Gut Microbiota, Diet, and Immunity: Investigating the Connections and Implications for Disease Development

A special issue of Biomedicines (ISSN 2227-9059). This special issue belongs to the section "Immunology and Immunotherapy".

Deadline for manuscript submissions: closed (30 September 2024) | Viewed by 21621

Special Issue Editor


E-Mail Website
Guest Editor
Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
Interests: diet-induced inflammation; nutrition and inflammation; diet and gut microbiota; oxidative stress and antioxidants; dietary bioactive compounds
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The interplay between gut microbiota, dietary factors, and immune responses has emerged as a critical area of research with profound implications for understanding disease development. This Special Issue delves into the intricate connections between the gut microbiota, dietary patterns/intakes/habits, and the immune system, aiming to shed light on the multifaceted relationships that contribute to health and disease.

The gut microbiota, a diverse ecosystem of microorganisms residing in the gastrointestinal tract, plays a pivotal role in maintaining homeostasis and influencing various physiological processes. Recent advances in microbiome research have unveiled the impact of diet on shaping the composition and functionality of gut microbes. This Special Issue explores how dietary choices/patterns/intakes/habits, including nutrients and bioactive compounds, modulate the gut microbiota and subsequently impact immune function.

Understanding the bidirectional communication between the gut microbiota and the immune system is crucial for unraveling the mechanisms underlying disease susceptibility and resilience. Articles featured in this Special Issue will cover a spectrum of topics, including the role of gut microbiota in immune system development, the influence of dietary patterns on microbial diversity, and the immunomodulatory effects of specific nutrients.

Furthermore, the implications of dysbiosis—an imbalance in the gut microbiota—on immune-related diseases will be addressed. Contributors will investigate how disruptions in the delicate equilibrium of the gut ecosystem, often driven by poor dietary habits, may contribute to the pathogenesis of various disorders, such as inflammatory bowel diseases, autoimmune conditions, and metabolic syndromes.

The overarching goal of this Special Issue is to consolidate current knowledge, present cutting-edge research findings, and stimulate further exploration into the complex interconnections between gut microbiota, diet, and immunity. By fostering a comprehensive understanding of these relationships, we aim to provide insights that may guide the development of targeted interventions and therapeutic strategies for preventing and managing diseases associated with a dysregulated gut microbiota and immune responses.

Researchers, clinicians, and professionals across disciplines are invited to contribute their expertise to advance our understanding of the pivotal role played by the gut microbiota, diet, and immunity in health and disease.

Contributions from every corner of biomedicine, such as biochemistry, molecular biology, physiology, pathology, microbiology, cell biology, toxicology, pharmacology, virology, bacteriology, immunology, genetics, and embryology, are welcome. However, reports must be based on serum/blood/circulating investigations and be chemically well defined. Outstanding cellular or animal models may be considered. Even if not associated with serum metabolites, gut microbiota results are welcome.

Dr. Farhad Vahid
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. Biomedicines 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 2600 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
  • diet
  • immunity
  • microbiome
  • dysbiosis
  • inflammatory bowel diseases
  • cancer
  • CVD
  • T2D
  • obesity
  • overweight
  • autoimmune diseases
  • metabolic syndromes
  • immunomodulation
  • microbial diversity
  • nutrient–gut microbiota interactions
  • probiotics
  • prebiotics
  • immune system development
  • disease susceptibility
  • therapeutic interventions
  • host–microbe interactions
  • gut–brain axis
  • microbial metabolites
  • precision nutrition

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Related Special Issue

Published Papers (14 papers)

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

Research

Jump to: Review, Other

14 pages, 1809 KiB  
Article
Causal Relationship Between Sjögren’s Syndrome and Gut Microbiota: A Two-Sample Mendelian Randomization Study
by Xinrun Wang, Minghui Liu and Weiping Xia
Biomedicines 2024, 12(10), 2378; https://doi.org/10.3390/biomedicines12102378 - 18 Oct 2024
Viewed by 138
Abstract
Background: Gut microbiota have been previously reported to be related to a variety of immune diseases. However, the causal connection between Sjögren’s syndrome (SS) and gut microbiota has yet to be clarified. Methods: We employed a two-sample Mendelian randomization (MR) analysis to evaluate [...] Read more.
Background: Gut microbiota have been previously reported to be related to a variety of immune diseases. However, the causal connection between Sjögren’s syndrome (SS) and gut microbiota has yet to be clarified. Methods: We employed a two-sample Mendelian randomization (MR) analysis to evaluate the causal connection between gut microbiota and SS, utilizing summary statistics from genome-wide association studies (GWASs) obtained from the MiBioGen and FinnGen consortia. The inverse variance weighted (IVW) approach represents the primary method of Mendelian randomization (MR) analysis. Sensitivity analysis was used to eliminate instrumental variables heterogeneity and horizontal pleiotropy. In addition, we performed an analysis using independent GWAS summary statistics for SS from the European Bioinformatics Institute (EBI) dataset for further verify our results. Results: IVW results demonstrated that the phylum Lentisphaerae (OR = 0.79, 95% CI: 0.63–0.99, p = 0.037), class Deltaproteobacteria (OR = 0.67, 95% CI: 0.47–0.96, p = 0.030), family Porphyromonadaceae (OR = 0.60, 95% CI: 0.38–0.94, p = 0.026), genus Eubacterium coprostanoligenes group (OR = 0.61, 95% CI: 0.4–0.93, p = 0.021), genus Blautia (OR = 0.62, 95% CI: 0.43–0.90, p = 0.012), genus Butyricicoccus (OR = 0.61, 95% CI: 0.42–0.90, p = 0.012), genus Escherichia.Shigella (OR = 0.7, 95% CI: 0.49–0.99, p = 0.045) and genus Subdoligranulum (OR = 0.61, 95% CI: 0.44–0.86, p = 0.005) exhibited protective effects on SS. Relevant heterogeneity of horizontal pleiotropy or instrumental variables was not detected. Furthermore, repeating our results with an independent cohort provided by the EBI dataset, only the genus Eubacterium coprostanoligenes group remained significantly associated with the protective effect on SS (OR = 0.41, 95% CI: 0.18–0.91, p = 0.029). Two-step MR analysis further revealed that genus Eubacterium coprostanoligenes group exerts its protective effect by reducing CXCL6 levels in SS (OR, 0.87; 95% CI = 0.76–0.99, p = 0.033). Conclusions: Our study using two-sample MR analysis identified a causal association between multiple genera and SS. A two-step MR result calculated that genus Eubacterium coprostanoligenes group mediated its protective effect by reducing CXCL6 levels in SS. However, the datasets available from the MiBioGen and FinnGen consortia do not provide sufficient information or comprehensive demographic data for subgroup analyses. Additional validation using various omics technologies is necessary to comprehend the development of SS in the intricate interplay between genes and the environment over a period of time. Full article
Show Figures

Figure 1

17 pages, 2023 KiB  
Article
The Combined Effect of Western Diet Consumption and Diclofenac Administration Alters the Gut Microbiota and Promotes Anastomotic Leakage in the Distal Colon
by Melissa N. N. Arron, Stijn Bluiminck, Richard P. G. ten Broek, Thomas H. A. Ederveen, Lindsay Alpert, Olga Zaborina, John C. Alverdy and Harry van Goor
Biomedicines 2024, 12(10), 2170; https://doi.org/10.3390/biomedicines12102170 - 24 Sep 2024
Viewed by 592
Abstract
Background: Obesity, Western diet (WD) consumption, and the use of non-steroidal anti-inflammatory drugs (NSAIDs) are co-occurring and modifiable factors associated with microbiome dysbiosis and anastomotic leakage. We studied the combined effect of a Western-type diet (WD) and diclofenac, a standard NSAID used in [...] Read more.
Background: Obesity, Western diet (WD) consumption, and the use of non-steroidal anti-inflammatory drugs (NSAIDs) are co-occurring and modifiable factors associated with microbiome dysbiosis and anastomotic leakage. We studied the combined effect of a Western-type diet (WD) and diclofenac, a standard NSAID used in surgical patients, on anastomotic healing and gut microbiota composition following distal colon resection. Methods: Forty-two rats were fed a WD for 6 weeks, after which they were randomized to either parenteral diclofenac 3 mg/kg/day or saline started on the day of surgery and continued for three days. The surgical procedure involved distal colon resection with anastomosis. Animals were sacrificed on postoperative day (POD)-3 or POD-5. Anastomotic healing was assessed and correlated with diclofenac treatment and gut microbiota composition, analyzed by 16S rRNA marker gene amplicon sequencing. Mucosal integrity of the anastomosis was evaluated by histological analysis. Results: Anastomotic leakage rate was 100 percent (8/8) in diclofenac-treated rats and 10 percent (1/10) in saline-treated controls on POD-5. Diclofenac administration in WD-fed animals induced a shift in microbiota composition, characterized by an increase in microbiota diversity on POD-5 and a significant 15-fold, 4-fold, and 16-fold increase of Proteobacteria, Bacteroidetes, and Verrucomicrobia, respectively. Diclofenac use in WD-fed animals caused mucosal erosion on POD-5, a phenomenon not observed in control animals. Conclusions: Consumption of a Western diet combined with diclofenac administration shifts the microbiota composition, associated with clinically relevant AL in the distal colon of rats. Full article
Show Figures

Figure 1

17 pages, 6504 KiB  
Article
Effects of Resistant-Starch-Encapsulated Probiotic Cocktail on Intestines Damaged by 5-Fluorouracil
by Jui-Ling Wang, Chin-Hsing Yeh, Shih-Hung Huang, Lawrence Shih-Hsin Wu and Miles Chih-Ming Chen
Biomedicines 2024, 12(8), 1912; https://doi.org/10.3390/biomedicines12081912 - 20 Aug 2024
Viewed by 594
Abstract
Probiotics and prebiotics have gained attention for their potential health benefits. However, their efficacy hinges on probiotic survival through the harsh gastrointestinal environment. Microencapsulation techniques provide a solution, with resistant starch (RS)-based techniques showing promise in maintaining probiotic viability. Specifically, RS-encapsulated probiotics significantly [...] Read more.
Probiotics and prebiotics have gained attention for their potential health benefits. However, their efficacy hinges on probiotic survival through the harsh gastrointestinal environment. Microencapsulation techniques provide a solution, with resistant starch (RS)-based techniques showing promise in maintaining probiotic viability. Specifically, RS-encapsulated probiotics significantly improved probiotic survival in gastric acid, bile salts, and simulated intestinal conditions. This study investigated the effects of a resistant-starch-encapsulated probiotic cocktail (RS-Pro) in the context of 5-fluorouracil (5-FU) chemotherapy, which frequently induces microbiota dysbiosis and intestinal mucositis. Female BALB/c mice were divided into three groups: a 5-FU group, a 5-FU+Pro group receiving free probiotics, and a 5-FU+RS-Pro group receiving RS-encapsulated probiotics. After 28 days of treatment, analyses were conducted on fecal microbiota, intestinal histology, peripheral blood cell counts, and body and organ weights. It was revealed by 16S rRNA MiSeq sequencing that 5-FU treatment disrupted gut microbiota composition, reduced microbial diversity, and caused dysbiosis. RS-Pro treatment restored microbial diversity and increased the population of beneficial bacteria, such as Muribaculaceae, which play roles in carbohydrate and polyphenol metabolism. Furthermore, 5-FU administration induced moderate intestinal mucositis, characterized by reduced cellularity and shortened villi. However, RS-Pro treatment attenuated 5-FU-induced intestinal damage, preserving villus length. Mild leukopenia observed in the 5-FU-treated mice was partially alleviated in 5-FU+Pro and 5-FU+RS-Pro groups. These findings suggest that RS-Pro may serve as an adjunct to chemotherapy, potentially reducing adverse effects and improving therapeutic outcomes in future clinical applications. Full article
Show Figures

Figure 1

16 pages, 6019 KiB  
Article
Diet and the Gut Microbiome as Determinants Modulating Metabolic Outcomes in Young Obese Adults
by Elena N. Livantsova, Georgy E. Leonov, Antonina V. Starodubova, Yurgita R. Varaeva, Aleksey A. Vatlin, Stanislav I. Koshechkin, Tatyana N. Korotkova and Dmitry B. Nikityuk
Biomedicines 2024, 12(7), 1601; https://doi.org/10.3390/biomedicines12071601 - 18 Jul 2024
Cited by 1 | Viewed by 1262
Abstract
Obesity, along with metabolic disorders such as dyslipidemia and insulin resistance, increases the risk of cardiovascular disease, diabetes, various cancers, and other non-communicable diseases, thereby contributing to higher mortality rates. The intestinal microbiome plays a crucial role in maintaining homeostasis and influencing human [...] Read more.
Obesity, along with metabolic disorders such as dyslipidemia and insulin resistance, increases the risk of cardiovascular disease, diabetes, various cancers, and other non-communicable diseases, thereby contributing to higher mortality rates. The intestinal microbiome plays a crucial role in maintaining homeostasis and influencing human metabolism. This study enrolled 82 young obese individuals, who were stratified into groups with or without metabolic disturbances. No significant differences in the alpha or beta diversity of the microbiota were observed among the groups. Insulin resistance was characterized by an increase in the number of Adlercreutzia and Dialister as well as a decrease in Collinsella, Coprococcus and Clostridiales. The dyslipidemia and dyslipidemia+insulin resistance groups had no significant differences in the gut microbiota. Dietary patterns also influenced microbial composition, with high protein intake increasing Leuconostoc and Akkermansia, and high fiber intake boosting Lactobacillus and Streptococcus. The genus Erwinia was associated with increases in visceral fat and serum glucose as well as a decrease in high-density lipoprotein cholesterol. Our findings highlight a significant association between gut microbiota composition and metabolic disturbances in young obese individuals, and they suggest that dietary modifications may promote a healthy microbiome and reduce the risk of developing metabolic disorders. Full article
Show Figures

Figure 1

11 pages, 2158 KiB  
Article
Exploring the Impact of Short Term Travel on Gut Microbiota and Probiotic Bacteria Mediated Stability
by Yiming Zhao, Chunyan Li, Kaijuan Wu, Hao Chen, Qingqun Wang, Ying Xiao, Siqi Yao, Ao Hong, Man Zhang, Shibo Lei, Wenyu Yang, Shukun Zhong, Abdulrahim Umar, Jing Huang and Zheng Yu
Biomedicines 2024, 12(7), 1378; https://doi.org/10.3390/biomedicines12071378 - 21 Jun 2024
Viewed by 935
Abstract
Although travelers are frequently accompanied by abdominal discomfort and even diarrhea, not every trip can cause this issue. Many studies have reported that intestinal microbes play an important role in it. However, little is known about the reason for the dynamics of these [...] Read more.
Although travelers are frequently accompanied by abdominal discomfort and even diarrhea, not every trip can cause this issue. Many studies have reported that intestinal microbes play an important role in it. However, little is known about the reason for the dynamics of these intestinal microbes. Here, we delved into the effects of short-term travel on the gut microbiota of 12 healthy individuals. A total of 72 fecal samples collected before and after one-week travel, alongside non-traveling controls, underwent amplicon sequencing and a series of bioinformatic analyses. We found that travel significantly increased intra-individual gut microbiota fluctuations without diarrhea symptoms. In addition, the initial composition of the gut microbiota before travel emerged as a crucial factor in understanding these fluctuations. Travelers with stable microbiota exhibited an enrichment of specific probiotic bacteria (Agathobaculum, Faecalibacterium, Bifidobacterium, Roseburia, Lactobacillus) before travel. Another batch of data validated their predictive role in distinguishing travelers with and without the gut microbial disorder. This work provided valuable insights into understanding the relationship between gut microbiota and travel. It offered a microbiota-centric perspective and a potential avenue for interventions to preserve gut health during travel. Full article
Show Figures

Figure 1

20 pages, 3577 KiB  
Article
Microbial Signatures in COVID-19: Distinguishing Mild and Severe Disease via Gut Microbiota
by Julia S. Galeeva, Dmitry E. Fedorov, Elizaveta V. Starikova, Alexander I. Manolov, Alexander V. Pavlenko, Oksana V. Selezneva, Ksenia M. Klimina, Vladimir A. Veselovsky, Maxim D. Morozov, Oleg O. Yanushevich, Natella I. Krikheli, Oleg V. Levchenko, Dmitry N. Andreev, Filipp S. Sokolov, Aleksey K. Fomenko, Mikhail K. Devkota, Nikolai G. Andreev, Andrey V. Zaborovskiy, Petr A. Bely, Sergei V. Tsaregorodtsev, Vladimir V. Evdokimov, Igor V. Maev, Vadim M. Govorun and Elena N. Ilinaadd Show full author list remove Hide full author list
Biomedicines 2024, 12(5), 996; https://doi.org/10.3390/biomedicines12050996 - 1 May 2024
Cited by 2 | Viewed by 1451
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has significantly impacted global healthcare, underscoring the importance of exploring the virus’s effects on infected individuals beyond treatments and vaccines. Notably, recent findings suggest that SARS-CoV-2 can infect the gut, thereby altering the gut microbiota. [...] Read more.
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has significantly impacted global healthcare, underscoring the importance of exploring the virus’s effects on infected individuals beyond treatments and vaccines. Notably, recent findings suggest that SARS-CoV-2 can infect the gut, thereby altering the gut microbiota. This study aimed to analyze the gut microbiota composition differences between COVID-19 patients experiencing mild and severe symptoms. We conducted 16S rRNA metagenomic sequencing on fecal samples from 49 mild and 43 severe COVID-19 cases upon hospital admission. Our analysis identified a differential abundance of specific bacterial species associated with the severity of the disease. Severely affected patients showed an association with Enterococcus faecium, Akkermansia muciniphila, and others, while milder cases were linked to Faecalibacterium prausnitzii, Alistipes putredinis, Blautia faecis, and additional species. Furthermore, a network analysis using SPIEC-EASI indicated keystone taxa and highlighted structural differences in bacterial connectivity, with a notable disruption in the severe group. Our study highlights the diverse impacts of SARS-CoV-2 on the gut microbiome among both mild and severe COVID-19 patients, showcasing a spectrum of microbial responses to the virus. Importantly, these findings align, to some extent, with observations from other studies on COVID-19 gut microbiomes, despite variations in methodologies. The findings from this study, based on retrospective data, establish a foundation for future prospective research to confirm the role of the gut microbiome as a predictive biomarker for the severity of COVID-19. Full article
Show Figures

Figure 1

17 pages, 12861 KiB  
Article
Characteristics of the Gut Microbiota in Regard to Atopic Dermatitis and Food Allergies of Children
by Alexandra I. Nekrasova, Irina G. Kalashnikova, Maria M. Bobrova, Anna V. Korobeinikova, Sirozhdin Yu. Bakoev, German A. Ashniev, Ekaterina S. Petryaikina, Alexander S. Nekrasov, Angelica V. Zagainova, Mariya V. Lukashina, Larisa R. Tolkacheva, Anastasia S. Zhdanova, Vladimir E. Mukhin, Vladimir S. Yudin, Anton A. Keskinov, Valentin V. Makarov, Sergey A. Kraevoy and Sergey M. Yudin
Biomedicines 2024, 12(3), 553; https://doi.org/10.3390/biomedicines12030553 - 1 Mar 2024
Cited by 1 | Viewed by 2137
Abstract
The gut microbiota plays an important role in maintaining human health, as well as in the development of various pathologies, as indicated by a large amount of research. One of the manifestations of an imbalance in the gut microbiome composition is the appearance [...] Read more.
The gut microbiota plays an important role in maintaining human health, as well as in the development of various pathologies, as indicated by a large amount of research. One of the manifestations of an imbalance in the gut microbiome composition is the appearance of various diseases or immune reactions, in particular, atopic dermatitis (AD) and/or food allergies (FA). In this research, using 16S NGS sequencing, it was found that the gut microbiome of children with food allergies and children with atopic dermatitis can be characterized as having higher inflammatory potential. Both groups exhibited an abundance of representatives from the Pasteurellaceae and Erysipelotrichaceae families, as well as a decrease in the relative number of representatives from the Barnesiellaceae family compared to healthy participants. In the group of participants with food allergies, there was a decrease in the relative number of Desulfovibrionaceae representatives and Bifidobacteriaceae family enrichment in relatively healthy participants. In addition, when comparing this group with patients with atopic dermatitis, it was revealed that a number of representatives of such families as Erysipelotrichaceae, Ruminococcaceae and Sutterellaceae prevailed. This information confirms that AD and FA correlate with changes in the composition of the gut microbiota. Further research is needed to determine the cause–effect connections and the effect of compounds derived from the microbiota on the AD and FA development and progression, as well as to create new probiotic drugs to prevent and modulate immune responses, including at an early age. Full article
Show Figures

Figure 1

Review

Jump to: Research, Other

15 pages, 820 KiB  
Review
Targeting the Intestinal Microbiota: A Novel Direction in the Treatment of Inflammatory Bowel Disease
by Jie Zhang, Huilin Gan, Xiaoyan Duan and Guangming Li
Biomedicines 2024, 12(10), 2340; https://doi.org/10.3390/biomedicines12102340 (registering DOI) - 15 Oct 2024
Viewed by 401
Abstract
Over the past decade, there has been a rapid increase in the incidence of inflammatory bowel disease. It has been suggested that multifactorial interactions of environmental factors, genetic factors, immune response and intestinal microbiota are involved in the pathogenesis of inflammatory bowel disease. [...] Read more.
Over the past decade, there has been a rapid increase in the incidence of inflammatory bowel disease. It has been suggested that multifactorial interactions of environmental factors, genetic factors, immune response and intestinal microbiota are involved in the pathogenesis of inflammatory bowel disease. It is widely recognized that the intestinal microbiota are essential for human metabolism, the immune system and pathogen resistance, and are integral to human health. Therefore, the dysbiosis of the microbiota is a critical step leading to intestinal mucosal damage and a key factor in the pathogenesis of inflammatory bowel disease. Regulating the microbiota through interventions such as enteral nutrition, fecal microbiota transplantation, and probiotic supplementation has the potential to prevent or even reverse intestinal dysbiosis, opening up new perspectives for the treatment of inflammatory bowel disease. Full article
Show Figures

Figure 1

12 pages, 1016 KiB  
Review
The Contribution of the Brain–Gut Axis to the Human Reward System
by Katerina Karaivazoglou, Ioanna Aggeletopoulou and Christos Triantos
Biomedicines 2024, 12(8), 1861; https://doi.org/10.3390/biomedicines12081861 - 15 Aug 2024
Viewed by 847
Abstract
The human reward network consists of interconnected brain regions that process stimuli associated with satisfaction and modulate pleasure-seeking behaviors. Impairments in reward processing have been implicated in several medical and psychiatric conditions, and there is a growing interest in disentangling the underlying pathophysiological [...] Read more.
The human reward network consists of interconnected brain regions that process stimuli associated with satisfaction and modulate pleasure-seeking behaviors. Impairments in reward processing have been implicated in several medical and psychiatric conditions, and there is a growing interest in disentangling the underlying pathophysiological mechanisms. The brain–gut axis plays a regulatory role in several higher-order neurophysiological pathways, including reward processing. In this context, the aim of the current review was to critically appraise research findings on the contribution of the brain–gut axis to the human reward system. Enteric neuropeptides, which are implicated in the regulation of hunger and satiety, such as ghrelin, PYY3–36, and glucagon-like peptide 1 (GLP-1), have been associated with the processing of food-related, alcohol-related, and other non-food-related rewards, maintaining a delicate balance between the body’s homeostatic and hedonic needs. Furthermore, intestinal microbiota and their metabolites have been linked to differences in the architecture and activation of brain reward areas in obese patients and patients with attention deficit and hyperactivity disorder. Likewise, bariatric surgery reduces hedonic eating by altering the composition of gut microbiota. Although existing findings need further corroboration, they provide valuable information on the pathophysiology of reward-processing impairments and delineate a novel framework for potential therapeutic interventions. Full article
Show Figures

Figure 1

14 pages, 541 KiB  
Review
The Potential Role of Butyrate in the Pathogenesis and Treatment of Autoimmune Rheumatic Diseases
by Carmela Coccia, Francesco Bonomi, Anna Lo Cricchio, Edda Russo, Silvia Peretti, Giulia Bandini, Gemma Lepri, Francesca Bartoli, Alberto Moggi-Pignone, Serena Guiducci, Francesco Del Galdo, Daniel E. Furst, Marco Matucci Cerinic and Silvia Bellando-Randone
Biomedicines 2024, 12(8), 1760; https://doi.org/10.3390/biomedicines12081760 - 5 Aug 2024
Viewed by 1234
Abstract
The gut microbiota is a complex ecosystem of microorganisms residing in the human gastrointestinal tract, playing a crucial role in various biological processes and overall health maintenance. Dysbiosis, an imbalance in the composition and function of the gut microbiota, is linked to systemic [...] Read more.
The gut microbiota is a complex ecosystem of microorganisms residing in the human gastrointestinal tract, playing a crucial role in various biological processes and overall health maintenance. Dysbiosis, an imbalance in the composition and function of the gut microbiota, is linked to systemic autoimmune diseases (SAD). Short-chain fatty acids (SCFAs), especially butyrate, produced by the gut microbiota through the fermentation of dietary fibers, play a significant role in immunomodulation and maintaining intestinal homeostasis. Butyrate is essential for colonocyte energy, anti-inflammatory responses, and maintaining intestinal barrier integrity. Studies show reduced butyrate-producing bacteria in SAD patients, suggesting that increasing butyrate levels could have therapeutic benefits. Butyrate’s anti-inflammatory effects and its potential therapeutic role have been studied in rheumatoid arthritis, Sjogren’s syndrome, systemic lupus erythematosus, systemic sclerosis, and Behçet’s disease. Despite promising in vitro and animal model results, human studies are limited, and the optimal strategies for modulating dysbiosis in SADs remain elusive. This review explores the current evidence on the immunoregulatory role of butyrate and its potential therapeutic effects in SAD. Full article
Show Figures

Figure 1

19 pages, 828 KiB  
Review
Inflammaging: The Next Challenge—Exploring the Role of Gut Microbiota, Environmental Factors, and Sex Differences
by Mario Caldarelli, Pierluigi Rio, Andrea Marrone, Vincenzo Giambra, Antonio Gasbarrini, Giovanni Gambassi and Rossella Cianci
Biomedicines 2024, 12(8), 1716; https://doi.org/10.3390/biomedicines12081716 - 1 Aug 2024
Viewed by 905
Abstract
The term ‘inflammaging’ has been coined to describe the chronic state of inflammation derived from ongoing cycles of tissue damage and the subsequent immune responses. This inflammatory status contributes to the decline of organs and physiological functions, accelerates the aging process, and increases [...] Read more.
The term ‘inflammaging’ has been coined to describe the chronic state of inflammation derived from ongoing cycles of tissue damage and the subsequent immune responses. This inflammatory status contributes to the decline of organs and physiological functions, accelerates the aging process, and increases the risk of age-related illnesses and death. During aging, the gut microbiota (GM) undergoes significant changes, including a decreased diversity of species, a decline in beneficial bacteria, and a rise in proinflammatory ones, resulting in persistent low-grade inflammation. Moreover, environmental factors, such as diet and medications, contribute to age-related changes in GM and immune function, preventing or promoting inflammaging. This narrative review aims to clarify the underlying mechanisms of inflammaging and to specifically investigate the influence of GM and several environmental factors on these mechanisms, while also exploring potential differences related to sex. Moreover, lifestyle and pharmacological interventions will be suggested to promote healthy aging. Full article
Show Figures

Figure 1

34 pages, 1730 KiB  
Review
Small Intestinal Bacterial Overgrowth (SIBO) and Twelve Groups of Related Diseases—Current State of Knowledge
by Paulina Roszkowska, Emilia Klimczak, Ewa Ostrycharz, Aleksandra Rączka, Iwona Wojciechowska-Koszko, Andrzej Dybus, Yeong-Hsiang Cheng, Yu-Hsiang Yu, Szymon Mazgaj and Beata Hukowska-Szematowicz
Biomedicines 2024, 12(5), 1030; https://doi.org/10.3390/biomedicines12051030 - 7 May 2024
Viewed by 5275
Abstract
The human gut microbiota creates a complex microbial ecosystem, characterized by its high population density, wide diversity, and complex interactions. Any imbalance of the intestinal microbiome, whether qualitative or quantitative, may have serious consequences for human health, including small intestinal bacterial overgrowth (SIBO). [...] Read more.
The human gut microbiota creates a complex microbial ecosystem, characterized by its high population density, wide diversity, and complex interactions. Any imbalance of the intestinal microbiome, whether qualitative or quantitative, may have serious consequences for human health, including small intestinal bacterial overgrowth (SIBO). SIBO is defined as an increase in the number of bacteria (103–105 CFU/mL), an alteration in the bacterial composition, or both in the small intestine. The PubMed, Science Direct, Web of Science, EMBASE, and Medline databases were searched for studies on SIBO and related diseases. These diseases were divided into 12 groups: (1) gastrointestinal disorders; (2) autoimmune disease; (3) cardiovascular system disease; (4) metabolic disease; (5) endocrine disorders; (6) nephrological disorders; (7) dermatological diseases; (8) neurological diseases (9); developmental disorders; (10) mental disorders; (11) genetic diseases; and (12) gastrointestinal cancer. The purpose of this comprehensive review is to present the current state of knowledge on the relationships between SIBO and these 12 disease groups, taking into account risk factors and the causal context. This review fills the evidence gap on SIBO and presents a biological–medical approach to the problem, clearly showing the groups and diseases having a proven relationship with SIBO, as well as indicating groups within which research should continue to be expanded. Full article
Show Figures

Figure 1

15 pages, 310 KiB  
Review
Intestinal Dysbiosis: Microbial Imbalance Impacts on Colorectal Cancer Initiation, Progression and Disease Mitigation
by Mary Garvey
Biomedicines 2024, 12(4), 740; https://doi.org/10.3390/biomedicines12040740 - 26 Mar 2024
Cited by 2 | Viewed by 2239
Abstract
The human gastrointestinal tract houses a diverse range of microbial species that play an integral part in many biological functions. Several preclinical studies using germ-free mice models have demonstrated that the gut microbiome profoundly influences carcinogenesis and progression. Colorectal cancer appears to be [...] Read more.
The human gastrointestinal tract houses a diverse range of microbial species that play an integral part in many biological functions. Several preclinical studies using germ-free mice models have demonstrated that the gut microbiome profoundly influences carcinogenesis and progression. Colorectal cancer appears to be associated with microbial dysbiosis involving certain bacterial species, including F. nucleatum, pks+ E. coli, and B. fragilis, with virome commensals also disrupted in patients. A dysbiosis toward these pro-carcinogenic species increases significantly in CRC patients, with reduced numbers of the preventative species Clostridium butyicum, Roseburia, and Bifidobacterium evident. There is also a correlation between Clostridium infection and CRC. F. nucleatum, in particular, is strongly associated with CRC where it is associated with therapeutic resistance and poor outcomes in patients. The carcinogenic mode of action of pathogenic bacteria in CRC is a result of genotoxicity, epigenetic alterations, ROS generation, and pro-inflammatory activity. The aim of this review is to discuss the microbial species and their impact on colorectal cancer in terms of disease initiation, progression, and metastasis. The potential of anticancer peptides as anticancer agents or adjuvants is also discussed, as novel treatment options are required to combat the high levels of resistance to current pharmaceutical options. Full article

Other

Jump to: Research, Review

9 pages, 1050 KiB  
Case Report
Complementary Therapeutic Effect of Fecal Microbiota Transplantation in Ulcerative Colitis after the Response to Anti-Tumor Necrosis Factor Alpha Agent Was Lost: A Case Report
by Jongbeom Shin, Ga Hyeon Baek, Boram Cha, Soo-Hyun Park, Jung-Hwan Lee, Jun-Seob Kim and Kye Sook Kwon
Biomedicines 2024, 12(4), 800; https://doi.org/10.3390/biomedicines12040800 - 3 Apr 2024
Viewed by 1404
Abstract
In patients with ulcerative colitis (UC), the development of an antidrug antibody (ADA) to anti-tumor necrosis factor (TNF)α agent is a crucial problem which aggravates the clinical course of the disease, being cited as one of the most common causes for discontinuing anti-TNFα [...] Read more.
In patients with ulcerative colitis (UC), the development of an antidrug antibody (ADA) to anti-tumor necrosis factor (TNF)α agent is a crucial problem which aggravates the clinical course of the disease, being cited as one of the most common causes for discontinuing anti-TNFα treatment. This is due to ADA eventually causing secondary LOR, leading to discontinuation of anti-TNFα treatment. Recently, research on the microbiome and relationship between worsening UC and dysbiosis has been conducted. Further, investigations on the association between the microbiome and secondary LOR are increasing. Here, we present the therapeutic effect of fecal microbiota transplantation (FMT) on a 42-year-old man with secondary LOR and high ADA levels. FMT has recently been used for the treatment of, and for overcoming, drug resistance through microbiome modification. Stool samples were collected from the patient before and 4 weeks after FMT. Symptoms, including hematochezia and Mayo endoscopy sub-scores, improved after FMT, while ADA levels decreased by one-third to less than half the value (29 ng/mL) compared to before FMT (79 ng/mL). Additionally, the trough level of infliximab became measurable, which reflects the improvement in the area under the concentration (AUC). Butyricicoccus, Faecalibacterium, Bifidobacterium, Ligilactobacillus, Alistipes, and Odoribacter, which regulate immune responses and alleviate inflammation, also increased after FMT. We report a case in which microbiome modification by FMT increased the AUC of anti-TNFα in a patient who developed secondary LOR during anti-TNFα treatment, thereby improving symptoms and mucosal inflammation. Full article
Show Figures

Figure 1

Back to TopTop