Displaying publications 21 - 40 of 232 in total

Abstract:
Sort:
  1. Khosravi Y, Dieye Y, Loke MF, Goh KL, Vadivelu J
    PLoS One, 2014;9(11):e112214.
    PMID: 25386948 DOI: 10.1371/journal.pone.0112214
    Helicobacter pylori (H. pylori) is a major gastric pathogen that has been associated with humans for more than 60,000 years. H. pylori causes different gastric diseases including dyspepsia, ulcers and gastric cancers. Disease development depends on several factors including the infecting H. pylori strain, environmental and host factors. Another factor that might influence H. pylori colonization and diseases is the gastric microbiota that was overlooked for long because of the belief that human stomach was a hostile environment that cannot support microbial life. Once established, H. pylori mainly resides in the gastric mucosa and interacts with the resident bacteria. How these interactions impact on H. pylori-caused diseases has been poorly studied in human. In this study, we analyzed the interactions between H. pylori and two bacteria, Streptococcus mitis and Lactobacillus fermentum that are present in the stomach of both healthy and gastric disease human patients. We have found that S. mitis produced and released one or more diffusible factors that induce growth inhibition and coccoid conversion of H. pylori cells. In contrast, both H. pylori and L. fermentum secreted factors that promote survival of S. mitis during the stationary phase of growth. Using a metabolomics approach, we identified compounds that might be responsible for the conversion of H. pylori from spiral to coccoid cells. This study provide evidences that gastric bacteria influences H. pylori physiology and therefore possibly the diseases this bacterium causes.
    Matched MeSH terms: Gastrointestinal Microbiome
  2. Yap TW, Gan HM, Lee YP, Leow AH, Azmi AN, Francois F, et al.
    PLoS One, 2016;11(3):e0151893.
    PMID: 26991500 DOI: 10.1371/journal.pone.0151893
    BACKGROUND: Accumulating evidence shows that Helicobacter pylori protects against some metabolic and immunological diseases in which the development of these diseases coincide with temporal or permanent dysbiosis. The aim of this study was to assess the effect of H. pylori eradication on the human gut microbiome.

    METHODS: As part of the currently on-going ESSAY (Eradication Study in Stable Adults/Youths) study, we collected stool samples from 17 H. pylori-positive young adult (18-30 years-old) volunteers. The same cohort was followed up 6, 12 and 18 months-post H. pylori eradication. The impact of H. pylori on the human gut microbiome pre- and post-eradication was investigated using high throughput 16S rRNA gene (V3-V4 region) sequencing using the Illumina Miseq followed by data analysis using Qiime pipeline.

    RESULTS: We compared the composition and diversity of bacterial communities in the fecal microbiome of the H. pylori-positive volunteers, before and after H. pylori eradication therapy. The 16S rRNA gene was sequenced at an average of 150,000-170,000 reads/sample. The microbial diversity were similar pre- and post-H. pylori eradication with no significant differences in richness and evenness of bacterial species. Despite that the general profile of the gut microbiome was similar pre- and post-eradication, some changes in the bacterial communities at the phylum and genus levels were notable, particularly the decrease in relative abundance of Bacterioidetes and corresponding increase in Firmicutes after H. pylori eradication. The significant increase of short-chain fatty acids (SCFA)-producing bacteria genera could also be associated with increased risk of metabolic disorders.

    CONCLUSIONS: Our preliminary stool metagenomics study shows that eradication of H. pylori caused perturbation of the gut microbiome and may indirectly affect the health of human. Clinicians should be aware of the effect of broad spectrum antibiotics used in H. pylori eradication regimen and be cautious in the clinical management of H. pylori infection, particularly in immunocompromised patients.

    Matched MeSH terms: Gastrointestinal Microbiome/drug effects*
  3. Khosravi Y, Bunte RM, Chiow KH, Tan TL, Wong WY, Poh QH, et al.
    Gut Microbes, 2016;7(1):48-53.
    PMID: 26939851 DOI: 10.1080/19490976.2015.1119990
    Helicobacter pylori have been shown to influence physiological regulation of metabolic hormones involved in food intake, energy expenditure and body mass. It has been proposed that inducing H. pylori-induced gastric atrophy damages hormone-producing endocrine cells localized in gastric mucosal layers and therefore alter their concentrations. In a recent study, we provided additional proof in mice under controlled conditions that H. pylori and gut microbiota indeed affects circulating metabolic gut hormones and energy homeostasis. In this addendum, we presented data from follow-up investigations that demonstrated H. pylori and gut microbiota-associated modulation of metabolic gut hormones was independent and precedes H. pylori-induced histopathological changes in the gut of H. pylori-infected mice. Thus, H. pylori-associated argumentation of energy homeostasis is not caused by injury to endocrine cells in gastric mucosa.
    Matched MeSH terms: Gastrointestinal Microbiome*
  4. Péré-Védrenne C, Flahou B, Loke MF, Ménard A, Vadivelu J
    Helicobacter, 2017 Sep;22 Suppl 1.
    PMID: 28891140 DOI: 10.1111/hel.12407
    The current article is a review of the most important and relevant literature published in 2016 and early 2017 on non-Helicobacter pylori Helicobacter infections in humans and animals, as well as interactions between H. pylori and the microbiota of the stomach and other organs. Some putative new Helicobacter species were identified in sea otters, wild boars, dogs, and mice. Many cases of Helicobacter fennelliae and Helicobacter cinaedi infection have been reported in humans, mostly in immunocompromised patients. Mouse models have been used frequently as a model to investigate human Helicobacter infection, although some studies have investigated the pathogenesis of Helicobacters in their natural host, as was the case for Helicobacter suis infection in pigs. Our understanding of both the gastric and gut microbiome has made progress and, in addition, interactions between H. pylori and the microbiome were demonstrated to go beyond the stomach. Some new approaches of preventing Helicobacter infection or its related pathologies were investigated and, in this respect, the probiotic properties of Saccharomyces, Lactobacillus and Bifidobacterium spp. were confirmed.
    Matched MeSH terms: Gastrointestinal Microbiome*
  5. Liu YW, Liong MT, Tsai YC
    J Microbiol, 2018 Sep;56(9):601-613.
    PMID: 30141154 DOI: 10.1007/s12275-018-8079-2
    Lactobacillus plantarum is a non-gas-producing lactic acid bacterium that is generally regarded as safe (GRAS) with Qualified Presumption of Safety (QPS) status. Although traditionally used for dairy, meat and vegetable fermentation, L. plantarum is gaining increasing significance as a probiotic. With the newly acclaimed gut-heart-brain axis, strains of L. plantarum have proven to be a valuable species for the development of probiotics, with various beneficial effects on gut health, metabolic disorders and brain health. In this review, the classification and taxonomy, and the relation of these with safety aspects are introduced. Characteristics of L. plantarum to fulfill the criteria as a probiotic are discussed. Emphasis are also given to the beneficial functions of L. plantarum in gut disorders such as inflammatory bowel diseases, metabolic syndromes, dyslipidemia, hypercholesteromia, obesity, and diabetes, and brain health aspects involving psychological disorders.
    Matched MeSH terms: Gastrointestinal Microbiome
  6. Pitisuttithum P, Chan WK, Piyachaturawat P, Imajo K, Nakajima A, Seki Y, et al.
    BMC Gastroenterol, 2020 Apr 06;20(1):88.
    PMID: 32252638 DOI: 10.1186/s12876-020-01240-z
    BACKGROUND: The Gut and Obesity in Asia (GOASIA) Workgroup was formed to study obesity and gastrointestinal diseases in the Asia Pacific region. We aimed to 1) compare the characteristics of elderly (i.e. age ≥ 60) vs. non-elderly patients with biopsy-proven nonalcoholic fatty liver disease (NAFLD); 2) identify predictors of advanced fibrosis in elderly patients with NAFLD; and 3) assess the performance of non-invasive fibrosis scores in the prediction of advance fibrosis in the elderly population.

    METHODS: We abstracted the data of 1008 patients with NAFLD from nine centers across eight countries. Characteristics of elderly and non-elderly patients with NAFLD were compared using 1:3 sex-matched analysis.

    RESULTS: Of the 1008 patients, 175 were elderly [age 64 (62-67) years], who were matched with 525 non-elderly patients [46 (36-54) years]. Elderly patients were more likely to have advanced fibrosis (35.4% vs. 13.3%; p 

    Matched MeSH terms: Gastrointestinal Microbiome
  7. Hartini Yusof, Mohamad Shafiq Aazmi, Teh Lay Kek, Mohd Zaki Salleh, Ili Ng Abdullah, Aminuddin Ahmad, et al.
    MyJurnal
    Obesity is a growing epidemic due to an accelerated phase of industrialization and urbanization with the overfed people
    now outnumbered the underfed. It is the major public health problem with a lot of research interest as it is associated
    with many complicated chronic disorders such as type-2 diabetes, cardiovascular diseases (CVD) and cancers. A global
    estimation of 2.8 million deaths per year is due to obesity and there are tremendous on-going efforts to identify hosts
    and environmental factors that infl uence the cause and pathogenesis of obesity. Concerted efforts from different research
    groups had successfully shown that obese subjects have altered composition of gut microbiota and transplantation of this
    microbiota infl uences body weight in the germ-free recipient mice. The advancement of technology had made possible
    the study of gut microbiota which was unculturable for better understanding of their impact to human health. Rapid
    deep sequencing of DNA at reasonable cost through various options of platforms followed by data analysis using robust
    bioinformatic tools are an important way of analysing the gut microbiome. Here we review the role of gut microbiota
    which modulates host’s metabolic functions and gene expression, facilitating the extraction and storage of energy from the
    ingested dietary substances and leading to body-weight gain. We will discuss on the different techniques used, focusing
    on the high-defi nition technologies for the determination of the composition, function and ecology of gut microbiota. This
    allows the appropriate selection of platform which becomes the key for success of subsequent research.
    Matched MeSH terms: Gastrointestinal Microbiome
  8. Williams AR, Krych L, Fauzan Ahmad H, Nejsum P, Skovgaard K, Nielsen DS, et al.
    PLoS One, 2017;12(10):e0186546.
    PMID: 29028844 DOI: 10.1371/journal.pone.0186546
    Polyphenols are a class of bioactive plant secondary metabolites that are thought to have beneficial effects on gut health, such as modulation of mucosal immune and inflammatory responses and regulation of parasite burdens. Here, we examined the interactions between a polyphenol-rich diet supplement and infection with the enteric nematode Ascaris suum in pigs. Pigs were fed either a basal diet or the same diet supplemented with grape pomace (GP), an industrial by-product rich in polyphenols such as oligomeric proanthocyanidins. Half of the animals in each group were then inoculated with A. suum for 14 days to assess parasite establishment, acquisition of local and systemic immune responses and effects on the gut microbiome. Despite in vitro anthelmintic activity of GP-extracts, numbers of parasite larvae in the intestine were not altered by GP-supplementation. However, the bioactive diet significantly increased numbers of eosinophils induced by A. suum infection in the duodenum, jejunum and ileum, and modulated gene expression in the jejunal mucosa of infected pigs. Both GP-supplementation and A. suum infection induced significant and apparently similar changes in the composition of the prokaryotic gut microbiota, and both also decreased concentrations of isobutyric and isovaleric acid (branched-chain short chain fatty acids) in the colon. Our results demonstrate that while a polyphenol-enriched diet in pigs may not directly influence A. suum establishment, it significantly modulates the subsequent host response to helminth infection. Our results suggest an influence of diet on immune function which may potentially be exploited to enhance immunity to helminths.
    Matched MeSH terms: Gastrointestinal Microbiome/drug effects*
  9. Yap PSX, Chong CW, Ahmad Kamar A, Yap IKS, Choo YM, Lai NM, et al.
    Sci Rep, 2021 01 14;11(1):1353.
    PMID: 33446779 DOI: 10.1038/s41598-020-80278-1
    Emerging evidence has shown a link between the perturbations and development of the gut microbiota in infants with their immediate and long-term health. To better understand the assembly of the gut microbiota in preterm infants, faecal samples were longitudinally collected from the preterm (n = 19) and term (n = 20) infants from birth until month 12. 16S rRNA gene sequencing (n = 141) and metabolomics profiling (n = 141) using nuclear magnetic resonance spectroscopy identified significant differences between groups in various time points. A panel of amino acid metabolites and central metabolism intermediates significantly correlated with the relative abundances of 8 species of bacteria were identified in the preterm group. In contrast, faecal metabolites of term infants had significantly higher levels of metabolites which are commonly found in milk such as fucose and β-hydroxybutyrate. We demonstrated that the early-life factors such as gestational age, birth weight and NICU exposures, exerted a sustained effect to the dynamics of gut microbial composition and metabolism of the neonates up to one year of age. Thus, our findings suggest that intervention at this early time could provide 'metabolic rescue' to preterm infants from aberrant initial gut microbial colonisation and succession.
    Matched MeSH terms: Gastrointestinal Microbiome*
  10. Tan SC, Chong CW, Yap IKS, Thong KL, Teh CSJ
    Sci Rep, 2020 Jun 02;10(1):8997.
    PMID: 32488118 DOI: 10.1038/s41598-020-65891-4
    The gastrointestinal tract of humans and swine consist of a wide range of bacteria which interact with hosts metabolism. Due to the differences in co-evolution and co-adaptation, a large fraction of the gut microbiome is host-specific. In this study, we evaluated the effect of close human-animal interaction to the faecal metagenome and metabonome of swine, farmer and human control. Three distinct clusters were observed based on T-RFLP-derived faecal microbial composition. However, 16S-inferred faecal microbiota and metabolic profiles showed that only human control was significantly different from the swine (P 
    Matched MeSH terms: Gastrointestinal Microbiome/genetics; Gastrointestinal Microbiome/physiology*
  11. Pathmanathan SG, Lawley B, McConnell M, Baird MA, Tannock GW
    Anaerobe, 2020 Feb;61:102112.
    PMID: 31629806 DOI: 10.1016/j.anaerobe.2019.102112
    Immuno-modulatory effects of infant gut bacteria were tested on poly(I:C) stimulated HT-29 intestinal epithelial cells. Blautia producta, Bacteroides vulgatus, Bacteroides fragilis and Bacteroides thetaiotaomicron decreased transcription of poly(I:C)-induced inflammatory genes. Modulation of basal level and poly(I:C)-induced IL-8 secretion varied between bacterial species, and between heat treated and non-heat treated bacterial cells.
    Matched MeSH terms: Gastrointestinal Microbiome*
  12. Mi Y, Chin YX, Cao WX, Chang YG, Lim PE, Xue CH, et al.
    Int J Biol Macromol, 2020 Mar 15;147:284-294.
    PMID: 31926226 DOI: 10.1016/j.ijbiomac.2020.01.072
    Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, has gradually emerged as a public health challenge worldwide. Carrageenan is a popular food additive that has been in use for decades. However, controversy exists regarding to the safety of carrageenan due to its exacerbation of colitis in experimental models. In this study, we studied the effects of vehicle and host intestinal microflora on carrageenan inflammatory properties in C57BL/6 J mice. We found that in high-fat diet model, native carrageenan in drinking water increased the disease activity index (DAI), myeloperoxidase (MPO) activity and the mRNA expression of TLR4 in colon, whereas carrageenan-supplemented diet has no visible effects. However, no signs of colitis were observed under low-fat diet regardless of the mode of vehicle used. Moreover, we discovered that carrageenan-induced colitis in high-fat diet model was robustly correlated with changes in the composition of gut microbiota, specifically Alistipes finegoldii and Bacteroides acidifaciens. Hence, we propose that the inflammatory property of carrageenan is influenced greatly by its intake form via modification of host intestinal microecology.
    Matched MeSH terms: Gastrointestinal Microbiome*
  13. Chin YX, Mi Y, Cao WX, Lim PE, Xue CH, Tang QJ
    Nutrients, 2019 May 21;11(5).
    PMID: 31117266 DOI: 10.3390/nu11051133
    Kappaphycus is a commercially important edible red alga widely cultivated for carrageenan production. Here, we aimed to investigate the anti-obesity mechanism of Kappaphycusalvarezii by comparing the effects of whole seaweed (T), extracted native κ-carrageenan (CGN), and the leftover fraction sans-carrageenan (SCGN) supplementations (5%, w/w) on diet-induced obese C57BL/6J mice. A high-fat diet induced both a raised body fat percentage and serum cholesterol level, increased adipocytes size, abnormal levels of adipocytokines, and promoted gut dysbiosis. Our results showed that, overall, both CGN and SCGN were more effective in reversing obesity and related metabolic syndromes to normal levels than T. Furthermore, these findings suggested that CGN- and SCGN-modulated gut dysbiosis induced by a high-fat diet, which may play an influencing role in adiponectin dysregulation. Our data also showed some evidence that CGN and SCGN have distinct effects on selected genes involved in lipid metabolism. In conclusion, both κ-carrageenan and SCGN have novel anti-obesity potential with possible different mechanisms of action.
    Matched MeSH terms: Gastrointestinal Microbiome
  14. Ismail IH, Boyle RJ, Licciardi PV, Oppedisano F, Lahtinen S, Robins-Browne RM, et al.
    Pediatr Allergy Immunol, 2016 12;27(8):838-846.
    PMID: 27590263 DOI: 10.1111/pai.12646
    BACKGROUND: An altered compositional signature and reduced diversity of early gut microbiota are linked to development of allergic disease. We investigated the relationship between dominant Bifidobacterium species during the early post-natal period and subsequent development of allergic disease in the first year of life.

    METHODS: Faecal samples were collected at age 1 week, 1 month and 3 months from 117 infants at high risk of allergic disease. Bifidobacterium species were analysed by quantitative PCR and terminal restriction fragment length polymorphism. Infants were examined at 3, 6 and 12 months, and skin prick test was performed at 12 months. Eczema was diagnosed according to the UK Working Party criteria.

    RESULTS: The presence of B. catenulatum at 3 months was associated with a higher risk of developing eczema (ORadj = 4.5; 95% CI: 1.56-13.05, padj = 0.005). Infants colonized with B. breve at 1 week (ORadj = 0.29; 95% CI: 0.09-0.95, padj = 0.04) and 3 months (ORadj = 0.15; 95% CI: 0.05-0.44, padj = 0.00001) had a reduced risk of developing eczema. Furthermore, the presence of B. breve at 3 months was associated with a lower risk of atopic sensitization at 12 months (ORadj = 0.38; 95% CI: 0.15-0.98, padj = 0.05). B. breve colonization patterns were influenced by maternal allergic status, household pets and number of siblings.

    CONCLUSIONS: Temporal variations in Bifidobacterium colonization patterns early in life are associated with later development of eczema and/or atopic sensitization in infants at high risk of allergic disease. Modulation of the early microbiota may provide a means to prevent eczema in high-risk infants.

    Matched MeSH terms: Gastrointestinal Microbiome/immunology
  15. Huët MAL, Wong LW, Goh CBS, Ong KS, Dwiyanto J, Reidpath D, et al.
    Braz J Microbiol, 2020 Dec;51(4):2067-2075.
    PMID: 32572838 DOI: 10.1007/s42770-020-00323-z
    Species of fungi belonging to the order Mucorales can be found everywhere in the environment. Gilbertella persicaria, which belongs to this order, have often been isolated from fruits and in water systems. However, there has been no report of isolation of this fungus from human samples. During a gut mycobiome study, from the Segamat community, Gilbertella persicaria was isolated from a human fecal sample and was characterized through a series of morphological assessment, biochemical tests, and molecular techniques. The isolate produced a white velvety surface that turned grayish after 24 h. Although no biofilm production was observed, the results indicated that the isolate could form calcium oxalate crystals, produced urease, and was resistant to low pH. The isolate was sensitive to amphotericin but resistant to voriconazole and itraconazole. The features of this fungus that could help in its survival in the human gut are also discussed.
    Matched MeSH terms: Gastrointestinal Microbiome*
  16. Ford AC, Mahadeva S, Carbone MF, Lacy BE, Talley NJ
    Lancet, 2020 11 21;396(10263):1689-1702.
    PMID: 33049222 DOI: 10.1016/S0140-6736(20)30469-4
    Dyspepsia is a complex of symptoms referable to the gastroduodenal region of the gastrointestinal tract and includes epigastric pain or burning, postprandial fullness, or early satiety. Approximately 80% of individuals with dyspepsia have no structural explanation for their symptoms and have functional dyspepsia. Functional dyspepsia affects up to 16% of otherwise healthy individuals in the general population. Risk factors include psychological comorbidity, acute gastroenteritis, female sex, smoking, use of non-steroidal anti-inflammatory drugs, and Helicobacter pylori infection. The pathophysiology remains incompletely understood, but it is probably related to disordered communication between the gut and the brain, leading to motility disturbances, visceral hypersensitivity, and alterations in gastrointestinal microbiota, mucosal and immune function, and CNS processing. Although technically a normal endoscopy is required to diagnose functional dyspepsia, the utility of endoscopy in all patients with typical symptoms is minimal; its use should be restricted to people aged 55 years and older, or to those with concerning features, such as weight loss or vomiting. As a result of our incomplete understanding of its pathophysiology, functional dyspepsia is difficult to treat and, in most patients, the condition is chronic and the natural history is one of fluctuating symptoms. Eradication therapy should be offered to patients with functional dyspepsia who test positive for Helicobacter pylori. Other therapies with evidence of effectiveness include proton pump inhibitors, histamine-2 receptor antagonists, prokinetics, and central neuromodulators. The role of psychological therapies is uncertain. As our understanding of the pathophysiology of functional dyspepsia increases, it is probable that the next decade will see the emergence of truly disease-modifying therapies for the first time.
    Matched MeSH terms: Gastrointestinal Microbiome
  17. Lubomski M, Tan AH, Lim SY, Holmes AJ, Davis RL, Sue CM
    J Neurol, 2020 Sep;267(9):2507-2523.
    PMID: 31041582 DOI: 10.1007/s00415-019-09320-1
    Recently, there has been a surge in awareness of the gastrointestinal microbiome (GM) and its role in health and disease. Of particular note is an association between the GM and Parkinson's disease (PD) and the realisation that the GM can act via a complex bidirectional communication between the gut and the brain. Compelling evidence suggests that a shift in GM composition may play an important role in the pathogenesis of PD by facilitating the characteristic ascending neurodegenerative spread of α-synuclein aggregates from the enteric nervous system to the brain. Here, we review evidence linking GM changes with PD, highlighting mechanisms supportive of pathological α-synuclein spread and intestinal inflammation in PD. We summarise existing patterns and correlations seen in clinical studies of the GM in PD, together with the impacts of non-motor symptoms, medications, lifestyle, diet and ageing on the GM. Roles of GM modulating therapies including probiotics and faecal microbiota transplantation are discussed. Encouragingly, alterations in the GM have repeatedly been observed in PD, supporting a biological link and highlighting it as a potential therapeutic target.
    Matched MeSH terms: Gastrointestinal Microbiome
  18. Li L, Su Y, Li F, Wang Y, Ma Z, Li Z, et al.
    BMC Microbiol, 2020 03 24;20(1):65.
    PMID: 32209070 DOI: 10.1186/s12866-020-01754-2
    BACKGROUND: It has recently been reported that intermittent fasting shapes the gut microbiota to benefit health, but this effect may be influenced to the exact fasting protocols. The purpose of this study was to assess the effects of different daily fasting hours on shaping the gut microbiota in mice. Healthy C57BL/6 J male mice were subjected to 12, 16 or 20 h fasting per day for 1 month, and then fed ad libitum for an extended month. Gut microbiota was analyzed by 16S rRNA gene-based sequencing and food intake was recorded as well.

    RESULTS: We found that cumulative food intake was not changed in the group with 12 h daily fasting, but significantly decreased in the 16 and 20 h fasting groups. The composition of gut microbiota was altered by all these types of intermittent fasting. At genus level, 16 h fasting led to increased level of Akkermansia and decreased level of Alistipes, but these effects disappeared after the cessation of fasting. No taxonomic differences were identified in the other two groups.

    CONCLUSIONS: These data indicated that intermittent fasting shapes gut microbiota in healthy mice, and the length of daily fasting interval may influence the outcome of intermittent fasting.

    Matched MeSH terms: Gastrointestinal Microbiome
  19. Hamid NH, Daud HM, Kayansamruaj P, Hassim HA, Mohd Yusoff MS, Abu Bakar SN, et al.
    Fish Shellfish Immunol, 2021 Jul;114:1-19.
    PMID: 33872754 DOI: 10.1016/j.fsi.2021.04.012
    This study evaluated the short- and long-term effects of dietary supplementation with Enterococcus hirae strain UPM02 on the growth performance, immunity, and disease resistance of hybrid catfish (Clarias gariepinus × Clarias macrocephalus) against Aeromonas hydrophila infection. In the long-term trial, fingerling fish were fed diets containing 0 (control), 2 × 105, or 2 × 107 CFU/g E. hirae UPM02 for 120 days. Administration of E. hirae UPM02 had significant effects on the specific growth rate (SGR), feed utilization efficiency, body indices (P 
    Matched MeSH terms: Gastrointestinal Microbiome
Filters
Contact Us

Please provide feedback to Administrator (afdal@afpm.org.my)

External Links