nutrients-logo

Journal Browser

Journal Browser

Health Benefits of Traditional and Next Generation Probiotics

A special issue of Nutrients (ISSN 2072-6643).

Deadline for manuscript submissions: closed (31 July 2019) | Viewed by 78881

Special Issue Editors


E-Mail Website
Guest Editor
Department of Animal Science, Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD Wageningen, The Netherlands
Interests: intestinal microbiota; host–microbe interactions; probiotics; lactic acid bacteria; genomics; host immunity and metabolism

E-Mail
Guest Editor
NIZO Food Research BV, 6710 BA Ede, The Netherlands
Interests: lactic acid bacteria; genomics; genetics; probiotics; host–microbe interaction; cell envelope components; food fermentation; anaerobic fermentation

Special Issue Information

Dear Colleagues,

Traditionally, lactic acid bacteria isolated from the human gut have been employed as probiotics. These probiotics have been reported to elicit health improvements in a variety of clinical trials that targeted a variety of health benefit areas. However, for many of these health benefit areas, the scientific evidence for probiotic benefits is limited and in some cases inconsistent between individual studies. Moreover, the rationale behind the application of lactic acid bacteria is typically driven by their safety for consumption rather than by their association with a healthy intestinal microbiome. In the current metagenomics era, other—typically anaerobic—genera have been associated with host health and disease, which stimulated the isolation, characterization, and production of these health-associated microbes that are commonly referred to as second-generation probiotics. This Special Issue aims to cover the current status and future potential of both traditional and second-generation probiotics.

Prof. Dr. Michiel Kleerebezem
Dr. Peter Bron
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

  • gastrointestinal microbiota
  • health-promoting microbes
  • probiotics
  • anaerobic fermentation

Published Papers (10 papers)

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

Research

Jump to: Review

13 pages, 1544 KiB  
Article
Worm-Based Alternate Assessment of Probiotic Intervention against Gut Barrier Infection
by Juil Kim and Yuseok Moon
Nutrients 2019, 11(9), 2146; https://doi.org/10.3390/nu11092146 - 8 Sep 2019
Cited by 12 | Viewed by 4553
Abstract
The epithelial barrier is the frontline defense against enteropathogenic bacteria and nutrition-linked xenobiotic stressors in the alimentary tract. In particular, enteropathogenic Escherichia coli (EPEC) insults the gut barrier and is increasingly implicated in chronic intestinal diseases such as inflammatory bowel disease. For the [...] Read more.
The epithelial barrier is the frontline defense against enteropathogenic bacteria and nutrition-linked xenobiotic stressors in the alimentary tract. In particular, enteropathogenic Escherichia coli (EPEC) insults the gut barrier and is increasingly implicated in chronic intestinal diseases such as inflammatory bowel disease. For the efficient development of intervention against barrier-linked distress, the present study provided a Caenorhabditis elegans-based assessment instead of extensive preclinical evaluations using mammalian models. In particular, EPEC infected the gut and shortened the lifespan of C. elegans, which was counteracted by colonization of E. coli strain Nissle 1917 (EcN). In addition to the competitive actions of EcN against EPEC, EcN improved the gut barrier integrity of worms via the Zonula occludens ortholog (Zoo-1) induction, which was verified in the murine infection and colitis model. The worm-based assessment provided a crucial methodology and important insights into the potent chronic events in the human gut barrier after the ingestion of probiotic candidates as a mucoactive dietary or therapeutic agent. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

12 pages, 278 KiB  
Article
Effects of Lactobacillus plantarum and Lactobacillus paracasei on the Peripheral Immune Response in Children with Celiac Disease Autoimmunity: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial
by Åsa Håkansson, Carin Andrén Aronsson, Charlotte Brundin, Elin Oscarsson, Göran Molin and Daniel Agardh
Nutrients 2019, 11(8), 1925; https://doi.org/10.3390/nu11081925 - 16 Aug 2019
Cited by 60 | Viewed by 7386
Abstract
Two Lactobacillus strains have proven anti-inflammatory properties by reducing pro-inflammatory responses to antigens. This randomized double-blind placebo-controlled trial tested the hypothesis that L. plantarum HEAL9 and L. paracasei 8700:2 suppress ongoing celiac disease autoimmunity in genetically at risk children on a gluten-containing diet [...] Read more.
Two Lactobacillus strains have proven anti-inflammatory properties by reducing pro-inflammatory responses to antigens. This randomized double-blind placebo-controlled trial tested the hypothesis that L. plantarum HEAL9 and L. paracasei 8700:2 suppress ongoing celiac disease autoimmunity in genetically at risk children on a gluten-containing diet in a longitudinally screening study for celiac disease. Seventy-eight children with celiac disease autoimmunity participated of whom 40 received 1010 CFU/day of L. plantarum HEAL9 and L. paracasei 8700:2 (probiotic group) and 38 children maltodextrin (placebo group) for six months. Blood samples were drawn at zero, three and six months and phenotyping of peripheral blood lymphocytes and IgA and IgG autoantibodies against tissue transglutaminase (tTG) were measured. In the placebo group, naïve CD45RA+ Th cells decreased (p = 0.002) whereas effector and memory CD45RO+ Th cells increased (p = 0.003). In contrast, populations of cells expressing CD4+CD25highCD45RO+CCR4+ increased in the placebo group (p = 0.001). Changes between the groups were observed for NK cells (p = 0.038) and NKT cells (p = 0.008). Median levels of IgA-tTG decreased more significantly over time in the probiotic (p = 0.013) than in the placebo (p = 0.043) group whereas the opposite was true for IgG-tTG (p = 0.062 respective p = 0.008). In conclusion, daily oral administration of L. plantarum HEAL9 and L. paracasei 8700:2 modulate the peripheral immune response in children with celiac disease autoimmunity. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
14 pages, 531 KiB  
Article
Early Probiotic Supplementation and the Risk of Celiac Disease in Children at Genetic Risk
by Ulla Uusitalo, Carin Andren Aronsson, Xiang Liu, Kalle Kurppa, Jimin Yang, Edwin Liu, Jennifer Skidmore, Christiane Winkler, Marian J. Rewers, William A. Hagopian, Jin-Xiong She, Jorma Toppari, Anette-G. Ziegler, Beena Akolkar, Jill M. Norris, Suvi M. Virtanen, Jeffrey P. Krischer, Daniel Agardh and on behalf of the TEDDY Study Group
Nutrients 2019, 11(8), 1790; https://doi.org/10.3390/nu11081790 - 2 Aug 2019
Cited by 22 | Viewed by 4945
Abstract
Probiotics are linked to positive regulatory effects on the immune system. The aim of the study was to examine the association between the exposure of probiotics via dietary supplements or via infant formula by the age of 1 year and the development of [...] Read more.
Probiotics are linked to positive regulatory effects on the immune system. The aim of the study was to examine the association between the exposure of probiotics via dietary supplements or via infant formula by the age of 1 year and the development of celiac disease autoimmunity (CDA) and celiac disease among a cohort of 6520 genetically susceptible children. Use of probiotics during the first year of life was reported by 1460 children. Time-to-event analysis was used to examine the associations. Overall exposure of probiotics during the first year of life was not associated with either CDA (n = 1212) (HR 1.15; 95%CI 0.99, 1.35; p = 0.07) or celiac disease (n = 455) (HR 1.11; 95%CI 0.86, 1.43; p = 0.43) when adjusting for known risk factors. Intake of probiotic dietary supplements, however, was associated with a slightly increased risk of CDA (HR 1.18; 95%CI 1.00, 1.40; p = 0.043) compared to children who did not get probiotics. It was concluded that the overall exposure of probiotics during the first year of life was not associated with CDA or celiac disease in children at genetic risk. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

17 pages, 1393 KiB  
Article
In Vitro Probiotic and Antioxidant Potential of Lactococcus lactis subsp. cremoris LL95 and Its Effect in Mice Behaviour
by Juliana B. Ramalho, Melina B. Soares, Cristiano C. Spiazzi, Diogo F. Bicca, Vanessa M. Soares, Juliano G. Pereira, Wladimir P. da Silva, Carla P. Sehn and Francielli W. S. Cibin
Nutrients 2019, 11(4), 901; https://doi.org/10.3390/nu11040901 - 22 Apr 2019
Cited by 32 | Viewed by 5259
Abstract
The composition of intestinal microbiota is widely believed to not only affect gut health but also influence behaviour. This study aimed to evaluate the probiotic characteristics, antioxidant activity, and antidepressant- and anxiolytic-like activities of Lactococcus lactis subsp. cremoris LL95. This strain showed probiotic [...] Read more.
The composition of intestinal microbiota is widely believed to not only affect gut health but also influence behaviour. This study aimed to evaluate the probiotic characteristics, antioxidant activity, and antidepressant- and anxiolytic-like activities of Lactococcus lactis subsp. cremoris LL95. This strain showed probiotic properties such as resistance in a simulated gastric tract model and survival at different concentrations of NaCl and bile salts. Moreover, antioxidant activity of LL95 was demonstrated through DPPH radical scavenging activity, scavenging of ABTS•+ radical and ferric ion reducing antioxidant power (FRAP) assays. Female C57BL/6 mice received LL95 orally at a dose of 109 UFC/day for 28 days. LL95 improved depressive- and anxiety-like behaviour, demonstrated by decreased immobility time in the tail suspension test and forced swim test and increased per cent of time spent in the open arms on the elevated plus maze. These findings indicate the potential antioxidant activity of LL95 and its role in behaviour, suggesting that probiotic may have therapeutic applications. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Graphical abstract

9 pages, 1668 KiB  
Article
Lactobacillus fermentum PC1 has the Capacity to Attenuate Joint Inflammation in Collagen-Induced Arthritis in DBA/1 Mice
by Meera Esvaran and Patricia L. Conway
Nutrients 2019, 11(4), 785; https://doi.org/10.3390/nu11040785 - 5 Apr 2019
Cited by 18 | Viewed by 3434
Abstract
Lactobacillus strains have shown efficacy in attenuating inflammation. This study evaluated the potential of Lactobacillus fermentum PC1 for the treatment of rheumatoid arthritis (RA) using a murine model of collagen-induced arthritis. On Day 1, healthy DBA/1 mice (six to eight weeks of age) [...] Read more.
Lactobacillus strains have shown efficacy in attenuating inflammation. This study evaluated the potential of Lactobacillus fermentum PC1 for the treatment of rheumatoid arthritis (RA) using a murine model of collagen-induced arthritis. On Day 1, healthy DBA/1 mice (six to eight weeks of age) were immunized, with 100 μg of Chicken Type 11 collagen emulsified in complete Freund’s adjuvant (CFA) by intradermal injection, at the base of the tail. On Day 21, the mice were immunized intraperitoneally with 100 μg of Bovine Type11 collagen in phosphate buffered saline (PBS). On Day 28, the mice were immunized intraperitoneally with 50 μg of lipopolysaccharide (LPS). Viable L. fermentum PC1 (1 × 109 colony forming units) was given daily from Day two until the end of the experiment. From Day 21 onwards, the mice were monitored daily for clinical signs of arthritis. On Day 44, the experiment was terminated. Paws were obtained for histology and serum for cytokine assays. L. fermentum PC1-fed mice had significantly reduced paw inflammation as well as decreased synovial infiltration and less cartilage damage. Circulating serum cytokine profiles revealed decreased IL-12 and increased anti-inflammatory cytokines, namely IL-4 and IL-10. Thus, early administration of L. fermentum PC1 could prove to be a valuable therapeutic agent in the management of RA. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

20 pages, 3993 KiB  
Article
Exopolysaccharide-Producing Bifidobacterium adolescentis Strains with Similar Adhesion Property Induce Differential Regulation of Inflammatory Immune Response in Treg/Th17 Axis of DSS-Colitis Mice
by Rui Yu, Fanglei Zuo, Huiqin Ma and Shangwu Chen
Nutrients 2019, 11(4), 782; https://doi.org/10.3390/nu11040782 - 4 Apr 2019
Cited by 73 | Viewed by 6117
Abstract
Intestinal bifidobacteria benefit human health by promoting and modulating the gut flora, and boosting therapeutic efficiency for chronic metabolic diseases and cancer. Recently, Bifidobacterium adolescentis strains with high adhesion to intestinal epithelial cells were associated with induction of T-helper 17 (Th17) cells in [...] Read more.
Intestinal bifidobacteria benefit human health by promoting and modulating the gut flora, and boosting therapeutic efficiency for chronic metabolic diseases and cancer. Recently, Bifidobacterium adolescentis strains with high adhesion to intestinal epithelial cells were associated with induction of T-helper 17 (Th17) cells in humans and rodents. Here, two B. adolescentis strains with similar adhesive ability but different aggregation properties were investigated for specific immunoregulatory effects, including the underlying cellular pathway, on macrophage and T-regulatory (Treg)/Th17 axis activation in vitro and in the colon of dextran sodium sulfate (DSS)-colitis mice in vivo. In-vitro, the auto-aggregative B. adolescentis strain IF1-11 induced significantly higher IL-6 and lower IL-10 secretion from immune cells, and it induced abundant Th17 cells. The non-aggregating strain IF1-03 induced significantly higher IL-10, less IL-6 and a high proportion of Treg/Th17 cells compared to total T cells. In vivo, orally administered IF1-03 protected DSS-colitis mice via activation of dendritic cells or macrophages and skewing of Treg/Th17 cells, consistent with Treg cell induction in vitro. IF1-03 exopolysaccharides showed a functional recognition pattern similar to IF1-03 for IL-10 cytokine secretion and Treg cell-differentiation induction, both dependent on the toll-like receptor 2–ERK/p38 MAPK-signaling cascade for macrophage activation. We suggest that B. adolescentis exopolysaccharide-associated enterocyte adhesion/aggregation phenotypes determine strain-specific adaptive immune responses in the gut via the macrophage-regulated Treg/Th17 axis. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

12 pages, 914 KiB  
Article
Human Gut Microbiome Response Induced by Fermented Dairy Product Intake in Healthy Volunteers
by Olesya Volokh, Natalia Klimenko, Yulia Berezhnaya, Alexander Tyakht, Polina Nesterova, Anna Popenko and Dmitry Alexeev
Nutrients 2019, 11(3), 547; https://doi.org/10.3390/nu11030547 - 4 Mar 2019
Cited by 56 | Viewed by 17517
Abstract
Accumulated data suggests that the gut microbiome can rapidly respond to changes in diet. Consumption of fermented dairy products (FDP) fortified with probiotic microbes may be associated with positive impact on human health. However, the extent and details of the possible impact of [...] Read more.
Accumulated data suggests that the gut microbiome can rapidly respond to changes in diet. Consumption of fermented dairy products (FDP) fortified with probiotic microbes may be associated with positive impact on human health. However, the extent and details of the possible impact of FDP consumption on gut community structure tends to vary across individuals. We used microbiome analysis to characterize changes in gut microbiota composition after 30 days of oral intake of a yoghurt fortified with Bifidobacterium animalis subsp. lactis BB-12. 16S rRNA gene sequencing was used to assess the gut microbial composition before and after FDP consumption in healthy adults (n = 150). Paired comparison of gut microbial content demonstrated an increase in presence of potentially beneficial bacteria, particularly, Bifidobacterium genus, as well as Adlercreutzia equolifaciens and Slackia isoflavoniconvertens. At a functional level, an increased capacity to metabolize lactose and synthesize amino acids was observed accompanied by a lowered potential for synthesis of lipopolysaccharides. Cluster analysis revealed that study volunteers segregated into two groups with post-intervention microbiota response that was dependent on the baseline microbial community structure. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

15 pages, 2872 KiB  
Article
In Vitro Antimicrobial Activity and Probiotic Potential of Bifidobacterium and Lactobacillus against Species of Clostridium
by Cinara R. A. V. Monteiro, Monique S. do Carmo, Bruna O. Melo, Matheus S. Alves, Camilla I. dos Santos, Sílvio G. Monteiro, Maria Rosa Q. Bomfim, Elizabeth S. Fernandes and Valério Monteiro-Neto
Nutrients 2019, 11(2), 448; https://doi.org/10.3390/nu11020448 - 21 Feb 2019
Cited by 61 | Viewed by 9798
Abstract
Many Clostridium species are found as commensal members of the intestinal microbiota. However, imbalances of the microbiota may lead to certain infections caused by these microorganisms, mainly Clostridium butyricum, Clostridium difficile, and Clostridium perfringens. In many cases, infection recurrence can [...] Read more.
Many Clostridium species are found as commensal members of the intestinal microbiota. However, imbalances of the microbiota may lead to certain infections caused by these microorganisms, mainly Clostridium butyricum, Clostridium difficile, and Clostridium perfringens. In many cases, infection recurrence can occur after antibiotics, indicating the need for novel therapeutic options that act on the pathogens and also restore the microbiota. Herein, the in vitro antimicrobial activity and probiotic potential of clinical and reference strains of Bifidobacterium and Lactobacillus were investigated against Clostridium species. Antimicrobial activity was evaluated by the agar spot test and inhibition of gas production. Then, the probiotic potential of selected strains was assessed by analyzing their coaggregation ability, adhesive properties to host cells and mucin, tolerance to acidic pH and bile salts, and antimicrobial susceptibility profiles. Lactobacillus plantarum ATCC 8014 was the most promising strain based on its inhibitory activity against Clostridium spp. Also, this strain met criteria to be considered a probiotic based on its coaggregation ability, adhesive properties, and tolerance to harsh pH and bile acid salt conditions. The results indicate that among the studied strains, L. plantarum ATCC 8014 presents probiotic potential for controlling infections induced by the studied Clostridium species and should be further evaluated in in vivo animal models. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

Review

Jump to: Research

26 pages, 320 KiB  
Review
Probiotics in Extraintestinal Diseases: Current Trends and New Directions
by Despoina E. Kiousi, Athanasios Karapetsas, Kyriaki Karolidou, Mihalis I. Panayiotidis, Aglaia Pappa and Alex Galanis
Nutrients 2019, 11(4), 788; https://doi.org/10.3390/nu11040788 - 5 Apr 2019
Cited by 49 | Viewed by 9013
Abstract
Probiotics are defined as live microorganisms that when administered in adequate amounts confer a health benefit to the host. Their positive supplementation outcomes on several gastrointestinal disorders are well defined. Nevertheless, their actions are not limited to the gut, but may also impart [...] Read more.
Probiotics are defined as live microorganisms that when administered in adequate amounts confer a health benefit to the host. Their positive supplementation outcomes on several gastrointestinal disorders are well defined. Nevertheless, their actions are not limited to the gut, but may also impart their beneficial effects at distant sites and organs. In this regard, in this review article we: (i) comprehensively describe the main mechanisms of action of probiotics at distant sites, including bones, skin, and brain; (ii) critically present their therapeutic potential against bone, skin, and neuronal diseases (e.g., osteoporosis, non-healing wounds and autoimmune skin illnesses, mood, behavior, memory, and cognitive impairments); (iii) address the current gaps in the preclinical and clinical research; and (iv) indicate new research directions and suggest future investigations. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
16 pages, 2008 KiB  
Review
The Efficacy of Probiotics, Prebiotic Inulin-Type Fructans, and Synbiotics in Human Ulcerative Colitis: A Systematic Review and Meta-Analysis
by Erola Astó, Iago Méndez, Sergi Audivert, Andreu Farran-Codina and Jordi Espadaler
Nutrients 2019, 11(2), 293; https://doi.org/10.3390/nu11020293 - 30 Jan 2019
Cited by 76 | Viewed by 10028
Abstract
Studies of probiotics, fructan-type prebiotics, and synbiotics in patients with ulcerative colitis (UC) show significant heterogeneity in methodology and results. Here, we study the efficacy of such interventions and the reasons for the heterogeneity of their results. Eligible random controlled trials were collected [...] Read more.
Studies of probiotics, fructan-type prebiotics, and synbiotics in patients with ulcerative colitis (UC) show significant heterogeneity in methodology and results. Here, we study the efficacy of such interventions and the reasons for the heterogeneity of their results. Eligible random controlled trials were collected from the PUBMED and SCOPUS databases. A total of 18 placebo-controlled and active treatment-controlled (i.e., mesalazine) studies were selected with a Jadad score ≥ 3, including 1491 patients with UC. Data for prebiotics and synbiotics were sparse and consequently these studies were excluded from the meta-analysis. The UC remission efficacy of probiotics was measured in terms of relative risk (RR) and odds ratio (OR). Significant effects were observed in patients with active UC whenever probiotics containing bifidobacteria were used, or when adopting the US Food and Drug Administration (FDA)-recommended scales (UC Disease Activity Index and Disease Activity Index). By the FDA recommended scales, the RR was 1.55 (CI95%: 1.13–2.15, p-value = 0.007, I2 = 29%); for bifidobacteria-containing probiotics, the RR was 1.73 (CI95%: 1.23–2.43, p-value = 0.002, I2 = 35%). No significant effects were observed on the maintenance of remission for placebo-controlled or mesalazine-controlled studies. We conclude that a validated scale is necessary to determine the state of patients with UC. However, probiotics containing bifidobacteria are promising for the treatment of active UC. Full article
(This article belongs to the Special Issue Health Benefits of Traditional and Next Generation Probiotics)
Show Figures

Figure 1

Back to TopTop