Gastrointestinal Microbiota and Gut Barrier Impact Human Health and Disease

A special issue of Microorganisms (ISSN 2076-2607). This special issue belongs to the section "Gut Microbiota".

Deadline for manuscript submissions: closed (15 February 2022) | Viewed by 117974

Special Issue Editor


E-Mail Website
Guest Editor
1. Department of Medicine and Translational Surgery, Catholic University of the Sacred Heart, Largo F. Vito 1, 00168 Rome, Italy
2. UOC Nutrizione Clinica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy
Interests: perioperative nutrition; ERAS (Enhanced Recovery After Surgery) programs; nutrition in cancer patients; disease-related malnutrition; gut microbiota and diet; sports nutrition
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues, 

Recent observational and intervention studies indicate that the gut microbiota, which is the community of microorganisms living in the gastrointestinal tract and a component of the gut barrier, is one of the key regulators of many physiological processes and pathological conditions of the human body.  

As we know, physiological conditions (pregnancy, delivery, weaning, aging, etc.) and several interventions may modify the complex interaction between gut-inhabitant flora (microbes, viruses, fungi, etc.) and the gut barrier in health and disease, like dietary regimens and diet components, the administration of drugs (particularly antibiotics), and recently, fecal microbiota transplantation, coadjuvant therapy in life-threatening infectious conditions.  

I kindly invite you to contribute to this Special Issue in Microorganisms, aiming to broadly cover gut microbiota/gut barrier interactions in maintaining health, and to outline their possible role in inflammatory, autoimmune, oncological, and neurodegenerative diseases. 

Dr. Maria Cristina Mele
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. Microorganisms is an international peer-reviewed open access monthly 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 2700 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

  • gut microbiota
  • gut barrier
  • neurodegenerative diseases
  • IBD
  • obesity
  • health
  • autoimmune diseases
  • cancer

Published Papers (25 papers)

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

Editorial

Jump to: Research, Review

4 pages, 206 KiB  
Editorial
Special Issue “Gastrointestinal Microbiota and Gut Barrier Impact Human Health and Disease”: Editorial
by Pauline Raoul, Marco Cintoni, Emanuele Rinninella and Maria Cristina Mele
Microorganisms 2023, 11(4), 985; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms11040985 - 10 Apr 2023
Viewed by 1130
Abstract
The increasing incidence of non-communicable diseases is a worldwide public health issue, and the role of gut microbiota is becoming evident [...] Full article

Research

Jump to: Editorial, Review

18 pages, 3223 KiB  
Article
Early Antibiotic Exposure Alters Intestinal Development and Increases Susceptibility to Necrotizing Enterocolitis: A Mechanistic Study
by Hala Chaaban, Maulin M. Patel, Kathryn Burge, Jeffrey V. Eckert, Cristina Lupu, Ravi S. Keshari, Robert Silasi, Girija Regmi, MaJoi Trammell, David Dyer, Steven J. McElroy and Florea Lupu
Microorganisms 2022, 10(3), 519; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms10030519 - 27 Feb 2022
Cited by 17 | Viewed by 3032
Abstract
Increasing evidence suggests that prolonged antibiotic therapy in preterm infants is associated with increased mortality and morbidities, such as necrotizing enterocolitis (NEC), a devastating gastrointestinal pathology characterized by intestinal inflammation and necrosis. While a clinical correlation exists between antibiotic use and the development [...] Read more.
Increasing evidence suggests that prolonged antibiotic therapy in preterm infants is associated with increased mortality and morbidities, such as necrotizing enterocolitis (NEC), a devastating gastrointestinal pathology characterized by intestinal inflammation and necrosis. While a clinical correlation exists between antibiotic use and the development of NEC, the potential causality of antibiotics in NEC development has not yet been demonstrated. Here, we tested the effects of systemic standard-of-care antibiotic therapy for ten days on intestinal development in neonatal mice. Systemic antibiotic treatment impaired the intestinal development by reducing intestinal cell proliferation, villi height, crypt depth, and goblet and Paneth cell numbers. Oral bacterial challenge in pups who received antibiotics resulted in NEC-like intestinal injury in more than half the pups, likely due to a reduction in mucous-producing cells affecting microbial–epithelial interactions. These data support a novel mechanism that could explain why preterm infants exposed to prolonged antibiotics after birth have a higher incidence of NEC and other gastrointestinal disorders. Full article
Show Figures

Figure 1

12 pages, 3027 KiB  
Article
The Gut Microbiome Alterations in Pediatric Patients with Functional Abdominal Pain Disorders
by Bassam Abomoelak, Veronica Pemberton, Chirajyoti Deb, Stephani Campion, Michelle Vinson, Jennifer Mauck, Joseph Manipadam, Sailendharan Sudakaran, Samit Patel, Miguel Saps, Hesham A. El Enshasy, Theodoros Varzakas and Devendra I. Mehta
Microorganisms 2021, 9(11), 2354; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9112354 - 15 Nov 2021
Cited by 8 | Viewed by 2556
Abstract
In this prospective longitudinal study, we enrolled 54 healthy pediatric controls and 28 functional abdominal pain disorders (FAPDs) pediatric patients (mean age was 11 ± 2.58 years old). Fecal samples and symptom questionnaires were obtained from all participants over the course of the [...] Read more.
In this prospective longitudinal study, we enrolled 54 healthy pediatric controls and 28 functional abdominal pain disorders (FAPDs) pediatric patients (mean age was 11 ± 2.58 years old). Fecal samples and symptom questionnaires were obtained from all participants over the course of the year. Clinical data assessment showed that FAPDs patients were more symptomatic than the control group. Microbiome analysis revealed that Phylum Bacteroidetes was higher in FAPDs compared to the control group (p < 0.05), while phylum Firmicutes was lower in FAPDs (p < 0.05). In addition, Verrucomicrobiota was higher in the control group than the FAPDs (p < 0.05). At the genus level the relative abundance of 72 bacterial taxa showed statistically significant differences between the two groups and at the school term levels. In the control group, Shannon diversity, Observed_species, and Simpson were higher than the FAPDs (p < 0.05), and beta diversity showed differences between the two groups (PERMANOVA = 2.38; p = 0.002) as well. Using linear discriminant analysis effect size (LEfSe), Enterobacteriaceae family and Megaspherae showed increased abundances in vacation term (LDA score > 2.0, LEfSe, p < 0.05). In the FAPDs group, the severity of symptoms (T-scores) correlated with 11 different taxa bacterial relative abundances using Pearson′s correlation and linear regression analyses. Our data showed that gut microbiome is altered in FAPDs compared to the control. Differences in other metrics such as alpha- and beta diversity were also reported between the two groups. Correlation of the severity of the disease (T-scores) correlated with gut microbiome. Finally, our findings support the use of Faecalibacterium/Bacteroides ratio as a potential diagnostic biomarker for FAPDs. Full article
Show Figures

Figure 1

17 pages, 1369 KiB  
Article
Fermentation Supernatants of Pleurotus eryngii Mushroom Ameliorate Intestinal Epithelial Barrier Dysfunction in Lipopolysaccharide-Induced Caco-2 Cells via Upregulation of Tight Junctions
by Georgia Saxami, Evangelia N. Kerezoudi, Evdokia K. Mitsou, Georgios Koutrotsios, Georgios I. Zervakis, Vasiliki Pletsa and Adamantini Kyriacou
Microorganisms 2021, 9(10), 2071; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9102071 - 01 Oct 2021
Cited by 4 | Viewed by 2422
Abstract
In recent years, modulation of gut microbiota through prebiotics has garnered interest as a potential to ameliorate intestinal barrier dysfunction. The aim of the study was to examine the in vitro effect of fermentation supernatants (FSs) from rich in β-glucan Pleurotus eryngii mushrooms [...] Read more.
In recent years, modulation of gut microbiota through prebiotics has garnered interest as a potential to ameliorate intestinal barrier dysfunction. The aim of the study was to examine the in vitro effect of fermentation supernatants (FSs) from rich in β-glucan Pleurotus eryngii mushrooms on the expression levels of tight junctions (TJs) genes in Caco-2 cells stimulated by bacterial lipopolysaccharides (LPS). Mushrooms were fermented using fecal inocula in an in vitro batch culture model. Caco-2 cells were subjected to LPS and FS treatment under three different conditions: pre-incubation with FS, co- and post-incubation. Reverse transcription PCR was applied to measure the expression levels of zonulin-1, occludin and claudin-1 genes. FSs from P. eryngii mushrooms led to a significant upregulation of the TJs gene expression in pre-incubation state, indicating potential preventive action. Down-regulation of all TJs gene expression levels was observed when the cells were challenged with LPS. The FS negative control (gut microbiota of each donor with no carbohydrate source) exhibited a significant upregulation of TJs expression levels compared to the cells that were challenged with LPS, for all three conditions. Overall, our data highlighted the positive and potential protective effects of P. eryngii mushrooms in upregulation of TJs’ genes. Full article
Show Figures

Figure 1

15 pages, 406 KiB  
Article
Descriptive Study of Gut Microbiota in Infected and Colonized Subjects by Clostridiodes difficile
by Pedro Sánchez-Pellicer, Vicente Navarro-López, Ruth González-Tamayo, Coral Llopis-Ruiz, Eva Núñez-Delegido, Beatriz Ruzafa-Costas, Laura Navarro-Moratalla and Juan Agüera-Santos
Microorganisms 2021, 9(8), 1727; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9081727 - 13 Aug 2021
Cited by 3 | Viewed by 2047
Abstract
Clostridiodes difficile can lead to a range of situations from the absence of symptoms (colonization) to severe diarrhea (infection). Disruption of gut microbiota provides an ideal environment for infection to occur. Comparison of gut microbiota of infected and colonized subjects could provide relevant [...] Read more.
Clostridiodes difficile can lead to a range of situations from the absence of symptoms (colonization) to severe diarrhea (infection). Disruption of gut microbiota provides an ideal environment for infection to occur. Comparison of gut microbiota of infected and colonized subjects could provide relevant information on susceptible groups or protectors to the development of infection, since the presence of certain genera could be related to the inhibition of transition from a state of colonization to infection. Through high-throughput sequencing of 16S rDNA gene, we performed alpha and beta diversity and composition studies on 15 infected patients (Group CDI), 15 colonized subjects (Group P), and 15 healthy controls (Group CTLR). A loss of alpha diversity and richness and a different structure have been evidenced in the CDI and P groups with respect to the CTRL group, but without significant differences between the first two. In CDI and P groups, there was a strong decrease in phylum Firmicutes and an expansion of potential pathogens. Likewise, there was a loss of inhibitory genus of C. difficile germination in infected patients that were partially conserved in colonized subjects. Therefore, infected and colonized subjects presented a gut microbiota that was completely different from that of healthy controls, although similar to each other. It is in composition where we found that colonized subjects, especially in minority genera, presented differences with respect to those infected. Full article
Show Figures

Figure 1

20 pages, 2561 KiB  
Article
Fecal Supernatant from Adult with Autism Spectrum Disorder Alters Digestive Functions, Intestinal Epithelial Barrier, and Enteric Nervous System
by Jacques Gonzales, Justine Marchix, Laetitia Aymeric, Catherine Le Berre-Scoul, Johanna Zoppi, Philippe Bordron, Marie Burel, Laetitia Davidovic, Jean-Romain Richard, Alexandru Gaman, Florian Lejuste, Julie Z. Brouillet, Françoise Le Vacon, Samuel Chaffron, Marion Leboyer, Hélène Boudin and Michel Neunlist
Microorganisms 2021, 9(8), 1723; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9081723 - 13 Aug 2021
Cited by 12 | Viewed by 4117
Abstract
Autism Spectrum Disorders (ASDs) are neurodevelopmental disorders defined by impaired social interactions and communication with repetitive behaviors, activities, or interests. Gastrointestinal (GI) disturbances and gut microbiota dysbiosis are frequently associated with ASD in childhood. However, it is not known whether microbiota dysbiosis in [...] Read more.
Autism Spectrum Disorders (ASDs) are neurodevelopmental disorders defined by impaired social interactions and communication with repetitive behaviors, activities, or interests. Gastrointestinal (GI) disturbances and gut microbiota dysbiosis are frequently associated with ASD in childhood. However, it is not known whether microbiota dysbiosis in ASD patients also occurs in adulthood. Further, the consequences of altered gut microbiota on digestive functions and the enteric nervous system (ENS) remain unexplored. Therefore, we studied, in mice, the ability offecal supernatant (FS) from adult ASD patients to induce GI dysfunctions and ENS remodeling. First, the analyses of the fecal microbiota composition in adult ASD patients indicated a reduced α-diversity and increased abundance of three bacterial 16S rRNA gene amplicon sequence variants compared to healthy controls (HC). The transfer of FS from ASD patients (FS–ASD) to mice decreased colonic barrier permeability by 29% and 58% compared to FS–HC for paracellular and transcellular permeability, respectively. These effects are associated with the reduced expression of the tight junction proteins JAM-A, ZO-2, cingulin, and proinflammatory cytokines TNFα and IL1β. In addition, the expression of glial and neuronal molecules was reduced by FS–ASD as compared to FS-HC in particular for those involved in neuronal connectivity (βIII-tubulin and synapsin decreased by 31% and 67%, respectively). Our data suggest that changes in microbiota composition in ASD may contribute to GI alterations, and in part, via ENS remodeling. Full article
Show Figures

Figure 1

13 pages, 1296 KiB  
Article
Colonization with Altered Schaedler Flora Impacts Leukocyte Adhesion in Mesenteric Ischemia-Reperfusion Injury
by Franziska Bayer, Stefanie Ascher, Klytaimnistra Kiouptsi, Jens M. Kittner, Roland H. Stauber and Christoph Reinhardt
Microorganisms 2021, 9(8), 1601; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9081601 - 27 Jul 2021
Cited by 12 | Viewed by 1932
Abstract
The microbiota impacts mesenteric ischemia-reperfusion injury, aggravating the interaction of leukocytes with endothelial cells in mesenteric venules. The role of defined gut microbiomes in this life-threatening pathology is unknown. To investigate how a defined model microbiome affects the adhesion of leukocytes in mesenteric [...] Read more.
The microbiota impacts mesenteric ischemia-reperfusion injury, aggravating the interaction of leukocytes with endothelial cells in mesenteric venules. The role of defined gut microbiomes in this life-threatening pathology is unknown. To investigate how a defined model microbiome affects the adhesion of leukocytes in mesenteric ischemia-reperfusion, we took advantage of gnotobiotic isolator technology and transferred altered Schaedler flora (ASF) from C3H/HeNTac to germ-free C57BL/6J mice. We were able to detect all eight bacterial taxa of ASF in fecal samples of colonized C57BL/6J mice by PCR. Applying qRT-PCR for quantification of species-specific 16S rDNA sequences of ASF bacteria, we found a major shift in the abundance of ASF 500, which was greater in C57BL/6J mice relative to the C3H/HeNTac founder breeding pair. Using high-speed epifluorescence intravital microscopy to visualize the venules of the small bowel mesentery, we found that gnotobiotic ASF-colonized mice showed reduced leukocyte adherence, both pre- and post-ischemia. Relative to germ-free mice, the counts of adhering leukocytes were increased pre-ischemia but did not significantly increase in ASF-colonized mice in the post-ischemic state. Collectively, our results suggest a protective role of the minimal microbial consortium ASF in mesenteric ischemia-reperfusion injury. Full article
Show Figures

Figure 1

18 pages, 1750 KiB  
Article
Composition and Functions of the Gut Microbiome in Pediatric Obesity: Relationships with Markers of Insulin Resistance
by Camila E. Orsso, Ye Peng, Edward C. Deehan, Qiming Tan, Catherine J. Field, Karen L. Madsen, Jens Walter, Carla M. Prado, Hein M. Tun and Andrea M. Haqq
Microorganisms 2021, 9(7), 1490; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9071490 - 13 Jul 2021
Cited by 15 | Viewed by 4563
Abstract
The gut microbiome is hypothesized to play a crucial role in the development of obesity and insulin resistance (IR); the pathways linking the microbiome to IR in pediatrics have yet to be precisely characterized. We aimed to determine the relationship between the gut [...] Read more.
The gut microbiome is hypothesized to play a crucial role in the development of obesity and insulin resistance (IR); the pathways linking the microbiome to IR in pediatrics have yet to be precisely characterized. We aimed to determine the relationship between the gut microbiome composition and metabolic functions and IR in children with obesity. In a cross-sectional study, fecal samples from children with obesity (10–16 years old) were collected for taxonomical and functional analysis of the fecal microbiome using shotgun metagenomics. The homeostatic model assessment for insulin resistance (HOMA-IR) was determined using fasting glucose and insulin. Associations between HOMA-IR and α-diversity measures as well as metabolic pathways were evaluated using Spearman correlations; relationships between HOMA-IR and β-diversity were assessed by permutational multivariate analysis of variance. Twenty-one children (nine males; median: age = 12.0 years; BMI z-score = 2.9; HOMA-IR = 3.6) completed the study. HOMA-IR was significantly associated with measures of α-diversity but not with β-diversity. Children with higher HOMA-IR exhibited lower overall species richness, Firmicutes species richness, and overall Proteobacteria species Shannon diversity. Furthermore, HOMA-IR was inversely correlated with the abundance of pathways related to the biosynthesis of lipopolysaccharides, amino acids, and short-chain fatty acids, whereas positive correlations between HOMA-IR and the peptidoglycan biosynthesis pathways were observed. In conclusion, insulin resistance was associated with decreased microbial α-diversity measures and abundance of genes related to the metabolic pathways. Our study provides a framework for understanding the microbial alterations in pediatric obesity. Full article
Show Figures

Figure 1

19 pages, 3408 KiB  
Article
Effects of Wine and Its Microbial-Derived Metabolites on Intestinal Permeability Using Simulated Gastrointestinal Digestion/Colonic Fermentation and Caco-2 Intestinal Cell Models
by Irene Zorraquín-Peña, Diego Taladrid, Alba Tamargo, Mariana Silva, Natalia Molinero, Dolores González de Llano, Begoña Bartolomé and M. Victoria Moreno-Arribas
Microorganisms 2021, 9(7), 1378; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9071378 - 24 Jun 2021
Cited by 15 | Viewed by 3289
Abstract
This paper explores the effects of wine polyphenols on intestinal permeability in in vitro conditions. A red wine (2500 mg/L of gallic acid equivalents) was sequentially subjected to gastrointestinal and colonic digestion in the Dynamic Gastrointestinal Simulator (simgi®) to obtain two [...] Read more.
This paper explores the effects of wine polyphenols on intestinal permeability in in vitro conditions. A red wine (2500 mg/L of gallic acid equivalents) was sequentially subjected to gastrointestinal and colonic digestion in the Dynamic Gastrointestinal Simulator (simgi®) to obtain two simulated fluids: intestinal-digested wine (IDW) and colonic-digested wine (CDW). The two fluids were incubated with Caco-2 cell monolayers grown in Transwell® inserts, and paracellular permeability was measured as transport of FITC-dextran. Non-significant decreases (p > 0.05) in paracellular permeability were found, which was attributed to the relatively low phenolic concentration in the solutions tested (15.6 and 7.8 mg of gallic acid equivalents/L for IDW and CDW, respectively) as quercetin (200 µM) and one of its microbial-derived phenolic metabolites, 3,4-dihydroxyphenylacetic acid (200 µM), led to significant decreases (p < 0.05). The expression of tight junction (TJ) proteins (i.e., ZO-1 and occludin) in Caco-2 cells after incubation with IDW and CDW was also determined. A slight increase in mRNA levels for occludin for both IDW and CDW fluids, albeit without statistical significance (p > 0.05), was observed. Analysis of the microbiome and microbial activity during wine colonic fermentation revealed relevant changes in the relative abundance of some families/genera (i.e., reduction in Bacteroides and an increase in Veillonella, Escherichia/Shigella and Akkermansia) as well as in the microbial production of SCFA (i.e., a significant increase in propionic acid in the presence of IDW), all of which might affect paracellular permeability. Both direct and indirect (microbiota-mediated) mechanisms might be involved in the protective effects of (wine) polyphenols on intestinal barrier integrity. Overall, this paper reinforces (wine) polyphenols as a promising dietary strategy to improve gut functionality, although further studies are needed to evaluate the effect on the intestinal barrier under different conditions. Full article
Show Figures

Figure 1

8 pages, 890 KiB  
Communication
New Variants of Squash Mosaic Viruses Detected in Human Fecal Samples
by Fabiola Villanova, Roberta Marcatti, Mayara Bertanhe, Vanessa dos Santos Morais, Flavio Augusto de Padua Milagres, Rafael Brustulin, Emerson Luiz Lima Araújo, Roozbeh Tahmasebi, Steven S. Witkin, Xutao Deng, Eric Delwart, Ester Cerdeira Sabino, Cassio Hamilton Abreu-Junior, Élcio Leal and Antonio Charlys da Costa
Microorganisms 2021, 9(7), 1349; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9071349 - 22 Jun 2021
Cited by 2 | Viewed by 1993
Abstract
Squash mosaic virus (SqMV) is a phytovirus that infects great diversity of plants worldwide. In Brazil, the SqMV has been identified in the states of Ceará, Maranhão, Piauí, Rio Grande do Norte, and Tocantins. The presence of non-pathogenic viruses in animals, such as [...] Read more.
Squash mosaic virus (SqMV) is a phytovirus that infects great diversity of plants worldwide. In Brazil, the SqMV has been identified in the states of Ceará, Maranhão, Piauí, Rio Grande do Norte, and Tocantins. The presence of non-pathogenic viruses in animals, such as phytoviruses, may not be completely risk-free. Similarities in gene repertories between these viruses and viruses that affect animal species have been reported. The present study describes the fully sequenced genomes of SqMV found in human feces, collected in Tocantins, and analyzes the viral profile by metagenomics in the context of diarrhea symptomatology. The complete SqMV genome was obtained in 39 of 253 analyzed samples (15.5%); 97.4% of them belonged to children under 5 years old. There was no evidence that the observed symptoms were related to the presence of SqMV. Of the different virus species detected in these fecal samples, at least 4 (rotavirus, sapovirus, norovirus, parechovirus) are widely known to cause gastrointestinal symptoms. The presence of SqMV nucleic acid in fecal samples is likely due to recent dietary consumption and it is not evidence of viral replication in the human intestinal cells. Identifying the presence of SqMV in human feces and characterization of its genome is a relevant precursor to determining whether and how plant viruses interact with host cells or microorganisms in the human gastrointestinal tract. Full article
Show Figures

Figure 1

16 pages, 2163 KiB  
Article
Diversity and Adaptations of Escherichia coli Strains: Exploring the Intestinal Community in Crohn’s Disease Patients and Healthy Individuals
by Maria N. Siniagina, Maria I. Markelova, Eugenia A. Boulygina, Alexander V. Laikov, Dilyara R. Khusnutdinova, Sayar R. Abdulkhakov, Natalia A. Danilova, Alfiya H. Odintsova, Rustam A. Abdulkhakov and Tatyana V. Grigoryeva
Microorganisms 2021, 9(6), 1299; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9061299 - 15 Jun 2021
Cited by 5 | Viewed by 2996
Abstract
Crohn’s disease (CD) is characterized by a chronic, progressive inflammation across the gastrointestinal tract with a series of exacerbations and remissions. A significant factor in the CD pathogenesis is an imbalance in gut microbiota composition, particularly the prevalence of Escherichia coli. In [...] Read more.
Crohn’s disease (CD) is characterized by a chronic, progressive inflammation across the gastrointestinal tract with a series of exacerbations and remissions. A significant factor in the CD pathogenesis is an imbalance in gut microbiota composition, particularly the prevalence of Escherichia coli. In the present study, the genomes of sixty-three E. coli strains from the gut of patients with CD and healthy subjects were sequenced. In addition, eighteen E. coli-like metagenome-assembled genomes (MAGs) were reconstructed from the shotgun-metagenome sequencing data of fecal samples. The comparative analysis revealed the similarity of E. coli genomes regardless of the origin of the strain. The strains exhibited similar genetic patterns of virulence, antibiotic resistance, and bacteriocin-producing systems. The study showed antagonistic activity of E. coli strains and the metabolic features needed for their successful competition in the human gut environment. These observations suggest complex bacterial interactions within the gut which may affect the host and cause intestinal damage. Full article
Show Figures

Figure 1

16 pages, 4396 KiB  
Article
Gut Dysbiosis and IL-21 Response in Patients with Severe COVID-19
by Mahejibin Khan, Bijina J. Mathew, Priyal Gupta, Garima Garg, Sagar Khadanga, Ashish Kumar Vyas and Anirudh K. Singh
Microorganisms 2021, 9(6), 1292; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9061292 - 13 Jun 2021
Cited by 32 | Viewed by 4440
Abstract
Background: The disease severity, ranging from being asymptomatic to having acute illness, and associated inflammatory responses has suggested that alterations in the gut microbiota may play a crucial role in the development of chronic disorders due to COVID-19 infection. This study describes gut [...] Read more.
Background: The disease severity, ranging from being asymptomatic to having acute illness, and associated inflammatory responses has suggested that alterations in the gut microbiota may play a crucial role in the development of chronic disorders due to COVID-19 infection. This study describes gut microbiota dysbiosis in COVID-19 patients and its implications relating to the disease. Design: A cross sectional prospective study was performed on thirty RT-PCR-confirmed COVID-19 patients admitted to the All India Institute of Medical Sciences, Bhopal, India, between September 10 and 20, 2020. Ten healthy volunteers were recruited as the control group. IFN, TNF, and IL-21 profiling was conducted using plasma samples, and gut bacterial analysis was performed after obtaining the metagenomics data of stool samples. Results: Patients with a variable COVID-19 severity showed distinct gut microflora and peripheral interleukin-21 levels. A low Firmicute/Bacteroidetes ratio, caused by the depletion of the fibre-utilizing bacteria, F. prausnitzii, B. Plebius, and Prevotella, and an increase in Bacteroidetes has associated gut microbiota dysbiosis with COVID-19 disease severity. Conclusions: The loss of the functional attributes of signature commensals in the gut, due to dysbiosis, is a predisposing factor of COVID-19 pathophysiology. Full article
Show Figures

Graphical abstract

14 pages, 3176 KiB  
Article
Comparative Gut Microbiome Differences between Ferric Citrate and Calcium Carbonate Phosphate Binders in Patients with End-Stage Kidney Disease
by Ping-Hsun Wu, Po-Yu Liu, Yi-Wen Chiu, Wei-Chun Hung, Yi-Ting Lin, Ting-Yun Lin, Szu-Chun Hung, Rachel Ann Delicano, Mei-Chuan Kuo and Chun-Ying Wu
Microorganisms 2020, 8(12), 2040; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8122040 - 20 Dec 2020
Cited by 15 | Viewed by 4399
Abstract
Gut dysbiosis in patients with chronic kidney disease (CKD) may induce chronic inflammation and increase morbidity. Phosphate-binding agents, generally used in patients with CKD, may potentially change the composition of the gut microbiota. This study aimed to compare the microbiota composition in hemodialysis [...] Read more.
Gut dysbiosis in patients with chronic kidney disease (CKD) may induce chronic inflammation and increase morbidity. Phosphate-binding agents, generally used in patients with CKD, may potentially change the composition of the gut microbiota. This study aimed to compare the microbiota composition in hemodialysis patients treated with ferric citrate or calcium carbonate. The stool microbiota was investigated in hemodialysis patients treated with ferric citrate (n = 8) and calcium carbonate (n = 46) using 16S rRNA gene amplicon sequencing profiling using linear discriminant analysis of effect size. Further predictive functional profiling of microbial communities was obtained with Tax4Fun in R. Hemodialysis patients treated with calcium carbonate had a significantly reduced microbial species diversity (Shannon index and Simpson index) and an increased microbial alteration ratio compared with patients treated with ferric citrate. A distinct microbial community structure was found in patients treated with ferric citrate, with an increased abundance of the Bacteroidetes phylum and a decreased abundance of the phylum Firmicutes. Members of the order Lactobacillales were enriched in patients treated with calcium carbonate, whereas taxa of the genera Ruminococcaceae UCG-004, Flavonifractor, and Cronobacter were enriched in patients treated with ferric citrate phosphate binder. In conclusion, Ferric citrate therapy results in a more diverse microbiome community compared to calcium carbonate therapy in hemodialysis patients with phosphate binder treatment. The gut microbiome reflects the phosphate binder choice in hemodialysis patients, further affecting the physiological environment in the gastrointestinal tract. Full article
Show Figures

Figure 1

13 pages, 6660 KiB  
Article
Effect of α-Hemolysin Producing E. coli in Two Different Mouse Strains in a DSS Model of Inflammatory Bowel Disease
by Hengameh Chloé Mirsepasi-Lauridsen, Carsten Struve, Andreas Munk Petersen and Karen Angeliki Krogfelt
Microorganisms 2020, 8(12), 1971; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8121971 - 11 Dec 2020
Cited by 5 | Viewed by 2273
Abstract
Background: Phylogroup B2 Escherichia coli have been associated with ulcerative colitis (UC). In this study, we aimed to compare colonization with the UC-associated E. coli p19A in different mice strains, to investigate the role of alpha hemolysin in a UC mouse model. Methods: [...] Read more.
Background: Phylogroup B2 Escherichia coli have been associated with ulcerative colitis (UC). In this study, we aimed to compare colonization with the UC-associated E. coli p19A in different mice strains, to investigate the role of alpha hemolysin in a UC mouse model. Methods: In this study, Sigirr −/− and C57BL/6 mice were chosen, and UC was induced by adding dextran sulfate sodium (DSS) to the drinking water. The mice were pre-treated with ciprofloxacin. p19A expressing luminescence and GFP, alpha-hemolysin knock out p19A-ΔhlyI II, and non-pathogenic lab E. coli DH10B were cultured in LB broth, and orally gavaged into the mice. Colonization with p19A WT was visualized using an in vivo imaging system. Results: p19A WT colonized the colon, ileum, Peyer’s patches, liver, and spleen of infected C57BL/6 and Sigirr −/− mice. A total of 99% of the p19A WT infected C57BL/6 mice and 29% of the p19A WT infected Sigirr −/− mice survived to the 4th post infection day. Conclusion: UC-associated E. coli p19A WT colonized the intestines of DSS-treated mice and caused extra-intestinal infection. Hemolysin is an important factor in this pathogenesis, since isogenic hemolysin mutants did not cause the same inflammation. Full article
Show Figures

Figure 1

19 pages, 2475 KiB  
Article
Impact of the Post-Transplant Period and Lifestyle Diseases on Human Gut Microbiota in Kidney Graft Recipients
by Nessrine Souai, Oumaima Zidi, Amor Mosbah, Imen Kosai, Jameleddine El Manaa, Naima Bel Mokhtar, Elias Asimakis, Panagiota Stathopoulou, Ameur Cherif, George Tsiamis and Soumaya Kouidhi
Microorganisms 2020, 8(11), 1724; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8111724 - 04 Nov 2020
Cited by 16 | Viewed by 2915
Abstract
Gaining long-term graft function and patient life quality remain critical challenges following kidney transplantation. Advances in immunology, gnotobiotics, and culture-independent molecular techniques have provided growing insights into the complex relationship of the microbiome and the host. However, little is known about the over [...] Read more.
Gaining long-term graft function and patient life quality remain critical challenges following kidney transplantation. Advances in immunology, gnotobiotics, and culture-independent molecular techniques have provided growing insights into the complex relationship of the microbiome and the host. However, little is known about the over time-shift of the gut microbiota in the context of kidney transplantation and its impact on both graft and health stability. Here we aimed to characterize the structure of gut microbiota within stable kidney graft recipients. We enrolled forty kidney transplant patients after at least three months of transplantation and compared them to eighteen healthy controls. The overall microbial community structure of the kidney transplanted group was clearly different from control subjects. We found lower relative abundances of Actinobacteria, Bacteroidetes, and Verrucomicrobia within the patient group and a higher abundance of Proteobacteria compared to the control group. Both richness and Shannon diversity indexes were significantly lower in the kidney graft recipients than in healthy controls. Post-graft period was positively correlated with the relative abundance of the Proteobacteria phylum, especially Escherichia.Shigella genus. Interestingly, only Parabacteroides was found to significantly differentiate patients that were not suffering from lifestyle diseases and those who suffer from post-graft complications. Furthermore, network analysis showed that the occurrence of lifestyle diseases was significantly linked with a higher number of negative interactions of Sutterella and Succinivibrio genera within patients. This study characterizes gut microbiome fluctuation in stable kidney transplant patients after a long post-allograft period. Analysis of fecal microbiota could be useful for nephrologists as a new clinical tool that can improve kidney allograft monitoring and outcomes. Full article
Show Figures

Figure 1

16 pages, 1427 KiB  
Article
Investigation into In Vitro and In Vivo Caenorhabditis elegans Models to Select Cheese Yeasts as Probiotic Candidates for their Preventive Effects against Salmonella Typhimurium
by Philippe Veisseire, Muriel Bonnet, Taous Saraoui, Cyril Poupet, Olivier Camarès, Marylise Gachinat, Cécile Callon, Guy Febvre, Christophe Chassard and Stéphanie Bornes
Microorganisms 2020, 8(6), 922; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8060922 - 18 Jun 2020
Cited by 8 | Viewed by 2719
Abstract
The design of multiscale strategies integrating in vitro and in vivo models is necessary for the selection of new probiotics. In this regard, we developed a screening assay based on the investigation of the potential of yeasts from cheese as probiotics against the [...] Read more.
The design of multiscale strategies integrating in vitro and in vivo models is necessary for the selection of new probiotics. In this regard, we developed a screening assay based on the investigation of the potential of yeasts from cheese as probiotics against the pathogen Salmonella Typhimurium UPsm1 (ST). Two yeasts isolated from raw-milk cheese (Saccharomyces cerevisiae 16, Sc16; Debaryomyces hansenii 25, Dh25), as well as S. cerevisiae subspecies boulardii (CNCM I-1079, Sb1079), were tested against ST by applying in vitro and in vivo tests. Adherence measurements to Caco-2 and HT29-MTX intestinal cells indicated that the two tested cheese yeasts presented a better adhesion than the probiotic Sb1079 as the control strain. Further, the Dh25 was the cheese yeast most likely to survive in the gastrointestinal tract. What is more, the modulation of the TransEpithelial Electrical Resistance (TEER) of differentiated Caco-2 cell monolayers showed the ability of Dh25 to delay the deleterious effects of ST. The influence of microorganisms on the in vivo model Caenorhabditis elegans was evaluated by measuring the longevity of the worm. This in vivo approach revealed that this yeast increased the worm’s lifespan and protected it against ST infection, confirming that this in vivo model can be useful for screening probiotic cheese yeasts. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

20 pages, 1222 KiB  
Review
GLP-1 and GLP-2 Orchestrate Intestine Integrity, Gut Microbiota, and Immune System Crosstalk
by Nyan Abdalqadir and Khosrow Adeli
Microorganisms 2022, 10(10), 2061; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms10102061 - 19 Oct 2022
Cited by 16 | Viewed by 7539
Abstract
The intestine represents the body’s largest interface between internal organs and external environments except for its nutrient and fluid absorption functions. It has the ability to sense numerous endogenous and exogenous signals from both apical and basolateral surfaces and respond through endocrine and [...] Read more.
The intestine represents the body’s largest interface between internal organs and external environments except for its nutrient and fluid absorption functions. It has the ability to sense numerous endogenous and exogenous signals from both apical and basolateral surfaces and respond through endocrine and neuronal signaling to maintain metabolic homeostasis and energy expenditure. The intestine also harbours the largest population of microbes that interact with the host to maintain human health and diseases. Furthermore, the gut is known as the largest endocrine gland, secreting over 100 peptides and other molecules that act as signaling molecules to regulate human nutrition and physiology. Among these gut-derived hormones, glucagon-like peptide 1 (GLP-1) and -2 have received the most attention due to their critical role in intestinal function and food absorption as well as their application as key drug targets. In this review, we highlight the current state of the literature that has brought into light the importance of GLP-1 and GLP-2 in orchestrating intestine–microbiota–immune system crosstalk to maintain intestinal barrier integrity, inflammation, and metabolic homeostasis. Full article
Show Figures

Figure 1

16 pages, 727 KiB  
Review
Risk Factors, Diagnosis, and Management of Clostridioides difficile Infection in Patients with Inflammatory Bowel Disease
by Livio Enrico Del Vecchio, Marcello Fiorani, Ege Tohumcu, Stefano Bibbò, Serena Porcari, Maria Cristina Mele, Marco Pizzoferrato, Antonio Gasbarrini, Giovanni Cammarota and Gianluca Ianiro
Microorganisms 2022, 10(7), 1315; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms10071315 - 29 Jun 2022
Cited by 6 | Viewed by 2254
Abstract
Clostridioides difficile infection (CDI) and inflammatory bowel disease (IBD) are two pathologies that share a bidirectional causal nexus, as CDI is known to have an aggravating effect on IBD and IBD is a known risk factor for CDI. The colonic involvement in IBD [...] Read more.
Clostridioides difficile infection (CDI) and inflammatory bowel disease (IBD) are two pathologies that share a bidirectional causal nexus, as CDI is known to have an aggravating effect on IBD and IBD is a known risk factor for CDI. The colonic involvement in IBD not only renders the host more prone to an initial CDI development but also to further recurrences. Furthermore, IBD flares, which are predominantly set off by a CDI, not only create a need for therapy escalation but also prolong hospital stay. For these reasons, adequate and comprehensive management of CDI is of paramount importance in patients with IBD. Microbiological diagnosis, correct evaluation of clinical status, and consideration of different treatment options (from antibiotics and fecal microbiota transplantation to monoclonal antibodies) carry pivotal importance. Thus, the aim of this article is to review the risk factors, diagnosis, and management of CDI in patients with IBD. Full article
Show Figures

Figure 1

15 pages, 832 KiB  
Review
Production of Indole and Indole-Related Compounds by the Intestinal Microbiota and Consequences for the Host: The Good, the Bad, and the Ugly
by Naouel Tennoune, Mireille Andriamihaja and François Blachier
Microorganisms 2022, 10(5), 930; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms10050930 - 28 Apr 2022
Cited by 28 | Viewed by 6718
Abstract
The intestinal microbiota metabolic activity towards the available substrates generates myriad bacterial metabolites that may accumulate in the luminal fluid. Among them, indole and indole-related compounds are produced by specific bacterial species from tryptophan. Although indole-related compounds are, first, involved in intestinal microbial [...] Read more.
The intestinal microbiota metabolic activity towards the available substrates generates myriad bacterial metabolites that may accumulate in the luminal fluid. Among them, indole and indole-related compounds are produced by specific bacterial species from tryptophan. Although indole-related compounds are, first, involved in intestinal microbial community communication, these molecules are also active on the intestinal mucosa, exerting generally beneficial effects in different experimental situations. After absorption, indole is partly metabolized in the liver into the co-metabolite indoxyl sulfate. Although some anti-inflammatory actions of indole on liver cells have been shown, indoxyl sulfate is a well-known uremic toxin that aggravates chronic kidney disease, through deleterious effects on kidney cells. Indoxyl sulfate is also known to provoke endothelial dysfunction. Regarding the central nervous system, emerging research indicates that indole at excessive concentrations displays a negative impact on emotional behavior. The indole-derived co-metabolite isatin appears, in pre-clinical studies, to accumulate in the brain, modulating brain function either positively or negatively, depending on the doses used. Oxindole, a bacterial metabolite that enters the brain, has shown deleterious effects on the central nervous system in experimental studies. Lastly, recent studies performed with indoxyl sulfate report either beneficial or deleterious effects depending once again on the dose used, with missing information on the physiological concentrations that are reaching the central nervous system. Any intervention aiming at modulating indole and indole-related compound concentrations in the biological fluids should crucially take into account the dual effects of these compounds according to the host tissues considered. Full article
Show Figures

Figure 1

29 pages, 1346 KiB  
Review
Food Additives, a Key Environmental Factor in the Development of IBD through Gut Dysbiosis
by Pauline Raoul, Marco Cintoni, Marta Palombaro, Luisa Basso, Emanuele Rinninella, Antonio Gasbarrini and Maria Cristina Mele
Microorganisms 2022, 10(1), 167; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms10010167 - 13 Jan 2022
Cited by 32 | Viewed by 17806
Abstract
Diet is a key environmental factor in inflammatory bowel disease (IBD) and, at the same time, represents one of the most promising therapies for IBD. Our daily diet often contains food additives present in numerous processed foods and even in dietary supplements. Recently, [...] Read more.
Diet is a key environmental factor in inflammatory bowel disease (IBD) and, at the same time, represents one of the most promising therapies for IBD. Our daily diet often contains food additives present in numerous processed foods and even in dietary supplements. Recently, researchers and national authorities have been paying much attention to their toxicity and effects on gut microbiota and health. This review aims to gather the latest data focusing on the potential role of food additives in the pathogenesis of IBDs through gut microbiota modulation. Some artificial emulsifiers and sweeteners can induce the dysbiosis associated with an alteration of the intestinal barrier, an activation of chronic inflammation, and abnormal immune response accelerating the onset of IBD. Even if most of these results are retrieved from in vivo and in vitro studies, many artificial food additives can represent a potential hidden driver of gut chronic inflammation through gut microbiota alterations, especially in a population with IBD predisposition. In this context, pending the confirmation of these results by large human studies, it would be advisable that IBD patients avoid the consumption of processed food containing artificial food additives and follow a personalized nutritional therapy prescribed by a clinical nutritionist. Full article
Show Figures

Figure 1

12 pages, 588 KiB  
Review
Akkermansia muciniphila, a New Generation of Beneficial Microbiota in Modulating Obesity: A Systematic Review
by Jumana Nabil Abuqwider, Gianluigi Mauriello and Mohammad Altamimi
Microorganisms 2021, 9(5), 1098; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9051098 - 20 May 2021
Cited by 40 | Viewed by 6698
Abstract
Obesity is a complex syndrome and is recognized as the ultimate pathway of many chronic diseases. Studies using Akkermansia muciniphila supplementation strategy have proved to be effective for the prevention and treatment of obesity and other metabolic disorders. Although there are studies that [...] Read more.
Obesity is a complex syndrome and is recognized as the ultimate pathway of many chronic diseases. Studies using Akkermansia muciniphila supplementation strategy have proved to be effective for the prevention and treatment of obesity and other metabolic disorders. Although there are studies that support the protective effect of this strategy, the effects on the prevention of obesity on humans are not clear yet and need more investigation. The aim of this study is to investigate the effect of A. muciniphila administration on modulating obesity. This systematic review was generated from articles published within the last 10 years. All articles were in English and included animal subjects. The review relied on the search engines Google Scholar, Pub Med, Web of Science and Medline using the following keywords: A. muciniphila, next-generation probiotic, new-generation probiotic, obesity, fat mass, body fat and lipid profile. The search has revealed 804 articles with relevant key words. After the exclusion of irrelevant articles, 10 studies were selected based on the criteria. These studies were randomized controlled trials that have shown that A. muciniphila modulates obesity by regulating metabolism and energy hemostasis and improving insulin sensitivity and glucose hemostasis. In addition, studies showed this microorganism enhances low grade inflammation by different mechanisms. Full article
Show Figures

Figure 1

15 pages, 860 KiB  
Review
The Role of Enterobacteriaceae in Gut Microbiota Dysbiosis in Inflammatory Bowel Diseases
by Valerio Baldelli, Franco Scaldaferri, Lorenza Putignani and Federica Del Chierico
Microorganisms 2021, 9(4), 697; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9040697 - 27 Mar 2021
Cited by 110 | Viewed by 11166
Abstract
Inflammatory bowel diseases (IBDs) are a group of chronic gastrointestinal inflammatory diseases with unknown etiology. There is a combination of well documented factors in their pathogenesis, including intestinal microbiota dysbiosis. The symbiotic microbiota plays important functions in the host, and the loss of [...] Read more.
Inflammatory bowel diseases (IBDs) are a group of chronic gastrointestinal inflammatory diseases with unknown etiology. There is a combination of well documented factors in their pathogenesis, including intestinal microbiota dysbiosis. The symbiotic microbiota plays important functions in the host, and the loss of beneficial microbes could favor the expansion of microbial pathobionts. In particular, the bloom of potentially harmful Proteobacteria, especially Enterobacteriaceae, has been described as enhancing the inflammatory response, as observed in IBDs. Herein, we seek to investigate the contribution of Enterobacteriaceae to IBD pathogenesis whilst considering the continuous expansion of the literature and data. Despite the mechanism of their expansion still remaining unclear, their expansion could be correlated with the increase in nitrate and oxygen levels in the inflamed gut and with the bile acid dysmetabolism described in IBD patients. Furthermore, in several Enterobacteriaceae studies conducted at a species level, it has been suggested that some adherent-invasive Escherichia coli (AIEC) play an important role in IBD pathogenesis. Overall, this review highlights the pivotal role played by Enterobacteriaceae in gut dysbiosis associated with IBD pathogenesis and progression. Full article
Show Figures

Figure 1

15 pages, 331 KiB  
Review
Gut and Reproductive Tract Microbiota Adaptation during Pregnancy: New Insights for Pregnancy-Related Complications and Therapy
by Martina De Siena, Lucrezia Laterza, Maria Valeria Matteo, Irene Mignini, Tommaso Schepis, Gianenrico Rizzatti, Gianluca Ianiro, Emanuele Rinninella, Marco Cintoni and Antonio Gasbarrini
Microorganisms 2021, 9(3), 473; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9030473 - 25 Feb 2021
Cited by 27 | Viewed by 3452
Abstract
Pregnancy is characterized by maternal adaptations that are necessary to create a welcoming and hospitable environment for the fetus. Studies have highlighted how the microbiota modulates several networks in humans through complex molecular interactions and how dysbiosis (defined as quantitative and qualitative alterations [...] Read more.
Pregnancy is characterized by maternal adaptations that are necessary to create a welcoming and hospitable environment for the fetus. Studies have highlighted how the microbiota modulates several networks in humans through complex molecular interactions and how dysbiosis (defined as quantitative and qualitative alterations of the microbiota communities) is related to human pathologies including gynecological diseases. This review analyzed how maternal uterine, vaginal, and gut microbiomes could impact on fetus health during the gestational period. We evaluated the role of a dysbiotic microbiota in preterm birth, chorioamnionitis, gestational diabetes mellitus and pre-eclampsia. For many years it has been hypothesized that newborns were sterile organisms but in the past few years this paradigm has been questioned through the demonstration of the presence of microbes in the placenta and meconium. In the future, we should go deeper into the concept of in utero colonization to better understand the role of microbiota through the phases of pregnancy. Numerous studies in the literature have already showed interesting results regarding the role of microbiota in pregnancy. This evidence gives us the hope that microbiota modulation could be a novel strategy to reduce the morbidity and mortality related to pregnancy complications in the future. Full article
19 pages, 734 KiB  
Review
Does Gut-Microbiome Interaction Protect against Obesity and Obesity-Associated Metabolic Disorders?
by Agnieszka Zawada, Anna Maria Rychter, Alicja Ewa Ratajczak, Agata Lisiecka-Masian, Agnieszka Dobrowolska and Iwona Krela-Kaźmierczak
Microorganisms 2021, 9(1), 18; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9010018 - 23 Dec 2020
Cited by 15 | Viewed by 4600
Abstract
More research has recently focused on the role of the gut microbiota in the development or course of numerous diseases, including non-communicable diseases. As obesity remains prevalent, the question arises as to what microbial changes are associated with increased obesity prevalence and what [...] Read more.
More research has recently focused on the role of the gut microbiota in the development or course of numerous diseases, including non-communicable diseases. As obesity remains prevalent, the question arises as to what microbial changes are associated with increased obesity prevalence and what kind of prevention and treatment approaches it could provide. Moreover, the influence of the gut-brain axis on obesity is also crucial, since it can affect metabolism and food intake. The quantitative and qualitative changes in the microbiota composition are called dysbiosis; however, in view of the current knowledge, it is difficult to conclude which microbial imbalances are adverse or beneficial. Increased numbers of pathological microorganisms were observed among patients with obesity and comorbidities associated with it, such as diabetes, cardiovascular disease, and insulin resistance. Our review provides current knowledge regarding changes in the intestinal microbiota associated with obesity and obesity-associated comorbidities. Nevertheless, given that dietary patterns and nutrients are two of the factors affecting the intestinal microbiota, we also discuss the role of different dietary approaches, vitamins, and minerals in the shaping of the intestinal microbiota. Full article
Show Figures

Figure 1

23 pages, 808 KiB  
Review
Gut Microbiota during Dietary Restrictions: New Insights in Non-Communicable Diseases
by Emanuele Rinninella, Marco Cintoni, Pauline Raoul, Gianluca Ianiro, Lucrezia Laterza, Loris Riccardo Lopetuso, Francesca Romana Ponziani, Antonio Gasbarrini and Maria Cristina Mele
Microorganisms 2020, 8(8), 1140; https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8081140 - 28 Jul 2020
Cited by 29 | Viewed by 8411
Abstract
In recent decades, there has been a growing interest in dietary restrictions for their promising effects on longevity and health span. Indeed, these strategies are supposed to delay the onset and burden of non-communicable diseases (NCDs) such as obesity, diabetes, cancer and neurological [...] Read more.
In recent decades, there has been a growing interest in dietary restrictions for their promising effects on longevity and health span. Indeed, these strategies are supposed to delay the onset and burden of non-communicable diseases (NCDs) such as obesity, diabetes, cancer and neurological and gastrointestinal inflammatory diseases. At the same time, the gut microbiota has been shown to play a crucial role in NCDs since it is actively involved in maintaining gut homeostasis through its impact on nutrients metabolism, gut barrier, and immune system. There is evidence that dietary restrictions could slow down age-related changes in the types and numbers of gut bacteria, which may counteract gut dysbiosis. The beneficial effects on gut microbiota may positively influence host metabolism, gut barrier permeability, and brain functions, and subsequently, postpone the onset of NCDs prolonging the health span. These new insights could lead to the development of novel strategies for modulating gut microbiota with the end goal of treating/preventing NCDs. This review provides an overview of animal and human studies focusing on gut microbiota variations during different types of dietary restriction, in order to highlight the close relationship between gut microbiota balance and the host’s health benefits induced by these nutritional regimens. Full article
Show Figures

Graphical abstract

Back to TopTop