-
1.
Efficacy of probiotics or synbiotics in critically ill patients: A systematic review and meta-analysis.
Lou, J, Cui, S, Huang, N, Jin, G, Chen, C, Fan, Y, Zhang, C, Li, J
Clinical nutrition ESPEN. 2024;59:48-62
-
-
-
Free full text
-
Plain language summary
The intestinal microbiota is a complex microbial community that plays an irreplaceable role in human life. Intestinal dysbiosis is very common in patients with critical illnesses. The aim of this study was to assess the efficacy and safety of probiotics or synbiotics in preventing ventilator associated pneumonia (VAP) in critically ill patients in the intensive care unit (ICU). This study was a systematic review and meta-analysis of thirty-three trials (n=4 retrospective studies and n=29 randomised controlled studies). A total of 7886 patients were grouped into the probiotics or synbiotics group (n= 4065) and control group (n= 3821). Results showed that probiotics or synbiotics significantly reduced the incidence of VAP and sepsis, as well as the duration of mechanical support, length of hospital stay, length of ICU stay, and ICU morality. Authors concluded that probiotics or synbiotics supplementation plays a beneficial role in critically ill patients and presents a novel approach to the management of critical diseases.
Abstract
BACKGROUND This latest systematic review and meta-analysis aim to examine the effects of probiotic and synbiotic supplementation in critically ill patients. METHODS Relevant articles were retrieved from PubMed, Embase, the Cochrane Database, and the Web of Science. The primary output measure was the incident of ventilator-associated pneumonia, and the secondary outputs were diarrhea, Clostridium diffusion infection (CDI), incident of sepsis, incident of hospital acquired pneumonia, duration of mechanical exploitation, ICU mortality rate, length of ICU stay, in hospital mortality, and length of hospital stay. Data were pooled and expressed as Relative Risk(RR) and Standardized Mean Difference (SMD) with a 95 % confidence interval (CI). RESULTS 33 studies were included in this systematic review and meta-analysis, with 4065 patients who received probiotics or synbiotics (treatment group) and 3821 patients who received standard care or placebo (control group). The pooled data from all included studies demonstrated that the treatment group has significantly reduced incidence of ventilation-associated pneumonia (VAP) (RR = 0.80; 95 % CI: 0.67-0.96; p = 0.021, I2 = 52.5 %) and sepsis (RR = 0.97; 95 % CI: 0.66-1.42; p = 0.032, I2 = 54.4 %), As well as significantly increased duration of mechanical exploitation (SMD = -0.47; 95 % CI: -0.74-0.20, p = 0.012, I2 = 63.4 %), ICU mobility (RR = 0.95; 95 % CI: 0.71-1.27; p = 0.004, I2 = 62.8 %), length of ICU stay (SMD = -0.29; 95 % CI: -0.58-0.01; p = 0.000, I2 = 82.3 %) and length of hospital stay (SMD = -0.33; 95 % CI: -0.57-0.08, p = 0.000, I2 = 74.2 %) than the control group. There were no significant differences in diarrhea, CDI, incidence of hospital acquired pneumonia, and in hospital mortality between the two groups. CONCLUSION Our meta-analysis showed that probiotic and synbiotic supplements are beneficial for critically ill patients as they significantly reduce the incidence of ventilator associated pneumonia and sepsis, as well as the duration of mechanical exploitation, length of hospital stay, length of ICU stay, and ICU mortality. However, this intervention has minimal impact on diarrhea, CDI, incidence of hospital acquired pneumonia, and in hospital mortality in critically ill patients.
-
2.
Effect of probiotics or prebiotics on thyroid function: A meta-analysis of eight randomized controlled trials.
Shu, Q, Kang, C, Li, J, Hou, Z, Xiong, M, Wang, X, Peng, H
PloS one. 2024;19(1):e0296733
-
-
-
Free full text
Plain language summary
The gut microbiome is thought to play a role in thyroid disorders, mediated by regulating iodine uptake, degradation and enterohepatic cycling of thyroid hormones, and differences in microbiome composition between patients with thyroid disorders and healthy individuals have been observed. The aim of this systematic review and meta-analysis was to evaluate the effect of pro-, pre- and synbiotics on thyroid function (thyroid stimulating hormone (TSH), free thyroxine (fT4) and free triiodothyronine (fT3) and thyroid stimulating hormone receptor antibody (TRAb)) in patients with and without thyroid disorders. 8 randomised controlled trials including 367 participants were included in the review and meta-analysis. Neither pro-, pre- nor synbiotics had a significant effect on TSH, fT4 or fT3 but pre- and probiotics lead to a significant reduction in TRAb in patients with Graves’ disease.
Abstract
BACKGROUND Microbiome-directed therapies are increasingly utilized to optimize thyroid function in both healthy individuals and those with thyroid disorders. However, recent doubts have been raised regarding the efficacy of probiotics, prebiotics, and synbiotics in improving thyroid function. This systematic review aimed to investigate the potential relationship between probiotics/prebiotics and thyroid function by analyzing the impact on thyroid hormone levels. METHODS We conducted a comprehensive systematic review and meta-analysis of randomized controlled trials that investigated the effects of probiotics, prebiotics, and synbiotics on free triiodothyronine (fT3), free thyroxine (fT4), thyroid stimulating hormone (TSH), and thyroid stimulating hormone receptor antibody (TRAb) levels. We searched for articles from PubMed, Scopus, Web of Science, and Embase up until April 1st, 2023, without any language restriction. Quantitative data analysis was performed using a random-effects model, with standardized mean difference (SMD) and 95% confidence interval as summary statistics. The methods and results were reported according to the PRISMA2020 statement. RESULTS A total of eight articles were included in this review. The meta-analysis showed no significant alterations in TSH (SMD: -0.01, 95% CI: -0.21, 0.20, P = 0.93; I2: 0.00%), fT4 (SMD: 0.04, 95% CI: -0.29, 0.21, P = 0.73; I2: 0.00%) or fT3 (SMD: 0.45, 95% CI: -0.14, 1.03, P = 0.43; I2: 78.00%), while a significant reduction in TRAb levels was observed (SMD: -0.85, 95% CI: -1.54, -0.15, P = 0.02; I2: 18.00%) following probiotics/prebiotics supplementation. No indication of publication bias was found. CONCLUSIONS Probiotics/prebiotics supplementation does not influence thyroid hormone levels, but may modestly reduce TRAb levels in patients with Graves' disease.
-
3.
Autoimmune Skin Disease - NED Infobite
Infobite 32_November 2022. Summaries of key research on autoimmune skin diseases
2024
Abstract
Psoriasis is an autoimmune inflammatory condition, affecting skin, nails and joints. This NED Infobite highlights research on the role of oral and gut microbiomes, including use of probiotics and synbiotics, key nutrients to employ and the importance of weight management. BANT's scientific NED InfoBites are designed to provide key elements of the latest research using plain language. They provide quick overviews on particular health issues and nutrition topics for a speedy introduction to the science. Visually attractive and easily shareable with clients and social media followers.
-
4.
IBS and the Microbiome - NED Infobite
Infobite 48_February 2024. Summaries of key research on irritable bowel syndrome and the microbiome
2024
Abstract
IBS is a functional intestinal disorder that can significantly affect quality of life. This NED Infobite includes studies looking at the efficacy of probiotics, synbiotics and dietary change at different life stages and the impacts on various IBS symptoms. BANT's scientific NED InfoBites are designed to provide key elements of the latest research using plain language. They provide quick overviews on particular health issues and nutrition topics for a speedy introduction to the science. Visually attractive and easily shareable with clients and social media followers.
-
5.
T2DM and the Microbiome - NED Infobite
Infobite 46_January 2024. Summaries of key research on type 2 diabetes mellitus and the microbiome
2024
Abstract
Accumulating studies have shown a strong correlation between Type 2 diabetes mellitus and the gut microbiome. This NED Infobite includes studies comparing probiotics and glucose-lowering drugs, the effects or probiotics and synbiotics on inflammation and oxidative stress in T2DM subjects, as well as the impacts of dietary interventions on gut bacterial profiles in those with dysregulated blood glucose markers. BANT's scientific NED InfoBites are designed to provide key elements of the latest research using plain language. They provide quick overviews on particular health issues and nutrition topics for a speedy introduction to the science. Visually attractive and easily shareable with clients and social media followers.
-
6.
The NED Journal - Microbiome in IBS, IBD and Health
Issue 3_April 2024 Gut Health. Peer-reviewed expert reviews and clinical insights from the Nutrition Evidence Editorial Board and Expert Review Panel
2024
Abstract
This 3rd edition of the NED Journal shines a light on the many and varied ways the microbiome interacts with human health, from different disease states and multiple life stages. In includes sections on the common diagnosis of irritable bowel syndrome, as well as the challenging autoimmune inflammatory bowel diseases. This NED Journal transforms the science to enable individualised science-based clinical decision making, with 23 peer-reviewed expert reviews. The NED Journal supports BANT members and other integrative and personalised medicine practitioners with their evidence-based practice.
-
7.
Functional response to a microbial synbiotic in the gastrointestinal system of children: a randomized clinical trial.
Tierney, BT, Versalovic, J, Fasano, A, Petrosino, JF, Chumpitazi, BP, Mayer, EA, Boetes, J, Smits, G, Parkar, SG, Voreades, N, et al
Pediatric research. 2023;93(7):2005-2013
-
-
-
Free full text
-
Plain language summary
The composition of the human gut microbiome has been identified as playing a role in regulating bowel movements in children. This includes functional constipation, which is characterised by infrequent bowel movements and associated phenotypes such as stool consistency, pain when defecating and bloating. The aim of this study was to determine the impact of a nine-strain (eight species) synbiotic (a prebiotic and defined microbial consortium) formulation (with the prebiotic comprising mixed-chain length oligosaccharides) on ameliorating constipation. This study was a multicentre, randomised, double-blind, and placebo-controlled with two parallel arms. Ninety-one healthy male/female subjects were recruited and randomly assigned to one of the two arms; treatment or placebo group. Results showed that: - compared to placebo, synbiotic use increased weekly bowel movements (WBMs) in constipated children. - there was an increased abundance of the administered probiotic species (bifidobacteria) in the treatment arm. - baseline microbial richness demonstrated potential as a predictive biomarker for response to intervention. Authors conclude that a synbiotic formulation may increase weekly WBMs in children who have low-frequency WBMs.
Abstract
BACKGROUND Oral microbial therapy has been studied as an intervention for a range of gastrointestinal disorders. Though research suggests that microbial exposure may affect the gastrointestinal system, motility, and host immunity in a pediatric population, data have been inconsistent, with most prior studies being in neither a randomized nor placebo-controlled setting. The aim of this randomized, placebo-controlled study was to evaluate the efficacy of a synbiotic on increasing weekly bowel movements (WBMs) in constipated children. METHODS Sixty-four children (3-17 years of age) were randomized to receive a synbiotic (n = 33) comprising mixed-chain length oligosaccharides and nine microbial strains, or placebo (n = 31) for 84 days. Stool microbiota was analyzed on samples collected at baseline and completion. The primary outcome was a change from baseline of WBMs in the treatment group compared to placebo. RESULTS Treatment increased (p < 0.05) the number of WBMs in children with low baseline WBMs, despite broadly distinctive baseline microbiome signatures. Sequencing revealed that low baseline microbial richness in the treatment group significantly anticipated improvements in constipation (p = 0.00074). CONCLUSIONS These findings suggest the potential for (i) multi-species-synbiotic interventions to improve digestive health in a pediatric population and (ii) bioinformatics-based methods to predict response to microbial interventions in children. IMPACT Synbiotic microbial treatment improved the number of spontaneous weekly bowel movements in children compared to placebo. Intervention induced an increased abundance of bifidobacteria in children, compared to placebo. All administered probiotic species were enriched in the gut microbiome of the intervention group compared to placebo. Baseline microbial richness demonstrated potential as a predictive biomarker for response to intervention.
-
8.
Effect of a Multispecies Synbiotic Supplementation on Body Composition, Antioxidant Status, and Gut Microbiomes in Overweight and Obese Subjects: A Randomized, Double-Blind, Placebo-Controlled Study.
Oraphruek, P, Chusak, C, Ngamukote, S, Sawaswong, V, Chanchaem, P, Payungporn, S, Suantawee, T, Adisakwattana, S
Nutrients. 2023;15(8)
-
-
-
Free full text
Plain language summary
Overweight and obesity can be attributed to a complex interplay of multiple factors that result in a dysregulated energy balance in the body, leading to an abnormal accumulation of adipose tissue or body fat. The aim of this study was to evaluate the impact of a specific multispecies synbiotic supplement, containing a blend of seven probiotic strains and fructooligosaccharides, on body composition, antioxidant levels, and gut microbiome composition in overweight and obese individuals. This study was a double-blind, placebo-controlled, randomised, parallel design study. Participants were randomly assigned to either a placebo (n = 36) or synbiotic (n = 36) group based on their age, gender, body weight, and body mass index (BMI). Results showed notable improvement in body composition (waist circumference and body fat percentage), antioxidant status, and gut microbiota in overweight and obese individuals over 12 weeks. However, at the end of the study, there were no significant differences in body weight, body mass index, waist circumference, or percentage of body fat between the synbiotic group and the placebo. Authors conclude that synbiotic consumption may be a viable strategy for promoting overall health in individuals with overweight or obesity. However, further research is needed to determine its long-term effects.
Abstract
Studies investigating the effect of multispecies synbiotic supplementation in obesity management are limited. The current study was performed to evaluate the effects of multispecies probiotics mixed with fructooligosaccharides on body composition, antioxidant status, and gut microbiome composition in overweight and obese individuals. We employed a randomized, double-blind, placebo-controlled trial design, in which 63 individuals aged 18-45 years were assigned to receive either a synbiotic supplement or placebo for 12 weeks. The synbiotic group consumed a daily dose of 37 × 109 colony-forming units (CFU) of a unique blend of seven different probiotics, along with 2 g of fructooligosaccharides, while the placebo group consumed 2 g of maltodextrin daily. Assessments were performed at baseline, week 6, and the end of the study. The results of the study indicated that synbiotic supplementation resulted in a significant reduction in waist circumference and body fat percentage compared to the baseline measurements, as observed at 12 weeks. At the end of the study, there were no significant differences observed in body weight, BMI, waist circumference, or percentage of body fat between the synbiotic group and the placebo group. An analysis of plasma antioxidant capacity revealed that synbiotic supplementation caused a significant increase in Trolox equivalent antioxidant capacity (TEAC) and a concomitant decrease in malondialdehyde (MDA) in the test group when compared to the placebo. For the gut microbiota analysis, synbiotic supplementation significantly decreased Firmicutes abundance and the Firmicutes/Bacteroidetes (F/B) ratio at week 12 as compared to the placebo group. Nevertheless, the synbiotic group did not exhibit any substantial alterations in other biochemical blood parameters compared to the placebo group. These findings suggest that multispecies synbiotic supplementation could be a beneficial strategy to improve body composition, antioxidant status, and gut microbiome composition in overweight and obese subjects.
-
9.
The Impact of Synbiotic Treatment on the Levels of Gut-Derived Uremic Toxins, Inflammation, and Gut Microbiome of Chronic Kidney Disease Patients-A Randomized Trial.
Mitrović, M, Stanković-Popović, V, Tolinački, M, Golić, N, Soković Bajić, S, Veljović, K, Nastasijević, B, Soldatović, I, Svorcan, P, Dimković, N
Journal of renal nutrition : the official journal of the Council on Renal Nutrition of the National Kidney Foundation. 2023;33(2):278-288
-
-
-
Free full text
-
Plain language summary
The gut microbiome consists of more than 100 trillion bacteria and plays an important role in normal body functioning. There is increasing evidence that gut microbiome alteration can affect multiple organ systems and also lead to numerous chronic diseases, such as chronic kidney disease (CKD). The aim of this study was to assess the efficacy and safety of synbiotic treatment in reducing the levels of gut-derived uremic toxins and serum inflammatory markers and its impact on gut microbiome, with controlled factors such as diet and antibiotic usage. This study was a single-centre, randomised, double-blind, placebo-controlled study. After a 2-week run-in period under prescribed diet, patients were randomised into 2 groups, one receiving synbiotic therapy and the other receiving placebo. Results showed that in comparison to placebo: - synbiotic treatment significantly altered levels of indoxyl sulfate [uremic toxin] and p-cresyl sulfate [uremic toxin] in the intervention arm, and - only the dynamic of total serum indoxyl sulfate was significant. Authors conclude that synbiotics are a safe and an effective therapeutic strategy that may help to decrease levels of uremic toxins and microinflammation in chronic kidney disease patients.
Abstract
OBJECTIVE Altering dysbiotic gut flora through synbiotic supplementation has recently been recognized as a potential treatment strategy to reduce the levels of gut-derived uremic toxins and decrease inflammation. Assessing its efficacy and safety has been the main goal of our randomized, double-blind, placebo-controlled study. METHODS A total of 34 nondialyzed chronic kidney disease patients, aged ≥18 years, with an estimated glomerular filtration rate between 15 and 45 mL/minute, were randomized either to an intervention group (n = 17), receiving synbiotic (Lactobacillus acidophilus, Lactobacillus casei, and Bifidobacterium lactis, 32 billion colony forming units per day plus 3.2 g of inulin), or control group (n = 17), receiving placebo during 12 weeks. The impact of treatment on the dynamic of serum levels of gut-derived uremic toxins, total serum indoxyl sulfate, p-cresyl sulfate, and trimethylamine N-oxide, was defined as the primary outcome of the study. Secondary outcomes included changes in the stool microbiome, serum interleukin-6 levels, high-sensitivity C-reactive protein, estimated glomerular filtration rate, albuminuria, diet, gastrointestinal symptom dynamics, and safety. Serum levels of uremic toxins were determined using ultraperformance liquid chromatography. The stool microbiome analysis was performed using the 16S ribosomal ribonucleic acid gene sequencing approach. RESULTS Synbiotic treatment significantly modified gut microbiome with Bifidobacteria, Lactobacillus, and Subdoligranulum genera enrichment and consequently reduced serum level of indoxyl sulfate (ΔIS -21.5% vs. 5.3%, P < .001), improved estimated glomerular filtration rate (ΔeGFR 12% vs. 8%, P = .029), and decreased level of high-sensitivity C-reactive protein (-39.5 vs. -8.5%, P < .001) in treated patients. Two patients of the intervention arm complained of increased flatulence. No other safety issues were noted. CONCLUSION Synbiotics could be available, safe, and an effective therapeutic strategy we could use in daily practice in order to decrease levels of uremic toxins and microinflammation in chronic kidney disease patients.
-
10.
Targeting the gut-lung axis by synbiotic feeding to infants in a randomized controlled trial.
Sjödin, KS, Sjödin, A, Ruszczyński, M, Kristensen, MB, Hernell, O, Szajewska, H, West, CE
BMC biology. 2023;21(1):38
-
-
-
Free full text
Plain language summary
Infants are at increased risk of infections, and respiratory tract infections are a leading cause of morbidity and mortality globally. Although the respiratory and gastrointestinal tracts are separate, they share a mucosal immune system called the “gut-lung axis.” The aim of this study was to compare the impacts of feeding prebiotic infant formula with the same prebiotic infant formula supplemented with probiotic Lactobacillus F19 (synbiotics) until 6 months of age on infant gut microbiota development in the first year of life. This study was a multicentre, double-blind randomised controlled study. Infants were randomised to control group - prebiotic formula or experimental group - synbiotic formula. Results showed additional benefit of feeding specific synbiotics to formula-fed infants over prebiotics only. In fact, synbiotic feeding led to the underrepresentation of Klebsiella [bacteria], enrichment of bifidobacteria, and slight increases in microbial degradation metabolites. Authors concluded that their findings support future clinical evaluation of synbiotic formula in the prevention of infections and associated antibiotic treatment as a primary outcome when breastfeeding is not feasible.
Abstract
BACKGROUND Formula-fed infants are at increased risk of infections. Due to the cross-talk between the mucosal systems of the gastrointestinal and respiratory tracts, adding synbiotics (prebiotics and probiotics) to infant formula may prevent infections even at distant sites. Infants that were born full term and weaned from breast milk were randomized to prebiotic formula (fructo- and galactooligosaccharides) or the same prebiotic formula with Lactobacillus paracasei ssp. paracasei F19 (synbiotics) from 1 to 6 months of age. The objective was to examine the synbiotic effects on gut microbiota development. RESULTS Fecal samples collected at ages 1, 4, 6, and 12 months were analyzed using 16S rRNA gene sequencing and a combination of untargeted gas chromatography-mass spectrometry/liquid chromatography-mass spectrometry. These analyses revealed that the synbiotic group had a lower abundance of Klebsiella, a higher abundance of Bifidobacterium breve compared to the prebiotic group, and increases in the anti-microbial metabolite d-3-phenyllactic acid. We also analyzed the fecal metagenome and antibiotic resistome in the 11 infants that had been diagnosed with lower respiratory tract infection (cases) and 11 matched controls using deep metagenomic sequencing. Cases with lower respiratory tract infection had a higher abundance of Klebsiella species and antimicrobial resistance genes related to Klebsiella pneumoniae, compared to controls. The results obtained using 16S rRNA gene amplicon and metagenomic sequencing were confirmed in silico by successful recovery of the metagenome-assembled genomes of the bacteria of interest. CONCLUSIONS This study demonstrates the additional benefit of feeding specific synbiotics to formula-fed infants over prebiotics only. Synbiotic feeding led to the underrepresentation of Klebsiella, enrichment of bifidobacteria, and increases in microbial degradation metabolites implicated in immune signaling and in the gut-lung and gut-skin axes. Our findings support future clinical evaluation of synbiotic formula in the prevention of infections and associated antibiotic treatment as a primary outcome when breastfeeding is not feasible. TRIAL REGISTRATION ClinicalTrials.gov NCT01625273 . Retrospectively registered on 21 June 2012.