Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
mBio ; 15(3): e0282123, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38376160

RESUMO

The cellular junctional architecture remodeling by Listeria adhesion protein-heat shock protein 60 (LAP-Hsp60) interaction for Listeria monocytogenes (Lm) passage through the epithelial barrier is incompletely understood. Here, using the gerbil model, permissive to internalin (Inl) A/B-mediated pathways like in humans, we demonstrate that Lm crosses the intestinal villi at 48 h post-infection. In contrast, the single isogenic (lap- or ΔinlA) or double (lap-ΔinlA) mutant strains show significant defects. LAP promotes Lm translocation via endocytosis of cell-cell junctional complex in enterocytes that do not display luminal E-cadherin. In comparison, InlA facilitates Lm translocation at cells displaying apical E-cadherin during cell extrusion and mucus expulsion from goblet cells. LAP hijacks caveolar endocytosis to traffic integral junctional proteins to the early and recycling endosomes. Pharmacological inhibition in a cell line and genetic knockout of caveolin-1 in mice prevents LAP-induced intestinal permeability, junctional endocytosis, and Lm translocation. Furthermore, LAP-Hsp60-dependent tight junction remodeling is also necessary for InlA access to E-cadherin for Lm intestinal barrier crossing in InlA-permissive hosts. IMPORTANCE: Listeria monocytogenes (Lm) is a foodborne pathogen with high mortality (20%-30%) and hospitalization rates (94%), particularly affecting vulnerable groups such as pregnant women, fetuses, newborns, seniors, and immunocompromised individuals. Invasive listeriosis involves Lm's internalin (InlA) protein binding to E-cadherin to breach the intestinal barrier. However, non-functional InlA variants have been identified in Lm isolates, suggesting InlA-independent pathways for translocation. Our study reveals that Listeria adhesion protein (LAP) and InlA cooperatively assist Lm entry into the gut lamina propria in a gerbil model, mimicking human listeriosis in early infection stages. LAP triggers caveolin-1-mediated endocytosis of critical junctional proteins, transporting them to early and recycling endosomes, facilitating Lm passage through enterocytes. Furthermore, LAP-Hsp60-mediated junctional protein endocytosis precedes InlA's interaction with basolateral E-cadherin, emphasizing LAP and InlA's cooperation in enhancing Lm intestinal translocation. This understanding is vital in combating the severe consequences of Lm infection, including sepsis, meningitis, encephalitis, and brain abscess.


Assuntos
Listeria monocytogenes , Listeria , Listeriose , Recém-Nascido , Feminino , Camundongos , Gravidez , Humanos , Animais , Listeria monocytogenes/genética , Caveolina 1/metabolismo , Cavéolas/metabolismo , Gerbillinae , Proteínas de Bactérias/metabolismo , Listeriose/metabolismo , Caderinas/genética
2.
Cell Rep ; 42(5): 112515, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37171960

RESUMO

Listeria adhesion protein (LAP) is a secreted acetaldehyde alcohol dehydrogenase (AdhE) that anchors to an unknown molecule on the Listeria monocytogenes (Lm) surface, which is critical for its intestinal epithelium crossing. In the present work, immunoprecipitation and mass spectrometry identify internalin B (InlB) as the primary ligand of LAP (KD ∼ 42 nM). InlB-deleted and naturally InlB-deficient Lm strains show reduced LAP-InlB interaction and LAP-mediated pathology in the murine intestine and brain invasion. InlB-overexpressing non-pathogenic Listeria innocua also displays LAP-InlB interplay. In silico predictions reveal that a pocket region in the C-terminal domain of tetrameric LAP is the binding site for InlB. LAP variants containing mutations in negatively charged (E523S, E621S) amino acids in the C terminus confirm altered binding conformations and weaker affinity for InlB. InlB transforms the housekeeping enzyme, AdhE (LAP), into a moonlighting pathogenic factor by fastening on the cell surface.


Assuntos
Listeria monocytogenes , Listeria , Animais , Camundongos , Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Listeria/metabolismo , Listeria monocytogenes/metabolismo , Membrana Celular/metabolismo , Álcool Desidrogenase/metabolismo
3.
Lett Appl Microbiol ; 76(2)2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-36737424

RESUMO

Zearalenone (ZEN) is an estrogenic mycotoxin produced by the Fusarium species and induces severe reproductive disorders in animals thus a major concern in the livestock industry. Probiotic bacteria treatments have been shown to inactivate mycotoxins, therefore, in this study, we investigated the effect of two commercial probiotic feed additives on the sequestration of ZEN. Commercial probiotic blends containing clay-based binder with Aspergillus niger, Bacillus licheniformis, Bacillus pumilus, and Bacillus subtilis at various proportions from BioMatrix International were incubated with ZEN in a time-dependent manner and then analyzed by Enzyme-Linked Immunosorbent Assay (ELISA) to quantify unbound ZEN. Sequestration of ZEN was further verified by using MCF-7 cell-based cytotoxicity and/or cell proliferation assays. ZEN, or probiotic mix, was nontoxic to MCF-7 cells. Probiotic blends decreased ZEN concentration by 45% (∼100 µg L-1) and prevented ZEN from inducing MCF-7 cell proliferation (20%-28% reduction). The probiotic feed supplements tested show a potential utility in ZEN neutralization.


Assuntos
Bacillus licheniformis , Fusarium , Micotoxinas , Zearalenona , Animais , Zearalenona/análise , Zearalenona/toxicidade
4.
NPJ Biofilms Microbiomes ; 7(1): 18, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33558519

RESUMO

Environmental cues promote microbial biofilm formation and physiological and genetic heterogeneity. In food production facilities, biofilms produced by pathogens are a major source for food contamination; however, the pathogenesis of biofilm-isolated sessile cells is not well understood. We investigated the pathogenesis of sessile Listeria monocytogenes (Lm) using cell culture and mouse models. Lm sessile cells express reduced levels of the lap, inlA, hly, prfA, and sigB and show reduced adhesion, invasion, translocation, and cytotoxicity in the cell culture model than the planktonic cells. Oral challenge of C57BL/6 mice with food, clinical, or murinized-InlA (InlAm) strains reveals that at 12 and 24 h post-infection (hpi), Lm burdens are lower in tissues of mice infected with sessile cells than those infected with planktonic cells. However, these differences are negligible at 48 hpi. Besides, the expressions of inlA and lap mRNA in sessile Lm from intestinal content are about 6.0- and 280-fold higher than the sessle inoculum, respectively, suggesting sessile Lm can still upregulate virulence genes shortly after ingestion (12 h). Similarly, exposure to simulated gastric fluid (SGF, pH 3) and intestinal fluid (SIF, pH 7) for 13 h shows equal reduction in sessile and planktonic cell counts, but induces LAP and InlA expression and pathogenic phenotypes. Our data show that the virulence of biofilm-isolated Lm is temporarily attenuated and can be upregulated in mice during the early stage (12-24 hpi) but fully restored at a later stage (48 hpi) of infection. Our study further demonstrates that in vitro cell culture assay is unreliable; therefore, an animal model is essential for studying the pathogenesis of biofilm-isolated bacteria.


Assuntos
Biofilmes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Fatores de Virulência/genética , Animais , Aderência Bacteriana , Proteínas de Bactérias/genética , Células CACO-2 , Modelos Animais de Doenças , Feminino , Microbiologia de Alimentos , Regulação Bacteriana da Expressão Gênica , Humanos , Listeria monocytogenes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Front Microbiol ; 11: 575615, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329436

RESUMO

Rapid detection of live pathogens is of paramount importance to ensure food safety. At present, nucleic acid-based polymerase chain reaction and antibody-based lateral flow assays are the primary methods of choice for rapid detection, but these are prone to interference from inhibitors, and resident microbes. Moreover, the positive results may neither assure virulence potential nor viability of the analyte. In contrast, the mammalian cell-based assay detects pathogen interaction with the host cells and is responsive to only live pathogens, but the short shelf-life of the mammalian cells is the major impediment for its widespread application. An innovative approach to prolong the shelf-life of mammalian cells by using formalin was undertaken. Formalin (4% formaldehyde)-fixed human ileocecal adenocarcinoma cell line, HCT-8 on 24-well tissue culture plates was used for the capture of viable pathogens while an antibody was used for specific detection. The specificity of the Mammalian Cell-based ImmunoAssay (MaCIA) was validated with Salmonella enterica serovar Enteritidis and Typhimurium as model pathogens and further confirmed against a panel of 15 S. Enteritidis strains, 8 S. Typhimurium, 11 other Salmonella serovars, and 14 non-Salmonella spp. The total detection time (sample-to-result) of MaCIA with artificially inoculated ground chicken, eggs, milk, and cake mix at 1-10 CFU/25 g was 16-21 h using a traditional enrichment set up but the detection time was shortened to 10-12 h using direct on-cell (MaCIA) enrichment. Formalin-fixed stable cell monolayers in MaCIA provide longer shelf-life (at least 14 weeks) for possible point-of-need deployment and multi-sample testing on a single plate.

6.
Nat Commun ; 11(1): 6344, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311493

RESUMO

Probiotic bacteria reduce the intestinal colonization of pathogens. Yet, their use in preventing fatal infection caused by foodborne Listeria monocytogenes (Lm), is inconsistent. Here, we bioengineered Lactobacillus probiotics (BLP) to express the Listeria adhesion protein (LAP) from a non-pathogenic Listeria (L. innocua) and a pathogenic Listeria (Lm) on the surface of Lactobacillus casei. The BLP strains colonize the intestine, reduce Lm mucosal colonization and systemic dissemination, and protect mice from lethal infection. The BLP competitively excludes Lm by occupying the surface presented LAP receptor, heat shock protein 60 and ameliorates the Lm-induced intestinal barrier dysfunction by blocking the nuclear factor-κB and myosin light chain kinase-mediated redistribution of the major epithelial junctional proteins. Additionally, the BLP increases intestinal immunomodulatory functions by recruiting FOXP3+T cells, CD11c+ dendritic cells and natural killer cells. Engineering a probiotic strain with an adhesion protein from a non-pathogenic bacterium provides a new paradigm to exclude pathogens and amplify their inherent health benefits.


Assuntos
Lacticaseibacillus casei/metabolismo , Listeria monocytogenes/efeitos dos fármacos , Listeriose/prevenção & controle , Probióticos/metabolismo , Probióticos/farmacologia , Administração Oral , Animais , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Antígeno CD11c , Linhagem Celular , Chaperonina 60/metabolismo , Células Dendríticas , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Intestinos/microbiologia , Células Matadoras Naturais , Lacticaseibacillus casei/genética , Listeria/genética , Listeria monocytogenes/genética , Listeria monocytogenes/crescimento & desenvolvimento , Camundongos , Quinase de Cadeia Leve de Miosina/metabolismo , NF-kappa B/metabolismo , Linfócitos T
7.
Trends Microbiol ; 27(5): 408-425, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30661918

RESUMO

The intestinal epithelial cell lining provides the first line of defense, yet foodborne pathogens such as Listeria monocytogenes can overcome this barrier; however, the underlying mechanism is not well understood. Though the host M cells in Peyer's patch and the bacterial invasion protein internalin A (InlA) are involved, L. monocytogenes can cross the gut barrier in their absence. The interaction of Listeria adhesion protein (LAP) with the host cell receptor (heat shock protein 60) disrupts the epithelial barrier, promoting bacterial translocation. InlA aids L. monocytogenes transcytosis via interaction with the E-cadherin receptor, which is facilitated by epithelial cell extrusion and goblet cell exocytosis; however, LAP-induced cell junction opening may be an alternative bacterial strategy for InlA access to E-cadherin and its translocation. Here, we summarize the strategies that L. monocytogenes employs to circumvent the intestinal epithelial barrier and compare and contrast these strategies with other enteric bacterial pathogens. Additionally, we provide implications of recent findings for food safety regulations.


Assuntos
Proteínas de Bactérias/metabolismo , Translocação Bacteriana , Interações Hospedeiro-Patógeno/imunologia , Mucosa Intestinal/microbiologia , Listeria monocytogenes/metabolismo , Animais , Aderência Bacteriana , Caderinas/metabolismo , Inocuidade dos Alimentos , Doenças Transmitidas por Alimentos/microbiologia , Doenças Transmitidas por Alimentos/prevenção & controle , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Listeria monocytogenes/patogenicidade
8.
Front Microbiol ; 9: 1352, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30034372

RESUMO

The emergence of bacterial resistance to therapeutic antibiotics limits options for treatment of common microbial diseases. Subinhibitory antibiotics dosing, often aid in the emergence of resistance, but its impact on pathogen's physiology and pathogenesis is not well understood. Here we investigated the effect of tunicamycin, a cell wall teichoic acid (WTA) biosynthesis inhibiting antibiotic at the subinhibitory dosage on Staphylococcus aureus and Listeria monocytogenes physiology, antibiotic cross-resistance, biofilm-formation, and virulence. Minimum inhibitory concentration (MIC) of tunicamycin to S. aureus and L. monocytogenes was 20-40 µg/ml and 2.5-5 µg/ml, respectively, and the subinhibitory concentration was 2.5-5 µg/ml and 0.31-0.62 µg/ml, respectively. Tunicamycin pre-exposure reduced cellular WTA levels by 18-20% and affected bacterial cell wall ultrastructure, cell membrane permeability, morphology, laser-induced colony scatter signature, and bacterial ability to form biofilms. It also induced a moderate level of cross-resistance to tetracycline, ampicillin, erythromycin, and meropenem for S. aureus, and ampicillin, erythromycin, vancomycin, and meropenem for L. monocytogenes. Pre-treatment of bacterial cells with subinhibitory concentrations of tunicamycin also significantly reduced bacterial adhesion to and invasion into an enterocyte-like Caco-2 cell line, which is supported by reduced expression of key virulence factors, Internalin B (InlB) and Listeria adhesion protein (LAP) in L. monocytogenes, and a S. aureus surface protein A (SasA) in S. aureus. Tunicamycin-treated bacteria or the bacterial WTA preparation suppressed NF-κB and inflammatory cytokine production (TNFα, and IL-6) from murine macrophage cell line (RAW 264.7) indicating the reduced WTA level possibly attenuates an inflammatory response. These results suggest that at the subinhibitory dosage, tunicamycin-mediated inhibition of WTA biosynthesis interferes with cell wall structure, pathogens infectivity and inflammatory response, and ability to form biofilms but promotes the development of antibiotic cross-resistance.

9.
Cell Host Microbe ; 23(4): 470-484.e7, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29606495

RESUMO

Intestinal epithelial cells are the first line of defense against enteric pathogens, yet bacterial pathogens, such as Listeria monocytogenes, can breach this barrier. We show that Listeria adhesion protein (LAP) induces intestinal epithelial barrier dysfunction to promote bacterial translocation. These disruptions are attributed to the production of pro-inflammatory cytokines TNF-α and IL-6, which is observed in mice challenged with WT and isogenic strains lacking the surface invasion protein Internalin A (ΔinlA), but not a lap- mutant. Additionally, upon engagement of its surface receptor Hsp60, LAP activates canonical NF-κB signaling, facilitating myosin light-chain kinase (MLCK)-mediated opening of the epithelial barrier via cellular redistribution of the epithelial junctional proteins claudin-1, occludin, and E-cadherin. Pharmacological inhibition of MLCK or NF-κB in cells or genetic ablation of MLCK in mice prevents mislocalization of junctional proteins and L. monocytogenes translocation. Thus, L. monocytogenes uses LAP to exploit epithelial defenses and cross the intestinal epithelial barrier.


Assuntos
Adesinas Bacterianas/metabolismo , Translocação Bacteriana , Interações Hospedeiro-Patógeno , Listeria monocytogenes/fisiologia , Adesinas Bacterianas/genética , Animais , Células CACO-2 , Chaperonina 60/genética , Chaperonina 60/metabolismo , Humanos , Interleucina-6/metabolismo , Listeria monocytogenes/genética , Camundongos , Células RAW 264.7 , Proteínas de Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Appl Environ Microbiol ; 82(11): 3256-3268, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26994085

RESUMO

UNLABELLED: In this study, we investigated whether a laser scatterometer designated BARDOT (bacterial rapid detection using optical scattering technology) could be used to directly screen colonies of Listeria monocytogenes, a model pathogen, with mutations in several known virulence genes, including the genes encoding Listeria adhesion protein (LAP; lap mutant), internalin A (ΔinlA strain), and an accessory secretory protein (ΔsecA2 strain). Here we show that the scatter patterns of lap mutant, ΔinlA, and ΔsecA2 colonies were markedly different from that of the wild type (WT), with >95% positive predictive values (PPVs), whereas for the complemented mutant strains, scatter patterns were restored to that of the WT. The scatter image library successfully distinguished the lap mutant and ΔinlA mutant strains from the WT in mixed-culture experiments, including a coinfection study using the Caco-2 cell line. Among the biophysical parameters examined, the colony height and optical density did not reveal any discernible differences between the mutant and WT strains. We also found that differential LAP expression in L. monocytogenes serotype 4b strains also affected the scatter patterns of the colonies. The results from this study suggest that BARDOT can be used to screen and enumerate mutant strains separately from the WT based on differential colony scatter patterns. IMPORTANCE: In studies of microbial pathogenesis, virulence-encoding genes are routinely disrupted by deletion or insertion to create mutant strains. Screening of mutant strains is an arduous process involving plating on selective growth media, replica plating, colony hybridization, DNA isolation, and PCR or immunoassays. We applied a noninvasive laser scatterometer to differentiate mutant bacterial colonies from WT colonies based on forward optical scatter patterns. This study demonstrates that BARDOT can be used as a novel, label-free, real-time tool to aid researchers in screening virulence gene-associated mutant colonies during microbial pathogenesis, coinfection, and genetic manipulation studies.


Assuntos
Proteínas de Bactérias/metabolismo , Técnicas Bacteriológicas/métodos , Lasers , Listeria monocytogenes/classificação , Proteínas de Membrana/deficiência , Propriedades de Superfície , Fatores de Virulência/deficiência , Proteínas de Bactérias/análise , Fenômenos Biofísicos , Listeria monocytogenes/química , Proteínas de Membrana/análise , Fatores de Virulência/análise
11.
PLoS One ; 10(8): e0135035, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26252374

RESUMO

We investigated the streptomycin-induced stress response in Salmonella enterica serovars with a laser optical sensor, BARDOT (bacterial rapid detection using optical scattering technology). Initially, the top 20 S. enterica serovars were screened for their response to streptomycin at 100 µg/mL. All, but four S. enterica serovars were resistant to streptomycin. The MIC of streptomycin-sensitive serovars (Enteritidis, Muenchen, Mississippi, and Schwarzengrund) varied from 12.5 to 50 µg/mL, while streptomycin-resistant serovar (Typhimurium) from 125-250 µg/mL. Two streptomycin-sensitive serovars (Enteritidis and Mississippi) were grown on brain heart infusion (BHI) agar plates containing sub-inhibitory concentration of streptomycin (1.25-5 µg/mL) and a streptomycin-resistant serovar (Typhimurium) was grown on BHI containing 25-50 µg/mL of streptomycin and the colonies (1.2 ± 0.1 mm diameter) were scanned using BARDOT. Data show substantial qualitative and quantitative differences in the colony scatter patterns of Salmonella grown in the presence of streptomycin than the colonies grown in absence of antibiotic. Mass-spectrometry identified overexpression of chaperonin GroEL, which possibly contributed to the observed differences in the colony scatter patterns. Quantitative RT-PCR and immunoassay confirmed streptomycin-induced GroEL expression while, aminoglycoside adenylyltransferase (aadA), aminoglycoside efflux pump (aep), multidrug resistance subunit acrA, and ribosomal protein S12 (rpsL), involved in streptomycin resistance, were unaltered. The study highlights suitability of the BARDOT as a non-invasive, label-free tool for investigating stress response in Salmonella in conjunction with the molecular and immunoassay methods.


Assuntos
Farmacorresistência Bacteriana , Salmonella typhimurium/efeitos dos fármacos , Estreptomicina/química , Ágar , Antibacterianos/química , Proteínas de Bactérias/genética , Humanos , Imunoensaio , Espectrometria de Massas , Testes de Sensibilidade Microbiana , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espalhamento de Radiação , Sorogrupo
12.
Int J Med Microbiol ; 303(1): 40-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23306065

RESUMO

Ehrlichia chaffeensis is a Gram-negative, obligate intracellular bacterium which causes the tick-borne disease human monocytic ehrlichiosis. In vertebrates, E. chaffeensis replicates in monocytes and macrophages. However, no clear cell or tissue tropism has been defined in arthropods. Our group identified two host genes that control E. chaffeensis replication and infection in vivo in Drosophila, Uridine cytidine kinase and separation anxiety. Using the UAS-GAL4 RNAi system, we generated F1 flies (UAS-gene of interestRNAi x tissue-GAL4 flies) that have Uck2 or san silenced in ubiquitous or tissue-specific fashion. When Uck2 or san were suppressed in the hemocytes or in the fat body, E. chaffeensis replicated poorly and caused significantly less severe infections. Silencing of these genes in the eyes, wings, or the salivary glands did not impact fly susceptibility or bacterial replication. Our data suggest that in Drosophila, E. chaffeensis replicates within the hemocytes, the insect homolog of mammalian macrophages, and in the fat body, the liver homolog of mammals.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/microbiologia , Ehrlichia chaffeensis/fisiologia , Ehrlichiose/microbiologia , Animais , Linhagem Celular , Cães , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/crescimento & desenvolvimento , Olho/microbiologia , Corpo Adiposo/microbiologia , Feminino , Inativação Gênica , Hemócitos/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/microbiologia , Masculino , Monócitos/microbiologia , Especificidade de Órgãos , Interferência de RNA , RNA Bacteriano/genética , Análise de Sobrevida , Uridina Quinase/genética , Asas de Animais/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA