nutrients-logo

Journal Browser

Journal Browser

Effects of Diet–Microbiome Interactions on Chronic Diseases

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

Deadline for manuscript submissions: closed (5 October 2023) | Viewed by 35362

Special Issue Editors

National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
Interests: gut microbiota; diabetes; cardiovascular diseases; resistance protein; diet intervention
1. School of Public Health & Management, Wenzhou Medical University, Wenzhou 325035, China
2. Institute of Lipids Medicine, Wenzhou Medical University, Wenzhou 325035, China
Interests: dietary pattern; food intake; nutrient biomarker; fatty acid; cardiometabolic diseases; mental disorders
State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330031, China
Interests: food nutrition and chemistry; development of functional food; food-derived bioactive molecules
Special Issues, Collections and Topics in MDPI journals
State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
Interests: probiotics; gut microbiome; gut health; immune diseases; metabolic syndrome; bacterial functional metabolites

Special Issue Information

Dear Colleagues,

The gut microbiome plays a fundamental role in the development of many chronic diseases ranging from metabolic diseases to cardiovascular diseases and cancers. Diet is the key determinant of the gut microbiota configuration, through a potential modulation of the abundance of specific species and their individual or collective functions. Dietary–microbiome interactions may have affected the development and progression of the above-mentioned chronic diseases. Though accumulating evidence has suggested that gut microbiota and microbial metabolites intermediate various health effects of diet on the host, high-quality cohort study and intervention studies are currently scarce and potential mechanisms are not well elucidated. Furthermore, the complexity of diet patterns and intestinal microflora makes it difficult to find true causal effects/relationships between diet, microbiome, and their interactions with chronic diseases.     

Based on this context, current data from population-based, animal, and in vitro studies, have evidenced the effects of diet–microbiome interactions on chronic diseases and elucidated the mechanisms of action involved in the effects on the host’s health, which would contribute to the prevention and treatment of chronic diseases through dietary intervention mediated by gut microbiota.  

We encourage researchers to submit relevant manuscripts including population-based and basic studies to this Special Issue of Nutrients.

Dr. Yong Xue
Dr. Bo Yang
Dr. Hongyan Li
Dr. Bo Yang
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Nutrients is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • gut microbiota
  • diet intervention
  • cardiovascular diseases
  • mental disorder
  • cancers
  • obesity-related metabolic diseases
  • phytochemicals
  • polysaccharide
  • resistance protein
  • fatty acids

Published Papers (11 papers)

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

Research

Jump to: Review

12 pages, 2011 KiB  
Article
Influence of Lactose Supplementation on Regulation of Streptococcus thermophilus on Gut Microbiota
by Peng Yu, Yuqi Pan, Zhiwen Pei, Min Guo, Bo Yang, Yuan-Kun Lee, Xiaoming Liu, Jianxin Zhao, Hao Zhang and Wei Chen
Nutrients 2023, 15(22), 4767; https://doi.org/10.3390/nu15224767 - 13 Nov 2023
Viewed by 1173
Abstract
It has been found that Streptococcus thermophilus (S. thermophilus) influenced the gut microbiota and host metabolism with strain specificity in C57BL/6J mice in the previous study, though it remains unclear whether lactose as a dietary factor associated with dairy consumption is [...] Read more.
It has been found that Streptococcus thermophilus (S. thermophilus) influenced the gut microbiota and host metabolism with strain specificity in C57BL/6J mice in the previous study, though it remains unclear whether lactose as a dietary factor associated with dairy consumption is involved as the mediator in the interaction. In the present study, integrated analysis of 16S rRNA gene sequencing and untargeted metabolomics by liquid chromatography–mass spectrometry of fecal samples in C57BL/6J mice was applied to evaluate the effect of lactose on the regulation of gut microbiota by two S. thermophilus strains (4M6 and DYNDL13-4). The results showed that the influence of lactose supplementation on gut microbiota induced by S. thermophilus ingestion was strain-specific. Although two S. thermophilus strains ingestion introduced similar perturbations in the fecal microbiota and gut microbial metabolism, the regulation of DYNDL13-4 on the gut microbiota and metabolism was more affected by lactose than 4M6. More specifically, lactose and 4M6 supplementation mainly enriched pathways of d-glutamine and d-glutamate metabolism, alanine, aspartate, and glutamate metabolism, and tryptophan and phenylalanine metabolism in the gut, whereas 4M6 only enriched tryptophan and phenylalanine metabolism. DYNDL13-4-L (DYNDL13-4 with lactose) had significant effects on sulfur, taurine, and hypotaurine metabolism in the gut and on phenylalanine, tyrosine, tryptophan biosynthesis, and linoleic acid metabolism in serum relative to the DYNDL13-4. Our study demonstrated the strain-specific effect of lactose and S. thermophilus supplementation on gut microbiota and host metabolism. However, considering the complexity of the gut microbiota, further research is necessary to provide insights to facilitate the design of personalized fermented milk products as a dietary therapeutic strategy for improving host health. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Graphical abstract

15 pages, 2821 KiB  
Article
Dietary Patterns Are Associated with the Gut Microbiome and Metabolic Syndrome in Mexican Postmenopausal Women
by Priscilla López-Montoya, Berenice Rivera-Paredez, Berenice Palacios-González, Sofia Morán-Ramos, Blanca E. López-Contreras, Samuel Canizales-Quinteros, Jorge Salmerón and Rafael Velázquez-Cruz
Nutrients 2023, 15(22), 4704; https://doi.org/10.3390/nu15224704 - 7 Nov 2023
Viewed by 1595
Abstract
Postmenopausal women are at an increased risk of developing metabolic syndrome (MetS) due to hormonal changes and lifestyle factors. Gut microbiota (GM) have been linked to the development of MetS, and they are influenced by dietary habits. However, the interactions between dietary patterns [...] Read more.
Postmenopausal women are at an increased risk of developing metabolic syndrome (MetS) due to hormonal changes and lifestyle factors. Gut microbiota (GM) have been linked to the development of MetS, and they are influenced by dietary habits. However, the interactions between dietary patterns (DP) and the GM of postmenopausal women, as well as their influence on MetS, still need to be understood. The present study evaluated the DP and microbiota composition of postmenopausal Mexican women with MetS and those in a control group. Diet was assessed using a food frequency questionnaire, and the GM were profiled using 16S rRNA gene sequencing. Greater adherence to a “healthy” DP was significantly associated with lower values of MetS risk factors. GM diversity was diminished in women with MetS, and it was negatively influenced by an “unhealthy” DP. Moreover, a higher intake of fats and proteins, as well as lower amounts of carbohydrates, showed a reduction in some of the short-chain fatty acid-producing genera in women with MetS, as well as increases in some harmful bacteria. Furthermore, Roseburia abundance was positively associated with dietary fat and waist circumference, which may explain 7.5% of the relationship between this macronutrient and MetS risk factors. These findings suggest that GM and diet interactions are important in the development of MetS in postmenopausal Mexican women. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

31 pages, 8610 KiB  
Article
The Antioxidant and Anti-Inflammatory Effects of the Main Carotenoids from Tomatoes via Nrf2 and NF-κB Signaling Pathways
by Wenxiu Ba, Wenzhen Xu, Zeyuan Deng, Bing Zhang, Liufeng Zheng and Hongyan Li
Nutrients 2023, 15(21), 4652; https://doi.org/10.3390/nu15214652 - 2 Nov 2023
Cited by 3 | Viewed by 1496
Abstract
Oxidative stress and inflammation are crucial factors in the development of cardiovascular diseases. In previous research, the oxidative stress and inflammation models have frequently been explored independently. In the current study, we investigated the antioxidant and anti-inflammatory effects of tomato extract and its [...] Read more.
Oxidative stress and inflammation are crucial factors in the development of cardiovascular diseases. In previous research, the oxidative stress and inflammation models have frequently been explored independently. In the current study, we investigated the antioxidant and anti-inflammatory effects of tomato extract and its two main carotenoids (lutein and lycopene) with various concentrations using a rat cardiomyocyte model of co-existing oxidative stress and persistent chronic inflammation. It was discovered that the antioxidant effects of 0.5–5 μM lutein, 0.5–5 μM lycopene, and 50–200 μg/mL tomato extract increased in a dose-dependent manner. However, the pro-oxidation effects emerged by measuring the antioxidant-related indices, including the levels of ROS, SOD, and GPX in H9c2 cells as concentrations exceeded those mentioned above. The anti-inflammatory effects of lutein, lycopene, and tomato extract were simultaneously strengthened with higher concentrations, potentially due to the suppression of the NF-κB signaling pathway. Furthermore, high concentrations of lutein, lycopene, and tomato extract potentially regulated Nrf2/HO-1 and NF-κB signaling pathways dependent on TGF-1β and IL-10 to demonstrate high concentrations of pro-oxidation and anti-inflammation effects. Our findings indicate that the dose–effect regulatory mechanisms of antioxidant and anti-inflammatory properties among lutein, lycopene, and tomato extract will be advantageous in developing more effective therapeutic strategies to prevent cardiovascular diseases. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

14 pages, 13737 KiB  
Article
Enterococcus faecium HDRsEf1 Promotes Systemic Th1 Responses and Enhances Resistance to SalmonellaTyphimurium Infection
by Jin Zhou, Tingyang Wang, Lele Fan, Hongde Xiao, Hui Ji, Naiji Zhou, Zutao Zhou, Huazhen Liu, Muhammad Akhtar, Yuncai Xiao and Deshi Shi
Nutrients 2023, 15(19), 4241; https://doi.org/10.3390/nu15194241 - 30 Sep 2023
Viewed by 1067
Abstract
The gut microbiota is known to regulate the immune system and thereby influence susceptibility to infection. In this study, we observed that the administration of Enterococcus faecium HDRsEf1 (HDRsEf1) led to an improvement in the development of the immune system. This was evidenced [...] Read more.
The gut microbiota is known to regulate the immune system and thereby influence susceptibility to infection. In this study, we observed that the administration of Enterococcus faecium HDRsEf1 (HDRsEf1) led to an improvement in the development of the immune system. This was evidenced by an increase in both the spleen index and the area of spleen white pulp. Specifically, the proportion of T helper (Th) 1 cells and the production of IFN-γ and IL-12 were significantly increased in the spleens of mice treated with HDRsEf1. In agreement with the in vivo results, we found that Th1-related cytokines, including IFN-γ and IL-12p70, were strongly induced in splenocytes treated with HDRsEf1. In addition, Th1 cell activation and high-level secretion of IL-12p70 were also confirmed by coculture of CD4+ T cells with bone marrow-derived dendritic cells treated with HDRsEf1. Moreover, the employment of HDRsEf1 was identified to augment resilience against systemic infection provoked by S. Typhimurium and stimulate the expression of the genes for TNFα and iNOS in the initial stage of infection, signifying that reinforced Th1 cells and IL-12 might activate macrophages for antibacterial safeguards. In summary, our study suggests that HDRsEf1 could act as an effective immunobiotic functional agent, promoting systemic Th1 immunological responses and priming defenses against infection. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

13 pages, 2676 KiB  
Article
Whole-Grain Highland Barley Attenuates Atherosclerosis Associated with NLRP3 Inflammasome Pathway and Gut Microbiota in ApoE−/− Mice
by Tong Wu, Qinye Yu, Yingting Luo, Zijian Dai, Yuhong Zhang, Chao Wang, Qun Shen and Yong Xue
Nutrients 2023, 15(19), 4186; https://doi.org/10.3390/nu15194186 - 28 Sep 2023
Cited by 1 | Viewed by 1165
Abstract
The efficacy and mechanism of highland barley in the treatment of atherosclerosis have received little attention. Herein, we aimed to explore whether highland barley supplementation can prevent atherosclerosis progression and improve gut microbiota disorder in apolipoprotein E knockout (ApoE−/−) mice. Male [...] Read more.
The efficacy and mechanism of highland barley in the treatment of atherosclerosis have received little attention. Herein, we aimed to explore whether highland barley supplementation can prevent atherosclerosis progression and improve gut microbiota disorder in apolipoprotein E knockout (ApoE−/−) mice. Male ApoE−/− mice were fed a high-fat diet with whole-grain highland barley (WHB) or refined highland barley for 18 weeks. WHB substantially inhibited the formation of atherosclerotic plaques, reduced serum tumor necrosis factor-α, and downregulated the expression of NLRP3 in the aorta. Furthermore, the 16S rRNA analysis revealed that highland barley supplementation helped to restore the dysregulation of the gut microbiota, as evidenced by an increase in the relative abundance of specific beneficial bacteria known for their anti-inflammatory properties, such as Lachnospiraceae, Lactobacillus, Muribaculaceae, and Bifidobacterium. Highland barley supplementation might alleviate atherosclerotic plaque formation by modulating the NLRP3 inflammasome pathway and the synthesis of anti-inflammatory metabolites by the gut microbiota. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

15 pages, 5597 KiB  
Article
Bacteriocin-Producing Lactiplantibacillus plantarum YRL45 Enhances Intestinal Immunity and Regulates Gut Microbiota in Mice
by Yushan Bu, Yisuo Liu, Tai Zhang, Yinxue Liu, Zhe Zhang and Huaxi Yi
Nutrients 2023, 15(15), 3437; https://doi.org/10.3390/nu15153437 - 3 Aug 2023
Cited by 6 | Viewed by 1781
Abstract
Bacteriocins production is one of important beneficial characteristics of probiotics, which has antibacterial property against intestinal pathogens and is helpful for regulating intestinal flora. To investigate the impact of bacteriocin-producing probiotics on gut microecology, bacteriocin-producing Lactiplantibacillus plantarum YRL45 was orally administered to mice. [...] Read more.
Bacteriocins production is one of important beneficial characteristics of probiotics, which has antibacterial property against intestinal pathogens and is helpful for regulating intestinal flora. To investigate the impact of bacteriocin-producing probiotics on gut microecology, bacteriocin-producing Lactiplantibacillus plantarum YRL45 was orally administered to mice. The results revealed that it promoted the release of cytokines and improved the phagocytic activity of peritoneal macrophages to activate the immune regulation system. L. plantarum YRL45 was conducive to maintaining the morphology of colon tissue without inflammation and increasing the ratio of villus height to crypt depth in the ileum. The gene expression levels of Muc2, ZO-1 and JAM-1 were significantly up-regulated in the ileum and colon, and the gene expression of Cramp presented an upward trend with L. plantarum YRL45 intervention. Moreover, L. plantarum YRL45 remarkably enhanced the levels of immunoglobulins sIgA, IgA and IgG in the intestine of mice. The 16S rRNA gene analysis suggested that L. plantarum YRL45 administration up-regulated the relative abundance of the beneficial bacteria Muribaculaceae and Akkermansia, down-regulated the abundance of the pathogenic bacteria Lachnoclostridium, and promoted the production of acetic acid, propionic acid and total short-chain fatty acids (SCFAs) in mice feces. Our findings indicated that L. plantarum YRL45 had the potential to be developed as a novel probiotic to regulate the intestinal barrier by altering gut microbiota to enhance intestinal immunity and ameliorate intestinal flora balance. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Graphical abstract

15 pages, 2040 KiB  
Article
Maternal Betaine Supplementation Mitigates Maternal High Fat Diet-Induced NAFLD in Offspring Mice through Gut Microbiota
by Liuqiao Sun, Xuying Tan, Xiaoping Liang, Hangjun Chen, Qian Ou, Qiongmei Wu, Xinxue Yu, Hanqing Zhao, Qiaoli Huang, Zehua Yi, Jun Wei, Feng Wu, Huilian Zhu and Lijun Wang
Nutrients 2023, 15(2), 284; https://doi.org/10.3390/nu15020284 - 6 Jan 2023
Cited by 10 | Viewed by 2730
Abstract
Maternal betaine supplementation has been proven to alleviate non-alcoholic fatty liver disease (NAFLD) in offspring caused by maternal high-fat diet (MHFD). The gut–liver axis plays an important role in NAFLD pathogenesis. However, whether maternal betaine supplementation can alleviate NAFLD in offspring by the [...] Read more.
Maternal betaine supplementation has been proven to alleviate non-alcoholic fatty liver disease (NAFLD) in offspring caused by maternal high-fat diet (MHFD). The gut–liver axis plays an important role in NAFLD pathogenesis. However, whether maternal betaine supplementation can alleviate NAFLD in offspring by the gut–liver axis is unknown. C57BL/6J mice were fed with high-fat diet for 4 weeks before mating, and supplemented with 1% betaine during pregnancy and lactation. After weaning, offspring mice were fed with standard diet to 10 weeks. Maternal betaine supplementation reduced hepatic triglyceride content and alleviated hepatic steatosis in offspring mice exposed to MHFD. Furthermore, the mRNA expression of PPARα, CPT1α and FATP2 was increased and TNFα was reduced by maternal betaine supplementation. Maternal betaine intake decreased the relative abundances of Proteobateria, Desulfovibrio and Ruminococcus, but increased the relative abundances of Bacteroides and Parabacteroides. Moreover, maternal betaine intake increased the concentrations of short-chain fatty acids (SCFAs), including acetic acid, butyric acid and valeric acid, in the feces. Gut microbiota and SCFAs were significantly correlated with hepatic triglyceride content and expression of the above genes. Maternal betaine intake had no effect on other gut microbiota-related metabolites (bile acid and trimethylamine-n-oxide). Altogether, maternal betaine supplementation ameliorated MHFD-induced NAFLD possibly through regulating gut microbiota and SCFAs in offspring mice. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

16 pages, 6436 KiB  
Article
Gut Microbiome and Serum Metabolome Profiles of Capsaicin with Cognitive Benefits in APP/PS1 Mice
by Jun Li, Xiaojun Liao, Xuedong Yin, Zimeng Deng, Guangfen Hu, Weiwei Zhang, Feng Jiang and Liang Zhao
Nutrients 2023, 15(1), 118; https://doi.org/10.3390/nu15010118 - 27 Dec 2022
Cited by 3 | Viewed by 3197
Abstract
Capsaicin, a natural bioactive component, has been reported to improve cognition and ameliorate the pathology of Alzheimer’s disease (AD). Studies have linked AD to alterations in gut microbiota composition and serum metabolites. In the present study, we examined the alterations in serum metabolome [...] Read more.
Capsaicin, a natural bioactive component, has been reported to improve cognition and ameliorate the pathology of Alzheimer’s disease (AD). Studies have linked AD to alterations in gut microbiota composition and serum metabolites. In the present study, we examined the alterations in serum metabolome and gut microbiome in APPswe/PS1dE9 (APP/PS1) mice treated with capsaicin. Capsaicin treatments resulted in a significant increase in the abundance of Akkermansia, Faecalibaculum, Unclassified_f_Atopobiaceae, and Gordonibacter and a significant decrease in the abundance of Adlercreutzia, Peptococcaceae, Alistipes, Oscillibacter and Erysipelatoclostridium. Furthermore, the species Akkermansia muciniphila (A. muciniphila) was significantly enriched in capsaicin-treated APP/PS1 mice (p = 0.0002). Serum metabolomic analysis showed that capsaicin-treated APP/PS1 mice had a significant higher level of tryptophan (Trp) metabolism and a significantly lower level of lipid metabolism compared with vehicle-treated mice. Capsaicin altered serum metabolites, including Kynurenine (Kyn), 5-Hydroxy-L-tryptophan (5-HIT), 5-Hydroxyindoleacetic acid (5-HIAA), indoxylsulfuric acid, lysophosphatidyl cholines (LysoPCs), and lysophosphatidyl ethanolamine (LysoPE). Significant correlations were observed between the gut bacteria and serum metabolite. With regard to the increased abundance of A. muciniphila and the ensuing rise in tryptophan metabolites, our data show that capsaicin alters both the gut microbiota and blood metabolites. By altering the gut microbiome and serum metabolome, a diet high in capsaicin may reduce the incidence and development of AD. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Graphical abstract

17 pages, 6450 KiB  
Article
Effects of Bifidobacterium with the Ability of 2′-Fucosyllactose Utilization on Intestinal Microecology of Mice
by Bingyong Mao, Zhujun He, Yang Chen, Catherine Stanton, Reynolds Paul Ross, Jianxin Zhao, Wei Chen and Bo Yang
Nutrients 2022, 14(24), 5392; https://doi.org/10.3390/nu14245392 - 19 Dec 2022
Cited by 1 | Viewed by 2066
Abstract
In breast milk, 2′-Fucosyllactose (2′FL) is the most abundant breast milk oligosaccharide and can selectively promote the proliferation of bifidobacteria. This study aimed to explore the effect of ifidobacterial with different utilization capacities of 2′FL on the intestinal microecology of mice. Furthermore, the [...] Read more.
In breast milk, 2′-Fucosyllactose (2′FL) is the most abundant breast milk oligosaccharide and can selectively promote the proliferation of bifidobacteria. This study aimed to explore the effect of ifidobacterial with different utilization capacities of 2′FL on the intestinal microecology of mice. Furthermore, the effects of ifidobacterial with different 2′FL utilization capabilities on mice gut microbiota under the competitive pressure of 2′FL as a carbon source were explored. Compared with the control group, 2′FL, Bifidobacterium (B.) bifidum M130R01M51 + 2′FL, B. longum subsp. Longum CCFM752, and CCFM752 + 2′FL treatments significantly decreased the food intake. Moreover, the water intake, body weight, and fecal water content in all groups showed no significant difference compared with the control group. The combination of B. longum subsp. longum CCFM752 and 2′FL can significantly increase the levels of pro-inflammatory and anti-inflammatory factors. B. bifidum M130R01M51 and mixed strains combined with 2′FL significantly increased the contents of acetic acid and isobutyric acid. The results showed that B. bifidum M130R01M51, B. breve FHuNCS6M1, B. longum subsp. longum CCFM752, and B. longum subsp. infantis SDZC2M4 combined with 2′FL significantly increased the species richness of the gut microbiota. Moreover, B. longum subsp. longum CCFM752 and B. longum subsp. infantis SDZC2M4 significantly increased the abundance of Faecalibaculum and Bifidobacterium, respectively. In conclusion, exploring the impact on intestinal microecology can provide theoretical guidance for the development of personalized prebiotics for different bifidobacteria, which has the potential to improve the ecological imbalance of infant gut microbiota. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

Review

Jump to: Research

20 pages, 1374 KiB  
Review
The Gut–Brain Axis in Schizophrenia: The Implications of the Gut Microbiome and SCFA Production
by Songhyun Ju, Yoonhwa Shin, Sunhee Han, Juhui Kwon, Tae Gyu Choi, Insug Kang and Sung Soo Kim
Nutrients 2023, 15(20), 4391; https://doi.org/10.3390/nu15204391 - 16 Oct 2023
Cited by 4 | Viewed by 4317
Abstract
Schizophrenia, a severe mental illness affecting about 1% of the population, manifests during young adulthood, leading to abnormal mental function and behavior. Its multifactorial etiology involves genetic factors, experiences of adversity, infection, and gene–environment interactions. Emerging research indicates that maternal infection or stress [...] Read more.
Schizophrenia, a severe mental illness affecting about 1% of the population, manifests during young adulthood, leading to abnormal mental function and behavior. Its multifactorial etiology involves genetic factors, experiences of adversity, infection, and gene–environment interactions. Emerging research indicates that maternal infection or stress during pregnancy may also increase schizophrenia risk in offspring. Recent research on the gut–brain axis highlights the gut microbiome’s potential influence on central nervous system (CNS) function and mental health, including schizophrenia. The gut microbiota, located in the digestive system, has a significant role to play in human physiology, affecting immune system development, vitamin synthesis, and protection against pathogenic bacteria. Disruptions to the gut microbiota, caused by diet, medication use, environmental pollutants, and stress, may lead to imbalances with far-reaching effects on CNS function and mental health. Of interest are short-chain fatty acids (SCFAs), metabolic byproducts produced by gut microbes during fermentation. SCFAs can cross the blood–brain barrier, influencing CNS activity, including microglia and cytokine modulation. The dysregulation of neurotransmitters produced by gut microbes may contribute to CNS disorders, including schizophrenia. This review explores the potential relationship between SCFAs, the gut microbiome, and schizophrenia. Our aim is to deepen the understanding of the gut–brain axis in schizophrenia and to elucidate its implications for future research and therapeutic approaches. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

20 pages, 1563 KiB  
Review
The Role of Gut Microbiota in Anxiety, Depression, and Other Mental Disorders as Well as the Protective Effects of Dietary Components
by Ruo-Gu Xiong, Jiahui Li, Jin Cheng, Dan-Dan Zhou, Si-Xia Wu, Si-Yu Huang, Adila Saimaiti, Zhi-Jun Yang, Ren-You Gan and Hua-Bin Li
Nutrients 2023, 15(14), 3258; https://doi.org/10.3390/nu15143258 - 23 Jul 2023
Cited by 12 | Viewed by 13538
Abstract
The number of individuals experiencing mental disorders (e.g., anxiety and depression) has significantly risen in recent years. Therefore, it is essential to seek prevention and treatment strategies for mental disorders. Several gut microbiota, especially Firmicutes and Bacteroidetes, are demonstrated to affect mental health [...] Read more.
The number of individuals experiencing mental disorders (e.g., anxiety and depression) has significantly risen in recent years. Therefore, it is essential to seek prevention and treatment strategies for mental disorders. Several gut microbiota, especially Firmicutes and Bacteroidetes, are demonstrated to affect mental health through microbiota–gut–brain axis, and the gut microbiota dysbiosis can be related to mental disorders, such as anxiety, depression, and other mental disorders. On the other hand, dietary components, including probiotics (e.g., Lactobacillus and Bifidobacterium), prebiotics (e.g., dietary fiber and alpha-lactalbumin), synbiotics, postbiotics (e.g., short-chain fatty acids), dairy products, spices (e.g., Zanthoxylum bungeanum, curcumin, and capsaicin), fruits, vegetables, medicinal herbs, and so on, could exert protective effects against mental disorders by enhancing beneficial gut microbiota while suppressing harmful ones. In this paper, the mental disorder-associated gut microbiota are summarized. In addition, the protective effects of dietary components on mental health through targeting the gut microbiota are discussed. This paper can be helpful to develop some dietary natural products into pharmaceuticals and functional foods to prevent and treat mental disorders. Full article
(This article belongs to the Special Issue Effects of Diet–Microbiome Interactions on Chronic Diseases)
Show Figures

Figure 1

Back to TopTop