ArticlePDF Available
Editorial
Recent Advances and Future Perspective in
Microbiota and Probiotics
Haruki Kitazawa,1Susana Alvarez,2Alexander Suvorov,3Vyacheslav Melnikov,4
Julio Villena,2and Borja Sánchez5
1Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science,
Tohoku University, Sendai 981-8555, Japan
2Laboratory of Immunobiotechnology, CERELA-CONOCET, 4000 Tucuman, Argentina
3Department of Molecular Microbiology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia
4International Science and Technology Center (ISTC), Moscow 127374, Russia
5Department of Microbiology and Biochemistry of Dairy Products, IPLA-CSIC, 32004 Ourense, Spain
Correspondence should be addressed to Haruki Kitazawa; haruki@bios.tohoku.ac.jp and Julio Villena; jcvillena@cerela.org.ar
Received 10 February 2015; Accepted 10 February 2015
Copyright © 2015 Haruki Kitazawa et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Recent studies have highlighted the critical role of intestinal
microbes on health. e various bacterial communities in
the gut have many functions including metabolic, barrier
eect, and trophic and immunological functions. e gut
microbiota therefore performs a large number of important
roles that dene the physiology of the host. Advances in the
understanding of microbiota interaction with the host have
irrevocably altered the view of mammalian metabolism and
gut biology. As stated by Kinross et al. [1], human gut biology
and metabolism is not only inuenced by the human genome,
butacoregutmicrobiomeexistwithinthehumangut,atleast
at a genomic or metabolic level, and this is fundamental to the
maintenance of health, the development of disease and human
metabolic processes.
e understanding of the gut microbiota and its activities
is essential for the generation of future personalized health-
care strategies. In this regard, there is a growing body of
evidence to support the potential use of selected bacterial
strains in the prevention and treatment of various human
and animal diseases. Numerous studies including dierent
probiotic strains have been performed in humans and animal
models to investigate their benecial eects [24]. Overall
there is encouraging evidence that specic probiotic strains
are valuable in the prevention and treatment of dierent dis-
eases and their successful application is related to the better
understanding of the cellular and molecular mechanisms of
probiotic action.
Studies have provided insight into the mechanisms by
which probiotic bacteria are able to regulate the colonization
and eradication of pathogens in the gut, including competi-
tion for limited nutrients in the intestine and modulation of
the mucosal immune system [4,5]. In addition, it has been
well established that probiotics are an important prophylactic
or therapeutic strategy for many mucosal and nonmucosal
immune-related conditions, such as inammatory bowel
diseases (IBDs), celiac disease, metabolic syndrome, and
diabetes [6]. In this regard, results from experiments in
animal models of IBDs overwhelmingly support a causal role
of the microbiota in these diseases. In this special issue, A.
Hevia et al. explored the levels of antibodies raised against
extracellular proteins produced by dierent food bacteria
from the genera Bidobacterium and Lactobacillus,inhealthy
individuals and IBDs patients. e authors found that IBD
patients appeared to have dierent immune responses to food
bacteria. e work could set the basis for developing systems
for early detection of IBD, based on the association of high
levels of antibodies developed against extracellular proteins
from lactic acid bacteria. On the other hand, data from
animal models of colitis have indicated that specic probiotic
Lactobacillus and Bidobacterium strains could prevent and
treat intestinal inammation. e study of R. Chauhan et
al. was undertaken to evaluate the antioxidative potentials
of Lactobacillus fermentum Lf1, a promising indigenous
probiotic Lactobacillus strain, to manage oxidative stress and
Hindawi Publishing Corporation
BioMed Research International
Volume 2015, Article ID 275631, 2 pages
http://dx.doi.org/10.1155/2015/275631
2BioMed Research International
modulation of lipid peroxidation in vitro and in vivo. e
authorsshowedinintestinalepithelialcellsculturesandin
a DSS colitis mouse model that the probiotic strain Lf1 was
able to increase the expression of antioxidative enzymes and
reduce colitis, indicating that probiotics could be explored
as a new strategy for IBD management through activation
of the antioxidant enzyme system. In addition, J. Breton
et al. studied the direct immune responses to alimentary
bers in murine model of experimental colitis. e study
strongly suggests that intrinsic, nonprebiotic-driven eects
of selected oligosaccharide and polysaccharide bers can
inuence immunomodulatory functions and that these bers
could be used to enhance dietary interventions for the
treatment of inammatory disorders such as IBD and other
diseases with an immune component. e use of bers
alone or in combination with selected probiotics (symbiotic
preparations) could be considered as a promise intervention
tool for inammatory diseases.
Fermented dairy foods result from the metabolic activ-
ity of complex and heterogeneous bacterial communities;
then these dairy fermented products contain a complex,
live microbial consortium mostly represented by lactic acid
bacteria, which enter the human body and reach the gas-
trointestinal tract, where they can transiently interact with
the resident gut microora of the host. e interplay between
these two microbial communities can greatly contribute
to human health. However, evaluation of the interaction
between these microbial populations has the obstacle of the
lack of simple model organisms suitable for these studies.
In this special issue, the work of E. Zanni et al. tested the
nematode Caenorhabditis elegans as a simple animal model
to evaluate the eects of a complex food-derived microbiota
on well characterized metabolic pathways. Authors provide
evidence that feeding C. elegans with a lactic acid bacteria
consortium inuences longevity, larval development, fertility,
lipid accumulation, and gene expression related to obesity in
this model organism, as supported by transcriptional analysis
of some genes involved in fat metabolism. e work clearly
demonstrates the applicability of C. elegans model in the eld
of host-microbiota interaction.
Currently, the use of genetically modied commensal
and lactic acid bacteria to deliver compounds of health
interest is gaining importance as an extension of the probiotic
concept [7]. Most of the works using recombinant friendly
bacteria are mainly related to vaccines. Several antigens from
pathogens have been expressed in genetically engineered
lactic acid bacteria and these recombinant bacteria have
been successfully used for inducing protective immunity
in animal models. However, recombinant friendly bacteria
canbealsousedforexploringnoveleectivestrategies
to deliver therapeutic molecules to the mucosal tissues in
order to avoid degradation. In this special issue P. Kumar
et al. studied the potential benecial eects of E. coli 16
expressing Vitreoscilla hemoglobin gene, associated with bac-
terial respiration under microaerobic condition, on carbon
tetrachloride induced toxicity in rats. e work showed
that the presence of Vitreoscil la hemoglobin gene improved
the growth and intestinal tract colonization of E. coli 16.
Moreover, recombinant E. coli 16 enhanced catalase activity
in rats, prevented absorption of carbon tetrachloride in the
intestine, and ameliorated hepatotoxicity. On the other hand,
S. Shigemori et al. developed a 𝛽-lactoglobulin-secreting
Lactococcus lactis and demonstrated that this recombinant
strain is able to inhibit dipeptidyl peptidase-IV (DPP-IV)
activity. DPP-IV is a serine protease and its endogenous phys-
iological substrates are incretins. e incretins are primarily
glucose-dependent insulinotropic polypeptide and glucagon-
like peptide-1, which drive insulin secretion in pancreatic
𝛽cells and suppress pancreatic glucagon production. us,
DPP-IVinhibitorsareusedinthemanagementoftype2
diabetes mellitus.
In summary, this special issue covers a range of diverse
topics related to microbiota and probiotic in gut health
and disease, thus highlighting the potential benecial role
friendly bacteria in human health. We hope the papers pub-
lished will serve to further highlight the potential application
of probiotics for the prevention and treatment of gut diseases,
as well as stimulating further research into the cellular and
molecular mechanisms of probiotic actions.
Haruki Kitazawa
Susana Alvarez
Alexander Suvorov
Vyac h eslav Melni k ov
Julio Villena
Borja S´
anchez
References
[1] J. M. Kinross, A. W. Darzi, and J. K. Nicholson, “Gut
microbiome-host interactions in health and disease, Genome
Medicine,vol.3,no.3,article14,2011.
[2] S. Lebeer, J. Vanderleyden, and S. C. J. de Keersmaecker,
“Host interactions of probiotic bacterial surface molecules:
comparison with commensals and pathogens, Nature Reviews
Microbiology,vol.8,no.3,pp.171184,2010.
[3] P. A. Bron, P. van Baarlen, and M. Kleerebezem, “Emerging
molecular insights into the interaction between probiotics and
the host intestinal mucosa, Nature Reviews Microbiology,vol.
10,no.1,pp.6678,2012.
[4] H. Kitazawa, J. Villena, and A. Susana, Probiotics: Immunobiotics
and Immunogenics,CRCPress,2013.
[5] N. Kamada, G. Y. Chen, N. Inohara, and G. N´
u˜
nez, “Control
of pathogens and pathobionts by the gut microbiota, Nature
Immunology,vol.14,no.7,pp.685690,2013.
[6] I. I. Ivanov and K. Honda, “Intestinal commensal microbes as
immune modulators, Cell Host & Microbe,vol.12,no.4,pp.
496–508, 2012.
[7] R. Mart´
ın,S.Miquel,J.Ulmer,N.Kechaou,P.Langella,and
L. G. Berm´
udez-Humar´
an, “Role of commensal and probiotic
bacteria in human health: a focus on inammatory bowel
disease, MicrobialCellFactories,vol.12,no.1,article71,2013.
... Pro and Syn perform an important role in the health of the host by maintaining the intestinal flora population, producing bacteriostatic agents, regulating the intestinal structure and function, and preventing the normal flora from turning into pathogenic bacteria. [8][9][10] Due to the clinical properties of Pro and Syn, they have been well used in the prevention of postoperative abdominal infections. Although more studies have reported the effect of Pro and Syn on post-colorectal resection infections, there are discrepancies in the results. ...
Article
Full-text available
Wound infection is a serious complication that impacts the prognosis of patients after colorectal surgery (CS). Probiotics and synbiotics (Pro and Syn) are live bacteria that produce bacteriostatic agents in the intestinal system and have a positive effect on postoperative wound infections. The purpose of this study was to evaluate the effect of Pro and Syn on complications of wound infection after CS. In November 2023, we searched relevant clinical trial reports from Pubmed, Cochrane Library, and Embase databases and screened the retrieved reports, extracted data, and finally analysed the data by using RevMan 5.3. A total of 12 studies with 1567 patients were included in the study. Pro and Syn significantly reduced total infection (OR, 0.44; 95% CI, 0.35, 0.56; p < 0.00001), surgical incision site infection (SSI) (OR, 0.61; 95% CI, 0.45, 0.81; p = 0.002), pneumonia (OR, 0.43; 95% CI, 0.25, 0.72; p = 0.001), urinary tract infection (OR, 0.28; 95% CI, 0.14, 0.56; p = 0.0003), and Pro and Syn did not reduce anastomotic leakage after colorectal surgery (OR, 0.84; 95% CI, 0.50, 1.41; p = 0.51). Pro and Syn can reduce postoperative wound infections in patients with colorectal cancer, which benefits patients' postoperative recovery.
... E. durans LAB18S has been characterized as a potential probiotic bacterium [12,28]. Many studies show the ability of probiotic strains to change the composition and control microbial population of the intestinal microbiota [26,29,30]. Moreover, synbiotic mixture of FOS, GOS and probiotics promoted the diversity and positively influenced the development of the gut microbiota in neonates [31,32]. ...
Article
Full-text available
The gut microbiome plays a critical role to all animals and humans health. Methods based on ex vivo cultures are time and cost-effective solutions for rapid evaluation of probiotic effects on microbiomes. In this study, we assessed whether the protein secretome from the potential probiotic Enterococcus durans LAB18S grown on fructoligosaccharides (FOS) and galactoligosaccharides (GOS) had specific effects on ex vivo cultured intestinal microbiome obtained from a healthy individual. Metaproteomics was used to evaluate changes in microbial communities of the human intestinal microbiome. Hierarchical clustering analysis revealed 654 differentially abundant proteins from the metaproteome samples, showing that gut microbial protein expression varied on the presence of different E. durans secretomes. Increased amount of Bacteroidetes phylum was observed in treatments with secretomes from E. durans cultures on FOS, GOS and albumin, resulting in a decrease of the Firmicutes to Bacteroidetes (F/B) ratio. The most functionally abundant bacterial taxa were Roseburia, Bacteroides, Alistipes and Faecalibacterium. The results suggest that the secretome of E. durans may have favorable effects on the intestinal microbial composition, stimulating growth and different protein expression of beneficial bacteria. These findings suggest that proteins secreted by E. durans growing on FOS and GOS have different effects on the modulation of gut microbiota functional activities during cultivation.
... Recent studies have shown that probiotic bacteria adjust the settlement and eradication of pathogens in the gut, and modulate the mucosal immune system [23,24]. Probiotics are regarded as a significant prophylactic or therapeutic approach for several mucosal and non-mucosal immune-related disorders, such as IBDs, celiac disease, metabolic syndrome, and diabetes [25]. Probiotics have been fruitfully employed for the treatment of antibiotic-associated diarrhea and have antimicrobial, antimutagenic, anti-diarrheal, and anticarcinogenic properties. ...
... O înțelegere mai profundă a microbiotei intestinale și a rolului acesteia este necesară pentru strategiile viitoare de asistență medicală. În acest sens, studii ample ale potențialei utilizări a speciilor de bacterii probiotice selectate și a tulpinilor acestora pentru prevenirea și tratarea numeroaselor boli umane și animale sunt necesare [338][339][340][341]. ...
Thesis
This is my PhD thesis. The aim of this research is to provide a very complex and complete overview of the use, action and value of probiotics in patients with MS and DZT2, on the parameters characteristic of the 2 pathologies, such as weight status, glycemic profile, blood pressure (BP) and lipid metabolism, but also on the intestinal microbiome. The reason we selected the current topic was the increased prevalence of MS and its associated diseases, given that many therapies are not controlled in terms of achieving goals; therefore, additional measures are needed. In addition, supplementation with probiotics is an easy way to give the patient another chance to control or at least improve his condition and improve his quality of life.
... Long-term impact. IMT usually generates profound changes in the recipient's Gut Microbiota, which are usually beneficial, in a safe procedure [51]. More studies must be done to determine its longevity. ...
Article
BACKGROUND Critically ill trauma patients are at an increased risk for infection, which can increase morbidity and mortality. The use of probiotic preparations for infection prevention is promising, yet the results of their effectiveness are mixed. OBJECTIVES To synthesize current research regarding the use of probiotics to prevent and possibly treat infection in the critically ill adult trauma population. METHODS DATA SOURCES The medicine and nursing databases PubMed, CINAHL, and ProQuest were searched for this review. STUDY SELECTION Studies addressed the efficacy of probiotics in the critically ill in reducing health care–associated infection rates, with particular emphasis on patients who experienced severe trauma. DATA EXTRACTION Studies were reviewed and included through the consensus of the authors. Each study's population, design, methodology, results, and conclusions were analyzed for relevance. Of 170 publications retrieved and reviewed, 21 articles were identified for use in this review, including original research, meta-analyses, and reviews. RESULTS DATA SYNTHESIS There is a lack of homogeneity of studies concerning probiotic species/strain selection and frequency and timing of dosing in the current body of literature. KEY THEMES There is also a lack of consensus on what constitutes a “health care–associated infection,” with few studies identifying specific types of infection and the role probiotics may play in preventing these infections. CONCLUSION Upon reviewing the current body of evidence, one cannot definitively conclude that probiotic supplementation in the critically-ill trauma population decreases health care–associated infection rates and improves outcomes, but most published evidence supports their use.
Article
Full-text available
Probiotic bacteria are known to have ability to tolerate inhospitable conditions experienced during food preparation, food storage, and gastrointestinal tract of consumer. As probiotics are living cells, they are adversely affected by the harsh environment of the carrier matrix as well as low pH, bile salts, oxidative stress, osmotic pressure, and commensal microflora of the host. To overcome the unfavorable environments, many probiotics switch on the cell-mediated protection mechanisms, which helps them to survive, acclimatize and remain operational in the harsh circumstances. In this review, we provide comprehensive understanding on the different stresses experienced by the probiotic when added in carrier food as well as during human gastrointestinal tract transit. Under such situation how these health beneficial bacteria protect themselves by activation of several defense systems and get adapted to the lethal environments. Graphical Abstract
Article
In 1954, the term “probiotics” was coined by Ferdinand Vergin in his article. Although there are many clinical reports on the use of pro/synbiotics and other microbial preparations to prevent postoperative infections and related complications in patients with Colorectal cancer (CRC), their effectiveness remains divided. Therefore, we collected relevant high-quality randomized controlled trial (RCT) studies and conducted systematic review and meta-analysis. We electronically searched online databases (the PubMed, EMBASE, MEDLINE, Cochrane Central Register of Controlled Trials (CENTRAL), Allied and Alternative Medieine (AMED), China National Knowledge Infrastructure (CNKI), Wanfang, and Weipu) for literature published until December 2020. These reports were rigorously screened, and the data extracted, assessed for risk of bias (ROB), and subjected to meta-analysis and subgroup analysis. Postoperative infections were the main criteria for outcomes. Nineteen high-quality articles were included, involving 1975 patients. Compared with the control group, the pro/synbiotics group had reduced total postoperative infections ((odds ratio)OR = 0.28, 95% (confidence interval)CI: 0.20; 0.39, p < 0.0001), which included surgical site infections (SSI) (OR = 0.43, 95% CI: 0.31; 0.58, p < 0.0001) and non-surgical site infections (non-SSI) (OR = 0.28 95% CI: 0.20; 0.39, p < 0.0001).What is more, in aspects of inflammatory factors, intestinal dysbiosis, non-infectious complications, and systemic symptoms, the treatment group was better than the control group. However, there were no differences in perineal infections (OR = 0.45, 95% CI: 0.13; 1.50, p = 0.1933), celiac infections (OR = 0.54, 95% CI: 0.11; 2.66, p = 0.4471), or systemic inflammatory response syndrome (SIRS) incidence (OR = 0.63, 95% CI: 0.31; 1.30, p = 0.2139), etc. There were no differences in intervention (probiotics or synbiotics), strain type (multistrain or non-multistrain probiotics), and intervention time (administration preoperatively or pre-and-postoperatively). Pro/synbiotics can effectively prevent postoperative infections and related complications in patients with CRC. The strain type and intervention time did not affect the treatment effects.
Chapter
The chapter summarizes the importance of gut microbiota and the relationship between gut microbiota and different inflammatory disease conditions. It also discusses how the commensals and pathogens modulate intestinal barrier functions directly and indirectly, which is the major factor contributing to gut inflammatory disorders. The brain–gut axis involving the hypothalamus–pituitary–adrenal axis and enteric nervous system has a major role in the inflammatory bowel disorder (IBD) pathogenesis. IBD, which is the most common among gut inflammatory disorders, involves a homeostatic imbalance in the microbiota, gut epithelium, and intestinal immune response. This chapter highlights various animal models used to study inflammation related to gut and the management of these pathologies by using different probiotic strains. The probiotics cause immune stimulation through dendritic cells, which further prevents pathogen translocation and strengthens the host immune system. Probiotics, prebiotics, fermentable carbohydrates, and antibiotics are some of the microbiota directed therapies used to modulate host metabolic and immune response in IBD. The most common bacterial strains used in the management of these disorders include Lactobacillus and Bifidobacterium strains which have shown good results in mice model. Understanding the mechanisms involved in disorders related to inflammation in the gut will further help in exploring possible therapies for their management.
Article
Full-text available
The human gut is one of the most complex ecosystems, composed of 1013-1014 microorganisms which play an important role in human health. In addition, some food products contain live bacteria which transit through our gastrointestinal tract and could exert beneficial effects on our health (known as probiotic effect). Among the numerous proposed health benefits attributed to commensal and probiotic bacteria, their capacity to interact with the host immune system is now well demonstrated. Currently, the use of recombinant lactic acid bacteria to deliver compounds of health interest is gaining importance as an extension of the probiotic concept. This review summarizes some of the recent findings and perspectives in the study of the crosstalk of both commensal and probiotic bacteria with the human host as well as the latest studies in recombinant commensal and probiotic bacteria. Our aim is to highlight the potential roles of recombinant bacteria in this ecosystem.
Article
Full-text available
A dense resident microbial community in the gut, referred as the commensal microbiota, coevolved with the host and is essential for many host physiological processes that include enhancement of the intestinal epithelial barrier, development of the immune system and acquisition of nutrients. A major function of the microbiota is protection against colonization by pathogens and overgrowth of indigenous pathobionts that can result from the disruption of the healthy microbial community. The mechanisms that regulate the ability of the microbiota to restrain pathogen growth are complex and include competitive metabolic interactions, localization to intestinal niches and induction of host immune responses. Pathogens, in turn, have evolved strategies to escape from commensal-mediated resistance to colonization. Thus, the interplay between commensals and pathogens or indigenous pathobionts is critical for controlling infection and disease. Understanding pathogen-commensal interactions may lead to new therapeutic approaches to treating infectious diseases.
Article
Full-text available
The gut microbiome is the term given to describe the vast collection of symbiotic microorganisms in the human gastrointestinal system and their collective interacting genomes. Recent studies have suggested that the gut microbiome performs numerous important biochemical functions for the host, and disorders of the microbiome are associated with many and diverse human disease processes. Systems biology approaches based on next generation 'omics' technologies are now able to describe the gut microbiome at a detailed genetic and functional (transcriptomic, proteomic and metabolic) level, providing new insights into the importance of the gut microbiome in human health, and they are able to map microbiome variability between species, individuals and populations. This has established the importance of the gut microbiome in the disease pathogenesis for numerous systemic disease states, such as obesity and cardiovascular disease, and in intestinal conditions, such as inflammatory bowel disease. Thus, understanding microbiome activity is essential to the development of future personalized strategies of healthcare, as well as potentially providing new targets for drug development. Here, we review recent metagenomic and metabonomic approaches that have enabled advances in understanding gut microbiome activity in relation to human health, and gut microbial modulation for the treatment of disease. We also describe possible avenues of research in this rapidly growing field with respect to future personalized healthcare strategies.
Article
Commensal bacteria are necessary for the development and maintenance of a healthy immune system. Harnessing the ability of microbiota to affect host immunity is considered an important therapeutic strategy for many mucosal and nonmucosal immune-related conditions, such as inflammatory bowel diseases (IBDs), celiac disease, metabolic syndrome, diabetes, and microbial infections. In addition to well-established immunostimulatory effects of the microbiota, the presence of individual mutualistic commensal bacteria with immunomodulatory effects has been described. These organisms are permanent members of the commensal microbiota and affect host immune homeostasis in specific ways. Identification of individual examples of such immunomodulatory commensals and understanding their mechanisms of interaction with the host will be invaluable in designing therapeutic strategies to reverse intestinal dysbiosis and recover immunological homeostasis.
Article
Probiotic bacteria can modulate immune responses in the host gastrointestinal tract to promote health. The genomics era has provided novel opportunities for the discovery and characterization of bacterial probiotic effector molecules that elicit specific responses in the intestinal system. Furthermore, nutrigenomic analyses of the response to probiotics have unravelled the signalling and immune response pathways which are modulated by probiotic bacteria. Together, these genomic approaches and nutrigenomic analyses have identified several bacterial factors that are involved in modulation of the immune system and the mucosal barrier, and have revealed that a molecular 'bandwidth of human health' could represent a key determinant in an individual's physiological responsiveness to probiotics. These approaches may lead to improved stratification of consumers and to subpopulation-level probiotic supplementation to maintain or improve health, or to reduce the risk of disease.
Article
How can probiotic bacteria transduce their health benefits to the host? Bacterial cell surface macromolecules are key factors in this beneficial microorganism-host crosstalk, as they can interact with host pattern recognition receptors (PRRs) of the gastrointestinal mucosa. In this Review, we highlight the documented signalling interactions of the surface molecules of probiotic bacteria (such as long surface appendages, polysaccharides and lipoteichoic acids) with PRRs. Research on host-probiotic interactions can benefit from well-documented host-microorganism studies that span the spectrum from pathogenicity to mutualism. Distinctions and parallels are therefore drawn with the interactions of similar molecules that are presented by gastrointestinal commensals and pathogens.