ijms-logo

Journal Browser

Journal Browser

Prebiotics and Probiotics: Healthy Biotools for Molecular Integrative and Modulation Approaches

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Bioactives and Nutraceuticals".

Deadline for manuscript submissions: closed (31 December 2022) | Viewed by 33706

Special Issue Editors


E-Mail Website
Guest Editor
1. Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain
2. Instituto de Investigación Biosanitaria (IBS), 18014 Granada, Spain
3. Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain
Interests: nutrition and dietetics nutritional; biochemistry; pharmacy; cell biology; molecular biology; immunology; regulation of the gene expression in prokaryotes and eukaryotes; pathogens; enzymology and biotechnology
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
1. Department of Microbiology, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain
2. Instituto de Investigación Biosanitaria (IBS), 18014 Granada, Spain
3. Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain
Interests: probiotics; next-generation probiotics; molecular microbiology; microbiome; culturomics and toxicomicrobiomics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The key role played by beneficial bacteria at the intestine through promoting overall human health is generally accepted. Helpful bacteria in the colon have many important functions that include increasing mineral homeostasis, strengthening the intestinal barrier, regulating the immune response and therefore human health and wellbeing. Hence, probiotics and prebiotics are food bioproducts that exert key functions in maintaining human gut eubiosis and are specially designed for this purpose.

After decades of basic and clinical research, together with the evolution of biotechnological and high-throughput technologies, a huge impact on understanding the effect of probiotics, prebiotics on the immune response, and antibacterial infection have been achieved. Thus, these biotools have become the main instruments to intervene and optimize several physiological functions of the microbiota. Moreover, considerable efforts have been made to influence the intestinal microbiota by dietary complementary means in such a way that the health of the host is positively affected.

Functional foods are those which provide benefits beyond their traditional nutritional value, either enhancing a function of the body or reducing the risk of a disease. One of the main types of functional foods are prebiotics, which are non-digestible foods (mostly oligosaccharides) that selectively stimulate the growth of a limited number of host-friendly colonic bacteria. From a chemical standpoint, resistance to human digestive enzymes and low absorption are key for these compounds to reach the distal parts of the gut, where they can be fermented by the microbiota, which in turn is selectively modified in the process. It is important to note that pathological alterations of the intestinal microbiota (dysbiosis) have been related to conditions such as obesity, diabetes, and fatty liver, and of course in inflammatory bowel disease and infection with pathogenic bacteria.

It is clear that the diversity of prebiotic substances, probiotic microbes, and the influences that they may respectively exert mean that any health-enhancement claims must be supported by experimental and clinical data. However, the molecular mechanisms and interactions of prebiotics and probiotics with their specific targets for immune responses are not well known, and such information will help to identify new alternative compounds as therapies against various pathologies.

The goal of this Special Issue is to further expand our knowledge of the molecular mechanism of prebiotics and probiotics, as well as to offer a complete and broad view of the current situation in the area, and its future projection, pointing out the new indications that will enrich this field.

In this Special Issue, we welcome innovative and original research articles, as well as high-quality review articles with pioneering opinions with a focus on probiotics and prebiotics and balance host health/disease, related to their antibacterial roles.

Prof. Dr. Abdelali Daddaoua
Prof. Dr. Margarita Aguilera
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Related Special Issue

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

3 pages, 191 KiB  
Editorial
Prebiotics and Probiotics: Healthy Biotools for Molecular Integrative and Modulation Approaches
by Margarita Aguilera and Abdelali Daddaoua
Int. J. Mol. Sci. 2023, 24(8), 7559; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24087559 - 20 Apr 2023
Cited by 2 | Viewed by 1064
Abstract
The scope of this Special Issue is to highlight and expand our knowledge on the molecular mechanisms of prebiotics and probiotics, as well as to offer a broad overview of current advancements and future directions in this research field [...] Full article

Research

Jump to: Editorial, Review

25 pages, 3542 KiB  
Article
Effects of a Novel Infant Formula on the Fecal Microbiota in the First Six Months of Life: The INNOVA 2020 Study
by Francisco Javier Ruiz-Ojeda, Julio Plaza-Diaz, Javier Morales, Guillermo Álvarez-Calatayud, Eric Climent, Ángela Silva, Juan F. Martinez-Blanch, María Enrique, Marta Tortajada, Daniel Ramon, Beatriz Alvarez, Empar Chenoll and Ángel Gil
Int. J. Mol. Sci. 2023, 24(3), 3034; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24033034 - 03 Feb 2023
Cited by 3 | Viewed by 2867
Abstract
Exclusive breastfeeding is highly recommended for infants for at least the first six months of life. However, for some mothers, it may be difficult or even impossible to do so. This can lead to disturbances in the gut microbiota, which in turn may [...] Read more.
Exclusive breastfeeding is highly recommended for infants for at least the first six months of life. However, for some mothers, it may be difficult or even impossible to do so. This can lead to disturbances in the gut microbiota, which in turn may be related to a higher incidence of acute infectious diseases. Here, we aimed to evaluate whether a novel starting formula versus a standard formula provides a gut microbiota composition more similar to that of breastfed infants in the first 6 months of life. Two hundred and ten infants (70/group) were enrolled in the study and completed the intervention until 12 months of age. For the intervention period, infants were divided into three groups: Group 1 received formula 1 (INN) with a lower amount of protein, a proportion of casein to whey protein ratio of about 70/30 by increasing the content of α-lactalbumin, and with double the amount of docosahexaenoic acid/arachidonic acid than the standard formula; INN also contained a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis). Group 2 received the standard formula (STD) and the third group was exclusively breastfed (BF) for exploratory analysis. During the study, visits were made at 21 days, 2, 4, and 6 months of age, with ±3 days for the visit at 21 days of age, ±1 week for the visit at 2 months, and ±2 weeks for the others. Here, we reveal how consuming the INN formula promotes a similar gut microbiota composition to those infants that were breastfed in terms of richness and diversity, genera, such as Bacteroides, Bifidobacterium, Clostridium, and Lactobacillus, and calprotectin and short-chain fatty acid levels at 21 days, 2 and 6 months. Furthermore, we observed that the major bacteria metabolic pathways were more alike between the INN formula and BF groups compared to the STD formula group. Therefore, we assume that consumption of the novel INN formula might improve gut microbiota composition, promoting a healthier intestinal microbiota more similar to that of an infant who receives exclusively human milk. Full article
Show Figures

Figure 1

14 pages, 4011 KiB  
Article
Effect of Probiotic Lactobacillus plantarum on Streptococcus mutans and Candida albicans Clinical Isolates from Children with Early Childhood Caries
by Yan Zeng, Ahmed Fadaak, Nora Alomeir, Yan Wu, Tong Tong Wu, Shuang Qing and Jin Xiao
Int. J. Mol. Sci. 2023, 24(3), 2991; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24032991 - 03 Feb 2023
Cited by 7 | Viewed by 2591
Abstract
Probiotics interfere with pathogenic microorganisms or reinstate the natural microbiome. Streptococcus mutans and Candida albicans are well-known emerging pathogenic bacteria/fungi for dental caries. In this study, three probiotic Lactobacilli strains (Lactobacillus plantarum 8014, L. plantarum 14917, and Lactobacillus salivarius 11741) were tested [...] Read more.
Probiotics interfere with pathogenic microorganisms or reinstate the natural microbiome. Streptococcus mutans and Candida albicans are well-known emerging pathogenic bacteria/fungi for dental caries. In this study, three probiotic Lactobacilli strains (Lactobacillus plantarum 8014, L. plantarum 14917, and Lactobacillus salivarius 11741) were tested on S. mutans and C. albicans clinical isolates using a multispecies biofilm model simulating clinical cariogenic conditions. The ten pairs of clinical isolates of S. mutans and C. albicans were obtained from children with severe early childhood caries. Our study findings show a remarkable inhibitory effect of L. plantarum 14917 on S. mutans and C. albicans clinical isolates, resulting in significantly reduced growth of S. mutans and C. albicans, a compromised biofilm structure with a significantly smaller microbial and extracellular matrix and a less virulent microcolony structure. FurTre, plantaricin, an antimicrobial peptide produced by L. plantarum, inhibited the growth of S. mutans and C. albicans. The mechanistic assessment indicated that L. plantarum 14917 had a positive inhibitory impact on the expression of S. mutans and C. albicans virulence genes and virulent structure, such as C. albicans hypha formation. Future utilization of L. plantarum 14917 and/or its antimicrobial peptide plantaricin could lead to a new paradigm shift in dental caries prevention. Full article
Show Figures

Figure 1

14 pages, 8711 KiB  
Article
Evaluation of Antimicrobial, Antiadhesive and Co-Aggregation Activity of a Multi-Strain Probiotic Composition against Different Urogenital Pathogens
by Patrizia Malfa, Laura Brambilla, Silvana Giardina, Martina Masciarelli, Diletta Francesca Squarzanti, Federica Carlomagno and Marisa Meloni
Int. J. Mol. Sci. 2023, 24(2), 1323; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24021323 - 10 Jan 2023
Cited by 12 | Viewed by 3103
Abstract
The urogenital microbiota is dominated by Lactobacillus that, together with Bifidobacterium, creates a physiological barrier counteracting pathogen infections. The aim of this study was to evaluate the efficacy of a multi-strain probiotic formulation (Lactiplantibacillus plantarum PBS067, Lacticaseibacillus rhamnosus LRH020, and Bifidobacterium [...] Read more.
The urogenital microbiota is dominated by Lactobacillus that, together with Bifidobacterium, creates a physiological barrier counteracting pathogen infections. The aim of this study was to evaluate the efficacy of a multi-strain probiotic formulation (Lactiplantibacillus plantarum PBS067, Lacticaseibacillus rhamnosus LRH020, and Bifidobacterium animalis subsp. lactis BL050) to inhibit adhesion and growth of urogenital pathogens. The antimicrobial and antiadhesive properties of the probiotic strains and their mixture were evaluated on human vaginal epithelium infected with Candida glabrata, Neisseria gonorrheae, Trichomonas vaginalis, and Escherichia coli-infected human bladder epithelium. The epithelial tissue permeability and integrity were assessed by transepithelial/transendothelial electrical resistance (TEER). Co-aggregation between probiotics and vaginal pathogens was also investigated to elucidate a possible mechanism of action. The multi-strain formulation showed a full inhibition of T. vaginalis, and a reduction in C. glabrata and N. gonorrheae growth. A relevant antimicrobial activity was observed for each single strain against E. coli. TEER results demonstrated that none of the strains have negatively impaired the integrity of the 3D tissues. All the probiotics and their mixture were able to form aggregates with the tested pathogens. The study demonstrated that the three strains and their mixture are effective to prevent urogenital infections. Full article
Show Figures

Figure 1

14 pages, 1688 KiB  
Article
High Acetate Concentration Protects Intestinal Barrier and Exerts Anti-Inflammatory Effects in Organoid-Derived Epithelial Monolayer Cultures from Patients with Ulcerative Colitis
by Sara Deleu, Kaline Arnauts, Lowie Deprez, Kathleen Machiels, Marc Ferrante, Geert R. B. Huys, Johan M. Thevelein, Jeroen Raes and Séverine Vermeire
Int. J. Mol. Sci. 2023, 24(1), 768; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24010768 - 01 Jan 2023
Cited by 12 | Viewed by 3313
Abstract
Short-chain fatty acids as well as their bacterial producers are of increasing interest in inflammatory bowel diseases. Although less studied compared to butyrate, acetate might also be of interest as it may be less toxic to epithelial cells, stimulate butyrate-producing bacteria by cross-feeding, [...] Read more.
Short-chain fatty acids as well as their bacterial producers are of increasing interest in inflammatory bowel diseases. Although less studied compared to butyrate, acetate might also be of interest as it may be less toxic to epithelial cells, stimulate butyrate-producing bacteria by cross-feeding, and have anti-inflammatory and barrier-protective properties. Moreover, one of the causative factors of the probiotic potency of Saccharomyces cerevisae var. boulardii is thought to be its high acetate production. Therefore, the objective was to preclinically assess the effects of high acetate concentrations on inflammation and barrier integrity in organoid-based monolayer cultures from ulcerative colitis patients. Confluent organoid-derived colonic epithelial monolayers (n = 10) were exposed to basolateral inflammatory stimulation or control medium. After 24 h, high acetate or control medium was administered apically for an additional 48 h. Changes in TEER were measured after 48 h. Expression levels of barrier genes and inflammatory markers were determined by qPCR. Pro-inflammatory proteins in the supernatant were quantified using the MSD platform. Increased epithelial resistance was observed with high acetate administration in both inflamed and non-inflamed conditions, together with decreased expression levels of IL8 and TNFα and CLDN1. Upon high acetate administration to inflamed monolayers, upregulation of HIF1α, MUC2, and MKI67, and a decrease of the majority of pro-inflammatory cytokines was observed. In our patient-derived human epithelial cell culture model, a protective effect of high acetate administration on epithelial resistance, barrier gene expression, and inflammatory protein production was observed. These findings open up new possibilities for acetate-mediated management of barrier defects and inflammation in IBD. Full article
Show Figures

Graphical abstract

17 pages, 3133 KiB  
Article
In Vitro Selection of Lactobacillus and Bifidobacterium Probiotic Strains for the Management of Oral Pathobiont Infections Associated to Systemic Diseases
by Paola Zanetta, Diletta Francesca Squarzanti, Alessia di Coste, Roberta Rolla, Paolo Aluffi Valletti, Massimiliano Garzaro, Valeria Dell’Era, Angela Amoruso, Marco Pane and Barbara Azzimonti
Int. J. Mol. Sci. 2022, 23(24), 16163; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232416163 - 18 Dec 2022
Cited by 2 | Viewed by 1460
Abstract
The human oral pathobionts Aggregatibacter actinomycetemcomitans, Streptococcus mitis and Streptococcus mutans, in dysbiosis-promoting conditions, lead to oral infections, which also represent a threat to human systemic health. This scenario may be worsened by antibiotic misuse, which favours multi-drug resistance, making the [...] Read more.
The human oral pathobionts Aggregatibacter actinomycetemcomitans, Streptococcus mitis and Streptococcus mutans, in dysbiosis-promoting conditions, lead to oral infections, which also represent a threat to human systemic health. This scenario may be worsened by antibiotic misuse, which favours multi-drug resistance, making the research on pathogen containment strategies more than crucial. Therefore, we aimed to in vitro select the most promising probiotic strains against oral pathogen growth, viability, biofilm formation, and co-aggregation capacity, employing both the viable probiotics and their cell-free supernatants (CFSs). Interestingly, we also assessed probiotic efficacy against the three-pathogen co-culture, mimicking an environment similar to that in vivo. Overall, the results showed that Lactobacillus CFSs performed better than the Bifidobacterium, highlighting Limosilactobacillus reuteri LRE11, Lacticaseibacillus rhamnosus LR04, Lacticaseibacillus casei LC04, and Limosilactobacillus fermentum LF26 as the most effective strains, opening the chance to deeper investigation of their action and CFS composition. Altogether, the methodologies presented in this study can be used for probiotic efficacy screenings, in order to better focus the research on a viable probiotic, or on its postbiotics, suitable in case of infections. Full article
Show Figures

Figure 1

23 pages, 1890 KiB  
Article
Exploring Next Generation Probiotics for Metabolic and Microbiota Dysbiosis Linked to Xenobiotic Exposure: Holistic Approach
by Alfonso Torres-Sánchez, Alicia Ruiz-Rodríguez, Pilar Ortiz, María Alejandra Moreno, Antonis Ampatzoglou, Agnieszka Gruszecka-Kosowska, Mercedes Monteoliva-Sánchez and Margarita Aguilera
Int. J. Mol. Sci. 2022, 23(21), 12917; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232112917 - 26 Oct 2022
Cited by 5 | Viewed by 2145
Abstract
Variation of gut microbiota in metabolic diseases seems to be related to dysbiosis induced by exposure to multiple substances called Microbiota Disrupting Chemicals (MDCs), which are present as environmental and dietary contaminants. Some recent studies have focused on elucidating the alterations of gut [...] Read more.
Variation of gut microbiota in metabolic diseases seems to be related to dysbiosis induced by exposure to multiple substances called Microbiota Disrupting Chemicals (MDCs), which are present as environmental and dietary contaminants. Some recent studies have focused on elucidating the alterations of gut microbiota taxa and their metabolites as a consequence of xenobiotic exposures to find possible key targets involved in the severity of the host disease triggered. Compilation of data supporting the triad of xenobiotic-microbiota-metabolic diseases would subsequently allow such health misbalances to be prevented or treated by identifying beneficial microbe taxa that could be Next Generation Probiotics (NGPs) with metabolic enzymes for MDC neutralisation and mitigation strategies. In this review, we aim to compile the available information and reports focused on variations of the main gut microbiota taxa in metabolic diseases associated with xenobiotic exposure and related microbial metabolite profiles impacting the host health status. We performed an extensive literature search using SCOPUS, Web of Science, and PubMed databases. The data retrieval and thorough analyses highlight the need for more combined metagenomic and metabolomic studies revealing signatures for xenobiotics and triggered metabolic diseases. Moreover, metabolome and microbiome compositional taxa analyses allow further exploration of how to target beneficial NGP candidates according to their alleged variability abundance and potential therapeutic significance. Furthermore, this holistic approach has identified limitations and the need of future directions to expand and integrate key knowledge to design appropriate clinical and interventional studies with NGPs. Apart from human health, the beneficial microbes and metabolites identified could also be proposed for various applications under One Health, such as probiotics for animals, plants and environmental bioremediation. Full article
Show Figures

Figure 1

15 pages, 1434 KiB  
Article
Isomelezitose Overproduction by Alginate-Entrapped Recombinant E. coli Cells and In Vitro Evaluation of Its Potential Prebiotic Effect
by Martin Garcia-Gonzalez, Fadia V. Cervantes, Ricardo P. Ipiales, Angeles de la Rubia, Francisco J. Plou and María Fernández-Lobato
Int. J. Mol. Sci. 2022, 23(20), 12682; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232012682 - 21 Oct 2022
Cited by 2 | Viewed by 1512
Abstract
In this work, the trisaccharide isomelezitose was overproduced from sucrose using a biocatalyst based on immobilized Escherichia coli cells harbouring the α-glucosidase from the yeast Metschnikowia reukaufii, the best native producer of this sugar described to date. The overall process for isomelezitose [...] Read more.
In this work, the trisaccharide isomelezitose was overproduced from sucrose using a biocatalyst based on immobilized Escherichia coli cells harbouring the α-glucosidase from the yeast Metschnikowia reukaufii, the best native producer of this sugar described to date. The overall process for isomelezitose production and purification was performed in three simple steps: (i) oligosaccharides synthesis by alginate-entrapped E. coli; (ii) elimination of monosaccharides (glucose and fructose) using alginate-entrapped Komagataella phaffii cells; and (iii) semi-preparative high performance liquid chromatography under isocratic conditions. As result, approximately 2.15 g of isomelezitose (purity exceeding 95%) was obtained from 15 g of sucrose. The potential prebiotic effect of this sugar on probiotic bacteria (Lactobacillus casei, Lactobacillus rhamnosus and Enterococcus faecium) was analysed using in vitro assays for the first time. The growth of all probiotic bacteria cultures supplemented with isomelezitose was significantly improved and was similar to that of cultures supplemented with a commercial mixture of fructo-oligosaccharides. In addition, when isomelezitose was added to the bacteria cultures, the production of organic acids (mainly butyrate) was significantly promoted. Therefore, these results confirm that isomelezitose is a potential novel prebiotic that could be included in healthier foodstuffs designed for human gastrointestinal balance maintenance. Full article
Show Figures

Figure 1

14 pages, 1251 KiB  
Article
Glucosylceramide Changes Bacterial Metabolism and Increases Gram-Positive Bacteria through Tolerance to Secondary Bile Acids In Vitro
by Huanghuang Dai, Akira Otsuka, Kurumi Tanabe, Teruyoshi Yanagita, Jiro Nakayama and Hiroshi Kitagaki
Int. J. Mol. Sci. 2022, 23(10), 5300; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105300 - 10 May 2022
Cited by 3 | Viewed by 2433
Abstract
Glucosylceramide is present in many foods, such as crops and fermented foods. Most glucosylceramides are not degraded or absorbed in the small intestine and pass through the large intestine. Glucosylceramide exerts versatile effects on colon tumorigenesis, skin moisture, cholesterol metabolism and improvement of [...] Read more.
Glucosylceramide is present in many foods, such as crops and fermented foods. Most glucosylceramides are not degraded or absorbed in the small intestine and pass through the large intestine. Glucosylceramide exerts versatile effects on colon tumorigenesis, skin moisture, cholesterol metabolism and improvement of intestinal microbes in vivo. However, the mechanism of action has not yet been fully elucidated. To gain insight into the effect of glucosylceramide on intestinal microbes, glucosylceramide was anaerobically incubated with the dominant intestinal microbe, Blautia coccoides, and model intestinal microbes. The metabolites of the cultured broth supplemented with glucosylceramide were significantly different from those of broth not treated with glucosylceramide. The number of Gram-positive bacteria was significantly increased upon the addition of glucosylceramide compared to that in the control. Glucosylceramide endows intestinal microbes with tolerance to secondary bile acid. These results first demonstrated that glucosylceramide plays a role in the modification of intestinal microbes. Full article
Show Figures

Figure 1

20 pages, 1458 KiB  
Article
Anti-Inflammatory Effect of an O-2-Substituted (1-3)-β-D-Glucan Produced by Pediococcus parvulus 2.6 in a Caco-2 PMA-THP-1 Co-Culture Model
by Sara Notararigo, Encarnación Varela, Anna Otal, María Antolín, Francisco Guarner and Paloma López
Int. J. Mol. Sci. 2022, 23(3), 1527; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23031527 - 28 Jan 2022
Cited by 7 | Viewed by 2808
Abstract
Bacterial β-glucans are exopolysaccharides (EPSs), which can protect bacteria or cooperate in biofilm formation or in bacterial cell adhesion. Pediococcus parvulus 2.6 is a lactic acid bacterium that produces an O-2-substituted (1-3)-β-D-glucan. The structural similarity of this EPS to active compounds such [...] Read more.
Bacterial β-glucans are exopolysaccharides (EPSs), which can protect bacteria or cooperate in biofilm formation or in bacterial cell adhesion. Pediococcus parvulus 2.6 is a lactic acid bacterium that produces an O-2-substituted (1-3)-β-D-glucan. The structural similarity of this EPS to active compounds such as laminarin, together with its ability to modulate the immune system and to adhere in vitro to human enterocytes, led us to investigate, in comparison with laminarin, its potential as an immunomodulator of in vitro co-cultured Caco-2 and PMA-THP-1 cells. O-2-substituted (1-3)-β-D-glucan synthesized by the GTF glycosyl transferase of Pediococcus parvulus 2.6 or that by Lactococcus lactis NZ9000[pGTF] were purified and used in this study. The XTT tests revealed that all β-glucans were non-toxic for both cell lines and activated PMA-THP-1 cells’ metabolisms. The O-2-substituted (1-3)-β-D-glucan modulated production and expression of IL-8 and the IL-10 in Caco-2 and PMA-THP-1 cells. Laminarin also modulated cytokine production by diminishing TNF-α in Caco-2 cells and IL-8 in PMA-THP-1. All these features could be considered with the aim to produce function foods, supplemented with laminarin or with another novel β-glucan-producing strain, in order to ameliorate an individual’s immune system response toward pathogens or to control mild side effects in remission patients affected by inflammatory bowel diseases. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

32 pages, 6752 KiB  
Review
Overview of the Importance of Biotics in Gut Barrier Integrity
by Aleksandra Maria Kocot, Elżbieta Jarocka-Cyrta and Natalia Drabińska
Int. J. Mol. Sci. 2022, 23(5), 2896; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052896 - 07 Mar 2022
Cited by 26 | Viewed by 8003
Abstract
Increased gut permeability is suggested to be involved in the pathogenesis of a growing number of disorders. The altered intestinal barrier and the subsequent translocation of bacteria or bacterial products into the internal milieu of the human body induce the inflammatory state. Gut [...] Read more.
Increased gut permeability is suggested to be involved in the pathogenesis of a growing number of disorders. The altered intestinal barrier and the subsequent translocation of bacteria or bacterial products into the internal milieu of the human body induce the inflammatory state. Gut microbiota maintains intestinal epithelium integrity. Since dysbiosis contributes to increased gut permeability, the interventions that change the gut microbiota and correct dysbiosis are suggested to also restore intestinal barrier function. In this review, the current knowledge on the role of biotics (probiotics, prebiotics, synbiotics and postbiotics) in maintaining the intestinal barrier function is summarized. The potential outcome of the results from in vitro and animal studies is presented, and the need for further well-designed randomized clinical trials is highlighted. Moreover, we indicate the need to understand the mechanisms by which biotics regulate the function of the intestinal barrier. This review is concluded with the future direction and requirement of studies involving biotics and gut barrier. Full article
Show Figures

Figure 1

Back to TopTop