Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Pediatr Res ; 93(1): 49-55, 2023 01.
Article in English | MEDLINE | ID: mdl-35505080

ABSTRACT

Our pilot RCT found that probiotic supplementation with the three-strain bifidobacterial product (B. breve M-16V, B. longum subsp. infantis M-63 and B. longum subsp. longum BB536) attenuates gut dysbiosis, increases stool short-chain fatty acid (SCFA) levels and improves the growth of head circumference in neonates with congenital gastrointestinal surgical conditions (CGISC). In this article, we have provided guidelines for designing future multicentre RCTs based on the experience gained from our pilot RCT. The recommendations include advice about sample size, potential confounders, outcomes of interest, probiotic strain selection, storage, dose, duration and microbial quality assurance, collection of stool samples, storage and analysis and reporting. Following these guidelines will increase the validity of future RCTs in this area and hence confidence in their results. IMPACT: Probiotic supplementation attenuates gut dysbiosis, increases stool short-chain fatty acid (SCFA) levels and improves the growth of head circumference in neonates with congenital gastrointestinal surgical conditions. The current review provides evidence-based guidelines to conduct adequately powered RCTs in this field.


Subject(s)
Gastrointestinal Diseases , Probiotics , Infant, Newborn , Humans , Dysbiosis , Probiotics/therapeutic use , Bifidobacterium , Feces/microbiology
2.
Sci Adv ; 8(16): eabm5944, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35452284

ABSTRACT

Dogs are uniquely associated with human dispersal and bring transformational insight into the domestication process. Dingoes represent an intriguing case within canine evolution being geographically isolated for thousands of years. Here, we present a high-quality de novo assembly of a pure dingo (CanFam_DDS). We identified large chromosomal differences relative to the current dog reference (CanFam3.1) and confirmed no expanded pancreatic amylase gene as found in breed dogs. Phylogenetic analyses using variant pairwise matrices show that the dingo is distinct from five breed dogs with 100% bootstrap support when using Greenland wolf as the outgroup. Functionally, we observe differences in methylation patterns between the dingo and German shepherd dog genomes and differences in serum biochemistry and microbiome makeup. Our results suggest that distinct demographic and environmental conditions have shaped the dingo genome. In contrast, artificial human selection has likely shaped the genomes of domestic breed dogs after divergence from the dingo.


Subject(s)
Canidae , Wolves , Animals , Australia , Breeding , Canidae/genetics , Dogs , Phylogeny , Wolves/genetics
3.
Article in English | MEDLINE | ID: mdl-35185013

ABSTRACT

OBJECTIVE: Evidence indicates that multistrain probiotics benefit preterm infants more than single-strain (SS) probiotics. We assessed the effects of SS versus triple-strain (TS) probiotic supplementation (PS) in extremely preterm (EP) infants. DESIGN: EP infants (gestational age (GA) <28 weeks) were randomly allocated to TS or SS probiotic, assuring blinding. Reference (REF) group was EP infants in the placebo arm of our previous probiotic trial. PS was commenced with feeds and continued until 37 weeks' corrected GA. Primary outcome was time to full feed (TFF: 150 mL/kg/day). Secondary outcomes included short-chain fatty acids and faecal microbiota collected at T1 (first week) and T2 (after 3 weeks of PS) using 16S ribosomal RNA gene sequencing. RESULTS: 173 EP (SS: 86, TS: 87) neonates with similar GA and birth weight (BW) were randomised. Median TFF was comparable (11 (IQR 8-16) vs 10 (IQR 8-16) days, p=0.92). Faecal propionate (SS, p<0.001, and TS, p=0.0009) and butyrate levels (TS, p=0.029) were significantly raised in T2 versus T1 samples. Secondary clinical outcomes were comparable. At T2, alpha diversity was comparable (p>0.05) between groups, whereas beta-diversity analysis revealed significant differences between PS and REF groups (both p=0.001). Actinobacteria were higher (both p<0.01), and Proteobacteria, Firmicutes and Bacteroidetes were lower in PS versus REF. Gammaproteobacteria, Clostridia and Negativicutes were lower in both PS versus REF. CONCLUSION: TFF in EP infants was similar between SS and TS probiotics. Both probiotics were effective in reducing dysbiosis (higher bifidobacteria and lower Gammaproteobacteria). Long-term significance of increased propionate and butyrate needs further studies. TRIAL REGISTRATION NUMBER: ACTRN 12615000940572.


Subject(s)
Infant, Extremely Premature , Probiotics , Bifidobacterium , Butyrates , Firmicutes , Humans , Infant , Infant, Newborn , Probiotics/therapeutic use , Propionates
4.
Pediatr Res ; 92(4): 1122-1131, 2022 10.
Article in English | MEDLINE | ID: mdl-34980887

ABSTRACT

OBJECTIVE: To evaluate whether probiotic supplementation attenuates gut-dysbiosis in neonates with congenital gastrointestinal surgical conditions (CGISC). METHODS: Sixty-one neonates (≥35 weeks gestation) with CGISC were randomised to receive daily supplementation with a triple-strain bifidobacterial probiotic (n = 30) or placebo (n = 31) until discharge. Stool microbiota was analysed using 16S ribosomal RNA gene sequencing on samples collected before (T1), 1 week (T2), and 2 weeks (T3) after supplementation and before discharge (T4). The primary outcome was the sum of the relative abundance of potentially pathogenic families of Clostridiaceae, Enterobacteriaceae, Enterococcaceae, Pseudomonaceae, Staphylococcaeae, Streptococcaceae, and Yersiniaceae at T3. RESULTS: The median gestational age [38 weeks (IQR: 37.1-38.9)] was similar in both groups. The probiotic group had lower rates of caesarean deliveries (40% versus 70%, p = 0.02). The relative abundance of potentially pathogenic families was lower in the probiotic group compared to placebo at T3 [(median: 50.4 (IQR: 26.6-67.6) versus 67.1 (IQR: 50.9-96.2); p = 0.04). Relative abundance of Bifidobacteriaceae was higher in the probiotic group at T3 [(median: 39.8 (IQR: 24.9-52.1) versus 0.03 (IQR 0.02-2.1); p < 0.001). Stratified analysis continued to show a higher abundance of Bifidobacteriaceae in the probiotic group, irrespective of the mode of delivery. CONCLUSIONS: Probiotic supplementation attenuated gut dysbiosis in neonates with CGISC. TRIAL REGISTRATION: http://www.anzctr.org.au (ACTRN12617001401347). IMPACT: Probiotic supplementation attenuates gut dysbiosis and improves stool short-chain fatty acid levels in neonates with congenital gastrointestinal surgical conditions. This is the second pilot RCT of probiotic supplementation in neonates with congenital gastrointestinal conditions. These findings will pave the way for conducting multicentre RCTs in this area.


Subject(s)
Gastrointestinal Diseases , Probiotics , Infant, Newborn , Pregnancy , Female , Humans , Infant , Dysbiosis , Pilot Projects , Probiotics/therapeutic use , Bifidobacterium , Fatty Acids, Volatile
5.
Pediatr Res ; 88(6): 878-886, 2020 12.
Article in English | MEDLINE | ID: mdl-32179871

ABSTRACT

BACKGROUND: There is limited information on gut microbiota of neonates with congenital gastrointestinal surgical conditions (CGISCs) available. METHODS: This study compared stool microbiota and short-chain fatty acids (SCFAs) of 37 term infants with CGISCs with 36 term healthy infants (HIs). Two stool samples were collected from each infant: as soon as possible after birth (week 1) and 10-14 days of life (week 2). RESULTS: Bacterial richness and alpha diversity were comparable between CGISCs and HIs at week 1 and week 2 (all p > 0.05). Beta diversity analysis revealed that at week 1, CGISCs had similar community structures to HIs (p = 0.415). However, by week 2, community structures of CGISCs were significantly different from HIs (p = 0.003). At week 1, there were no significant differences in the relative abundances of genera Bifidobacterium and Bacteroides between CGISCs and HIs. At week 2, the relative abundance of Bifidobacterium was significantly lower in CGISCs (mean percentage 7.21 ± 13.49 vs. 28.96 ± 19.6; p = 0.002). Bacteroides were also less abundant in the CGISC group (mean percentage 0.12 ± 0.49 vs. 6.59 ± 8.62; p = 0.039). Relative abundance of genera Pseudomonas and Escherichia-Shigella were higher in CGISCs. At week 2, stool concentrations of all SCFAs were lower in CGISCs (all p < 0.001). CONCLUSIONS: During hospitalization, neonates with CGISCs develop gut dysbiosis and deficiency of SCFAs. IMPACT: During hospitalisation, neonates with congenital gastrointestinal surgical conditions develop gut dysbiosis with deficiency of Bifidobacteria and Bacteroides and increased abundance of Escherichia-Shigella and Pseudomonas. They also have low levels of short chain fatty acids in their stools compared to healthy infants. This is the first study evaluating the gut microbiota using 16S ribosomal RNA sequencing methods and stool short chain fatty acids in neonates with congenital gastrointestinal surgical conditions and comparing them to healthy infants. The findings of this study will pave the way for randomised trials of bifidobacterial supplementation in neonates with congenital gastrointestinal surgical conditions.


Subject(s)
Gastrointestinal Diseases/complications , Gastrointestinal Microbiome , Bacteroides , Bifidobacterium , Calibration , Escherichia coli , Fatty Acids, Volatile/metabolism , Feces/microbiology , Female , Gas Chromatography-Mass Spectrometry , Gastrointestinal Diseases/congenital , Hospitalization , Humans , Infant, Extremely Premature , Infant, Newborn , Infant, Premature , Linear Models , Male , Polymerase Chain Reaction , Prospective Studies , Pseudomonas , RNA, Ribosomal, 16S , Risk Factors , Shigella , Treatment Outcome
6.
Nutrients ; 11(4)2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30959746

ABSTRACT

Lactobacillus strains have shown efficacy in attenuating inflammation. This study evaluated the potential of Lactobacillus fermentum PC1 for the treatment of rheumatoid arthritis (RA) using a murine model of collagen-induced arthritis. On Day 1, healthy DBA/1 mice (six to eight weeks of age) were immunized, with 100 µg of Chicken Type 11 collagen emulsified in complete Freund's adjuvant (CFA) by intradermal injection, at the base of the tail. On Day 21, the mice were immunized intraperitoneally with 100 µg of Bovine Type11 collagen in phosphate buffered saline (PBS). On Day 28, the mice were immunized intraperitoneally with 50 µg of lipopolysaccharide (LPS). Viable L. fermentum PC1 (1 × 108 colony forming units) was given daily from Day two until the end of the experiment. From Day 21 onwards, the mice were monitored daily for clinical signs of arthritis. On Day 44, the experiment was terminated. Paws were obtained for histology and serum for cytokine assays. L. fermentum PC1-fed mice had significantly reduced paw inflammation as well as decreased synovial infiltration and less cartilage damage. Circulating serum cytokine profiles revealed decreased IL-12 and increased anti-inflammatory cytokines, namely IL-4 and IL-10. Thus, early administration of L. fermentum PC1 could prove to be a valuable therapeutic agent in the management of RA.


Subject(s)
Arthritis, Experimental/chemically induced , Collagen Type II/toxicity , Inflammation/drug therapy , Limosilactobacillus fermentum/physiology , Probiotics/pharmacology , Animals , Arthritis, Experimental/therapy , Joints/drug effects , Joints/pathology , Male , Mice , Mice, Inbred DBA
7.
BMC Res Notes ; 11(1): 567, 2018 Aug 08.
Article in English | MEDLINE | ID: mdl-30089517

ABSTRACT

OBJECTIVE: It is well established that polyethylene (PE) wear particles induce macrophage production of cytokines and mediators associated with the pathogenesis of inflammatory osteolysis. The objective of this study was to examine the potential of three Lactobacillus strains to attenuate the TNF-α cytokine response of macrophages exposed to Ceridust 3615 PE particles. An in vitro experimental model using the RAW 246.7 macrophage cell line and PE particles was utilized. RESULTS: Lactobacillus strains were found to modulate the cytokines in a strain and dose specific manner. Only the Lactobacillus acidophilus strain that was tested was able to attenuate PE particle-induced TNF-α production by RAW 246.7 macrophages. This effect was independent of IL-10 cytokine levels since all three strains of lactobacilli yielded comparable levels of IL-10. It was concluded that some, but not all, Lactobacillus strains may be useful in reducing the risk of inflammatory osteolysis and that further studies in appropriate in vivo models are warranted. Furthermore, this in vitro model can be used to evaluate the inflammatory potential of new materials being tested for use as joint implants.


Subject(s)
Inflammation , Lactobacillus , Macrophages/drug effects , Animals , Cytokines , Mice , Osteolysis , Polyethylene/toxicity , Tumor Necrosis Factor-alpha
8.
Vaccines (Basel) ; 4(3)2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27447674

ABSTRACT

This study examined the influences of the dosage of the adjuvant, the nature of the antigen and the host genetics on the capacity of L. fermentum PC1 (PC1) to function as an oral adjuvant. BALB/c and DBA/1 mice were vaccinated with either ovalbumin (OVA) or Salmonella Typhimurium on days 0 and 14, Mice were also dosed with the PC1 (108 CFU or 10(11) CFU per dose per mouse) with the antigens (days 0 and 14) and alone (days -1 and 13). The higher PC1 dose elicited a greater specific serum IgG2a response than IgG1 for both antigens and mice strains, indicating a Th1-biased humoral immune response. The Th1 bias was also observed at the cellular level with greater specific IFN-γ levels than IL-4 and IL-10 with both antigen types and mouse strains. With the particulate antigen, the lower dose of PC1 elicited a Th1 bias at the cellular level, but a balanced Th1/Th2 response at the systemic humoral level. With the soluble antigen, a strong Th1-biased response occurred at the cellular level while the systemic humoral response was Th2-biased. In conclusion, PC1 at the higher dose was an excellent Th1 adjuvant, which was unaffected by the nature of the antigen or the host's genetic background.

9.
J Matern Fetal Neonatal Med ; 29(23): 3751-5, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26821074

ABSTRACT

BACKGROUND: Gut development, function and colonisation are impaired in animal models of prematurity with intrauterine growth restriction (IUGR). The effect of Bifidobacterium breve (B. breve) supplementation on faecal bifidobacteria in small for gestational age (SGA: birth weight <10th centile due to IUGR) preterm infants is not known. OBJECTIVE: We compared B. breve M-16V supplementation effect on faecal bifidobacteria in preterm (<33 weeks) SGA versus non-SGA infants in the two arms of our randomised controlled trial. RESULTS: There were no baseline differences in the proportion of detectable B. breve counts between SGA versus non-SGA infants [probiotic: 7 (33%) versus 22 (42%), p = 0.603; placebo: 1 (6%) versus 1 (2%), p = 0.429]. B. breve counts did not differ between SGA and non-SGA infants in response to treatment (p = 0.589), after adjusting for baseline count (p < 0.001) and treatment allocation (p < 0.001). An interaction term between growth status and treatment showed negligible change (p = 0.938). Probiotic treated SGA infants reached full feeds earlier than SGA controls (HR 2.00, 95% CI 1.05-3.82, p = 0.035): Median (IQR): 16 (12-26) versus 19 (11-25) days, after adjustment for age at starting feeds and gestation <28 weeks. CONCLUSION: Response to B. breve M-16V supplementation was not significantly different in preterm (<33 weeks) SGA versus non-SGA infants.


Subject(s)
Bifidobacterium breve , Feces/microbiology , Fetal Growth Retardation/microbiology , Infant, Extremely Premature , Infant, Premature, Diseases/microbiology , Probiotics/therapeutic use , Bacterial Load , Double-Blind Method , Female , Fetal Growth Retardation/therapy , Gestational Age , Humans , Infant, Newborn , Infant, Premature, Diseases/therapy , Male , Proportional Hazards Models , Statistics, Nonparametric
10.
PLoS One ; 9(3): e89511, 2014.
Article in English | MEDLINE | ID: mdl-24594833

ABSTRACT

BACKGROUND: Probiotic supplementation significantly reduces the risk of necrotising enterocolitis (NEC) and all cause mortality in preterm neonates. Independent quality assessment is important before introducing routine probiotic supplementation in this cohort. AIM: To assess product quality, and confirm that Bifidobacterium breve (B. breve) M-16V supplementation will increase fecal B. breve counts without adverse effects. METHODS AND PARTICIPANTS: Strain identity (16S rRNA gene sequencing), viability over 2 year shelf-life were confirmed, and microbial contamination of the product was ruled out. In a controlled trial preterm neonates (Gestation <33 weeks) ready to commence or on feeds for <12 hours were randomly allocated to either B. breve M-16V (3×109 cfu/day) or placebo (dextrin) supplementation until the corrected age 37 weeks. Stool samples were collected before (S1) and after 3 weeks of supplementation (S2) for studying fecal B. breve levels using quantitative PCR (Primary outcome). Secondary outcomes included total fecal bifidobacteria and NEC≥Stage II. Categorical and continuous outcomes were analysed using Chi-square and Mann-Whitney tests, and McNemar and Wilcoxon signed-rank tests for paired comparisons. RESULTS: A total of 159 neonates (Probiotic: 79, Placebo: 80) were enrolled. Maternal and neonatal demographic characteristics were comparable between the groups. The proportion of neonates with detectable B. breve increased significantly post intervention: Placebo: [S1:2/66 (3%), S2: 25/66 (38%), p<0.001] Probiotic: [S1: 29/74 (40%), S2: 67/74 (91%), p<0.001]. Median S1 B. breve counts in both groups were below detection (<4.7 log cells x g(-1)), increasing significantly in S2 for the probiotic group (log 8.6) while remaining <4.7 log in the control group (p<0.001). There were no adverse effects including probiotic sepsis and no deaths. NEC≥Stage II occurred in only 1 neonate (placebo group). CONCLUSION: B. breve M-16V is a suitable probiotic strain for routine use in preterm neonates. TRIAL REGISTRATION: Australia New Zealand Clinical Trial Registry ACTRN 12609000374268.


Subject(s)
Bifidobacterium , Enterocolitis, Necrotizing/prevention & control , Feces/microbiology , Infant, Newborn, Diseases/prevention & control , Infant, Premature , Probiotics , Base Sequence , Bifidobacterium/genetics , DNA Primers , DNA, Bacterial/genetics , Humans , Infant, Newborn , Polymerase Chain Reaction , Treatment Outcome
11.
J Microbiol Methods ; 87(2): 165-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21906633

ABSTRACT

Community-level selection is an important concept in evolutionary biology and has been predicted to arise in systems that are spatially structured. Here we develop an experimental model for spatially-structured bacterial communities based on coaggregating strains and test their relative fitness under a defined selection pressure. As selection we apply protozoan grazing in a defined, continuous culturing system. We demonstrate that a slow-growing bacterial strain Blastomonas natatoria 2.1, which forms coaggregates with Micrococcus luteus, can outcompete a fast-growing, closely related strain Blastomonas natatoria 2.8 under conditions of protozoan grazing. The competitive benefit provided by spatial structuring has implications for the evolution of natural bacterial communities in the environment.


Subject(s)
Biological Evolution , Micrococcus/growth & development , Models, Biological , Ochromonas/physiology , Selection, Genetic , Sphingomonadaceae/growth & development , Ecosystem
SELECTION OF CITATIONS
SEARCH DETAIL
...