nutrients-logo

Journal Browser

Journal Browser

Prebiotics and Probiotics in the Management of Chronic Inflammatory Diseases

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

Deadline for manuscript submissions: closed (31 January 2021) | Viewed by 51933

Special Issue Editor


E-Mail Website
Guest Editor
Center for Infection and Immunity of Lille, Institut Pasteur de Lille, INSERM U1019-CNRS UMR9017; 1 rue du Pr Calmette ; 59019 LILLE - FRANCE
Interests: Gut microbiota; probiotics; inflammatory bowel diseases; metabolic diseases; immune regulation; microbial metabolites

Special Issue Information

Dear Colleagues,

The mammalian gut is densely colonized by a complex and dynamic microbial ecosystem, composed of archeae, eukaryotes, viruses, and mainly bacteria. This gut microbiota plays a crucial role in shaping the host physiology, not only at the metabolic level but also by educating and regulating the immune system and protecting the gut barrier from invading pathogens. Even though the gut microbiota is highly resilient under physiological conditions, it can be altered temporarily or permanently, a process called dysbiosis, consequently disturbing intestinal homeostasis. Alterations in microbiota composition together with reduced bacterial diversity are frequently related to the development of intra- and extra-intestinal chronic inflammatory diseases, such as inflammatory bowel disease (IBD), obesity, type 1 and 2 diabetes, liver diseases, cancer, and allergy and neuronal disorders. Therefore, targeting microbiota dysbiosis is offering novel and attractive therapeutic approaches. With this Special Issue, we aim to provide an overview of the recent advances in the research concerning the use of prebiotics, which are non-digestible substances able to favor the growth and/or activity of health-promoting bacteria, and of probiotics, which are defined as “live microorganisms that when administered in adequate amounts confer a health benefit on the host”, in the management of inflammatory disorders. This will allow us to discuss their impact on the regulation of host physiology processes and, notably, their abilities to regulate immune responses and metabolic homeostasis and also neuro-inflammation and lung inflammation by acting on the gut–brain and gut–lung axes, respectively. Finally, we will present how the gut microbiota per se is now envisaged as a source of novel health-promoting bacteria and how the combination of pro- and pre-biotics, as potential synbiotics, is able to provide synergistic effects. It will be important to decipher the mechanisms involved and to identify potential bacterial metabolites. This could pave the way for the development of potential postbiotics able to promote beneficial biological activities and to bypass the difficulties in the regulatory issues of health claim applications of such nutritional intervention.

Despite numerous studies that have highlighted interesting results with such microbiota-targeting therapeutic approaches, major concerns remain to be addressed regarding the appropriate therapy and the selection criteria according to the host physiology, the targeted diseases, the impact of the diet, and the endogenous microbiota composition, leading to the concept of personalized therapy. Original research articles and reviews (systematic reviews and meta-analyses) are welcome.

Dr. Corinne Grangette
Guest Editor

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

  • prebiotics
  • probiotics
  • symbiotics
  • postbiotics
  • live biotherapeutics
  • gut microbiota
  • chronic inflammatory diseases
  • dysbiosis
  • immune and metabolic homeostasis
  • metabolites
  • nutrition

Published Papers (9 papers)

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

Research

Jump to: Review

14 pages, 1803 KiB  
Article
Anti-Inflammatory and Immune Modulatory Effects of Synbio-Glucan in an Atopic Dermatitis Mouse Model
by Yoon-Hwan Kim, Min Soo Kang, Tae Hyeong Kim, Yunho Jeong, Jin-Ok Ahn, Jung Hoon Choi and Jin-Young Chung
Nutrients 2021, 13(4), 1090; https://doi.org/10.3390/nu13041090 - 26 Mar 2021
Cited by 3 | Viewed by 2417
Abstract
Many trials have been conducted to treat atopic dermatitis (AD), but these therapies are generally unsuccessful because of their insufficiency or side effects. This study examined the efficacy of β-glucan derived from oats with fermented probiotics (called Synbio-glucan) on an AD-induced mouse model. [...] Read more.
Many trials have been conducted to treat atopic dermatitis (AD), but these therapies are generally unsuccessful because of their insufficiency or side effects. This study examined the efficacy of β-glucan derived from oats with fermented probiotics (called Synbio-glucan) on an AD-induced mouse model. For the experiment, Nc/Nga mice were exposed to a house dust mite extract (HDM) to induce AD. The mice were placed in one of four groups: positive control group, Synbio-glucan topical treatment group, Synbio-glucan dietary treatment group, and Synbio-glucan topical + dietary treatment group. The experiment revealed no significant difference in the serum IgE concentration among the groups. Serum cytokine antibody arrays showed that genes related to the immune response were enriched. A significant difference in the skin lesion scores was observed between the groups. Compared to the control group tissue, skin lesions were alleviated in the Synbio-glucan topical treatment group and Synbio-glucan dietary treatment group. Interestingly, almost normal structures were observed within the skin lesions in the Synbio-glucan topical + dietary treatment group. Overall, the β-glucan extracted from oats and fermented probiotic mixture is effective in treating atopic dermatitis. Full article
Show Figures

Figure 1

16 pages, 854 KiB  
Article
Impact of 2′-Fucosyllactose on Gut Microbiota Composition in Adults with Chronic Gastrointestinal Conditions: Batch Culture Fermentation Model and Pilot Clinical Trial Findings
by Jennifer Joan Ryan, Andrea Monteagudo-Mera, Nikhat Contractor and Glenn R. Gibson
Nutrients 2021, 13(3), 938; https://doi.org/10.3390/nu13030938 - 14 Mar 2021
Cited by 20 | Viewed by 6555
Abstract
Intestinal dysbiosis has been described in patients with certain gastrointestinal conditions including irritable bowel syndrome (IBS) and ulcerative colitis. 2′-fucosyllactose (2′-FL), a prebiotic human milk oligosaccharide, is considered bifidogenic and butyrogenic. To assess prebiotic effects of 2′-FL, alone or in combination with probiotic [...] Read more.
Intestinal dysbiosis has been described in patients with certain gastrointestinal conditions including irritable bowel syndrome (IBS) and ulcerative colitis. 2′-fucosyllactose (2′-FL), a prebiotic human milk oligosaccharide, is considered bifidogenic and butyrogenic. To assess prebiotic effects of 2′-FL, alone or in combination with probiotic strains (potential synbiotics), in vitro experiments were conducted on stool from healthy, IBS, and ulcerative colitis adult donors. In anaerobic batch culture fermenters, Bifidobacterium and Eubacterium rectale-Clostridium coccoides counts, and short-chain fatty acids (SCFAs) including butyrate increased during fermentation with 2′-FL and some of the 2′-FL/probiotic combinations. In a subsequent open-label pilot trial, the effect of a 2′-FL-containing nutritional formula was evaluated in twelve adults with IBS or ulcerative colitis. Gastrointestinal Quality of Life Index (GIQLI) total and gastrointestinal symptoms domain scores, stool counts of Bifidobacterium and Faecalibacterium prausnitzii, and stool SCFAs including butyrate, increased after six weeks of intervention. Consistent with documented effects of 2′-FL, the batch culture fermentation experiments demonstrated bifidogenic and butyrogenic effects of 2′-FL during fermentation with human stool samples. Consumption of the 2′-FL-containing nutritional formula by adults with IBS or ulcerative colitis was associated with improvements in intra- and extra-intestinal symptoms, and bifidogenic and butyrogenic effects. Full article
Show Figures

Figure 1

16 pages, 1431 KiB  
Article
The Effectiveness and Safety of Multi-Strain Probiotic Preparation in Patients with Diarrhea-Predominant Irritable Bowel Syndrome: A Randomized Controlled Study
by Barbara Skrzydło-Radomańska, Beata Prozorow-Król, Halina Cichoż-Lach, Emilia Majsiak, Joanna Beata Bierła, Ewelina Kanarek, Agnieszka Sowińska and Bożena Cukrowska
Nutrients 2021, 13(3), 756; https://doi.org/10.3390/nu13030756 - 26 Feb 2021
Cited by 39 | Viewed by 5935
Abstract
The aim of this randomized double-blind placebo-controlled study was to evaluate the effectiveness and safety of multi-strain probiotic in adults with diarrhea-predominant irritable bowel syndrome (IBS-D). The patients were randomized to receive a mixture of Lactobacillus, Bifidobacterium, and Streptococcus thermophilus strains [...] Read more.
The aim of this randomized double-blind placebo-controlled study was to evaluate the effectiveness and safety of multi-strain probiotic in adults with diarrhea-predominant irritable bowel syndrome (IBS-D). The patients were randomized to receive a mixture of Lactobacillus, Bifidobacterium, and Streptococcus thermophilus strains or placebo for eight weeks. Primary endpoints included changes in symptom severity and improvement assessed with the IBS Severity Scoring System (IBS-SSS) and Global Improvement Scale (IBS-GIS). The probiotic in comparison with placebo significantly improved the IBS symptom severity (the change of total IBS-SSS score from baseline −165.8 ± 78.9 in the probiotic group and −105.6 ± 60.2 in the placebo group, p = 0.005) and in the specific scores related to the severity of pain (p = 0.015) and the quality of life (p = 0.016) after eight weeks of intervention. The probiotic group indicated an improvement in symptoms with the use of the IBS-GIS compared with the placebo group after four (p = 0.04) and eight weeks (p = 0.003). The occurrence of adverse events did not differ between study groups. In conclusion, the multi-strain probiotic intervention resulted in a significant improvement in IBS symptoms evaluated with the use of both IBS-SSS and IBS-GIS scales. The results suggest that the studied probiotic preparation is well tolerated and safe and can offer benefits for patients with IBS-D. (registration number in Clinicaltrials.gov NCT 04662957). Full article
Show Figures

Figure 1

23 pages, 4715 KiB  
Article
Multiple Selection Criteria for Probiotic Strains with High Potential for Obesity Management
by Jeanne Alard, Benoit Cudennec, Denise Boutillier, Véronique Peucelle, Amandine Descat, Raphaël Decoin, Sarah Kuylle, Amin Jablaoui, Moez Rhimi, Isabelle Wolowczuk, Bruno Pot, Anne Tailleux, Emmanuelle Maguin, Sophie Holowacz and Corinne Grangette
Nutrients 2021, 13(3), 713; https://doi.org/10.3390/nu13030713 - 24 Feb 2021
Cited by 24 | Viewed by 3561
Abstract
Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We [...] Read more.
Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action. Full article
Show Figures

Graphical abstract

19 pages, 963 KiB  
Article
Effects of Synbiotic Supplementation on Chronic Inflammation and the Gut Microbiota in Obese Patients with Type 2 Diabetes Mellitus: A Randomized Controlled Study
by Akio Kanazawa, Masanori Aida, Yasuto Yoshida, Hideyoshi Kaga, Takehiro Katahira, Luka Suzuki, Shoko Tamaki, Junko Sato, Hiromasa Goto, Kosuke Azuma, Tomoaki Shimizu, Takuya Takahashi, Yuichiro Yamashiro and Hirotaka Watada
Nutrients 2021, 13(2), 558; https://doi.org/10.3390/nu13020558 - 8 Feb 2021
Cited by 54 | Viewed by 7215
Abstract
The aim of this study was to investigate the effects of 24-week synbiotic supplementation on chronic inflammation and the gut microbiota in obese patients with type 2 diabetes. We randomized 88 obese patients with type 2 diabetes to one of two groups for [...] Read more.
The aim of this study was to investigate the effects of 24-week synbiotic supplementation on chronic inflammation and the gut microbiota in obese patients with type 2 diabetes. We randomized 88 obese patients with type 2 diabetes to one of two groups for 24 weeks: control or synbiotic (Lacticaseibacillus paracasei strain Shirota (previously Lactobacillus casei strain Shirota) and Bifidobacterium breve strain Yakult, and galactooligosaccharides). The primary endpoint was the change in interleukin-6 from baseline to 24 weeks. Secondary endpoints were evaluation of the gut microbiota in feces and blood, fecal organic acids, high-sensitivity C-reactive protein, lipopolysaccharide-binding protein, and glycemic control. Synbiotic administration for 24 weeks did not significantly affect changes in interleukin-6 from baseline to 24 weeks (0.35 ± 1.99 vs. −0.24 ± 1.75 pg/mL, respectively). Relative to baseline, however, at 24 weeks after synbiotic administration there were positive changes in the counts of Bifidobacterium and total lactobacilli, the relative abundances of Bifidobacterium species such as Bifidobacterium adolescentis and Bifidobacterium pseudocatenulatum, and the concentrations of acetic and butyric acids in feces. No significant changes in inflammatory markers were found in the synbiotic group compared to the control group. However, synbiotic administration at least partially improved the gut environment in obese patients with type 2 diabetes. Full article
Show Figures

Figure 1

10 pages, 876 KiB  
Article
Clinical Significance of Probiotics for Children with Idiopathic Nephrotic Syndrome
by Tadashi Yamaguchi, Shoji Tsuji, Shohei Akagawa, Yuko Akagawa, Jiro Kino, Sohsaku Yamanouchi, Takahisa Kimata, Masaki Hashiyada, Atsushi Akane and Kazunari Kaneko
Nutrients 2021, 13(2), 365; https://doi.org/10.3390/nu13020365 - 26 Jan 2021
Cited by 18 | Viewed by 4109
Abstract
We previously reported that a decrease in butyrate-producing bacteria in the gut is a potential cause of regulatory T cell (Treg) abnormalities in children with idiopathic nephrotic syndrome (INS). Therefore, we hypothesized that administration of butyrate-producing bacteria might reduce INS relapse and the [...] Read more.
We previously reported that a decrease in butyrate-producing bacteria in the gut is a potential cause of regulatory T cell (Treg) abnormalities in children with idiopathic nephrotic syndrome (INS). Therefore, we hypothesized that administration of butyrate-producing bacteria might reduce INS relapse and the need for immunosuppressants in these patients. Twenty patients in remission from INS (median age 5.3 years, 15 boys) were enrolled in the study and assigned to receive either daily oral treatment with a preparation of 3 g Clostridium butyricum or no probiotic treatment. The number of relapses and requirement for immunosuppressive agents were compared between the two groups. In the probiotic treatment group, analyses of the gut microbiota and Treg measurements were also performed. Probiotic-treated patients experienced fewer INS relapses per year compared with non-probiotic-treated patients (p = 0.016). Further, administration of rituximab in the probiotic treatment group was significantly less frequent compared with the non-probiotic-treated group (p = 0.025). In the probiotic treatment group, analyses before and after probiotic treatment revealed the significant increases in the relative abundance of butyrate-producing bacteria (p = 0.017) and blood Treg counts (p = 0.0065). Thus, oral administration of butyrate-producing bacteria during INS remission may reduce the frequency of relapse and the need for immunosuppressive agents. Full article
Show Figures

Graphical abstract

11 pages, 1152 KiB  
Article
Effects of Multi-Strain Probiotics on Immune Responses and Metabolic Balance in Helicobacter pylori-Infected Mice
by Chun-Che Lin, Wei-Chiao Huang, Chiu-Hsian Su, Wei-De Lin, Wen-Tzu Wu, Bi Yu and Yuan-Man Hsu
Nutrients 2020, 12(8), 2476; https://doi.org/10.3390/nu12082476 - 17 Aug 2020
Cited by 22 | Viewed by 4329
Abstract
Chronic inflammation caused by Helicobacter pylori infection increases the risk of developing gastric cancer. Even though the prevalence of H. pylori infection has been decreased in many regions, the development of antibiotic resistance strains has increased the difficulty of eradicating H. pylori. [...] Read more.
Chronic inflammation caused by Helicobacter pylori infection increases the risk of developing gastric cancer. Even though the prevalence of H. pylori infection has been decreased in many regions, the development of antibiotic resistance strains has increased the difficulty of eradicating H. pylori. Therefore, exploring alternative approaches to combat H. pylori infection is required. It is well-known that probiotic therapy can improve H. pylori clearance. In this study, H. pylori-infected mice were treated with Lactobacillus fermentum P2 (P2), L. casei L21 (L21), L. rhamnosus JB3 (JB3), or a mixture including the aforementioned three (multi-LAB) for three days. All the lactic acid producing bacteria (LAB) treatments decreased H. pylori loads in the stomach and vacA gene expression, H. pylori specific immunoglobulin (Ig) A, and IgM levels in stomach homogenates, as well as serum levels of interferon-gamma and interleukin-1 beta. The multi-LAB and JB3 treatments further restored the superoxide dismutase and catalase activities suppressed by H. pylori infection. Furthermore, H. pylori infection decreased serum concentrations of 15 kinds of amino acids as well as palmitic acid. The multi-LAB treatment was able to recover the serum levels of alanine, arginine, aspartate, glycine, and tryptophan, which are all important in modulating immune functions. In addition, butyric acid, valeric acid, palmitic acid, palmitoleic acid, stearic acid, and oleic acid levels were increased. In this study, multi-LAB revealed its ability to adjust the composition of metabolites to improve health. To date, the mechanisms underlying how LAB strains crosstalk with the host are not fully understood. Identifying the mechanisms which are regulated by LABs will facilitate the development of effective therapies for infection in the future. Full article
Show Figures

Figure 1

Review

Jump to: Research

21 pages, 1130 KiB  
Review
Evidence for the Contribution of Gut Microbiota to Age-Related Anabolic Resistance
by Matthew D. Watson, Brett L. Cross and Gregory J. Grosicki
Nutrients 2021, 13(2), 706; https://doi.org/10.3390/nu13020706 - 23 Feb 2021
Cited by 18 | Viewed by 4994
Abstract
Globally, people 65 years of age and older are the fastest growing segment of the population. Physiological manifestations of the aging process include undesirable changes in body composition, declines in cardiorespiratory fitness, and reductions in skeletal muscle size and function (i.e., sarcopenia) that [...] Read more.
Globally, people 65 years of age and older are the fastest growing segment of the population. Physiological manifestations of the aging process include undesirable changes in body composition, declines in cardiorespiratory fitness, and reductions in skeletal muscle size and function (i.e., sarcopenia) that are independently associated with mortality. Decrements in muscle protein synthetic responses to anabolic stimuli (i.e., anabolic resistance), such as protein feeding or physical activity, are highly characteristic of the aging skeletal muscle phenotype and play a fundamental role in the development of sarcopenia. A more definitive understanding of the mechanisms underlying this age-associated reduction in anabolic responsiveness will help to guide promyogenic and function promoting therapies. Recent studies have provided evidence in support of a bidirectional gut-muscle axis with implications for aging muscle health. This review will examine how age-related changes in gut microbiota composition may impact anabolic response to protein feeding through adverse changes in protein digestion and amino acid absorption, circulating amino acid availability, anabolic hormone production and responsiveness, and intramuscular anabolic signaling. We conclude by reviewing literature describing lifestyle habits suspected to contribute to age-related changes in the microbiome with the goal of identifying evidence-informed strategies to preserve microbial homeostasis, anabolic sensitivity, and skeletal muscle with advancing age. Full article
Show Figures

Graphical abstract

20 pages, 1051 KiB  
Review
Gut Hormones in Health and Obesity: The Upcoming Role of Short Chain Fatty Acids
by Habeeb Alhabeeb, Ali AlFaiz, Emad Kutbi, Dayel AlShahrani, Abdullah Alsuhail, Saleh AlRajhi, Nemer Alotaibi, Khalid Alotaibi, Saad AlAmri, Saleh Alghamdi and Naji AlJohani
Nutrients 2021, 13(2), 481; https://doi.org/10.3390/nu13020481 - 31 Jan 2021
Cited by 38 | Viewed by 11428
Abstract
We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action [...] Read more.
We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action towards understanding the underlying mechanisms behind the development of obesity as well as developing more effective treatments and interventions. Appetite is carefully regulated in humans via the interaction between the central nervous system and peripheral hormones. This involves a delicate balance in external stimuli, circulating satiating and appetite stimulating hormones, and correct functioning of neuronal signals. Any changes in this equilibrium can lead to an imbalance in energy intake versus expenditure, which often leads to overeating, and potentially weight gain resulting in overweight or obesity. Several lines of research have shown imbalances in gut hormones are found in those who are overweight or obese, which may be contributing to their condition. Therefore, this review examines the evidence for targeting gut hormones in the treatment of obesity by discussing how their dysregulation influences food intake, the potential possibility of altering the circulating levels of these hormones for treating obesity, as well as the role of short chain fatty acids and protein as novel treatments. Full article
Show Figures

Graphical abstract

Back to TopTop