Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 14941, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696924

RESUMO

Up to 25% of the US population harbor Clostridioides difficile in the gut. Following antibiotic disruption of the gut microbiota, C. difficile can act as an opportunistic pathogen and induce potentially lethal infections. Consequently, reducing the colonization of C. difficile in at-risk populations is warranted, prompting us to identify and characterize a probiotic candidate specifically targeting C. difficile colonization. We identified Bacillus velezensis DSM 33864 as a promising strain to reduce C. difficile levels in vitro. We further investigated the effects of B. velezensis DSM 33864 in an assay including human fecal medium and in healthy or clindamycin-treated mouse models of C. difficile colonization. The addition of B. velezensis DSM 33864 to human fecal samples was shown to reduce the colonization of C. difficile in vitro. This was supported in vivo where orally administered B. velezensis DSM 33864 spores reduced C. difficile levels in clindamycin-treated mice. The commensal microbiota composition or post-antibiotic reconstitution was not impacted by B. velezensis DSM 33864 in human fecal samples, short-, or long-term administration in mice. In conclusion, oral administration of B. velezensis DSM 33864 specifically reduced C. difficile colonization in vitro and in vivo without adversely impacting the commensal gut microbiota composition.


Assuntos
Clostridioides difficile , Microbioma Gastrointestinal , Humanos , Animais , Camundongos , Clindamicina/farmacologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Clostridioides
2.
Gut Microbes ; 15(1): 2192547, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36945120

RESUMO

Growing evidence supports the use of probiotics to prevent or mitigate obesity-related dysmetabolism and non-alcoholic fatty liver disease (NAFLD). However, frequent reports of responders versus non-responders to probiotic treatment warrant a better understanding of key modifiers of host-microbe interactions. The influence of host diet on probiotic efficacy, in particular against metabolic diseases, remains elusive. We fed C57BL6/J mice a low fat reference diet or one of two energy-matched high fat and high sucrose diets for 12 weeks; a classical high fat diet (HFD) and a customized fast food-mimicking diet (FFMD). During the studies, mice fed either obesogenic diet were gavaged daily with one of two probiotic lactic acid bacteria (LAB) strains previously classified as Lactobaccillus, namely Limosilactobacillus reuteri (L. reuteri)or Lacticaseibacillus paracaseisubsp. paracasei (L. paracasei), or vehicle. The tested probiotics exhibited a reproducible efficacy but dichotomous response according to the obesogenic diets used. Indeed, L. paracaseiprevented weight gain, improved insulin sensitivity, and protected against NAFLD development in mice fed HFD, but not FFMD. Conversely, L. reuteri improved glucoregulatory capacity, reduced NAFLD development, and increased distal gut bile acid levels associated with changes in predicted functions of the gut microbiota exclusively in the context of FFMD-feeding. We found that the probiotic efficacy of two LAB strains is highly dependent on experimental obesogenic diets. These findings highlight the need to carefully consider the confounding impact of diet in order to improve both the reproducibility of preclinical probiotic studies and their clinical research translatability.


Assuntos
Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Probióticos , Animais , Camundongos , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Reprodutibilidade dos Testes , Obesidade/microbiologia , Probióticos/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Camundongos Endogâmicos C57BL
3.
Neurogastroenterol Motil ; 35(1): e14477, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36178333

RESUMO

BACKGROUND: Stress is an exacerbator of irritable bowel syndrome (IBS) symptoms, and anxiety and depression are co-morbidities. Bifidobacterium longum strains 1714® and 35642® attenuate stress responses in healthy people and reduce symptoms in IBS, respectively. Here, we explore relationships between the psychological and visceral effects of the two strains (COMBO) in IBS subjects and biomarkers of stress and inflammation. METHODS: We recruited 40 patients with IBS (Rome III) and mild to moderate anxiety (HADS-A) and/or depression (HADS-D) and 57 asymptomatic female controls with low or moderate stress. IBS patients were fed COMBO (1 × 109 cfu/day) for 8 weeks with an 8-week washout. IBS symptoms, psychometric measures, salivary cortisol awakening response (CAR), and plasma inflammatory biomarkers were assessed every 4 weeks. KEY RESULTS: Compared to healthy controls, IBS subjects had a blunted CAR. Treatment with COMBO restored CAR and improved IBS symptoms compared to baseline during the treatment phase. The COMBO reduced HADS-D, HADS-A score, and TNF-α, while sleep quality improved significantly from baseline to the end of the intervention. Surprisingly, these parameters improved further once treatment ended and maintained this improvement by Week 16. CONCLUSIONS AND INFERENCES: These findings suggest that the stress response is a major driver of IBS symptoms. The time course of the beneficial effect of COMBO on IBS symptoms suggests that this is achieved through a restoration of the stress response. In contrast, the time course of the effects of COMBO on anxiety and depression in IBS paralleled an anti-inflammatory effect as indicated by a reduction in circulating levels of TNF-α.


Assuntos
Síndrome do Intestino Irritável , Probióticos , Humanos , Feminino , Síndrome do Intestino Irritável/psicologia , Fator de Necrose Tumoral alfa , Ansiedade/psicologia , Comorbidade , Probióticos/uso terapêutico
4.
Gut Microbes ; 13(1): 1988836, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34693864

RESUMO

Colitis is characterized by colonic inflammation and impaired gut health. Both features aggravate obesity and insulin resistance. Host defense peptides (HDPs) are key regulators of gut homeostasis and generally malfunctioning in above-mentioned conditions. We aimed here to improve bowel function in diet-induced obesity and chemically induced colitis through daily oral administration of lysozyme, a well-characterized HDP, derived from Acremonium alcalophilum.C57BL6/J mice were fed either low-fat reference diet or HFD ± daily gavage of lysozyme for 12 weeks, followed by metabolic assessment and evaluation of colonic microbiota encroachment. To further evaluate the efficacy of intestinal inflammation, we next supplemented chow-fed BALB/c mice with lysozyme during Dextran Sulfate Sodium (DSS)-induced colitis in either conventional or microbiota-depleted mice. We assessed longitudinal microbiome alterations by 16S amplicon sequencing in both models.Lysozyme dose-dependently alleviated intestinal inflammation in DSS-challenged mice and further protected against HFD-induced microbiota encroachment and fasting hyperinsulinemia. Observed improvements of intestinal health relied on a complex gut flora, with the observation that microbiota depletion abrogated lysozyme's capacity to mitigate DSS-induced colitis.Akkermansia muciniphila associated with impaired gut health in both models, a trajectory that was mitigated by lysozyme administration. In agreement with this notion, PICRUSt2 analysis revealed specific pathways consistently affected by lysozyme administration, independent of vivarium, disease model and mouse strain.Taking together, lysozyme leveraged the gut microbiota to curb DSS-induced inflammation, alleviated HFD-induced gastrointestinal disturbances and lowered fasting insulin levels in obese mice. Collectively, these data present A. alcalophilum-derived lysozyme as a promising candidate to enhance gut health.


Assuntos
Acremonium/enzimologia , Colite/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Muramidase/administração & dosagem , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Colite/induzido quimicamente , Colite/microbiologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muramidase/metabolismo
5.
Am J Physiol Endocrinol Metab ; 317(1): E42-E52, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30860877

RESUMO

Overnutrition is the principal cause of insulin resistance (IR) and dyslipidemia, which drive nonalcoholic fatty liver disease (NAFLD). Overnutrition is further linked to disrupted bowel function, microbiota alterations, and change of function in gut-lining cell populations, including Paneth cells of the small intestine. Paneth cells regulate microbial diversity through expression of antimicrobial peptides, particularly human α-defensin-5 (HD-5), and have shown repressed secretory capacity in human obesity. Mice were fed a 60% high-fat diet for 13 wk and subsequently treated with physiologically relevant amounts of HD-5 (0.001%) or vehicle for 10 wk. The glucoregulatory capacity was determined by glucose tolerance tests and measurements of corresponding insulin concentrations both before and during intervention. Gut microbiome composition was examined by 16S rRNA gene amplicon sequencing. HD-5-treated mice exhibited improved glucoregulatory capacity along with an ameliorated plasma and liver lipid profile. This was accompanied by specific decrease in jejunal inflammation and gut microbiota alterations including increased Bifidobacterium abundances, which correlated inversely with metabolic dysfunctions. This study provides proof of concept for the use of human defensins to improve host metabolism by mitigating the triad cluster of dyslipidemia, IR, and NAFLD.


Assuntos
Metabolismo dos Carboidratos/efeitos dos fármacos , Dislipidemias/tratamento farmacológico , Glucose/metabolismo , Obesidade/tratamento farmacológico , alfa-Defensinas/uso terapêutico , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Dieta Hiperlipídica , Dislipidemias/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/etiologia , Obesidade/metabolismo , Celulas de Paneth/metabolismo , alfa-Defensinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA