Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 482
Filtrar
1.
PLoS Pathog ; 18(5): e1010511, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35605029

RESUMO

Hematogenous dissemination is a critical step in the evolution of local infection to systemic disease. The Lyme disease (LD) spirochete, which efficiently disseminates to multiple tissues, has provided a model for this process, in particular for the key early event of pathogen adhesion to the host vasculature. This occurs under shear force mediated by interactions between bacterial adhesins and mammalian cell-surface proteins or extracellular matrix (ECM). Using real-time intravital imaging of the Lyme spirochete in living mice, we previously identified BBK32 as the first LD spirochetal adhesin demonstrated to mediate early vascular adhesion in a living mouse; however, deletion of bbk32 resulted in loss of only about half of the early interactions, suggesting the existence of at least one other adhesin (adhesin-X) that promotes early vascular interactions. VlsE, a surface lipoprotein, was identified long ago by its capacity to undergo rapid antigenic variation, is upregulated in the mammalian host and required for persistent infection in immunocompetent mice. In immunodeficient mice, VlsE shares functional overlap with OspC, a multi-functional protein that displays dermatan sulfate-binding activity and is required for joint invasion and colonization. In this research, using biochemical and genetic approaches as well as intravital imaging, we have identified VlsE as adhesin-X; it is a dermatan sulfate (DS) adhesin that efficiently promotes transient adhesion to the microvasculature under shear force via its DS binding pocket. Intravenous inoculation of mice with a low-passage infectious B. burgdorferi strain lacking both bbk32 and vlsE almost completely eliminated transient microvascular interactions. Comparative analysis of binding parameters of VlsE, BBK32 and OspC provides a possible explanation why these three DS adhesins display different functionality in terms of their ability to promote early microvascular interactions.


Assuntos
Adesinas Bacterianas , Variação Antigênica , Antígenos de Bactérias , Proteínas de Bactérias , Borrelia burgdorferi , Lipoproteínas , Doença de Lyme , Microvasos , Adesinas Bacterianas/genética , Adesinas Bacterianas/imunologia , Animais , Variação Antigênica/genética , Variação Antigênica/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Aderência Bacteriana/genética , Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Borrelia burgdorferi/genética , Borrelia burgdorferi/imunologia , Dermatan Sulfato/imunologia , Lipoproteínas/genética , Lipoproteínas/imunologia , Doença de Lyme/genética , Doença de Lyme/imunologia , Doença de Lyme/microbiologia , Mamíferos , Camundongos , Microvasos/imunologia , Microvasos/microbiologia , Resistência ao Cisalhamento
2.
Nat Commun ; 12(1): 6664, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795263

RESUMO

Crohn's disease is an inflammatory disease of the gastrointestinal tract characterized by an aberrant response to microbial and environmental triggers. This includes an altered microbiome dominated by Enterobacteriaceae and in particular adherent-invasive E. coli (AIEC). Clinical evidence implicates periods of psychological stress in Crohn's disease exacerbation, and disturbances in the gut microbiome might contribute to the pathogenic mechanism. Here we show that stress-exposed mice develop ileal dysbiosis, dominated by the expansion of Enterobacteriaceae. In an AIEC colonisation model, stress-induced glucocorticoids promote apoptosis of CD45+CD90+ cells that normally produce IL-22, a cytokine that is essential for the maintenance of ileal mucosal barrier integrity. Blockade of glucocorticoid signaling or administration of recombinant IL-22 restores mucosal immunity, prevents ileal dysbiosis, and blocks AIEC expansion. We conclude that psychological stress impairs IL-22-driven protective immunity in the gut, which creates a favorable niche for the expansion of pathobionts that have been implicated in Crohn's disease. Importantly, this work also shows that immunomodulation can counteract the negative effects of psychological stress on gut immunity and hence disease-associated dysbiosis.


Assuntos
Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Estresse Psicológico/imunologia , Animais , Aderência Bacteriana/imunologia , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Disbiose/microbiologia , Enterobacteriaceae/classificação , Enterobacteriaceae/genética , Enterobacteriaceae/imunologia , Escherichia coli/imunologia , Escherichia coli/fisiologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Íleo/imunologia , Íleo/microbiologia , Íleo/patologia , Interleucinas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Receptores de Glucocorticoides/imunologia , Receptores de Glucocorticoides/metabolismo , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismo
3.
Immunohorizons ; 5(10): 870-883, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702763

RESUMO

Citrobacter rodentium is a murine pathogenic bacterium that adheres to intestinal epithelial cells, resulting in loss of microvilli and pedestal formation, and alters multiple cellular processes, including actin dynamics. Translocated intimin receptor (Tir), one of its virulence factors, functions as receptor for intimin, a bacterial adhesin, thereby mediating bacterial adhesion to epithelial cells. Although robust immune responses are induced to eliminate pathogenic bacteria in the host, they are suppressed against harmless commensal bacteria. The mechanism(s) underlying such a differentiation remains unclear. This study sought to determine the roles of intimate adhesion in the induction of specific immune responses upon C. rodentium infection. To this end, microbiota-depleted mice were infected with the Tir-F strain expressing full-length Tir or mutant strains expressing the C-terminal truncated Tir that is defective in intimin binding and host cell actin polymerization. There were no differences in the colonization kinetics and Abs responses against C. rodentium LPS among the strains, whereas Abs against the virulence factors were only produced on Tir-F infection. Although there were no differences in the virulence factors mRNA expression levels, colonic hyperplasia, and bacterial translocation to the systemic organs irrespective of the strain, adhesion to colonic epithelial cells was reduced in the mutant strain-infected mice. Furthermore, transcriptomic analysis indicated that robust inflammatory and immune responses were only induced in the Tir-F-infected group and were suppressed in the mutant-infected groups. Taken together, these findings suggest that Tir-mediated intimate adhesion induces inflammatory and immune responses, resulting in the induction of virulence factor-specific Abs.


Assuntos
Aderência Bacteriana/imunologia , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Mucosa Intestinal/patologia , Fatores de Virulência/metabolismo , Adesinas Bacterianas/metabolismo , Animais , Aderência Bacteriana/genética , Linhagem Celular Tumoral , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Feminino , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Mutação , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Organismos Livres de Patógenos Específicos
4.
mBio ; 12(3): e0017321, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34154415

RESUMO

Bacteria that colonize animals must overcome, or coexist, with the reactive oxygen species products of inflammation, a front-line defense of innate immunity. Among these is the neutrophilic oxidant bleach, hypochlorous acid (HOCl), a potent antimicrobial that plays a primary role in killing bacteria through nonspecific oxidation of proteins, lipids, and DNA. Here, we report that in response to increasing HOCl levels, Escherichia coli regulates biofilm production via activation of the diguanylate cyclase DgcZ. We identify the mechanism of DgcZ sensing of HOCl to be direct oxidation of its regulatory chemoreceptor zinc-binding (CZB) domain. Dissection of CZB signal transduction reveals that oxidation of the conserved zinc-binding cysteine controls CZB Zn2+ occupancy, which in turn regulates the catalysis of c-di-GMP by the associated GGDEF domain. We find DgcZ-dependent biofilm formation and HOCl sensing to be regulated in vivo by the conserved zinc-coordinating cysteine. Additionally, point mutants that mimic oxidized CZB states increase total biofilm. A survey of bacterial genomes reveals that many pathogenic bacteria that manipulate host inflammation as part of their colonization strategy possess CZB-regulated diguanylate cyclases and chemoreceptors. Our findings suggest that CZB domains are zinc-sensitive regulators that allow host-associated bacteria to perceive host inflammation through reactivity with HOCl. IMPORTANCE Immune cells are well equipped to eliminate invading bacteria, and one of their primary tools is the synthesis of bleach, hypochlorous acid (HOCl), the same chemical used as a household disinfectant. In this work, we present findings showing that many host-associated bacteria possess a bleach-sensing protein that allows them to adapt to the presence of this chemical in their environment. We find that the bacterium Escherichia coli responds to bleach by hunkering down and producing a sticky matrix known as biofilm, which helps it aggregate and adhere to surfaces. This behavior may play an important role in pathogenicity for E. coli and other bacteria, as it allows the bacteria to detect and adapt to the weapons of the host immune system.


Assuntos
Aderência Bacteriana/genética , Biofilmes/crescimento & desenvolvimento , GMP Cíclico/análogos & derivados , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Inflamação/genética , Transdução de Sinais , Bactérias/classificação , Bactérias/genética , Bactérias/imunologia , Bactérias/metabolismo , Aderência Bacteriana/imunologia , Biofilmes/efeitos dos fármacos , GMP Cíclico/genética , GMP Cíclico/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/imunologia , Escherichia coli/metabolismo , Proteínas de Escherichia coli/imunologia , Proteínas de Escherichia coli/metabolismo , Genoma Bacteriano , Ácido Hipocloroso/farmacologia , Inflamação/imunologia
5.
Dev Comp Immunol ; 122: 104108, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33909995

RESUMO

BLAST searches against databases for the bullfrog (Rana catesbeiana), using the collectin sequence previously identified in tadpoles, revealed the presence of at least 20 members of the collectin gene family. Phylogenetic analysis demonstrated that the bullfrog possesses expanded gene subfamilies encoding mannose-binding lectin (MBL) and pulmonary surfactant-associated protein D (PSAPD). Two collectins, of 20 kDa (PSAPD1) and 25 kDa (PSAPD6), were purified as a mixture from adult bullfrog plasma using affinity chromatography. These collectins were present as an oligomer of ~400 kDa in their native state, and showed Ca2+-dependent carbohydrate binding with different sugar preferences. Affinity-purified collectins showed weak E. coli agglutination and bactericidal activities, compared with those of plasma. Although both PSAPD1 and PSAPD6 genes were predominantly expressed in the liver, PSAPD1 transcripts were abundant in adults whereas PSAPD6 transcripts were abundant in tadpoles. The findings indicate that two gene subfamilies in the collectin family have diverged structurally, functionally and transcriptionally in the bullfrog. Rapid expansion of the collectin family in bullfrogs may reflect the onset of sub-functionalization of the prototype MBL gene towards tetrapod MBL and PSAPDs, and may be one means of natural adaptation in the innate immune system to various pathogens in both aquatic and terrestrial environments.


Assuntos
Carboidratos/imunologia , Imunidade Inata/imunologia , Lectina de Ligação a Manose/sangue , Proteína D Associada a Surfactante Pulmonar/sangue , Rana catesbeiana/metabolismo , Aglutinação/imunologia , Animais , Aderência Bacteriana/imunologia , Metabolismo dos Carboidratos/imunologia , Colectinas/sangue , Colectinas/genética , Colectinas/metabolismo , Escherichia coli/imunologia , Imunidade Inata/genética , Larva/imunologia , Lectina de Ligação a Manose/genética , Lectina de Ligação a Manose/metabolismo , Filogenia , Proteína D Associada a Surfactante Pulmonar/genética , Proteína D Associada a Surfactante Pulmonar/metabolismo
6.
Biochem Soc Trans ; 49(2): 617-627, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33704415

RESUMO

The human skin microbiota forms a key barrier against skin pathogens and is important in modulating immune responses. Recent studies identify lactobacilli as endogenous inhabitants of healthy skin, while inflammatory skin conditions are often associated with a disturbed skin microbiome. Consequently, lactobacilli-based probiotics are explored as a novel treatment of inflammatory skin conditions through their topical skin application. This review focuses on the potential beneficial role of lactobacilli (family Lactobacillaceae) in the skin habitat, where they can exert multifactorial local mechanisms of action against pathogens and inflammation. On one hand, lactobacilli have been shown to directly compete with skin pathogens through adhesion inhibition, production of antimicrobial metabolites, and by influencing pathogen metabolism. The competitive anti-pathogenic action of lactobacilli has already been described mechanistically for common different skin pathogens, such as Staphylococcus aureus, Cutibacterium acnes, and Candida albicans. On the other hand, lactobacilli also have an immunomodulatory capacity associated with a reduction in excessive skin inflammation. Their influence on the immune system is mediated by bacterial metabolites and cell wall-associated or excreted microbe-associated molecular patterns (MAMPs). In addition, lactobacilli can also enhance the skin barrier function, which is often disrupted as a result of infection or in inflammatory skin diseases. Some clinical trials have already translated these mechanistic insights into beneficial clinical outcomes, showing that topically applied lactobacilli can temporarily colonize the skin and promote skin health, but more and larger clinical trials are required to generate in vivo mechanistic insights and in-depth skin microbiome analysis.


Assuntos
Antibiose/imunologia , Candida albicans/imunologia , Inflamação/imunologia , Lactobacillus/imunologia , Pele/imunologia , Staphylococcus aureus/imunologia , Antibiose/fisiologia , Aderência Bacteriana/imunologia , Bacteriocinas/imunologia , Bacteriocinas/metabolismo , Candida albicans/fisiologia , Humanos , Sistema Imunitário/imunologia , Sistema Imunitário/microbiologia , Inflamação/microbiologia , Lactobacillus/metabolismo , Lactobacillus/fisiologia , Pele/microbiologia , Pele/patologia , Staphylococcus aureus/fisiologia
7.
Methods Mol Biol ; 2291: 253-272, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33704757

RESUMO

Therapeutic antibodies (Abs) inhibiting bacterial adhesion to host epithelia are an attractive option to reduce the load of Shiga toxin-producing E. coli (STEC) in the intestine of the patient and also in the bovine reservoir, thereby minimizing the risk of STEC contamination in the food chain. Of particular interest are recombinant single-domain Ab fragments called nanobodies (Nbs) derived from the variable domain of camelid heavy chain-only antibodies (VHH). The outer membrane adhesin intimin and the translocated intimin receptor (Tir) are essential for the attachment of STEC to host epithelia. In addition, EspA filaments of the bacterial type III protein secretion system are needed for Tir translocation into the host cell. Given their importance for bacterial adhesion and colonization, we developed Nbs against intimin, Tir and EspA proteins of STEC serotype O157:H7. Here, we report the screening methods used to isolate inhibitory Nbs blocking intimin-Tir protein-protein interaction, actin-pedestal formation, and intimate adhesion of STEC to epithelial cells in vitro. First, we describe how VHH gene repertoires can be produced as Nbs secreted by E. coli using the α-hemolysin (HlyA) protein secretion system. Next, we report the methods for identification of inhibitors of intimin-Tir protein-protein interaction and of STEC intimate adhesion to HeLa cells in culture. These methods can be adapted for the screening of Nbs against different adhesin-receptor complexes to block the adhesion of other pathogens to host cells.


Assuntos
Adesinas Bacterianas/imunologia , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/imunologia , Células Epiteliais , Escherichia coli O157/imunologia , Proteínas de Escherichia coli/imunologia , Receptores de Superfície Celular/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Bovinos , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Escherichia coli O157/patogenicidade , Humanos
8.
Chembiochem ; 22(12): 2124-2133, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33755306

RESUMO

Group B Streptococcus (GBS) is an encapsulated Gram-positive human pathogen that causes invasive infections in pregnant hosts and neonates, as well as immunocompromised individuals. Colonization of the human host requires the ability to adhere to mucosal surfaces and circumnavigate the nutritional challenges and antimicrobial defenses associated with the innate immune response. Biofilm formation is a critical process to facilitate GBS survival and establishment of a replicative niche in the vertebrate host. Previous work has shown that the host responds to GBS infection by producing the innate antimicrobial glycoprotein lactoferrin, which has been implicated in repressing bacterial growth and biofilm formation. Additionally, lactoferrin is highly abundant in human breast milk and could serve a protective role against invasive microbial pathogens. This study demonstrates that human breast milk lactoferrin has antimicrobial and anti-biofilm activity against GBS and inhibits its adherence to human gestational membranes. Together, these results indicate that human milk lactoferrin could be used as a prebiotic chemotherapeutic strategy to limit the impact of bacterial adherence and biofilm formation on GBS-associated disease outcomes.


Assuntos
Antibacterianos/farmacologia , Lactoferrina/imunologia , Leite Humano/química , Streptococcus agalactiae/efeitos dos fármacos , Antibacterianos/química , Aderência Bacteriana/efeitos dos fármacos , Aderência Bacteriana/imunologia , Biofilmes/efeitos dos fármacos , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Lactoferrina/química , Testes de Sensibilidade Microbiana , Streptococcus agalactiae/imunologia
9.
Front Immunol ; 12: 766970, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095842

RESUMO

In the lumen of blood vessels, there are large numbers of erythrocytes, which are approximately 95% of the total blood cells. Although the function of erythrocytes is to transport oxygen in the organism, recent studies have shown that mammalian and teleost erythrocytes are involved in the immune response against bacterial infections. However, the immune mechanisms used by avian erythrocytes are not yet clear. Here, we demonstrated that erythrocytes from goose have the ability to phagocytose as well as conduct antimicrobial activity. Firstly, we revealed the phagocytosis or adhesion activity of goose erythrocytes for latex beads 0.1-1.0 µm in diameter by fluorescence microscopy, and scanning and transmission electron microscopy. The low cytometry results also proved that goose erythrocytes had a wide range of phagocytic or adhesion activity for different bacteria. Followed, the low cytometry analysis data further explored that the goose erythrocytes contain the ability to produce reactive oxygen species (ROS) and inducible nitric oxide synthase (iNOS) in response to bacterial stimulation, and also up-regulated the expression of NOX family includes NOX1 and NOX5. Finally, we also found that goose erythrocytes showed a powerful antibacterial activity against all the three bacteria, meanwhile the stimulation of three kinds of bacteria up-regulated the expression of inflammatory factors, and increased the production of antioxidant enzymes to protect the cells from oxidative damage. Herein, our results demonstrate that goose Erythrocytes possess a certain phagocytic capacity and antioxidant system, and that the antimicrobial activity of erythrocytes can occurred through the production of unique respiratory burst against foreign pathogenic bacteria, which provides new clues to the interaction between bacteria and avian erythrocytes.


Assuntos
Antibacterianos/imunologia , Eritrócitos/imunologia , Gansos/imunologia , Fagocitose/imunologia , Explosão Respiratória/imunologia , Animais , Antioxidantes/metabolismo , Bactérias/imunologia , Aderência Bacteriana/imunologia , Imunidade/imunologia , Inflamação/imunologia , Estresse Oxidativo/imunologia , Fagócitos/imunologia , Espécies Reativas de Oxigênio/imunologia
10.
J Microbiol Immunol Infect ; 54(3): 420-428, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31706823

RESUMO

BACKGROUND/PURPOSE: SraP is a serine-rich repeat protein (SRRP) from Staphylococcus aureus that binds to sialylated receptors to promote bacterial adhesion to and invasion into host epithelial cells, mediated by the l-lectin module of its ligand-binding region. METHODS: The sequence encoding the L-lectin module of SraP was inserted into pET28a plasmid, and the recombinant protein was purified by His label affinity chromatography. A monoclonal antibody (mAb) against the l-lectin module was obtained and confirmed by enzyme-linked immunosorbent assay and western blotting. The effect of the mAb on S. aureus adhesion and invasion was assessed in A549 cells and mice subjected to S. aureus challenge. RESULTS: We successfully obtained a mAb against the l-lectin module of SraP. Pre-incubation with the mAb dramatically inhibited the bacteria's ability to adhere to and invade A549 cells. Moreover, mice administered mAb through tail vein injection had significantly fewer bacteria in the blood. CONCLUSION: The anti-SraPL-Lectin mAb significantly reduced the adherence and invasion of S. aureus to host cells. This study lays the foundation for the future development of the l-lectin module of SraP as a target for the prevention and treatment of S. aureus infection. Our findings suggest that specific subdomains of SRRPs may represent potential antibacterial drug targets for intervention.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Aderência Bacteriana/imunologia , Proteínas de Bactérias/imunologia , Lectinas/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/química , Staphylococcus aureus/imunologia , Células A549 , Animais , Anticorpos Monoclonais/uso terapêutico , Proteínas de Bactérias/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Infecções Estafilocócicas/prevenção & controle
11.
Gastroenterology ; 160(4): 1301-1314.e8, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33227279

RESUMO

BACKGROUND & AIMS: Although Clostridioides difficile infection (CDI) is known to involve the disruption of the gut microbiota, little is understood regarding how mucus-associated microbes interact with C difficile. We hypothesized that select mucus-associated bacteria would promote C difficile colonization and biofilm formation. METHODS: To create a model of the human intestinal mucus layer and gut microbiota, we used bioreactors inoculated with healthy human feces, treated with clindamycin and infected with C difficile with the addition of human MUC2-coated coverslips. RESULTS: C difficile was found to colonize and form biofilms on MUC2-coated coverslips, and 16S rRNA sequencing showed a unique biofilm profile with substantial cocolonization with Fusobacterium species. Consistent with our bioreactor data, publicly available data sets and patient stool samples showed that a subset of patients with C difficile infection harbored high levels of Fusobacterium species. We observed colocalization of C difficile and F nucleatum in an aggregation assay using adult patients and stool of pediatric patients with inflammatory bowel disease and in tissue sections of patients with CDI. C difficile strains were found to coaggregate with F nucleatum subspecies in vitro; an effect that was inhibited by blocking or mutating the adhesin RadD on Fusobacterium and removal of flagella on C difficile. Aggregation was shown to be unique between F nucleatum and C difficile, because other gut commensals did not aggregate with C difficile. Addition of F nucleatum also enhanced C difficile biofilm formation and extracellular polysaccharide production. CONCLUSIONS: Collectively, these data show a unique interaction of between pathogenic C difficile and F nucleatum in the intestinal mucus layer.


Assuntos
Adesinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidade , Infecções por Clostridium/imunologia , Fusobacterium nucleatum/imunologia , Microbioma Gastrointestinal/imunologia , Adesinas Bacterianas/genética , Aderência Bacteriana/imunologia , Biofilmes , Reatores Biológicos/microbiologia , Clostridioides difficile/genética , Clostridioides difficile/imunologia , Clostridioides difficile/metabolismo , Infecções por Clostridium/microbiologia , Fezes/microbiologia , Flagelos/genética , Flagelos/metabolismo , Fusobacterium nucleatum/metabolismo , Células HT29 , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Mucina-2/metabolismo
12.
Nihon Saikingaku Zasshi ; 75(2): 185-194, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-33361654

RESUMO

Countless numbers of bacteria inhabit the intestinal tract. One of the important functions of gut microbiota is the "colonization resistance" against infection by pathogenic microorganisms. However, detailed mechanism of the colonization resistance of intestinal bacteria is still largely unknown. We tried to identify molecular and cellular mechanism of it and found that antigen presentation by dendritic cells is required for the induction of intestinal segmented filamentous bacteria (SFB)-induced T helper 17 (Th17) cells that contribute to the protection against infection by Citrobacter rodentium. We further identified that gut Th17 cells selectively recognize antigens derived from SFB. We also revealed that SFB induce α1,2-fucose, one of carbohydrate chains, expressed on the intestinal epithelial cells mediated by group 3 innate lymphoid cells. Epithelial α1,2-fucose protected against infection by pathogenic bacterium Salmonella typhimurium. Furthermore, it was found that intestinal bacteria inhibit colonization of the pathogenic fungus Candida albicans as well as pathogenic bacteria. From these studies, detailed mechanism of "colonization resistance" against pathogenic microorganisms by intestinal bacteria has been clarified.


Assuntos
Candida albicans/patogenicidade , Citrobacter rodentium/patogenicidade , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Sistema Imunitário/imunologia , Mucosa Intestinal/microbiologia , Salmonella typhimurium/patogenicidade , Células Th17/imunologia , Animais , Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Aderência Bacteriana/imunologia , Candida albicans/imunologia , Citrobacter rodentium/imunologia , Células Dendríticas/imunologia , Fucose/metabolismo , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Salmonella typhimurium/imunologia
13.
Front Immunol ; 11: 2192, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072084

RESUMO

During the last years probiotics gained the attention of clinicians for their use in the prevention and treatment of multiple diseases. Probiotics main mechanisms of action include enhanced mucosal barrier function, direct antagonism with pathogens, inhibition of bacterial adherence and invasion capacity in the intestinal epithelium, boosting of the immune system and regulation of the central nervous system. It is accepted that there is a mutual communication between the gut microbiota and the liver, the so-called "microbiota-gut-liver axis" as well as a reciprocal communication between the intestinal microbiota and the central nervous system through the "microbiota-gut-brain axis." Moreover, recently the "gut-lung axis" in bacterial and viral infections is considerably discussed for bacterial and viral infections, as the intestinal microbiota amplifies the alveolar macrophage activity having a protective role in the host defense against pneumonia. The importance of the normal human intestinal microbiota is recognized in the preservation of health. Disease states such as, infections, autoimmune conditions, allergy and other may occur when the intestinal balance is disturbed. Probiotics seem to be a promising approach to prevent and even reduce the symptoms of such clinical states as an adjuvant therapy by preserving the balance of the normal intestinal microbiota and improving the immune system. The present review states globally all different disorders in which probiotics can be given. To date, Stronger data in favor of their clinical use are provided in the prevention of gastrointestinal disorders, antibiotic-associated diarrhea, allergy and respiratory infections. We hereby discuss the role of probiotics in the reduction of the respiratory infection symptoms and we focus on the possibility to use them as an adjuvant to the therapeutic approach of the pandemic COVID-19. Nevertheless, it is accepted by the scientific community that more clinical studies should be undertaken in large samples of diseased populations so that the assessment of their therapeutic potential provide us with strong evidence for their efficacy and safety in clinical use.


Assuntos
Bactérias/imunologia , Betacoronavirus/imunologia , Infecções por Coronavirus , Microbioma Gastrointestinal/imunologia , Pandemias , Pneumonia Viral , Probióticos/uso terapêutico , Aderência Bacteriana/imunologia , Encéfalo/imunologia , Encéfalo/microbiologia , COVID-19 , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/microbiologia , Infecções por Coronavirus/terapia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Pneumonia Viral/epidemiologia , Pneumonia Viral/imunologia , Pneumonia Viral/microbiologia , Pneumonia Viral/terapia , SARS-CoV-2
14.
Nat Commun ; 11(1): 5188, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33057023

RESUMO

Mycoplasma pneumoniae is a bacterial human pathogen that causes primary atypical pneumonia. M. pneumoniae motility and infectivity are mediated by the immunodominant proteins P1 and P40/P90, which form a transmembrane adhesion complex. Here we report the structure of P1, determined by X-ray crystallography and cryo-electron microscopy, and the X-ray structure of P40/P90. Contrary to what had been suggested, the binding site for sialic acid was found in P40/P90 and not in P1. Genetic and clinical variability concentrates on the N-terminal domain surfaces of P1 and P40/P90. Polyclonal antibodies generated against the mostly conserved C-terminal domain of P1 inhibited adhesion of M. pneumoniae, and serology assays with sera from infected patients were positive when tested against this C-terminal domain. P40/P90 also showed strong reactivity against human infected sera. The architectural elements determined for P1 and P40/P90 open new possibilities in vaccine development against M. pneumoniae infections.


Assuntos
Adesinas Bacterianas/imunologia , Aderência Bacteriana/imunologia , Mycoplasma pneumoniae/imunologia , Pneumonia por Mycoplasma/imunologia , Adesinas Bacterianas/isolamento & purificação , Adesinas Bacterianas/ultraestrutura , Microscopia Crioeletrônica , Cristalografia por Raios X , Mycoplasma pneumoniae/isolamento & purificação , Mycoplasma pneumoniae/patogenicidade , Pneumonia por Mycoplasma/sangue , Pneumonia por Mycoplasma/microbiologia , Domínios Proteicos/imunologia
15.
PLoS One ; 15(9): e0238425, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32960889

RESUMO

OBJECTIVE: To evaluate the effects of Bifidobacterium animalis subsp. lactis HN019 (HN019) on clinical periodontal parameters (plaque accumulation and gingival bleeding), on immunocompetence of gingival tissues [expression of beta-defensin (BD)-3, toll-like receptor 4 (TLR4), cluster of differentiation(CD)-57 and CD-4], and on immunological properties of saliva (IgA levels) in non-surgical periodontal therapy in generalized chronic periodontitis (GCP) patients. Adhesion to buccal epithelial cells (BEC) and the antimicrobial properties of HN019 were also investigated. MATERIALS AND METHODS: Thirty patients were recruited and monitored clinically at baseline (before scaling and root planing-SRP) and after 30 and 90 days. Patients were randomly assigned to Test (SRP+Probiotic, n = 15) or Control (SRP+Placebo, n = 15) group. Probiotic lozenges were used for 30 days. Gingival tissues and saliva were immunologically analyzed. The adhesion of HN019 with or without Porphyromonas gingivalis in BEC and its antimicrobial properties were investigated in in vitro assays. Data were statistically analyzed (p<0.05). RESULTS: Test group presented lower plaque index (30 days) and lower marginal gingival bleeding (90 days) when compared with Control group. Higher BD-3, TLR4 and CD-4 expressions were observed in gingival tissues in Test group than in Control group. HN019 reduced the adhesion of P. gingivalis to BEC and showed antimicrobial potential against periodontopathogens. CONCLUSION: Immunological and antimicrobial properties of B. lactis HN019 make it a potential probiotic to be used in non-surgical periodontal therapy of patients with GCP. CLINICAL RELEVANCE: B. lactis HN019 may be a potential probiotic to improve the effects of non-surgical periodontal therapy. Name of the registry and registration number (ClinicalTrials.gov): "Effects of probiotic therapy in the treatment of periodontitis"-NCT03408548.


Assuntos
Bifidobacterium animalis/imunologia , Periodontite Crônica/terapia , Probióticos/uso terapêutico , Adulto , Aderência Bacteriana/imunologia , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/terapia , Periodontite Crônica/imunologia , Periodontite Crônica/microbiologia , Método Duplo-Cego , Feminino , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunoglobulina A Secretora/metabolismo , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/imunologia , Mucosa Bucal/microbiologia , Porphyromonas gingivalis/patogenicidade , Saliva/imunologia
16.
Nat Commun ; 11(1): 3545, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669564

RESUMO

Group A Streptococcus (GAS) infection causes a range of diseases, but vaccine development is hampered by the high number of serotypes. Here, using reverse vaccinology the authors identify SPy_2191 as a cross-protective vaccine candidate. From 18 initially identified surface proteins, only SPy_2191 is conserved, surface-exposed and inhibits both GAS adhesion and invasion. SPy_2191 immunization in mice generates bactericidal antibodies resulting in opsonophagocytic killing of prevalent and invasive GAS serotypes of different geographical regions, including M1 and M49 (India), M3.1 (Israel), M1 (UK) and M1 (USA). Resident splenocytes show higher interferon-γ and tumor necrosis factor-α secretion upon antigen re-stimulation, suggesting activation of cell-mediated immunity. SPy_2191 immunization significantly reduces streptococcal load in the organs and confers ~76-92% protection upon challenge with invasive GAS serotypes. Further, it significantly suppresses GAS pharyngeal colonization in mice mucosal infection model. Our findings suggest that SPy_2191 can act as a universal vaccine candidate against GAS infections.


Assuntos
Proteínas de Bactérias/imunologia , Proteção Cruzada/imunologia , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Animais , Aderência Bacteriana/imunologia , Linhagem Celular , Clonagem Molecular , Modelos Animais de Doenças , Feminino , Humanos , Imunogenicidade da Vacina , Camundongos , Testes de Neutralização , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Sorogrupo , Infecções Estreptocócicas/microbiologia , Vacinas Estreptocócicas/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
17.
mBio ; 11(4)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32694142

RESUMO

Streptococcus pyogenes (group A Streptococcus [GAS]) is an important human pathogen causing a broad spectrum of diseases and associated with significant global morbidity and mortality. Almost all GAS isolates express a surface hyaluronic acid capsule, a virulence determinant that facilitates host colonization and impedes phagocyte killing. However, recent epidemiologic surveillance has reported a sustained increase in both mucosal and invasive infections caused by nonencapsulated GAS, which questions the indispensable role of hyaluronic acid capsule in GAS pathogenesis. In this study, we found that pilus of M4 GAS not only significantly promotes biofilm formation, adherence, and cytotoxicity to human upper respiratory tract epithelial cells and keratinocytes, but also promotes survival in human whole blood and increased virulence in murine models of invasive infection. T4 antigen, the pilus backbone protein of M4 GAS, binds haptoglobin, an abundant human acute-phase protein upregulated upon infection and inflammation, on the bacterial surface. Haptoglobin sequestration reduces the susceptibility of nonencapsulated M4 GAS to antimicrobial peptides released from activated neutrophils and platelets. Our results reveal a previously unappreciated virulence-promoting role of M4 GAS pili, in part mediated by co-opting the biology of haptoglobin to mitigate host antimicrobial defenses.IMPORTANCE Group A Streptococcus (GAS) is a strict human pathogen causing more than 700 million infections globally each year. The majority of the disease-causing GAS are encapsulated, which greatly guarantees survival and dissemination in the host. Emergence of the capsule-negative GAS, such as M4 GAS, in recent epidemiologic surveillance alarms the necessity to elucidate the virulence determinants of these pathogens. Here, we found that M4 pili play an important role in promoting M4 GAS adherence and cytotoxicity to human pharyngeal epithelial cells and keratinocytes. The same molecule also significantly enhanced M4 GAS survival and replication in human whole blood and experimental murine infection. T4 antigen, which composes the backbone of M4 pili, was able to sequester the very abundant serum protein haptoglobin to further confer M4 GAS resistance to antibacterial substances released by neutrophils and platelets.


Assuntos
Proteínas de Bactérias/metabolismo , Fímbrias Bacterianas/imunologia , Evasão da Resposta Imune , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/patogenicidade , Animais , Aderência Bacteriana/imunologia , Biofilmes/crescimento & desenvolvimento , Células Sanguíneas/microbiologia , Feminino , Fímbrias Bacterianas/classificação , Células HaCaT , Haptoglobinas/metabolismo , Humanos , Queratinócitos/microbiologia , Camundongos , Camundongos Endogâmicos ICR , Neutrófilos/microbiologia , Fenótipo , Infecções Estreptocócicas/sangue , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Virulência , Fatores de Virulência/metabolismo
18.
Infect Immun ; 88(9)2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32540869

RESUMO

Human rhinovirus (hRV) is frequently detected in the upper respiratory tract, and symptomatic infection is associated with an increased nasopharyngeal bacterial load, with subsequent development of secondary bacterial diseases. Nontypeable Haemophilus influenzae (NTHI) is a commensal bacterial species of the human nasopharynx; however, in the context of prior or concurrent upper respiratory tract viral infection, this bacterium commonly causes multiple diseases throughout the upper and lower respiratory tracts. The present study was conducted to determine the mechanism(s) by which hRV infection promotes the development of NTHI-induced diseases. We showed that hRV infection of polarized primary human airway epithelial cells resulted in increased adherence of NTHI, due in part to augmented expression of CEACAM1 and ICAM1, host cell receptors to which NTHI binds via engagement of multiple adhesins. Antibody blockade of these host cell receptors significantly reduced NTHI adherence. With a specific focus on the NTHI type IV pilus (T4P), which we have previously shown binds to ICAM1, an essential adhesin and virulence determinant, we next showed that T4P-directed antibody blockade significantly reduced NTHI adherence to hRV-infected airway cells and, further, that expression of this adhesin was required for the enhanced adherence observed. Collectively, these data provide a mechanism by which "the common cold" promotes diseases due to NTHI, and they add further support for the use of PilA (the majority subunit of T4P) as a vaccine antigen, since antibodies directed against PilA are expected to limit the notably increased bacterial load associated with hRV coinfection and thereby to prevent secondary NTHI-induced diseases of the respiratory tract.


Assuntos
Adesinas Bacterianas/imunologia , Aderência Bacteriana/imunologia , Células Epiteliais/imunologia , Proteínas de Fímbrias/imunologia , Haemophilus influenzae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Rhinovirus/imunologia , Adesinas Bacterianas/genética , Anticorpos Neutralizantes/farmacologia , Antígenos CD/genética , Antígenos CD/imunologia , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Proteínas de Fímbrias/genética , Regulação da Expressão Gênica/imunologia , Haemophilus influenzae/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno/genética , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Cultura Primária de Células , Ligação Proteica , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Rhinovirus/crescimento & desenvolvimento , Transdução de Sinais
19.
Methods Mol Biol ; 2136: 271-278, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32430828

RESUMO

The critical first step of Group A Streptococcus (GAS) pathogenesis is adhesion to the host pharyngeal and skin epithelial cell surfaces (Brouwer et al., FEBS Lett 590:3739-3757, 2016). Host-cell adhesion assays provide a straightforward model to study these host-pathogen interactions. Here, we describe the culturing of immortalized cell lines into monolayers to mimic host epithelia. Various GAS strains can then be added to study their adhesion properties. In addition, we describe the use of antibodies raised against the cell-surface components of GAS to study if these are able to neutralize the binding of GAS to the cell lines. This provides an indication if these cell-surface components are involved in adhesion and if antibodies generated against them function through neutralization.


Assuntos
Aderência Bacteriana/imunologia , Aderência Bacteriana/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Proteínas de Bactérias/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Epitélio/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Modelos Biológicos , Faringe , Pele/metabolismo , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/metabolismo
20.
J Clin Invest ; 130(2): 927-941, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31687974

RESUMO

Successful infection by mucosal pathogens requires overcoming the mucus barrier. To better understand this key step, we performed a survey of the interactions between human respiratory mucus and the human pathogen Streptococcus pneumoniae. Pneumococcal adherence to adult human nasal fluid was seen only by isolates expressing pilus-1. Robust binding was independent of pilus-1 adhesive properties but required Fab-dependent recognition of RrgB, the pilus shaft protein, by naturally acquired secretory IgA (sIgA). Pilus-1 binding by specific sIgA led to bacterial agglutination, but adherence required interaction of agglutinated pneumococci and entrapment in mucus particles. To test the effect of these interactions in vivo, pneumococci were preincubated with human sIgA before intranasal challenge in a mouse model of colonization. sIgA treatment resulted in rapid immune exclusion of pilus-expressing pneumococci. Our findings predict that immune exclusion would select for nonpiliated isolates in individuals who acquired RrgB-specific sIgA from prior episodes of colonization with piliated strains. Accordingly, genomic data comparing isolates carried by mothers and their children showed that mothers are less likely to be colonized with pilus-expressing strains. Our study provides a specific example of immune exclusion involving naturally acquired antibody in the human host, a major factor driving pneumococcal adaptation.


Assuntos
Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/imunologia , Proteínas de Bactérias/imunologia , Imunoglobulina A Secretora/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Mucosa Nasal , Streptococcus pneumoniae/imunologia , Humanos , Mucosa Nasal/imunologia , Mucosa Nasal/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...