Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 139(3): 485-98, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19836068

RESUMO

The gastrointestinal tract of mammals is inhabited by hundreds of distinct species of commensal microorganisms that exist in a mutualistic relationship with the host. How commensal microbiota influence the host immune system is poorly understood. We show here that colonization of the small intestine of mice with a single commensal microbe, segmented filamentous bacterium (SFB), is sufficient to induce the appearance of CD4(+) T helper cells that produce IL-17 and IL-22 (Th17 cells) in the lamina propria. SFB adhere tightly to the surface of epithelial cells in the terminal ileum of mice with Th17 cells but are absent from mice that have few Th17 cells. Colonization with SFB was correlated with increased expression of genes associated with inflammation and antimicrobial defenses and resulted in enhanced resistance to the intestinal pathogen Citrobacter rodentium. Thus, manipulation of this commensal-regulated pathway may provide new opportunities for enhancing mucosal immunity and treating autoimmune disease.


Assuntos
Bactérias Gram-Positivas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Diferenciação Celular , Citrobacter rodentium/imunologia , Bactérias Gram-Positivas/fisiologia , Imunidade nas Mucosas/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Mucosa Intestinal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/imunologia , Mucosa/microbiologia , Proteína Amiloide A Sérica/metabolismo , Organismos Livres de Patógenos Específicos , Simbiose , Interleucina 22
2.
Appl Environ Microbiol ; 86(13)2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32332136

RESUMO

Immunoglobulin A (IgA) is essential for defense of the intestinal mucosa against harmful pathogens. Previous studies have shown that Bacteroidetes, the major phylum of gut microbiota together with Firmicutes, impact IgA production. However, the relative abundances of species of Bacteroidetes responsible for IgA production were not well understood. In the present study, we identified some specific Bacteroidetes species that were associated with gut IgA induction by hsp60-based profiling of species distribution among Bacteroidetes The levels of IgA and the expression of the gene encoding activation-induced cytidine deaminase (AID) in the large intestine lamina propria, which is crucial for class switch recombination from IgM to IgA, were increased in soluble high-fiber diet (sHFD)-fed mice. We found that Bacteroides acidifaciens was the most abundant Bacteroidetes species in both sHFD- and normal diet-fed mice. In addition, the gut IgA levels were associated with the relative abundance of Bacteroides fragilis group species such as Bacteroides faecis, Bacteroides caccae, and Bacteroides acidifaciens Conversely, the ratio of B. acidifaciens to other Bacteroidetes species was reduced in insoluble high-fiber diet fed- and no-fiber diet-fed mice. To investigate whether B. acidifaciens increases IgA production, we generated B. acidifaciens monoassociated mice and found increased gut IgA production and AID expression. Collectively, soluble dietary fiber increases the ratio of gut Bacteroides fragilis group, such as B. acidifaciens, and IgA production. This might improve gut immune function, thereby protecting against bowel pathogens and reducing the incidence of inflammatory bowel diseases.IMPORTANCE Immunoglobulin A (IgA) is essential for defense of the intestinal mucosa against harmful pathogens. Gut microbiota impact IgA production, but the specific species responsible for IgA production remain largely elusive. Previous studies have shown that IgA and Bacteroidetes, the major phyla of gut microbiota, were increased in soluble high-fiber diet-fed mice. We show here that the levels of IgA in the gut and the expression of activation-induced cytidine deaminase (AID) in the large intestine lamina propria, which is crucial for class switch recombination from IgM to IgA, were correlated with the abundance of Bacteroides fragilis group species such as Bacteroides faecis, Bacteroides caccae, and Bacteroides acidifaciensB. acidifaciens monoassociated mice increased gut IgA production and AID expression. Soluble dietary fiber may improve gut immune function, thereby protecting against bowel pathogens and reducing inflammatory bowel diseases.


Assuntos
Bacteroides fragilis/fisiologia , Fibras na Dieta/metabolismo , Imunoglobulina A/biossíntese , Animais , Chaperonina 60 , Fibras na Dieta/administração & dosagem , Feminino , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais
3.
Surg Today ; 50(8): 920-930, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32062787

RESUMO

PURPOSE: Resistin-like molecule beta (RELMß) is a small cysteine-rich protein secreted by colonic epithelial cells. RELMß mRNA and protein expressions are dramatically induced by bacterial exposure in germ-free mice. We hypothesized that RELMß has antimicrobial activity. METHODS: The antimicrobial activity of RELMß was screened by an agar spot test and confirmed by a liquid broth test. The amount of RELMß in human stools was semi-quantified by Western blot analysis. The induction of RELMß mRNA and protein expression by bacteria was measured by quantitative RT-PCR using LS174T cells. Electron microscopic immunohistochemistry was performed using polyclonal anti-RELMß antibody. RESULTS: RELMß showed antimicrobial activity against S. aureus and all MRSAs examined in a dose- and pH-dependent fashion. Western blot study showed that the amount of RELMß in healthy human stools was comparable to that exhibiting antimicrobial activity in vitro. Both RELMß mRNA and protein expression were induced by heat-inactivated S. aureus, but not by E. coli in LS174T cells. Electron microscopic immunohistochemistry showed that RELMß bound to the cell surface of S. aureus, followed by destruction of the bacterial cytoplasm. CONCLUSIONS: RELMß is a colonic antimicrobial protein and its antibacterial activity is species selective. Because RELMß is abundant in healthy human stool, RELMß may modulate gut flora.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Farmacorresistência Bacteriana , Humanos , Testes de Sensibilidade Microbiana/métodos
4.
Immunity ; 29(2): 261-71, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18656387

RESUMO

Immunoglobulin A (IgA) is generated in the gut by both T cell-dependent and T cell-independent processes. The sites and the mechanisms for T cell-independent IgA synthesis remain elusive. Here we show that isolated lymphoid follicles (ILFs) were sites where induction of activation-induced cytidine deaminase (AID) and IgA class switching of B cells took place in the absence of T cells. We also show that formation of ILFs was regulated by interactions between lymphoid tissue-inducer cells expressing the nuclear receptor ROR gamma t (ROR gamma t(+)LTi cells) and stromal cells (SCs). Activation of SCs by ROR gamma t(+)LTi cells through lymphotoxin (LT)-beta receptor (LT beta R) and simultaneously by bacteria through TLRs induced recruitment of dendritic cells (DCs) and B cells and formation of ILFs. These findings provide insight into the crosstalk between bacteria, ROR gamma t(+)LTi cells, SCs, DCs, and B cells required for ILF formation and establish a critical role of ILFs in T cell-independent IgA synthesis in gut.


Assuntos
Linfócitos B/imunologia , Trato Gastrointestinal/imunologia , Imunoglobulina A/biossíntese , Tecido Linfoide/imunologia , Linfócitos T/imunologia , Animais , Linfócitos B/metabolismo , Comunicação Celular , Citidina Desaminase/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Trato Gastrointestinal/metabolismo , Imunoglobulina A/imunologia , Switching de Imunoglobulina , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Células Estromais/imunologia , Células Estromais/metabolismo , Linfócitos T/metabolismo
5.
Nature ; 469(7331): 543-7, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21270894

RESUMO

The human gut is colonized with a wide variety of microorganisms, including species, such as those belonging to the bacterial genus Bifidobacterium, that have beneficial effects on human physiology and pathology. Among the most distinctive benefits of bifidobacteria are modulation of host defence responses and protection against infectious diseases. Nevertheless, the molecular mechanisms underlying these effects have barely been elucidated. To investigate these mechanisms, we used mice associated with certain bifidobacterial strains and a simplified model of lethal infection with enterohaemorrhagic Escherichia coli O157:H7, together with an integrated 'omics' approach. Here we show that genes encoding an ATP-binding-cassette-type carbohydrate transporter present in certain bifidobacteria contribute to protecting mice against death induced by E. coli O157:H7. We found that this effect can be attributed, at least in part, to increased production of acetate and that translocation of the E. coli O157:H7 Shiga toxin from the gut lumen to the blood was inhibited. We propose that acetate produced by protective bifidobacteria improves intestinal defence mediated by epithelial cells and thereby protects the host against lethal infection.


Assuntos
Acetatos/metabolismo , Bifidobacterium/metabolismo , Infecções por Escherichia coli/prevenção & controle , Escherichia coli O157/fisiologia , Animais , Bifidobacterium/genética , Chlorocebus aethiops , Infecções por Escherichia coli/microbiologia , Perfilação da Expressão Gênica , Genoma Bacteriano , Camundongos , Dados de Sequência Molecular , Células Vero
6.
Nature ; 462(7270): 226-30, 2009 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19907495

RESUMO

The mucosal immune system forms the largest part of the entire immune system, containing about three-quarters of all lymphocytes and producing grams of secretory IgA daily to protect the mucosal surface from pathogens. To evoke the mucosal immune response, antigens on the mucosal surface must be transported across the epithelial barrier into organized lymphoid structures such as Peyer's patches. This function, called antigen transcytosis, is mediated by specialized epithelial M cells. The molecular mechanisms promoting this antigen uptake, however, are largely unknown. Here we report that glycoprotein 2 (GP2), specifically expressed on the apical plasma membrane of M cells among enterocytes, serves as a transcytotic receptor for mucosal antigens. Recombinant GP2 protein selectively bound a subset of commensal and pathogenic enterobacteria, including Escherichia coli and Salmonella enterica serovar Typhimurium (S. Typhimurium), by recognizing FimH, a component of type I pili on the bacterial outer membrane. Consistently, these bacteria were colocalized with endogenous GP2 on the apical plasma membrane as well as in cytoplasmic vesicles in M cells. Moreover, deficiency of bacterial FimH or host GP2 led to defects in transcytosis of type-I-piliated bacteria through M cells, resulting in an attenuation of antigen-specific immune responses in Peyer's patches. GP2 is therefore a previously unrecognized transcytotic receptor on M cells for type-I-piliated bacteria and is a prerequisite for the mucosal immune response to these bacteria. Given that M cells are considered a promising target for oral vaccination against various infectious diseases, the GP2-dependent transcytotic pathway could provide a new target for the development of M-cell-targeted mucosal vaccines.


Assuntos
Adesinas de Escherichia coli/metabolismo , Antígenos de Bactérias/metabolismo , Células Epiteliais/imunologia , Proteínas de Fímbrias/metabolismo , Imunidade nas Mucosas/imunologia , Glicoproteínas de Membrana/metabolismo , Nódulos Linfáticos Agregados/citologia , Adesinas de Escherichia coli/genética , Adesinas de Escherichia coli/imunologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Linhagem Celular , Células Epiteliais/metabolismo , Escherichia coli/imunologia , Escherichia coli/metabolismo , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/imunologia , Proteínas Ligadas por GPI , Glicoproteínas , Células HeLa , Humanos , Intestinos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/imunologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Especificidade por Substrato
7.
Biosci Biotechnol Biochem ; 78(10): 1669-76, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25273132

RESUMO

The intestine harbors a substantial number of commensal bacteria that provide considerable benefits to the host. Epidemiologic studies have identified associations between alterations in the composition of the intestinal microbiota and the development of allergic disease. However, the cellular and molecular mechanisms underlying these effects remain to be determined. Here, we show that heat-killed commensal bacteria suppressed degranulation of mast cells in vitro in a MyD88-independent manner. In particular, Enterococcus faecalis showed the strongest suppression of degranulation through partial inhibition of Ca(2+) signaling upon the high affinity IgE receptor (FcεRI) cross-linking.


Assuntos
Degranulação Celular , Enterococcus faecalis/fisiologia , Mastócitos/citologia , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Feminino , Espaço Intracelular/metabolismo , Camundongos , Transdução de Sinais
8.
J Immunol ; 184(9): 4646-53, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20304829

RESUMO

Naive T lymphocytes recirculate through the lymph-vascular system and enter and exit lymphoid organs. Using mice expressing the photoconvertible fluorescence protein Kaede, we demonstrated that naive T cells seek to interact with endogenous Ags after migrating to the lymphoid organs. The interaction with endogenous Ags transiently induces CD69 expression on T cells, which prolongs retention in the lymphoid organs. Cells that fail to express CD69 or lose CD69 expression migrate to other lymphoid organs. Functionally, CD69(+)-naive CD4(+) T cells exhibit faster and greater cytokine production than do CD69(-) naive CD4(+) T cells. These results indicate that CD4(+) T cells continuously migrate to interact with endogenous Ags, and such an interaction plays an important role in the Ag reactivity of naive CD4(+) T cells.


Assuntos
Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Lectinas Tipo C/biossíntese , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Regulação para Cima/imunologia , Animais , Antígenos CD/administração & dosagem , Antígenos CD/fisiologia , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Citocinas/biossíntese , Lectinas Tipo C/administração & dosagem , Lectinas Tipo C/deficiência , Lectinas Tipo C/fisiologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fase de Repouso do Ciclo Celular/imunologia , Timo/citologia , Timo/imunologia , Timo/metabolismo , Regulação para Cima/genética
9.
Int J Syst Evol Microbiol ; 61(Pt 4): 898-902, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20495031

RESUMO

A fructophilic lactic acid bacterium, designated strain F214-1(T), was isolated from a flower of Tropaeolum majus in South Africa. Based on phylogenetic analysis of 16S rRNA gene sequences, the strain formed a subcluster with Fructobacillus ficulneus and Fructobacillus pseudoficulneus and, based on recA gene sequences, the strain formed a subcluster with F. ficulneus. DNA-DNA hybridization studies showed that strain F214-1(T) was phylogenetically distinct from its closest relatives. Acid was produced from the fermentation of d-glucose, d-fructose and d-mannitol only. d-Fructose was the preferred sole carbon and energy source and was fermented more rapidly than d-glucose. Growth of the strain on d-glucose under anaerobic conditions was very weak but external electron acceptors such as oxygen and pyruvate enhanced growth on d-glucose. Lactic acid and acetic acid were produced from d-glucose in equimolar amounts. Ethanol was produced at very low levels, despite the strain's obligately heterofermentative metabolism. Based on these data, strain F214-1(T) represents a novel species of fructophilic bacteria in the genus Fructobacillus, for which the name Fructobacillus tropaeoli sp. nov. is proposed. The type strain is F214-1(T) ( = JCM 16675(T)  = DSM 23246(T)).


Assuntos
Frutose/metabolismo , Ácido Láctico/metabolismo , Leuconostocaceae/classificação , Leuconostocaceae/isolamento & purificação , Tropaeolum/microbiologia , Ácido Acético/metabolismo , Análise por Conglomerados , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Glucose/metabolismo , Leuconostocaceae/genética , Leuconostocaceae/metabolismo , Manitol/metabolismo , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Filogenia , RNA Ribossômico 16S/genética , Recombinases Rec A/genética , Análise de Sequência de DNA , África do Sul
10.
Immunobiology ; 226(2): 152056, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33535092

RESUMO

Mast cells, which express the high-affinity IgE receptor (FcεRI) on their surface, play a crucial role in inducing allergic inflammation. Since mast cells are activated by crosslinking of FcεRI with IgE and allergens, the cell surface expression level of FcεRI is an important factor in determining the sensitivity to allergens. Recently, the involvement of gut microbiota in the prevalence and regulation of allergy has attracted attention but the precise underlying mechanisms are not fully understood. In this study, the effect of intestinal bacteria on cell surface expression of FcεRI was examined. Bacteroides acidifaciens type A 43 specifically suppressed cell surface expression of FcεRI on mouse bone marrow-derived mast cells (BMMCs) without reduction in FcεRI α and ß-chain mRNA and total protein expression. The suppressive effect required sustained exposure to this bacterium, with a corresponding reduction in Erk activation. Inhibition of Erk decreased cell surface distribution of FcεRI in BMMCs, at least in part, through facilitated endocytosis of FcεRI. These results indicate that B. acidifaciens type A 43 suppresses cell surface expression of FcεRI on mast cells in a post-translational manner via inhibition of Erk. The suppression of FcεRI expression on mast cells by specific bacteria might be the underlying mechanism involved in the regulation of allergy by gut microbiota.


Assuntos
Bacteroides , Mastócitos/imunologia , Receptores de IgE/imunologia , Animais , Células Cultivadas , Feminino , Microbioma Gastrointestinal , Intestinos/microbiologia , Camundongos Endogâmicos C57BL , Processamento de Proteína Pós-Traducional , Receptores de IgE/genética
11.
J Bacteriol ; 192(4): 1165-6, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20008064

RESUMO

Escherichia coli SE15 (O150:H5) is a human commensal bacterium recently isolated from feces of a healthy adult and classified into E. coli phylogenetic group B2, which includes the majority of extraintestinal pathogenic E. coli. Here, we report the finished and annotated genome sequence of this organism.


Assuntos
DNA Bacteriano/genética , Escherichia coli/genética , Genoma Bacteriano , Análise de Sequência de DNA , Adulto , Técnicas de Tipagem Bacteriana , DNA Bacteriano/química , Escherichia coli/classificação , Escherichia coli/isolamento & purificação , Fezes/microbiologia , Genótipo , Humanos , Dados de Sequência Molecular , Adulto Jovem
12.
FASEB J ; 23(8): 2514-20, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19276171

RESUMO

The innate immune system plays an important role in protecting organs that are continuous with the outer surface of the body from bacterial infection. The antibacterial factors involved in this system have been sought in exocrine glands, particularly in the mammary glands. Because milk produced in the mammary glands is enriched in various nutrients, supporting the proliferation of bacteria, mammary glands appear to be at the greatest risk of bacterial infection and proliferation. Here, we show that mouse milk contains L-amino acid oxidase (LAO), a lactating mammary gland-specific protein that displays antibacterial activity in vitro through the production of hydrogen peroxide from free amino acids. We produced LAO-disrupted mouse lines to define the physiological properties and importance of the protein in vivo. The LAO-knockout mice were healthy and had normal mammary gland development; however, the antibacterial activity normally observed in milk from wild-type mice was absent from the milk of knockout mice. The content of free amino acids targeted by LAO was very low in wild-type milk, whereas these amino acids were abundant in LAO-knockout milk. Knockout mice exhibited weak resistance to an intramammary bacterial challenge compared to their wild-type counterparts. Further, preadministration of wild-type milk whey reduced the severity of bacterial infection in LAO-knockout mice. These results demonstrate that milk LAO protects the mammary gland against bacterial infection, and this antibacterial effect may be due to the generation of hydrogen peroxide by using free amino acids abundantly present in milk.


Assuntos
Imunidade Inata , L-Aminoácido Oxidase/imunologia , Glândulas Mamárias Animais/enzimologia , Glândulas Mamárias Animais/imunologia , Animais , Infecções Bacterianas/enzimologia , Infecções Bacterianas/imunologia , Infecções Bacterianas/prevenção & controle , Feminino , L-Aminoácido Oxidase/deficiência , L-Aminoácido Oxidase/genética , Lactação/imunologia , Lactação/metabolismo , Glândulas Mamárias Animais/microbiologia , Mastite/enzimologia , Mastite/imunologia , Mastite/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Leite/química , Leite/enzimologia , Leite/imunologia , Infecções Estafilocócicas/enzimologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade
13.
Antonie Van Leeuwenhoek ; 97(2): 107-17, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19911297

RESUMO

Previous reports have shown that Escherichia coli O157:H7 infection is strongly modified by intestinal microbes. In this paper, we examined whether bifidobacteria protect against E. coli O157:H7 infections using gnotobiotic mice di-associated with Bifidobacterium strains (6 species, 9 strains) and E. coli O157:H7. Seven days after oral administration of each Bifidobacterium strain, the mice were orally infected with E. coli O157:H7 and their mortality was examined. Bifidobacterium longum subsp. infantis 157F-4-1 (B. infantis 157F) and B. longum subsp. longum NCC2705 (B. longum NS) protected against the lethal infection, while mice associated with all other Bifidobacterium strains, including type strains of B. longum subsp. infantis and B. longum subsp. longum, died. There were no significant differences in the numbers of E. coli O157:H7 in the faeces among the Bifidobacterium-associated mouse groups. However, the Shiga toxin concentrations in the cecal contents and sera of the GB mice associated with B. infantis 157F and B. longum NS were significantly lower than those of the other groups. However, there were no significant differences in the volatile fatty acid concentrations and histopathological lesions between these two groups. These data suggest that some strains of B. longum subsp. longum/infantis can protect against the lethal infections of E. coli O157:H7 by preventing Shiga toxin production in the cecum and/or Shiga toxin transfer from the intestinal lumen to the bloodstream.


Assuntos
Bifidobacterium/fisiologia , Infecções por Escherichia coli/prevenção & controle , Escherichia coli O157/crescimento & desenvolvimento , Trato Gastrointestinal/microbiologia , Animais , Antibiose , Bifidobacterium/crescimento & desenvolvimento , Ceco/química , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Fezes/microbiologia , Feminino , Vida Livre de Germes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Toxina Shiga/análise , Análise de Sobrevida
14.
J Vet Med Sci ; 72(5): 621-5, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20009425

RESUMO

We have previously reported the action of whey acidic protein (WAP) inhibiting the proliferation of mouse mammary epithelial cells in the experiments utilizing in vivo and in vitro systems. We report herein the bacteriostatic activity of WAP. Western blot analysis demonstrated successful isolation of WAP from whey fractions of rat milk by column chromatography. The WAP fraction inhibited the growth of Staphylococcus aureus JCM2413 in a dose-dependent manner, but did not inhibit the growth of Escherichia coli. The bacteriostatic activity of WAP was highest at pH 6.6 and was not affected by the presence of 150 mM NaCl. A scanning electron micrograph of bacteria treated with WAP exhibited the disruption of the bacterial cell walls.


Assuntos
Antibacterianos/farmacologia , Proteínas do Leite/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Microscopia Eletrônica de Varredura , Leite/química , Proteínas do Leite/isolamento & purificação , Ratos , Proteínas Recombinantes/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/ultraestrutura
15.
Free Radic Res ; 54(1): 64-75, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31905044

RESUMO

Bleomycin is a glycopeptide antibiotic that is widely employed in the therapy of a range of lymphomas and germ cell tumours. But the therapeutic efficacy of bleomycin is limited by development of lung fibrosis. The cytotoxicity of bleomycin is mostly ascribed to mitochondrial DNA (mtDNA) damage, while a protective effect of metformin against bleomycin-induced lung fibrosis results from the inhibition of mitochondrial complex I. Since mitochondria and bacteria have certain similarities in structure and function, we used Escherichia coli for simplification in the present work to investigate the relationship between metformin and bleomycin with apparently opposite effects on mitochondrial DNA damage. Bleomycin lethality to E. coli was ameliorated by metformin treatment accompanying further increase of the level of reactive oxygen species. Catalase but not superoxide dismutases attenuated the protective effect of metformin. Meanwhile, treatment with hydrogen peroxide enhanced the protection, indicating that metformin may protect E. coli from bleomycin-induced bactericide via enhanced generation of hydrogen peroxide. Moreover, silibinin, a hepatoprotective polyphenolic flavonoid attenuates the cytotoxicity of bleomycin to E. coli via enhanced generation of hydrogen peroxide as well. This bacterial model in place of mitochondria can provide us with easier screening for the molecules with capability of reducing the bleomycin side effect.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Bleomicina/efeitos adversos , Atividade Bactericida do Sangue/efeitos dos fármacos , Escherichia coli/patogenicidade , Peróxido de Hidrogênio/química , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Humanos , Hipoglicemiantes/farmacologia , Metformina/farmacologia
16.
Biosci Biotechnol Biochem ; 73(2): 372-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19202287

RESUMO

The gut mucosal immune system is crucial in host defense against infection by pathogenic microbacteria and viruses via the production of IgA. Previous studies have shown that intestinal commensal bacteria enhance mucosal IgA production. However, it is poorly understood how these bacteria induce IgA production and which genera of intestinal commensal bacteria induce IgA production effectively. In this study, we compared the immunomodulatory effects of Bacteroides and Lactobacillus on IgA production by Peyer's patches lymphocytes. IgA production by Peyer's patches lymphocytes co-cultured with Bacteroides was higher than with Lactobacillus. In addition, the expression of activation-induced cytidine deaminase increased in co-culture with Bacteroides but not with Lactobacillus. We found that intestinal commensal bacteria elicited IgA production. In particular, Bacteroides induced the differentiation of Peyer's patches B cell into IgA(+) B cells by increasing activation-induced cytidine deaminase expression.


Assuntos
Linfócitos B/metabolismo , Bacteroides/fisiologia , Citidina Desaminase/genética , Regulação Enzimológica da Expressão Gênica , Imunoglobulina A/biossíntese , Lactobacillus/fisiologia , Nódulos Linfáticos Agregados/imunologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/microbiologia , Diferenciação Celular , Técnicas de Cocultura , Interleucina-5/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/citologia , Regulação para Cima
17.
J Vet Med Sci ; 71(9): 1247-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19801909

RESUMO

The presence of 11 virulence-associated genes in Campylobacter jejuni isolated from chickens at different stages of their development was examined by PCR. Multiple groups of C. jejuni were colonized in one chicken at the same time. Two hundred thirty one C. jejuni in total were isolated from the same group at four different ages and 12 groups of C. jejuni possessing of 11 virulence-associated genes were observed. Eleven, eight, five and three groups of C. jejuni were detected at 21, 28, 42 and 56 days after hatching, respectively. The variation of Campylobacter groups was reduced as the chicken developed and one group of C. jejuni became predominant.


Assuntos
Envelhecimento , Infecções por Campylobacter/veterinária , Campylobacter jejuni/genética , Campylobacter jejuni/patogenicidade , Galinhas , Doenças das Aves Domésticas/microbiologia , Animais , Infecções por Campylobacter/microbiologia , Virulência
18.
DNA Res ; 15(3): 151-61, 2008 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-18487258

RESUMO

Lactobacillus reuteri is a heterofermentative lactic acid bacterium that naturally inhabits the gut of humans and other animals. The probiotic effects of L. reuteri have been proposed to be largely associated with the production of the broad-spectrum antimicrobial compound reuterin during anaerobic metabolism of glycerol. We determined the complete genome sequences of the reuterin-producing L. reuteri JCM 1112(T) and its closely related species Lactobacillus fermentum IFO 3956. Both are in the same phylogenetic group within the genus Lactobacillus. Comparative genome analysis revealed that L. reuteri JCM 1112(T) has a unique cluster of 58 genes for the biosynthesis of reuterin and cobalamin (vitamin B(12)). The 58-gene cluster has a lower GC content and is apparently inserted into the conserved region, suggesting that the cluster represents a genomic island acquired from an anomalous source. Two-dimensional nuclear magnetic resonance (2D-NMR) with (13)C(3)-glycerol demonstrated that L. reuteri JCM 1112(T) could convert glycerol to reuterin in vivo, substantiating the potential of L. reuteri JCM 1112(T) to produce reuterin in the intestine. Given that glycerol is shown to be naturally present in feces, the acquired ability to produce reuterin and cobalamin is an adaptive evolutionary response that likely contributes to the probiotic properties of L. reuteri.


Assuntos
Ilhas Genômicas , Gliceraldeído/análogos & derivados , Limosilactobacillus fermentum/genética , Limosilactobacillus reuteri/genética , Propano/metabolismo , Vitamina B 12/biossíntese , Mapeamento Cromossômico , Genoma Bacteriano , Gliceraldeído/metabolismo , Limosilactobacillus fermentum/metabolismo , Limosilactobacillus reuteri/metabolismo , Redes e Vias Metabólicas/genética , Modelos Biológicos , Família Multigênica , Filogenia , Vitamina B 12/genética
19.
Bioresour Technol ; 99(7): 2687-93, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17572083

RESUMO

The objectives of this study were to investigate the fate of microorganisms by using cultivation methods as well as DNA analyses in a commercial microbiological additive (MA) in the course of the composting. Almost all the predominant species in the microbial succession during composting process determined by polymerase chain reaction-denaturing gradient gel electrophoresis (PCR-DGGE) were in disagreement with those determined by the clone library method. None of the microbial species in the composting stages corresponded to the microorganisms identified in the MA either by the cultivation method or DNA analysis. The results in regard to predominant microorganisms of the MA detected from the liquid medium by the PCR-DGGE did not correspond with those detected from the MA itself and composting processes. Although no evidence was found that predominant species in the MA itself dominate in the composting process, predominant species diversity in the MA itself was markedly changed after culturing at different thermophilic temperatures. These results suggested that cultivable microorganisms in the MA did not become predominant in the composting process: however, some microorganisms that are detected from the MA itself by the DNA analysis may act effectively in the composting process.


Assuntos
Esterco , Solo , Sequência de Bases , DNA/análise , DNA/genética , Primers do DNA , Eletroforese em Gel de Poliacrilamida , RNA Ribossômico 16S/genética
20.
FEMS Microbiol Lett ; 365(8)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29546276

RESUMO

This study demonstrates that growth of Staphylococcus aureus in the presence of salicylate reduces ultraviolet C (UVC)-induced cell death and increases the generation of reactive oxygen species (ROS). In addition, compounds that scavenge ROS (N-acetylcysteine, glutathione, catalase and superoxide dismutase) reverse the increased UVC survival induced by growth in the presence of salicylate, while ROS donors (tert-butylhydroperoxide, H2O2 and NaClO) enhance survival of salicylate challenged cultures. Collectively, these findings suggest that ROS production induced by growth in the presence of salicylate protects S. aureus from UVC-induced cell death.


Assuntos
Espécies Reativas de Oxigênio/metabolismo , Salicilatos/metabolismo , Staphylococcus aureus/efeitos da radiação , Catalase/genética , Catalase/metabolismo , Glutationa/metabolismo , Viabilidade Microbiana/efeitos da radiação , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA