Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(10): e0269093, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36315510

RESUMO

Klebsiella pneumoniae is the causative agent of a variety of severe infections. Many K. pneumoniae strains are resistant to multiple antibiotics, and this situation creates a need for new antibacterial molecules. K. pneumoniae pathogenicity relies largely on its ability to escape phagocytosis and intracellular killing by phagocytic cells. Interfering with these escape mechanisms may allow to decrease bacterial virulence and to combat infections. In this study, we used Dictyostelium discoideum as a model phagocyte to screen a collection of 1,099 chemical compounds. Phg1A KO D. discoideum cells cannot feed upon K. pneumoniae bacteria, unless bacteria bear mutations decreasing their virulence. We identified 3 non-antibiotic compounds that restored growth of phg1A KO cells on K. pneumoniae, and we characterized the mode of action of one of them, 5-ethyl-2'-deoxyuridine (K2). K2-treated bacteria were more rapidly killed in D. discoideum phagosomes than non-treated bacteria. They were more sensitive to polymyxin and their outer membrane was more accessible to a hydrophobic fluorescent probe. These results suggest that K2 acts by rendering the membrane of K. pneumoniae accessible to antibacterial effectors. K2 was effective on three different K. pneumoniae strains, and acted at concentrations as low as 3 µM. K2 has previously been used to treat viral infections but its precise molecular mechanism of action in K. pneumoniae remains to be determined.


Assuntos
Dictyostelium , Infecções por Klebsiella , Humanos , Klebsiella pneumoniae/genética , Dictyostelium/microbiologia , Fagócitos , Antibacterianos , Infecções por Klebsiella/microbiologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-29552544

RESUMO

Environmental bacteria of the genus Legionella naturally parasitize free-living amoebae. Upon inhalation of bacteria-laden aerosols, the opportunistic pathogens grow intracellularly in alveolar macrophages and can cause a life-threatening pneumonia termed Legionnaires' disease. Intracellular replication in amoebae and macrophages takes place in a unique membrane-bound compartment, the Legionella-containing vacuole (LCV). LCV formation requires the bacterial Icm/Dot type IV secretion system, which translocates literally hundreds of "effector" proteins into host cells, where they modulate crucial cellular processes for the pathogen's benefit. The mechanism of LCV formation appears to be evolutionarily conserved, and therefore, amoebae are not only ecologically significant niches for Legionella spp., but also useful cellular models for eukaryotic phagocytes. In particular, Acanthamoeba castellanii and Dictyostelium discoideum emerged over the last years as versatile and powerful models. Using genetic, biochemical and cell biological approaches, molecular interactions between amoebae and Legionella pneumophila have recently been investigated in detail with a focus on the role of phosphoinositide lipids, small and large GTPases, autophagy components and the retromer complex, as well as on bacterial effectors targeting these host factors.


Assuntos
Acanthamoeba/microbiologia , Dictyostelium/microbiologia , Modelos Animais de Doenças , Legionella/metabolismo , Doença dos Legionários/microbiologia , Doença dos Legionários/veterinária , Acanthamoeba castellanii/microbiologia , Amoeba/microbiologia , Animais , Autofagia , Proteínas de Bactérias/metabolismo , Avaliação Pré-Clínica de Medicamentos , Evolução Molecular , GTP Fosfo-Hidrolases , Interações Hospedeiro-Patógeno/fisiologia , Legionella/patogenicidade , Legionella pneumophila/metabolismo , Macrófagos/microbiologia , Fosfatidilinositóis/metabolismo , Proteômica , Sistemas de Secreção Tipo IV/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia
3.
PLoS One ; 12(7): e0181121, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28727774

RESUMO

Tuberculosis remains one of the major threats to public health worldwide. Given the prevalence of multi drug resistance (MDR) in Mycobacterium tuberculosis strains, there is a strong need to develop new anti-mycobacterial drugs with modes of action distinct from classical antibiotics. Inhibitors of mycobacterial virulence might target new molecular processes and may represent a potential new therapeutic alternative. In this study, we used a Dictyostelium discoideum host model to assess virulence of Mycobacterium marinum and to identify compounds inhibiting mycobacterial virulence. Among 9995 chemical compounds, we selected 12 inhibitors of mycobacterial virulence that do not inhibit mycobacterial growth in synthetic medium. Further analyses revealed that 8 of them perturbed functions requiring an intact mycobacterial cell wall such as sliding motility, bacterial aggregation or cell wall permeability. Chemical analogs of two compounds were analyzed. Chemical modifications altered concomitantly their effect on sliding motility and on mycobacterial virulence, suggesting that the alteration of the mycobacterial cell wall caused the loss of virulence. We characterized further one of the selected compounds and found that it inhibited the ability of mycobacteria to replicate in infected cells. Together these results identify new antimycobacterial compounds that represent new tools to unravel the molecular mechanisms controlling mycobacterial pathogenicity. The isolation of compounds with anti-virulence activity is the first step towards developing new antibacterial treatments.


Assuntos
Dictyostelium/microbiologia , Mycobacterium marinum/efeitos dos fármacos , Virulência/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Mycobacterium marinum/patogenicidade , Mycobacterium marinum/fisiologia , Mycobacterium marinum/ultraestrutura , Bibliotecas de Moléculas Pequenas
4.
J Med Chem ; 59(24): 10917-10928, 2016 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-26730986

RESUMO

Here, we report on the design, synthesis, and biological evaluation of 4-thiazolidinone (rhodanine) derivatives targeting Mycobacterial tuberculosis (Mtb) trans-2-enoyl-acyl carrier protein reductase (InhA). Compounds having bulky aromatic substituents at position 5 and a tryptophan residue at position N-3 of the rhodanine ring were the most active against InhA, with IC50 values ranging from 2.7 to 30 µM. The experimental data showed consistent correlations with computational studies. Their antimicrobial activity was assessed against Mycobacterium marinum (Mm) (a model for Mtb), Pseudomonas aeruginosa (Pa), Legionella pneumophila (Lp), and Enterococcus faecalis (Ef) by using anti-infective, antivirulence, and antibiotic assays. Nineteen out of 34 compounds reduced Mm virulence at 10 µM. 33 exhibited promising antibiotic activity against Mm with a MIC of 0.21 µM and showed up to 89% reduction of Lp growth in an anti-infective assay at 30 µM. 32 showed high antibiotic activity against Ef, with a MIC of 0.57 µM.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Desenho de Fármacos , Oxirredutases/antagonistas & inibidores , Rodanina/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Relação Dose-Resposta a Droga , Enterococcus faecalis/efeitos dos fármacos , Legionella pneumophila/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Mycobacterium marinum/efeitos dos fármacos , Oxirredutases/isolamento & purificação , Oxirredutases/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Rodanina/síntese química , Rodanina/química , Relação Estrutura-Atividade
5.
Microbiology (Reading) ; 161(7): 1392-406, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25873585

RESUMO

Legionella pneumophila is a facultative intracellular bacterium, which upon inhalation can cause a potentially fatal pneumonia termed Legionnaires' disease. The opportunistic pathogen grows in environmental amoebae and mammalian macrophages within a unique membrane-bound compartment, the 'Legionella-containing vacuole'. Bacteria are exposed to many environmental cues including small signalling molecules from eukaryotic cells. A number of pathogenic bacteria sense and respond to catecholamine hormones, such as adrenalin and noradrenalin, a process mediated via the QseBC two-component system in some bacteria. In this study, we examined the effect of adrenergic compounds on L. pneumophila, and discovered that the adrenergic receptor antagonists benoxathian, naftopidil, propranolol and labetalol, as well as the QseC sensor kinase inhibitor LED209, reduced the growth of L. pneumophila in broth or amoebae, while replication in macrophages was enhanced. Growth restriction was common to members of the genus Legionella and Mycobacterium, and was observed for L. pneumophila in the replicative but not stationary phase of the biphasic life cycle. Deletion of the L. pneumophila qseBC genes indicated that growth inhibition by adrenergics or LED209 is mediated only to a minor extent by this two-component system, implying the presence of other adrenergic sensing systems. This study identifies adrenergic molecules as novel inhibitors of extra- and intracellular growth of Legionella and reveals LED209 as a potential lead compound to combat infections with Legionella or Mycobacterium spp.


Assuntos
Antagonistas Adrenérgicos/metabolismo , Antibacterianos/metabolismo , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/crescimento & desenvolvimento , Acanthamoeba castellanii/efeitos dos fármacos , Acanthamoeba castellanii/microbiologia , Animais , Linhagem Celular , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Mycobacterium/efeitos dos fármacos , Mycobacterium/crescimento & desenvolvimento
6.
ACS Infect Dis ; 1(7): 327-38, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-27622823

RESUMO

The causative agent of Legionnaires' disease, Legionella pneumophila, grows in environmental amoebae and mammalian macrophages within a distinct compartment, the 'Legionella-containing vacuole' (LCV). Intracellular bacteria are protected from many antibiotics, and thus are notoriously difficult to eradicate. To identify novel compounds that restrict intracellular bacterial replication, we previously developed an assay based on a coculture of amoebae and GFP-producing L. pneumophila. This assay was used to screen a pathway-based, highly diverse chemical library, referred to as the Sinergia library. In this work, we chose to focus on a group of 11 hit compounds, the majority of which originated from the query molecule CN585, a compound that targets the protein phosphatase calcineurin. Further studies on 78 related compound variants revealed crucial structural attributes, namely a triple-ring scaffold with a central triazine moiety, substituted in positions 3 and 5 by two piperidine or pyrrolidine rings, and in position 1 by an amine group bearing a single aliphatic chain moiety. The most effective compound, ZINC00615682, inhibited intracellular replication of L. pneumophila with an IC50 of approximately 20 nM in Acanthamoeba castellanii and slightly less efficiently in Dictyostelium discoideum or macrophages. Pharmacological and genetic attempts to implicate calcineurin in the intracellular replication of L. pneumophila failed. Taken together, these results show that the amoebae-based screen and structure-activity relationship analysis is suitable for the identification of novel inhibitors of the intracellular replication of L. pneumophila. The most potent compound identified in this study targets (an) as yet unidentified host factor(s).

7.
Small GTPases ; 5(3): 1-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25496424

RESUMO

Legionella spp. are amoebae-resistant environmental bacteria that replicate in free-living protozoa in a distinct compartment, the Legionella-containing vacuole (LCV). Upon transmission of Legionella pneumophila to the lung, the pathogens employ an evolutionarily conserved mechanism to grow in LCVs within alveolar macrophages, thus triggering a severe pneumonia termed Legionnaires' disease. LCV formation is a complex and robust process, which requires the bacterial Icm/Dot type IV secretion system and involves the amazing number of 300 different translocated effector proteins. LCVs interact with the host cell's endosomal and secretory vesicle trafficking pathway. Accordingly, in a proteomics approach as many as 12 small Rab GTPases implicated in endosomal and secretory vesicle trafficking were identified and validated as LCV components. Moreover, the small GTPase Ran and its effector protein RanBP1 have been found to decorate the pathogen vacuole. Ran regulates nucleo-cytoplasmic transport, spindle assembly, and cytokinesis, as well as the organization of non-centrosomal microtubules. In L. pneumophila-infected amoebae or macrophages, Ran and RanBP1 localize to LCVs, and the small GTPase is activated by the Icm/Dot substrate LegG1. Ran activation by LegG1 leads to microtubule stabilization and promotes intracellular pathogen vacuole motility and bacterial growth, as well as chemotaxis and migration of Legionella-infected cells.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/fisiologia , Vacúolos/fisiologia , Proteína ran de Ligação ao GTP/metabolismo , Amoeba/microbiologia , Interações Hospedeiro-Patógeno , Legionella pneumophila/enzimologia , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Microtúbulos/metabolismo , Vacúolos/microbiologia
8.
Cell Microbiol ; 16(1): 15-26, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24168696

RESUMO

The severe pneumonia known as Legionnaires' disease occurs following infection by the Gram-negative bacterium Legionella pneumophila. Normally resident in fresh-water sources, Legionella are subject to predation by eukaryotic phagocytes such as amoeba and ciliates. To counter this, L. pneumophila has evolved a complex system of effector proteins which allow the bacteria to hijack the phagocytic vacuole, hiding and replicating within their erstwhile killers. These same mechanisms allow L. pneumophila to hijack another phagocyte, lung-based macrophages, which thus avoids a vital part of the immune system and leads to infection. The course of infection can be divided into five main categories: pathogen uptake, formation of the replication-permissive vacuole, intracellular replication, host cell response, and bacterial exit. L. pneumophila effector proteins target every stage of this process, interacting with secretory, endosomal, lysosomal, retrograde and autophagy pathways, as well as with mitochondria. Each of these steps can be studied in protozoa or mammalian cells, and the knowledge gained can be readily applied to human pathogenicity. Here we describe the manner whereby L. pneumophila infects host protozoa, the various techniques which are available to analyse these processes and the implications of this model for Legionella virulence and the pathogenesis of Legionnaires' disease.


Assuntos
Alveolados , Células Eucarióticas/microbiologia , Interações Hospedeiro-Patógeno , Legionella pneumophila/fisiologia , Doença dos Legionários , Animais , Humanos , Modelos Teóricos
9.
J Virol ; 88(5): 2690-703, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24352465

RESUMO

UNLABELLED: Prion diseases are a group of fatal and incurable neurodegenerative diseases affecting both humans and animals. The principal mechanism of these diseases involves the misfolding the host-encoded cellular prion protein, PrP(C), into the disease-associated isoform, PrP(Sc). Familial forms of human prion disease include those associated with the mutations G114V and A117V, which lie in the hydrophobic domain of PrP. Here we have studied the murine homologues (G113V and A116V) of these mutations using cell-based and animal models of prion infection. Under normal circumstances, the mutant forms of PrP(C) share similar processing, cellular localization, and physicochemical properties with wild-type mouse PrP (MoPrP). However, upon exposure of susceptible cell lines expressing these mutants to infectious prions, very low levels of protease-resistant aggregated PrP(Sc) are formed. Subsequent mouse bioassay revealed high levels of infectivity present in these cells. Thus, these mutations appear to limit the formation of aggregated PrP(Sc), giving rise to the accumulation of a relatively soluble, protease sensitive, prion species that is highly neurotoxic. Given that these mutations lie next to the glycine-rich region of PrP that can abrogate prion infection, these findings provide further support for small, protease-sensitive prion species having a significant role in the progression of prion disease and that the hydrophobic domain is an important determinant of PrP conversion. IMPORTANCE: Prion diseases are transmissible neurodegenerative diseases associated with an infectious agent called a prion. Prions are comprised of an abnormally folded form of the prion protein (PrP) that is normally resistant to enzymes called proteases. In humans, prion disease can occur in individuals who inherited mutations in the prion protein gene. Here we have studied the effects of two of these mutations and show that they influence the properties of the prions that can be formed. We show that the mutants make highly infectious prions that are more sensitive to protease treatment. This study highlights a certain region of the prion protein as being involved in this effect and demonstrates that prions are not always resistant to protease treatment.


Assuntos
Mutação , Príons/genética , Príons/metabolismo , Domínios e Motivos de Interação entre Proteínas , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Códon , Expressão Gênica , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Dados de Sequência Molecular , Peptídeo Hidrolases/metabolismo , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Doenças Priônicas/genética , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Príons/química , Proteólise , Alinhamento de Sequência
10.
PLoS One ; 8(9): e74813, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058631

RESUMO

Legionella pneumophila is a ubiquitous fresh-water bacterium which reproduces within its erstwhile predators, environmental amoeba, by subverting the normal pathway of phagocytosis and degradation. The molecular mechanisms which confer resistance to amoeba are apparently conserved and also allow replication within macrophages. Thus, L. pneumophila can act as an 'accidental' human pathogen and cause a severe pneumonia known as Legionnaires' disease. The intracellular localisation of L. pneumophila protects it from some antibiotics, and this fact must be taken into account to develop new anti-bacterial compounds. In addition, the intracellular lifestyle of L. pneumophila may render the bacteria susceptible to compounds diminishing bacterial virulence and decreasing intracellular survival and replication of this pathogen. The development of a single infection cycle intracellular replication assay using GFP-producing L. pneumophila and Acanthamoebacastellanii amoeba is reported here. This fluorescence-based assay allows for continuous monitoring of intracellular replication rates, revealing the effect of bacterial gene deletions or drug treatment. To examine how perturbations of the host cell affect L. pneumophila replication, several known host-targeting compounds were tested, including modulators of cytoskeletal dynamics, vesicle scission and Ras GTPase localisation. Our results reveal a hitherto unrealized potential antibiotic property of the ß-lactone-based Ras depalmitoylation inhibitor palmostatin M, but not the closely related inhibitor palmostatin B. Further characterisation indicated that this compound caused specific growth inhibition of Legionella and Mycobacterium species, suggesting that it may act on a common bacterial target.


Assuntos
Antibacterianos/farmacologia , Espaço Intracelular/microbiologia , Legionella pneumophila/efeitos dos fármacos , Animais , Linhagem Celular , Proteínas de Fluorescência Verde/metabolismo , Humanos , Concentração Inibidora 50 , Espaço Intracelular/efeitos dos fármacos , Legionella pneumophila/crescimento & desenvolvimento , Camundongos , Testes de Sensibilidade Microbiana , Peso Molecular , Mycobacterium/efeitos dos fármacos , Mycobacterium/crescimento & desenvolvimento , Propiolactona/análogos & derivados , Propiolactona/farmacologia , Especificidade da Espécie , Sulfonas/farmacologia
11.
PLoS Pathog ; 9(9): e1003598, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068924

RESUMO

The causative agent of Legionnaires' disease, Legionella pneumophila, uses the Icm/Dot type IV secretion system (T4SS) to form in phagocytes a distinct "Legionella-containing vacuole" (LCV), which intercepts endosomal and secretory vesicle trafficking. Proteomics revealed the presence of the small GTPase Ran and its effector RanBP1 on purified LCVs. Here we validate that Ran and RanBP1 localize to LCVs and promote intracellular growth of L. pneumophila. Moreover, the L. pneumophila protein LegG1, which contains putative RCC1 Ran guanine nucleotide exchange factor (GEF) domains, accumulates on LCVs in an Icm/Dot-dependent manner. L. pneumophila wild-type bacteria, but not strains lacking LegG1 or a functional Icm/Dot T4SS, activate Ran on LCVs, while purified LegG1 produces active Ran(GTP) in cell lysates. L. pneumophila lacking legG1 is compromised for intracellular growth in macrophages and amoebae, yet is as cytotoxic as the wild-type strain. A downstream effect of LegG1 is to stabilize microtubules, as revealed by conventional and stimulated emission depletion (STED) fluorescence microscopy, subcellular fractionation and Western blot, or by microbial microinjection through the T3SS of a Yersinia strain lacking endogenous effectors. Real-time fluorescence imaging indicates that LCVs harboring wild-type L. pneumophila rapidly move along microtubules, while LCVs harboring ΔlegG1 mutant bacteria are stalled. Together, our results demonstrate that Ran activation and RanBP1 promote LCV formation, and the Icm/Dot substrate LegG1 functions as a bacterial Ran activator, which localizes to LCVs and promotes microtubule stabilization, LCV motility as well as intracellular replication of L. pneumophila.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Legionella pneumophila/fisiologia , Macrófagos/microbiologia , Microtúbulos/metabolismo , Fagossomos/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Ativação Enzimática , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/genética , Inativação Gênica , Humanos , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Legionella pneumophila/ultraestrutura , Doença dos Legionários/imunologia , Doença dos Legionários/metabolismo , Doença dos Legionários/microbiologia , Doença dos Legionários/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Camundongos , Proteínas dos Microtúbulos/química , Proteínas dos Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Mutação , Fagocitose , Fagossomos/enzimologia , Fagossomos/ultraestrutura , Polimerização , Estabilidade Proteica , Transporte Proteico , Replicação Viral , Proteína ran de Ligação ao GTP/antagonistas & inibidores , Proteína ran de Ligação ao GTP/genética
12.
Cell Host Microbe ; 14(1): 38-50, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23870312

RESUMO

The bacteria causing Legionnaires' disease, Legionella pneumophila, replicate intracellularly within unique Legionella-containing vacuoles (LCVs). LCV formation involves a type IV secretion system (T4SS) that translocates effector proteins into host cells. We show that the T4SS effector RidL localizes to LCVs, supports intracellular bacterial growth, and alters retrograde trafficking, in which selected proteins are transported from endosomes to the Golgi. The retromer complex that mediates retrograde trafficking localizes to LCVs independently of RidL and restricts intracellular bacterial growth. RidL binds the Vps29 retromer subunit and the lipid PtdIns(3)P, which localizes retromer components to membranes. Additionally, specific retromer cargo receptors and sorting nexins that mediate protein capture and membrane remodeling preferentially localize to LCVs in the absence of ridL. Ectopic RidL production inhibits retrograde trafficking, and L. pneumophila blocks retrograde transport at endosome exit sites in a ridL-dependent manner. Collectively, these findings suggest that RidL inhibits retromer function to promote intracellular bacterial replication.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/metabolismo , Doença dos Legionários/microbiologia , Vacúolos/microbiologia , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Endossomos/metabolismo , Humanos , Legionella pneumophila/genética , Camundongos , Transporte Proteico , Vacúolos/metabolismo
13.
Environ Microbiol Rep ; 3(3): 286-96, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23761274

RESUMO

Bacteria of the genus Legionella persist in a wide range of environmental habitats, including biofilms, protozoa and nematodes. Legionellaceae are 'accidental' human pathogens that upon inhalation cause a severe pneumonia termed 'Legionnaires' disease'. The interactions of L. pneumophila with eukaryotic hosts are governed by the Icm/Dot type IV secretion system (T4SS) and more than 150 'effector proteins', which subvert signal transduction pathways and promote the formation of the replication-permissive 'Legionella-containing vacuole'. The Icm/Dot T4SS is essential to infect free-living protozoa, such as the amoeba Dictyostelium discoideum, as well as the nematode Caenorhabditis elegans, or mammalian macrophages. To adapt to different niches, L. pneumophila not only responds to exogenous cues, but also to endogenous signals, such as the α-hydroxyketone compound LAI-1 (Legionella autoinducer-1). The long-term adaptation of Legionella spp. is based on extensive horizontal DNA transfer. In fact, Legionella spp. have acquired canonical 'genomic islands' of prokaryotic origin, but also a number of eukaryotic genes. Since many aspects of Legionella virulence against environmental predators and immune phagocytes are similar, an understanding of Legionella ecology provides valuable insights into the pathogenesis of legionellaceae for humans.

14.
J Virol ; 84(13): 6678-86, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20427543

RESUMO

Prion diseases are a group of transmissible, invariably fatal neurodegenerative diseases that affect both humans and animals. According to the protein-only hypothesis, the infectious agent is a prion (proteinaceous infectious particle) that is composed primarily of PrP(Sc), the disease-associated isoform of the cellular prion protein, PrP. PrP(Sc) arises from the conformational change of the normal, glycosylphosphatidylinositol (GPI)-anchored protein, PrP(C). The mechanism by which this process occurs, however, remains enigmatic. Rabbits are one of a small number of mammalian species reported to be resistant to prion infection. Sequence analysis of rabbit PrP revealed that its C-terminal amino acids differ from those of PrP from other mammals and may affect the anchoring of rabbit PrP through its GPI anchor. Using a cell culture model, this study investigated the effect of the rabbit PrP-specific C-terminal amino acids on the addition of the GPI anchor to PrP(C), PrP(C) localization, and PrP(Sc) formation. The incorporation of rabbit-specific C-terminal PrP residues into mouse PrP did not affect the addition of a GPI anchor or the localization of PrP. However, these residues did inhibit PrP(Sc) formation, suggesting that these rabbit-specific residues interfere with a C-terminal PrP(Sc) interaction site.


Assuntos
Glicosilfosfatidilinositóis/metabolismo , Imunidade Inata , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Doenças Priônicas , Sequência de Aminoácidos , Animais , Sítios de Ligação , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Coelhos , Alinhamento de Sequência
15.
J Biol Chem ; 285(26): 20213-23, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20356832

RESUMO

Prion diseases are associated with the misfolding of the endogenously expressed prion protein (designated PrP(C)) into an abnormal isoform (PrP(Sc)) that has infectious properties. The hydrophobic domain of PrP(C) is highly conserved and contains a series of glycine residues that show perfect conservation among all species, strongly suggesting it has functional and evolutionary significance. These glycine residues appear to form repeats of the GXXXG protein-protein interaction motif (two glycines separated by any three residues); the retention of these residues is significant and presumably relates to the functionality of PrP(C). Mutagenesis studies demonstrate that minor alterations to this highly conserved region of PrP(C) drastically affect the ability of cells to uptake and replicate prion infection in both cell and animal bioassay. The localization and processing of mutant PrP(C) are not affected, although in vitro and in vivo studies demonstrate that this region is not essential for interaction with PrP(Sc), suggesting these residues provide conformational flexibility. These data suggest that this region of PrP(C) is critical in the misfolding process and could serve as a novel, species-independent target for prion disease therapeutics.


Assuntos
Motivos de Aminoácidos , Glicina/genética , Proteínas PrPC/genética , Proteínas PrPSc/genética , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Glicina/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Microdomínios da Membrana/metabolismo , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/metabolismo , Ligação Proteica , Dobramento de Proteína , Homologia de Sequência de Aminoácidos , Transfecção
16.
Biochem J ; 428(2): 169-82, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20337594

RESUMO

Prion diseases are fatal transmissible neurodegenerative diseases that result from structural conversion of the prion protein into a disease-associated isoform. The prion protein contains a single disulfide bond. Our analysis of all NMR structures of the prion protein (total of 440 structures over nine species) containing an explicit disulfide bond reveals that the bond exists predominantly in a stable low-energy state, but can also adopt a high-energy configuration. The side chains of two tyrosine residues and one phenylalanine residue control access of solvent to the disulfide bond. Notably, the side chains rotate away from the disulfide bond in the high-energy state, exposing the disulfide bond to solvent. The importance of these aromatic residues for protein function was analysed by mutating them to alanine residues and analysing the properties of the mutant proteins using biophysical and cell biological approaches. Whereas the mutant protein behaved similarly to wild-type prion protein in recombinant systems, the mutants were retained in the endoplasmic reticulum of mammalian cells and degraded by the proteasomal system. The cellular behaviour of the aromatic residue mutants was similar to the cellular behaviour of a disulfide bond mutant prion protein in which the cysteine residues were replaced with alanine, a result which is consistent with an unstable disulfide bond in the aromatic residue mutants. These observations suggest that the conformation of the prion protein disulfide bond may have implications for correct maturation and function of this protein.


Assuntos
Transporte Biológico/fisiologia , Dissulfetos/química , Príons/química , Príons/metabolismo , Solventes/química , Animais , Transporte Biológico/genética , Bovinos , Linhagem Celular , Cricetinae , Eletroforese em Gel de Poliacrilamida , Humanos , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Príons/genética , Conformação Proteica , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Suínos
17.
J Neurochem ; 103(5): 1709-20, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17944867

RESUMO

Prion diseases such as bovine spongiform encephalopathy in cattle and Creutzfeldt-Jakob disease in humans are associated with the misfolding and accumulation of an abnormal conformation of the host-encoded prion protein (PrP). Despite intensive research efforts conducted on PrP, the toxic agent involved in neurodegeneration is as yet unidentified. Several potential candidates have been proposed, each of which may be relevant to subsets of the broad array of prion diseases. In this study, we review current knowledge on neurotoxic PrP species, including the importance of a central hydrophobic domain for mediating neurotoxicty.


Assuntos
Neurotoxinas , Proteínas PrPC/patogenicidade , Proteínas PrPSc/patogenicidade , Doenças Priônicas/etiologia , Animais , Modelos Animais de Doenças , Humanos , Modelos Biológicos , Proteínas PrPC/genética , Proteínas PrPSc/genética , Doenças Priônicas/metabolismo , Isoformas de Proteínas/fisiologia
18.
FEBS J ; 272(23): 6120-30, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16302975

RESUMO

SOCS3 is a negative regulator of cytokine signalling that inhibits Janus kinase-signal transduction and activator of transcription (JAK-STAT) mediated signal tranduction by binding to phosphorylated tyrosine residues on intracellular subunits of various cytokine receptors, as well as possibly the JAK proteins. SOCS3 consists of a short N-terminal sequence followed by a kinase inhibitory region, an extended SH2 domain and a C-terminal suppressor of cytokine signalling (SOCS) box. SOCS3 and the related protein, cytokine-inducible SH2-containing protein, are unique among the SOCS family of proteins in containing a region of mostly low complexity sequence, between the SH2 domain and the C-terminal SOCS box. Using NMR, we assigned and determined the secondary structure of a murine SOCS3 construct. The SH2 domain, unusually, consists of 140 residues, including an unstructured insertion of 35 residues. This insertion fits the criteria for a PEST sequence and is not required for phosphotyrosine binding, as shown by isothermal titration calorimetry. Instead, we propose that the PEST sequence has a functional role unrelated to phosphotyrosine binding, possibly mediating efficient proteolytic degradation of the protein. The latter half of the kinase inhibitory region and the entire extended SH2 subdomain form a single alpha-helix. The mapping of the true SH2 domain, and the location of its C terminus more than 50 residues further downstream than predicted by sequence homology, explains a number of previously unexpected results that have shown the importance of residues close to the SOCS box for phosphotyrosine binding.


Assuntos
Estrutura Secundária de Proteína , Proteínas Supressoras da Sinalização de Citocina/química , Domínios de Homologia de src , Sequência de Aminoácidos , Animais , Clonagem Molecular , Humanos , Camundongos , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Fosfotirosina/metabolismo , Alinhamento de Sequência , Transdução de Sinais/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...