Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
BMC Complement Med Ther ; 23(1): 367, 2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37853370

RESUMO

INTRODUCTION: Infants who are born from mothers with HIV (infants who are HIV exposed but uninfected; iHEU) are at higher risk of morbidity and display multiple immune alterations compared to infants who are HIV-unexposed (iHU). Easily implementable strategies to improve immunity of iHEU, and possibly subsequent clinical health outcomes, are needed. iHEU have altered gut microbiome composition and bifidobacterial depletion, and relative abundance of Bifidobacterium infantis has been associated with immune ontogeny, including humoral and cellular vaccine responses. Therefore, we will assess microbiological and immunological phenotypes and clinical outcomes in a randomized, double-blinded trial of B. infantis Rosell®-33 versus placebo given during the first month of life in South African iHEU. METHODS: This is a parallel, randomised, controlled trial. Two-hundred breastfed iHEU will be enrolled from the Khayelitsha Site B Midwife Obstetric Unit in Cape Town, South Africa and 1:1 randomised to receive 8 × 109 CFU B. infantis Rosell®-33 daily or placebo for the first 4 weeks of life, starting on day 1-3 of life. Infants will be followed over 36 weeks with extensive collection of meta-data and samples. Primary outcomes include gut microbiome composition and diversity, intestinal inflammation and microbial translocation and cellular vaccine responses. Additional outcomes include biological (e.g. gut metabolome and T cell phenotypes) and clinical (e.g. growth and morbidity) outcome measures. DISCUSSION: The results of this trial will provide evidence whether B. infantis supplementation during early life could improve health outcomes for iHEU. ETHICS AND DISSEMINATION: Approval for this study has been obtained from the ethics committees at the University of Cape Town (HREC Ref 697/2022) and Seattle Children's Research Institute (STUDY00003679). TRIAL REGISTRATION: Pan African Clinical Trials Registry Identifier: PACTR202301748714019. TRIALS: gov: NCT05923333. PROTOCOL VERSION: Version 1.8, dated 18 July 2023.


Assuntos
Infecções por HIV , Vacinas , Feminino , Humanos , Lactente , Gravidez , Bifidobacterium longum subspecies infantis , Suplementos Nutricionais , Infecções por HIV/tratamento farmacológico , Ensaios Clínicos Controlados Aleatórios como Assunto , África do Sul
2.
Pediatr Res ; 94(6): 1887-1905, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37460707

RESUMO

BACKGROUND: Bifidobacterium infantis has special abilities to utilise human milk oligosaccharides. Hence we hypothesised that probiotic supplements containing B. infantis may confer greater benefits to preterm infants than probiotic supplements without B. infantis. METHODS: A systematic review with meta-analysis was conducted according to standard guidelines. We selected RCTs evaluating probiotics compared to placebo or no treatment in preterm and/or low birth weight infants. Probiotic effects on Necrotizing Enterocolitis (NEC), Late Onset Sepsis (LOS) and Mortality were analysed separately for RCTs in which the supplemented probiotic product contained B. infantis and those that did not contain B. infantis. RESULTS: 67 RCTs were included (n = 14,606), of which 16 used probiotics containing B. infantis (Subgroup A) and 51 RCTs did not (Subgroup B) Meta-analysis of all RCTs indicated that probiotics reduced the risk of NEC, LOS, and mortality. The subgroup meta-analysis demonstrated greater reduction in the incidence of NEC in subgroup A than subgroup B [(relative risk in subgroup A: 0.38; 95% CI, 0.27-0.55) versus (0.67; 95% CI, 0.55-0.81) in subgroup B; p value for subgroup difference: 0.01]. CONCLUSIONS: These results provide indirect evidence that probiotic supplements that include B. infantis may be more beneficial for preterm infants. Well-designed RCTs are necessary to confirm these findings. IMPACT: Evidence is emerging that beneficial effects of probiotics are species and strain specific. This systematic review analyses if B. infantis supplementation provides an advantage to preterm infants. This is the first systematic review evaluating the effects of probiotics containing B. infantis in preterm infants. The results of this systematic review provides indirect evidence that probiotics that include B. infantis may be more beneficial for preterm infants. These results will help in guiding future research and clinical practice for using B. infantis as a probiotic in preterm infants.


Assuntos
Enterocolite Necrosante , Probióticos , Sepse , Lactente , Recém-Nascido , Humanos , Recém-Nascido Prematuro , Bifidobacterium longum subspecies infantis , Probióticos/uso terapêutico , Suplementos Nutricionais , Recém-Nascido de Baixo Peso , Enterocolite Necrosante/prevenção & controle , Sepse/prevenção & controle , Sepse/microbiologia
3.
J Pediatr Gastroenterol Nutr ; 75(4): 535-542, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35881967

RESUMO

OBJECTIVES: To compare the impact of two probiotic supplements on fecal microbiota and metabolites, as well as on gut inflammation in human milk-fed preterm infants. METHODS: In this single-center observational cohort study, we assessed the effects of Bifidobacterium longum subsp. infantis or Lactobacillus reuteri supplementation on the infant gut microbiota by 16S rRNA gene sequencing and fecal metabolome by 1 H nuclear magnetic resonance spectroscopy. Fecal calprotectin was measured as a marker of enteric inflammation. Aliquots of human or donor milk provided to each infant were also assessed to determine human milk oligosaccharide (HMO) content. RESULTS: As expected, each probiotic treatment was associated with increased proportions of the respective bacterial taxon. Fecal HMOs were significantly higher in L. reuteri fed babies despite similar HMO content in the milk consumed. Fecal metabolites associated with bifidobacteria fermentation products were significantly increased in B. infantis supplemented infants. Fecal calprotectin was lower in infants receiving B. infantis relative to L. reuteri ( P < 0.01, Wilcoxon rank-sum test) and was negatively associated with the microbial metabolite indole-3-lactate (ILA). CONCLUSIONS: This study demonstrates that supplementing an HMO-catabolizing Bifidobacterium probiotic results in increased microbial metabolism of milk oligosaccharides and reduced intestinal inflammation relative to a noncatabolizing Lactobacillus probiotic in human milk-fed preterm infants. In this context, Bifidobacterium may provide greater benefit in human milk-fed infants via activation of the microbiota-metabolite-immune axis.


Assuntos
Microbiota , Probióticos , Bifidobacterium , Bifidobacterium longum subspecies infantis/metabolismo , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Inflamação , Complexo Antígeno L1 Leucocitário/metabolismo , Oligossacarídeos/metabolismo , RNA Ribossômico 16S
4.
BMJ Open ; 11(11): e052449, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34753762

RESUMO

INTRODUCTION: The Global Platform for the Prevention of Autoimmune Diabetes-SINT1A Study is designed as a randomised, placebo-controlled, double-blind, multicentre, multinational, primary prevention study aiming to assess whether daily administration of Bifidobacterium infantis from age 7 days to 6 weeks until age 12 months to children with elevated genetic risk for type 1 diabetes reduces the cumulative incidence of beta-cell autoantibodies in childhood. METHODS AND ANALYSIS: Infants aged 7 days to 6 weeks from Germany, Poland, Belgium, UK and Sweden are eligible for study participation if they have a >10.0% expected risk for developing multiple beta-cell autoantibodies by age 6 years as determined by genetic risk score or family history and HLA genotype. Infants are randomised 1:1 to daily administration of B. infantis EVC001 or placebo until age 12 months and followed for a maximum of 5.5 years thereafter. The primary outcome is the development of persistent confirmed multiple beta-cell autoantibodies. Secondary outcomes are (1) Any persistent confirmed beta-cell autoantibody, defined as at least one confirmed autoantibody in two consecutive samples, including insulin autoantibodies, glutamic acid decarboxylase, islet tyrosine phosphatase 2 or zinc transporter 8, (2) Diabetes, (3) Transglutaminase autoantibodies associated with coeliac disease, (4) Respiratory infection rate in first year of life during supplementation and (5) Safety. Exploratory outcomes include allergy, antibody response to vaccines, alterations of the gut microbiome or blood metabolome, stool pH and calprotectin. ETHICS AND DISSEMINATION: The study was approved by the local ethical committees of the Technical University Munich, Medical Faculty, the Technische Universität Dresden, the Medizinische Hochschule Hannover, the Medical University of Warsaw, EC Research UZ Leuven and the Swedish ethical review authority. The results will be disseminated through peer-reviewed journals and conference presentations and will be openly shared after completion of the study. TRIAL REGISTRATION NUMBER: NCT04769037.


Assuntos
Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Autoimunidade , Bifidobacterium longum subspecies infantis , Criança , Diabetes Mellitus Tipo 1/prevenção & controle , Suplementos Nutricionais , Humanos , Lactente , Estudos Multicêntricos como Assunto , Ensaios Clínicos Controlados Aleatórios como Assunto
5.
Food Funct ; 11(5): 3964-3974, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32400787

RESUMO

Ulcerative colitis (UC) is a chronic inflammatory bowel disease whose prevalence is increasing globally. A synbiotic has probiotic and prebiotic components and is regarded as a promising candidate for alleviating UC-associated inflammation. The purpose of this study is to determine whether there is an additive efficacy between the probiotic Bifidobacterium infantis (B. infantis) and the prebiotic xylooligosaccharide (XOS) against UC. C57BL/6 mice were treated with B. infantis, XOS, or synbiotic (combination of B. infantis and XOS) for 21 d. During the final 7 d of treatment, the mice were administered dextran sulfate sodium (DSS) dissolved in drinking water to induce colitis. All treatments decreased the disease activity index (DAI) and pathological scores, and synbiotic treatment was more efficacious than either the probiotic or prebiotic alone. Compared with the DSS-induced colitis group, all treatment groups significantly downregulated the proinflammatory cytokines TNF-α and IL-1ß, and synbiotic treatment significantly upregulated the anti-inflammatory cytokine IL-10 in the colon tissues. Furthermore, all treatments significantly reduced the NLR family pyrin domain containing 3 (NLRP3) inflammasome mRNA level in the colon tissues. All treatments significantly inhibited oxidative stress and increased zonula occludens-1 (ZO-1), occludin, and claudin-1 tight junction (TJ) molecule mRNA levels in the colon tissues. Therefore, the observed efficacy of synbiotics against colitis may be explained by the additive combination of the direct anti-inflammatory effects of the probiotic and prebiotic components and their ability to fortify colonic epithelial barrier integrity. Our findings suggest that a synbiotic is a promising dietary supplement or functional food for the effective management of UC.


Assuntos
Bifidobacterium longum subspecies infantis , Colite Ulcerativa/induzido quimicamente , Sulfato de Dextrana/toxicidade , Glucuronatos/administração & dosagem , Oligossacarídeos/administração & dosagem , Simbióticos , Animais , Claudina-1/genética , Claudina-1/metabolismo , Citocinas/genética , Citocinas/metabolismo , Suplementos Nutricionais , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ocludina/genética , Ocludina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
6.
Med Sci Monit ; 26: e920583, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32249275

RESUMO

BACKGROUND Bifidobacteria are among the probiotics used in treating intestinal diseases and are rarely used for allergic asthma treatment. The present study investigated the mechanism of B. infantis in treating allergic asthma in mice. MATERIAL AND METHODS A total of 40 male Balb/c mice were randomized into control, ovalbumin (OVA), montelukast (Mon), and B. infantis (B10) groups, and allergic asthma was induced in the OVA, Mon, and B10 groups. Airway reactivity was measured on day 29 by methacholine at various doses. The numbers of total cells and inflammatory cells in bronchoalveolar lavage fluid (BALF) were counted by blood cell counter and Diff-Quik staining. Hematoxylin-eosin (HE) staining was performed to observe inflammatory cell infiltration in lung tissues. Total IgE and OVA-specific IgE in serum were measured by ELISA. Mucin 5AC expression was detected by Western blot to evaluate airway obstruction. The levels of Th1 (IFN-γ, IL-2) and Th2 (IL-4, IL-5, IL-13) cytokines in BALF and tissues were detected by ELISA and qRT-PCR, respectively. RESULTS The mice in the OVA group had airway hyperreactivity, while the symptoms in the B10 group and Mon group were effectively relieved. B10 reduced the number of inflammatory cells in BALF as well as inflammatory cell infiltration in tissues. Moreover, the levels of total serum IgE, OVA-specific IgE, and Mucin 5AC were increased in the OVA group, but were reduced in the Mon group and B10 group. B. infantis increased the levels of Th1 cytokines and decreased those of Th2 cytokines. CONCLUSIONS B. infantis can reduce the infiltration of inflammatory cells induced by OVA-specific antibodies in mice. B. infantis has therapeutic effects on allergic asthma by promoting Th1 and inhibiting Th2 immune responses.


Assuntos
Asma/terapia , Bifidobacterium longum subspecies infantis , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/imunologia , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Imunoglobulina E/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Distribuição Aleatória , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th2/efeitos dos fármacos , Células Th2/imunologia
7.
Nutrients ; 12(3)2020 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-32235769

RESUMO

Objective: To evaluate the nutrition-related effects of prophylactic Lactobacillus acidophilus/Bifidobacterium infantis probiotics on the outcomes of preterm infants <29 weeks of gestation that receive human milk and/or formula nutrition. We hypothesize that human-milk-fed infants benefit from probiotics in terms of sepsis prevention and growth. METHODS: We performed an observational study of the German Neonatal Network (GNN) over a period of six years, between 1 January, 2013 and 31 December, 2018. Prophylactic probiotic use of L. acidophilus/B. infantis was evaluated in preterm infants <29 weeks of gestation (n = 7516) in subgroups stratified to feeding type: (I) Exclusively human milk (HM) of own mother and/or donors (HM group, n = 1568), (II) HM of own mother and/or donor and formula (Mix group, n = 5221), and (III) exclusive exposure to formula (F group, n = 727). The effect of probiotics on general outcomes and growth was tested in univariate models and adjusted in linear/logistic regression models. RESULTS: 5954 (76.5%) infants received L. acidophilus/B. infantis prophylactically for the prevention of necrotizing enterocolitis (NEC). Probiotic use was associated with improved growth measures in the HM group (e.g., weight gain velocity in g/day: effect size B = 0.224; 95% CI: 2.82-4.35; p < 0.001) but not in the F group (effect size B = -0.06; 95% CI: -3.05-0.28; p = 0.103). The HM group had the lowest incidence of clinical sepsis (34.0%) as compared to the Mix group (35.5%) and the F group (40.0%). Only in the Mix group, probiotic supplementation proved to be protective against clinical sepsis (OR 0.69; 95% CI: 0.59-0.79; p < 0.001). CONCLUSION: Our observational data indicate that the exposure to L. acidophilus/B. infantis probiotics may promote growth in exclusively HM-fed infants as compared to formula-fed infants. To exert a sepsis-preventive effect, probiotics seem to require human milk.


Assuntos
Bifidobacterium longum subspecies infantis , Suplementos Nutricionais , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido Prematuro/crescimento & desenvolvimento , Recém-Nascido Prematuro/fisiologia , Lactobacillus acidophilus , Leite Humano , Probióticos/administração & dosagem , Enterocolite Necrosante/prevenção & controle , Feminino , Idade Gestacional , Humanos , Fórmulas Infantis , Recém-Nascido , Masculino , Profilaxia Pré-Exposição , Sepse/prevenção & controle
8.
Benef Microbes ; 10(7): 729-739, 2019 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-31965842

RESUMO

Formula-fed infants are more susceptible to infectious diseases because they lack the maternal immune factors transferred from breast milk, while their own immune system is still immature. As timely probiotic administration was suggested to promote immune system development in formula-fed infants, this study aimed at assessing the safety and the effects of a probiotic supplement (Bifidobacterium infantis R0033, Bifidobacterium bifidum R0071, and Lactobacillus helveticus R0052) on mucosal immune competence and digestive function in formula-fed infants. Healthy infants (3.5-6 months old) were randomised to receive either probiotic- (n=66) or placebo-supplemented (n=66) formula once a day for four weeks. In the probiotics group, faecal secretory immunoglobulin A (SIgA) levels remained similar between visit 2 (baseline; V2) and visit 3 (end-of-treatment; V3), but decreased in the placebo group. Changes in SIgA levels following treatment (log10ΔV3-V2 [95%CI]) between the probiotic and placebo groups were statistically significant (23 ng/dl [-57;102] and -137 ng/dl [-212;-62], respectively (P=0.0044; ANCOVA)). While log10ΔV3-V2 [95%CI] for salivary SIgA levels increased in both groups, this trend was more pronounced in the probiotics than in the placebo group with an increase of 123 ng/dl [9;236] and 37 ng/dL [-72;147], respectively (P=0.2829; ANCOVA). The weekly average number of stools/day was significantly higher in the probiotics group compared to placebo during the last week of treatment for the per protocol population. There was no difference in microbiota composition or anthropometric parameters between groups. No serious adverse event was reported, and all adverse events were mild and unrelated to the product or study. Our results show that formula-fed infants receiving probiotics maintained higher faecal SIgA levels at the end of the four-week treatment period, suggesting a positive effect of probiotics on SIgA production. This study demonstrates the safety of this probiotic formulation in infants. Formula-fed infants may benefit from probiotics supplementation to sustain the development of mucosal immunity.


Assuntos
Suplementos Nutricionais , Imunoglobulina A Secretora/análise , Fórmulas Infantis , Intestinos/imunologia , Intestinos/microbiologia , Probióticos , Bifidobacterium bifidum , Bifidobacterium longum subspecies infantis , Método Duplo-Cego , Fezes/microbiologia , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina A Secretora/imunologia , Lactente , Lactobacillus helveticus , Masculino , Saliva/imunologia
9.
Int Immunopharmacol ; 61: 290-296, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29908492

RESUMO

BACKGROUND: Guillain-Barré syndrome (GBS) is a rare, autoimmune-mediated disease. The use of Bifidobacterium is reportedly effective in alleviating GBS since they act by regulating T helper (Th) cells. OBJECTIVES: In this study, we explored the differentiation of T helper cell subsets in patients with GBS. We also evaluated the effect of GBS on Bifidobacterium levels in patients and the likely protective influence of this bacterium in alleviating the disease in an animal model. MATERIALS AND METHODS: We used flow cytometry, and real-time polymerase chain reaction (PCR) to determine the T cell subsets differentiation among 30 GBS patients and 20 healthy controls (HC). The concentration of Bifidobacterium was assayed by real-time PCR. Experimental autoimmune neuritis (EAN) animal model was established to support the protective role of Bifidobacterium in GBS. RESULTS: The expression of Th cells, Th2 and Th17 in the patients was significantly higher than that in the HC, while Treg cells decreased substantially. Moreover, the levels of Bifidobacterium in the GBS patients were considerably lower than those in the HC, the concentration of Bifidobacterium correlating with Th2 and Th17 subsets negatively. Treatment with Bifidobacterium significantly reduced the levels of Th2 and Th17 and promoted the levels of Treg cells. CONCLUSIONS: We concluded from this study that Bifidobacterium alleviated GBS by regulating Th cells, although in-depth studies might be required to fully understand the mechanism of action.


Assuntos
Bifidobacterium longum subspecies infantis/imunologia , Terapia Biológica , Síndrome de Guillain-Barré/terapia , Neurite Autoimune Experimental/terapia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Animais , Autoimunidade , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Endogâmicos , Subpopulações de Linfócitos T/microbiologia , Linfócitos T Reguladores/microbiologia
10.
J Gastroenterol Hepatol ; 32 Suppl 1: 69-72, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28244666

RESUMO

Irritable bowel syndrome (IBS) is heterogeneous. Patients need proper assessment and explanation of IBS pathophysiology and appropriate therapies. A low FODMAP (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) diet effectively reduces symptoms in 75% of patients. Best treatment for those nonresponsive will depend on the pathophysiological basis for symptom genesis, with the following possible abnormalities: (i) Visceral hypersensitivity and/or enhanced gut-brain communication: a low FODMAP diet is mainly targeted for this patient group. A dietitian may also recommend antispasmodic agents, including peppermint oil. Another dietary treatment is a low food chemical diet, although this diet is often extremely limited, and therefore, not suited for some populations. Psychological therapies are also clinically beneficial. (ii) Altered motility: in patients with fast transit, a dietitian may recommend a reduction in all FODMAPs or targeted monosaccharides and disaccharides, which are more osmotic in nature. If not effective, patients may benefit from psyllium, which has an exceptional water-holding capacity aimed to promote more formed stools. Patients with slow or uncoordinated transit are often more difficult to treat. Dietary interventions have some success and usually comprise a combination of adequate fiber and fluid, osmotic laxatives, and stimulating agents such as caffeine, senna, and exercise. (iii) Altered microbiome: supplementary probiotics and prebiotics have weak evidence of efficacy with some notable exceptions. A dietitian may trial supplementary Bifidobacterium infantis or oligosaccharides, usually as an adjunct therapy. Guidance from a dietitian will encompass dietary methods to treat IBS but additionally identify where dietary treatment is not indicated to ensure that diet is correctly used and patients are not nutritionally or psychologically compromised.


Assuntos
Dieta com Restrição de Carboidratos , Síndrome do Intestino Irritável/dietoterapia , Bifidobacterium longum subspecies infantis , Dissacarídeos/administração & dosagem , Dissacarídeos/efeitos adversos , Fermentação , Motilidade Gastrointestinal , Humanos , Síndrome do Intestino Irritável/etiologia , Síndrome do Intestino Irritável/fisiopatologia , Laxantes/administração & dosagem , Mentha piperita , Monossacarídeos/administração & dosagem , Monossacarídeos/efeitos adversos , Oligossacarídeos/administração & dosagem , Oligossacarídeos/efeitos adversos , Parassimpatolíticos/administração & dosagem , Óleos de Plantas/administração & dosagem , Polímeros/administração & dosagem , Polímeros/efeitos adversos , Processos Psicoterapêuticos , Psyllium/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA