Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Int. microbiol ; 26(4): 907-915, Nov. 2023. graf
Artigo em Inglês | IBECS | ID: ibc-227479

RESUMO

The prevention of biofilm formation plays a pivotal role in managing Helicobacter pylori inside the body and the environment. This study showed in vitro potentials of two recently isolated probiotic strains, Bacillus sp. 1630F and Enterococcus sp. 7C37, to form biofilm and combat H. pylori attachment to the abiotic and biotic surfaces. Lactobacillus casei and Bifidobacterium bifidum were used as the reference probiotics. The biofilm rates were the highest in the solid–liquid interface for Lactobacillus and Bifidobacterium and the air–liquid interface for Bacillus and Enterococcus. The highest tolerances to the environmental conditions were observed during the biofilm formations of Enterococcus and Bifidobacterium (pH), Enterococcus and Bacillus (bile), and Bifidobacterium and Lactobacillus (NaCl) on the polystyrene and glass substratum, respectively. Biofilms occurred more quickly by Bacillus and Enterococcus strains than reference strains on the polystyrene and glass substratum, respectively. Enterococcus (competition) and Bacillus (exclusion) achieved the most inhibition of H. pylori biofilm formations on the polystyrene and AGS cells, respectively. Expression of luxS was promoted by Bacillus (exclusion, 3.2 fold) and Enterococcus (competition, 2.0 fold). Expression of ropD was decreased when H. pylori biofilm was excluded by Bacillus (0.4 fold) and Enterococcus (0.2 fold) cells. This study demonstrated the ability of Bacillus and Enterococcus probiotic bacteria to form biofilm and combat H. pylori biofilm formation.(AU)


Assuntos
Humanos , Bacillus , Enterococcus , Helicobacter pylori , Probióticos , Poliestirenos , Biofilmes , Microbiologia , Técnicas Microbiológicas , Infecções por Bifidobacteriales
2.
Anaerobe ; 73: 102483, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34839008

RESUMO

Clinical infections by Bifidobacterium species rarely developed. We report two cases of bacteremic pneumonia caused by B. pseudocatenulatum and B. dentium, respectively, in patients vulnerable to aspiration. These cases suggested the potential for cause of serious pneumonia caused by Bifidobacterium species, in patients with high risk of aspiration.


Assuntos
Bacteriemia , Infecções por Bifidobacteriales , Bifidobacterium , Pneumonia , Bacteriemia/diagnóstico , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Infecções por Bifidobacteriales/diagnóstico , Infecções por Bifidobacteriales/tratamento farmacológico , Infecções por Bifidobacteriales/microbiologia , Humanos
3.
Biomolecules ; 11(8)2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34439760

RESUMO

BACKGROUND: Accumulating evidence indicates that the gut microbiota can synthesize neurotransmitters as well as impact host-derived neurotransmitter levels. In the past, it has been challenging to decipher which microbes influence neurotransmitters due to the complexity of the gut microbiota. METHODS: To address whether a single microbe, Bifidobacterium dentium, could regulate important neurotransmitters, we examined Bifidobacteria genomes and explored neurotransmitter pathways in secreted cell-free supernatant using LC-MS/MS. To determine if B. dentium could impact neurotransmitters in vivo, we mono-associated germ-free mice with B. dentium ATCC 27678 and examined fecal and brain neurotransmitter concentrations. RESULTS: We found that B. dentium possessed the enzymatic machinery to generate γ-aminobutyric acid (GABA) from glutamate, glutamine, and succinate. Consistent with the genome analysis, we found that B. dentium secreted GABA in a fully defined microbial media and elevated fecal GABA in B. dentium mono-associated mice compared to germ-free controls. We also examined the tyrosine/dopamine pathway and found that B. dentium could synthesize tyrosine, but could not generate L-dopa, dopamine, norepinephrine, or epinephrine. In vivo, we found that B. dentium mono-associated mice had elevated levels of tyrosine in the feces and brain. CONCLUSIONS: These data indicate that B. dentium can contribute to in vivo neurotransmitter regulation.


Assuntos
Bifidobacterium/metabolismo , Neurotransmissores/metabolismo , Animais , Infecções por Bifidobacteriales/metabolismo , Encéfalo/metabolismo , Calibragem , Cromatografia Líquida , Microbioma Gastrointestinal , Genoma , Intestinos/patologia , Masculino , Camundongos , Microbiota , Espectrometria de Massas em Tandem , Tirosina/metabolismo
4.
J Nippon Med Sch ; 88(3): 171-177, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-33692290

RESUMO

Atopic dermatitis (AD) is a chronic eczematous disease characterized by T helper 2 (Th2) -shifted allergic immunity, skin barrier impairment, and pruritus. Oral intake of certain nutrients might help regulate AD. Serum 25-hydroxyvitamin D levels are often low in patients with AD, and oral vitamin D supplementation improves AD. Vitamin D increases regulatory T (Treg) cells, which promote tolerance to allergens and prevent allergic inflammation by inducing expression of filaggrin and cathelicidin in keratinocytes. Vitamin A strengthens Treg cells by inducing expression of forkhead box P3 and inhibits mediator release from mast cells and eosinophils. Serum levels of γ-linolenic acid and its metabolite, dihomo-γ-linolenic acid, are low in patients with AD, and oral γ-linolenic acid improves AD through anti-inflammatory prostaglandin D1 and E1 derived from dihomo-γ-linolenic acid. Eicosapentaenoic acid and docosahexaenoic acid ameliorate AD by suppressing production of leukotriene B4, increasing ceramides in the stratum corneum, and through their metabolites, resolvin E1 and D1, which resolve inflammation. The probiotics Lactobacillus and Bifidobacteria improve the intestinal permeability barrier and induce Treg cells. Zinc levels in serum, hair, and erythrocytes are diminished in patients with AD. Zinc induces forkhead box P3 expression and increases Treg cells, and zinc-finger protein A20 suppresses nuclear factor-κB-dependent expression of inflammatory cytokines and cell-adhesion molecules. Oral supplementation of the above nutrients might have therapeutic or preventive roles in AD.


Assuntos
Dermatite Atópica , Estado Nutricional , Probióticos/administração & dosagem , Vitamina D , Zinco , Infecções por Bifidobacteriales , Bifidobacterium , Dermatite Atópica/tratamento farmacológico , Proteínas Filagrinas , Humanos , Inflamação , Lactobacillus , Linfócitos T Reguladores , Resultado do Tratamento , Vitamina D/sangue , Ácido gama-Linolênico
5.
Saudi J Kidney Dis Transpl ; 31(5): 898-904, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33229754

RESUMO

Recurrent urinary tract infection (UTI) in children is a well-known risk factor of chronic kidney disease. Periurethral area is normally inhabited by non-pathogenic flora, such as Bifidobacterium sp., and pathogenic flora from gastrointestinal tract, such as Escherichia coli (E. coli), which can cause UTI. Dysbiosis between pathogenic and non-pathogenic bacteria leads to infections, but studies regarding dysbiosis and recurrent UTI have not yet been documented. To estimate the proportional differences between gastrointestinal E. coli and Bifidobacterium sp. in children with recurrent UTI, a cross-sectional study was conducted in children from age six months to <18 years old diagnosed with recurrent UTI in Dr. Cipto Mangunkusumo Hospital. Healthy children matched in gender and age were recruited as control group. Stool samples were obtained from all the children in the two groups. Stool DNA was extracted using real-time polymerized chain reaction method to count E. coli and Bifidobacterium sp. proportion. Children with recurrent UTI had significantly higher proportion of E. coli compared to control group (10.97 vs. 4.74; P = 0.014) and lower proportion of Bifidobacterium sp. (6.54 vs. 9.33; P = 0.594). In children with recurrent UTI group, E. coli proportion was found higher than Bifidobacterium sp. although not statistically significant (10.97 vs. 6.54; P = 0.819). In healthy controls, Bifidobacterium sp. proportion was significantly higher than E. coli (4.74 vs. 9.33; P = 0.021). The total amount of E. coli (996,004 vs. 1,099,271; P = 0.798) and Bifidobacterium sp. (835,921 vs. 1,196,991; P = 0.711) were higher in secondary UTI compared to the simple UTI. Proportion of E. coli is higher in children with recurrent UTI than in healthy children. The proportion of E. coli is higher than Bifidobacterium sp in the colon of children with recurrent UTI.


Assuntos
Bifidobacterium , Colo/microbiologia , Disbiose , Escherichia coli , Infecções Urinárias/epidemiologia , Adolescente , Infecções por Bifidobacteriales/epidemiologia , Infecções por Bifidobacteriales/microbiologia , Criança , Pré-Escolar , Estudos Transversais , Disbiose/epidemiologia , Disbiose/microbiologia , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Fezes/microbiologia , Feminino , Humanos , Lactente , Masculino , Infecções Urinárias/microbiologia
6.
Anaerobe ; 65: 102247, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32771620

RESUMO

Most species of the genus Bifidobacterium contain the gene cluster PFNA, which is presumably involved in the species-specific communication between bacteria and their hosts. The gene cluster PFNA consists of five genes including fn3, which codes for a protein containing two fibronectin type III domains. Each fibronectin domain contains sites similar to cytokine-binding sites of human receptors. Based on this finding we assumed that this protein would bind specifically to human cytokines in vitro. We cloned a fragment of the fn3 gene (1503 bp; 501 aa) containing two fibronectin domains, from the strain B. longum subsp. longum GT15. After cloning the fragment into the expression vector pET16b and expressing it in E. coli, the protein product was purified to a homogenous state for further analysis. Using the immunoferment method, we tested the purified fragment's ability to bind the following human cytokines: IL-1ß, IL-6, IL-10, TNFα. We developed a sandwich ELISA system to detect any specific interactions between the purified protein and any of the studied cytokines. We found that the purified protein fragment only binds to TNFα.


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium/metabolismo , Domínio de Fibronectina Tipo III , Fibronectinas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas de Bactérias/química , Infecções por Bifidobacteriales/metabolismo , Infecções por Bifidobacteriales/microbiologia , Bifidobacterium/genética , Biologia Computacional/métodos , Citocinas/metabolismo , Fibronectinas/química , Interações Hospedeiro-Patógeno , Humanos , Família Multigênica , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
7.
Int Immunol ; 32(10): 629-636, 2020 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-32564086

RESUMO

The gut microbiota strongly impacts the physiology and pathology in the host. To understand the complex interactions between host and gut microbiota, an 'integrated omics' approach has been employed, where exhaustive analyses for the different layers of cellular functions, such as epigenomics, transcriptomics and metabolomics, in addition to metagenomics, are combined. With this approach, the mechanisms whereby short-chain fatty acids (SCFAs) regulate host defense and the immune system have been elucidated. In a gnotobiotic mouse model of enterohemorrhagic Escherichia coli infection, Bifidobacterium-derived acetate can protect from infection-mediated death by changing the gene expression profile of colonic epithelial cells. It has also been shown that gut microbiota-derived butyrate enhances colonic regulatory T-cell differentiation through its epigenetic modulatory ability via histone deacetylase inhibition. SCFAs are involved in many other immunomodulatory effects as well as host pathophysiological conditions. Dysbiosis in the gut has been implicated in the pathogenesis of many diseases. Although the causal relationship of gut microbial dysbiosis and/or metabolites with pathogenesis is mostly unknown, mechanistic insights have been elucidated in some cases. Metabolism in the gut microbiota and host liver produces trimethylamine N-oxide, which is known to aggravate atherosclerosis, and a secondary bile acid deoxycholate, which reportedly induces non-alcoholic steatohepatitis-related hepatocellular carcinoma. It has been reported that secondary bile acids could also induce the differentiation of peripherally derived regulatory T cells in the gut. Further studies on the interactions between the host and gut microbiota could lead to the development of new therapeutic strategies as well as in preventive medicine.


Assuntos
Infecções por Bifidobacteriales/imunologia , Microbioma Gastrointestinal/imunologia , Linfócitos T Reguladores/imunologia , Animais , Infecções por Bifidobacteriales/metabolismo , Disbiose/imunologia , Disbiose/metabolismo , Humanos , Linfócitos T Reguladores/metabolismo
8.
Allergol. immunopatol ; 48(2): 107-115, mar.-abr. 2020. tab
Artigo em Inglês | IBECS | ID: ibc-191812

RESUMO

BACKGROUND: It is possible that imbalances in the composition of the gut microbiota or the relationship of the microbiota with the host may be implicated in the origin of allergy. Therefore, we studied the intestinal microbiota of children with atopic dermatitis (AD). METHODS: Cross-sectional study with 81 children aged 5-11; 23 with AD and 58 controls. Surveys were conducted to obtain demographic, socioeconomic and neonatal data. Diagnosis of AD was made based on the International Study of Asthma and Allergies in Childhood (ISAAC) questionnaire. Eubacteria, Bacteroidetes, Firmicutes, B. fragilis, E. coli, Lactobacillus spp., S. aureus, E. faecalis, Salmonella spp., M. smithii, Bifidobacterium spp., C. difficile and C. perfringens were quantified using real-time PCR. RESULTS: The analysis showed an association between presence of C. difficile (OR: 5.88; 95 % CI: 1.24; 27.98), greater abundance of bifidobacteria (OR: 11.09; 95 % CI: 2.14; 57.39) and a lower abundance of lactobacilli (OR: 0.07; 95 % CI: 0.01; 0.51) in the gut microbiota of children with AD. Counts of Eubacteria (0,05 × 103 and 8.49 × 103), B. fragilis (0.72 × 109 and 4.5 × 109), Lactobacillus spp. (0.02 × 108 and 0.38 × 108), E. coli (0.13 × 109 and 1.52 × 109) and M. smithii (0.02×108 and 0.31 × 108) were lower in children with AD (P < 0.05). CONCLUSIONS: This study confirmed that children living in the metropolitan area of São Paulo (Brazil) with AD have a different microbiota pattern with higher prevalence of C. difficile, lower abundance of Lactobacillus and greater abundance of bifidobacteria, regardless of socioeconomic status


No disponible


Assuntos
Humanos , Masculino , Feminino , Pré-Escolar , Criança , Microbioma Gastrointestinal/imunologia , Dermatite Atópica/complicações , Doenças Respiratórias/epidemiologia , Microbioma Gastrointestinal/genética , Brasil/epidemiologia , Inquéritos e Questionários , Estudos Transversais , Infecções por Clostridium/imunologia , Lactobacillus/imunologia , Infecções por Bifidobacteriales/imunologia , Estado Nutricional/imunologia , Doenças Respiratórias/imunologia , Modelos Logísticos
9.
Front Immunol ; 10: 2348, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31632412

RESUMO

Since bifidobacteria are among the pioneering colonizers of the human infant gut, their interaction with their host is believed to start soon following birth. Several members of the Bifidobacterium genus are purported to exert various health-promoting effects at local and systemic levels, e.g., limiting pathogen colonization/invasion, influencing gut homeostasis, and influencing the immune system through changes in innate and/or adaptive immune responses. This has promoted extensive research efforts to shed light on the precise mechanisms by which bifidobacteria are able to stimulate and interact with the host immune system. These studies uncovered a variety of secreted or surface-associated molecules that act as essential mediators for the establishment of a bifidobacteria-host immune system dialogue, and that allow interactions with mucosa-associated immune cells. Additionally, the by-products generated from bifidobacterial carbohydrate metabolism act as vectors that directly and indirectly trigger the host immune response, the latter by stimulating growth of other commensal microorganisms such as propionate- or butyrate-producing bacteria. This review is aimed to provide a comprehensive overview on the wide variety of strategies employed by bifidobacteria to engage with the host immune system.


Assuntos
Infecções por Bifidobacteriales/imunologia , Infecções por Bifidobacteriales/microbiologia , Bifidobacterium/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Sistema Imunitário/imunologia , Imunomodulação , Infecções por Bifidobacteriales/metabolismo , Bifidobacterium/classificação , Código de Barras de DNA Taxonômico , Matriz Extracelular/metabolismo , Microbioma Gastrointestinal , Homeostase , Humanos , Sistema Imunitário/metabolismo , Metaboloma , Metabolômica/métodos , Polissacarídeos Bacterianos/metabolismo , Probióticos
10.
New Microbiol ; 42(4): 237-239, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31609456

RESUMO

Alloscardovia omnicolens is a recently-reported microorganism with unknown pathogenic implications. It has been isolated in various clinical localizations but not in the endocervix. We isolated A. omnicolens in an endocervical sample from a 31-yr-old patient with preterm premature rupture of membranes (PPROM) in week 33+3 of pregnancy. The main risk of PPROM is prematurity and the possibility of developing infectious chorioamnionitis, which can be lethal for the mother and newborn. This is the first report of an association between A. omnicolens and PPROM, although its pathogenic role has not yet been elucidated.


Assuntos
Actinobacteria , Infecções por Bifidobacteriales , Corioamnionite , Ruptura Prematura de Membranas Fetais , Actinobacteria/fisiologia , Adulto , Antibacterianos/uso terapêutico , Infecções por Bifidobacteriales/complicações , Infecções por Bifidobacteriales/tratamento farmacológico , Infecções por Bifidobacteriales/microbiologia , Infecções por Bifidobacteriales/patologia , Colo do Útero/microbiologia , Corioamnionite/tratamento farmacológico , Corioamnionite/microbiologia , Feminino , Ruptura Prematura de Membranas Fetais/etiologia , Ruptura Prematura de Membranas Fetais/microbiologia , Idade Gestacional , Humanos , Gravidez , Nascimento Prematuro , Resultado do Tratamento
11.
Sci Rep ; 9(1): 8692, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213639

RESUMO

Bifidobacteria are one of the most abundant bacterial groups in the infant gut microbiota and are closely associated with infant health and can potentially affect health in later life. However, the details regarding the source of bifidobacteria have yet to be completely elucidated. This study aimed to assess neonatal oral fluid (OF) as a transmission route for bifidobacteria to the infant gut during delivery. Neonatal OF and infant feces (IF) were collected immediately and one month after birth from 15 healthy vaginally delivered newborns. Bifidobacterium strains were isolated from OF and IF samples, and the similarity of strains between the OF-IF pairs was evaluated based on the average nucleotide identity (ANI) value. The 16S rRNA gene sequencing results revealed the presence of Bifidobacteriaceae at >1% relative abundance in all OF samples. Bifidobacterium strains were isolated from OF (9/15) and IF (11/15) samples, and those sharing high genomic homology (ANI values >99.5%) between the neonatal OF and IF samples were present in one-third of the OF-IF pairs. The results of this study indicate that viable bifidobacteria are present in neonatal OF and that OF at birth is a possible transmission route of bifidobacteria to the infant gut.


Assuntos
Bifidobacterium/isolamento & purificação , Fezes/microbiologia , Trato Gastrointestinal/microbiologia , Boca/microbiologia , Saliva/microbiologia , Infecções por Bifidobacteriales/microbiologia , Infecções por Bifidobacteriales/transmissão , Bifidobacterium/classificação , Bifidobacterium/genética , Análise por Conglomerados , Feminino , Microbioma Gastrointestinal/genética , Humanos , Lactente , Recém-Nascido , Masculino , RNA Ribossômico 16S/genética , Análise de Sequência de DNA/métodos , Especificidade da Espécie
12.
13.
Pediatrics ; 143(2)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30674610

RESUMO

BACKGROUND: The intestinal microbiome in early infancy affects immunologic development and thus may affect vaccine memory, though few prospective studies have examined such associations. We examined the association of Bifidobacterium levels in early infancy with memory responses to early vaccination measured at 2 years of age. METHODS: In this prospective observational study, we examined the association of Bifidobacterium abundance in the stool of healthy infants at 6 to 15 weeks of age, near the time of vaccination, with T-cell and antibody responses measured at 6 weeks, 15 weeks, and 2 years of age. Infants were vaccinated with Bacillus Calmette-Guérin (BCG) (at birth), oral polio virus (at birth and at 6, 10, and 14 weeks), tetanus toxoid (TT) (at 6, 10, and 14 weeks), and hepatitis B virus (at 6, 10, and 14 weeks). Fecal Bifidobacterium was measured at 6, 11, and 15 weeks. Bifidobacterium species and subspecies were measured at 6 weeks. RESULTS: Mean Bifidobacterium abundance in early infancy was positively associated with the CD4 T-cell responses to BCG, TT, and hepatitis B virus at 15 weeks, with CD4 responses to BCG and TT at 2 years, and with plasma TT-specific immunoglobulin G and stool polio-specific immunoglobulin A at 2 years. Similar associations were seen for the predominant subspecies, Bifidobacterium longum subspecies infantis. CONCLUSIONS: Bifidobacterium abundance in early infancy may increase protective efficacy of vaccines by enhancing immunologic memory. This hypothesis could be tested in clinical trials of interventions to optimize Bifidobacterium abundance in appropriate populations.


Assuntos
Vacina BCG/administração & dosagem , Infecções por Bifidobacteriales/diagnóstico , Infecções por Bifidobacteriales/prevenção & controle , Bifidobacterium/efeitos dos fármacos , Vacinação/métodos , Infecções por Bifidobacteriales/epidemiologia , Bifidobacterium/fisiologia , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Estudos Prospectivos , Resultado do Tratamento , Vacinação/tendências
14.
Int Arch Allergy Immunol ; 177(4): 342-349, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30205386

RESUMO

BACKGROUND: Altered regulatory immune responses to microbial stimuli and intestinal colonization of beneficial bacteria early in life may contribute to the development of allergic diseases (e.g., atopic dermatitis [AD]). However, few reports have investigated these factors simultaneously. The purpose of this study was to analyze neonatal immune responses to microbial stimuli as well as intestinal colonization of beneficial bacteria, in relation to the development of AD in a birth cohort. METHODS: Pregnant women were recruited, and their infants were followed up until 7 months of age. Levels of interleukin (IL)-10 released from cord-blood mononuclear cells (CBMCs) stimulated with heat-killed gram-positive bacteria (Bifidobacterium bifidum and Lactobacillus rhamnosus GG) and Lactobacillus-derived peptidoglycan were measured. Fecal Bifidobacterium counts at 4 days and 1 month were quantified using real-time polymerase chain reaction. The development of AD was determined by means of a questionnaire at 7 months of age. RESULTS: The levels of released IL-10 were significantly lower in infants with AD (n = 17) than in infants without AD (n = 53) for all stimuli. In infants with fecal Bifidobacterium, the incidence of AD was inversely associated with the release of IL-10 from cord blood mononuclear cells. CONCLUSION: Our findings suggest that impaired IL-10 production in response to microbial stimuli at birth may be associated with an increased risk of developing infantile AD, even in infants with early colonization of intestinal bifidobacteria.


Assuntos
Infecções por Bifidobacteriales/imunologia , Bifidobacterium/fisiologia , Dermatite Atópica/imunologia , Sangue Fetal/fisiologia , Leucócitos Mononucleares/imunologia , Células Cultivadas , Estudos de Coortes , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Interleucina-10/metabolismo , Masculino , Mães , Gravidez , Estudos Prospectivos , Fatores de Risco
15.
J Indian Soc Pedod Prev Dent ; 36(3): 290-295, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30246752

RESUMO

AIM: This study aimed to quantitatively assess the levels of Scardovia wiggsiae in caries-free and early childhood caries (ECC)- and severe ECC (SECC)-affected children using real-time polymerase chain reaction (RT-PCR). METHODS: Forty-five children aged <71 months were randomly recruited from the Outpatient Clinic at the Department of Pedodontics and Preventive Dentistry at Subharti Dental College and Hospital, Meerut, India. Fifteen children suffering from ECC, 15 with SECC, and 15 children without ECC were enrolled in the study. About 1-2 mL of unstimulated saliva was collected and subjected to microbial analysis using RT-PCR. RESULTS: The SECC group (n = 15) was found to have significantly higher mean relative 16s rRNA expression of S. wiggsiae (3.67) than both ECC (n = 15) and controls (n = 15) (1.69 and 0.85, respectively). S. wiggsiae was detected in 86.7% of the SECC and 60% ECC group and was detected negligibly in the control (caries free) group. The correlation of decayed, missing, or filled surface levels with 16s rRNA levels showed significant positive correlation with 16S rRNA in both ECC and SECC patients. CONCLUSION: Salivary levels of S. wiggsiae were significantly associated with ECC in children. S. wiggsiae represents a new frontier in the microbial etiology of ECC. This may lead to the development of new antimicrobial agents targeted to this organism and improve the treatment of ECC.


Assuntos
Infecções por Bifidobacteriales/diagnóstico , Bifidobacterium/isolamento & purificação , Cárie Dentária/microbiologia , Reação em Cadeia da Polimerase em Tempo Real , Saliva/microbiologia , Bifidobacterium/genética , Criança , Pré-Escolar , Humanos , Índia , RNA Ribossômico 16S/análise
16.
Am J Pathol ; 188(12): 2839-2852, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30243655

RESUMO

Evidence indicates a complex link between microbiota, tumor characteristics, and host immunity in the tumor microenvironment. In experimental studies, bifidobacteria appear to modulate intestinal epithelial cell differentiation. Accumulating evidence suggests that bifidobacteria may enhance the antitumor immunity and efficacy of immunotherapy. We hypothesized that the amount of bifidobacteria in colorectal carcinoma tissue might be associated with tumor differentiation and higher immune response to colorectal cancer. Using a molecular pathologic epidemiology database of 1313 rectal and colon cancers, we measured the amount of Bifidobacterium DNA in carcinoma tissue by a quantitative PCR assay. The multivariable regression model was used to adjust for potential confounders, including microsatellite instability status, CpG island methylator phenotype, long-interspersed nucleotide element-1 methylation, and KRAS, BRAF, and PIK3CA mutations. Intratumor bifidobacteria were detected in 393 cases (30%). The amount of bifidobacteria was associated with the extent of signet ring cells (P = 0.002). Compared with Bifidobacterium-negative cases, multivariable odd ratios for the extent of signet ring cells were 1.29 (95% CI, 0.74-2.24) for Bifidobacterium-low cases and 1.87 (95% CI, 1.16-3.02) for Bifidobacterium-high cases (Ptrend = 0.01). The association between intratumor bifidobacteria and signet ring cells suggests a possible role of bifidobacteria in determining distinct tumor characteristics or as an indicator of dysfunctional mucosal barrier in colorectal cancer.


Assuntos
Infecções por Bifidobacteriales/microbiologia , Bifidobacterium/genética , Biomarcadores Tumorais/genética , Carcinoma de Células em Anel de Sinete/patologia , Neoplasias Colorretais/patologia , DNA Bacteriano/genética , Adulto , Idoso , Infecções por Bifidobacteriales/genética , Infecções por Bifidobacteriales/patologia , Carcinoma de Células em Anel de Sinete/genética , Carcinoma de Células em Anel de Sinete/microbiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Estudos Prospectivos , Taxa de Sobrevida , Microambiente Tumoral
17.
Sci Rep ; 8(1): 13958, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30228375

RESUMO

Gut microbiota of breast-fed infants are generally rich in bifidobacteria. Recent studies show that infant gut-associated bifidobacteria can assimilate human milk oligosaccharides (HMOs) specifically among the gut microbes. Nonetheless, little is known about how bifidobacterial-rich communities are shaped in the gut. Interestingly, HMOs assimilation ability is not related to the dominance of each species. Bifidobacterium longum susbp. longum and Bifidobacterium breve are commonly found as the dominant species in infant stools; however, they show limited HMOs assimilation ability in vitro. In contrast, avid in vitro HMOs consumers, Bifidobacterium bifidum and Bifidobacterium longum subsp. infantis, are less abundant in infant stools. In this study, we observed altruistic behaviour by B. bifidum when incubated in HMOs-containing faecal cultures. Four B. bifidum strains, all of which contained complete sets of HMO-degrading genes, commonly left HMOs degradants unconsumed during in vitro growth. These strains stimulated the growth of other Bifidobacterium species when added to faecal cultures supplemented with HMOs, thereby increasing the prevalence of bifidobacteria in faecal communities. Enhanced HMOs consumption by B. bifidum-supplemented cultures was also observed. We also determined the complete genome sequences of B. bifidum strains JCM7004 and TMC3115. Our results suggest B. bifidum-mediated cross-feeding of HMOs degradants within bifidobacterial communities.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Bifidobacteriales/metabolismo , Bifidobacterium/metabolismo , Fezes/microbiologia , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Adulto , Proteínas de Bactérias/genética , Infecções por Bifidobacteriales/microbiologia , Bifidobacterium/classificação , Bifidobacterium/genética , Células Cultivadas , Pré-Escolar , Suplementos Nutricionais , Feminino , Microbioma Gastrointestinal , Genoma Bacteriano , Humanos , Lactente , Masculino
18.
J. physiol. biochem ; 74(1): 101-110, feb. 2018. tab, graf
Artigo em Inglês | IBECS | ID: ibc-178922

RESUMO

Nowadays, the strong relationship between diet and health is well known. Although the primary role of diet is to provide nutrients to fulfill metabolic requirements, the use of foods to improve health and the state of well-being is an idea increasingly accepted by society in the last three decades. During the last years, an important number of scientific advances have been achieved in this field and, although in some situations, it is difficult to establish a distinction between "harmful" and "good" bacteria, experts agree in classifying the genera Bifidobacterium and Lactobacillus as beneficial bacteria. Thus, several strategies can be used to stimulate the proliferation of these beneficial intestinal bacteria, being one of them the consumption of prebiotics. The development of new prebiotics, with added functionality, is one of the most serious challenges shared not only by the scientific community but also by the food industry. The objective of this work was to evaluate the potential prebiotic effect of red and white grape residues, both obtained during the winemaking process. For such purpose, an in vitro study with pure cultures of Lactobacillus and Bifidobacterium was first conducted. Secondly, a study with mixed cultures using human fecal inocula was carried out in a simulator of the distal part of the colon. The obtained results showed an increase in the Lactobacillus and Bifidobacterium population, indicating that these ingredients are serious candidates to be considered as prebiotics


Assuntos
Humanos , Animais , Masculino , Feminino , Microbioma Gastrointestinal , Modelos Biológicos , Pigmentos Biológicos/metabolismo , Componentes Aéreos da Planta/química , Extratos Vegetais/metabolismo , Prebióticos/economia , Vitis/química , Vitis/metabolismo , Infecções por Bifidobacteriales/metabolismo , Bifidobacterium/classificação , Bifidobacterium/isolamento & purificação , Fezes/microbiologia , Resíduos Industriais/análise , Resíduos Industriais/economia , Lactobacillus/classificação , Viabilidade Microbiana , Componentes Aéreos da Planta/metabolismo , Extratos Vegetais/economia
19.
Rev. argent. microbiol ; 49(4): 328-331, Dec. 2017. ilus
Artigo em Espanhol | LILACS | ID: biblio-1041797

RESUMO

La especie Bifidobacterium scardovii está constituida por bacilos gram positivos anaerobios facultativos, cuyo desarrollo es estimulado por el CO2 y la anaerobiosis. Excepcionalmente se la ha asociado a infecciones humanas. Se presenta el caso de un paciente anoso con infección urinaria por B. scardovii y Enterococcus faecalis, ambos microorganismos aislados en 2 urocultivos consecutivos. El bacilo no desarrolló en los medios de cultivo habituales, pero sí en agar chocolate en CO2 y en agar Brucella suplementado, incubados durante 72 h a 35°C. La coloración de Gram alertó acerca de su presencia al observarse abundantes bacilos gram positivos irregulares con extremos bifurcados en forma de Y, y escasos cocos gram positivos. Es importante la coloración de Gram en orinas con piuria y la siembra en medios enriquecidos por tiempos prolongados. En este caso, sin el resultado del Gram y con el desarrollo de E. faecalis, no hubiésemos advertido la presencia del agente mayoritario.


Bifidobacterium scardovii species consists of facultative anaerobic gram-positive rods whose growth is stimulated by CO2 and anaerobiosis. Exceptionally it has been associated with infections in humans. An elderly male patient with a urinary tract infection due to B. scardovii and Enterococcus faecalis is presented here; both microorganisms were isolated from two consecutive urine samples. The bacillus did not grow on standard media, but on chocolate agar incubated in CO2 and on supplemented Brucella agar in an anaerobic atmosphere, incubated for 72 h at 35°C. Gram staining with abundant irregular gram-positive rods with Y-shaped ends and some gram-positive cocci alerted to its presence. The importance of the Gram stain test in urine samples with pyuria and the growth on enriched media for long periods is highlighted here. In this case, if we had not had the Gram stain test results, and had considered only the E. faecalis growth, we would have lost the major etiologic agent.


Assuntos
Idoso , Humanos , Masculino , Infecções Urinárias , Bifidobacterium , Infecções por Bifidobacteriales , Infecções Urinárias/microbiologia , Urina , Bifidobacterium/isolamento & purificação , Infecções por Bifidobacteriales/microbiologia , Anaerobiose
20.
Sci Rep ; 7(1): 7408, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28785042

RESUMO

The longevity-promoting benefits of lactobacilli were hypothesized as early as 1907. Although the anti-aging effects of lactic acid bacteria (LAB) have been observed in nematodes, rodents and humans for over a century, the mechanisms underlying the effects of probiotics on aging have rarely been assessed. Using the Caenorhabditis elegans (C. elegans) model, various studies have elucidated the role of different signaling cascades, especially the DAF-16 cascade, on lifespan extension by LAB. In this study, the mechanisms through which Bifidobacterium longum strain BB68 affects the longevity of C. elegans were assessed. The lifespan of nematodes increased by 28% after worms were fed BB68, and this extension of lifespan was completely lost in backgrounds containing a mutated DAF-16 gene. High levels of DAF-16 (in the daf-16 (mu86); muIs61 strain) nuclear accumulation and high expression of the SOD-3 gene (a DAF-16-specific target gene) were observed as a result of BB68 treatment. Immunofluorescence microscopy revealed that TIR-1 and JNK-1 are involved in the phosphorylation and activation of DAF-16. Thus, BB68 increased the longevity of nematodes by activating the TIR-1 - JNK-1 - DAF-16 signaling pathway, and the cell wall component of BB68 contributed to longevity.


Assuntos
Bifidobacterium longum/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Caenorhabditis elegans/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Longevidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Infecções por Bifidobacteriales , Proteínas de Caenorhabditis elegans/análise , Núcleo Celular/química , Microscopia de Fluorescência , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Superóxido Dismutase/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...