Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (355)

Search Parameters:
Keywords = diet–microbiota relationship

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 2171 KiB  
Review
Induction of Autophagy as a Therapeutic Breakthrough for NAFLD: Current Evidence and Perspectives
by Yanke Liu, Mingkang Zhang and Yazhi Wang
Biology 2025, 14(8), 989; https://doi.org/10.3390/biology14080989 (registering DOI) - 4 Aug 2025
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a clinicopathological syndrome characterised by hepatic steatosis in the absence of significant alcohol consumption or other specific causes of liver injury. It has become one of the leading causes of liver dysfunction worldwide. However, the precise pathophysiological [...] Read more.
Nonalcoholic fatty liver disease (NAFLD) is a clinicopathological syndrome characterised by hepatic steatosis in the absence of significant alcohol consumption or other specific causes of liver injury. It has become one of the leading causes of liver dysfunction worldwide. However, the precise pathophysiological mechanisms underlying NAFLD remain unclear, and effective therapeutic strategies are still under investigation. Autophagy, a vital intracellular process in eukaryotic cells, enables the degradation and recycling of cytoplasmic components through a membrane trafficking pathway. Recent studies have demonstrated a strong association between impaired or deficient autophagy and the development and progression of NAFLD. Restoring autophagic function may represent a key approach to mitigating hepatocellular injury. Nevertheless, due to the complexity of autophagy regulation and its context-dependent effects on cellular function, therapeutic strategies targeting autophagy in NAFLD remain limited. This review aims to summarise the relationship between autophagy and NAFLD, focusing on autophagy as a central mechanism. We discuss the latest research advances regarding interventions such as diet and exercise, pharmacological therapies (including modern pharmacological therapy and plant-derived compounds), and other approaches (such as hormones, nanoparticles, gut microbiota, and vitamins). Furthermore, we briefly highlight potential autophagy-related molecular targets that may offer novel therapeutic insights for NAFLD management. Full article
(This article belongs to the Section Medical Biology)
Show Figures

Figure 1

21 pages, 1024 KiB  
Review
The Impact of Environmental Factors on the Secretion of Gastrointestinal Hormones
by Joanna Smarkusz-Zarzecka, Lucyna Ostrowska and Marcelina Radziszewska
Nutrients 2025, 17(15), 2544; https://doi.org/10.3390/nu17152544 - 2 Aug 2025
Viewed by 216
Abstract
The enteroendocrine system of the gastrointestinal (GI) tract is the largest endocrine organ in the human body, playing a central role in the regulation of hunger, satiety, digestion, and energy homeostasis. Numerous factors—including dietary components, physical activity, and the gut microbiota—affect the secretion [...] Read more.
The enteroendocrine system of the gastrointestinal (GI) tract is the largest endocrine organ in the human body, playing a central role in the regulation of hunger, satiety, digestion, and energy homeostasis. Numerous factors—including dietary components, physical activity, and the gut microbiota—affect the secretion of GI hormones. This study aims to analyze how these factors modulate enteroendocrine function and influence systemic metabolic regulation. This review synthesizes the current scientific literature on the physiology and distribution of enteroendocrine cells and mechanisms of hormone secretion in response to macronutrients, physical activity, and microbial metabolites. Special attention is given to the interactions between gut-derived signals and central nervous system pathways involved in appetite control. Different GI hormones are secreted in specific regions of the digestive tract in response to meal composition and timing. Macronutrients, particularly during absorption, stimulate hormone release, while physical activity influences hormone concentrations, decreasing ghrelin and increasing GLP-1, PYY, and leptin levels. The gut microbiota, through fermentation and metabolite production (e.g., SCFAs and bile acids), modulates enteroendocrine activity. Species such as Akkermansia muciniphila are associated with improved gut barrier integrity and enhanced GLP-1 secretion. These combined effects contribute to appetite regulation and energy balance. Diet composition, physical activity, and gut microbiota are key modulators of gastrointestinal hormone secretion. Their interplay significantly affects appetite regulation and metabolic health. A better understanding of these relationships may support the development of personalized strategies for managing obesity and related disorders. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Graphical abstract

13 pages, 436 KiB  
Opinion
It Is Time to Consider the Lost Battle of Microdamaged Piezo2 in the Context of E. coli and Early-Onset Colorectal Cancer
by Balázs Sonkodi
Int. J. Mol. Sci. 2025, 26(15), 7160; https://doi.org/10.3390/ijms26157160 - 24 Jul 2025
Viewed by 337
Abstract
The recent identification of early-onset mutational signatures with geographic variations by Diaz-Gay et al. is a significant finding, since early-onset colorectal cancer has emerged as an alarming public health challenge in the past two decades, and the pathomechanism remains unclear. Environmental risk factors, [...] Read more.
The recent identification of early-onset mutational signatures with geographic variations by Diaz-Gay et al. is a significant finding, since early-onset colorectal cancer has emerged as an alarming public health challenge in the past two decades, and the pathomechanism remains unclear. Environmental risk factors, including lifestyle and diet, are highly suspected. The identification of colibactin from Escherichia coli as a potential pathogenic source is a major step forward in addressing this public health challenge. Therefore, the following opinion manuscript aims to outline the likely onset of the pathomechanism and the critical role of acquired Piezo2 channelopathy in early-onset colorectal cancer, which skews proton availability and proton motive force regulation toward E. coli within the microbiota–host symbiotic relationship. In addition, the colibactin produced by the pks island of E. coli induces host DNA damage, which likely interacts at the level of Wnt signaling with Piezo2 channelopathy-induced pathological remodeling. This transcriptional dysregulation eventually leads to tumorigenesis of colorectal cancer. Mechanotransduction converts external physical cues to inner chemical and biological ones. Correspondingly, the proposed quantum mechanical free-energy-stimulated ultrafast proton-coupled tunneling, initiated by Piezo2, seems to be the principal and essential underlying novel oscillatory signaling that could be lost in colorectal cancer onset. Hence, Piezo2 channelopathy not only contributes to cancer initiation and impaired circadian regulation, including the proposed hippocampal ultradian clock, but also to proliferation and metastasis. Full article
(This article belongs to the Special Issue Advanced Research of Gut Microbiota and Toxins)
Show Figures

Figure 1

13 pages, 4863 KiB  
Article
p53 Protein Stability Plays a Crucial Role in NaB-Mediated Apoptosis in Colorectal Cancer Cells
by Jeong Yeon Lee and Hyunju Kim
Curr. Issues Mol. Biol. 2025, 47(8), 579; https://doi.org/10.3390/cimb47080579 - 22 Jul 2025
Viewed by 332
Abstract
Colorectal cancer (CRC) is associated with factors such as an unhealthy diet, physical inactivity, obesity, diabetes, and chronic inflammatory conditions like inflammatory bowel disease (IBD), as well as TP53 mutations, which are observed in a broad spectrum of CRC. Additionally, alteration in the [...] Read more.
Colorectal cancer (CRC) is associated with factors such as an unhealthy diet, physical inactivity, obesity, diabetes, and chronic inflammatory conditions like inflammatory bowel disease (IBD), as well as TP53 mutations, which are observed in a broad spectrum of CRC. Additionally, alteration in the composition of the gut microbiome community and metabolism plays a significant role in the development of colorectal cancer and its therapeutic effects. It is well known that treatment with sodium butyrate (NaB), an intestinal microbial metabolite, can induce apoptosis by activating histone deacetylase (HDAC) in cancer cells. Therefore, this study examined the relationship between NaB-induced apoptosis and p53 protein level in colorectal cancer cells. Treatment with NaB triggered cell death in the HCT116 cell line. Furthermore, a notable elevation in p53 protein level was detected following treatment with a high concentration of NaB, compared to both the control group and the low concentration NaB. Furthermore, apoptotic cell death was diminished in a p53-deficient cell line (HCT 116 p53−/−) and p53 protein expression was more stabilized. Although p53 mRNA expression was not affected, acetylation of p53 protein was clearly observed by high concentration NaB treatment. To demonstrate the relationship between p53 acetylation and cell death, HT29 cells were treated with a high concentration of NaB. In HT29 cells with a mutation in the p53 gene, increased cell viability, overproduction p53 protein, and hyperacetylation of p53 were observed compared to the control. The results of this study suggest that p53 protein expression plays an important role in the effectiveness of therapy utilizing gut microbiota metabolites. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

15 pages, 1142 KiB  
Article
The Estimated Intake of S100B Relates to Microbiota Biodiversity in Different Diets
by Tehreema Ghaffar, Veronica Volpini, Serena Platania, Olga Vassioukovitch, Alessandra Valle, Federica Valeriani, Fabrizio Michetti and Vincenzo Romano Spica
Biomolecules 2025, 15(7), 1047; https://doi.org/10.3390/biom15071047 - 18 Jul 2025
Viewed by 362
Abstract
The S100B protein, known for its role in the central and enteric nervous systems, has recently been identified in dietary sources such as milk, dairy products, fruits, and vegetables. Given its potential interaction with the gut microbiota, this study explores the relationship between [...] Read more.
The S100B protein, known for its role in the central and enteric nervous systems, has recently been identified in dietary sources such as milk, dairy products, fruits, and vegetables. Given its potential interaction with the gut microbiota, this study explores the relationship between dietary intake of S100B and microbiota biodiversity across different diets. A comprehensive study was conducted, estimating S100B concentrations in 13 dietary patterns recommended in different countries. This is the first study to provide a comparative estimation of S100B exposure from the diet and to explore its potential ecological and epidemiological relevance. The association between S100B levels and microbiota biodiversity was statistically analyzed, showing a direct correlation. Microbial diversity was assessed using the Shannon index, based on data extracted from studies reporting microbiota composition across dietary patterns. Additionally, the relative risk of Crohn’s disease was assessed in different populations to examine potential links between dietary patterns, S100B, and chronic disease prevention. A moderate positive correlation (R2 = 0.537) was found between S100B concentration and Shannon index, suggesting that diets higher in S100B (e.g., Mediterranean diet) were associated with higher microbial alpha-diversity. Furthermore, Western-style diets, with the lowest S100B levels, exhibited a higher relative risk for Crohn’s disease (R2 = 0.780). These findings highlight the potential role of dietary S100B content in modulating gut microbiota diversity and reducing chronic disease risk. Full article
(This article belongs to the Section Biomacromolecules: Proteins, Nucleic Acids and Carbohydrates)
Show Figures

Graphical abstract

13 pages, 1189 KiB  
Review
The Role of the Gut Microbiota in Mental Health and Cognitive Function in Patients with Coronary Atherosclerosis
by Paulina Helisz, Karolina Krupa-Kotara, Weronika Gwioździk and Joanna Głogowska-Ligus
Nutrients 2025, 17(14), 2311; https://doi.org/10.3390/nu17142311 - 14 Jul 2025
Viewed by 531
Abstract
The gut microbiota plays an important role in maintaining the body’s homeostasis, and its disruption has been linked to the pathogenesis of coronary atherosclerosis and cognitive decline. This review attempted to assess whether the composition of the gut microbiota differs significantly according to [...] Read more.
The gut microbiota plays an important role in maintaining the body’s homeostasis, and its disruption has been linked to the pathogenesis of coronary atherosclerosis and cognitive decline. This review attempted to assess whether the composition of the gut microbiota differs significantly according to the severity of coronary atherosclerosis and whether the presence of specific cytokines and inflammatory markers in the microbiota of patients with atherosclerosis may correlate with cognitive impairment. In addition, it considered whether increased dietary fiber intake may contribute to lower levels of inflammatory markers compared to a low-fiber diet. This review included publications from 2015 to 2024, searched in the PubMed and Scopus databases. Only studies meeting the quality criteria were included. The pooled data indicate that intestinal dysbiosis can lead to increased intestinal barrier permeability and lipopolysaccharide (LPS) translocation, which promotes chronic inflammation. This process plays an important role in both atherosclerosis and neurodegeneration. In addition, some studies indicate a beneficial effect of dietary fiber in reducing inflammatory markers. The conclusions of this review highlight the need for further, well-designed studies to identify the causal relationship between the microbiota, its metabolites, atherosclerosis, and cognitive deficits, which may provide the basis for new therapeutic strategies. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota, and Gastrointestinal Disease)
Show Figures

Figure 1

25 pages, 1644 KiB  
Review
The Role of Gut Microbiota in the Development and Treatment of Obesity and Overweight: A Literature Review
by Gabriela Augustynowicz, Maria Lasocka, Hubert Paweł Szyller, Marta Dziedziak, Agata Mytych, Joanna Braksator and Tomasz Pytrus
J. Clin. Med. 2025, 14(14), 4933; https://doi.org/10.3390/jcm14144933 - 11 Jul 2025
Viewed by 634
Abstract
The gut microbiota, dominated by bacteria from the Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria phyla, plays an essential role in fermenting indigestible carbohydrates, regulating metabolism, synthesizing vitamins, and maintaining immune functions and intestinal barrier integrity. Dysbiosis is associated with obesity development. Shifts in the [...] Read more.
The gut microbiota, dominated by bacteria from the Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria phyla, plays an essential role in fermenting indigestible carbohydrates, regulating metabolism, synthesizing vitamins, and maintaining immune functions and intestinal barrier integrity. Dysbiosis is associated with obesity development. Shifts in the ratio of Firmicutes to Bacteroidetes, particularly an increase in Firmicutes, may promote enhanced energy storage, appetite dysregulation, and increased inflammatory processes linked to insulin resistance and other metabolic disorders. The purpose of this literature review is to summarize the current state of knowledge on the relationship between the development and treatment of obesity and overweight and the gut microbiota. Current evidence suggests that probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT) can influence gut microbiota composition and metabolic parameters, including body weight and BMI. The most promising effects are observed with probiotic supplementation, particularly when combined with prebiotics, although efficacy depends on strain type, dose, and duration. Despite encouraging preclinical findings, FMT has shown limited and inconsistent results in human studies. Diet and physical activity are key modulators of the gut microbiota. Fiber, plant proteins, and omega-3 fatty acids support beneficial bacteria, while diets low in fiber and high in saturated fats promote dysbiosis. Aerobic exercise increases microbial diversity and supports growth of favorable bacterial strains. While microbiota changes do not always lead to immediate weight loss, modulating gut microbiota represents an important aspect of obesity prevention and treatment strategies. Further research is necessary to better understand the mechanisms and therapeutic potential of these interventions. Full article
(This article belongs to the Special Issue Metabolic Syndrome and Its Burden on Global Health)
Show Figures

Figure 1

28 pages, 933 KiB  
Review
Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders
by Shabnam Nohesara, Hamid Mostafavi Abdolmaleky, Ahmad Pirani and Sam Thiagalingam
Cells 2025, 14(13), 1027; https://doi.org/10.3390/cells14131027 - 4 Jul 2025
Viewed by 781
Abstract
Neuroinflammation is a hallmark of many neuropsychiatric disorders (NPD), which are among the leading causes of disability worldwide. Emerging evidence highlights the significant role of the gut microbiota (GM)–immune system–brain axis in neuroinflammation and the pathogenesis of NPD, primarily through epigenetic mechanisms. Gut [...] Read more.
Neuroinflammation is a hallmark of many neuropsychiatric disorders (NPD), which are among the leading causes of disability worldwide. Emerging evidence highlights the significant role of the gut microbiota (GM)–immune system–brain axis in neuroinflammation and the pathogenesis of NPD, primarily through epigenetic mechanisms. Gut microbes and their metabolites influence immune cell activity and brain function, thereby contributing to neuroinflammation and the development and progression of NPD. The enteric nervous system, the autonomic nervous system, neuroendocrine signaling, and the immune system all participate in bidirectional communication between the gut and the brain. Importantly, the interaction of each of these systems with the GM influences epigenetic pathways. Here, we first explore the intricate relationship among intestinal microbes, microbial metabolites, and immune cell activity, with a focus on epigenetic mechanisms involved in NPD pathogenesis. Next, we provide background information on the association between inflammation and epigenetic aberrations in the context of NPD. Additionally, we review emerging therapeutic strategies—such as prebiotics, probiotics, methyl-rich diets, ketogenic diet, and medications—that may modulate the GM–immune system–brain axis via epigenetic regulation for the prevention or treatment of NPD. Finally, we discuss the challenges and future directions in investigating the critical role of this axis in mental health. Full article
Show Figures

Figure 1

23 pages, 2655 KiB  
Review
The Role of Nutrition in HIV-Associated Neurocognitive Disorders: Mechanisms, Risks, and Interventions
by Carlotta Siddi, Jihane Balla, Christy Agbey, Paola Fadda and Simona Dedoni
Life 2025, 15(6), 982; https://doi.org/10.3390/life15060982 - 19 Jun 2025
Viewed by 1705
Abstract
HIV-associated neurocognitive disorders (HANDs) refer to a range of cognitive deficits that afflict people living with the Human Immunodeficiency Virus (HIV). The fundamental processes of HAND include persistent inflammation, immunological activation, and direct viral impact on the central nervous system. Emerging research shows [...] Read more.
HIV-associated neurocognitive disorders (HANDs) refer to a range of cognitive deficits that afflict people living with the Human Immunodeficiency Virus (HIV). The fundamental processes of HAND include persistent inflammation, immunological activation, and direct viral impact on the central nervous system. Emerging research shows that nutritional status, especially food consumption and body weight, is critical in determining the course and severity of HAND. Malnutrition exacerbates neurocognitive impairment by increasing inflammation and oxidative stress, while obesity may contribute to HAND through the promotion of metabolic disruption, gut microbiota alterations, and systemic inflammation. Additionally, the introduction of antiretroviral treatment (ART) has substantially enhanced the prognosis of people living with HIV by lowering viral load and improving immune function. However, depending on the regimen, ART can cause changes in body weight, which may influence the progression of HAND. This emphasizes the intricate interplay between HIV, nutrition, body weight, and neurocognitive health. As a result, various dietary approaches are currently being investigated to improve the quality of life of individuals with HIV and possibly help prevent neurocognitive decline in this population. This review aims to elucidate the relationship between nutrition and neurocognitive function in individuals living with HIV, shedding light on aspects of HANDs related to diet, body weight fluctuations, and metabolic syndrome. It explores the shift from current pharmacological treatments to innovative non-pharmacological interventions, including specific dietary strategies, to support overall health and cognitive well being in HIV-positive people. Full article
Show Figures

Figure 1

15 pages, 2920 KiB  
Article
Grazing Intensities Regulated the Effects of Seasonal Dietary Pattern on Gut Bacterial Community Composition of Sheep
by Pengzhen Li, Zhenhao Zhang, Thomas A. Monaco, Yao Dong and Yuping Rong
Microorganisms 2025, 13(6), 1392; https://doi.org/10.3390/microorganisms13061392 - 14 Jun 2025
Viewed by 382
Abstract
Gut microbiota “enterotypes” are strongly associated with diet and host health. For grazing animals, plant species richness and nutrient content of vegetation may alter the food supply and diet composition of animals. Understanding this relationship is critical to clarify the adaption of gut [...] Read more.
Gut microbiota “enterotypes” are strongly associated with diet and host health. For grazing animals, plant species richness and nutrient content of vegetation may alter the food supply and diet composition of animals. Understanding this relationship is critical to clarify the adaption of gut microbiota to changes in vegetation quantity and quality in grassland ecosystems. Here, we studied the relationship between dietary and gut microbiota composition of sheep (lambs) over a growing season in a grassland ecosystem in northern China. Variation in vegetation composition among grazing intensities was greatest in September: and sheep preferred forbs and Rosaceae throughout the grazing period in all grazing treatments, yet their preference for Fabaceae was reduced in HG treatments in September. Grazing intensity and seasonal variations in food resource availability influenced dietary patterns, which in turn affected gut bacterial community composition. Enterotype 1, dominated by Christensenellaceae_R_7_group and Clostridia_UCG_014_unclassified, predominated during the warm season (July) for both LG and HG treatments. In contrast, Enterotype 2, dominated by Escherichia_Shigella, prevailed during the cool season (September) in HG. Diversity of Enterotype 1 exceeded (p < 0.001) that of Enterotype 2. For MG, Enterotype 1 and Enterotype 2 were evenly distributed over the grazing period. Our results highlight the importance of regulating grazing intensity to maintain the balance and health of gut microbiota according to temporal changes in plant nutrients and aboveground biomass of grassland ecosystems. Full article
(This article belongs to the Special Issue Advances in Diet–Host–Gut Microbiome Interactions)
Show Figures

Graphical abstract

19 pages, 835 KiB  
Review
Kidney-Gut Axis in Chronic Kidney Disease: Therapeutic Perspectives from Microbiota Modulation and Nutrition
by Shu Wakino, Kazuhiro Hasegawa, Masanori Tamaki, Masanori Minato and Taizo Inagaki
Nutrients 2025, 17(12), 1961; https://doi.org/10.3390/nu17121961 - 9 Jun 2025
Viewed by 1159
Abstract
Chronic kidney disease (CKD) has a high prevalence worldwide, with an increasing incidence. One of the mechanisms of CKD progression involves a disordered inter-organ relationship between the kidneys and the intestine, known as the kidney-gut axis. In CKD, two pathological gut conditions—disturbed gut [...] Read more.
Chronic kidney disease (CKD) has a high prevalence worldwide, with an increasing incidence. One of the mechanisms of CKD progression involves a disordered inter-organ relationship between the kidneys and the intestine, known as the kidney-gut axis. In CKD, two pathological gut conditions—disturbed gut microbiota composition called uremic dysbiosis and leaky gut—contribute to the progression of CKD. Dysbiosis is associated with the increased production of gut-derived uremic toxins, leaky gut, and chronic systemic inflammation, leading to worsening uremia, which in turn aggravates the gut condition. This vicious cycle should be a target of the therapeutic strategy against CKD. The modulation of uremic dysbiosis, including prebiotics, probiotics, and synbiotics, has been a typical treatment approach, although clinical evidence for their efficacy has been insufficient. Some non-antibiotic drugs have an impact on human gut bacteria that are believed to play a role in their clinical efficacy on kidney function. Nutrition therapies, including a low-protein diet, dietary fiber, a Mediterranean diet, and whole grains, positively influence gut microbiota composition and have been linked to a decreased risk of CKD. Novel strategies are currently being explored, involving the use of postbiotics, microbiome sequencing techniques, and fecal microbiota transplantation, although clinical application remains to be tested. Human trials investigating the above-mentioned interventions remain inconclusive due to several limitations, including dietary variability and genetic factors. Future research should focus on the development of more effective probiotics, prebiotics, and microbial metabolism-modifying drugs, not only for CKD but for other systemic diseases as well. Full article
Show Figures

Figure 1

14 pages, 2125 KiB  
Article
Fermented Apple Juice Reduces the Susceptibility of Offspring Mice to Food Allergy Exacerbated by Maternal High-Fat Diet
by Jing Ma, Jian Yu, Yining Jia, Zining Luo, Xin Yang, Huzhong Li and Fangyu Long
Nutrients 2025, 17(11), 1927; https://doi.org/10.3390/nu17111927 - 4 Jun 2025
Viewed by 666
Abstract
Background: Food allergy (FA) is associated with dietary habits, antibiotic use, living environment, and delivery method. Pregnancy and lactation represent critical periods for neonatal immune system development. Methods: This study investigated the relationship between maternal dietary habits and FA risk in offspring. [...] Read more.
Background: Food allergy (FA) is associated with dietary habits, antibiotic use, living environment, and delivery method. Pregnancy and lactation represent critical periods for neonatal immune system development. Methods: This study investigated the relationship between maternal dietary habits and FA risk in offspring. Pregnant C57BL/6J mice (8-week-old males and females) were fed either a high-fat diet (HFD) or HFD supplemented with fermented apple juice (FAJ) during pregnancy and lactation. Offspring were nursed by their respective dams until weaning at 21 days postpartum, followed by ovalbumin (OVA) sensitization. Lipid profiles, acylcarnitines, immunological, and histopathological analyses were performed. Gut microbiota composition and serum markers were also assessed. Results: The findings indicated that maternal HFD had a negative impact on OVA-sensitized offspring mice. Early-life FAJ intervention modulated gut microbiota alterations and alleviated maternal HFD-worsened allergic symptoms through Th1/Th2 and Th17/Treg immunity balance and intestinal barrier repair. Maternal serum triglyceride and total cholesterol levels, along with gut microbiota profiles, significantly influenced offspring gut microbiota composition. Moreover, reduced short-chain and medium-chain acylcarnitines in offspring may be associated with increased allergy risk. Conclusions: Maternal HFD during pregnancy and lactation disrupted gut microbiota balance and exacerbated offspring FA susceptibility. These findings provide a scientific foundation for developing early-life FA prevention strategies. Full article
Show Figures

Graphical abstract

28 pages, 1526 KiB  
Review
Microbiota-Accessible Borates as Novel and Emerging Prebiotics for Healthy Longevity: Current Research Trends and Perspectives
by Andrei Biţă, Ion Romulus Scorei, Marvin A. Soriano-Ursúa, George Dan Mogoşanu, Ionela Belu, Maria Viorica Ciocîlteu, Cristina Elena Biţă, Gabriela Rău, Cătălina Gabriela Pisoschi, Maria-Victoria Racu, Iurie Pinzaru, Alejandra Contreras-Ramos, Roxana Kostici, Johny Neamţu, Viorel Biciuşcă and Dan Ionuţ Gheonea
Pharmaceuticals 2025, 18(6), 766; https://doi.org/10.3390/ph18060766 - 22 May 2025
Viewed by 1678
Abstract
Precision nutrition-targeted gut microbiota (GM) may have therapeutic potential not only for age-related diseases but also for slowing the aging process and promoting longer healthspan. Recent studies have shown that restoring a healthy symbiosis of GM by counteracting dysbiosis (DYS) through precise nutritional [...] Read more.
Precision nutrition-targeted gut microbiota (GM) may have therapeutic potential not only for age-related diseases but also for slowing the aging process and promoting longer healthspan. Recent studies have shown that restoring a healthy symbiosis of GM by counteracting dysbiosis (DYS) through precise nutritional intervention is becoming a major target for extending healthspan. Microbiota-accessible borate (MAB) complexes, such as boron (B)–pectins (rhamnogalacturonan–borate) and borate–phenolic esters (diester chlorogenoborate), have a significant impact on healthy host–microbiota symbiosis (HMS). The mechanism of action of MABs involves the biosynthesis of the autoinducer-2–borate (AI-2B) signaling molecule, B fortification of the mucus gel layer by the MABs diet, inhibition of pathogenic microbes, and reversal of GM DYS, strengthening the gut barrier structure, enhancing immunity, and promoting overall host health. In fact, the lack of MAB complexes in the human diet causes reduced levels of AI-2B in GM, inhibiting the Firmicutes phylum (the main butyrate-producing bacteria), with important effects on healthy HMS. It can now be argued that there is a relationship between MAB-rich intake, healthy HMS, host metabolic health, and longevity. This could influence the deployment of natural prebiotic B-based nutraceuticals targeting the colon in the future. Our review is based on the discovery that MAB diet is absolutely necessary for healthy HMS in humans, by reversing DYS and restoring eubiosis for longer healthspan. Full article
Show Figures

Graphical abstract

28 pages, 2708 KiB  
Review
The Role of G Protein-Coupled Receptors in the Regulation of Orthopaedic Diseases by Gut Microbiota
by Peng Sun, Jinchao Liu, Guannan Chen and Yilan Guo
Nutrients 2025, 17(10), 1702; https://doi.org/10.3390/nu17101702 - 16 May 2025
Cited by 1 | Viewed by 842
Abstract
Exercise and diet modulate the gut microbiota, which is involved in the regulation of orthopaedic diseases and synthesises a wide range of metabolites that modulate cellular function and play an important role in bone development, remodelling and disease. G protein-coupled receptors (GPCRs), the [...] Read more.
Exercise and diet modulate the gut microbiota, which is involved in the regulation of orthopaedic diseases and synthesises a wide range of metabolites that modulate cellular function and play an important role in bone development, remodelling and disease. G protein-coupled receptors (GPCRs), the largest family of transmembrane receptors in the human body, interact with gut microbial metabolites to regulate relevant pathological processes. This paper provides a review of different dietary and exercise effects on the pathogenic gut microbiota and their metabolites associated with GPCRs in orthopaedic diseases. RESULTS: Generally, metabolites produced by gut microbiota contribute to the maintenance of bone health by activating the corresponding GPCRs, which are involved in bone metabolism, regulation of immune response, and maintenance of gut flora homeostasis. Exercise and diet can influence gut microbiota, and an imbalance in gut microbiota homeostasis can trigger a series of adverse immune and metabolic responses by affecting GPCR function, ultimately leading to the onset and progression of various orthopaedic diseases. Understanding these relationships is crucial for elucidating the pathogenesis of orthopaedic diseases and developing personalised probiotic-based therapeutic strategies. In the future, we should further explore how to prevent and treat orthopaedic diseases through GPCR-based modulation of gut microbes and their interactions. The development of substances that precisely modulate gut microbes through different exercises and diets will provide more effective interventions to improve bone health in patients. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

19 pages, 3221 KiB  
Review
Exercise, Diet, and Brain Health: From the Perspective of Gut Microbiota Regulation
by Li Zhang, Renhe Liu, Zheyi Song and Xin Zhang
Nutrients 2025, 17(10), 1686; https://doi.org/10.3390/nu17101686 - 15 May 2025
Viewed by 1577
Abstract
The existing body of evidence has highlighted gut microbiota as a versatile regulator of body wellness affecting not only multiple physiological metabolisms but also the function of remote organs. Emerging studies revealed a reciprocal relationship between physical exercise and intestinal microbiota, suggesting that [...] Read more.
The existing body of evidence has highlighted gut microbiota as a versatile regulator of body wellness affecting not only multiple physiological metabolisms but also the function of remote organs. Emerging studies revealed a reciprocal relationship between physical exercise and intestinal microbiota, suggesting that physical exercise could enhance gut health, including regulating intestinal barrier integrity, increasing microbial diversity, and promoting beneficial microbial metabolism. Furthermore, the beneficial outcomes of exercise on the intestine may also promote brain health through the gut–brain axis. Diet is an important factor in boosting exercise performance and also greatly impacts the structure of gut microbiota. Abundant research has reported that diet alongside exercise could exert beneficial effects on metabolism, immune regulation, and the neuropsychiatric system. In this paper, we used a narrative review, primarily searching PubMed, Web of Science, and Elsevier, to review the existing research on how moderate-intensity exercise promotes gut health, and we introduced the effects of exercise on the nervous system through the gut–brain axis. We also proposed dietary strategies targeting the regulation of gut microbiota to provide guidelines for boosting brain health. This review highlights that moderate exercise and a healthy diet promote gut and brain health. Full article
Show Figures

Graphical abstract

Back to TopTop