Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol Res ; 2021: 8030297, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34337079

RESUMO

The prevalence of inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), increases gradually worldwide in the past decades. IBD is generally associated with the change of the immune system and gut microbiota, and the conventional treatments usually result in some side effects. Bifidobacterium longum, as colonizing bacteria in the intestine, has been demonstrated to be capable of relieving colitis in mice and can be employed as an alternative or auxiliary way for treating IBD. Here, the mechanisms of the Bifidobacterium longum in the treatment of IBD were summarized based on previous cell and animal studies and clinical trials testing bacterial therapies. This review will be served as a basis for future research on IBD treatment.


Assuntos
Bifidobacterium longum/imunologia , Colite Ulcerativa/dietoterapia , Doença de Crohn/dietoterapia , Microbioma Gastrointestinal/imunologia , Probióticos/administração & dosagem , Animais , Ensaios Clínicos como Assunto , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Doença de Crohn/imunologia , Doença de Crohn/patologia , Modelos Animais de Doenças , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia
2.
Front Immunol ; 11: 595877, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33304352

RESUMO

As permanent residents of the normal gut microbiota, bifidobacteria have evolved to adapt to the host's immune response whose priority is to eliminate pathogenic agents. The mechanisms that ensure the survival of commensals during inflammation and maintain the stability of the core component of the normal gut microbiota in such conditions remain poorly understood. We propose a new in vitro approach to study the mechanisms of resistance to immune response factors based on high-throughput sequencing followed by transcriptome analysis. This approach allowed us to detect differentially expressed genes associated with inflammation. In this study, we demonstrated that the presence of the pro-inflammatory cytokines IL-6 and TNFα to the growth medium of the B. longum subsp. longum GT15 strain changes the latter's growth rate insignificantly while affecting the expression of certain genes. We identified these genes and performed a COG and a KEGG pathway enrichment analysis. Using phylogenetic profiling we predicted the operons of genes whose expression was triggered by the cytokines TNFα and IL-6 in vitro. By mapping the transcription start points, we experimentally validated the predicted operons. Thus, in this study, we predicted the genes involved in a putative signaling pathway underlying the mechanisms of resistance to inflammatory factors in bifidobacteria. Since bifidobacteria are a major component of the human intestinal microbiota exhibiting pronounced anti-inflammatory properties, this study is of great practical and scientific relevance.


Assuntos
Bifidobacterium longum , Regulação Bacteriana da Expressão Gênica , Interleucina-6/imunologia , Fator de Necrose Tumoral alfa/imunologia , Bifidobacterium longum/genética , Bifidobacterium longum/crescimento & desenvolvimento , Bifidobacterium longum/imunologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Redes Reguladoras de Genes , Genoma Bacteriano , Inflamação/imunologia
3.
J Microbiol Biotechnol ; 29(11): 1739-1744, 2019 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-31546293

RESUMO

The present study evaluates the immunomodulatory effect of Bifidobacterium longum KACC 91563 in murine primary splenocytes and macrophages. B. longum KACC 91563 regulated Tand B-cell proliferation and inhibited the Th1 (IL-2, IFN-γ)/Th2 (IL-4, IL-10) cytokine imbalance and immune cytokine production. Moreover, immunoglobulin E (IgE) levels were significantly lower after treatment with B. longum KACC 91563. These findings suggest that B. longum KACC 91563 could modulate the systemic immune system toward both IgE production and regulation of the Th1/Th2 balance.


Assuntos
Bifidobacterium longum/imunologia , Imunomodulação , Macrófagos/imunologia , Baço/imunologia , Animais , Células Cultivadas , Citocinas/metabolismo , Imunoglobulina E/metabolismo , Linfócitos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/microbiologia , Equilíbrio Th1-Th2
4.
Artigo em Inglês | MEDLINE | ID: mdl-31380297

RESUMO

The consumption of probiotics and fermented foods has been very popular in recent decades. The primary aim of our study was to evaluate the effect of probiotics on the gut microbiota and the changes in inflammatory cytokines after an average of 6.7 weeks of probiotic administration among normal pregnant women. Thirty-two healthy pregnant women at 32 weeks of gestation were recruited and divided into two groups. The probiotic group ingested combined probiotics until after birth. The base characteristics of the probiotics and control groups showed no significant differences. The structure of the fecal microbiota at the genus level varied during the third trimester, and administration of probiotics had no influence on the composition of the fecal microbiota however, many highly abundant taxa and core microbiota at the genus level changed in the probiotic group when compared to the control group. The analysis of cytokines showed that IL-5, IL-6, TNF-α, and GM-CSF had equal levels between the baseline and control groups but were significantly increased after probiotic administration (baseline = control < probiotics). Additionally, levels of IL-1ß, IL-2, IL-12, and IFN-γ significantly increased among the three groups (baseline < control < probiotics). This result demonstrated that probiotics helped to shift the anti-inflammatory state to a pro-inflammatory state. The correlation analysis outcome suggested that the relationship between the microbiota and the cytokines was not strain-dependent. The gut microbiota varied during the third trimester. The probiotics demonstrated immunomodulation effects that helped to switch over to a pro-inflammatory immune state in the third trimester, which was important for labor.


Assuntos
Bifidobacterium longum/imunologia , Suplementos Nutricionais , Microbioma Gastrointestinal/imunologia , Lactobacillus delbrueckii/imunologia , Probióticos/administração & dosagem , Streptococcus thermophilus/imunologia , Adulto , Bifidobacterium longum/genética , Estudos de Casos e Controles , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/genética , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunidade Inata , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Lactobacillus delbrueckii/genética , Aprendizado de Máquina , Gravidez , Terceiro Trimestre da Gravidez , Streptococcus thermophilus/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
5.
JCI Insight ; 3(8)2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29669931

RESUMO

Obesity is a risk factor for osteoarthritis (OA), the greatest cause of disability in the US. The impact of obesity on OA is driven by systemic inflammation, and increased systemic inflammation is now understood to be caused by gut microbiome dysbiosis. Oligofructose, a nondigestible prebiotic fiber, can restore a lean gut microbial community profile in the context of obesity, suggesting a potentially novel approach to treat the OA of obesity. Here, we report that - compared with the lean murine gut - obesity is associated with loss of beneficial Bifidobacteria, while key proinflammatory species gain in abundance. A downstream systemic inflammatory signature culminates with macrophage migration to the synovium and accelerated knee OA. Oligofructose supplementation restores the lean gut microbiome in obese mice, in part, by supporting key commensal microflora, particularly Bifidobacterium pseudolongum. This is associated with reduced inflammation in the colon, circulation, and knee and protection from OA. This observation of a gut microbiome-OA connection sets the stage for discovery of potentially new OA therapeutics involving strategic manipulation of specific microbial species inhabiting the intestinal space.


Assuntos
Microbioma Gastrointestinal/fisiologia , Inflamação/microbiologia , Obesidade/microbiologia , Osteoartrite/microbiologia , Animais , Bifidobacterium longum/imunologia , Bifidobacterium longum/metabolismo , Disbiose/microbiologia , Humanos , Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/complicações , Obesidade/metabolismo , Obesidade/patologia , Oligossacarídeos/metabolismo , Osteoartrite/etiologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Transcriptoma/genética
6.
Science ; 359(6371): 104-108, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29302014

RESUMO

Anti-PD-1-based immunotherapy has had a major impact on cancer treatment but has only benefited a subset of patients. Among the variables that could contribute to interpatient heterogeneity is differential composition of the patients' microbiome, which has been shown to affect antitumor immunity and immunotherapy efficacy in preclinical mouse models. We analyzed baseline stool samples from metastatic melanoma patients before immunotherapy treatment, through an integration of 16S ribosomal RNA gene sequencing, metagenomic shotgun sequencing, and quantitative polymerase chain reaction for selected bacteria. A significant association was observed between commensal microbial composition and clinical response. Bacterial species more abundant in responders included Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium. Reconstitution of germ-free mice with fecal material from responding patients could lead to improved tumor control, augmented T cell responses, and greater efficacy of anti-PD-L1 therapy. Our results suggest that the commensal microbiome may have a mechanistic impact on antitumor immunity in human cancer patients.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunoterapia/métodos , Melanoma/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Neoplasias Cutâneas/terapia , Animais , Anticorpos Monoclonais/uso terapêutico , Bifidobacterium longum/classificação , Bifidobacterium longum/genética , Bifidobacterium longum/imunologia , Bifidobacterium longum/isolamento & purificação , Enterococcus faecium/classificação , Enterococcus faecium/genética , Enterococcus faecium/imunologia , Enterococcus faecium/isolamento & purificação , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Melanoma/imunologia , Camundongos , RNA Ribossômico 16S/genética , Neoplasias Cutâneas/imunologia , Linfócitos T/imunologia
7.
Hum Vaccin Immunother ; 14(1): 159-162, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29048978

RESUMO

Despite the revolutionary progress of immune checkpoint inhibitors (CPIs) for cancer immunotherapy, CPIs are effective only in a subset of patients. Combining CPIs and cancer vaccines to achieve better clinical outcomes is a reasonable approach since CPI enhances cancer vaccine-induced tumor-associated antigen (TAA) specific CTL. Among the various TAAs so far identified, WT1 protein is one of the most promising TAAs as a cancer vaccine target. Until now clinical trials of WT1 vaccine have demonstrated only modest clinical efficacy. These WT1 vaccines were based on peptides or dendritic cells (DCs), and there was no oral cancer vaccine. Recently, we developed a WT1 oral cancer vaccine using a recombinant Bifidobacterium displaying WT1 protein, which can efficiently deliver WT1 protein to the gut immune system, and we demonstrated that this oral cancer vaccine had a significant anti-tumor effect in a C1498-WT1 murine leukemia syngeneic tumor model. The WT1 protein displayed in this vaccine consists of about 70% of the WT1 amino acid sequence including multiple known CD4 and CD8 T-cell epitopes of WT1. In this commentary, we introduce our recent data indicating the superior anti-tumor effect of a WT1 oral cancer vaccine delivering WT1 protein to the gut immune system compared to a peptide vaccine.


Assuntos
Bifidobacterium longum/imunologia , Vacinas Anticâncer/uso terapêutico , Imunoterapia/métodos , Leucemia/terapia , Oligopeptídeos/imunologia , Proteínas Repressoras/imunologia , Administração Oral , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Trato Gastrointestinal/imunologia , Humanos , Leucemia/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Repressoras/genética , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico , Proteínas WT1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA