Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G969-G981, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33787352

RESUMO

Treg deficiency causes a lethal, CD4+ T cell-driven autoimmune disease called IPEX syndrome (immunodysregulation, polyendocrinopathy, and enteropathy, with X-linked inheritance) in humans and in the scurfy (SF) mouse, a mouse model of the disease. Feeding Limosilactobacillus reuteri DSM 17938 (LR 17938, LR) to SF mice reprograms the gut microbiota, reduces disease progression, and prolongs lifespan. However, the efficacy and mechanism of LR, compared with other probiotics, in producing these effects is unknown. We compared LR with Lacticaseibacillus rhamnosus GG (LGG), an extensively investigated probiotic. LR was more effective than LGG in prolonging survival. Both probiotics restored the fecal microbial alpha diversity, but they produced distinct fecal bacterial clusters and differentially modulated microbial relative abundance (RA). LR increased the RA of phylum_Firmicutes, genus_Oscillospira whereas LR reduced phylum_Bacteroidetes, genus_Bacteroides and genus_Parabacteroides, reversing changes attributed to the SF phenotype. LGG primarily reduced the RA of genus_Bacteroides. Both LR and LGG reduced the potentially pathogenic taxon class_γ-proteobacteria. Plasma metabolomics revealed substantial differences among 696 metabolites. We observed similar changes of many clusters of metabolites in SF mice associated with treatment with either LR or LGG. However, a unique effect of LR was to increase the abundance of plasma adenosine metabolites such as inosine, which we previously showed had immune modulatory effects. In conclusion: 1) different probiotics produce distinct signatures in the fecal microbial community in mice with Treg deficiency; and 2) when comparing different probiotics, there are strain-specific microbial products with different anti-inflammatory properties, reinforcing the concept that "one size does not fit all" in the treatment of autoimmune disease.NEW & NOTEWORTHY In the treatment of Treg-deficiency-induced autoimmunity, Limosilactobacillus reuteri DSM 17938 (LR) showed greater efficacy than Lacticaseibacillus rhamnosus GG (LGG). The study demonstrated that two different probiotics produce distinct signatures in the fecal microbial community in mice with Treg deficiency, but with many similarities in global plasma metabolites in general. However, there are strain-specific microbial products with different anti-inflammatory properties, reinforcing the concept that "one size does not fit all" in the treatment of autoimmune disease.


Assuntos
Diabetes Mellitus Tipo 1/congênito , Diarreia/microbiologia , Microbioma Gastrointestinal/imunologia , Doenças Genéticas Ligadas ao Cromossomo X/microbiologia , Doenças do Sistema Imunitário/congênito , Lacticaseibacillus rhamnosus , Limosilactobacillus reuteri , Linfócitos T Reguladores/imunologia , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/microbiologia , Diarreia/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Doenças do Sistema Imunitário/metabolismo , Doenças do Sistema Imunitário/microbiologia , Camundongos , Camundongos Transgênicos , Probióticos , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia
2.
Microbiome ; 7(1): 145, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31699146

RESUMO

BACKGROUND: Regulatory T cell (Treg) deficiency leads to IPEX syndrome, a lethal autoimmune disease, in Human and mice. Dysbiosis of the gut microbiota in Treg-deficient scurfy (SF) mice has been described, but to date, the role of the gut microbiota remains to be determined. RESULTS: To examine how antibiotic-modified microbiota can inhibit Treg deficiency-induced lethal inflammation in SF mice, Treg-deficient SF mice were treated with three different antibiotics. Different antibiotics resulted in distinct microbiota and metabolome changes and led to varied efficacy in prolonging lifespan and reducing inflammation in the liver and lung. Moreover, antibiotics altered plasma levels of several cytokines, especially IL-6. By analyzing gut microbiota and metabolome, we determined the microbial and metabolomic signatures which were associated with the antibiotics. Remarkably, antibiotic treatments restored the levels of several primary and secondary bile acids, which significantly reduced IL-6 expression in RAW macrophages in vitro. IL-6 blockade prolonged lifespan and inhibited inflammation in the liver and lung. By using IL-6 knockout mice, we further identified that IL-6 deletion provided a significant portion of the protection against inflammation induced by Treg dysfunction. CONCLUSION: Our results show that three antibiotics differentially prolong survival and inhibit lethal inflammation in association with a microbiota-IL-6 axis. This pathway presents a potential avenue for treating Treg deficiency-mediated autoimmune disorders.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1/congênito , Diarreia , Disbiose/microbiologia , Microbioma Gastrointestinal , Doenças Genéticas Ligadas ao Cromossomo X , Doenças do Sistema Imunitário/congênito , Inflamação , Animais , Antibacterianos/farmacologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/microbiologia , Doença Crônica , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/microbiologia , Diarreia/imunologia , Diarreia/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Doenças Genéticas Ligadas ao Cromossomo X/imunologia , Doenças Genéticas Ligadas ao Cromossomo X/microbiologia , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/microbiologia , Inflamação/imunologia , Inflamação/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/citologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-31565666

RESUMO

BACKGROUND AND OBJECTIVE: Breast milk has many growth-promoting and immune-active components, including transforming growth factor-ß, lactoferrin, lysozyme, immunoglobulin A, and prebiotics such as the human milk oligosaccharides. Treatment with Lactobacillus reuteri DSM 17938 (LR), a probiotic with immunomodulatory functions, significantly increases regulatory T cells (Tregs) in the intestinal mucosa of newborn suckling rats. In humans, treatment with LR of infants with colic reduces crying optimally if the infants are breast-fed. Therefore, we examined the effects of human breast milk (HBM) on LR-associated immune modulation. METHODS: Newborn rats were divided into 8 feeding groups, including dam-fed ± LR (106 CFU/kg bw/day, daily), formula-fed ± LR, formula with 20% (v/v) HBM-fed ± LR, and HBM-fed ± LR. Pups were fed by gavage from d1 to d3 of age. Subsequently, we measured intestinal immune cell profiles, including Tregs and tolerogenic dendritic cells (tDCs) by flow cytometry. We also measured inflammatory cytokine and chemokine levels of interleukin (IL)-1ß and cytokine-induced neutrophil chemoattratant (CINC)-1 in intestinal tissue lysates by ELISA. RESULTS AND CONCLUSION: (1) Formula feeding increased intestinal CD3+ T cells, CD4+ helper T (TH) cells and CD11c+ DCs, pro-inflammatory effects which were reversed by HBM. (2) When comparing HBM-fed with formula-fed newborns, HBM supplementation produced a lower percentage of CD4+ TH cells and a higher percentage of CD8+ (cytotoxic) T cells, while reducing protein levels of IL-1ß and CINC-1 in the intestine. (3) Probiotic LR feeding maximally stimulated the percentage of intestinal Tregs and tDCs when the pups were fed HBM. In conclusion, HBM reduced formula-induced intestinal gut immune activation, and the addition of LR further promoted immune tolerance.

4.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G824-G838, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31482733

RESUMO

Early administration of Lactobacillus reuteri DSM 17938 (LR) prevents necrotizing enterocolitis and inhibits regulatory T-cell (Treg)-deficiency-associated autoimmunity in mice. In humans, LR reduces crying time in breastfed infants with colic, modifies severity in infants with acute diarrheal illnesses, and improves pain in children with functional bowel disorders. In healthy breastfed newborns with evolving microbial colonization, it is unclear if early administration of LR can modulate gut microbiota and their metabolites in such a way as to promote homeostasis. We gavaged LR (107 colony-forming units/day, daily) to C57BL/6J mice at age of day 8 for 2 wk. Both male and female mice were investigated in these experiments. We found that feeding LR did not affect clinical phenotype or inflammatory biomarkers in plasma and stool, but LR increased the proportion of Foxp3+ regulatory T cells (Tregs) in the intestine. LR also increased bacterial diversity and the relative abundance of p_Firmicutes, f_Lachnospiraceae, f_Ruminococcaceae, and genera Clostridium and Candidatus arthromitus, while decreasing the relative abundance of p_Bacteriodetes, f_Bacteroidaceae, f_Verrucomicrobiaceae, and genera Bacteroides, Ruminococcus, Akkermansia, and Sutterella. Finally, LR exerted a major impact on the plasma metabolome, upregulating amino acid metabolites formed via the urea, tricarboxylic acid, and methionine cycles and increasing tryptophan metabolism. In conclusion, early oral administration of LR to healthy breastfed mice led to microbial and metabolic changes which could be beneficial to general health.NEW & NOTEWORTHY Oral administration of Lactobacillus reuteri DSM 17938 (LR) to healthy breastfed mice promotes intestinal immune tolerance and is linked to proliferation of beneficial gut microbiota. LR upregulates plasma metabolites that are involved in the urea cycle, the TCA cycle, methionine methylation, and the polyamine pathway. Herein, we show that LR given to newborn mice specifically increases levels of tryptophan metabolites and the purine nucleoside adenosine that are known to enhance tolerance to inflammatory stimuli.


Assuntos
Microbioma Gastrointestinal , Intestinos , Limosilactobacillus reuteri , Probióticos/administração & dosagem , Linfócitos T Reguladores , Triptofano/metabolismo , Adenosina/metabolismo , Administração Oral , Animais , Animais Recém-Nascidos , Intervenção Médica Precoce/métodos , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Intestinos/microbiologia , Intestinos/fisiologia , Limosilactobacillus reuteri/imunologia , Limosilactobacillus reuteri/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Interações Microbianas/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
5.
Front Immunol ; 10: 385, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30899262

RESUMO

The gut microbiome plays an important role in immune function and has been implicated in multiple sclerosis (MS). However, how and if the modulation of microbiota can prevent or treat MS remain largely unknown. In this study, we showed that probiotic Lactobacillus reuteri DSM 17938 (L. reuteri) ameliorated the development of murine experimental autoimmune encephalomyelitis (EAE), a widely used animal model of MS, a model which is primarily mediated by TH17 and TH1 cells. We discovered that L. reuteri treatment reduced TH1/TH17 cells and their associated cytokines IFN-γ/IL-17 in EAE mice. We also showed that the loss of diversity of gut microbiota induced by EAE was largely restored by L. reuteri treatment. Taxonomy-based analysis of gut microbiota showed that three "beneficial" genera Bifidobacterium, Prevotella, and Lactobacillus were negatively correlated with EAE clinical severity, whereas the genera Anaeroplasma, Rikenellaceae, and Clostridium were positively correlated with disease severity. Notably, L. reuteri treatment coordinately altered the relative abundance of these EAE-associated taxa. In conclusion, probiotic L. reuteri changed gut microbiota to modulate immune responses in EAE, making it a novel candidate in future studies to modify the severity of MS.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/microbiologia , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Probióticos/farmacologia , Animais , Encefalomielite Autoimune Experimental/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL
6.
J Pediatr ; 203: 55-61.e3, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30177353

RESUMO

OBJECTIVE: To dissect potential confounding effects of breast milk and formula feeding on crying + fussing, fecal calprotectin, and gut microbiota in babies with colic. We hypothesized that infant colic is associated with gut inflammation linked to intestinal dysbiosis. STUDY DESIGN: A nested case-control design of 3 of our studies was used to analyze clinical and laboratory data at presentation, comparing babies with colic with controls. All investigators other than the biostatistician were blinded during data analysis. Subjects were recruited based on their age and crying + fussy time. We screened 65 infants, 37 with colic, as defined by Barr diary (crying + fussing time >3 hours daily), who were compared with 28 noncolicky infants. RESULTS: Fecal calprotectin was elevated in babies with colic. For each mode of infant feeding (breast milk, formula, or breast + formula), infants' fecal calprotectin was higher in babies with colic. Infants with colic had similar levels of fecal alpha diversity (richness) when compared with controls, and alpha diversity was lower in breast-fed babies. Beta diversity at the phylum level revealed significant differences in microbial population. A phylum difference resulted from reduced Actinobacteria (95% of which are Bifidobacilli) in babies with colic. Species significantly associated with colic were Acinetobacter and Lactobacillus iners. CONCLUSIONS: Colic is linked with gut inflammation (as determined by fecal calprotectin) and dysbiosis, independent of mode of feeding, with fewer Bifidobacilli. TRIAL REGISTRATION: Clinicaltrials.gov: NCT01279265 and NCT01849991.


Assuntos
Cólica/complicações , Disbiose/diagnóstico , Fezes/química , Inflamação/diagnóstico , Complexo Antígeno L1 Leucocitário/análise , Acinetobacter/isolamento & purificação , Aleitamento Materno , Estudos de Casos e Controles , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal , Humanos , Lactente , Fórmulas Infantis , Recém-Nascido , Lactobacillus/isolamento & purificação , Masculino
7.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G231-G240, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29648878

RESUMO

Lactobacillus reuteri DSM 17938 (LR 17938) has been shown to reduce the incidence and severity of necrotizing enterocolitis (NEC). It is unclear if preventing NEC by LR 17938 is mediated by Toll-like receptor 2 (TLR2), which is known to mediate proinflammatory responses to bacterial cell wall components. NEC was induced in newborn TLR2-/- or wild-type (WT) mice by the combination of gavage-feeding cow milk-based formula and exposure to hypoxia and cold stress. Treatment groups were administered formula supplemented with LR 17938 or placebo (deMan-Rogosa-Sharpe media). We observed that LR 17938 significantly reduced the incidence of NEC and reduced the percentage of activated effector CD4+T cells, while increasing Foxp3+ regulatory T cells in the intestinal mucosa of WT mice with NEC, but not in TLR2-/- mice. Dendritic cell (DC) activation by LR 17938 was mediated by TLR2. The percentage of tolerogenic DC in the intestine of WT mice was increased by LR 17938 treatment during NEC, a finding not observed in TLR2-/- mice. Furthermore, gut levels of proinflammatory cytokines IL-1ß and IFN-γ were decreased after treatment with LR 17938 in WT mice but not in TLR2-/- mice. In conclusion, the combined in vivo and in vitro findings suggest that TLR2 receptors are involved in DC recognition and DC-priming of T cells to protect against NEC after oral administration of LR 17938. Our studies further clarify a major mechanism of probiotic LR 17938 action in preventing NEC by showing that neonatal immune modulation of LR 17938 is mediated by a mechanism requiring TLR2. NEW & NOTEWORTHY Lactobacillus reuteri DSM 17938 (LR 17938) has been shown to protect against necrotizing enterocolitis (NEC) in neonates and in neonatal animal models. The role of Toll-like receptor 2 (TLR2) as a sensor for gram-positive probiotics, activating downstream anti-inflammatory responses is unclear. Our current studies examined TLR2 -/- mice subjected to experimental NEC and demonstrated that the anti-inflammatory effects of LR 17938 are mediated via a mechanism requiring TLR2.


Assuntos
Enterocolite Necrosante , Mucosa Intestinal/imunologia , Intestinos/patologia , Limosilactobacillus reuteri , Receptor 2 Toll-Like/imunologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/farmacologia , Modelos Animais de Doenças , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/prevenção & controle , Interleucina-1beta/imunologia , Limosilactobacillus reuteri/imunologia , Limosilactobacillus reuteri/fisiologia , Camundongos , Probióticos/farmacologia , Linfócitos T Reguladores/imunologia
8.
Front Immunol ; 8: 1680, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29270168

RESUMO

The lack of a functional Foxp3 transcription factor and regulatory T (Treg) cells causes lethal, CD4+ T cell-driven autoimmune diseases in scurfy (SF) mice and humans. Recent studies have shown that adenosine A2A receptor activation limits inflammation and tissue damage, thereby playing an anti-inflammatory role. However, the role of the adenosine A2A receptor in the development of disease in SF mice remains unclear. Using a genetic approach, we found that adenosine A2A receptor deletion in SF mice (SF[Formula: see text]) does not affect early life events, the development of a lymphoproliferative disorder, or hyper-production of pro-inflammatory cytokines seen in the Treg-deficiency state. As shown previously, Lactobacillus reuteri DSM 17938 treatment prolonged survival and reduced multiorgan inflammation in SF mice. In marked contrast, A2A receptor deletion completely blocked these beneficial effects of L. reuteri in SF mice. Altogether, these results suggest that although absence of the adenosine A2A receptor does not affect the development of disease in SF mice, it plays a critical role in the immunomodulation by L. reuteri in Treg-deficiency disease. The adenosine A2A receptor and its activation may have a role in treating other Treg dysfunction-mediated autoimmune diseases.

9.
J Pediatr ; 191: 170-178.e2, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28969890

RESUMO

OBJECTIVE: To assess the safety of probiotic Lactobacillus reuteri strain Deutsche Sammlung von Mikroorganismen (DSM) 17938 with daily administration to healthy infants with colic and to determine the effect of L reuteri strain DSM 17938 on crying, fussing, inflammatory, immune, and microbiome variables. STUDY DESIGN: We performed a controlled, double-blinded, phase 1 safety and tolerability trial in healthy breast-fed infants with colic, aged 3 weeks to 3 months, randomly assigned to L reuteri strain DSM 17938 (5 × 108 colony-forming units daily) or placebo for 42 days and followed for 134 days. RESULTS: Of 117 screened infants, 20 were randomized to L reuteri strain DSM 17938 or placebo (sunflower oil) (in a 2:1 ratio) with 80% retention. Eleven of the 20 (55%) presented with low absolute neutrophil counts (<1500/mm3), which resolved in all subjects by day 176. L reuteri strain DSM 17938 produced no severe adverse events and did not significantly change crying time, plasma bicarbonate, or inflammatory biomarkers. Fecal calprotectin decreased rapidly in both groups. In the infants with dominant fecal gram negatives (Klebsiella, Proteus, and Veillonella), resolution of colic was associated with marked decreases in these organisms. CONCLUSIONS: Daily administration of L reuteri strain DSM 17938 appears to be safe in newborn infants with colic, including those with neutropenia, which frequently coexists. A placebo response of 66% suggests that many infants with colic will have resolution within 3 weeks. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01849991.


Assuntos
Cólica/terapia , Limosilactobacillus reuteri , Probióticos/uso terapêutico , Biomarcadores/metabolismo , Cólica/diagnóstico , Cólica/metabolismo , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Masculino , Resultado do Tratamento
10.
J Exp Med ; 214(1): 107-123, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27994068

RESUMO

Regulatory T (T reg) cell deficiency causes lethal, CD4+ T cell-driven autoimmune diseases. Stem cell transplantation is used to treat these diseases, but this procedure is limited by the availability of a suitable donor. The intestinal microbiota drives host immune homeostasis by regulating the differentiation and expansion of T reg, Th1, and Th2 cells. It is currently unclear if T reg cell deficiency-mediated autoimmune disorders can be treated by targeting the enteric microbiota. Here, we demonstrate that Foxp3+ T reg cell deficiency results in gut microbial dysbiosis and autoimmunity over the lifespan of scurfy (SF) mouse. Remodeling microbiota with Lactobacillus reuteri prolonged survival and reduced multiorgan inflammation in SF mice. L. reuteri changed the metabolomic profile disrupted by T reg cell deficiency, and a major effect was to restore levels of the purine metabolite inosine. Feeding inosine itself prolonged life and inhibited multiorgan inflammation by reducing Th1/Th2 cells and their associated cytokines. Mechanistically, the inhibition of inosine on the differentiation of Th1 and Th2 cells in vitro depended on adenosine A2A receptors, which were also required for the efficacy of inosine and of L. reuteri in vivo. These results reveal that the microbiota-inosine-A2A receptor axis might represent a potential avenue for combatting autoimmune diseases mediated by T reg cell dysfunction.


Assuntos
Autoimunidade , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Receptor A2A de Adenosina/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular , Feminino , Inosina/farmacologia , Masculino , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Células Th1/citologia , Células Th2/citologia
11.
Int J Clin Exp Pathol ; 9(2): 899-909, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29805726

RESUMO

L-selectin (CD62L) is normally highly expressed in naïve T cells. The expression levels of CD62L have been reported to be decreased on T cells during the inflammatory state. It is currently unknown whether the frequency of CD62L+ T cell subsets in the peripheral blood can be used as a marker to indicate is disease severity during inflammation. Our study evaluated whether circulating CD62L+ T cell subsets correlate with the severity of disease by testing an autoimmune condition of scurfy (sf) mouse associated with multi-organ inflammation due to regulatory T cell deficiency. We observed that scurfy mice spontaneously developed an inflammatory phenotype with a significant decrease in the percentage of CD62L-expressing CD4+ T and CD8+ T cells in the peripheral blood. The percentage of CD62L+CD4+ T and CD62L+CD8+ T cells negatively correlated with disease severity, as determined by the weight of spleen and liver, as well as the mean area of lymphocyte infiltrates in lung and liver. The percentage of CD8+ T cells also correlated directly with these markers of disease severity. To conclude, our results support the concept that circulating CD62L-expressing T cells may be used as markers of disease severity in sf mice which is equivalent to a syndrome characterized by immune dysregulation with polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX syndrome) in humans, or in other autoimmune or inflammatory conditions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA