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

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
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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,777)

Search Parameters:
Keywords = gut-dysbiosis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
38 pages, 2249 KiB  
Review
Microbiome in Neuroblastoma: A Virgin Island in the World of Onco-Microbiome
by Ashwath Keshav Giri, Poorvi Subramanian, Loganayaki Periyasamy, Sivaroopan Aravindan and Natarajan Aravindan
Cells 2025, 14(15), 1218; https://doi.org/10.3390/cells14151218 (registering DOI) - 7 Aug 2025
Abstract
The composition of the gut and/or tumor microbiome has been intricately involved in the onset of carcinogenesis, tumor progression, therapy response, and patient outcomes in diverse solid cancers. The microbiome type, composition, and their metabolome have been functionally implicated in the multifarious cellular [...] Read more.
The composition of the gut and/or tumor microbiome has been intricately involved in the onset of carcinogenesis, tumor progression, therapy response, and patient outcomes in diverse solid cancers. The microbiome type, composition, and their metabolome have been functionally implicated in the multifarious cellular processes, transformation, proliferation, tumor immune evasion, cellular migration, etc. Despite such compelling evidence on the role of microbiome interactions in cancer, the realization of their role in neuroblastoma (NB), the deadly extracranial tumor in infants is few and fragmentary. This review comprehends the composition, diversity, and significance of microbiota in human health. Further, this review discusses the microbiota composition, their mode of action, and their signaling flow through and cellular processes in diverse cancers including NB. Precisely, this study for the first time has realized the functional relevance and clinical significance of the gut and tumor microbiome for NB. Interestingly, large cohort clinical and preclinical in vivo models of NB realized the following: gut microbiota predicts the risk for NB; postnatal (and or not maternal transmission) microbiome rearrangements; gut microbial effect on NB pathogenesis; tumor-altering gut microbial composition; microbial composition predicts treatment outcomes in NB; prebiotic remedies for stabilizing NB-associated microbial rearrangements; microbial composition in tumor-infiltrating microbiota predicts NB outcomes. Full article
(This article belongs to the Special Issue Signaling Pathways and Mechanisms in Cancer Therapy Resistance)
Show Figures

Figure 1

21 pages, 1426 KiB  
Review
Physical Activity and Metabolic Disorders—What Does Gut Microbiota Have to Do with It?
by Aneta Sokal-Dembowska, Ewelina Polak-Szczybyło, Kacper Helma, Patrycja Musz, Maciej Setlik, Weronika Fic, Dawid Wachowiak and Sara Jarmakiewicz-Czaja
Curr. Issues Mol. Biol. 2025, 47(8), 630; https://doi.org/10.3390/cimb47080630 (registering DOI) - 7 Aug 2025
Abstract
Obesity, type 2 diabetes mellitus (T2DM) and steatohepatitis associated with metabolic dysfunction (MASLD) are on the rise and pose serious health challenges worldwide. In recent years, researchers have gained a better understanding of the important role of the gut microbiota in the development [...] Read more.
Obesity, type 2 diabetes mellitus (T2DM) and steatohepatitis associated with metabolic dysfunction (MASLD) are on the rise and pose serious health challenges worldwide. In recent years, researchers have gained a better understanding of the important role of the gut microbiota in the development and progression of these diseases. Intestinal dysbiosis can contribute to the occurrence of increased intestinal permeability, inflammation and reduced numbers of commensal bacteria. In obesity, these changes contribute to chronic low-grade inflammation and deregulated metabolism. In MASLD, gut microbiota dysbiosis can promote liver fibrosis and impair bile acid metabolism, while in T2DM, they are associated with impaired glycemic control and insulin resistance. Regular physical activity has a positive effect on the composition of the gut microbiota, increasing its diversity, modulating its metabolic functions, strengthening the intestinal barrier and reducing inflammation. These findings suggest that exercise and microbiota-targeted interventions may play an important role in the prevention and treatment of metabolic diseases. Full article
(This article belongs to the Special Issue Metabolic Interactions Between the Gut Microbiome and Organism)
Show Figures

Figure 1

24 pages, 639 KiB  
Review
A Systemic Perspective of the Link Between Microbiota and Cardiac Health: A Literature Review
by Ionica Grigore, Oana Roxana Ciobotaru, Delia Hînganu, Gabriela Gurau, Dana Tutunaru and Marius Valeriu Hînganu
Life 2025, 15(8), 1251; https://doi.org/10.3390/life15081251 - 7 Aug 2025
Abstract
Cardiovascular diseases (CVDs) are the leading global cause of death, with long-term hospitalization becoming increasingly frequent in advanced or chronic cases. In this context, the interplay between systemic factors such as lipid metabolism, circulating metabolites, gut microbiota, and oral health is gaining attention [...] Read more.
Cardiovascular diseases (CVDs) are the leading global cause of death, with long-term hospitalization becoming increasingly frequent in advanced or chronic cases. In this context, the interplay between systemic factors such as lipid metabolism, circulating metabolites, gut microbiota, and oral health is gaining attention for its potential role in influencing inflammation, cardiometabolic risk, and long-term outcomes. Despite their apparent independence, these domains are increasingly recognized as interconnected and influential in cardiovascular pathophysiology. Methods: This narrative review was conducted by analyzing studies published between 2015 and 2024 from databases including PubMed, Scopus, and Web of Science. Keywords such as “lipid profile,” “metabolomics,” “gut microbiota,” “oral health,” and “cardiovascular disease” were used. Original research, meta-analyses, and reviews relevant to hospitalized cardiac patients were included. A critical integrative approach was applied to highlight cross-domain connections. Results and Discussion: Evidence reveals significant interrelations between altered lipid profiles, gut dysbiosis (including increased TMAO levels), metabolic imbalances, and oral inflammation. Each component contributes to a systemic pro-inflammatory state that worsens cardiovascular prognosis, particularly in long-term hospitalized patients. Despite isolated research in each domain, there is a paucity of studies integrating all four. The need for interdisciplinary diagnostic models and preventive strategies is emphasized, especially in populations with frailty or immobilization. Conclusions: Monitoring lipid metabolism, metabolomic shifts, gut microbial balance, and oral status should be considered part of comprehensive cardiovascular care. Gut microbiota exerts a dual role in cardiac health: when balanced, it supports anti-inflammatory and metabolic homeostasis; when dysbiotic, it contributes to systemic inflammation and worsened cardiac outcomes. Future research should aim to develop integrative screening tools and personalized interventions that address the multifactorial burden of disease. A systemic approach may improve both short- and long-term outcomes in this complex and vulnerable patient population. Full article
(This article belongs to the Special Issue The Emerging Role of Microbiota in Health and Diseases)
Show Figures

Figure 1

20 pages, 4612 KiB  
Article
Effect of a Gluten-Free Diet on the Intestinal Microbiota of Women with Celiac Disease
by M. Mar Morcillo Serrano, Paloma Reche-Sainz, Daniel González-Reguero, Marina Robas-Mora, Rocío de la Iglesia, Natalia Úbeda, Elena Alonso-Aperte, Javier Arranz-Herrero and Pedro A. Jiménez-Gómez
Antibiotics 2025, 14(8), 785; https://doi.org/10.3390/antibiotics14080785 - 2 Aug 2025
Viewed by 235
Abstract
Background/Objectives: Celiac disease (CD) is an autoimmune disorder characterized by small intestinal enteropathy triggered by gluten ingestion, often associated with gut dysbiosis. The most effective treatment is strict adherence to a gluten-free diet (GFD), which alleviates symptoms. This study uniquely integrates taxonomic, [...] Read more.
Background/Objectives: Celiac disease (CD) is an autoimmune disorder characterized by small intestinal enteropathy triggered by gluten ingestion, often associated with gut dysbiosis. The most effective treatment is strict adherence to a gluten-free diet (GFD), which alleviates symptoms. This study uniquely integrates taxonomic, functional, and resistance profiling to evaluate the gut microbiota of women with CD on a GFD. Methods: To evaluate the long-term impact of a GFD, this study analyzed the gut microbiota of 10 women with CD on a GFD for over a year compared to 10 healthy controls with unrestricted diets. Taxonomic diversity (16S rRNA gene sequencing and the analysis of α and β-diversity), metabolic functionality (Biolog EcoPlates®), and antibiotic resistance profiles (Cenoantibiogram) were assessed. Results: Metagenomic analysis revealed no significant differences in taxonomic diversity but highlighted variations in the abundance of specific bacterial genera. Women with CD showed increased proportions of Bacteroides, Streptococcus, and Clostridium, associated with inflammation, but also elevated levels of beneficial genera such as Roseburia, Oxalobacter, and Paraprevotella. Despite no significant differences in metabolic diversity, higher minimum inhibitory concentrations (MICs) in women in the healthy control group suggest that dietary substrates in unrestricted diets may promote the proliferation of fast-growing bacteria capable of rapidly developing and disseminating antibiotic resistance mechanisms. Conclusions: These findings indicate that prolonged adherence to a GFD in CD supports remission of gut dysbiosis, enhances microbiota functionality, and may reduce the risk of antibiotic resistance, emphasizing the importance of dietary management in CD. Full article
(This article belongs to the Special Issue Antibiotic Resistance: A One-Health Approach, 2nd Edition)
Show Figures

Graphical abstract

30 pages, 1428 KiB  
Review
The Oral–Gut Microbiota Axis Across the Lifespan: New Insights on a Forgotten Interaction
by Domenico Azzolino, Margherita Carnevale-Schianca, Luigi Santacroce, Marica Colella, Alessia Felicetti, Leonardo Terranova, Roberto Carlos Castrejón-Pérez, Franklin Garcia-Godoy, Tiziano Lucchi and Pier Carmine Passarelli
Nutrients 2025, 17(15), 2538; https://doi.org/10.3390/nu17152538 - 1 Aug 2025
Viewed by 243
Abstract
The oral–gut microbiota axis is a relatively new field of research. Although most studies have focused separately on the oral and gut microbiota, emerging evidence has highlighted that the two microbiota are interconnected and may influence each other through various mechanisms shaping systemic [...] Read more.
The oral–gut microbiota axis is a relatively new field of research. Although most studies have focused separately on the oral and gut microbiota, emerging evidence has highlighted that the two microbiota are interconnected and may influence each other through various mechanisms shaping systemic health. The aim of this review is therefore to provide an overview of the interactions between oral and gut microbiota, and the influence of diet and related metabolites on this axis. Pathogenic oral bacteria, such as Porphyromonas gingivalis and Fusobacterium nucleatum, can migrate to the gut through the enteral route, particularly in individuals with weakened gastrointestinal defenses or conditions like gastroesophageal reflux disease, contributing to disorders like inflammatory bowel disease and colorectal cancer. Bile acids, altered by gut microbes, also play a significant role in modulating these microbiota interactions and inflammatory responses. Oral bacteria can also spread via the bloodstream, promoting systemic inflammation and worsening some conditions like cardiovascular disease. Translocation of microorganisms can also take place from the gut to the oral cavity through fecal–oral transmission, especially within poor sanitary conditions. Some metabolites including short-chain fatty acids, trimethylamine N-oxide, indole and its derivatives, bile acids, and lipopolysaccharides produced by both oral and gut microbes seem to play central roles in mediating oral–gut interactions. The complex interplay between oral and gut microbiota underscores their crucial role in maintaining systemic health and highlights the potential consequences of dysbiosis at both the oral and gastrointestinal level. Some dietary patterns and nutritional compounds including probiotics and prebiotics seem to exert beneficial effects both on oral and gut microbiota eubiosis. A better understanding of these microbial interactions could therefore pave the way for the prevention and management of systemic conditions, improving overall health outcomes. Full article
(This article belongs to the Special Issue Exploring the Lifespan Dynamics of Oral–Gut Microbiota Interactions)
Show Figures

Figure 1

12 pages, 788 KiB  
Article
Gut Microbial Composition on Dienogest Therapy in Patients with Endometriosis
by Veronika Pronina, Pavel Denisov, Vera Muravieva, Alexey Skorobogatiy, Ksenia Zhigalova, Galina Chernukha, Gennady Sukhikh and Tatiana Priputnevich
Microbiol. Res. 2025, 16(8), 169; https://doi.org/10.3390/microbiolres16080169 - 1 Aug 2025
Viewed by 232
Abstract
Endometriosis is a chronic inflammatory condition affecting approximately 10% of women of reproductive age, characterized by pelvic pain, dysmenorrhea, and infertility. Emerging evidence suggests a potential link between gut microbiota dysbiosis and endometriosis pathogenesis, mediated through hormonal regulation, immune modulation, and systemic inflammation. [...] Read more.
Endometriosis is a chronic inflammatory condition affecting approximately 10% of women of reproductive age, characterized by pelvic pain, dysmenorrhea, and infertility. Emerging evidence suggests a potential link between gut microbiota dysbiosis and endometriosis pathogenesis, mediated through hormonal regulation, immune modulation, and systemic inflammation. Dienogest (DNG) is widely used for endometriosis management, but its effects on gut microbiota remain underexplored. This study investigates the impact of DNG on gut microbial composition in endometriosis patients, aiming to elucidate its therapeutic mechanisms beyond hormonal modulation. DNG therapy led to a significant reduction in the Bacillota/Bacteroidota ratio (p = 0.0421), driven by decreased Staphylococcus spp. (p = 0.0244) and increased commensal bacteria such as Lactobacillus spp. and Collinsella aerofaciens (p = 0.049). Species richness and alpha diversity indices showed a non-significant upward trend. Notably, C. aerofaciens, a butyrate producer linked to gut barrier integrity, was detected twice as frequently during therapy. The study also observed reductions in facultative anaerobes like Enterococcus spp. and a trend toward higher titers of beneficial Bacteroidota. This study provides the first evidence that DNG therapy modulates gut microbiota in endometriosis patients, favoring a composition associated with anti-inflammatory and barrier-protective effects. The observed shifts—reduced opportunistic pathogens and increased symbionts—suggest a novel mechanism for DNG’s efficacy, potentially involving the microbial regulation of estrogen metabolism and immune responses. Full article
Show Figures

Figure 1

23 pages, 766 KiB  
Review
Pathophysiological Links Between Inflammatory Bowel Disease and Cardiovascular Disease: The Role of Dysbiosis and Emerging Biomarkers
by Roko Šantić, Nikola Pavlović, Marko Kumrić, Marino Vilović and Joško Božić
Biomedicines 2025, 13(8), 1864; https://doi.org/10.3390/biomedicines13081864 - 31 Jul 2025
Viewed by 148
Abstract
This review introduces a novel integrative framework linking gut dysbiosis, systemic inflammation, and cardiovascular risk in patients with inflammatory bowel disease (IBD). We highlight emerging biomarkers, including short-chain fatty acids (SCFAs), calprotectin, and zonulin, that reflect alterations in the gut microbiome and increased [...] Read more.
This review introduces a novel integrative framework linking gut dysbiosis, systemic inflammation, and cardiovascular risk in patients with inflammatory bowel disease (IBD). We highlight emerging biomarkers, including short-chain fatty acids (SCFAs), calprotectin, and zonulin, that reflect alterations in the gut microbiome and increased intestinal permeability, which contribute to cardiovascular pathology. Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide, and recent evidence identifies IBD, encompassing ulcerative colitis (UC) and Crohn’s disease (CD), as a significant non-traditional risk factor for CVD. This review synthesizes current knowledge on how dysbiosis-driven inflammation in IBD patients exacerbates endothelial dysfunction, hypercoagulability, and atherosclerosis, even in the absence of traditional risk factors. Additionally, we discuss how commonly used IBD therapies may modulate cardiovascular risk. Understanding these multifactorial mechanisms and validating reliable biomarkers are essential for improving cardiovascular risk stratification and guiding targeted prevention strategies in this vulnerable population. Full article
Show Figures

Figure 1

10 pages, 478 KiB  
Review
Chewing Matters: Masticatory Function, Oral Microbiota, and Gut Health in the Nutritional Management of Aging
by Monia Lettieri, Alessio Rosa, Fabrizio Spataro, Giovanni Capria, Paolo Barnaba, Marco Gargari and Mirko Martelli
Nutrients 2025, 17(15), 2507; https://doi.org/10.3390/nu17152507 - 30 Jul 2025
Viewed by 350
Abstract
Aging is a multifactorial process that affects various physiological functions, including masticatory performance, which is crucial for oral health and nutritional well-being. Impaired masticatory function, often due to factors such as tooth loss, reduced salivation, or muscle atrophy, can lead to significant nutritional [...] Read more.
Aging is a multifactorial process that affects various physiological functions, including masticatory performance, which is crucial for oral health and nutritional well-being. Impaired masticatory function, often due to factors such as tooth loss, reduced salivation, or muscle atrophy, can lead to significant nutritional challenges and compromise the overall health of elderly individuals. Recent research has illuminated the interconnectedness of masticatory function, oral microbiota, and gut health, suggesting that altered chewing ability may disrupt oral microbial communities, which in turn affect gastrointestinal health and systemic inflammation. This commentary review provides a comprehensive analysis of the role of masticatory function in aging, exploring its impact on the oral microbiota, gut health, and broader nutritional status. We discuss the potential consequences of impaired mastication, including malnutrition, dysbiosis, and gastrointestinal disorders, and explore possible strategies for improving masticatory function and maintaining a healthy gut microbiome through interventions like dietary modifications, oral care, and rehabilitation. We aim to underscore the importance of integrating masticatory function management into the broader context of aging-related healthcare, promoting holistic, multidisciplinary approaches to support nutritional needs and quality of life in older adults. Full article
(This article belongs to the Special Issue Exploring the Lifespan Dynamics of Oral–Gut Microbiota Interactions)
Show Figures

Graphical abstract

22 pages, 1013 KiB  
Review
Genomic Alterations and Microbiota Crosstalk in Hepatic Cancers: The Gut–Liver Axis in Tumorigenesis and Therapy
by Yuanji Fu, Jenny Bonifacio-Mundaca, Christophe Desterke, Íñigo Casafont and Jorge Mata-Garrido
Genes 2025, 16(8), 920; https://doi.org/10.3390/genes16080920 - 30 Jul 2025
Viewed by 246
Abstract
Background/Objectives: Hepatic cancers, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are major global health concerns due to rising incidence and limited therapeutic success. While traditional risk factors include chronic liver disease and environmental exposures, recent evidence underscores the significance of genetic alterations and [...] Read more.
Background/Objectives: Hepatic cancers, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are major global health concerns due to rising incidence and limited therapeutic success. While traditional risk factors include chronic liver disease and environmental exposures, recent evidence underscores the significance of genetic alterations and gut microbiota in liver cancer development and progression. This review aims to integrate emerging knowledge on the interplay between host genomic changes and gut microbial dynamics in the pathogenesis and treatment of hepatic cancers. Methods: We conducted a comprehensive review of current literature on genetic and epigenetic drivers of HCC and CCA, focusing on commonly mutated genes such as TP53, CTNNB1, TERT, IDH1/2, and FGFR2. In parallel, we evaluated studies addressing the gut–liver axis, including the roles of dysbiosis, microbial metabolites, and immune modulation. Key clinical and preclinical findings were synthesized to explore how host–microbe interactions influence tumorigenesis and therapeutic response. Results: HCC and CCA exhibit distinct but overlapping genomic landscapes marked by recurrent mutations and epigenetic reprogramming. Alterations in the gut microbiota contribute to hepatic inflammation, genomic instability, and immune evasion, potentially enhancing oncogenic signaling pathways. Furthermore, microbiota composition appears to affect responses to immune checkpoint inhibitors. Emerging therapeutic strategies such as probiotics, fecal microbiota transplantation, and precision oncology based on mutational profiling demonstrate potential for personalized interventions. Conclusions: The integration of host genomics with microbial ecology provides a promising paradigm for advancing diagnostics and therapies in liver cancer. Targeting the gut–liver axis may complement genome-informed strategies to improve outcomes for patients with HCC and CCA. Full article
(This article belongs to the Special Issue Feature Papers in Microbial Genetics and Genomics)
Show Figures

Figure 1

16 pages, 636 KiB  
Review
The Gut–Endometriosis Axis: Genetic Mechanisms and Public Health Implications
by Efthalia Moustakli, Nektaria Zagorianakou, Stylianos Makrydimas, Emmanouil D. Oikonomou, Andreas Miltiadous and George Makrydimas
Genes 2025, 16(8), 918; https://doi.org/10.3390/genes16080918 - 30 Jul 2025
Viewed by 491
Abstract
Background/Objectives: Endometriosis is a chronic, estrogen-driven gynecological disorder affecting approximately 10% of reproductive-aged women worldwide, with significant physical, psychosocial, and socioeconomic impacts. Recent research suggests a possible involvement of the gut microbiome in endometriosis disease mechanisms through immune manipulation, estrogen metabolism, and [...] Read more.
Background/Objectives: Endometriosis is a chronic, estrogen-driven gynecological disorder affecting approximately 10% of reproductive-aged women worldwide, with significant physical, psychosocial, and socioeconomic impacts. Recent research suggests a possible involvement of the gut microbiome in endometriosis disease mechanisms through immune manipulation, estrogen metabolism, and inflammatory networks. This narrative review aims to summarize current evidence on gut microbiota changes in endometriosis patients, explore the mechanisms by which gut dysbiosis contributes to disease progression, and examine epidemiological links between gastrointestinal health and endometriosis risk. Methods: A narrative review was conducted to synthesize available literature on the compositional changes in gut microbiota associated with endometriosis. The review also evaluated studies investigating potential mechanisms and epidemiological patterns connecting gut health with endometriosis development and severity. Results: Alterations in gut microbiota composition were observed in endometriosis patients, suggesting roles in immune dysregulation, estrogen metabolism, and inflammation. Potential gut-oriented interventions, including dietary changes, probiotics, and lifestyle modifications, emerged as promising management options. However, methodological variability and research gaps remain barriers to clinical translation. Conclusions: Integrating gut microbiome research into endometriosis management holds potential for improving early diagnosis, patient outcomes, and healthcare system sustainability. The study emphasizes the need for further research to address existing challenges and to develop public health strategies that incorporate microbiome-based interventions in population-level endometriosis care. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

25 pages, 1199 KiB  
Review
Gut-Microbiota-Derived Metabolites and Probiotic Strategies in Colorectal Cancer: Implications for Disease Modulation and Precision Therapy
by Yi-Chu Yang, Shih-Chang Chang, Chih-Sheng Hung, Ming-Hung Shen, Ching-Long Lai and Chi-Jung Huang
Nutrients 2025, 17(15), 2501; https://doi.org/10.3390/nu17152501 - 30 Jul 2025
Viewed by 535
Abstract
The human gut microbiota significantly influences host health through its metabolic products and interaction with immune, neural, and metabolic systems. Among these, short-chain fatty acids (SCFAs), especially butyrate, play key roles in maintaining gut barrier integrity, modulating inflammation, and supporting metabolic regulation. Dysbiosis [...] Read more.
The human gut microbiota significantly influences host health through its metabolic products and interaction with immune, neural, and metabolic systems. Among these, short-chain fatty acids (SCFAs), especially butyrate, play key roles in maintaining gut barrier integrity, modulating inflammation, and supporting metabolic regulation. Dysbiosis is increasingly linked to diverse conditions such as gastrointestinal, metabolic, and neuropsychiatric disorders, cardiovascular diseases, and colorectal cancer (CRC). Probiotics offer therapeutic potential by restoring microbial balance, enhancing epithelial defenses, and modulating immune responses. This review highlights the physiological functions of gut microbiota and SCFAs, with a particular focus on butyrate’s anti-inflammatory and anti-cancer effects in CRC. It also examines emerging microbial therapies like probiotics, synbiotics, postbiotics, and engineered microbes. Emphasis is placed on the need for precision microbiome medicine, tailored to individual host–microbiome interactions and metabolomic profiles. These insights underscore the promising role of gut microbiota modulation in advancing preventive and personalized healthcare. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota, and Gastrointestinal Disease)
Show Figures

Graphical abstract

26 pages, 3684 KiB  
Article
Creation of Zinc (II)-Complexed Green Tea and Its Effects on Gut Microbiota by Daily Green Tea Consumption
by Tsukasa Orita, Daichi Ijiri, De-Xing Hou and Kozue Sakao
Molecules 2025, 30(15), 3191; https://doi.org/10.3390/molecules30153191 - 30 Jul 2025
Viewed by 388
Abstract
Although Zn (II)-(−)-Epigallocatechin gallate (EGCg) complex (Zn-EGCg) is known for its promising bioactivities, little attention has been paid to its incorporation into daily green tea consumption. In this study, we aimed to incorporate Zn (II) into green tea extract to promote the formation [...] Read more.
Although Zn (II)-(−)-Epigallocatechin gallate (EGCg) complex (Zn-EGCg) is known for its promising bioactivities, little attention has been paid to its incorporation into daily green tea consumption. In this study, we aimed to incorporate Zn (II) into green tea extract to promote the formation of Zn-EGCg complex within the tea matrix. We then investigated how the formation of Zn-complexed green tea extract (Zn-GTE) influences the gut microbiota in a Western diet (WD)-fed mouse model. Structural analyses using ultraviolet–visible spectroscopy (UV–Vis), Fourier-transform infrared spectroscopy (FT-IR), proton nuclear magnetic resonance (1H NMR), and powder X-ray diffraction (PXRD) suggested that Zn (II) interacted with hydroxyl groups of polyphenols within the extract, consistent with Zn-EGCg formation, although the complex could not be unequivocally identified. Under intake levels equivalent to daily consumption, Zn-GTE administration restored WD-induced reductions in alpha-diversity and resulted in a distinct microbial composition compared to treatment with green tea extract (GTE) or Zn alone, as shown by beta-diversity analysis. Linear discriminant analysis Effect Size (LEfSe) analysis revealed increased abundances of bacterial taxa belonging to o_Clostridiales, o_Bacteroidales, and f_Rikenellaceae, and decreased abundances of g_Akkermansia in the Zn-GTE group compared to the GTE group. These findings highlight that Zn-GTE, prepared via Zn (II) supplementation to green tea, may exert distinct microbiota-modulating effects compared to its individual components. This study provides new insights into the role of dietary metal–polyphenol complexes, offering a food-based platform for studying metal–polyphenol interactions under physiologically relevant conditions. Full article
(This article belongs to the Special Issue Health Benefits and Applications of Bioactive Phenolic Compounds)
Show Figures

Graphical abstract

28 pages, 1184 KiB  
Review
Immune Modulation by Microbiota and Its Possible Impact on Polyomavirus Infection
by Giorgia Cianci, Gloria Maini, Matteo Ferraresi, Giulia Pezzi, Daria Bortolotti, Sabrina Rizzo, Silvia Beltrami and Giovanna Schiuma
Pathogens 2025, 14(8), 747; https://doi.org/10.3390/pathogens14080747 - 30 Jul 2025
Viewed by 396
Abstract
Polyomaviruses are a family of small DNA viruses capable of establishing persistent infections, and they can pose significant pathogenic risks in immunocompromised hosts. While traditionally studied in the context of viral reactivation and immune suppression, recent evidence has highlighted the gut microbiota as [...] Read more.
Polyomaviruses are a family of small DNA viruses capable of establishing persistent infections, and they can pose significant pathogenic risks in immunocompromised hosts. While traditionally studied in the context of viral reactivation and immune suppression, recent evidence has highlighted the gut microbiota as a critical regulator of host immunity and viral pathogenesis. This review examines the complex interactions between polyomaviruses, the immune system, and intestinal microbiota, emphasizing the role of short-chain fatty acids (SCFAs) in modulating antiviral responses. We explore how dysbiosis may facilitate viral replication, reactivation, and immune escape and also consider how polyomavirus infection can, in turn, alter microbial composition. Particular attention is given to the Firmicutes/Bacteroidetes ratio as a potential biomarker of infection risk and immune status. Therapeutic strategies targeting the microbiota, including prebiotics, probiotics, and fecal microbiota transplantation (FMT), are discussed as innovative adjuncts to immune-based therapies. Understanding these tri-directional interactions may offer new avenues for mitigating disease severity and improving patient outcomes during viral reactivation. Full article
Show Figures

Figure 1

14 pages, 618 KiB  
Review
Management of Neonates in the Special Care Nursery and Its Impact on the Developing Gut Microbiota: A Comprehensive Clinical Review
by Ravisha Srinivasjois, Shripada Rao and Gavin Pereira
Microorganisms 2025, 13(8), 1772; https://doi.org/10.3390/microorganisms13081772 - 29 Jul 2025
Viewed by 420
Abstract
The first few days following the birth are a vulnerable time for the neonate. Sick infants experience various interventions during their stay in the neonatal unit in order to stay alive and grow. Acquisition of gut microbes is critical for the short- and [...] Read more.
The first few days following the birth are a vulnerable time for the neonate. Sick infants experience various interventions during their stay in the neonatal unit in order to stay alive and grow. Acquisition of gut microbes is critical for the short- and long-term health of the neonate. At a time when the gut microbiome is starting to take shape, crucial interventions directed at improving the growth, development and survival of the neonate impact its development. Events prior to and after the birth of the neonate, such as maternal conditions, antibiotic exposure, type of feeds, supplemental probiotics, and neonatal intensive care environment, contribute significantly to shaping the gut microbiome over the first few weeks and maintain its healthy balance crucial for long-term health. In this comprehensive review, we address common interventions the neonate is exposed to in its journey and their impact on gut microbiome, and discuss various interventions that minimize the dysbiosis of the gut. Full article
(This article belongs to the Collection Feature Papers in Gut Microbiota Research)
Show Figures

Figure 1

17 pages, 1908 KiB  
Article
BDE-47 Disrupts Gut Microbiota and Exacerbates Prediabetic Conditions in Mice: Therapeutic Potential of Grape Exosomes and Antioxidants
by Zaoling Liu, Fang Cao, Aerna Qiayimaerdan, Nilupaer Aisikaer, Zulipiya Zunong, Xiaodie Ma and Yale Yu
Toxics 2025, 13(8), 640; https://doi.org/10.3390/toxics13080640 - 29 Jul 2025
Viewed by 222
Abstract
Background: BDE-47, a pervasive environmental pollutant detected in >90% of human serum samples, is increasingly linked to metabolic disorders. This study investigates the specific impact of BDE-47 exposure on the gut microbiota in prediabetic mice and evaluates the efficacy of therapeutic interventions [...] Read more.
Background: BDE-47, a pervasive environmental pollutant detected in >90% of human serum samples, is increasingly linked to metabolic disorders. This study investigates the specific impact of BDE-47 exposure on the gut microbiota in prediabetic mice and evaluates the efficacy of therapeutic interventions in mitigating these effects. Objectives: To determine whether BDE-47 exposure induces diabetogenic dysbiosis in prediabetic mice and to assess whether dietary interventions, such as grape exosomes and an antioxidant cocktail, can restore a healthy microbiota composition and mitigate diabetes risk. Methods: In this study, a prediabetic mouse model was established in 54 male SPF-grade C57BL/6J mice through a combination of high-sugar and high-fat diet feeding with streptozotocin injection. Oral glucose tolerance tests (OGTT) were conducted on day 7 and day 21 post-modeling to assess the establishment of the model. The criteria for successful model induction were defined as fasting blood glucose levels below 7.8 mmol/L and 2 h postprandial glucose levels between 7.8 and 11.1 mmol/L. Following confirmation of model success, a 3 × 3 factorial design was applied to allocate the experimental animals into groups based on two independent factors: BDE-47 exposure and exosome intervention. The BDE-47 exposure factor consisted of three dose levels—none, high-dose, and medium-dose—while the exosome intervention factor included three modalities—none, Antioxidant Nutrients Intervention, and Grape Exosomes Intervention. Fresh fecal samples were collected from mice two days prior to sacrifice. Cecal contents and segments of the small intestine were collected and transferred into 1.5 mL cryotubes. All sequences were clustered into operational taxonomic units (OTUs) based on defined similarity thresholds. To compare means across multiple groups, a two-way analysis of variance (ANOVA) was employed. The significance level was predefined at α = 0.05, and p-values < 0.05 were considered statistically significant. Bar charts and line graphs were generated using GraphPad Prism version 9.0 software, while statistical analyses were performed using SPSS version 20.0 software. Results: The results of 16S rDNA sequencing analysis of the microbiome showed that there was no difference in the α diversity of the intestinal microbiota in each group of mice (p > 0.05), but there was a difference in the Beta diversity (p < 0.05). At the gate level, the abundances of Proteobacteria, Campylobacterota, Desulfobacterota, and Fusobacteriota in the medium-dose BDE-7 group were higher than those in the model control group (p < 0.05). The abundance of Patellar bacteria was lower than that of the model control group (p < 0.05). The abundances of Proteobacteria and Campylobacterota in the high-dose BDE-7 group were higher than those in the model control group (p < 0.05). The abundance of Planctomycetota and Patescibacteria was lower than that of the model control group (p < 0.05), while the abundance of Campylobacterota in the grape exosome group was higher than that of the model control group (p < 0.05). The abundance of Patescibacteria was lower than that of the model control group (p < 0.05), while the abundance of Firmicutes and Fusobacteriota in the antioxidant nutrient group was higher than that of the model control group (p < 0.05). However, the abundance of Verrucomicrobiota and Patescibacteria was lower than that of the model control group (p < 0.05). At the genus level, the abundances of Bacteroides and unclassified Lachnospiraceae in the high-dose BDE-7 group were higher than those in the model control group (p < 0.05). The abundance of Lachnospiraceae NK4A136_group and Lactobacillus was lower than that of the model control group (p < 0.05). The abundance of Veillonella and Helicobacter in the medium-dose BDE-7 group was higher than that in the model control group (p < 0.05), while the abundance of Lactobacillus was lower (p < 0.05). The abundance of genera such as Lentilactobacillus and Faecalibacterium in the grape exosome group was higher than that in the model control group (p < 0.05). The abundance of Alloprevotella and Bacteroides was lower than that of the model control group (p < 0.05). In the antioxidant nutrient group, the abundance of Lachnospiraceae and Hydrogenophaga was higher than that in the model control group (p < 0.05). However, the abundance of Akkermansia and Coriobacteriaceae UCG-002 was significantly lower than that of the model control group (p < 0.05). Conclusions: BDE-47 induces diabetogenic dysbiosis in prediabetic mice, which is reversible by dietary interventions. These findings suggest that microbiota-targeted strategies may effectively mitigate the diabetes risk associated with environmental pollutant exposure. Future studies should further explore the mechanisms underlying these microbiota changes and the long-term health benefits of such interventions. Full article
Show Figures

Figure 1

Back to TopTop