Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Microbiol ; 121(3): 497-512, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38130174

RESUMO

Legionella pneumophila, the causative agent of a life-threatening pneumonia, intracellularly replicates in a specialized compartment in lung macrophages, the Legionella-containing vacuole (LCV). Secreted proteins of the pathogen govern important steps in the intracellular life cycle including bacterial egress. Among these is the type II secreted PlaA which, together with PlaC and PlaD, belongs to the GDSL phospholipase family found in L. pneumophila. PlaA shows lysophospholipase A (LPLA) activity which increases after secretion and subsequent processing by the zinc metalloproteinase ProA within a disulfide loop. Activity of PlaA contributes to the destabilization of the LCV in the absence of the type IVB-secreted effector SdhA. We here present the 3D structure of PlaA which shows a typical α/ß-hydrolase fold and reveals that the uncleaved disulfide loop forms a lid structure covering the catalytic triad S30/D278/H282. This leads to reduction of substrate access before activation; however, the catalytic site gets more accessible when the disulfide loop is processed. After structural modeling, a similar activation process is suggested for the GDSL hydrolase PlaC, but not for PlaD. Furthermore, the size of the PlaA substrate-binding site indicated preference toward phospholipids comprising ~16 carbon fatty acid residues which was verified by lipid hydrolysis, suggesting a molecular ruler mechanism. Indeed, mutational analysis changed the substrate profile with respect to fatty acid chain length. In conclusion, our analysis revealed the structural basis for the regulated activation and substrate preference of PlaA.


Assuntos
Legionella pneumophila , Lisofosfolipase , Lisofosfolipase/genética , Lisofosfolipase/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Proteínas de Bactérias/metabolismo , Dissulfetos/metabolismo , Vacúolos/metabolismo , Ácidos Graxos/metabolismo , Relação Estrutura-Atividade
2.
Mol Microbiol ; 121(3): 578-592, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38308564

RESUMO

Pathogenic Rhodococcus equi release the virulence-associated protein A (VapA) within macrophage phagosomes. VapA permeabilizes phagosome and lysosome membranes and reduces acidification of both compartments. Using biophysical techniques, we found that VapA interacts with model membranes in four steps: (i) binding, change of mechanical properties, (ii) formation of specific membrane domains, (iii) permeabilization within the domains, and (iv) pH-specific transformation of domains. Biosensor data revealed that VapA binds to membranes in one step at pH 6.5 and in two steps at pH 4.5 and decreases membrane fluidity. The integration of VapA into lipid monolayers was only significant at lateral pressures <20 mN m-1 indicating preferential incorporation into membrane regions with reduced integrity. Atomic force microscopy of lipid mono- and bilayers showed that VapA increased the surface heterogeneity of liquid disordered domains. Furthermore, VapA led to the formation of a new microstructured domain type and, at pH 4.5, to the formation of 5 nm high domains. VapA binding, its integration and lipid domain formation depended on lipid composition, pH, protein concentration and lateral membrane pressure. VapA-mediated permeabilization is clearly distinct from that caused by classical microbial pore formers and is a key contribution to the multiplication of Rhodococcus equi in phagosomes.


Assuntos
Rhodococcus equi , Proteína Estafilocócica A , Virulência , Proteína Estafilocócica A/metabolismo , Fatores de Virulência/metabolismo , Rhodococcus equi/metabolismo , Proteínas de Bactérias/metabolismo , Lipídeos
3.
PLoS Pathog ; 18(8): e1010771, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35960766

RESUMO

ESX type VII secretion systems are complex secretion machineries spanning across the mycobacterial membrane and play an important role in pathogenicity, nutrient uptake and conjugation. We previously reported the role of ESX-4 in modulating Mycobacterium abscessus intracellular survival. The loss of EccB4 was associated with limited secretion of two effector proteins belonging to the WXG-100 family, EsxU and EsxT, and encoded by the esx-4 locus. This prompted us to investigate the function of M. abscessus EsxU and EsxT in vitro and in vivo. Herein, we show that EsxU and EsxT are substrates of ESX-4 and form a stable 1:1 heterodimer that permeabilizes artificial membranes. While expression of esxU and esxT was up-regulated in M. abscessus-infected macrophages, their absence in an esxUT deletion mutant prevented phagosomal membrane disruption while maintaining M. abscessus in an unacidified phagosome. Unexpectedly, the esxUT deletion was associated with a hyper-virulent phenotype, characterised by increased bacterial loads and mortality in mouse and zebrafish infection models. Collectively, these results demonstrate that the presence of EsxU and EsxT dampens survival and persistence of M. abscessus during infection.


Assuntos
Mycobacterium abscessus , Mycobacterium marinum , Mycobacterium tuberculosis , Mycobacterium , Sistemas de Secreção Tipo VII , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Camundongos , Mycobacterium/genética , Mycobacterium abscessus/genética , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/genética , Sistemas de Secreção Tipo VII/genética , Sistemas de Secreção Tipo VII/metabolismo , Peixe-Zebra/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34183393

RESUMO

Antimicrobial peptides (AMPs) contribute to an effective protection against infections. The antibacterial function of AMPs depends on their interactions with microbial membranes and lipids, such as lipopolysaccharide (LPS; endotoxin). Hyperinflammation induced by endotoxin is a key factor in bacterial sepsis and many other human diseases. Here, we provide a comprehensive profile of peptide-mediated LPS neutralization by systematic analysis of the effects of a set of AMPs and the peptide antibiotic polymyxin B (PMB) on the physicochemistry of endotoxin, macrophage activation, and lethality in mice. Mechanistic studies revealed that the host defense peptide LL-32 and PMB each reduce LPS-mediated activation also via a direct interaction of the peptides with the host cell. As a biophysical basis, we demonstrate modifications of the structure of cholesterol-rich membrane domains and the association of glycosylphosphatidylinositol (GPI)-anchored proteins. Our discovery of a host cell-directed mechanism of immune control contributes an important aspect in the development and therapeutic use of AMPs.


Assuntos
Catelicidinas/farmacologia , Membrana Celular/metabolismo , Interações Hospedeiro-Patógeno , Lipopolissacarídeos/farmacologia , Testes de Neutralização , Polimixina B/farmacologia , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Transporte Biológico/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Colesterol/metabolismo , Feminino , Células HEK293 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Inflamação/patologia , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos
5.
PLoS Pathog ; 17(11): e1010052, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34788341

RESUMO

The methyltransferase FliB posttranslationally modifies surface-exposed ɛ-N-lysine residues of flagellin, the protomer of the flagellar filament in Salmonella enterica (S. enterica). Flagellin methylation, reported originally in 1959, was recently shown to enhance host cell adhesion and invasion by increasing the flagellar hydrophobicity. The role of FliB in this process, however, remained enigmatic. In this study, we investigated the properties and mechanisms of FliB from S. enterica in vivo and in vitro. We show that FliB is an S-adenosylmethionine (SAM) dependent methyltransferase, forming a membrane associated oligomer that modifies flagellin in the bacterial cytosol. Using X-band electron paramagnetic resonance (EPR) spectroscopy, zero-field 57Fe Mössbauer spectroscopy, methylation assays and chromatography coupled mass spectrometry (MS) analysis, we further found that FliB contains an oxygen sensitive [4Fe-4S] cluster that is essential for the methyl transfer reaction and might mediate a radical mechanism. Our data indicate that the [4Fe-4S] cluster is coordinated by a cysteine rich motif in FliB that is highly conserved among multiple genera of the Enterobacteriaceae family.


Assuntos
Proteínas de Bactérias/metabolismo , Flagelina/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Lisina/metabolismo , Metiltransferases/metabolismo , S-Adenosilmetionina/metabolismo , Salmonella typhi/enzimologia , Proteínas de Bactérias/genética , Flagelina/química , Proteínas Ferro-Enxofre/genética , Lisina/química , Metilação , Metiltransferases/genética
6.
Nature ; 532(7597): 64-8, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27027296

RESUMO

Cytolytic proteins and peptide toxins are classical virulence factors of several bacterial pathogens which disrupt epithelial barrier function, damage cells and activate or modulate host immune responses. Such toxins have not been identified previously in human pathogenic fungi. Here we identify the first, to our knowledge, fungal cytolytic peptide toxin in the opportunistic pathogen Candida albicans. This secreted toxin directly damages epithelial membranes, triggers a danger response signalling pathway and activates epithelial immunity. Membrane permeabilization is enhanced by a positive charge at the carboxy terminus of the peptide, which triggers an inward current concomitant with calcium influx. C. albicans strains lacking this toxin do not activate or damage epithelial cells and are avirulent in animal models of mucosal infection. We propose the name 'Candidalysin' for this cytolytic peptide toxin; a newly identified, critical molecular determinant of epithelial damage and host recognition of the clinically important fungus, C. albicans.


Assuntos
Candida albicans/metabolismo , Candida albicans/patogenicidade , Citotoxinas/metabolismo , Proteínas Fúngicas/toxicidade , Micotoxinas/toxicidade , Fatores de Virulência/metabolismo , Cálcio/metabolismo , Candida albicans/imunologia , Candidíase/metabolismo , Candidíase/microbiologia , Candidíase/patologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citotoxinas/genética , Citotoxinas/toxicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Mucosa/microbiologia , Mucosa/patologia , Micotoxinas/genética , Micotoxinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Virulência/efeitos dos fármacos , Fatores de Virulência/genética , Fatores de Virulência/toxicidade
7.
Int J Mol Sci ; 22(3)2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540553

RESUMO

The polypeptide Pep19-2.5 (Aspidasept®) has been described to act efficiently against infection-inducing bacteria by binding and neutralizing their most potent toxins, i.e., lipopolysaccharides (LPS) and lipoproteins/peptides (LP), independent of the resistance status of the bacteria. The mode of action was described to consist of a primary Coulomb/polar interaction of the N-terminal region of Pep19-2.5 with the polar region of the toxins followed by a hydrophobic interaction of the C-terminal region of the peptide with the apolar moiety of the toxins. However, clinical development of Aspidasept as an anti-sepsis drug requires an in-depth characterization of the interaction of the peptide with the constituents of the human immune system and with other therapeutically relevant compounds such as antibiotics and non-steroidal anti-inflammatory drugs (NSAIDs). In this contribution, relevant details of primary and secondary pharmacodynamics, off-site targets, and immunogenicity are presented, proving that Pep19-2.5 may be readily applied therapeutically against the deleterious effects of a severe bacterial infection.


Assuntos
Anti-Infecciosos/farmacologia , Anti-Inflamatórios/farmacologia , Endotoxemia/tratamento farmacológico , Inflamação , Peptídeos/farmacologia , Animais , Anti-Infecciosos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Endotoxemia/imunologia , Humanos , Lipopolissacarídeos , Camundongos , Peptídeos/uso terapêutico
8.
Cell Microbiol ; 21(1): e12958, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30251327

RESUMO

Professional phagocytic cells such as macrophages are a central part of innate immune defence. They ingest microorganisms into membrane-bound compartments (phagosomes), which acidify and eventually fuse with lysosomes, exposing their contents to a microbicidal environment. Gram-positive Rhodococcus equi can cause pneumonia in young foals and in immunocompromised humans. The possession of a virulence plasmid allows them to subvert host defence mechanisms and to multiply in macrophages. Here, we show that the plasmid-encoded and secreted virulence-associated protein A (VapA) participates in exclusion of the proton-pumping vacuolar-ATPase complex from phagosomes and causes membrane permeabilisation, thus contributing to a pH-neutral phagosome lumen. Using fluorescence and electron microscopy, we show that VapA is also transferred from phagosomes to lysosomes where it permeabilises the limiting membranes for small ions such as protons. This permeabilisation process is different from that of known membrane pore formers as revealed by experiments with artificial lipid bilayers. We demonstrate that, at 24 hr of infection, virulent R. equi is contained in a vacuole, which is enriched in lysosome material, yet possesses a pH of 7.2 whereas phagosomes containing a vapA deletion mutant have a pH of 5.8 and those with virulence plasmid-less sister strains have a pH of 5.2. Experimentally neutralising the macrophage endocytic system allows avirulent R. equi to multiply. This observation is mirrored in the fact that virulent and avirulent R. equi multiply well in extracts of purified lysosomes at pH 7.2 but not at pH 5.1. Together these data indicate that the major function of VapA is to generate a pH-neutral and hence growth-promoting intracellular niche. VapA represents a new type of Gram-positive virulence factor by trafficking from one subcellular compartment to another, affecting membrane permeability, excluding proton-pumping ATPase, and consequently disarming host defences.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Fagossomos/microbiologia , ATPases Translocadoras de Prótons/antagonistas & inibidores , Rhodococcus equi/crescimento & desenvolvimento , Rhodococcus equi/metabolismo , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Microscopia Eletrônica , Microscopia de Fluorescência , Virulência
9.
Biophys J ; 117(10): 1805-1819, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31676134

RESUMO

The activity of antimicrobial peptides (AMPs) has been investigated extensively using model membranes composed of phospholipids or lipopolysaccharides in aqueous environments. However, from a biophysical perspective, there is a large scientific interest regarding the direct interaction of membrane-active peptides with whole bacteria. Working with living bacteria limits the usability of experimental setups and the interpretation of the resulting data because of safety risks and the overlap of active and passive effects induced by AMPs. We killed or inactivated metabolic-active bacteria using γ-irradiation or sodium azide, respectively. Microscopy, flow cytometry, and SYTOX green assays showed that the cell envelope remained intact to a high degree at the minimal bactericidal dose. Furthermore, the tumor-necrosis-factor-α-inducing activity of the lipopolysaccharides and the chemical lipid composition was unchanged. Determining the binding capacity of AMPs to the bacterial cell envelope by calorimetry is difficult because of an overlapping of the binding heat and metabolic activities of the bacteria-induced by the AMPs. The inactivation of all active processes helps to decipher the complex thermodynamic information. From the isothermal titration calorimetry (ITC) results, we propose that the bacterial membrane potential (Δψ) is possibly an underestimated modulator of the AMP activity. The negative surface charge of the outer leaflet of the outer membrane of Gram-negative bacteria is already neutralized by peptide concentrations below the minimal inhibitory concentration. This proves that peptide aggregation on the bacterial membrane surface plays a decisive role in the degree of antimicrobial activity. This will not only enable many biophysical approaches for the investigation between bacteria and membrane-active peptides in the future but will also make it possible to compare biophysical parameters of active and inactive bacteria. This opens up new possibilities to better understand the active and passive interaction processes between AMPs and bacteria.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/efeitos da radiação , Raios gama , Viabilidade Microbiana/efeitos dos fármacos , Viabilidade Microbiana/efeitos da radiação , Adsorção , Bactérias/ultraestrutura , Fenômenos Biofísicos , Membrana Celular/efeitos dos fármacos , Membrana Celular/efeitos da radiação , Membrana Celular/ultraestrutura , Potenciais da Membrana/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Fosfolipídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Termodinâmica
10.
Biochemistry ; 58(19): 2447-2462, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31016971

RESUMO

Cathelicidins are a family of host defense antimicrobial peptides in mammalian species. Among them, LL-37 is the only peptide of this family found in humans. Although LL-37 has been intensively investigated in the past, the mode of exerting its bactericidal activity through the specific interactions with bacterial membranes remains elusive. In this work, we combined microbiological and computational approaches with a tool box of experimental biophysical techniques, including conventional and surface-enhanced infrared absorption spectroscopy as well as fluorescence spectroscopy to characterize the structural and dynamic properties of LL-37 and shorter variants adsorbed on POPC/POPG (9:1) lipid bilayers as mimics of bacterial membranes. First, microbiological assays demonstrate that, while LL-32 and, in a lesser degree, LL-37 show hemolysis and antimicrobial activity, LL-20 remains practically inactive. Second, by comparing experimental and computational data of LL-37 with LL-20, we explained the bactericidal activity of the active peptide core as a consequence of an increased flexibility of the peptide structure, leading to reactive dangling charged side chains. Third, permeabilization assays showed a concentration-dependent membrane disruption activity of LL-37 and LL-32: at high peptide concentrations, LL-32 shows higher activity than LL-37, while, at low peptide concentrations, both peptides show similar activities. Responsible for this behavior is the C-terminal VPRTES tail (Ct-VPRTES tail), which, according to atomistic simulations, is able to promote the insertion of the peptide in the membrane and plays an essential role in controlling ordered peptide oligomerization on the surface of the membrane.


Assuntos
Antibacterianos/metabolismo , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Catelicidinas/metabolismo , Catelicidinas/farmacologia , Bicamadas Lipídicas/metabolismo , Sequência de Aminoácidos , Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/química , Catelicidinas/química , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Lipossomos/metabolismo , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Conformação Proteica em alfa-Hélice , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Espectroscopia por Absorção de Raios X
11.
Adv Exp Med Biol ; 1117: 111-129, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30980356

RESUMO

Antimicrobial peptides (AMPs) are in the focus of scientific research since the 1990s. In most cases, the main aim was laid on the design of AMP to kill bacteria effectively, with particular emphasis on broadband action and independency on antibiotic resistance. However, so far no approved drug on the basis of AMP has entered the market.Our approach of constructing AMP, called synthetic anti-lipopolysaccharide peptides (SALPs), on the basis of inhibiting the inflammatory action of lipopolysaccharide (LPS, endotoxin) from Gram-negative bacteria was focused on the neutralization of the decisive toxins. These are, beside LPS from Gram-negative bacteria, the lipoproteins (LP) from Gram-positive origin. Although some of the SALPs have an antibacterial action, the most important property is the high-affinity binding to LPS and LP, whether as constituent of the bacteria or in free form which prevents the damaging inflammation, that could otherwise lead to life-threatening septic shock. Most importantly, the SALP may inhibit inflammation independently of the resistance status of the bacteria, and so far the repeated use of the peptides apparently does not cause resistance of the attacking pathogens.In this chapter, an overview is given over the variety of possible applications in the field of fighting against severe bacterial infections, from the use in systemic infection/inflammation up to various topical applications such as anti-biofilm action and severe skin and soft tissue infections.


Assuntos
Antibacterianos/química , Moléculas com Motivos Associados a Patógenos/antagonistas & inibidores , Peptídeos/química , Infecções Bacterianas/tratamento farmacológico , Endotoxinas , Bactérias Gram-Negativas , Humanos , Lipopolissacarídeos
12.
Ann Surg ; 268(2): 348-356, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28288070

RESUMO

OBJECTIVE: To evaluate (1) levels of the host-defense/antimicrobial peptide LL-37 in patients with trauma and hemorrhagic shock (HS) and (2) the effects of a synthetic host-defense peptide; Pep19-4LF on multiple organ failure (MOF) associated with HS. BACKGROUND: HS is a common cause of death in severely injured patients. There is no specific therapy that reduces HS-associated MOF. METHODS: (1) LL-37 was measured in 47 trauma/HS patients admitted to an urban major trauma center. (2) Male Wistar rats were submitted to HS (90 min, target mean arterial pressure: 27-32 mm Hg) or sham operation. Rats were treated with Pep19-4LF [66 (n = 8) or 333 µg/kg ·â€Šh (n = 8)] or vehicle (n = 12) for 4 hours following resuscitation. RESULTS: Plasma LL-37 was 12-fold higher in patients with trauma/HS compared to healthy volunteers. HS rats treated with Pep19-4LF (high dose) had a higher mean arterial pressure at the end of the 4-hour resuscitation period (79 ±â€Š4 vs 54 ±â€Š5 mm Hg) and less renal dysfunction, liver injury, and lung inflammation than HS rats treated with vehicle. Pep19-4LF enhanced (kidney/liver) the phosphorylation of (1) protein kinase B and (2) endothelial nitric oxide synthase. Pep19-4LF attenuated the HS-induced (1) translocation of p65 from cytosol to nucleus, (2) phosphorylation of IκB kinase on Ser, and (3) phosphorylation of IκBα on Ser resulting in inhibition of nuclear factor kappa B and formation of proinflammatory cytokines. Pep19-4LF prevented the release of tumor necrosis factor alpha caused by heparan sulfate in human mononuclear cells by binding to this damage-associated molecular pattern. CONCLUSIONS: Trauma-associated HS results in release of LL-37. The synthetic host-defense/antimicrobial peptide Pep19-4LF attenuates the organ injury/dysfunction associated with HS.


Assuntos
Anti-Infecciosos/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/sangue , Insuficiência de Múltiplos Órgãos/prevenção & controle , Peptídeos/uso terapêutico , Substâncias Protetoras/uso terapêutico , Choque Hemorrágico/tratamento farmacológico , Ferimentos e Lesões/complicações , Animais , Biomarcadores/sangue , Estudos de Casos e Controles , Terapia Combinada , Humanos , Masculino , Insuficiência de Múltiplos Órgãos/etiologia , Ratos , Ratos Wistar , Ressuscitação , Choque Hemorrágico/sangue , Choque Hemorrágico/complicações , Choque Hemorrágico/diagnóstico , Resultado do Tratamento , Catelicidinas
13.
Cell Microbiol ; 19(2)2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27450700

RESUMO

Streptococcus suis is an important meningitis-causing pathogen in pigs and humans. Neutrophil extracellular traps (NETs) have been identified as host defense mechanism against different pathogens. Here, NETs were detected in the cerebrospinal fluid (CSF) of S. suis-infected piglets despite the presence of active nucleases. To study NET-formation and NET-degradation after transmigration of S. suis and neutrophils through the choroid plexus epithelial cell barrier, a previously described model of the human blood-CSF barrier was used. NETs and respective entrapment of streptococci were recorded in the "CSF compartment" despite the presence of active nucleases. Comparative analysis of S. suis wildtype and different S. suis nuclease mutants did not reveal significant differences in NET-formation or bacterial survival. Interestingly, transcript expression of the human cathelicidin LL-37, a NET-stabilizing factor, increased after transmigration of neutrophils through the choroid plexus epithelial cell barrier. In good accordance, the porcine cathelicidin PR-39 was significantly increased in CSF of piglets with meningitis. Furthermore, we confirmed that PR-39 is associated with NETs in infected CSF and inhibits neutrophil DNA degradation by bacterial nucleases. In conclusion, neutrophils form NETs after breaching the infected choroid plexus epithelium, and those NETs may be protected by antimicrobial peptides against bacterial nucleases.


Assuntos
Líquido Cefalorraquidiano/imunologia , Líquido Cefalorraquidiano/microbiologia , Armadilhas Extracelulares/imunologia , Neutrófilos/imunologia , Infecções Estreptocócicas/veterinária , Streptococcus suis/imunologia , Doenças dos Suínos/patologia , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica , Catelicidinas/análise , Técnicas de Cultura de Células , Líquido Cefalorraquidiano/química , Líquido Cefalorraquidiano/citologia , Desoxirribonucleases/deficiência , Desoxirribonucleases/metabolismo , Viabilidade Microbiana , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/patologia , Suínos , Doenças dos Suínos/imunologia
14.
Biochim Biophys Acta ; 1858(5): 1034-43, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26851776

RESUMO

Mycobacteria can cause different severe health problems, including tuberculosis (TB). The treatment of TB with conventional antibiotics is successful, however, the number of multi-drug and extensively-drug resistant Mycobacterium tuberculosis strains increases. Moreover, many classical antimycobacterial antibiotics have severe side effects. Therefore, antimicrobial peptides (AMPs) seem to be good candidates for new therapeutic strategies. On the one hand AMPs can be used as a single drug or in combination with conventional antibiotics to directly kill mycobacteria, or on the other hand to act as immunstimulatory agents. This review summarizes the findings on the role of endogenous human AMPs being involved in TB, the antimycobacterial activity of various AMPs, and the molecular modes of action. Most active AMPs interact with the mycobacterial cell envelope and in particular with the mycomembrane and the plasma membrane. The mycomembrane is a very rigid membrane probably leading to a lower activity of the AMPs against mycobacteria as compared to other Gram-negative or Gram-positive bacteria. For some AMPs also other targets have been identified. Because of the complex environment of intracellular mycobacteria being trapped in the phagosome, within the macrophage, within the granuloma, within the lung, the external administration of AMPs in the latent phase of TB is a challenge. However, in the acute phase the AMPs can attack mycobacteria in a direct way. This article is part of a Special Issue entitled: Antimicrobial peptides edited by Karl Lohner and Kai Hilpert.


Assuntos
Adjuvantes Imunológicos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Antituberculosos/farmacologia , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Adjuvantes Imunológicos/biossíntese , Adjuvantes Imunológicos/síntese química , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/síntese química , Antituberculosos/síntese química , Antituberculosos/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Parede Celular/química , Parede Celular/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Testes de Sensibilidade Microbiana , Complexo Mycobacterium avium/química , Complexo Mycobacterium avium/efeitos dos fármacos , Complexo Mycobacterium avium/crescimento & desenvolvimento , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fagossomos/efeitos dos fármacos , Fagossomos/microbiologia , Tuberculose Pulmonar/tratamento farmacológico , Tuberculose Pulmonar/microbiologia
15.
J Struct Biol ; 194(1): 68-77, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26828112

RESUMO

Lipopolysaccharides (LPS) belong to the strongest immune-modulating compounds known in nature, and are often described as pathogen-associated molecular patterns (PAMPs). In particular, at higher concentrations they are responsible for sepsis and the septic shock syndrome associated with high lethality. Since most data are indicative that LPS aggregates are the bioactive units, their supramolecular structures are considered to be of outmost relevance for deciphering the molecular mechanisms of its bioactivity. So far, however, most of the data available addressing this issue, were published only for the lipid part (lipid A) and the core-oligosaccharide containing rough LPS, representing the bioactive unit. By contrast, it is well known that most of the LPS specimen identified in natural habitats contain the smooth-form (S-form) LPS, which carry additionally a high-molecular polysaccharide (O-chain). To fill this lacuna and going into a more natural system, here various wild-type (smooth form) LPS including also some LPS fractions were investigated by small-angle X-ray scattering with synchrotron radiation to analyze their aggregate structure. Furthermore, the influence of a recently designed synthetic anti-LPS peptide (SALP) Pep19-2.5 on the aggregate structure, on the binding thermodynamics, and on the cytokine-inducing activity of LPS were characterized, showing defined aggregate changes, high affinity binding and inhibition of cytokine secretion. The data obtained are suitable to refine our view on the preferences of LPS for non-lamellar structures, representing the highest bioactive forms which can be significantly influenced by the binding with neutralizing peptides such as Pep19-2.5.


Assuntos
Anticorpos Neutralizantes/química , Enterobacteriaceae/química , Lipopolissacarídeos/química , Peptídeos/química , Sequência de Aminoácidos , Anticorpos Neutralizantes/imunologia , Calorimetria/métodos , Células Cultivadas , Enterobacteriaceae/genética , Enterobacteriaceae/imunologia , Humanos , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Peptídeos/imunologia , Peptídeos/farmacologia , Ligação Proteica , Espalhamento a Baixo Ângulo , Espectroscopia de Infravermelho com Transformada de Fourier , Termodinâmica , Fator de Necrose Tumoral alfa/metabolismo , Difração de Raios X
16.
J Allergy Clin Immunol ; 136(5): 1295-301.e1-5, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26037551

RESUMO

BACKGROUND: Peanut is one of the most hazardous sources of food allergens. Unknown allergens are still hidden in the complex lipophilic matrix. These allergens need to be discovered to allow estimation of the allergenic risk for patients with peanut allergy and to further improve diagnostic measures. OBJECTIVE: We performed detection, isolation, and characterization of novel peanut allergens from lipophilic peanut extract. METHODS: Extraction of roasted peanuts were performed under defined extraction conditions and examined by means of 2-dimensional PAGE. Subsequently, chromatographic methods were adapted to isolate low-molecular-weight components. Proteins were studied by using SDS-PAGE and immunoblotting with sera from patients with peanut allergy. For allergen identification protein sequencing, homology search and mass spectrometry were applied. Functional characterization for allergenicity was performed by using the basophil activation assay and for antimicrobial activity by using inhibition assays of different bacteria and fungi. RESULTS: IgE-reactive proteins of 12, 11, and 10 kDa were first detected after chloroform/methanol extraction in the flow through of hydrophobic interaction chromatography. The proteins were able to activate basophils of patients with peanut allergy. N-terminal sequencing and homology search in the expressed sequence tag database identified the allergens as peanut defensins, which was confirmed by using mass spectrometry. On microbial cell cultures, the peanut defensins showed inhibitory effects on the mold strains of the genera Cladosporium and Alternaria but none on bacteria. CONCLUSIONS: We identified defensins as novel peanut allergens (Ara h 12 and Ara h 13) that react in particular with IgE of patients with severe peanut allergy. Their antimicrobial activity is solely antifungal.


Assuntos
Alérgenos/imunologia , Arachis/imunologia , Basófilos/imunologia , Defensinas/imunologia , Hipersensibilidade a Amendoim/imunologia , Extratos Vegetais/imunologia , Alérgenos/isolamento & purificação , Alternaria/efeitos dos fármacos , Antifúngicos/imunologia , Antifúngicos/isolamento & purificação , Antifúngicos/farmacologia , Defensinas/isolamento & purificação , Defensinas/farmacologia , Eletroforese em Gel de Poliacrilamida , Humanos , Immunoblotting , Imunoglobulina E/metabolismo , Espectrometria de Massas , Hipersensibilidade a Amendoim/diagnóstico , Extratos Vegetais/isolamento & purificação , Homologia de Sequência de Aminoácidos
17.
Traffic ; 14(3): 321-36, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23231467

RESUMO

Here we describe a novel approach for the isolation and biochemical characterization of pathogen-containing compartments from primary cells: We developed a lipid-based procedure to magnetically label the surface of bacteria and visualized the label by scanning and transmission electron microscopy (SEM, TEM). We performed infection experiments with magnetically labeled Mycobacterium avium, M. tuberculosis and Listeria monocytogenes and isolated magnetic bacteria-containing phagosomes using a strong magnetic field in a novel free-flow system. Magnetic labeling of M. tuberculosis did not affect the virulence characteristics of the bacteria during infection experiments addressing host cell activation, phagosome maturation delay and replication in macrophages in vitro. Biochemical analyses of the magnetic phagosome-containing fractions provided evidence of an enhanced presence of bacterial antigens and a differential distribution of proteins involved in the endocytic pathway over time as well as cytokine-dependent changes in the phagosomal protein composition. The newly developed method represents a useful approach to characterize and compare pathogen-containing compartments, in order to identify microbial and host cell targets for novel anti-infective strategies.


Assuntos
Imãs , Fagossomos/microbiologia , Coloração e Rotulagem/métodos , Humanos , Lipídeos/química , Listeria monocytogenes/isolamento & purificação , Macrófagos/microbiologia , Macrófagos/ultraestrutura , Imãs/química , Microscopia Eletrônica de Transmissão e Varredura , Microscopia de Fluorescência , Mycobacterium/isolamento & purificação , Fagossomos/ultraestrutura
18.
Biochim Biophys Acta ; 1838(10): 2739-44, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25016054

RESUMO

Antimicrobial peptides (AMPs) are important components of the innate immune system of animals, plants, fungi and bacteria and are recently under discussion as promising alternatives to conventional antibiotics. We have investigated two cecropin-like synthetic peptides, Gm1, which corresponds to the natural overall uncharged Galleria mellonella native peptide and ΔGm1, a modified overall positively charged Gm1 variant. We have analysed these peptides for their potential to inhibit the endotoxin-induced secretion of tumour necrosis factor-α (TNF-α) from human mononuclear cells. Furthermore, in a conventional microbiological assay, the ability of these peptides to inhibit the growth of the rough mutant bacteria Salmonella enterica Minnesota R60 and the polymyxin B-resistant Proteus mirabilis R45 was investigated and atomic force microscopy (AFM) measurements were performed to characterize the morphology of the bacteria treated by the two peptides. We have also studied their cytotoxic properties in a haemolysis assay to clarify potential toxic effects. Our data revealed for both peptides minor anti-inflammatory (anti-endotoxin) activity, but demonstrated antimicrobial activity with differences depending on the endotoxin composition of the respective bacteria. In accordance with the antimicrobial assay, AFM data revealed a stronger morphology change of the R45 bacteria than for the R60. Furthermore, Gm1 had a stronger effect on the bacteria than ΔGm1, leading to a different morphology regarding indentations and coalescing of bacterial structures. The findings verify the biophysical measurements with the peptides on model systems. Both peptides lack any haemolytic activity up to an amount of 100µg/ml, making them suitable as new anti-infective agents.


Assuntos
Antibacterianos , Peptídeos Catiônicos Antimicrobianos , Endotoxemia/tratamento farmacológico , Proteínas de Insetos , Leucócitos Mononucleares/metabolismo , Mariposas/química , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Células Cultivadas , Endotoxemia/induzido quimicamente , Endotoxemia/metabolismo , Endotoxemia/patologia , Feminino , Humanos , Proteínas de Insetos/química , Proteínas de Insetos/farmacologia , Leucócitos Mononucleares/patologia , Lipopolissacarídeos/toxicidade , Masculino , Fator de Necrose Tumoral alfa/metabolismo
19.
Biochim Biophys Acta ; 1838(10): 2728-38, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25017800

RESUMO

Natural occurring antimicrobial peptides (AMPs) are important components of the innate immune system of animals and plants. They are considered to be promising alternatives to conventional antibiotics. Here we present a comparative study of two synthetic peptides: Gm1, corresponding to the natural overall uncharged peptide from Galleria mellonella (Gm) and ΔGm1, a modified overall positively charged Gm1 variant. We have studied the interaction of the peptides with lipid membranes composed of different kinds of lipopolysaccharides (LPS) and dimyristoylphosphatidylglycerol (DMPG), in some cases also dimyristoylphosphatidylethanolamine (DMPE) as representative lipid components of Gram-negative bacterial membranes, by applying Fourier-transform infrared spectroscopy (FTIR), Förster resonance energy transfer spectroscopy (FRET), differential scanning calorimetry (DSC) and isothermal titration calorimetry (ITC). Gm1 generates a destabilizing effect on the gel to liquid crystalline phase transition of the acyl chains of the lipids, as deduced from a decrease in the phase transition temperature and enthalpy, suggesting a fluidization, whereas ΔGm1 led to the opposite behavior. Further, FTIR analysis of the functional groups of the lipids participating in the interaction with the peptides indicated a shift in the band position and intensity of the asymmetric PO2(-) stretching vibration originating from the lipid phosphate groups, a consequence of the sterical changes in the head group region. Interestingly, FRET spectroscopy showed a similar intercalation of both peptides into the DMPG and LPS, but much less into the DMPE membrane systems. These results are discussed in the light of a possible use of the peptides as antimicrobial and anti-endotoxin drugs.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Bactérias/química , Membrana Celular/química , Dimiristoilfosfatidilcolina/química , Proteínas de Insetos/química , Membranas Artificiais , Modelos Químicos , Animais , Anti-Infecciosos/química , Mariposas
20.
Nat Chem Biol ; 9(1): 37-42, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23143413

RESUMO

Human pathogens often produce soluble protein toxins that generate pores inside membranes, resulting in the death of target cells and tissue damage. In pathogenic amoebae, this has been exemplified with amoebapores of the enteric protozoan parasite Entamoeba histolytica. Here we characterize acanthaporin, to our knowledge the first pore-forming toxin to be described from acanthamoebae, which are free-living, bacteria-feeding, unicellular organisms that are opportunistic pathogens of increasing importance and cause severe and often fatal diseases. We isolated acanthaporin from extracts of virulent Acanthamoeba culbertsoni by tracking its pore-forming activity, molecularly cloned the gene of its precursor and recombinantly expressed the mature protein in bacteria. Acanthaporin was cytotoxic for human neuronal cells and exerted antimicrobial activity against a variety of bacterial strains by permeabilizing their membranes. The tertiary structures of acanthaporin's active monomeric form and inactive dimeric form, both solved by NMR spectroscopy, revealed a currently unknown protein fold and a pH-dependent trigger mechanism of activation.


Assuntos
Acanthamoeba/química , Proteínas de Protozoários/química , Proteínas de Protozoários/fisiologia , Acanthamoeba/patogenicidade , Sequência de Aminoácidos , Dimerização , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA