Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 545
Filtrar
1.
Nat Rev Microbiol ; 20(3): 161-173, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34548639

RESUMO

Bacteria have developed a large array of motility mechanisms to exploit available resources and environments. These mechanisms can be broadly classified into swimming in aqueous media and movement over solid surfaces. Swimming motility involves either the rotation of rigid helical filaments through the external medium or gyration of the cell body in response to the rotation of internal filaments. On surfaces, bacteria swarm collectively in a thin layer of fluid powered by the rotation of rigid helical filaments, they twitch by assembling and disassembling type IV pili, they glide by driving adhesins along tracks fixed to the cell surface and, finally, non-motile cells slide over surfaces in response to outward forces due to colony growth. Recent technological advances, especially in cryo-electron microscopy, have greatly improved our knowledge of the molecular machinery that powers the various forms of bacterial motility. In this Review, we describe the current understanding of the physical and molecular mechanisms that allow bacteria to move around.


Assuntos
Fenômenos Fisiológicos Bacterianos , Movimento/fisiologia , Adesinas Bacterianas/fisiologia , Animais , Bactérias , Microscopia Crioeletrônica/métodos , Fímbrias Bacterianas/fisiologia
2.
PLoS Biol ; 18(12): e3000986, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33378358

RESUMO

Clustering of the enteropathogenic Escherichia coli (EPEC) type III secretion system (T3SS) effector translocated intimin receptor (Tir) by intimin leads to actin polymerisation and pyroptotic cell death in macrophages. The effect of Tir clustering on the viability of EPEC-infected intestinal epithelial cells (IECs) is unknown. We show that EPEC induces pyroptosis in IECs in a Tir-dependent but actin polymerisation-independent manner, which was enhanced by priming with interferon gamma (IFNγ). Mechanistically, Tir clustering triggers rapid Ca2+ influx, which induces lipopolysaccharide (LPS) internalisation, followed by activation of caspase-4 and pyroptosis. Knockdown of caspase-4 or gasdermin D (GSDMD), translocation of NleF, which blocks caspase-4 or chelation of extracellular Ca2+, inhibited EPEC-induced cell death. IEC lines with low endogenous abundance of GSDMD were resistant to Tir-induced cell death. Conversely, ATP-induced extracellular Ca2+ influx enhanced cell death, which confirmed the key regulatory role of Ca2+ in EPEC-induced pyroptosis. We reveal a novel mechanism through which infection with an extracellular pathogen leads to pyroptosis in IECs.


Assuntos
Cálcio/metabolismo , Proteínas de Escherichia coli/metabolismo , Piroptose/fisiologia , Receptores de Superfície Celular/metabolismo , Actinas/metabolismo , Adesinas Bacterianas/metabolismo , Adesinas Bacterianas/fisiologia , Análise por Conglomerados , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/metabolismo , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Células HeLa , Humanos , Mucosa Intestinal/metabolismo , Intestinos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Transporte Proteico , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/fisiologia , Sistemas de Secreção Tipo III/metabolismo
3.
J Bacteriol ; 202(18)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32631946

RESUMO

Mechanisms by which cells attach to a surface and form a biofilm are diverse and differ greatly among organisms. The Gram-negative gammaproteobacterium Pseudomonas fluorescens attaches to a surface through the localization of the large type 1-secreted RTX adhesin LapA to the outer surface of the cell. LapA localization to the cell surface is controlled by the activities of a periplasmic protease, LapG, and an inner membrane-spanning cyclic di-GMP-responsive effector protein, LapD. A previous study identified a second, LapA-like protein encoded in the P. fluorescens Pf0-1 genome: Pfl01_1463. Here, we identified specific growth conditions under which Pfl01_1463, here called MapA (medium adhesion protein A) is a functional adhesin contributing to biofilm formation. This adhesin, like LapA, appears to be secreted through a Lap-related type 1 secretion machinery, and its localization is controlled by LapD and LapG. However, differing roles of LapA and MapA in biofilm formation are achieved, at least in part, through the differences in the sequences of the two adhesins and different distributions of the expression of the lapA and mapA genes within a biofilm. LapA-like proteins are broadly distributed throughout the Proteobacteria, and furthermore, LapA and MapA are well conserved among other Pseudomonas species. Together, our data indicate that the mechanisms by which a cell forms a biofilm and the components of a biofilm matrix can differ depending on growth conditions and the matrix protein(s) expressed.IMPORTANCE Adhesins are critical for the formation and maturation of bacterial biofilms. We identify a second adhesin in P. fluorescens, called MapA, which appears to play a role in biofilm maturation and whose regulation is distinct from the previously reported LapA adhesin, which is critical for biofilm initiation. Analysis of bacterial adhesins shows that LapA-like and MapA-like adhesins are found broadly in pseudomonads and related organisms, indicating that the utilization of different suites of adhesins may be broadly important in the Gammaproteobacteria.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Pseudomonas fluorescens/fisiologia , Membrana Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Periplasma/metabolismo
4.
Sci Rep ; 10(1): 9752, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546842

RESUMO

The two lectins LecA from Pseudomonas aeruginosa and the B-subunit of Shiga toxin from Shigella dysenteriae (StxB) share the glycosphingolipid globotriaosylceramide (Gb3) as receptor. Counterintuitively, we found that LecA and StxB segregated into different domains after recognizing Gb3 at the plasma membrane of cells. We hypothesized that the orientation of the carbohydrate head group of Gb3 embedded in the lipid bilayer differentially influences LecA and StxB binding. To test this hypothesis, we reconstituted lectin-Gb3 interaction using giant unilamellar vesicles and were indeed able to rebuild LecA and StxB segregation. Both, the Gb3 fatty acyl chain structure and the local membrane environment, modulated Gb3 recognition by LecA and StxB. Specifically, StxB preferred more ordered membranes compared to LecA. Based on our findings, we propose comparing staining patterns of LecA and StxB as an alternative method to assess membrane order in cells. To verify this approach, we re-established that the apical plasma membrane of epithelial cells is more ordered than the basolateral plasma membrane. Additionally, we found that StxB recognized Gb3 at the primary cilium and the periciliary membrane, whereas LecA only bound periciliary Gb3. This suggests that the ciliary membrane is of higher order than the surrounding periciliary membrane.


Assuntos
Adesinas Bacterianas/metabolismo , Ligação Proteica/fisiologia , Toxinas Shiga/metabolismo , Adesinas Bacterianas/fisiologia , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Glicoesfingolipídeos/metabolismo , Lectinas/metabolismo , Lectinas/fisiologia , Ligantes , Bicamadas Lipídicas/química , Ligação Proteica/genética , Pseudomonas aeruginosa , Toxina Shiga/metabolismo , Shigella dysenteriae , Triexosilceramidas/metabolismo , Lipossomas Unilamelares/metabolismo
5.
Curr Opin Microbiol ; 54: 37-42, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32035372

RESUMO

Kingella kingae is a gram-negative coccobacillus that is a fastidious commensal organism in the oropharynx and is being recognized increasingly as a common cause of osteoarticular infections and other invasive diseases in young children. The pathogenesis of K. kingae disease begins with bacterial adherence to respiratory epithelium, followed by translocation across the epithelial barrier, survival in the bloodstream, and dissemination to distant sites, including bones, joints, and the endocardium, among others. Characterization of the determinants of K. kingae pathogenicity has revealed a novel model of adherence that involves the interplay of type IV pili, a non-pilus adhesin, and a polysaccharide capsule and a novel model of resistance to serum killing and neutrophil killing that involves complementary functions of a polysaccharide capsule and an exopolysaccharide. These models likely apply to other bacterial pathogens as well.


Assuntos
Kingella kingae/patogenicidade , Infecções por Neisseriaceae/microbiologia , Fatores de Virulência/fisiologia , Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Cápsulas Bacterianas/fisiologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Sangue/microbiologia , Atividade Bactericida do Sangue , Pré-Escolar , Fímbrias Bacterianas/química , Regulação Bacteriana da Expressão Gênica , Humanos , Lactente , Kingella kingae/genética , Kingella kingae/crescimento & desenvolvimento , Infecções por Neisseriaceae/imunologia , Neutrófilos/imunologia , Polissacarídeos Bacterianos/fisiologia , Mucosa Respiratória/microbiologia , Virulência/genética , Fatores de Virulência/genética
6.
Med Microbiol Immunol ; 209(3): 277-299, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31784893

RESUMO

The capacity of pathogenic microorganisms to adhere to host cells and avoid clearance by the host immune system is the initial and most decisive step leading to infections. Bacteria have developed different strategies to attach to diverse host surface structures. One important strategy is the adhesion to extracellular matrix (ECM) proteins (e.g., collagen, fibronectin, laminin) that are highly abundant in connective tissue and basement membranes. Gram-negative bacteria express variable outer membrane proteins (adhesins) to attach to the host and to initiate the process of infection. Understanding the underlying molecular mechanisms of bacterial adhesion is a prerequisite for targeting this interaction by "anti-ligands" to prevent colonization or infection of the host. Future development of such "anti-ligands" (specifically interfering with bacteria-host matrix interactions) might result in the development of a new class of anti-infective drugs for the therapy of infections caused by multidrug-resistant Gram-negative bacteria. This review summarizes our current knowledge about the manifold interactions of adhesins expressed by Gram-negative bacteria with ECM proteins and the use of this information for the generation of novel therapeutic antivirulence strategies.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Proteínas da Matriz Extracelular/fisiologia , Fibronectinas/fisiologia , Bactérias Gram-Negativas/fisiologia , Interações entre Hospedeiro e Microrganismos , Bactérias Gram-Negativas/patogenicidade , Humanos
7.
Nat Commun ; 10(1): 4644, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604911

RESUMO

In mammalian cells, the internal and external leaflets of the plasma membrane (PM) possess different phospholipids. Phosphatidylserine (PS) is normally confined to the inner (cytoplasmic) membrane leaflet. Here we report that the adhesin CPn0473 of the human pathogenic bacterium Chlamydia pneumoniae (Cpn) binds to the PM of human cells and induces PS externalization but unexpectedly not apoptosis. PS externalization is increased in human cells exposed to infectious Cpn cells expressing increased CPn0473 and reduced in exposure to Cpn expressing decreased CPn0473. CPn0473 binds specifically to synthetic membranes carrying PS and stimulates pore formation. Asymmetric giant unilamellar vesicles (GUVs) in which PS is restricted to the inner leaflet reveal that CPn0473 induces PS externalization in the absence of other proteins. Thus our identification of CPn0473 as a bacterial PS translocator capable of specific and apoptosis-independent PS externalization during infection extends the spectrum of mechanisms intracellular pathogens use to enter host cells.


Assuntos
Adesinas Bacterianas/fisiologia , Chlamydophila pneumoniae/fisiologia , Fosfatidilserinas/metabolismo , Adesinas Bacterianas/metabolismo , Interações entre Hospedeiro e Microrganismos , Humanos , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo
8.
Microb Pathog ; 137: 103748, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31521802

RESUMO

Salmonellosis is a serious threat to human and animal health. Salmonella adhesion to the host cell is an initial and most crucial step in the pathogenesis of salmonellosis. Many factors are involved in the adhesion process of Salmonella infection. Fimbriae are one of the most important factors in the adhesion of Salmonella. The Salmonella fimbriae are assembled in three types of assembly pathways: chaperon-usher, nucleation-precipitation, and type IV fimbriae. These assembly pathways lead to multiple types of fimbriae. Salmonella fimbriae bind to host cell receptors to initiate adhesion. So far, many receptors have been identified, such as Toll-like receptors. However, several receptors that may be involved in the adhesive mechanism of Salmonella fimbriae are still un-identified. This review aimed to summarize the types of Salmonella fimbriae produced by different assembly pathways and their role in adhesion. It also enlisted previously discovered receptors involved in adhesion. This review might help readers to develop a comprehensive understanding of Salmonella fimbriae, their role in adhesion, and recently developed strategies to counter Salmonella infection.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana/fisiologia , Fímbrias Bacterianas/fisiologia , Salmonella/fisiologia , Adesinas Bacterianas/genética , Animais , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/fisiologia , Genes Bacterianos , Humanos , Salmonella/genética , Infecções por Salmonella , Receptores Toll-Like
9.
Trends Microbiol ; 27(11): 927-941, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31375310

RESUMO

The microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are a family of proteins that are defined by the presence of two adjacent IgG-like folded subdomains. These promote binding to ligands by mechanisms that involve major conformational changes exemplified by the binding to fibrinogen by the 'dock-lock-latch' mechanism or to collagen by the 'collagen hug'. Clumping factors A and B are two such MSCRAMMs that have several important roles in the pathogenesis of Staphylococcus aureus infections. MSCRAMM architecture, ligand binding, and roles in infection and colonization are examined with a focus on recent developments with clumping factors.


Assuntos
Adesinas Bacterianas/fisiologia , Cocos Gram-Positivos/fisiologia , Adesinas Bacterianas/química , Proteínas de Bactérias/química , Proteínas de Transporte/química , Parede Celular/química , Parede Celular/fisiologia , Coagulase/química , Humanos , Ligantes , Ligação Proteica , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Staphylococcus epidermidis/fisiologia
10.
Curr Biol ; 29(4): 616-630.e5, 2019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30744974

RESUMO

The selective pressure by infectious agents is a major driving force in the evolution of humans and other mammals. Members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family serve as receptors for bacterial pathogens of the genera Haemophilus, Helicobacter, Neisseria, and Moraxella, which engage CEACAMs via distinct surface adhesins. While microbial attachment to epithelial CEACAMs facilitates host colonization, recognition by CEACAM3, a phagocytic receptor expressed by granulocytes, eliminates CEACAM-binding bacteria. Sequence analysis of primate CEACAM3 orthologs reveals that this innate immune receptor is one of the most rapidly evolving human proteins. In particular, the pathogen-binding extracellular domain of CEACAM3 shows a high degree of non-synonymous versus synonymous nucleotide exchanges, indicating an exceptionally strong positive selection. Using CEACAM3 domains derived from different primates, we find that the amino acid alterations found in CEACAM3 translate into characteristic binding patterns for bacterial adhesins. One such amino acid residue is F62 in human and chimp CEACAM3, which is not present in other primates and which is critical for binding the OMP P1 adhesin of Haemophilus aegyptius. Incorporation of the F62-containing motif into gorilla CEACAM3 results in a gain-of-function phenotype with regard to phagocytosis of H. aegyptius. Moreover, CEACAM3 polymorphisms found in human subpopulations widen the spectrum of recognized bacterial adhesins, suggesting an ongoing multivariate selection acting on this innate immune receptor. The species-specific detection of diverse bacterial adhesins helps to explain the exceptionally fast evolution of CEACAM3 within the primate lineage and provides an example of Red Queen dynamics in the human genome.


Assuntos
Adesinas Bacterianas/fisiologia , Antígeno Carcinoembrionário/genética , Evolução Molecular , Gorilla gorilla/genética , Imunidade Inata/genética , Pan troglodytes/genética , Receptores Imunológicos/genética , Animais , Fenômenos Fisiológicos Bacterianos , Antígeno Carcinoembrionário/imunologia , Gorilla gorilla/imunologia , Humanos , Pan troglodytes/imunologia , Receptores Imunológicos/imunologia
11.
J R Soc Interface ; 15(146)2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30232243

RESUMO

Dense bacterial communities, known as biofilms, can have functional spatial organization driven by self-organizing chemical and physical interactions between cells, and their environment. In this work, we investigated intercellular adhesion, a pervasive property of bacteria in biofilms, to identify effects on the internal structure of bacterial colonies. We expressed the self-recognizing ag43 adhesin protein in Escherichia coli to generate adhesion between cells, which caused aggregation in liquid culture and altered microcolony morphology on solid media. We combined the adhesive phenotype with an artificial colony patterning system based on plasmid segregation, which marked clonal lineage domains in colonies grown from single cells. Engineered E. coli were grown to colonies containing domains with varying adhesive properties, and investigated with microscopy, image processing and computational modelling techniques. We found that intercellular adhesion elongated the fractal-like boundary between cell lineages only when both domains within the colony were adhesive, by increasing the rotational motion during colony growth. Our work demonstrates that adhesive intercellular interactions can have significant effects on the spatial organization of bacterial populations, which can be exploited for biofilm engineering. Furthermore, our approach provides a robust platform to study the influence of intercellular interactions on spatial structure in bacterial populations.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Biofilmes , Algoritmos , Adesão Celular , Simulação por Computador , Escherichia coli , Proteínas de Escherichia coli/fisiologia , Fractais , Modelos Biológicos , Movimento (Física) , Fenótipo , Plasmídeos
12.
Mol Oral Microbiol ; 33(5): 364-377, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29939498

RESUMO

Acinetobacter baumannii is a nosocomial, opportunistic pathogen that causes several serious conditions including meningitis, septicemia, endocarditis, and pneumonia. It can be found in the oral biofilm, which may be a reservoir for pneumonia and chronic obstructive pulmonary disease. Subgingival colonization by A. baumannii is associated with chronic and aggressive periodontitis as well as refractory periodontal disease. Porphyromonas gingivalis, a keystone periodontal pathogen localized to subgingival plaque, is also implicated in several chronic conditions including aspiration pneumonia. Although both bacteria are found together in subgingival plaque and can cause multiple polymicrobial infections, nothing is known about the interactions between these two important human pathogens. In this study, we used RNA sequencing to understand the transcriptional response of both species as they adapt to heterotypic communities. Among the differentially regulated genes were those encoding a number of important virulence factors for both species including adhesion, biofilm formation, and protein secretion. Additionally, the presence of A. baumannii increased the abundance of P. gingivalis in model dual-species communities. Collectively these results suggest that both P. gingivalis and A. baumannii adapt to each other and have synergistic potential for increased pathogenicity. In identifying the mechanisms that promote pathogenicity and refractory disease, novel approaches to mitigate polymicrobial synergistic interactions may be developed to treat or prevent associated diseases.


Assuntos
Acinetobacter baumannii/genética , Adesinas Bacterianas/fisiologia , Biofilmes/crescimento & desenvolvimento , Interações Microbianas , Porphyromonas gingivalis/genética , Acinetobacter baumannii/patogenicidade , Adesinas Bacterianas/genética , Placa Dentária/microbiologia , Perfilação da Expressão Gênica , Humanos , Porphyromonas gingivalis/patogenicidade , Análise de Sequência de RNA , Fatores de Virulência/metabolismo
13.
Infect Immun ; 86(7)2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29661931

RESUMO

Streptococcus gordonii is an early colonizer of the oral cavity. Although a variety of S. gordonii adherence mechanisms have been described, current dogma is that the major receptor for S. gordonii is sialic acid. However, as many bacterial species in the oral cavity produce neuraminidase that can cleave terminal sialic acid, it is unclear whether S. gordonii relies on sialic acid for adherence to oral surfaces or if this species has developed alternative binding strategies. Previous studies have examined adherence to immobilized glycoconjugates and identified binding to additional glycans, but no prior studies have defined the contribution of these different glycan structures in adherence to oral epithelial cells. We determined that the majority of S. gordonii strains tested did not rely on sialic acid for efficient adherence. In fact, adherence of some strains was significantly increased following neuraminidase treatment. Further investigation of representative strains that do not rely on sialic acid for adherence revealed binding not only to sialic acid via the serine-rich repeat protein GspB but also to ß-1,4-linked galactose. Adherence to this carbohydrate occurs via an unknown adhesin distinct from those utilized by Streptococcus oralis and Streptococcus pneumoniae Demonstrating the potential biological relevance of binding to this cryptic receptor, we established that S. oralis increases S. gordonii adherence in a neuraminidase-dependent manner. These data suggest that S. gordonii has evolved to simultaneously utilize both terminal and cryptic receptors in response to the production of neuraminidase by other species in the oral environment.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Proteínas de Transporte/fisiologia , Ácido N-Acetilneuramínico/fisiologia , Neuraminidase/biossíntese , Streptococcus gordonii/fisiologia , Galactose/metabolismo , Hemaglutininas Virais , Humanos , Mucosa Bucal/microbiologia , Streptococcus oralis/fisiologia
14.
Mol Immunol ; 93: 47-54, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29145158

RESUMO

Although Vibrio cholerae colonizes the small intestine and induces acute inflammatory responses, less is known about the molecular mechanisms of V. cholerae-induced inflammatory responses in the intestine. We recently reported that OmpU, one of the most abundant outer membrane proteins of V. cholerae, plays an important role in the innate immunity of the whole bacteria. In this study, we evaluated the role of OmpU in induction of IL-8, a representative chemokine that recruits various inflammatory immune cells, in the human intestinal epithelial cell (IEC) line, HT-29. Recombinant OmpU (rOmpU) of V. cholerae induced IL-8 expression at the mRNA and protein levels in a dose- and time-dependent manner. Interestingly, IL-8 was secreted through both apical and basolateral sides of the polarized HT-29 cells upon apical exposure to rOmpU but not upon basolateral exposure. rOmpU-induced IL-8 expression was inhibited by interference of lipid raft formation with nystatin, but not by blocking the formation of clathrin-coated pits with chlorpromazine. In addition, rOmpU-induced IL-8 expression was mediated via ERK1/2 and p38 kinase pathways, but not via JNK signaling pathway. Finally, V. cholerae lacking ompU elicited decreased IL-8 expression and adherence to HT-29 cells compared to the parental strain. Collectively, these results suggest that V. cholerae OmpU might play an important role in intestinal inflammation by inducing IL-8 expression in human IECs.


Assuntos
Adesinas Bacterianas/fisiologia , Células Epiteliais/metabolismo , Interleucina-8/biossíntese , Adesinas Bacterianas/genética , Adesão Celular , Polaridade Celular , Clorpromazina/farmacologia , Relação Dose-Resposta a Droga , Regulação Bacteriana da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Reporter , Células HT29 , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Nistatina/farmacologia , Polimixina B/farmacologia , RNA Mensageiro/biossíntese , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transdução de Sinais , Vibrio cholerae/patogenicidade , Virulência
15.
Artigo em Inglês | MEDLINE | ID: mdl-29164073

RESUMO

Streptococcus equi subsp. zooepidemicus (S. zooepidemicus) is an opportunistic pathogen of several species including humans. S. zooepidemicus is found on mucus membranes of healthy horses, but can cause acute and chronic endometritis. Recently S. zooepidemicus was found able to reside in the endometrium for prolonged periods of time. Thus, we hypothesized that an intracellular phase may be part of the S. zooepidemicus pathogenesis and investigated if S. zooepidemicus was able to invade and survive inside epithelial cells. HEp-2 and HeLa cell lines were co-cultured with two S. zooepidemicus strains (1-4a and S31A1) both originating from the uterus of mares suffering from endometritis. Cells were fixed at different time points during the 23 h infection assay and field emission scanning electron microscopy (FESEM) was used to characterize adhesion and invasion mechanisms. The FESEM images showed three morphologically different types of invasion for both bacterial strains. The main port of entry was through large invaginations in the epithelial cell membrane. Pili-like bacterial appendages were observed when the S. zooepidemicus cells were in close proximity to the epithelial cells indicating that attachment and invasion were active processes. Adherent and intracellular S. zooepidemicus, and bacteria in association with lysosomes was determined by immunofluorescence staining techniques and fluorescence microscopy. Quantification of intracellular bacteria was determined in penicillin protection assays. Both S. zooepidemicus strains investigated were able to invade epithelial cells although at different magnitudes. The immunofluorescence data showed significantly higher adhesion and invasion rates for strain 1-4a when compared to strain S31A1. S. zooepidemicus was able to survive intracellularly, but the survival rate decreased over time in the cell culture system. Phagosome-like compartments containing S. zooepidemicus at some stages fused with lysosomes to form a phagolysosome. The results indicate that an intracellular phase may be one way S. zooepidemicus survives in the host, and could in part explain how S. zooepidemicus can cause recurrent/persistent infections. Future studies should reveal the ability of S. zooepidemicus to internalize and survive in primary equine endometrial cells and during in vivo conditions.


Assuntos
Adesinas Bacterianas/fisiologia , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Infecções Estreptocócicas/microbiologia , Streptococcus/patogenicidade , Animais , Linhagem Celular , Técnicas de Cocultura , Contagem de Colônia Microbiana , Endometrite/microbiologia , Endometrite/veterinária , Feminino , Células HeLa , Células Hep G2 , Doenças dos Cavalos/microbiologia , Cavalos , Humanos , Lisossomos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Infecções Estreptocócicas/veterinária , Streptococcus/classificação , Streptococcus/crescimento & desenvolvimento , Streptococcus/isolamento & purificação
16.
Nihon Saikingaku Zasshi ; 72(4): 219-227, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-29109335

RESUMO

Many bacteria symbiotic and parasitic in humans are included in the genera Bacteroides, Prevotella, Porphyromonas and others, which belong to the phylum Bacteroidetes. We have been studying gingipain, a major secretory protease of Porphyromonas gingivalis which is a periodontopathogenic bacterium belonging to the genus Porphyromonas, and pili which contribute to host colonization in the bacterium. In the process, it was found that gingipain was secreted by a system not reported previously. Furthermore, this secretion system was found to exist widely in the Bacteroidetes phylum bacteria and closely related to the gliding motility of bacteroidete bacteria, and it was named the Por secretion system (later renamed the type IX secretion system). Regarding P. gingivalis pili, it was found that the pilus protein is transported as a lipoprotein to the cell surface, and the pilus formation occurs due to degradation by arginine-gingipain. Pili with this novel formation mechanism was found to be widely present in bacteria belonging to the class Bacteroidia in the phylum Bacteroidetes and was named the type V pili.


Assuntos
Adesinas Bacterianas/fisiologia , Sistemas de Secreção Bacterianos , Bacteroidetes/metabolismo , Bacteroidetes/patogenicidade , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/fisiologia , Fímbrias Bacterianas/fisiologia , Animais , Arginina/fisiologia , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Bacteroidetes/genética , Bacteroidetes/fisiologia , Membrana Celular/metabolismo , Fímbrias Bacterianas/metabolismo , Genoma Bacteriano , Cisteína Endopeptidases Gingipaínas , Humanos , Lipoproteínas/metabolismo , Camundongos , Periodontite/microbiologia , Porphyromonas gingivalis , Transporte Proteico
17.
Microb Pathog ; 113: 176-180, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29038054

RESUMO

Erysipelothrix rhusiopathiae is the causative agent of animal erysipelas and human erysipeloid. The major protective antigen SpaA was suggested to play important roles in E. rhusiopathiae adhesion to host cells, but there is no specific study on SpaA pathogenic roles in adhesion. In this study we characterized direct and indirect roles of SpaA in E. rhusiopathiae adhesion to porcine endothelial cells. Recombinant E. rhusiopathiae SpaA (rSpaA) successfully binded to porcine iliac arterial endothelial cells. rSpaA protein pre-incubating endothelial cells or rSpaA antiserum pre-incubating E. rhusiopathiae significantly decreased E. rhusiopathiae adhesion to endothelial cells. rSpaA successfully binded host plasminogen and fibronectin, and rSpaA antiserum significantly decreased plasminogen-recruitment activity but not fibronectin-recruitment activity of E. rhusiopathiae. In conclusion, SpaA acts as adhesin in E. rhusiopathiae adhesion to host cells, and SpaA binding activity to host plasminogen highly likely play roles in this adhesion.


Assuntos
Adesinas Bacterianas/fisiologia , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Células Endoteliais/microbiologia , Erysipelothrix/fisiologia , Adesinas Bacterianas/imunologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Aderência Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Fibronectinas/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Artéria Ilíaca/microbiologia , Plasminogênio/metabolismo , Ligação Proteica/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Suínos , Fatores de Virulência/fisiologia
18.
J Infect Dis ; 216(11): 1452-1459, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-28968845

RESUMO

Clostridium difficile infection (CDI) is an important hospital-acquired infection resulting from the germination of spores in the intestine as a consequence of antibiotic-mediated dysbiosis of the gut microbiota. Key to this is CotE, a protein displayed on the spore surface and carrying 2 functional elements, an N-terminal peroxiredoxin and a C-terminal chitinase domain. Using isogenic mutants, we show in vitro and ex vivo that CotE enables binding of spores to mucus by direct interaction with mucin and contributes to its degradation. In animal models of CDI, we show that when CotE is absent, both colonization and virulence were markedly reduced. We demonstrate here that the attachment of spores to the intestine is essential in the development of CDI. Spores are usually regarded as biochemically dormant, but our findings demonstrate that rather than being simply agents of transmission and dissemination, spores directly contribute to the establishment and promotion of disease.


Assuntos
Adesinas Bacterianas/fisiologia , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Esporos Bacterianos/química , Animais , Proteínas de Bactérias/genética , Quitinases/metabolismo , Clostridioides difficile/genética , Clostridioides difficile/metabolismo , Contagem de Colônia Microbiana , Cricetinae , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Parasita/fisiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Mesocricetus , Camundongos , Mucinas/metabolismo , Mutação , Peroxirredoxinas/metabolismo , Esporos Bacterianos/genética , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/patogenicidade , Virulência
19.
PLoS One ; 12(6): e0179601, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28665944

RESUMO

Bacterial adhesion to collagen, the most abundant protein in humans, is a critical step in the initiation and persistence of numerous bacterial infections. In this study, we explore the collagen binding mechanism of the multi-modular cell wall anchored collagen adhesin (CNA) in Staphylococcus aureus and examine how applied mechanical forces can modulate adhesion ability. The common structural-functional elements and domain organization of CNA are present across over 50 genera of bacteria. Through the use of molecular dynamics models and normal mode analysis, we shed light on the CNA's structural and conformational dynamics and its interactions with collagen that lead to collagen binding. Our results suggest that the linker region, CNA165-173, acts as a hinge exhibiting bending, extensional, and torsional modes of structural flexibility and its residues are key in the interaction of the CNA-collagen complex. Steered molecular dynamics simulations were conducted with umbrella sampling. During the course of these simulations, the 'locking' latch from the CNA N2 domain was dissociated from its groove in the CNA N1 domain, implying the importance of the latch for effective ligand binding. Finally, we observed that the binding efficiency of the CNA N1-N2 domains to collagen decreases greatly with increasing tensile force application to the collagen peptides. Thus, CNA and similar adhesins might preferentially bind to sites in which collagen fibers are cleaved, such as in wounded, injured, or inflamed tissues, or in which the collagenous tissue is less mature. As alternative techniques for control of bacterial infection are in-demand due to the rise of bacterial antibiotic resistance, results from our computational studies with respect to the mechanoregulation of the collagen binding site may inspire new therapeutics and engineering solutions by mechanically preventing colonization and/or further pathogenesis.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana/fisiologia , Colágeno/fisiologia , Adesinas Bacterianas/química , Adesinas Bacterianas/metabolismo , Colágeno/química , Colágeno/metabolismo , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Resistência à Tração
20.
Front Biosci (Elite Ed) ; 9(2): 266-275, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28199190

RESUMO

Increased resistance and survival, as well as immune evasion, play a significant role in the pathogenicity of Acinetobacter baumannii. Here, we report on the adhesion of the bacterium to epithelial cells and formation of biofilm on abiotic surfaces. We identificed autotransporter (AT) genes that encode homologues (Fha-like) of the two-partner secretion system (TPS) in A.baumannii (ATCC19606) which we designate as FhaB (exoprotein) and FhaC (transporter) and demonstrate that these novel genes, are under the control of distinct regulatable promoters within either the same (FhaBC) or two distinct (FhaB and FhaC) cells. The expression of this gene in outer membrane protein (OM) showed them to be deficient in the adherence to A546 cells. FhaB is involved in hydrophobicity of A. baumannii ATCC19606 while FhaBC is associated with biofilm formation. The vaccinogenic potential of FHA-like proteins offers use of these targets as novel therapeutic strategies to limit A.baumannii associated morbidity and mortality.


Assuntos
Acinetobacter baumannii/patogenicidade , Adesinas Bacterianas/fisiologia , Acinetobacter baumannii/genética , Acinetobacter baumannii/fisiologia , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Sequência de Aminoácidos , Aderência Bacteriana/genética , Biofilmes , Clonagem Molecular , Biologia Computacional , Sequência Conservada , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Interações Hidrofóbicas e Hidrofílicas , Proteínas Recombinantes de Fusão , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...