nutrients-logo

Journal Browser

Journal Browser

Prebiotics and Probiotics in Immune Health

A special issue of Nutrients (ISSN 2072-6643). This special issue belongs to the section "Prebiotics and Probiotics".

Deadline for manuscript submissions: closed (15 December 2022) | Viewed by 40955

Special Issue Editors


E-Mail Website
Guest Editor
French National Institute for Agriculture, Food, and Environment (INRAE), Paris-Saclay University, UMR MICALIS, 78 350 Jouy-en-josas, France
Interests: prebiotic, microbiota, intestine, health

E-Mail Website
Guest Editor
French National Institute for Agriculture, Food, and Environment (INRAE) | INRAE Centre de Recherche Angers-Nantes, UR1268 BIA, Site de la Géraudière, CS 71627, F-44 316 Nantes CEDEX 3, France
Interests: allergy; prebiotic; immune system; microbiota; prevention; early life

Special Issue Information

Dear Colleagues,

Prebiotics and/or probiotics supplementation are strategies for the treatment of noncommunicable diseases and for strengthen our immune system and our microbiota. Probiotics have been defined by the Food and Agriculture Organization of the United Nations and the WHO (FAO/WHO) as “live microorganisms which when administered in adequate amounts confer a health benefit on the host”. The definition of prebiotic has been recently updated by hte ISAPP as “a substrate that is selectively utilized by host microorganisms conferring a health benefit”.

These fields have exploded in recent years due to the increased knowledge of the human gut microbiota and the awareness about the health implication of dysbiosis.

Both prebiotic and probiotic have been used for different applications and currently, their mechanistic effects are more understood.

This Special Issue of Nutrients aims to present a collection of papers to give an overview on new findings and concept in the field of probiotics and prebiotics, including results on next generation probiotics, the impact of interventions at different stages (pregnancy, early life, adult, elderly), and on different pathological situations.

Prof. Dr. Claire Cherbuy
Prof. Dr. Marie Bodinier
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. Nutrients is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). 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.

Keywords

  • prebiotic
  • probiotic
  • immune system
  • microbiota
  • health

Published Papers (12 papers)

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

Research

Jump to: Review

15 pages, 2632 KiB  
Article
Immune-Enhancing Effects of Limosilactobacillus fermentum in BALB/c Mice Immunosuppressed by Cyclophosphamide
by SukJin Kim, Hwan Hee Lee, Chang-Ho Kang, Hyojeung Kang and Hyosun Cho
Nutrients 2023, 15(4), 1038; https://0-doi-org.brum.beds.ac.uk/10.3390/nu15041038 - 19 Feb 2023
Cited by 2 | Viewed by 1910
Abstract
This study evaluates the immune-enhancing effects of Limosilactobacillus fermentum on cyclophosphamide (CP)-induced immunosuppression in BALB/c mice. In vitro, the expressions of pro-inflammatory cytokines and MAPK signaling molecules in Raw264.7 cells were analyzed by ELISA and Western blot analysis. Moreover, cell proliferation, surface receptor [...] Read more.
This study evaluates the immune-enhancing effects of Limosilactobacillus fermentum on cyclophosphamide (CP)-induced immunosuppression in BALB/c mice. In vitro, the expressions of pro-inflammatory cytokines and MAPK signaling molecules in Raw264.7 cells were analyzed by ELISA and Western blot analysis. Moreover, cell proliferation, surface receptor expression, and cytotoxicity of NK-92 cells were examined by Cell Counting Kit-8, CytoTox96 assay, and flow cytometry, respectively. To investigate the immune-enhancing effects of selected L. fermentum strains in vivo, these strains were orally administered to BALB/c mice for 2 weeks, and CP was intraperitoneally injected. Then, liver, spleen, and whole blood were isolated from each animal. Administration of single L. fermentum strains or their mixture sustained the spleen weight, the counts of white blood cells compared to non-fed group. Splenocyte proliferation and NK cytotoxicity were significantly increased in all L. fermentum-fed groups. The frequency of B220+ cells was also significantly enhanced in splenocytes isolated from L. fermentum groups. In addition, the production of cytokines (TNF-α, IFN-γ) and antibodies was recovered in splenocyte supernatants isolated from L. fermentum groups. In conclusion, L. fermentum could be a suitable functional food additive for immune-enhancing effect. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

14 pages, 1377 KiB  
Article
Efficacy and Safety of MED-01 Probiotics on Vaginal Health: A 12-Week, Multicenter, Randomized, Double-Blind, Placebo-Controlled Clinical Trial
by Sung-Ho Park, Eun Sil Lee, Sung Taek Park, Soo Young Jeong, Yeoul Yun, YongGyeong Kim, Yulah Jeong, Chang-Ho Kang and Hyun Jin Choi
Nutrients 2023, 15(2), 331; https://0-doi-org.brum.beds.ac.uk/10.3390/nu15020331 - 09 Jan 2023
Cited by 5 | Viewed by 2968
Abstract
Bacterial vaginosis (BV) is the most common disease in women of childbearing age and is caused by the growth of abnormal microbiota in the vagina. Probiotic consumption can be an effective alternative treatment to preserve or improve vaginal health. In the present study, [...] Read more.
Bacterial vaginosis (BV) is the most common disease in women of childbearing age and is caused by the growth of abnormal microbiota in the vagina. Probiotic consumption can be an effective alternative treatment to preserve or improve vaginal health. In the present study, MED-01, a complex of five strains of probiotic candidates isolated from the vagina of Korean women, was used. This study was designed as a 12-week, randomized, multicenter, double-blind, placebo-controlled clinical trial to evaluate the efficacy and safety of MED-01 on vaginal health. A total of 101 reproductive-aged women with a Nugent score of 4–6 took MED-01 (5.0 × 109 CFU) or a placebo once a day, and 76 participants completed the procedure. MED-01 significantly reduced the Nugent score compared with the placebo. Quantitative PCR analysis confirmed that Lactobacillus plantarum was significantly increased in the vagina, whereas harmful bacteria such as Mobiluncus spp., Gardnerella vaginalis, and Atopobium vaginae were suppressed after 12 weeks of MED-01 ingestion. No adverse events to the test food supplements were observed in the participants. These results confirmed that MED-01 can be used as a probiotic for treating BV, as it improves the vaginal microbiota. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

15 pages, 2245 KiB  
Article
Impaired Mucosal Homeostasis in Short-Term Fiber Deprivation Is Due to Reduced Mucus Production Rather Than Overgrowth of Mucus-Degrading Bacteria
by Annelieke Overbeeke, Michaela Lang, Bela Hausmann, Margarete Watzka, Georgi Nikolov, Jasmin Schwarz, Gudrun Kohl, Kim De Paepe, Kevin Eislmayr, Thomas Decker, Andreas Richter and David Berry
Nutrients 2022, 14(18), 3802; https://0-doi-org.brum.beds.ac.uk/10.3390/nu14183802 - 15 Sep 2022
Cited by 1 | Viewed by 2671
Abstract
The gut mucosal environment is key in host health; protecting against pathogens and providing a niche for beneficial bacteria, thereby facilitating a mutualistic balance between host and microbiome. Lack of dietary fiber results in erosion of the mucosal layer, suggested to be a [...] Read more.
The gut mucosal environment is key in host health; protecting against pathogens and providing a niche for beneficial bacteria, thereby facilitating a mutualistic balance between host and microbiome. Lack of dietary fiber results in erosion of the mucosal layer, suggested to be a result of increased mucus-degrading gut bacteria. This study aimed to use quantitative analyses to investigate the diet-induced imbalance of mucosal homeostasis. Seven days of fiber-deficiency affected intestinal anatomy and physiology, seen by reduced intestinal length and loss of the colonic crypt-structure. Moreover, the mucus layer was diminished, muc2 expression decreased, and impaired mucus secretion was detected by stable isotope probing. Quantitative microbiome profiling of the gut microbiota showed a diet-induced reduction in bacterial load and decreased diversity across the intestinal tract, including taxa with fiber-degrading and butyrate-producing capabilities. Most importantly, there was little change in the absolute abundance of known mucus-degrading bacteria, although, due to the general loss of taxa, relative abundance would erroneously indicate an increase in mucus degraders. These findings underscore the importance of using quantitative methods in microbiome research, suggesting erosion of the mucus layer during fiber deprivation is due to diminished mucus production rather than overgrowth of mucus degraders. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

11 pages, 478 KiB  
Article
Study Protocol for a Randomised Controlled Trial Investigating the Effects of Maternal Prebiotic Fibre Dietary Supplementation from Mid-Pregnancy to Six Months’ Post-Partum on Child Allergic Disease Outcomes
by Debra J. Palmer, Jeffrey Keelan, Johan Garssen, Karen Simmer, Maria C. Jenmalm, Ravisha Srinivasjois, Desiree Silva and Susan L. Prescott
Nutrients 2022, 14(13), 2753; https://0-doi-org.brum.beds.ac.uk/10.3390/nu14132753 - 02 Jul 2022
Cited by 2 | Viewed by 2717
Abstract
Infant allergy is the most common early manifestation of an increasing propensity for inflammation and immune dysregulation in modern environments. Refined low-fibre diets are a major risk for inflammatory diseases through adverse effects on the composition and function of gut microbiota. This has [...] Read more.
Infant allergy is the most common early manifestation of an increasing propensity for inflammation and immune dysregulation in modern environments. Refined low-fibre diets are a major risk for inflammatory diseases through adverse effects on the composition and function of gut microbiota. This has focused attention on the potential of prebiotic dietary fibres to favourably change gut microbiota, for local and systemic anti-inflammatory effects. In pregnancy, the immunomodulatory effects of prebiotics may also have benefits for the developing fetal immune system, and provide a potential dietary strategy to reduce the risk of allergic disease. Here, we present the study protocol for a double-blinded, randomised controlled trial investigating the effects of maternal prebiotics supplementation on child allergic disease outcomes. Eligible pregnant women have infants with a first-degree relative with a history of medically diagnosed allergic disease. Consented women are randomised to consume either prebiotics (galacto-oligosaccharides and fructo-oligosaccharides) or placebo (maltodextrin) powder daily from 18–20 weeks’ gestation to six months’ post-partum. The target sample size is 652 women. The primary outcome is infant medically diagnosed eczema; secondary outcomes include allergen sensitisation, food allergies and recurrent wheeze. Breast milk, stool and blood samples are collected at multiple timepoints for further analysis. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

19 pages, 3833 KiB  
Article
Leuconostoc mesenteroides LVBH107 Antibacterial Activity against Porphyromonas gingivalis and Anti-Inflammatory Activity against P. gingivalis Lipopolysaccharide-Stimulated RAW 264.7 Cells
by Chang Luan, Jiaqing Yan, Ning Jiang, Chuang Zhang, Xu Geng, Zhengqiang Li and Chen Li
Nutrients 2022, 14(13), 2584; https://0-doi-org.brum.beds.ac.uk/10.3390/nu14132584 - 22 Jun 2022
Cited by 8 | Viewed by 2811
Abstract
Probiotics, active microorganisms benefiting human health, currently serve as nutritional supplements and clinical treatments. Periodontitis, a chronic infectious oral disease caused by Porphyromonas gingivalis (P. gingivalis), activates the host immune response to release numerous proinflammatory cytokines. Here, we aimed to clarify [...] Read more.
Probiotics, active microorganisms benefiting human health, currently serve as nutritional supplements and clinical treatments. Periodontitis, a chronic infectious oral disease caused by Porphyromonas gingivalis (P. gingivalis), activates the host immune response to release numerous proinflammatory cytokines. Here, we aimed to clarify Leuconostoc mesenterica (L. mesenteroides) LVBH107 probiotic effects based on the inhibition of P. gingivalis activities while also evaluating the effectiveness of an in vitro P. gingivalis lipopolysaccharide-stimulated RAW 264.7 cell-based inflammation mode. L. mesenteroides LVBH107 survived at acid, bile salts, lysozyme, and hydrogen peroxide conditions, auto-aggregated and co-aggregated with P. gingivalis, exhibited strong hydrophobicity and electrostatic action, and strongly adhered to gingival epithelial and HT-29 cells (thus exhibiting oral tissue adherence and colonization abilities). Moreover, L. mesenteroides LVBH107 exhibited sensitivity to antibiotics erythromycin, doxycycline, minocycline, ampicillin, and others (thus indicating it lacked antibiotic resistance plasmids), effectively inhibited P. gingivalis biofilm formation and inflammation (in vitro inflammation model), reduced the secretion of pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) and inflammatory mediators (NO and PGE2), and decreased the expression levels of inflammation related genes. Thus, L. mesenterica LVBH107 holds promise as a probiotic that can inhibit P. gingivalis biofilm formation and exert anti-inflammatory activity to maintain oral health. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

34 pages, 5812 KiB  
Article
Lentils and Yeast Fibers: A New Strategy to Mitigate Enterotoxigenic Escherichia coli (ETEC) Strain H10407 Virulence?
by Thomas Sauvaitre, Florence Van Herreweghen, Karen Delbaere, Claude Durif, Josefien Van Landuyt, Khaled Fadhlaoui, Ségolène Huille, Frédérique Chaucheyras-Durand, Lucie Etienne-Mesmin, Stéphanie Blanquet-Diot and Tom Van de Wiele
Nutrients 2022, 14(10), 2146; https://0-doi-org.brum.beds.ac.uk/10.3390/nu14102146 - 21 May 2022
Viewed by 2401
Abstract
Dietary fibers exhibit well-known beneficial effects on human health, but their anti-infectious properties against enteric pathogens have been poorly investigated. Enterotoxigenic Escherichia coli (ETEC) is a major food-borne pathogen that causes acute traveler’s diarrhea. Its virulence traits mainly rely on adhesion to an [...] Read more.
Dietary fibers exhibit well-known beneficial effects on human health, but their anti-infectious properties against enteric pathogens have been poorly investigated. Enterotoxigenic Escherichia coli (ETEC) is a major food-borne pathogen that causes acute traveler’s diarrhea. Its virulence traits mainly rely on adhesion to an epithelial surface, mucus degradation, and the secretion of two enterotoxins associated with intestinal inflammation. With the increasing burden of antibiotic resistance worldwide, there is an imperious need to develop novel alternative strategies to control ETEC infections. This study aimed to investigate, using complementary in vitro approaches, the inhibitory potential of two dietary-fiber-containing products (a lentil extract and yeast cell walls) against the human ETEC reference strain H10407. We showed that the lentil extract decreased toxin production in a dose-dependent manner, reduced pro-inflammatory interleukin-8 production, and modulated mucus-related gene induction in ETEC-infected mucus-secreting intestinal cells. We also report that the yeast product reduced ETEC adhesion to mucin and Caco-2/HT29-MTX cells. Both fiber-containing products strengthened intestinal barrier function and modulated toxin-related gene expression. In a complex human gut microbial background, both products did not elicit a significant effect on ETEC colonization. These pioneering data demonstrate the promising role of dietary fibers in controlling different stages of the ETEC infection process. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

15 pages, 1556 KiB  
Article
Description of a Newly Isolated Blautia faecis Strain and Its Benefit in Mouse Models of Post-Influenza Secondary Enteric and Pulmonary Infections
by Sophie Verstraeten, Valentin Sencio, Audrey Raise, Eugénie Huillet, Séverine Layec, Lucie Deruyter, Séverine Heumel, Sandrine Auger, Véronique Robert, Philippe Langella, Laurent Beney, François Trottein and Muriel Thomas
Nutrients 2022, 14(7), 1478; https://0-doi-org.brum.beds.ac.uk/10.3390/nu14071478 - 01 Apr 2022
Cited by 6 | Viewed by 2400
Abstract
The expanding knowledge on the systemic influence of the human microbiome suggests that fecal samples are underexploited sources of new beneficial strains for extra-intestinal health. We have recently shown that acetate, a main circulating microbiota-derived molecule, reduces the deleterious effects of pulmonary Streptococcus [...] Read more.
The expanding knowledge on the systemic influence of the human microbiome suggests that fecal samples are underexploited sources of new beneficial strains for extra-intestinal health. We have recently shown that acetate, a main circulating microbiota-derived molecule, reduces the deleterious effects of pulmonary Streptococcus pneumoniae and enteric Salmonella enterica serovar Typhimurium bacterial post-influenza superinfections. Considering the beneficial and broad effects of acetate, we intended to isolate a commensal strain, producing acetate and potentially exploitable in the context of respiratory infections. We designed successive steps to select intestinal commensals that are extremely oxygen-sensitive, cultivable after a freezing process, without a proinflammatory effect on IL-8 induction, and producing acetate. We have identified the Blautia faecis DSM33383 strain, which decreased the TNFα-induced production of IL-8 by the intestinal epithelial cell line HT-29. The beneficial effect of this bacterial strain was further studied in two preclinical models of post-influenza Streptococcus pneumoniae (S.p) and Salmonella enterica serovar Typhimurium (S.t) superinfection. The intragastrical administration of Blautia faecis DSM33383 led to protection in influenza-infected mice suffering from an S.p. and, to a lesser extent, from an S.t secondary infection. Altogether, this study showed that Blautia faecis DSM33383 could be a promising candidate for preventive management of respiratory infectious diseases. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

Review

Jump to: Research

25 pages, 1338 KiB  
Review
Pre-, pro-, syn-, and Postbiotics in Infant Formulas: What Are the Immune Benefits for Infants?
by Anaïs Lemoine, Patrick Tounian, Karine Adel-Patient and Muriel Thomas
Nutrients 2023, 15(5), 1231; https://0-doi-org.brum.beds.ac.uk/10.3390/nu15051231 - 28 Feb 2023
Cited by 9 | Viewed by 5308
Abstract
The first objective of infant formulas is to ensure the healthy growth of neonates and infants, as the sole complete food source during the first months of life when a child cannot be breastfed. Beyond this nutritional aspect, infant nutrition companies also try [...] Read more.
The first objective of infant formulas is to ensure the healthy growth of neonates and infants, as the sole complete food source during the first months of life when a child cannot be breastfed. Beyond this nutritional aspect, infant nutrition companies also try to mimic breast milk in its unique immuno-modulating properties. Numerous studies have demonstrated that the intestinal microbiota under the influence of diet shapes the maturation of the immune system and influences the risk of atopic diseases in infants. A new challenge for dairy industries is, therefore, to develop infant formulas inducing the maturation of immunity and the microbiota that can be observed in breastfed delivered vaginally, representing reference infants. Streptococcus thermophilus, Lactobacillus reuteri DSM 17938, Bifidobacterium breve (BC50), Bifidobacterium lactis Bb12, Lactobacillus fermentum (CECT5716), and Lactobacillus rhamnosus GG (LGG) are some of the probiotics added to infant formula, according to a literature review of the past 10 years. The most frequently used prebiotics in published clinical trials are fructo-oligosaccharides (FOSs), galacto-oligosaccharides (GOSs), and human milk oligosaccharides (HMOs). This review sums up the expected benefits and effects for infants of pre-, pro-, syn-, and postbiotics added to infant formula regarding the microbiota, immunity, and allergies. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

19 pages, 6302 KiB  
Review
Effects of Microecological Regulators on Rheumatoid Arthritis: A Systematic Review and Meta-Analysis of Randomized, Controlled Trials
by Tong Wu, Yanhong Li, Yinlan Wu, Xiuping Liang, Yu Zhou, Zehui Liao, Ji Wen, Lu Cheng, Yubin Luo and Yi Liu
Nutrients 2023, 15(5), 1102; https://0-doi-org.brum.beds.ac.uk/10.3390/nu15051102 - 22 Feb 2023
Cited by 1 | Viewed by 2027
Abstract
In this study, the available data from published randomized, controlled trials (RCTs) of the use of intestinal microecological regulators as adjuvant therapies to relieve the disease activity of rheumatoid arthritis (RA) are systematically compared. An English literature search was performed using PubMed, Embase, [...] Read more.
In this study, the available data from published randomized, controlled trials (RCTs) of the use of intestinal microecological regulators as adjuvant therapies to relieve the disease activity of rheumatoid arthritis (RA) are systematically compared. An English literature search was performed using PubMed, Embase, Scopus, Web of Science and the Cochrane Central Registry of Controlled Trials and supplemented by hand searching reference lists. Three independent reviewers screened and assessed the quality of the studies. Among the 2355 citations identified, 12 RCTs were included. All data were pooled using a mean difference (MD) with a 95% CI. The disease activity score (DAS) showed a significant improvement following microecological regulators treatment (MD (95% CI) of −1.01 (−1.81, −0.2)). A borderline significant reduction in the health assessment questionnaire (HAQ) scores was observed (MD (95% CI) of −0.11 (−0.21, −0.02)). We also confirmed the known effects of probiotics on inflammatory parameters such as the C-reactive protein (CRP) (MD −1.78 (95% CI −2.90, −0.66)) and L-1β (MD −7.26 (95% CI −13.03, −1.50)). No significant impact on visual analogue scale (VAS) of pain and erythrocyte sedimentation rate (ESR) reduction was observed. Intestinal microecological regulators supplementation could decrease RA activity with a significant effect on DAS28, HAQ and inflammatory cytokines. Nevertheless, these findings need further confirmation in large clinical studies with greater consideration of the confounding variables of age, disease duration, and individual medication regimens. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

28 pages, 1545 KiB  
Review
Immunomodulation of B Lymphocytes by Prebiotics, Probiotics and Synbiotics: Application in Pathologies
by Anaïs Rousseaux, Carole Brosseau and Marie Bodinier
Nutrients 2023, 15(2), 269; https://0-doi-org.brum.beds.ac.uk/10.3390/nu15020269 - 05 Jan 2023
Cited by 11 | Viewed by 3765
Abstract
Introduction: Prebiotics, probiotics and synbiotics are known to have major beneficial effects on human health due to their ability to modify the composition and the function of the gut mucosa, the gut microbiota and the immune system. These components largely function in a [...] Read more.
Introduction: Prebiotics, probiotics and synbiotics are known to have major beneficial effects on human health due to their ability to modify the composition and the function of the gut mucosa, the gut microbiota and the immune system. These components largely function in a healthy population throughout different periods of life to confer homeostasis. Indeed, they can modulate the composition of the gut microbiota by increasing bacteria strands that are beneficial for health, such as Firmicute and Bifidobacteria, and decreasing harmful bacteria, such as Enteroccocus. Their immunomodulation properties have been extensively studied in different innate cells (dendritic cells, macrophages, monocytes) and adaptive cells (Th, Treg, B cells). They can confer a protolerogenic environment but also modulate pro-inflammatory responses. Due to all these beneficial effects, these compounds have been investigated to prevent or to treat different diseases, such as cancer, diabetes, allergies, autoimmune diseases, etc. Regarding the literature, the effects of these components on dendritic cells, monocytes and T cells have been studied and presented in a number of reviews, but their impact on B-cell response has been less widely discussed. Conclusions: For the first time, we propose here a review of the literature on the immunomodulation of B-lymphocytes response by prebiotics, probiotics and synbiotics, both in healthy conditions and in pathologies. Discussion: Promising studies have been performed in animal models, highlighting the potential of prebiotics, probiotics and synbiotics intake to treat or to prevent diseases associated with B-cell immunomodulation, but this needs to be validated in humans with a full characterization of B-cell subsets and not only the humoral response. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

23 pages, 3291 KiB  
Review
Pectin in Metabolic Liver Disease
by Wanchao Hu, Anne-Marie Cassard and Dragos Ciocan
Nutrients 2023, 15(1), 157; https://0-doi-org.brum.beds.ac.uk/10.3390/nu15010157 - 29 Dec 2022
Cited by 5 | Viewed by 4378
Abstract
Alterations in the composition of the gut microbiota (dysbiosis) are observed in nutritional liver diseases, including non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) and have been shown to be associated with the severity of both. Editing the composition of the [...] Read more.
Alterations in the composition of the gut microbiota (dysbiosis) are observed in nutritional liver diseases, including non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) and have been shown to be associated with the severity of both. Editing the composition of the microbiota by fecal microbiota transfer or by application of probiotics or prebiotics/fiber in rodent models and human proof-of-concept trials of NAFLD and ALD have demonstrated its possible contribution to reducing the progression of liver damage. In this review, we address the role of a soluble fiber, pectin, in reducing the development of liver injury in NAFLD and ALD through its impact on gut bacteria. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

23 pages, 1419 KiB  
Review
Prebiotics and the Human Gut Microbiota: From Breakdown Mechanisms to the Impact on Metabolic Health
by Cassandre Bedu-Ferrari, Paul Biscarrat, Philippe Langella and Claire Cherbuy
Nutrients 2022, 14(10), 2096; https://0-doi-org.brum.beds.ac.uk/10.3390/nu14102096 - 17 May 2022
Cited by 26 | Viewed by 6001
Abstract
The colon harbours a dynamic and complex community of microorganisms, collectively known as the gut microbiota, which constitutes the densest microbial ecosystem in the human body. These commensal gut microbes play a key role in human health and diseases, revealing the strong potential [...] Read more.
The colon harbours a dynamic and complex community of microorganisms, collectively known as the gut microbiota, which constitutes the densest microbial ecosystem in the human body. These commensal gut microbes play a key role in human health and diseases, revealing the strong potential of fine-tuning the gut microbiota to confer health benefits. In this context, dietary strategies targeting gut microbes to modulate the composition and metabolic function of microbial communities are of increasing interest. One such dietary strategy is the use of prebiotics, which are defined as substrates that are selectively utilised by host microorganisms to confer a health benefit. A better understanding of the metabolic pathways involved in the breakdown of prebiotics is essential to improve these nutritional strategies. In this review, we will present the concept of prebiotics, and focus on the main sources and nature of these components, which are mainly non-digestible polysaccharides. We will review the breakdown mechanisms of complex carbohydrates by the intestinal microbiota and present short-chain fatty acids (SCFAs) as key molecules mediating the dialogue between the intestinal microbiota and the host. Finally, we will review human studies exploring the potential of prebiotics in metabolic diseases, revealing the personalised responses to prebiotic ingestion. In conclusion, we hope that this review will be of interest to identify mechanistic factors for the optimization of prebiotic-based strategies. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Immune Health)
Show Figures

Figure 1

Back to TopTop