Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.449
Filtrar
1.
Cell Biol Toxicol ; 40(1): 64, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39096436

RESUMO

BACKGROUND AND PURPOSE: Colorectal cancer (CRC) is a widespread malignancy with a complex and not entirely elucidated pathogenesis. This study aims to explore the role of Bifidobacterium in the urea cycle (UC) and its influence on the progression of CRC, a topic not extensively studied previously. EXPERIMENTAL APPROACH: Utilizing both bioinformatics and experimental methodologies, this research involved analyzing bacterial abundance in CRC patients in comparison to healthy individuals. The study particularly focused on the abundance of BA. Additionally, transcriptomic data analysis and cellular experiments were conducted to investigate the impact of Bifidobacterium on ammonia metabolism and mitochondrial function, specifically examining its regulation of the key UC gene, ALB. KEY RESULTS: The analysis revealed a significant decrease in Bifidobacterium abundance in CRC patients. Furthermore, Bifidobacterium was found to suppress ammonia metabolism and induce mitochondrial dysfunction through the regulation of the ALB gene, which is essential in the context of UC. These impacts contributed to the suppression of CRC cell proliferation, a finding corroborated by animal experimental results. CONCLUSIONS AND IMPLICATIONS: This study elucidates the molecular mechanism by which Bifidobacterium impacts CRC progression, highlighting its role in regulating key metabolic pathways. These findings provide potential targets for novel therapeutic strategies in CRC treatment, emphasizing the importance of microbiota in cancer progression.


Assuntos
Bifidobacterium , Neoplasias Colorretais , Ureia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Bifidobacterium/metabolismo , Humanos , Ureia/metabolismo , Animais , Proliferação de Células , Amônia/metabolismo , Camundongos , Mitocôndrias/metabolismo , Linhagem Celular Tumoral , Masculino , Microbioma Gastrointestinal/fisiologia , Feminino
2.
Commun Biol ; 7(1): 943, 2024 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-39098939

RESUMO

To enhance health benefits, a probiotic can be co-administered with a metabolizable prebiotic forming a synergistic synbiotic. We assessed the synergies resulting from combining Bifidobacterium longum subsp. infantis LMG 11588 and an age-adapted blend of six human milk oligosaccharides (HMOs) in ex vivo colonic incubation bioreactors seeded with fecal background microbiota from infant and toddler donors. When HMOs were combined with B. infantis LMG 11588, they were rapidly and completely consumed. This resulted in increased short chain fatty acid (SCFA) production compared to the summed SCFA production from individual ingredients (synergy). Remarkably, HMOs were partially consumed for specific infant donors in the absence of B. infantis LMG 11588, yet all donors showed increased SCFA production upon B. infantis LMG 11588 supplementation. We found specific bacterial taxa associated with the differential response pattern to HMOs. Our study shows the importance of carefully selecting pre- and probiotic into a synergistic synbiotic that could benefit infants.


Assuntos
Bifidobacterium longum subspecies infantis , Ácidos Graxos Voláteis , Leite Humano , Oligossacarídeos , Humanos , Leite Humano/metabolismo , Leite Humano/química , Oligossacarídeos/metabolismo , Ácidos Graxos Voláteis/metabolismo , Lactente , Bifidobacterium longum subspecies infantis/metabolismo , Bifidobacterium/metabolismo , Probióticos/administração & dosagem , Microbioma Gastrointestinal , Fezes/microbiologia , Feminino , Simbióticos/administração & dosagem , Pré-Escolar
3.
Med Sci (Basel) ; 12(3)2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39189204

RESUMO

The progression of chronic obstructive pulmonary disease (COPD) is characterized by functional changes in the airways. The lung-gut axis and gut microbiota (GM) have been linked to the pathophysiology of airway diseases. Regarding COPD, studies have shown that GM alterations could be related the stages of this disease. However, the relationship between GM and clinical, biochemical and immunological parameters in patients with COPD are not well understood. The aim of this study was to compare the relative abundance of specific groups of beneficial gut bacteria between COPD patients and healthy controls (CTLs) in order to evaluate relationships with metabolic and inflammatory markers in COPD. METHODS: We included 16 stable COPD patients and 16 healthy volunteer CTLs. The relative abundances of Bifidobacterium spp. (Bf) and Akkermansia muciniphila (Akk) bacteria and the Bacteroidetes and Firmicutes phyla were assessed by qPCR. Pulmonary function was evaluated by spirometry, biochemical parameters by colorimetric methods and plasma cytokine levels by cytometric bead array analysis. RESULTS: The Firmicutes/Bacteroides ratio was related to emergency hospital visits and six-minute walk test (6MWT) results. Furthermore, the relative abundance of Bf was associated with plasma concentrations of glucose, triglycerides, HDL-C and IL-10. In addition, Firmicutes levels and the Firmicutes/Bacteroidetes ratio were associated with the IL-12/IL-10 ratio, while Akk abundance was linked to IL-12 levels. CONCLUSIONS: The present findings suggest that the abundance of beneficial bacteria in the GM could influence clinical presentation and immunoregulation in COPD.


Assuntos
Microbioma Gastrointestinal , Doença Pulmonar Obstrutiva Crônica , Humanos , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Akkermansia , Bifidobacterium , Estudos de Casos e Controles , Citocinas/sangue
4.
Compr Rev Food Sci Food Saf ; 23(5): e13431, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39165140

RESUMO

Human milk oligosaccharides (HMOs) are an evolutionarily significant advantage bestowed by mothers for facilitating the development of the infant's gut microbiota. They can avoid absorption in the stomach and small intestine, reaching the colon successfully, where they engage in close interactions with gut microbes. This process also enables HMOs to exert additional prebiotic effects, including regulating the mucus layer, promoting physical growth and brain development, as well as preventing and mitigating conditions such as NEC, allergies, and diarrhea. Here, we comprehensively review the primary ways by which gut microbiota, including Bifidobacteria and other genera, utilize HMOs, and we classify them into five central pathways. Furthermore, we emphasize the metabolic benefits of bacteria consuming HMOs, particularly the recently identified intrinsic link between HMOs and the metabolic conversion of tryptophan to indole and its derivatives. We also examine the extensive probiotic roles of HMOs and their recent research advancements, specifically concentrating on the unsummarized role of HMOs in regulating the mucus layer, where their interaction with the gut microbiota becomes crucial. Additionally, we delve into the principal tools used for functional mining of new HMOs. In conclusion, our study presents a thorough analysis of the interaction mechanism between HMOs and gut microbiota, emphasizing the cooperative utilization of HMOs by gut microbiota, and provides an overview of the subsequent probiotic effects of this interaction. This review provides new insights into the interaction of HMOs with the gut microbiota, which will inform the mechanisms by which HMOs function.


Assuntos
Microbioma Gastrointestinal , Leite Humano , Oligossacarídeos , Prebióticos , Humanos , Microbioma Gastrointestinal/fisiologia , Leite Humano/química , Leite Humano/microbiologia , Oligossacarídeos/química , Probióticos , Lactente , Bactérias/metabolismo , Bifidobacterium/fisiologia
5.
Animal ; 18(8): 101243, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39106554

RESUMO

The performance of dairy cows is influenced by the microbial communities hosted within their digestive tract. While the rumen microbiota has long been associated with host phenotypes, the impact of the faecal microbiota remains elusive. In this study, we collected 697 faecal samples from commercial Holstein cows and analysed them with 16S rRNA gene analyses. For each animal, routinely recorded data, i.e., milk yield, fat yield, protein yield, fat content, protein content, and an aggregate production trait (pINEL) based on the French economic dairy index, were available to assess the links between the faecal microbiota and host production. Our findings revealed a strong and significant association between the structure of the bacterial and prokaryote community (ß-diversity) and dairy production. In addition, differential abundance analyses identified 48 genera whose abundances were significantly associated with pINEL, milk, fat and protein yield. Among these genera, the increased abundance of Bifidobacterium, and particularly an amplicon sequence variant with a 16S rRNA V3-V4 gene region identical to B. globosum and B. pseudolongum, was found to be the most important for high-yielding animals. Bifidobacterium seemed to be a potential key member of the bovine faecal microbiota that should be further investigated. Conversely, the p-1088-a5 gut group genus was found more abundant in low-productive cows. In conclusion, this study demonstrates significant associations between the faecal microbiota and the performance of dairy cows at the whole lactation scale. A better understanding of the physiology of the gut microbiota could help to improve dairy cow production.


Assuntos
Bifidobacterium , Fezes , Leite , RNA Ribossômico 16S , Animais , Bovinos/microbiologia , Fezes/microbiologia , Leite/microbiologia , Leite/química , Feminino , RNA Ribossômico 16S/análise , Microbioma Gastrointestinal , Lactação , Indústria de Laticínios
6.
Gut Microbes ; 16(1): 2387139, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39106231

RESUMO

Bacteriocins are broad or narrow-spectrum antimicrobial compounds that have received significant scientific attention due to their potential to treat infections caused by antibiotic-resistant pathogenic bacteria. The genome of Bifidobacterium pseudocatenulatum MM0196, an antimicrobial-producing, fecal isolate from a healthy pregnant woman, was shown to contain a gene cluster predicted to encode Pseudocin 196, a novel lantibiotic, in addition to proteins involved in its processing, transport and immunity. Following antimicrobial assessment against various indicator strains, protease-sensitive Pseudocin 196 was purified to homogeneity from cell-free supernatant. MALDI TOF mass spectrometry confirmed that the purified antimicrobial compound corresponds to a molecular mass of 2679 Da, which is consistent with that deduced from its genetic origin. Pseudocin 196 is classified as a lantibiotic based on its similarity to lacticin 481, a lanthionine ring-containing lantibiotic produced by Lactococcus lactis. Pseudocin 196, the first reported bacteriocin produced by a B. pseudocatenulatum species of human origin, was shown to inhibit clinically relevant pathogens, such as Clostridium spp. and Streptococcus spp. thereby highlighting the potential application of this strain as a probiotic to treat and prevent bacterial infections.


Assuntos
Antibacterianos , Bacteriocinas , Bifidobacterium , Bacteriocinas/farmacologia , Bacteriocinas/genética , Bacteriocinas/metabolismo , Bacteriocinas/química , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/metabolismo , Bifidobacterium/genética , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/metabolismo , Feminino , Clostridium/genética , Clostridium/efeitos dos fármacos , Clostridium/metabolismo , Fezes/microbiologia , Streptococcus/efeitos dos fármacos , Streptococcus/genética , Streptococcus/metabolismo , Gravidez , Família Multigênica , Testes de Sensibilidade Microbiana , Genoma Bacteriano , Probióticos/farmacologia
7.
Sci Rep ; 14(1): 19199, 2024 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-39160300

RESUMO

The main purpose was to determine the abundance of dominant phyla, Bifidobacterium spp., and Lactobacillus in breast milk of obese mothers versus normal-weights in fourth month of lactation in Iranian population. Sixty health women at the fourth month of breastfeeding, aged 18-40 years, were included and categorized based on body mass index (BMI) to the obese (BMI ≥ 30 kg/m2) and normal-weights (18.5 ≤ BMI ≤ 24.9). Bacterial DNA was extracted and qPCR of the 16S region was performed after human milk donation in a sterile condition. A multiple linear mixed model was used to determine the effective factors on the phyla population. Bifidobacterium spp. was significantly higher in milk of normal-weight group than the obese. The current weight showed a significant effect on the Actinobacteria abundance in milk. The Bacteroidetes and Firmicutes were significantly lower in mother's milk with cesarean section (p = 0.04). Pre-pregnancy obesity decreased the Firmicutes and Lactobacillus abundance in maternal milk (p = 0.04 and p = 0.01). The Actinobacteria and Bifidobacterium spp. showed a significant effect on infant's height (p = 0.008 and p = 0.04). The maternal current and pre-pregnancy weight showed an important effect on abundance of Actinobacteria and Bifidobacterium spp., as the good phyla and genus in milk which are associated with the infant's height.


Assuntos
Lactação , Leite Humano , Obesidade , Probióticos , Humanos , Feminino , Leite Humano/microbiologia , Adulto , Obesidade/microbiologia , Adulto Jovem , Adolescente , Bifidobacterium/isolamento & purificação , Bifidobacterium/genética , Aleitamento Materno , Índice de Massa Corporal , Lactobacillus/isolamento & purificação , Lactobacillus/genética , Gravidez , Irã (Geográfico)
8.
BMC Med Genomics ; 17(1): 216, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39160503

RESUMO

BACKGROUND: There is growing evidence for a relationship between gut microbiota and hepatic encephalopathy (HE). However, the causal nature of the relationship between gut microbiota and HE has not been thoroughly investigated. METHOD: This study utilized the large-scale genome-wide association studies (GWAS) summary statistics to evaluate the causal association between gut microbiota and HE risk. Specifically, two-sample Mendelian randomization (MR) approach was used to identify the causal microbial taxa for HE. The inverse variance weighted (IVW) method was used as the primary MR analysis. Sensitive analyses were performed to validate the robustness of the results. RESULTS: The IVW method revealed that the genus Bifidobacterium (OR = 0.363, 95% CI: 0.139-0.943, P = 0.037), the family Bifidobacteriaceae (OR = 0.359, 95% CI: 0.133-0.950, P = 0.039), and the order Bifidobacteriales (OR = 0.359, 95% CI: 0.133-0.950, P = 0.039) were negatively associated with HE. However, no causal relationship was observed among them after the Bonferroni correction test. Neither heterogeneity nor horizontal pleiotropy was found in the sensitivity analysis. CONCLUSION: Our MR study demonstrated a potential causal association between Bifidobacterium, Bifidobacteriaceae, and Bifidobacteriales and HE. This finding may provide new therapeutic targets for patients at risk of HE in the future.


Assuntos
Microbioma Gastrointestinal , Estudo de Associação Genômica Ampla , Encefalopatia Hepática , Análise da Randomização Mendeliana , Humanos , Encefalopatia Hepática/genética , Encefalopatia Hepática/microbiologia , Bifidobacterium/genética
9.
Bull Exp Biol Med ; 177(3): 313-317, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39126543

RESUMO

Inbred mouse strains KK.Cg-a/a and KK.Cg-Ay/a known as genetic models of type 2 diabetes mellitus significantly surpassed the control strain C57BL/6J in the body weight, relative weight of extractable fat, and basal blood glucose levels. Real-timePCR of fecal samples from KK.Cg-a/a and KK.Cg-Ay/a mice revealed dysbiosis typical of type 2 diabetes mellitus in humans and animals. Long-term intragastric administration of a suspension of Hafnia alvei bacteria had no effect on the above morphometric and biochemical parameters. At the same time, recovery of the Bacteroides spp. population in KK.Cg-Ay/a mice and a decrease in the number of Bifidobacterium spp. in KK.Cg-a/a mice were observed. The possibility of therapeutic use of the probiotic based on H. alvei is discussed.


Assuntos
Diabetes Mellitus Tipo 2 , Fezes , Microbioma Gastrointestinal , Hafnia alvei , Camundongos Endogâmicos C57BL , Probióticos , Animais , Diabetes Mellitus Tipo 2/microbiologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Microbioma Gastrointestinal/genética , Camundongos , Probióticos/administração & dosagem , Hafnia alvei/genética , Fezes/microbiologia , Masculino , Bacteroides/genética , Bifidobacterium/genética , Glicemia/metabolismo , Peso Corporal , Disbiose/microbiologia , Modelos Animais de Doenças
10.
Bull Exp Biol Med ; 177(3): 333-338, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39126545

RESUMO

We studied the effect of enteral administration of the glucocorticoid deflazacort (DFC, 1.2 mg/kg per day, 28 days) on the state of skeletal muscles and tissue ultrastructure, as well as the composition of the colon microbiota in dystrophin-deficient mdx mice. DFC has been shown to reduce the intensity of degeneration/regeneration cycles in muscle fibers of mdx mice. This effect of DFC was accompanied by normalization of the size of sarcomeres of skeletal muscles of mdx mice, improvement of the ultrastructure of the subsarcolemmal population of mitochondria, and an increase in the number of organelles, as well as normalization of the number of contact interactions between the sarcoplasmic reticulum and mitochondria. In addition, DFC had a corrective effect on the colon microbiota of mdx mice, which manifested in an increase in the number of the Bifidobacterium genus microorganisms and a decrease in the level of E. coli with reduced enzymatic activity.


Assuntos
Colo , Microbioma Gastrointestinal , Glucocorticoides , Camundongos Endogâmicos mdx , Músculo Esquelético , Pregnenodionas , Animais , Camundongos , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/patologia , Colo/ultraestrutura , Pregnenodionas/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/ultraestrutura , Músculo Esquelético/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Glucocorticoides/farmacologia , Distrofina/genética , Distrofina/deficiência , Distrofina/metabolismo , Bifidobacterium/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/ultraestrutura , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura
11.
Nat Commun ; 15(1): 6937, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138170

RESUMO

The honeybee gut microbiome is crucial for degrading diverse pollen glycans. Yet it is unclear how this process shapes the interactions among bacteria. Here, we demonstrate a conditional mutualistic interaction between strains of two honeybee gut bacteria Bifidobacterium asteroides and Gilliamella apicola. When co-occurring in vitro and in vivo, Bifidobacterium provides complementary demethylation service to promote Gilliamella growth on methylated homogalacturonan, an enriched polysaccharide of pectin. In exchange, Gilliamella shares digestive products with Bifidobacterium, through which a positive interaction is established. This positive interaction vanishes when Bifidobacterium is not required on a non-methylated diet. Results from biochemical and gene expression analyses combined with model simulation further suggest that the ratio change of the two major homogalacturonan breakdown products, galacturonic acid (GalA) and di-GalA, determines the bacterial interaction. This study unravels how glycan metabolism may shape the interactions between honeybee gut bacteria.


Assuntos
Bifidobacterium , Microbioma Gastrointestinal , Pectinas , Simbiose , Abelhas/microbiologia , Animais , Pectinas/metabolismo , Microbioma Gastrointestinal/fisiologia , Simbiose/fisiologia , Bifidobacterium/metabolismo , Bifidobacterium/genética , Polissacarídeos/metabolismo , Ácidos Hexurônicos
12.
J Transl Med ; 22(1): 657, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39010088

RESUMO

BACKGROUND: The combination of immune checkpoint inhibitors with radiotherapy can enhance the immunomodulation by RT and reduce the growth of distant unirradiated tumors (abscopal effect); however, the results are still not very satisfactory. Therefore, new treatment options are needed to enhance this effect. Our previous study showed that the combination of Bifidobacterium (Bi) and its specific monoclonal antibody (mAb) could target and alleviate hypoxia at the tumor site and act as a radiosensitizer. In this study, we explored the anti-tumor efficacy of quadruple therapy (Bi + mAb and RT + αPD-1). The current study also aimed to probe into the complex immune mechanisms underlying this phenomenon. METHODS: Constructed 4T1 breast and CT26 colon cancer tumor models. A comprehensive picture of the impact of constructed quadruple therapy was provided by tumor volume measurements, survival analysis, PET/CT imaging, immune cell infiltration analysis and cytokine expression levels. RESULTS: The abscopal effect was further amplified in the "cold" tumor model and prolonged survival in tumor-bearing mice. Bi can colonized in primary and secondary tumors and direct the mAb to reach the tumor site, activate complement, enhance the ADCC effect and initiate the innate immune response. Then combined with αPD-1 and radiotherapy to stimulate adaptive immune response and synergize with cytokines to expand the immune efficacy and generate effective anti-tumor immune response. CONCLUSIONS: Bi was used as an artificially implanted anaerobic target to cause a transient "infection" at the tumor, causing the tumor to become locally inflamed and "hot", and at the same time, mAb was used to target Bi to enhance the local immune effect of the tumor, and then combined with radiotherapy and αPD-1 to amplify the abscopal effect in multiple dimensions. Therefore, the present study provided a new idea for the multipotent immune-activating function of antibody-targeted anaerobic bacteria for the RT treatment of extensively metastasized cancer patients.


Assuntos
Anticorpos Monoclonais , Camundongos Endogâmicos BALB C , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Feminino , Bactérias Anaeróbias/imunologia , Camundongos , Bifidobacterium , Citocinas/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias/radioterapia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Terapia Combinada
13.
Nutrients ; 16(13)2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38999862

RESUMO

Irritable bowel syndrome (IBS) is a common gastrointestinal disorder with gut microbiota imbalance playing a significant role. There are increasing numbers of research studies exploring treatment options involving probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT), but it is still uncertain which treatment option is superior. The research was conducted on various databases and unpublished trial data (up to February 2023). Randomized controlled trials (RCTs) were screened for adult patients with IBS comparing interventions with placebo. Probiotics, prebiotics, synbiotics, and FMT were assessed for their impact using mean difference and Bayesian network meta-analysis. Out of 6528 articles, 54 were included for probiotics, 7 for prebiotics/synbiotics, and 6 for FMT. Probiotics showed improvement in IBS symptoms, particularly with Bifidobacterium and Lactobacillus strains. Prebiotics and synbiotics did not show significant improvement. Network meta-analysis indicated the favorable effects of probiotics (OR = 0.53, 95% CI, 0.48 to 0.59) and FMT (OR = 0.46, 95% CI, 0.33 to 0.64) on IBS, with no serious adverse events reported. In short, probiotics and FMT are effective for managing IBS, with Bifidobacterium and Lactobacillus being dominant strains. However, the most effective probiotic combination or strain remains unclear, while prebiotics and synbiotics did not show significant improvement.


Assuntos
Transplante de Microbiota Fecal , Síndrome do Intestino Irritável , Metanálise em Rede , Prebióticos , Probióticos , Simbióticos , Síndrome do Intestino Irritável/terapia , Síndrome do Intestino Irritável/microbiologia , Humanos , Prebióticos/administração & dosagem , Probióticos/uso terapêutico , Probióticos/administração & dosagem , Simbióticos/administração & dosagem , Resultado do Tratamento , Microbioma Gastrointestinal , Ensaios Clínicos Controlados Aleatórios como Assunto , Bifidobacterium , Adulto , Feminino , Lactobacillus , Masculino
14.
BMC Genomics ; 25(1): 718, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39054474

RESUMO

INTRODUCTION: Our understanding of particular gut microbiota members such as Bifidobacterium and Enterococcus in low-middle-income countries remains very limited, particularly early life strain-level beneficial traits. This study addresses this gap by exploring a collection of bacterial strains isolated from the gut of Zimbabwean infants; comparing their genomic characteristics with strains isolated from infants across North America, Europe, and other regions of Africa. MATERIALS AND METHOD: From 110 infant stool samples collected in Harare, Zimbabwe, 20 randomly selected samples were used to isolate dominant early-life gut microbiota members Bifidobacterium and Enterococcus. Isolated strains were subjected to whole genome sequencing and bioinformatics analysis including functional annotation of carbohydrates, human milk oligosaccharide (HMO) and protein degradation genes and clusters, and the presence of antibiotic resistance genes (ARGs). RESULTS: The study observed some location-based clustering within the main five identified taxonomic groups. Furthermore, there were varying and overall species-specific numbers of genes belonging to different GH families encoded within the analysed dataset. Additionally, distinct strain- and species-specific variances were identified in the potential of Bifidobacterium for metabolizing HMOs. Analysis of putative protease activity indicated a consistent presence of gamma-glutamyl hydrolases in Bifidobacterium, while Enterococcus genomes exhibited a high abundance of aspartyl peptidases. Both genera harboured resistance genes against multiple classes of antimicrobial drugs, with Enterococcus genomes containing a higher number of ARGs compared to Bifidobacterium, on average. CONCLUSION: This study identified promising probiotic strains within Zimbabwean isolates, offering the potential for early-life diet and microbial therapies. However, the presence of antibiotic resistance genes in infant-associated microbes raises concerns for infection risk and next-stage probiotic development. Further investigation in larger cohorts, particularly in regions with limited existing data on antibiotic and probiotic use, is crucial to validate these initial insights. IMPACT STATEMENT: This research represents the first investigation of its kind in the Zimbabwean context, focusing on potential probiotic strains within the early-life gut microbiota. By identifying local probiotic strains, this research can contribute to the development of probiotic interventions that are tailored to the Zimbabwean population, which can help address local health challenges and promote better health outcomes for infants. Another essential aspect of the study is the investigation of antimicrobial resistance genes present in Zimbabwean bacterial strains. Antimicrobial resistance is a significant global health concern, and understanding the prevalence and distribution of resistance genes in different regions can help inform public health policies and interventions.


Assuntos
Bifidobacterium , Enterococcus , Microbioma Gastrointestinal , Humanos , Zimbábue , Lactente , Microbioma Gastrointestinal/genética , Enterococcus/genética , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Bifidobacterium/efeitos dos fármacos , Genômica , Genoma Bacteriano , Fezes/microbiologia , Sequenciamento Completo do Genoma , Estudos de Coortes , Filogenia
15.
Food Res Int ; 191: 114720, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39059916

RESUMO

Mangosteen (Garcinia mangostana L.) is a tasty, polyphenol-rich tropical fruit. The edible part is highly appreciated by its aroma, taste and texture. The non-edible part, rich in polyphenols, has been traditionally used in Thai medicine. In this work, flavonoids and phenolic acid/derivatives were identified in mangosteen extracts (ME) from edible and non-edible portions. We first studied the effects of MEs on the growth, metabolism, antioxidant capacity, biofilm formation and antimicrobial capacity of eight bifidobacteria and lactobacilli strains from intestinal origin and two commercial probiotic strains (BB536 and GG). ME concentrations higher than 10-20 % were inhibitory for all strains. However, ME concentrations of 5 % significantly (P < 0.01) increased all strains antioxidant capacity, reduced biofilm-formation, and enhanced inhibition against Gram-positive pathogens. To apply these knowledge, bifunctional fermented milk products were elaborated with 5 % ME and individual strains, which were selected taking into account their growth with ME, and the widest range of values on antioxidant capacity, biofilm formation and antimicrobial activity (bifidobacteria INIA P2 and INIA P467, lactobacilli INIA P459 and INIA P708, and reference strain GG). Most strains survived well manufacture, refrigerated storage and an in vitro simulation of major conditions encountered in the gastrointestinal tract. As expected, products supplemented with ME showed higher polyphenol content and antioxidant capacity levels than control. After sensory evaluation, products containing strains INIA P2, INIA P708 and GG outstood as best.


Assuntos
Antioxidantes , Biofilmes , Produtos Fermentados do Leite , Garcinia mangostana , Lactobacillus , Extratos Vegetais , Extratos Vegetais/farmacologia , Garcinia mangostana/química , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Antioxidantes/farmacologia , Lactobacillus/efeitos dos fármacos , Lactobacillus/metabolismo , Produtos Fermentados do Leite/microbiologia , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Probióticos , Flavonoides/farmacologia , Flavonoides/análise , Humanos , Frutas/química , Frutas/microbiologia , Fermentação , Hidroxibenzoatos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Polifenóis/farmacologia
16.
Nutrients ; 16(14)2024 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-39064788

RESUMO

Bifidobacterium animalis subsp. lactis GCL2505 in combination with inulin has been shown to have several health benefits, including an improvement in the intestinal microbiota and a reduction in human visceral fat. Previous studies have suggested that the visceral fat reduction of GCL2505 and inulin may be achieved by improving daily energy expenditure. This parallel, placebo-controlled, randomized, double-blind study was conducted to evaluate the effects of GCL2505 and inulin on resting energy expenditure (REE) in overweight or mildly obese Japanese adults (n = 44). Participants ingested 1 × 1010 colony forming units of GCL2505 and 5.0 g of inulin daily for 4 weeks. REE score at week 4 was set as the primary endpoint. At week 4, the REE score of the GCL2505 and inulin group was significantly higher than that of the placebo group, with a difference of 84.4 kcal/day. In addition, fecal bifidobacteria counts were significantly increased in the GCL2505 and inulin group. Our results indicated that the intake of GCL2505 and inulin improves energy balance, which is known to be a major factor of obesity, by modulating the microbiota in the gut. This is the first report to demonstrate the effects of probiotics and dietary fiber on REE in humans.


Assuntos
Fibras na Dieta , Fezes , Microbioma Gastrointestinal , Inulina , Obesidade , Probióticos , Humanos , Método Duplo-Cego , Masculino , Feminino , Probióticos/administração & dosagem , Fibras na Dieta/administração & dosagem , Fibras na Dieta/farmacologia , Pessoa de Meia-Idade , Adulto , Inulina/administração & dosagem , Inulina/farmacologia , Fezes/microbiologia , Microbioma Gastrointestinal/fisiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Obesidade/microbiologia , Obesidade/dietoterapia , Metabolismo Energético , Bifidobacterium , Sobrepeso/microbiologia , Sobrepeso/dietoterapia , Bifidobacterium animalis , Japão , Metabolismo Basal/efeitos dos fármacos
17.
Pharmacol Rep ; 76(4): 740-753, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38951480

RESUMO

Asthma is a lifelong condition with varying degrees of severity and susceptibility to symptom control. Recent studies have examined the effects of individual genus, species, and strains of probiotic microorganisms on the course of asthma. The present review aims to provide an overview of current knowledge on the use of probiotic microorganisms, mainly bacteria of the genus Lactobacillus and Bifidobacterium, in asthma prevention and treatment. Recent data from clinical trials and mouse models of allergic asthma indicate that probiotics have therapeutic potential in this condition. Animal studies indicate that probiotic microorganisms demonstrate anti-inflammatory activity, attenuate airway hyperresponsiveness (AHR), and reduce airway mucus secretion. A randomized, double-blind, placebo-controlled human trials found that combining multi-strain probiotics with prebiotics yielded promising outcomes in the treatment of clinical manifestations of asthma. It appears that probiotic supplementation is safe and significantly reduces the frequency of asthma exacerbations, as well as improved forced expiratory volume and peak expiratory flow parameters, and greater attenuation of inflammation. Due to the small number of available clinical trials, and the use of a wide range of probiotic microorganisms and assessment methods, it is not possible to draw clear conclusions regarding the use of probiotics as asthma treatments.


Assuntos
Asma , Probióticos , Probióticos/uso terapêutico , Probióticos/administração & dosagem , Asma/terapia , Asma/prevenção & controle , Humanos , Animais , Bifidobacterium , Lactobacillus
18.
Nutrients ; 16(13)2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38999750

RESUMO

(1) Background: Recently, academic studies are demonstrating that the cholesterol-lowering effects of pectin oligosaccharides (POSs) are correlated to intestinal flora. However, the mechanisms of POS on cholesterol metabolisms are limited, and the observations of intestinal flora are lacking integrative analyses. (2) Aim and methods: To reveal the regulatory mechanisms of POS on cholesterol metabolism via an integrative analysis of the gut microbiota, the changes in gut microbiota structure and metabolite composition after POS addition were investigated using Illumina MiSeq sequencing and non-targeted metabolomics through in vitro gut microbiota fermentation. (3) Results: The composition of fecal gut flora was adjusted positively by POS. POS increased the abundances of the cholesterol-related bacterial groups Bacteroidetes, Bifidobacterium and Lactobacillus, while it decreased conditional pathogenic Escherichia coli and Enterococcus, showing good prebiotic activities. POS changed the composition of gut microbiota fermentation metabolites (P24), causing significant changes in 221 species of fermentation metabolites in a non-targeted metabolomics analysis and promoting the production of short-chain fatty acids. The abundances of four types of cholesterol metabolism-related metabolites (adenosine monophosphate, cyclic adenosine monophosphate, guanosine and butyrate) were significantly higher in the P24 group than those in the control group without POS addition. (4) Conclusion: The abovementioned results may explain the hypocholesterolemic effects of POS and promotion effects on cholesterol efflux of P24. These findings indicated that the potential regulatory mechanisms of citrus POS on cholesterol metabolism are modulated by cholesterol-related gut microbiota and specific metabolites.


Assuntos
Colesterol , Fezes , Fermentação , Microbioma Gastrointestinal , Oligossacarídeos , Pectinas , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Pectinas/farmacologia , Pectinas/metabolismo , Colesterol/metabolismo , Oligossacarídeos/farmacologia , Fezes/microbiologia , Humanos , Prebióticos , Masculino , Metabolômica , Ácidos Graxos Voláteis/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium/efeitos dos fármacos , Feminino , Bactérias/metabolismo , Bactérias/efeitos dos fármacos , Bactérias/classificação , Citrus
19.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-39038938

RESUMO

With the increasing prevalence of age-related chronic diseases burdening healthcare systems, there is a pressing need for innovative management strategies. Our study focuses on the gut microbiota, essential for metabolic, nutritional, and immune functions, which undergoes significant changes with aging. These changes can impair intestinal function, leading to altered microbial diversity and composition that potentially influence health outcomes and disease progression. Using advanced metagenomic sequencing, we explore the potential of personalized probiotic supplements in 297 older adults by analyzing their gut microbiota. We identified distinctive Lactobacillus and Bifidobacterium signatures in the gut microbiota of older adults, revealing probiotic patterns associated with various population characteristics, microbial compositions, cognitive functions, and neuroimaging results. These insights suggest that tailored probiotic supplements, designed to match individual probiotic profile, could offer an innovative method for addressing age-related diseases and functional declines. Our findings enhance the existing evidence base for probiotic use among older adults, highlighting the opportunity to create more targeted and effective probiotic strategies. However, additional research is required to validate our results and further assess the impact of precision probiotics on aging populations. Future studies should employ longitudinal designs and larger cohorts to conclusively demonstrate the benefits of tailored probiotic treatments.


Assuntos
Envelhecimento , Suplementos Nutricionais , Microbioma Gastrointestinal , Probióticos , Probióticos/uso terapêutico , Probióticos/administração & dosagem , Humanos , Idoso , Feminino , Masculino , Idoso de 80 Anos ou mais , Pessoa de Meia-Idade , Lactobacillus/genética , Metagenômica/métodos , Bifidobacterium
20.
NPJ Biofilms Microbiomes ; 10(1): 59, 2024 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-39034349

RESUMO

The dominant bacteria in the hindgut of calves play an important role in their growth and health, which could even lead to lifelong consequences. However, the identification of core probiotics in the hindgut and its mechanism regulating host growth remain unclear. Here, a total of 1045 fecal samples were analyzed by 16S rRNA gene sequencing from the 408 Holstein dairy calves at the age of 0, 14, 28, 42, 56, and 70 days to characterize the dynamic changes of core taxa. Moreover, the mechanisms of nutrient metabolism of calf growth regulated by core bacteria were investigated using multi-omics analyses. Finally, fecal microbiota transplantation (FMT) in mice were conducted to illustrate the potential beneficial effects of core bacteria. Four calf enterotypes were identified and enterotypes dominated by Bifidobacterium and Oscillospiraceae_UCG-005 were representative. The frequency of enterotype conversion shifted from variable to stable. The close relationship observed between phenotype and enterotype, revealing a potential pro-growth effect of Bifidobacterium, might be implemented by promoting the use of carbohydrate, activating the synthesis of volatile fatty acids, amino acids and vitamin B6, and inhibiting methane production in the hindgut. The FMT results indicated the beneficial effect of Bifidobacterium on host growth and hindgut development. These results support the notion that the Bifidobacterium-dominated fecal microbiome would be an important driving force for promoting the host growth in the early life. Our findings provide new insights into the potential probiotic mining and application strategies to promote the growth of young animals or improve their growth retardation.


Assuntos
Bifidobacterium , Transplante de Microbiota Fecal , Fezes , Microbioma Gastrointestinal , RNA Ribossômico 16S , Animais , Fezes/microbiologia , Bovinos , RNA Ribossômico 16S/genética , Bifidobacterium/genética , Bifidobacterium/crescimento & desenvolvimento , Camundongos , Transplante de Microbiota Fecal/métodos , Fenótipo , Probióticos/administração & dosagem , Filogenia , DNA Bacteriano/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...