Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 324
Filtrar
1.
Benef Microbes ; 15(3): 241-258, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38688490

RESUMO

Aging is a physiological and immunological process involving the deterioration of human health, characterised by the progressive alteration of organs and their functions. The speed and extent of such decline are dependent on lifestyle, environment, and genetic factors. Moreover, with advancing age, humans become progressively more fragile and prone to acute and chronic diseases. Although the intestinal microbiota is predisposed to perturbations that accompany aging and frailty, it is generally accepted that the gut microbiota engages in multiple interactions that affect host health throughout the host life span. In the current study, an exhaustive in silico investigation of gut-associated bifidobacteria in healthy individuals from birth to old age revealed that Bifidobacterium longum subsp. longum is the most prevalent member, especially during infancy and in centenarians. Moreover, B. longum subsp. longum genome reconstruction and strain tracing among human gut microbiomes allowed the identification of prototypes of this taxon in the human gut microbiota of healthy elderly individuals. Such analyses guided culturomics attempts to isolate B. longum subsp. longum strains that matched the genomic content of B. longum subsp. longum prototypes from healthy elderly individuals. The molecular effects of selected B. longum subsp. longum strains on the human host were further investigated using in vitro microbe-host interactions, revealing differences in the host immune system transcriptome, with a reduction in gene expression of inflammation-related cytokines. These intriguing findings support the potential anti-aging effects of elderly associated prototypes of B. longum subsp. longum.


Assuntos
Bifidobacterium , Microbioma Gastrointestinal , Imunidade Inata , Humanos , Bifidobacterium/genética , Bifidobacterium/imunologia , Idoso de 80 Anos ou mais , Idoso , Lactente , Envelhecimento/imunologia , Pré-Escolar , Adulto , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Masculino , Feminino , Genoma Bacteriano/genética , Interações entre Hospedeiro e Microrganismos/imunologia
2.
São Paulo; s.n; s.n; 2024. 103 p tab, graf.
Tese em Português | LILACS | ID: biblio-1563094

RESUMO

Os sintomas depressivos durante a gravidez e o período pós-parto (PP) são prevalentes e podem ter implicações profundas para o bem-estar materno e infantil. Evidências emergentes sugerem que a microbiota intestinal pode desempenhar um papel na regulação do humor. Este estudo explora a relação entre a composição da microbiota intestinal e os sintomas depressivos em mulheres grávidas e no pós-parto com diferentes intensidade de sintomas. Foram recrutadas gestantes que faziam acompanhamento nos hospitais HCFMUSP e HU- USP. A partir do preenchimento do questionário de Escala de Edimburgo as participantes foram triadas para os grupos de sintomas ausentes ou leves (AL) e sintomas graves ou moderados (MG). Para a análise de microbiota, as participantes forneceram amostras de fezes em três momentos diferentes. Uma no terceiro trimestre de gestação (G) e duas no período pós-parto. A primeira amostra deste período foi coletada durante a internação do pós-parto (P1), e a segunda durante a consulta de retorno um mês após o parto (P2). A composição da microbiota intestinal foi analisada usando técnicas de sequenciamento de alto rendimento e os ácidos graxos de cadeia curta (AGCC) foram quantificados por cromatografia gasosa acoplada à espectrometria de massas (GC-MS). Análises bioinformáticas e estatísticas foram realizadas utilizando os softwares QIIME 2 (2022.2) e R (4.3.1) para identificar possíveis associações entre a composição da microbiota intestinal e a gravidade dos sintomas depressivos. Os resultados indicam que a familia Enterobacteriacea aparece com maior abundância nas mulheres do grupo MG, especialmente durante o período P1 (p<0,05) e que há uma diminuição significativa (p<0,05) de sintomas depressivos nas participantes do grupo MG desde sua triagem até o fim do acompanhamento do estudo, indicando que conduta terapêutica está sendo eficaz. Apesar de não ter sido estabelecida diferença estatística na abundância relativa da microbiota entre os grupos durante a gestação e nos índices de alfa e beta diversidade entre grupos e entre os períodos, é possivel observar uma tendência de mudança de microbiota ao longo do tratamento com aumento do gênero Bifidobacterium, diminuição da familia Enterobacteraceae e é possivel observar uma aparente correlação inversa entre a diminuição da intensidade de sintomas depressivos e o aumento da abundância dos gêneros Bifidobacterium e Clostridium, além do aumento das concentrações de AGCC. Em conclusão, a composição da microbiota intestinal parece ser influenciada pela gravidade dos sintomas depressivos em mulheres grávidas e no pós-parto. Pesquisas adicionais são necessárias para explorar a relação entre a microbiota intestinal e a depressão perinatal e determinar as implicações clínicas dessas descobertas para a saúde materna e infantil.


Depressive symptoms during pregnancy and the postpartum period (PP) are prevalent and can have profound implications for maternal and infant well-being. Emerging evidence suggests that the gut microbiota may play a role in mood regulation. This study explores the relationship between gut microbiota composition and depressive symptoms in pregnant and postpartum women with different symptom severities. A cohort of pregnant women were recruited from HCFMUSP and HU-USP. Participants completed standardized depression assessment tools and were allocate in groups of absent or mild depressive symptoms (AL) and moderate or severe depressive symptoms (MG) and provided stool samples in three different time periods. One at the third gestation trimester (G) and two at the postpartum period. The first sample from this period was collected during postpartum hospitalization(P1), and the second during the onemonth postpartum follow-up appointment (P2). Their gut microbiota composition was analyzed using high-throughput sequencing techniques and Gas chromatography mass spectrometry (GS-MS) for quantification of short-chain fatty acids (SCFAs). Bioinformatic and statistical analyses were performed using softwares QIIME 2 (2022.2) and R (4.3.1) to identify potential associations between gut microbiota composition and depressive symptom severity. Findings that the Enterobacteriaceae family appears more abundantly in women of the MG group, especially during period P1 (p<0.05), and that there is a significant decrease (p<0.05) in depressive symptoms among the participants of the MG group from their screening to the end of the study follow-up, suggesting that the therapeutic approach is effective. Although no statistical differences in alpha and beta diversity indices were established between groups and across periods, it is possible to observe a trend of microbiota change during the treatment, with an increase in the Bifidobacterium genus, a decrease in the Enterobacteriaceae family, and an apparent inverse correlation between the reduction in the intensity of depressive symptoms and the increased abundance of the Bifidobacterium, Clostridium, and Dorea genera, as well as an increase in the concentrations of SCFAs. In conclusion, composition of gut microbiota appears to be influenced by the severity of depressive symptoms in pregnant and postpartum women. Further research is warranted to explore links between gut microbiota and perinatal depression and to determine the clinical implications of these findings for maternal and infant health


Assuntos
Humanos , Feminino , Gravidez , Gravidez , Depressão/patologia , Microbioma Gastrointestinal/imunologia , Obstetrícia/classificação , Encaminhamento e Consulta/classificação , Espectrometria de Massas/métodos , Bifidobacterium/imunologia , Cromatografia Gasosa/instrumentação , Hospitais/classificação , Bem-Estar do Lactente/classificação , Cromatografia Gasosa-Espectrometria de Massas/métodos , Bem-Estar Materno/classificação
3.
Probiotics Antimicrob Proteins ; 15(4): 868-879, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35113319

RESUMO

Sensing of the intestinal microbiota by the host immune system is important to induce protective immune responses. Hence, modification of the gut microbiota might be able to prevent or treat allergies, mediated by proinflammatory Th2 immune responses. The aim was to investigate the ex vivo immunomodulatory effects of the synbiotics Pollagen® and Kallergen®, containing the probiotic bacterial strains Lactobacillus, Lacticaseibacillus and Bifidobacterium, in the context of grass pollen allergy. Peripheral blood mononuclear cells (PBMCs) from grass pollen-allergic patients and healthy controls were stimulated with grass pollen extract (GPE) and synbiotics and Gata3 expression and cytokine secretion analyzed. Monocyte-derived dendritic cells (MoDCs) cells were matured in the presence of GPE and synbiotics, co-cultured with autologous naïve T cells and maturation markers and cytokine secretion analyzed. GPE stimulation of PBMCs from grass pollen-allergic patients resulted in a significant higher production of the Th2 cytokines IL-4, IL-5, IL-9 and IL-13 compared to healthy controls. Gata3+CD4+ T cell induction was independent of the allergic status. The synbiotics promoted IL-10 and IFN-γ secretion and downregulated the GPE-induced Th2-like phenotype. Co-culturing naïve T cells with MoDCs, matured in the presence of GPE and synbiotics, shifted the GPE-induced Th2 cytokine release towards Th1-Th17-promoting conditions in allergic subjects. The investigated synbiotics are effective in downregulating the GPE-induced Th2 immune response in PBMCs from grass pollen-allergic patients as well as in autologous MoDC-T cell stimulation assays. In addition to increased IL-10 release, the data indicates a shift from a Th2- to a more Th1- and Th17-like phenotype.


Assuntos
Bifidobacterium , Células Dendríticas , Leucócitos Mononucleares , Rinite Alérgica Sazonal , Simbióticos , Humanos , Bifidobacterium/imunologia , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Lacticaseibacillus/imunologia , Lactobacillus/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Poaceae/imunologia , Pólen/imunologia , Rinite Alérgica Sazonal/imunologia , Rinite Alérgica Sazonal/microbiologia , Imunomodulação/imunologia , Células Cultivadas
4.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055188

RESUMO

In a previous study, we uncovered three immune-responsive patterns of gut microbes using an in vitro mesenteric lymph node cell suspension model, abbreviated as the MLN model hereafter. We used Akkermansia muciniphila and Clostridium butyricum as the first group directly inducing an immune response, Bifidobacterium sp. and Bacteroides sp. as the second group evoking an immune response with the help of stimuli (anti-CD3 and anti-CD28 antibodies), and Lactobacillus sp. as the third group blunting the immune response with or without stimuli. Our group previously clarified the immune-activation characteristics of A. muciniphila and linked its in vivo immune induction effect in GF and SPF mice under homeostasis. In the present study, we supplemented the characteristics of C. butyricum and B. bifidum in the in vitro MLN model and addressed the specific elements of the model. Finally, we used an in vivo TNBS-challenge model to show the functional differences between these species with different response patterns in vitro. The results showed that C. butyricum and B. bifidum evoked an immune response in vitro in a dose-dependent and strain-unique manner. Although TLR2, rather than TLR4, is indispensable for immune activation in the present in vitro model, it may not involve interaction between TLR2 and bacterial ligands. Like the PBMC model, the present in vitro MLN model is highly dependent on cell resources and should be given more attention when used to conduct a quantitative comparison. Finally, a mixture of two strong immunogenic strains, A. muciniphila and C. butyricum, significantly increased the mortality of TNBS-challenged (2,4,6-trinitrobenzene sulfonic acid, TNBS) mice, indicating a possible link between the in vitro MLN model and in vivo functional evaluation. However, more evidence is needed to clarify the associations and underlying mechanisms.


Assuntos
Bifidobacterium/imunologia , Clostridium butyricum/imunologia , Linfonodos/citologia , Ácido Trinitrobenzenossulfônico/efeitos adversos , Animais , Técnicas de Cocultura , Linfonodos/imunologia , Linfonodos/microbiologia , Masculino , Mesentério , Camundongos , Modelos Biológicos , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
5.
Front Immunol ; 12: 736196, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867956

RESUMO

The potential therapeutic effects of probiotic bacteria in rheumatoid arthritis (RA) remain controversial. Thus, this study aimed to discover potential therapeutic bacteria based on the relationship between the gut microbiome and rheumatoid factor (RF) in RA. Bacterial genomic DNA was extracted from the fecal samples of 93 RA patients and 16 healthy subjects. Microbiota profiling was conducted through 16S rRNA sequencing and bioinformatics analyses. The effects of Bifidobacterium strains on human peripheral blood mononuclear cells and collagen-induced arthritis (CIA) mice were assessed. Significant differences in gut microbiota composition were observed in patients with different RF levels. The relative abundance of Bifidobacterium and Collinsella was lower in RF-high than in RF-low and RF-negative RA patients, while the relative abundance of Clostridium of Ruminococcaceae family was higher in RF-high than in RF-low and RF-negative patients. Among 10 differentially abundant Bifidobacterium, B. longum RAPO exhibited the strongest ability to inhibit IL-17 secretion. Oral administration of B. longum RAPO in CIA mice, obese CIA, and humanized avatar model significantly reduced RA incidence, arthritis score, inflammation, bone damage, cartilage damage, Th17 cells, and inflammatory cytokine secretion. Additionally, B. longum RAPO significantly inhibited Th17 cells and Th17-related genes-IL-17A, IRF4, RORC, IL-21, and IL-23R-in the PBMCs of rheumatoid arthritis patients. Our findings suggest that B. longum RAPO may alleviate RA by inhibiting the production of IL-17 and other proinflammatory mediators. The safety and efficacy of B. longum RAPO in patients with RA and other autoimmune disorders merit further investigation.


Assuntos
Artrite Reumatoide/imunologia , Artrite Reumatoide/terapia , Bifidobacterium/imunologia , Bifidobacterium/isolamento & purificação , Microbioma Gastrointestinal/imunologia , Probióticos/uso terapêutico , Fator Reumatoide/sangue , Adulto , Animais , Artrite Experimental/imunologia , Artrite Experimental/terapia , Bifidobacterium/genética , Biodiversidade , Estudos de Casos e Controles , Feminino , Microbioma Gastrointestinal/genética , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Endogâmicos NOD , Camundongos Obesos , Camundongos SCID , Pessoa de Meia-Idade , Células Th17/imunologia
6.
Front Immunol ; 12: 741371, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34650564

RESUMO

Delivery by cesarean section (CS) is linked to an increased incidence of food allergies in children and affects early gut microbiota colonization. Furthermore, emerging evidence has connected disordered intestinal microbiota to food allergies. Here, we investigated the impact of CS on a rat model for food allergy to ovalbumin (OVA). Rats delivered by CS were found to be more responsive to OVA sensitization than vaginally born ones, displaying a greater reduction in rectal temperature upon challenge, worse diarrhea, and higher levels of OVA-specific antibodies and histamine. 16S rRNA sequencing of feces revealed reduced levels of Lactobacillus and Bifidobacterium in the CS rats. Preventative supplementation with a probiotic combination containing Lactobacillus and Bifidobacterium could protect CS rats against an allergic response to OVA, indicating that the microbiota dysbiosis contributes to CS-related response. Additionally, probiotic intervention early in life might help to rebuild aberrant Th2 responses and tight junction proteins, both of which have been linked to CS-related high allergic reactions. Taken together, this study shows that disordered intestinal microbiota plays an essential role in the pathogenesis of food allergy mediated by CS. More importantly, interventions that modulate the microbiota composition in early life are therapeutically relevant for CS-related food allergies.


Assuntos
Bifidobacterium/imunologia , Cesárea/estatística & dados numéricos , Disbiose/prevenção & controle , Hipersensibilidade Alimentar/prevenção & controle , Microbioma Gastrointestinal/imunologia , Lactobacillus/imunologia , Probióticos/administração & dosagem , RNA Ribossômico 16S/genética , Células Th2/imunologia , Alérgenos/imunologia , Animais , Bifidobacterium/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Hipersensibilidade Alimentar/epidemiologia , Humanos , Imunoglobulina E/sangue , Lactobacillus/genética , Masculino , Ovalbumina/imunologia , Gravidez , Ratos , Ratos Sprague-Dawley , Junções Íntimas/metabolismo
7.
Ann Allergy Asthma Immunol ; 127(2): 183-190, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33971355

RESUMO

OBJECTIVE: To present a comprehensive, clinically focused scoping review of therapeutic agents and practices comprising the future of allergic rhinitis (AR) management. DATA SOURCES: A review of the published literature was performed using the PubMed database, published abstracts, and virtual presentations from scientific meetings and posted results on ClinicalTrials.gov. STUDY SELECTIONS: Primary manuscripts with trial results, case reports, case series, and clinical trial data from ClinicalTrials.gov, PubMed, and articles highlighting expert perspectives on management of AR were selected. RESULTS: Telemedicine, social media, and mHealth facilitate integrated care for AR management. Pharmacotherapy remains the standard of care for AR management; however, treatment combinations are recommended. Intralymphatic immunotherapy and peptide immunotherapy are the most promising new allergen immunotherapy options. Studies of targeted biologics for AR are ongoing. Probiotics may be beneficial for AR management, particularly Bifidobacterium spp, and as an add-on to allergen immunotherapy. CONCLUSION: AR is a chronic and often comorbid condition that requires integrated care for optimal management. New formulations and combinations of existing AR therapies are the most promising and merit future research.


Assuntos
Alérgenos/administração & dosagem , Antialérgicos/uso terapêutico , Dessensibilização Imunológica/métodos , Probióticos/uso terapêutico , Rinite Alérgica/terapia , Bifidobacterium/imunologia , Gerenciamento Clínico , Fluticasona/uso terapêutico , Humanos , Injeções Intralinfáticas , Ftalazinas/uso terapêutico , Rinite Alérgica/imunologia
8.
J Microbiol Biotechnol ; 30(12): 1793-1800, 2020 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-33144551

RESUMO

Bifidobacterium strains can provide several health benefits, such as antimicrobial and immunomodulatory effects. Some strains inhibit growth or cell adhesion of pathogenic bacteria, including multidrug-resistant bacteria, and their antibacterial activity can be intensified when combined with certain antibiotics. In addition, some strains of bifidobacteria reduce viral infectivity, leading to less epithelial damage of intestinal tissue, lowering the virus shedding titer, and controlling the release of antiviral substances. Furthermore, bifidobacteria can modulate the immune system by increasing immunoglobulins, and inducing or reducing pro- or antiinflammatory cytokines, respectively. In particular, these anti-inflammatory effects are helpful in the treatment of patients who are already suffering from infection or inflammatory diseases. This review summarizes the antimicrobial effects and mechanisms, and immunomodulatory effects of Bifidobacterium strains, suggesting the potential of bifidobacteria as an alternative or complementary treatment option.


Assuntos
Antibacterianos/farmacologia , Bifidobacterium/imunologia , Imunomodulação , Probióticos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Antibiose , Antivirais/farmacologia , Bactérias , Citocinas , Farmacorresistência Bacteriana Múltipla , Células Epiteliais/microbiologia , Trato Gastrointestinal/microbiologia , Humanos , Intestinos/microbiologia
9.
Proc Natl Acad Sci U S A ; 117(44): 27509-27515, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33077598

RESUMO

Immune checkpoint-blocking antibodies that attenuate immune tolerance have been used to effectively treat cancer, but they can also trigger severe immune-related adverse events. Previously, we found that Bifidobacterium could mitigate intestinal immunopathology in the context of CTLA-4 blockade in mice. Here we examined the mechanism underlying this process. We found that Bifidobacterium altered the composition of the gut microbiota systematically in a regulatory T cell (Treg)-dependent manner. Moreover, this altered commensal community enhanced both the mitochondrial fitness and the IL-10-mediated suppressive functions of intestinal Tregs, contributing to the amelioration of colitis during immune checkpoint blockade.


Assuntos
Doenças Autoimunes/prevenção & controle , Bifidobacterium/imunologia , Microbioma Gastrointestinal/imunologia , Probióticos/administração & dosagem , Linfócitos T Reguladores/imunologia , Animais , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Tolerância Imunológica , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Camundongos Knockout , Linfócitos T Reguladores/metabolismo
10.
Curr Opin Allergy Clin Immunol ; 20(2): 138-148, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32004178

RESUMO

PURPOSE OF REVIEW: The body's largest microbial community, the gut microbiome, is in contact with mucosal surfaces populated with epithelial, immune, endocrine and nerve cells, all of which sense and respond to microbial signals. These mutual interactions have led to a functional coevolution between the microbes and human physiology. Examples of coadaptation are anaerobes Bifidobacteria and Bacteroides, which have adjusted their metabolism to dietary components of human milk, and infant immune development, which has evolved to become reliant on the presence of beneficial microbes. Current research suggests that specific composition of the early-life gut microbiome aligns with the maturation of host immunity. Disruptions of natural microbial succession patterns during gut colonization are a consistent feature of immune-mediated diseases, including atopy and asthma. RECENT FINDINGS: Here, we catalog recent birth cohorts documenting associations between immune dysregulation and microbial alterations, and summarize the evidence supporting the role of the gut microbiome as an etiological determinant of immune-mediated allergic diseases. SUMMARY: Ecological concepts that describe microbial dynamics in the context of the host environment, and a portray of immune and neuroendocrine signaling induced by host-microbiome interactions, have become indispensable in describing the molecular role of early-life microbiome in atopy and asthma susceptibility.


Assuntos
Asma/imunologia , Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Leite Humano/imunologia , Asma/microbiologia , Bacteroides/imunologia , Bifidobacterium/imunologia , Aleitamento Materno , Desenvolvimento Infantil/fisiologia , Suscetibilidade a Doenças , Exposição Ambiental/efeitos adversos , Humanos , Lactente , Neuroimunomodulação/imunologia , Sistemas Neurossecretores/imunologia
11.
Biomed Res Int ; 2019: 2323540, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31119156

RESUMO

Bifidobacterium animalis subsp. lactis IPLA 20020 and Lactobacillus gasseri IPLA 20212, two strains isolated from human samples, were evaluated for safety and influence over the intestinal microbiota and cytokine production by the intestinal tissue of adult BALB/c mice. Mice were divided into four groups receiving during 8 days PBS or a suspension of each strain, prepared fresh or lyophilized (bifidobacteria), at an amount of 4x108 viable cells/day. This dose could be comparable to the probiotic intake of a human adult who consumed about 100-200 mL of functional fermented milk per day, considering the usual level of probiotics in commercial products. No microbial translocation to liver or alterations in food intake, weight, and behavior were observed in treated mice. Intestinal content of secretory immunoglobulin A (s-IgA) was not affected, discarding any adverse effect on the mucosa-associated immunity. The profile of intestinal proinflammatory/regulatory cytokines after intervention evidenced that the microbial strain administered and its cellular state (fresh or lyophilized) as well as the host tissue analyzed (small or large intestine) influenced the immune response and suggests a moderate shift towards a T helper 1 profile (Th1) in the large intestine after the administration of both strains. Changes on relative levels of some intestinal microbial groups were evidenced after intervention. It is noteworthy that butyrate was positively associated with a balanced pro-Th1 immune response. Therefore, B. animalis subsp. lactis IPLA20020 and L. gasseri IPLA 20212 could be considered potential probiotic candidates to be included in functional foods for balancing the intestinal immune response.


Assuntos
Bifidobacterium/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas/imunologia , Lactobacillus/imunologia , Animais , Bifidobacterium/crescimento & desenvolvimento , Fermentação , Humanos , Imunomodulação/genética , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Lactobacillus/crescimento & desenvolvimento , Camundongos , Probióticos , Células Th1/imunologia , Células Th1/microbiologia
12.
Benef Microbes ; 10(2): 199-209, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30860402

RESUMO

Previously, we reported that the non-viable immunomodulatory Bifidobacterium infantis MCC12 and Bifidobacterium breve MCC1274 strains (paraimmunobiotic bifidobacteria) were able to increase the protection against rotavirus infection in bovine intestinal epithelial (BIE) cells. In order to gain insight into the influence of paraimmunobiotic bifidobacteria on the innate antiviral immune response of BIE cells, their effect on the transcriptomic response triggered by Toll-like receptor 3 (TLR3) activation was investigated. By using microarray technology and qPCR analysis, we obtained a global overview of the immune genes involved in the innate antiviral immune response in BIE cells. Activation of TLR3 by poly(I:C) in BIE cells significantly increased the expression of interferon (IFN)-α and IFN-ß, several interferon-stimulated genes, cytokines, and chemokines. It was also observed that both paraimmunobiotic bifidobacteria differently modulated immune genes expression in poly(I:C)-challenged BIE cells. Most notable changes were found in genes involved in antiviral defence (IFN-ß, MX1, OAS1X, MDA5, TLR3, STAT2, STAT3), cytokines (interleukin (IL)-6), and chemokines (CCL2, CXCL2, CXCL6) that were significantly increased in bifidobacteria-treated BIE cells. B. infantis MCC12 and B. breve MCC1274 showed quantitative and qualitative differences in their capacities to modulate the innate antiviral immune response in BIE cells. B. breve MCC1274 was more efficient than the MCC12 strain to improve the production of type I IFNs and antiviral factors, an effect that could be related to its higher ability to protect against rotavirus replication in BIE cells. Interestingly, B. infantis MCC12 showed a remarkable anti-inflammatory effect. The MCC12 strain was more efficient to reduce the expression of inflammatory cytokines and chemokines (IL-16, IL-20, CX3CL1) when compared with B. breve MCC1274. These results provided valuable information for the deeper understanding of the antiviral immune response of intestinal epithelial cells as well as the host-paraimmunobiotic interaction in the bovine host.


Assuntos
Bifidobacterium/imunologia , Células Epiteliais/imunologia , Perfilação da Expressão Gênica , Imunidade Inata , Mucosa Intestinal/imunologia , Probióticos/metabolismo , Rotavirus/imunologia , Animais , Bovinos , Linhagem Celular , Fatores Imunológicos/metabolismo , Modelos Biológicos , Reação em Cadeia da Polimerase em Tempo Real
13.
Ann Neurol ; 83(6): 1147-1161, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29679417

RESUMO

OBJECTIVE: Effect of a probiotic on the gut microbiome and peripheral immune function in healthy controls and relapsing-remitting multiple sclerosis (MS) patients. METHODS: MS patients (N = 9) and controls (N = 13) were orally administered a probiotic containing Lactobacillus, Bifidobacterium, and Streptococcus twice-daily for two months. Blood and stool specimens were collected at baseline, after completion of the 2-month treatment, and 3 months after discontinuation of therapy. Frozen peripheral blood mononuclear cells (PBMCs) were used for immune cell profiling. Stool samples were used for 16S rRNA profiling and metabolomics. RESULTS: Probiotic administration increased the abundance of several taxa known to be depleted in MS such as Lactobacillus. We found that probiotic use decreased the abundance of taxa previously associated with dysbiosis in MS, including Akkermansia and Blautia. Predictive metagenomic analysis revealed a decrease in the abundance of several KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways associated with altered gut microbiota function in MS patients, such as methane metabolism, following probiotic supplementation. At the immune level, probiotic administration induced an anti-inflammatory peripheral immune response characterized by decreased frequency of inflammatory monocytes, decreased mean fluorescence intensity (MFI) of CD80 on classical monocytes, as well as decreased human leukocyte antigen (HLA) D related MFI on dendritic cells. Probiotic administration was also associated with decreased expression of MS risk allele HLA-DQA1 in controls. Probiotic-induced increase in abundance of Lactobacillus and Bifidobacterium was associated with decreased expression of MS risk allele HLA.DPB1 in controls. INTERPRETATION: Our results suggest that probiotics could have a synergistic effect with current MS therapies. Ann Neurol 2018.


Assuntos
Bifidobacterium/imunologia , Microbiota/imunologia , Esclerose Múltipla/genética , Probióticos/metabolismo , Adulto , Bifidobacterium/genética , Feminino , Microbioma Gastrointestinal/fisiologia , Humanos , Lactobacillus/genética , Lactobacillus/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Microbiota/genética , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , RNA Ribossômico 16S/genética , Adulto Jovem
14.
Nat Rev Clin Oncol ; 15(6): 382-396, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29636538

RESUMO

Discoveries made in the past 5 years indicate that the composition of the intestinal microbiota has a major influence on the effectiveness of anticancer immunosurveillance and thereby contributes to the therapeutic activity of immune-checkpoint inhibitors that target cytotoxic T lymphocyte protein 4 (CTLA-4) or the programmed cell death protein 1 (PD-1)-programmed cell death 1 ligand 1 (PD-L1) axis, as well as the activity of immunogenic chemotherapies. Herein, we highlight some of the bacteria, such as Akkermansia muciniphila, Bacteroides fragilis, Bifidobacterium spp. and Faecalibacterium spp., that have been associated with favourable anticancer immune responses in both preclinical tumour models and patients with cancer. Importantly, these bacteria also seem to have a positive influence on general health, thus reducing the incidence of metabolic disorders and a wide range of chronic inflammatory pathologies. We surmise that a diverse and propitious microbial ecosystem favours organismal homeostasis, particularly at the level of the cancer-immune dialogue.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade Inata , Neoplasias/microbiologia , Neoplasias/terapia , Antígeno B7-H1/imunologia , Antígeno B7-H1/uso terapêutico , Bacteroides fragilis/imunologia , Bacteroides fragilis/metabolismo , Bifidobacterium/imunologia , Bifidobacterium/metabolismo , Antígeno CTLA-4/imunologia , Antígeno CTLA-4/uso terapêutico , Faecalibacterium/imunologia , Faecalibacterium/metabolismo , Humanos , Monitorização Imunológica , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/uso terapêutico
15.
Proc Natl Acad Sci U S A ; 115(1): 157-161, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29255057

RESUMO

Antibodies that attenuate immune tolerance have been used to effectively treat cancer, but they can also trigger severe autoimmunity. To investigate this, we combined anti-CTLA-4 treatment with a standard colitis model to give mice a more severe form of the disease. Pretreatment with an antibiotic, vancomycin, provoked an even more severe, largely fatal form, suggesting that a Gram-positive component of the microbiota had a mitigating effect. We then found that a commonly used probiotic, Bifidobacterium, could largely rescue the mice from immunopathology without an apparent effect on antitumor immunity, and this effect may be dependent on regulatory T cells.


Assuntos
Bifidobacterium/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Colite/imunologia , Colite/terapia , Animais , Anticorpos Neutralizantes/efeitos adversos , Anticorpos Neutralizantes/farmacologia , Antígeno CTLA-4/imunologia , Colite/induzido quimicamente , Colite/genética , Camundongos , Camundongos Transgênicos , Vancomicina/efeitos adversos , Vancomicina/farmacologia
16.
ISME J ; 11(12): 2834-2847, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28837128

RESUMO

Internally transcribed spacer (ITS) rRNA profiling is a novel tool for detailed analysis of microbial populations at low taxonomic ranks. Here we exploited this approach to explore species-level biogeography of the Bifidobacterium genus across 291 adult mammals. These include humans and 13 other primates, domesticated animals, such as dogs, cats, cows, sheep, goats, horses and pigs, and 46 additional species. The collected profiles revealed the presence of 89 putative novel bifidobacterial taxa in addition to 45 previously described species. Remarkably, in contrast to what is currently known for many gut commensals, we did not observe host-specialization among bifidobacterial species but rather their widespread distribution across mammals. Moreover, ITS rRNA profiling of wild relatives of domesticated dogs, rabbits and pigs clearly indicates that domestication and close contact with humans have impacted on the composition of the fecal bifidobacterial population. These data were complemented by analysis of bifidobacterial communities in milk of eight mammalian families, showing that bifidobacteria represent prototypical early gut microbiota members which are inherited by newborns from their lactating mother. Thus this study highlights the role of bifidobacteria as pioneering gut colonizers of a wide range of mammals.


Assuntos
Bifidobacterium/classificação , Bifidobacterium/imunologia , Bifidobacterium/isolamento & purificação , Mamíferos/microbiologia , Adulto , Animais , Bifidobacterium/genética , Gatos , Bovinos , Cães , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal , Cavalos , Humanos , Mamíferos/classificação , Coelhos , Ovinos , Suínos
17.
Microbiol Spectr ; 5(3)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28643627

RESUMO

Bifidobacteria are members of the intestinal microbiota of mammals and other animals, and some strains are able to exert health-promoting effects. The genus Bifidobacterium belongs to the Actinobacteria phylum. Firmicutes, Bacteroidetes, and Actinobacteria constitute the most abundant phyla in the human intestinal microbiota, Firmicutes and Bacteroidetes being predominant in adults, and Actinobacteria in breast-fed infants, where bifidobacteria can reach levels higher than 90% of the total bacterial population. They are among the first microbial colonizers of the intestines of newborns, and play key roles in the development of their physiology, including maturation of the immune system and use of dietary components. Indeed, some nutrients, such as human milk oligosaccharides, are important drivers of bifidobacterial development. Some Bifidobacterium strains are considered probiotic microorganisms because of their beneficial effects, and they have been included as bioactive ingredients in functional foods, mainly dairy products, as well as in food supplements and pharma products, alone, or together with, other microbes or microbial substrates. Well-documented scientific evidence of their activities is currently available for bifidobacteria-containing preparations in some intestinal and extraintestinal pathologies. In this review, we focus on the role of bifidobacteria as members of the human intestinal microbiota and their use as probiotics in the prevention and treatment of disease.


Assuntos
Bifidobacterium/classificação , Bifidobacterium/fisiologia , Gastroenteropatias/microbiologia , Microbioma Gastrointestinal , Intestinos/microbiologia , Adulto , Fatores Etários , Antibacterianos/uso terapêutico , Bifidobacterium/imunologia , Clostridioides difficile/patogenicidade , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/terapia , Laticínios/microbiologia , Diarreia/microbiologia , Diarreia/terapia , Tratamento Farmacológico , Enterocolite Necrosante/microbiologia , Enterocolite Necrosante/terapia , Gastroenteropatias/terapia , Trato Gastrointestinal/microbiologia , Infecções por Helicobacter/terapia , Helicobacter pylori/patogenicidade , Humanos , Lactente , Recém-Nascido , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/terapia , Síndrome do Intestino Irritável/microbiologia , Síndrome do Intestino Irritável/terapia , Interações Microbianas , Probióticos/uso terapêutico
18.
Cytokine ; 97: 141-148, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28648868

RESUMO

Lactobacilli and bifidobacteria play a primary role in modulation of gut immunity. By considering that microbiota composition depends on various factors, including diet, we asked whether functional differences could characterize faecal populations of lactobacilli and bifidobacteria isolated from individuals with different dietary habits. 155 healthy volunteers who followed omnivorous, ovo-lacto-vegetarian or vegan diets were recruited at four Italian centres (Turin, Parma, Bologna and Bari). Faecal samples were collected; lactobacilli and bifidobacteria were isolated on selective media and their immunomodulatory activity was tested in mouse dendritic cells (DCs). Pre-incubation with lactobacilli increased LPS-induced expression of the maturation markers CD80 and CD86, whereas pre-incubation with bifidobacteria decreased such expression. Analysis of the cytokine profile indicated that strains of both genera induced down-regulation of IL-12 and up-regulation of IL-10, whereas expression of TNF-α was not modulated. Notably, analysis of anti-inflammatory potential (IL-10/IL-12 ratio) showed that lactobacilli evoked a greater anti-inflammatory effect than did bifidobacteria in the omnivorous group (P<0.05). We also found significantly reduced anti-inflammatory potential in the bacterial strains isolated from Bari's volunteers in comparison with those from the cognate groups from the other centres. In conclusion, lactobacilli and bifidobacteria showed a genus-specific ability of modulating in vitro innate immunity associated with a specific dietary habit. Furthermore, the geographical area had a significant impact on the anti-inflammatory potential of some components of faecal microbiota.


Assuntos
Bifidobacterium/imunologia , Células Dendríticas/imunologia , Dieta Vegana , Dieta , Microbioma Gastrointestinal/imunologia , Imunomodulação , Lactobacillus/imunologia , Animais , Antígeno B7-1/genética , Antígeno B7-2/genética , Bifidobacterium/isolamento & purificação , Citocinas/genética , Células Dendríticas/microbiologia , Regulação para Baixo , Fezes/microbiologia , Humanos , Interleucina-10/genética , Interleucina-12/genética , Lactobacillus/isolamento & purificação , Camundongos , Fator de Necrose Tumoral alfa/genética , Regulação para Cima , Vegetarianos
19.
Cancer Immunol Immunother ; 66(6): 787-798, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28299466

RESUMO

Several types of vaccine-delivering tumor-associated antigens (TAAs) have been developed in basic and clinical research. Wilms' tumor 1 (WT1), identified as a gene responsible for pediatric renal neoplasm, is one of the most promising TAA for cancer immunotherapy. Peptide and dendritic cell-based WT1 cancer vaccines showed some therapeutic efficacy in clinical and pre-clinical studies but as yet no oral WT1 vaccine can be administrated in a simple and easy way. In the present study, we constructed a novel oral cancer vaccine using a recombinant Bifidobacterium longum displaying WT1 protein. B. longum 420 was orally administered into mice inoculated with WT1-expressing tumor cells for 4 weeks to examine anti-tumor effects. To analyze the WT1-specific cellular immune responses to oral B. longum 420, mice splenocytes were isolated and cytokine production and cytotoxic activities were determined. Oral administrations of B. longum 420 significantly inhibited WT1-expressing tumor growth and prolonged survival in mice. Immunohistochemical study and immunological assays revealed that B. longum 420 substantially induced tumor infiltration of CD4+T and CD8+T cells, systemic WT1-specific cytokine production, and cytotoxic activity mediated by WT1-epitope specific cytotoxic T lymphocytes, with no apparent adverse effects. Our novel oral cancer vaccine safely induced WT1-specific cellular immunity via activation of the gut mucosal immune system and achieved therapeutic efficacy with several practical advantages over existing non-oral vaccines.


Assuntos
Bifidobacterium/imunologia , Vacinas Anticâncer/administração & dosagem , Imunoterapia , Neoplasias Bucais/imunologia , Neoplasias Bucais/terapia , Proteínas WT1/imunologia , Animais , Vacinas Anticâncer/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Bucais/metabolismo , Neoplasias Bucais/mortalidade , Taxa de Sobrevida , Linfócitos T Citotóxicos/imunologia
20.
Cytokine ; 90: 80-87, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27863334

RESUMO

Enterocytes are actively involved in the defense against pathogens and they limit penetration of commensal microbes into tissues. They also have an important role in gut immunity as enterocytes confer mucosal dendritic cell specialisation. On the other hand, the microbiota is directly involved in the development and modulation of the intestinal immune system. Particularly, lactobacilli and bifidobacteria play a primary role in shaping the immune response. We further explored this issue by evaluating whether functional differences in Caco-2 cells could characterise faecal populations of lactobacilli (155 samples) and bifidobacteria (110 samples) isolated from three dietary cohorts (omnivores, ovo-lacto-vegetarians and vegans) recruited at four Italian centres (Turin, Parma, Bologna and Bari). According to our findings, tested bacteria were unable to modulate expression of IL-8, IL-10, TGF-ß or thymic stromal lymphopoietin (TSLP) cytokines in unstimulated Caco-2 cells. Conversely, in phorbol 12-myristate 13-acetate and ionomycin (PMA/Io) stimulated Caco-2 cells, lactobacilli from the omnivorous group and all bifidobacteria significantly down-regulated IL-8. Notably, both genera also lowered the TSLP expression in stimulated Caco-2 cells, regardless of the diet regimen. By further examining these data on the basis of geographical origin, we found that lactobacilli from the vegetarian group recruited in Bari, significantly up-regulated this cytokine. In conclusion, we highlighted a peculiar immune-modulatory activity profile for lactobacilli on enterocytes undergoing a stimulatory signal, which was associated with a specific dietary habit. Furthermore, the geographical area had a significant impact on the inflammatory potential of members of the Lactobacillus genus.


Assuntos
Bifidobacterium/imunologia , Citocinas/imunologia , Comportamento Alimentar , Preferências Alimentares , Interleucina-8/imunologia , Lactobacillus/imunologia , Adolescente , Adulto , Bifidobacterium/isolamento & purificação , Células CACO-2 , Feminino , Humanos , Lactobacillus/isolamento & purificação , Masculino , Pessoa de Meia-Idade , Acetato de Tetradecanoilforbol/farmacologia , Linfopoietina do Estroma do Timo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA