Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 294
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
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.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(2): 152-156, 2019 Mar.
Artigo em Zh | 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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Mol Ther ; 22(7): 1266-1274, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24590046

RESUMO

Bacterial therapies, designed to manufacture therapeutic proteins directly within tumors, could eliminate cancers that are resistant to other therapies. To be effective, a payload protein must be secreted, diffuse through tissue, and efficiently kill cancer cells. To date, these properties have not been shown for a single protein. The gene for Staphylococcus aureus α-hemolysin (SAH), a pore-forming protein, was cloned into Escherichia coli. These bacteria were injected into tumor-bearing mice and volume was measured over time. The location of SAH relative to necrosis and bacterial colonies was determined by immunohistochemistry. In culture, SAH was released and killed 93% of cancer cells in 24 hours. Injection of SAH-producing bacteria reduced viable tissue to 9% of the original tumor volume. By inducing cell death, SAH moved the boundary of necrosis toward the tumor edge. SAH diffused 6.8 ± 0.3 µm into tissue, which increased the volume of affected tissue from 48.6 to 3,120 µm(3). A mathematical model of molecular transport predicted that SAH efficacy is primarily dependent on colony size and the rate of protein production. As a payload protein, SAH will enable effective bacterial therapy because of its ability to diffuse in tissue, kill cells, and expand tumor necrosis.


Assuntos
Proteínas Hemolisinas/metabolismo , Neoplasias Mamárias Animais/terapia , Necrose/etiologia , Staphylococcus aureus/metabolismo , Animais , Feminino , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/fisiologia , Humanos , Células MCF-7 , Masculino , Camundongos , Staphylococcus aureus/genética
14.
Subcell Biochem ; 80: 63-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798008

RESUMO

Cholesterol-dependent cytolysins (CDCs) constitute a family of pore forming toxins secreted by Gram-positive bacteria. These toxins form transmembrane pores by inserting a large ß-barrel into cholesterol-containing membrane bilayers. Binding of water-soluble CDCs to the membrane triggers the formation of oligomers containing 35-50 monomers. The coordinated insertion of more than seventy ß-hairpins into the membrane requires multiple structural conformational changes. Perfringolysin O (PFO), secreted by Clostridium perfringens, has become the prototype for the CDCs. In this chapter, we will describe current knowledge on the mechanism of PFO cytolysis, with special focus on cholesterol recognition, oligomerization, and the conformational changes involved in pore formation.


Assuntos
Toxinas Bacterianas/química , Membrana Celular/química , Citotoxinas , Proteínas Hemolisinas/química , Proteínas Hemolisinas/fisiologia , Sequência de Aminoácidos , Animais , Colesterol/química , Colesterol/metabolismo , Citotoxinas/química , Citotoxinas/fisiologia , Humanos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína
15.
Subcell Biochem ; 80: 161-95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798012

RESUMO

The cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins that are produced by numerous Gram-positive bacterial pathogens. These toxins are released in the extracellular environment as water-soluble monomers or dimers that bind to cholesterol-rich membranes and assemble into large pore complexes. Depending upon their concentration, the nature of the host cell and membrane (cytoplasmic or intracellular) they target, the CDCs can elicit many different cellular responses. Among the CDCs, listeriolysin O (LLO), which is a major virulence factor of the facultative intracellular pathogen Listeria monocytogenes, is involved in several stages of the intracellular lifecycle of the bacterium and displays unique characteristics. It has long been known that following L. monocytogenes internalization into host cells, LLO disrupts the internalization vacuole, enabling the bacterium to replicate into the host cell cytosol. LLO is then used by cytosolic bacteria to spread from cell to cell, avoiding bacterial exposure to the extracellular environment. Although LLO is continuously produced during the intracellular lifecycle of L. monocytogenes, several processes limit its toxicity to ensure the survival of infected cells. It was previously thought that LLO activity was limited to mediating vacuolar escape during bacterial entry and cell to cell spreading. This concept has been challenged by compelling evidence suggesting that LLO secreted by extracellular L. monocytogenes perforates the host cell plasma membrane, triggering important host cell responses. This chapter provides an overview of the well-established intracellular activity of LLO and the multiple roles attributed to LLO secreted by extracellular L. monocytogenes.


Assuntos
Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Listeria monocytogenes/patogenicidade , Animais , Toxinas Bacterianas/química , Colesterol/metabolismo , Citotoxinas/química , Citotoxinas/metabolismo , Citotoxinas/fisiologia , Regulação Bacteriana da Expressão Gênica , Proteínas de Choque Térmico/química , Proteínas Hemolisinas/química , Humanos , Listeria monocytogenes/genética , Modelos Moleculares , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Fatores de Virulência/química , Fatores de Virulência/fisiologia
16.
Subcell Biochem ; 80: 271-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798017

RESUMO

Proteins with membrane-attack complex/perforin (MACPF) domains are found in almost all kingdoms of life, and they have a variety of biological roles, including defence and attack, organism development, and cell adhesion and signalling. The distribution of these proteins in fungi appears to be restricted to some Pezizomycotina and Basidiomycota species only, in correlation with another group of proteins with unknown biological function, known as aegerolysins. These two protein groups coincide in only a few species, and they might operate in concert as cytolytic bi-component pore-forming agents. Representative proteins here include pleurotolysin B, which has a MACPF domain, and the aegerolysin-like protein pleurotolysin A, and the very similar ostreolysin A, which have been purified from oyster mushroom (Pleurotus ostreatus). These have been shown to act in concert to perforate natural and artificial lipid membranes with high cholesterol and sphingomyelin content. The aegerolysin-like proteins provide the membrane cholesterol/sphingomyelin selectivity and recruit oligomerised pleurotolysin B molecules, to create a membrane-inserted pore complex. The resulting protein structure has been imaged with electron microscopy, and it has a 13-meric rosette-like structure, with a central lumen that is ~4-5 nm in diameter. The opened transmembrane pore is non-selectively permeable for ions and smaller neutral solutes, and is a cause of cytolysis of a colloid-osmotic type. The biological significance of these proteins for the fungal life-style is discussed.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Proteínas Fúngicas/fisiologia , Proteínas Hemolisinas/fisiologia , Perforina/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Sequência de Aminoácidos , Animais , Complexo de Ataque à Membrana do Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Humanos , Dados de Sequência Molecular , Perforina/química , Filogenia , Pleurotus/genética , Pleurotus/patogenicidade , Proteínas Citotóxicas Formadoras de Poros/química , Multimerização Proteica/fisiologia , Homologia de Sequência de Aminoácidos
17.
Infect Immun ; 82(6): 2460-71, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24686060

RESUMO

Virulent Streptococcus suis serotype 2 strains are invasive extracellular bacteria causing septicemia and meningitis in piglets and humans. One objective of this study was to elucidate the function of complement in innate immune defense against S. suis. Experimental infection of wild-type (WT) and C3(-/-) mice demonstrated for the first time that the complement system protects naive mice against invasive mucosal S. suis infection. S. suis WT but not an unencapsulated mutant caused mortality associated with meningitis and other pathologies in C3(-/-) mice. The capsule contributed also substantially to colonization of the upper respiratory tract. Experimental infection of C3(-/-) mice with a suilysin mutant indicated that suilysin expression facilitated an early disease onset and the pathogenesis of meningitis. Flow cytometric analysis revealed C3 antigen deposition on the surface of ca. 40% of S. suis WT bacteria after opsonization with naive WT mouse serum, although to a significantly lower intensity than on the unencapsulated mutant. Ex vivo multiplication in murine WT and C3(-/-) blood depended on capsule but not suilysin expression. Interestingly, S. suis invasion of inner organs was also detectable in C5aR(-/-) mice, suggesting that chemotaxis and activation of immune cells via the anaphylatoxin receptor C5aR is, in addition to opsonization, a further important function of the complement system in defense against mucosal S. suis infection. In conclusion, we unequivocally demonstrate here the importance of complement against mucosal S. suis serotype 2 infection and that the capsule of this pathogen is also involved in escape from complement-independent immunity.


Assuntos
Cápsulas Bacterianas/fisiologia , Proteínas do Sistema Complemento/fisiologia , Proteínas Hemolisinas/fisiologia , Streptococcus suis/fisiologia , Anafilatoxinas/fisiologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Interações Hospedeiro-Patógeno/fisiologia , Imunidade Inata/fisiologia , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cavidade Nasal/microbiologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/mortalidade , Streptococcus suis/patogenicidade , Virulência
18.
Southeast Asian J Trop Med Public Health ; 45(6): 1365-75, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26466422

RESUMO

Vibrio cholerae non-O1/non-O139 is capable of producing sporadic outbreaks of cholera-like diarrhea; however, the pathogenic mechanisms of this bacterium remain unclear. The objectives of this study were to: 1) compare the apoptosis induction and cytotoxicity between hlyA-positive and hlyA-negative strains of V. cholerae non-O1/non-O139; 2) clarify the molecular mechanisms by which these strains induce apoptosis; and 3) compare clinical and environmental V. cholerae non-O1/non-O139 isolates with respect to cytotoxicity and ability to induce apoptosis. Using cytotoxicity and apoptosis assays, it was shown that hlyA-positive strains of V. cholerae non-O1/non-O139 had significantly higher cytotoxic activity (70.6%) and levels of apoptosis induction (59.6%) than hlyA- negative strains (37.0% and 37.5%, respectively). Western blot analyses revealed that hlyA-positive strains had significantly increased expression of Bax; active caspase-3 and -9; and significantly decreased expression of NF-κB and Bcl-2 relative to hlyA-negative strains. Expression of BID did not differ significantly between hlyA-positive and negative strains. The truncated BID was not found, indicating that V. cholerae non-O1/non-O139 induces apoptosis through a mitochondria- dependent apoptosis pathway and not an extrinsic pathway. V. cholerae non-O1/ non-O139 isolated from clinical sources exhibited significantly higher cytotoxic activity (79%) and levels of apoptosis induction (65.2%) than bacteria isolated from environmental sources (63% and 54.6%, respectively), suggesting that the clini- cal isolates may have other virulence-associated genes besides hlyA. Our results indicate that hlyA products play a role in cytotoxicity and apoptosis induction and that a mitochondria-dependent apoptosis pathway is involved.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas Hemolisinas/fisiologia , Vibrio cholerae não O1/fisiologia , Animais , Apoptose/fisiologia , Proteínas de Bactérias/genética , Células CHO , Sobrevivência Celular/fisiologia , Cricetinae , Cricetulus , Proteínas Hemolisinas/genética , Interações Hospedeiro-Patógeno , Vibrio cholerae não O1/genética , Vibrio cholerae não O1/patogenicidade
19.
Cell Microbiol ; 14(6): 949-64, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22321539

RESUMO

The endoplasmic reticulum (ER) responds to perturbation of homeostasis with stress. To maintain ER function, a signalling-circuitry has evolved which, when engaged, attempts to reduce a surplus of unfolded proteins by triggering the unfolded protein response (UPR). Several studies have implicated UPR in viral infections, neurodegenerative disorders and metabolic diseases but UPR has not yet been widely linked to bacterial infections. Here we demonstrate that the facultative intracellular pathogen Listeria monocytogenes (Lm) induces ER expansion and UPR prior to host cell entry. Lm activated protein kinase RNA-like ER kinase (PERK) evidenced by the phosphorylation of the α-subunit of eukaryotic translation initiation factor-2 (eIF2α), inositol-requiring protein-1 (IRE1) as shown by detection of spliced X-box binding protein-1 (XBP1) and activating transcription factor-6 (ATF6) as demonstrated by depletion of its inactive form. A mutant LmΔhly strain that did not produce listeriolysin (LLO) lacked the UPR response. Conversely purified LLO activated UPR. Sustained infection with Lm resulted in apoptosis. Induction of ER stress by thapsigargin or tunicamycin reduced intracellular bacterial number. Our findings suggest that UPR plays an important role in the cell autonomous defence responses to bacterial infection.


Assuntos
Interações Hospedeiro-Patógeno , Listeria monocytogenes/fisiologia , Resposta a Proteínas não Dobradas , Fator 6 Ativador da Transcrição/metabolismo , Animais , Apoptose , Toxinas Bacterianas/metabolismo , Cálcio/metabolismo , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/patologia , Chaperona BiP do Retículo Endoplasmático , Inibidores Enzimáticos/farmacologia , Células HeLa , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/fisiologia , Humanos , Imunidade Inata , Listeria monocytogenes/imunologia , Camundongos , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição de Fator Regulador X , Tapsigargina/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tunicamicina/farmacologia , Regulação para Cima , Proteína 1 de Ligação a X-Box
20.
Rev Argent Microbiol ; 45(2): 119-30, 2013.
Artigo em Espanhol | MEDLINE | ID: mdl-23876275

RESUMO

Staphylococcus aureus is the most prevalent mastitis pathogen in Argentina and worldwide. Lack of effectiveness of traditional control measures based on milking hygiene and antibiotic therapy against this organism has led to the development of alternatives directed to prevent the disease. Among them, the manipulation of host immune mechanisms through vaccination has been explored. The identification of virulence factors able to stimulate host immune defenses is key to developing a rational vaccine. S. aureus has multiple virulence factors that interact with the host at different stages of an intramammary infection. The use of some of these factors as immunogens has been shown to elicit protective responses in the host. The structure, function, and use as immunogens of S. aureus virulence factors considered to be relevant at different stages of intrammamary infections caused by this organism are reviewed in this article.


Assuntos
Mastite Bovina/imunologia , Mastite Bovina/microbiologia , Staphylococcus aureus/patogenicidade , Fatores de Virulência/imunologia , Animais , Toxinas Bacterianas , Biofilmes , Bovinos , Coagulase/fisiologia , Feminino , Fibronectinas/fisiologia , Proteínas Hemolisinas/fisiologia , Esfingomielina Fosfodiesterase/fisiologia , Staphylococcus aureus/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA