip3A, que codifican proteínas insecticidas lepidóptero-específicas. Se excluyeron las cepas gemelas. Se detectaron solo diez perfiles diferentes entre los 80 B. thuringiensis seleccionados. Dos de estos perfiles, el cry1Aa, cry1Ac, crylIa, cry2Aa, cry2Ab y vip3Aa (35/80) y el cry1Aa, cry1Ab, cry1Ac, crylIa, cry2Aa, cry2Ab y vip3Aa (25/80), comprendieron el 75% de las cepas seleccionadas. La existencia de esta baja diversidad es una rareza, ya que en la mayor parte de las colecciones estudiadas se ha descrito una gran diversidad de perfiles de genes de toxinas insecticidas. El perfil detectado con mayor frecuencia se obtuvo principalmente de cepas procedentes de suelo (el 70% de los de esa fuente lo tenían), también fue mayoritario entre los procedentes de polvo de producto almacenado (59%) y en los que procedían de telas de arana (50%). En cambio, el perfil cry1Aa, cry1Ab, cry1Ac, crylIa, cry2Aa, cry2Ab y vip3Aa se detectó principalmente en las cepas aisladas de hojas (40%) y de larvas de insectos muertos (50%). Seis de los perfiles identificados fueron encontrados en cepas aisladas de polvo de producto almacenado y de hojas, lo que indica una mayor diversidad de perfiles en estas fuentes que en otras. Se identificaron algunas cepas con alta actividad insecticida contra larvas de Epinotia aporema (Lepidoptera), hallazgo importante para explorar en el futuro estrategias microbianas para el control de esta plaga en la región.


Assuntos
Animais , Bacillus thuringiensis , Toxinas Bacterianas , Genes Bacterianos , Proteínas Hemolisinas , Argentina , Bacillus thuringiensis/genética , Bacillus thuringiensis/patogenicidade , Proteínas de Bactérias , Toxinas Bacterianas/genética , Controle Biológico de Vetores , Endotoxinas , Proteínas Hemolisinas/fisiologia , Proteínas Hemolisinas/genética , Larva , Lepidópteros
16.
Rev Argent Microbiol ; 49(3): 273-281, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28576334

RESUMO

A total of 268 Bacillus thuringiensis strains obtained from different sources of Argentina were analyzed to determine the diversity and distribution of the cry1, cry2, cry8, cry9 and vip3A genes encoding for lepidopteran-specific insecticidal proteins. Twin strains were excluded. Ten different profiles were detected among the 80 selected B. thuringiensis strains. Two of these profiles (cry1Aa, cry1Ac, cry1Ia, cry2Aa, cry2Ab and vip3Aa (35/80), and cry1Aa, cry1Ab, cry1Ac, cry1Ia, cry2Aa, cry2Ab and vip3Aa (25/80)) pooled 75% of the strains. The existence of this low diversity is rare, since in most of the studied collections a great diversity of insecticidal toxin gene profiles has been described. In addition, the most frequently detected profile was also most frequently derived from soil (70%), stored product dust (59%) and spider webs (50%). In contrast, the cry1Aa, cry1Ab, cry1Ac, cry1Ia, cry2Aa, cry2Ab and vip3Aa profiles were mainly detected in strains isolated from leaves (40%) and dead insect larvae (50%). Six of the identified insecticidal toxin gene profiles were discovered in strains isolated from stored product dust and leaves indicating higher diversity of profiles in these kinds of sources than in others. Some strains with high insecticidal activity against Epinotia aporema (Lepidoptera) larvae were identified, which is important to explore future microbial strategies for the control of this crop pest in the region.


Assuntos
Bacillus thuringiensis , Toxinas Bacterianas , Genes Bacterianos , Proteínas Hemolisinas , Animais , Argentina , Bacillus thuringiensis/genética , Bacillus thuringiensis/patogenicidade , Proteínas de Bactérias , Toxinas Bacterianas/genética , Endotoxinas , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/fisiologia , Larva , Lepidópteros , Controle Biológico de Vetores
17.
Cell Microbiol ; 19(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27739224

RESUMO

Bacterial pathogens can interfere during infection with host cell organelles, such as mitochondria, the endoplasmic reticulum-Golgi system or nuclei. As important cellular functions are often compartmentalized in these organelles, their targeting allows pathogens to manipulate key host functions during infection. Here, we identify lysosomes as a new class of organelles targeted by the pathogenic bacterium Listeria monocytogenes. We demonstrate that extracellular Listeria, via secretion of the pore-forming toxin listeriolysin O, alters lysosomal integrity in epithelial cells but not in macrophages. Listeriolysin O induces lysosomal membrane permeabilization and release of lysosomal content, such as cathepsins proteases, which remain transiently active in the host cytosol. We furthermore show that other bacterial pore-forming toxins, such as perfringolysin O and pneumolysin, also induce lysosomes alteration. Together, our data unveil a novel activity of bacterial cholesterol-dependent cytolysins.


Assuntos
Células Epiteliais/microbiologia , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Listeria monocytogenes/fisiologia , Listeriose/microbiologia , Lisossomos/fisiologia , Animais , Toxinas Bacterianas , Células CACO-2 , Permeabilidade da Membrana Celular , Células HeLa , Células Hep G2 , Humanos , Listeriose/patologia , Lisossomos/microbiologia , Camundongos , Proteólise , Células RAW 264.7
18.
Sci Rep ; 6: 29000, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27383371

RESUMO

Streptococcus agalactiae can cause urinary tract infection (UTI) including cystitis and asymptomatic bacteriuria (ABU). The early host-pathogen interactions that occur during S. agalactiae UTI and subsequent mechanisms of disease pathogenesis are poorly defined. Here, we define the early interactions between human bladder urothelial cells, monocyte-derived macrophages, and mouse bladder using uropathogenic S. agalactiae (UPSA) 807 and ABU-causing S. agalactiae (ABSA) 834 strains. UPSA 807 adhered, invaded and killed bladder urothelial cells more efficiently compared to ABSA 834 via mechanisms including low-level caspase-3 activation, and cytolysis, according to lactate dehydrogenase release measures and cell viability. Severe UPSA 807-induced cytotoxicity was mediated entirely by the bacterial ß-hemolysin/cytolysin (ß-H/C) because an ß-H/C-deficient UPSA 807 isogenic mutant, UPSA 807ΔcylE, was not cytotoxic in vitro; the mutant was also significantly attenuated for colonization in the bladder in vivo. Analysis of infection-induced cytokines, including IL-8, IL-1ß, IL-6 and TNF-α in vitro and in vivo revealed that cytokine and chemokine responses were dependent on expression of ß-H/C that also elicited severe bladder neutrophilia. Thus, virulence of UPSA 807 encompasses adhesion to, invasion of and killing of bladder cells, pro-inflammatory cytokine/chemokine responses that elicit neutrophil infiltration, and ß-H/C-mediated subversion of innate immune-mediated bacterial clearance from the bladder.


Assuntos
Proteínas de Bactérias/fisiologia , Bacteriúria/microbiologia , Proteínas Hemolisinas/fisiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/patogenicidade , Infecções Urinárias/microbiologia , Aderência Bacteriana/genética , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Técnicas de Tipagem Bacteriana , Caspase 3/fisiologia , Cistite/microbiologia , Citocinas/biossíntese , Ativação Enzimática , Feminino , Proteínas Hemolisinas/deficiência , Proteínas Hemolisinas/genética , Hemólise , Humanos , Inflamação , L-Lactato Desidrogenase/análise , Pessoa de Meia-Idade , Infiltração de Neutrófilos , Especificidade da Espécie , Streptococcus agalactiae/classificação , Streptococcus agalactiae/isolamento & purificação , Células U937/microbiologia , Urotélio/microbiologia , Virulência/genética
19.
Nano Lett ; 15(10): 6965-73, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26302195

RESUMO

Listeriolysin O (LLO) is the major virulence factor of Listeria monocytogenes and a member of the cholesterol-dependent cytolysin (CDC) family. Gram-positive pathogenic bacteria produce water-soluble CDC monomers that bind cholesterol-dependent to the lipid membrane of the attacked cell or of the phagosome, oligomerize into prepores, and insert into the membrane to form transmembrane pores. However, the mechanisms guiding LLO toward pore formation are poorly understood. Using electron microscopy and time-lapse atomic force microscopy, we show that wild-type LLO binds to membranes, depending on the presence of cholesterol and other lipids. LLO oligomerizes into arc- or slit-shaped assemblies, which merge into complete rings. All three oligomeric assemblies can form transmembrane pores, and their efficiency to form pores depends on the cholesterol and the phospholipid composition of the membrane. Furthermore, the dynamic fusion of arcs, slits, and rings into larger rings and their formation of transmembrane pores does not involve a height difference between prepore and pore. Our results reveal new insights into the pore-forming mechanism and introduce a dynamic model of pore formation by LLO and other CDC pore-forming toxins.


Assuntos
Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Lipídeos/fisiologia , Toxinas Bacterianas , Proteínas de Choque Térmico/ultraestrutura , Proteínas Hemolisinas/ultraestrutura , Listeria monocytogenes/patogenicidade , Microscopia de Força Atômica , Microscopia Eletrônica , Virulência
20.
Vet Res ; 46: 45, 2015 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-25928761

RESUMO

Streptococcus (S.) suis is one of the most important pathogens in pigs causing meningitis, arthritis, endocarditis and serositis. Furthermore, it is also an emerging zoonotic agent. In our previous work we identified a highly specific IgM protease in S. suis, designated Ide(Ssuis) . The objective of this study was to characterize the function of Ide(Ssuis) in the host-pathogen interaction. Edman-sequencing revealed that Ide(Ssuis) cleaves the heavy chain of the IgM molecule between constant domain 2 and 3. As the C1q binding motif is located in the C3 domain, we hypothesized that Ide(Ssuis) is involved in complement evasion. Complement-mediated hemolysis induced by porcine hyperimmune sera containing erythrocyte-specific IgM was abrogated by treatment of these sera with recombinant Ide(Ssuis) . Furthermore, expression of Ide(Ssuis) reduced IgM-triggered complement deposition on the bacterial surface. An infection experiment of prime-vaccinated growing piglets suggested attenuation in the virulence of the mutant 10Δide(Ssuis). Bactericidal assays confirmed a positive effect of Ide(Ssuis) expression on bacterial survival in porcine blood in the presence of high titers of specific IgM. In conclusion, this study demonstrates that Ide(Ssuis) is a novel complement evasion factor, which is important for bacterial survival in porcine blood during the early adaptive (IgM-dominated) immune response.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas do Sistema Complemento/fisiologia , Proteínas Hemolisinas/fisiologia , Imunoglobulina M/fisiologia , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Doenças dos Suínos/imunologia , Animais , Cápsulas Bacterianas/fisiologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Suínos , Doenças dos Suínos/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...