scholarly journals In VivoSelection To Identify Bacterial Strains with Enhanced Ecological Performance in Synbiotic Applications

2015 ◽  
Vol 81 (7) ◽  
pp. 2455-2465 ◽  
Author(s):  
Janina A. Krumbeck ◽  
María X. Maldonado-Gomez ◽  
Inés Martínez ◽  
Steven A. Frese ◽  
Thomas E. Burkey ◽  
...  

ABSTRACTOne strategy for enhancing the establishment of probiotic bacteria in the human intestinal tract is via the parallel administration of a prebiotic, which is referred to as a synbiotic. Here we present a novel method that allows a rational selection of putative probiotic strains to be used in synbiotic applications:in vivoselection (IVS). This method consists of isolating candidate probiotic strains from fecal samples following enrichment with the respective prebiotic. To test the potential of IVS, we isolated bifidobacteria from human subjects who consumed increasing doses of galactooligosaccharides (GOS) for 9 weeks. A retrospective analysis of the fecal microbiota of one subject revealed an 8-fold enrichment inBifidobacterium adolescentisstrain IVS-1 during GOS administration. The functionality of GOS to support the establishment of IVS-1 in the gastrointestinal tract was then evaluated in rats administered the bacterial strain alone, the prebiotic alone, or the synbiotic combination. Strain-specific quantitative real-time PCR showed that the addition of GOS increasedB. adolescentisIVS-1 abundance in the distal intestine by nearly 2 logs compared to rats receiving only the probiotic. Illumina 16S rRNA sequencing not only confirmed the increased establishment of IVS-1 in the intestine but also revealed that the strain was able to outcompete the residentBifidobacteriumpopulation when provided with GOS. In conclusion, this study demonstrated that IVS can be used to successfully formulate a synergistic synbiotic that can substantially enhance the establishment and competitiveness of a putative probiotic strain in the gastrointestinal tract.

2010 ◽  
Vol 105 (6) ◽  
pp. 887-894 ◽  
Author(s):  
Łukasz Grześkowiak ◽  
Erika Isolauri ◽  
Seppo Salminen ◽  
Miguel Gueimonde

Production and manufacturing methods and the food carrier may influence the properties of probiotic strains, and have an impact on the outcome of clinical intervention studies. The aim of the present study was to establish whether the properties of a specific probiotic strain,Lactobacillus rhamnosusGG, may differ depending on the product and source of the strain. In total, fifteen differentL. rhamnosusisolates, among them fourteen labelled asL.rhamnosusGG, were isolated from specific probiotic products. The micro-organisms were phenotypically and genotypically characterised. Their adhesion properties were compared using the human intestinal mucus model, and the ability of the isolates to influence model pathogen adhesion to human colonic mucus was assessed. AllL. rhamnosusisolates used were confirmed as members of the speciesL. rhamnosus. Except the reference strain OL, allL.rhamnosusisolates showed randomly amplified polymorphic DNA, enterobacterial repetitive intergenic consensus and pulsed-field gel electrophoresis profiles identical to that ofL. rhamnosusGG (ATCC 53103). AllL.rhamnosusisolates showed similar tolerance to acid and were able to bind to human colonic mucus. However, pathogen exclusion by inhibition and competition varied significantly among the differentL. rhamnosusisolates and pathogens tested. The results suggest that different sources of the same probiotic may have significantly altered strain properties. This should be considered inin vivostudies on human subjects and also for quality control of probiotic products.


2006 ◽  
Vol 72 (4) ◽  
pp. 3042-3045 ◽  
Author(s):  
Francesca Valerio ◽  
Palmira De Bellis ◽  
Stella Lisa Lonigro ◽  
Lorenzo Morelli ◽  
Angelo Visconti ◽  
...  

ABSTRACT The ability of potentially probiotic strains of Lactobacillus plantarum and Lactobacillus paracasei to survive on artichokes for at least 90 days was shown. The anchorage of bacterial strains to artichokes improved their survival in simulated gastrointestinal digestion. L. paracasei IMPC2.1 was further used in an artichoke human feeding study involving four volunteers, and it was shown that the organism could be recovered from stools.


2006 ◽  
Vol 72 (9) ◽  
pp. 5990-5997 ◽  
Author(s):  
Tom Vanhoutte ◽  
Vicky De Preter ◽  
Evie De Brandt ◽  
Kristin Verbeke ◽  
Jean Swings ◽  
...  

ABSTRACT Diet is a major factor in maintaining a healthy human gastrointestinal tract, and this has triggered the development of functional foods containing a probiotic and/or prebiotic component intended to improve the host's health via modulation of the intestinal microbiota. In this study, a long-term placebo-controlled crossover feeding study in which each subject received several treatments was performed to monitor the effect of a prebiotic substrate (i.e., lactulose), a probiotic organism (i.e., Saccharomyces boulardii), and their synbiotic combination on the fecal microbiota of three groups of 10 healthy human subjects differing in prebiotic dose and/or intake of placebo versus synbiotic. For this purpose, denaturing gradient gel electrophoresis (DGGE) analysis of 16S rRNA gene amplicons was used to detect possible changes in the overall bacterial composition using the universal V3 primer and to detect possible changes at the subpopulation level using group-specific primers targeting the Bacteroides fragilis subgroup, the genus Bifidobacterium, the Clostridium lituseburense group (cluster XI), and the Clostridium coccoides-Eubacterium rectale group (cluster XIVa). Although these populations remained fairly stable based on DGGE profiling, one pronounced change was observed in the universal fingerprint profiles after lactulose ingestion. Band position analysis and band sequencing revealed that a band appearing or intensifying following lactulose administration could be assigned to the species Bifidobacterium adolescentis. Subsequent analysis with real-time PCR (RT-PCR) indicated a statistically significant increase (P < 0.05) in total bifidobacteria in one of the three subject groups after lactulose administration, whereas a similar but nonsignificant trend was observed in the other two groups. Combined RT-PCR results from two subject groups indicated a borderline significant increase (P = 0.074) of B. adolescentis following lactulose intake. The probiotic yeast S. boulardii did not display any detectable universal changes in the DGGE profiles, nor did it influence the bifidobacterial levels. This study highlighted the capacity of an integrated approach consisting of DGGE analysis and RT-PCR to monitor and quantify pronounced changes in the fecal microbiota of healthy subjects upon functional food administration.


2012 ◽  
Vol 3 (2) ◽  
pp. 137-144 ◽  
Author(s):  
F. Vieira de Souza ◽  
R. Roque ◽  
J.L. Silva Moreira ◽  
M. Resende de Souza ◽  
J.R. Nicoli ◽  
...  

The aim of this study was to assess the potential horizontal transfer of genetic traits for antibiotic resistance between lactobacilli isolated from the chicken gut, both in vitro and in vivo. Thirty-seven Lactobacillus spp. strains isolated from the gizzard, small and large intestines and caeca of free-range broiler chickens showed multi-drug resistance as assessed by disc diffusion assays. The minimum inhibitory concentration (MIC) for vancomycin, tetracycline, erythromycin and chloramphenicol was determined in De Man, Rogosa and Sharpe broth in a microplate assay. Almost all the lactobacilli isolates were resistant to vancomycin (except strains belonging to the Lactobacillus acidophilus group) and to tetracycline (MIC≥128 μg/ml). Only five strains were resistant to erythromycin, and six to chloramphenicol. The transfer rate in filter mating experiments performed using L. acidophilus strain 4M14E (EmR), Lactobacillus vaginalis strain 5M14E (CmR), Lactobacillus salivarius strain 5C14C (EmR), and the 4G14L and 3C14C strains of Lactobacillus reuteri (CmR) showed a frequency of approximately 1×104 cfu/ml of double-resistant transconjugants for the different combinations. The exception was the L. salivarius 5C14C (EmR) and L. vaginalis 5M14E (CmR) mating combination, which produced no transconjugants. In vivo experiments performed in gnotobiotic mice by mating L. acidophilus 4M14E (EmR) with L. reuteri 3C14C (CmR), L. reuteri 4G14L (CmR) or L. vaginalis 5M14E (CmR) resulted in transconjugants at 3.95±0.29, 3.16±0.33, and 4.55±1.52 log10 cfu/g of faeces, respectively. Taken together, these data suggest that genetic exchange may occur between native bacterial strains within the gastrointestinal tract of chickens, which might maintain a dynamic gene pool conferring antibiotic resistance upon indigenous microbiota components, even in the absence of the pathogens. This possibility must be taken into account as a complementary criterion when lactobacilli are screened for probiotic use.


2013 ◽  
Vol 58 (3) ◽  
pp. 1603-1614 ◽  
Author(s):  
Suresh Kumar Gorla ◽  
Nina N. McNair ◽  
Guangyi Yang ◽  
Song Gao ◽  
Ming Hu ◽  
...  

ABSTRACTCryptosporidiumparasites are a major cause of diarrhea and malnutrition in the developing world, a frequent cause of waterborne disease in the developed world, and a potential bioterrorism agent. Currently, available treatment is limited, andCryptosporidiumdrug discovery remains largely unsuccessful. As a result, the pharmacokinetic properties required forin vivoefficacy have not been established. We have been engaged in aCryptosporidiumdrug discovery program targeting IMP dehydrogenase (CpIMPDH). Here, we report the activity of eight potent and selective inhibitors ofCpIMPDH in the interleukin-12 (IL-12) knockout mouse model, which mimics acute human cryptosporidiosis. Two compounds displayed significant antiparasitic activity, validatingCpIMPDH as a drug target. The best compound, P131 (250 mg/kg of body weight/day), performed equivalently to paromomycin (2,000 mg/kg/day) when administered in a single dose and better than paromomycin when administered in three daily doses. One compound, A110, appeared to promoteCryptosporidiuminfection. The pharmacokinetic, uptake, and permeability properties of the eight compounds were measured. P131 had the lowest systemic distribution but accumulated to high concentrations within intestinal cells. A110 had the highest systemic distribution. These observations suggest that systemic distribution is not required, and may be a liability, forin vivoantiparasitic activity. Intriguingly, A110 caused specific alterations in fecal microbiota that were not observed with P131 or vehicle alone. Such changes may explain how A110 promotes parasitemia. Collectively, these observations suggest a blueprint for the development of anticryptosporidial therapy.


2018 ◽  
Author(s):  
Jiayi He ◽  
Lingyi Wu ◽  
Zhen Wu ◽  
Daodong Pan ◽  
Yuxing Guo ◽  
...  

AbstractProbiotic candidateL. reuteriwas screened out forin vivoexperiments based on a relatively higher gastrointestinal tolerance and moderate adhesiveness. As results shown inin-vivoexperiments, a significantly higher level of IL-12 at low-dose group was found both in females and males. Higher levels of T-lymphocytes were also observed in females compared to control group, however, males displayed a reduction expcept for CD8-positive cells in ileum. In comparison to the control group, the relative abundance of phylotypes in the phylumBacteroidetes(genus ofBacteroides,Prevotella) andFirmicutes(genus ofClostridiumIV) exihibited a reserve shift between sexes afterL. reuteriintervened. Meanwhile, the relative abundance of several taxa (Acetobacteroides,Lactobcaillus,bacillus) also differed markedly in sexes at low-dose group, together with microbiota diversity, as indicated by Shannon index.ImportanceSexual dimorphism has triggered researchers’ attention. However, the relationship between immune parameters and gut microbiota caused byLactobacillusat different dosage are not fully elucidated. In present research, the possible probiotic role ofL. reuteriDMSZ 8533 on immunomodulation and effect on fecal microbiota composition were investigated. Our findings demonstrate the importance of L. reuteri DMSZ 8533 as a potential probiotic strain with an immunomodulatory effect, which also alters the microflora composition depending on the sex of the host.


2015 ◽  
Vol 81 (17) ◽  
pp. 5993-6002 ◽  
Author(s):  
Winschau F. van Zyl ◽  
Shelly M. Deane ◽  
Leon M. T. Dicks

ABSTRACTLactic acid bacteria (LAB) are natural inhabitants of the gastrointestinal tract (GIT) of humans and animals, and some LAB species receive considerable attention due to their health benefits. Although many papers have been published on probiotic LAB, only a few reports have been published on the migration and colonization of the cells in the GIT. This is due mostly to the lack of efficient reporter systems. In this study, we report on the application of the fluorescent mCherry protein in thein vivotagging of the probiotic strainsEnterococcus mundtiiST4SA andLactobacillus plantarum423. ThemCherrygene, encoding a red fluorescent protein (RFP), was integrated into a nonfunctional region on the genome ofL. plantarum423 by homologous recombination. In the case ofE. mundtiiST4SA, themCherrygene was cloned into the pGKV223D LAB/Escherichia coliexpression vector. Expression of themCherrygene did not alter the growth rate of the two strains and had no effect on bacteriocin production. Both strains colonized the cecum and colon of mice.


1991 ◽  
Vol 74 (4) ◽  
pp. 606-619 ◽  
Author(s):  
Frank A. Rodden ◽  
Herbert Wiegandt ◽  
Bernard L. Bauer

✓ Gangliosides are complex glycolipids found on the outer surface of most cell membranes: they are particularly concentrated in tissues of the nervous system. Gangliosides form part of the immunological identity of mammalian cells and are involved in a variety of cell-surface phenomena such as cell-substrate binding and receptor functions. In tumorous tissue, the ganglioside composition is altered, sometimes in direct proportion to the degree of malignancy. The literature on the glycosphingolipid composition and immunology of intracranial tumors is reviewed. Some gangliosides induce neuritogenesis and exhibit a trophic effect on nerve cells grown in vitro. In vivo, a particular ganglioside, GM1, reduces cerebral edema and accelerates recovery from injury (traumatic and ischemic) to the peripheral and central nervous systems of laboratory animals. Preliminary clinical studies have shown that treatment with gangliosides may have corresponding effects on lesions of the human peripheral nervous system. Gangliosides have not been tested in human subjects with brain injury.


2016 ◽  
Vol 82 (9) ◽  
pp. 2669-2675 ◽  
Author(s):  
Aaron W. Miller ◽  
Kelly F. Oakeson ◽  
Colin Dale ◽  
M. Denise Dearing

ABSTRACTDiet is one of the primary drivers that sculpts the form and function of the mammalian gut microbiota. However, the enormous taxonomic and metabolic diversity held within the gut microbiota makes it difficult to isolate specific diet-microbe interactions. The objective of the current study was to elucidate interactions between the gut microbiota of the mammalian herbivoreNeotoma albigulaand dietary oxalate, a plant secondary compound (PSC) degraded exclusively by the gut microbiota. We quantified oxalate degradation inN. albigulafed increasing amounts of oxalate over time and tracked the response of the fecal microbiota using high-throughput sequencing. The amount of oxalate degradedin vivowas linearly correlated with the amount of oxalate consumed. The addition of dietary oxalate was found to impact microbial species diversity by increasing the representation of certain taxa, some of which are known to be capable of degrading oxalate (e.g.,Oxalobacterspp.). Furthermore, the relative abundances of 117 operational taxonomic units (OTU) exhibited a significant correlation with oxalate consumption. The results of this study indicate that dietary oxalate induces complex interactions within the gut microbiota that include an increase in the relative abundance of a community of bacteria that may contribute either directly or indirectly to oxalate degradation in mammalian herbivores.


mSystems ◽  
2019 ◽  
Vol 4 (4) ◽  
Author(s):  
Alexander D. Naydich ◽  
Shannon N. Nangle ◽  
Johannes J. Bues ◽  
Disha Trivedi ◽  
Nabeel Nissar ◽  
...  

ABSTRACTEngineering synthetic circuits into intestinal bacteria to sense, record, and respond toin vivosignals is a promising new approach for the diagnosis, treatment, and prevention of disease. However, because the design of disease-responsive circuits is limited by a relatively small pool of known biosensors, there is a need for expanding the capacity of engineered bacteria to sense and respond to the host environment. Here, we apply a robust genetic memory circuit inEscherichia colito identify new bacterial biosensor triggers responding in the healthy and diseased mammalian gut, which may be used to construct diagnostic or therapeutic circuits. We developed a pipeline for rapid systems-level library construction and screening, using next-generation sequencing and computational analysis, which demonstrates remarkably reliable identification of responsive biosensor triggers from pooled libraries. By testing libraries of potential triggers—each consisting of a promoter and ribosome binding site (RBS)—and using RBS variation to augment the range of trigger sensitivity, we identify and validate triggers that selectively activate our synthetic memory circuit during transit through the gut. We further identify biosensor triggers with increased response in the inflamed gut through comparative screening of one of our libraries in healthy mice and those with intestinal inflammation. Our results demonstrate the power of systems-level screening for the identification of novel biosensor triggers in the gut and provide a platform for disease-specific screening that is capable of contributing to both the understanding and clinical management of intestinal illness.IMPORTANCEThe gut is a largely obscure and inaccessible environment. The use of live, engineered probiotics to detect and respond to disease signalsin vivorepresents a new frontier in the management of gut diseases. Engineered probiotics have also shown promise as a novel mechanism for drug delivery. However, the design and construction of effective strains that respond to thein vivoenvironment is hindered by our limited understanding of bacterial behavior in the gut. Our work expands the pool of environmentally responsive synthetic circuits for the healthy and diseased gut, providing insight into host-microbe interactions and enabling future development of increasingly complex biosensors. This method also provides a framework for rapid prototyping of engineered systems and for application across bacterial strains and disease models, representing a practical step toward the construction of clinically useful synthetic tools.


Sign in / Sign up

Export Citation Format

Share Document