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

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 (1,677)

Search Parameters:
Keywords = gut health and microbiome

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 2242 KB  
Review
Bioactive Peptides from Quinoa (Chenopodium quinoa Willd.) as Modulators of the Gut Microbiome: A Scoping Review of Preclinical Evidence
by Nicolás Caicedo, Yamil Liscano and Jose Oñate-Garzón
Nutrients 2025, 17(20), 3215; https://doi.org/10.3390/nu17203215 - 14 Oct 2025
Abstract
Background: Gut microbiome dysbiosis is implicated in numerous chronic diseases. While quinoa possesses a rich nutritional profile with prebiotic potential, the specific capacity of its bioactive peptides to modulate gut microbial communities is not well understood. This scoping review systematically maps the preclinical [...] Read more.
Background: Gut microbiome dysbiosis is implicated in numerous chronic diseases. While quinoa possesses a rich nutritional profile with prebiotic potential, the specific capacity of its bioactive peptides to modulate gut microbial communities is not well understood. This scoping review systematically maps the preclinical evidence on the gut microbiome modulatory effects of quinoa-derived bioactive peptides to identify mechanisms, characterize their therapeutic potential, and guide future clinical translation. Methods: Following PRISMA-ScR guidelines, we searched six databases for preclinical studies investigating quinoa-derived peptides or hydrolysates and their effects on gut microbiota. Results: From 834 records, 19 studies met the inclusion criteria. Quinoa interventions demonstrated consistent effects, with 83% of studies reporting enhancement of beneficial genera and 67% an increase in alpha diversity. Disease-specific microbial signatures were observed; for instance, obesity models showed a reduced Firmicutes/Bacteroidetes ratio, while colitis models exhibited decreased Proteobacteria. Butyrate production was consistently enhanced. Methodologically, peptide generation has evolved from traditional fermentation toward more efficient enzymatic hydrolysis. Conclusions: Preclinical evidence strongly suggests that quinoa-derived bioactive peptides act as robust, context-dependent modulators of the gut microbiome. These findings position quinoa as a promising functional ingredient for precision gut health interventions, though clinical translation requires standardized preparations and validation in human trials. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

17 pages, 1103 KB  
Article
In Vitro Probiotic Modulation of Specific Dietary Complex Sugar Consumption in Fecal Cultures in Infants
by Daniela Mollova, Vesselin Baev and Ilia Iliev
Microorganisms 2025, 13(10), 2352; https://doi.org/10.3390/microorganisms13102352 - 14 Oct 2025
Abstract
Establishing the relative stability of the gastrointestinal microbiome after birth is a long and complex process, and it occurs under various influences. The human gut microbiome plays a crucial role in influencing an individual’s health and well-being across all stages of life. Breastfeeding, [...] Read more.
Establishing the relative stability of the gastrointestinal microbiome after birth is a long and complex process, and it occurs under various influences. The human gut microbiome plays a crucial role in influencing an individual’s health and well-being across all stages of life. Breastfeeding, the introduction of solid food at a certain stage after birth, and the type of food largely determine the composition of the developing microbiome. The influence of probiotics on the early development of the microbiome is gaining increasing interest. The method of in vitro co-cultivation with probiotic strains provides a clearer picture of the influence of these microorganisms on the community and the functional changes that the infant’s microbiome undergoes. We used fecal samples to study this influence by conducting metagenomic sequencing to determine the composition of the microbiome and a series of cultivations to determine the absorption of various fibers and prebiotic sugars from breast milk. We found statistically significant differences in the absorption of prebiotic sugars isolated from breast milk, as well as better absorption of several substrates in the presence of a probiotic strain. Full article
(This article belongs to the Special Issue Role of Dietary Nutrients in the Modulation of Gut Microbiota)
Show Figures

Figure 1

23 pages, 672 KB  
Review
The Association Between Maternal Diet and the Human Milk Microbiome: A Review of Evidence and Methodological Challenges
by Azhar S. Sindi
Microorganisms 2025, 13(10), 2347; https://doi.org/10.3390/microorganisms13102347 - 14 Oct 2025
Abstract
The human milk (HM) microbiome plays an important role in shaping the infant gut microbiota, with potential implications for immune development and both short- and long-term health. Among the maternal and infant factors influencing HM microbial composition, maternal diet represents a modifiable determinant. [...] Read more.
The human milk (HM) microbiome plays an important role in shaping the infant gut microbiota, with potential implications for immune development and both short- and long-term health. Among the maternal and infant factors influencing HM microbial composition, maternal diet represents a modifiable determinant. However, evidence regarding the impact of diet on the HM microbiota remains limited, and the methodological quality of available studies is variable. This review synthesises findings from 15 observational and interventional studies, critically evaluating dietary assessment approaches, milk collection protocols, microbiome analysis methods, and control of confounding factors. Current evidence suggests that maternal intake of macronutrients, micronutrients, and bioactive compounds may influence HM bacterial composition and functional potential, though results are inconsistent. Key limitations across studies include small sample sizes, short intervention periods, lack of appropriate control groups, variable aseptic sampling methods, inadequate contamination controls, and insufficient adjustment for confounders. To advance the field, we recommend larger, multicentre randomised controlled trials with longer intervention durations, incorporation of dietary biomarkers, standardised HM collection and processing protocols, and advanced multi-omics approaches. Strengthening methodological rigour is essential to generate robust evidence that can guide dietary interventions aimed at optimising the HM microbiota and improving infant health outcomes. Full article
(This article belongs to the Special Issue Microbiome Research: Past, Present, and Future)
Show Figures

Figure 1

13 pages, 1486 KB  
Article
Nutritional and Physiological Demands Shape the Gut Microbiome of Female World Tour Cyclists
by Toon Ampe, Lieselot Decroix, Kevin De Pauw, Romain Meeusen, Thomas Demuyser and Bart Roelands
Microorganisms 2025, 13(10), 2345; https://doi.org/10.3390/microorganisms13102345 - 13 Oct 2025
Abstract
This cross-sectional study investigated whether elite female World Tour cyclists have a specific gut microbiome compared to non-athlete female controls, potentially resulting from the unique physiological and dietary demands of high-level endurance cycling. Fourteen female cyclists and thirteen matched controls provided fecal samples [...] Read more.
This cross-sectional study investigated whether elite female World Tour cyclists have a specific gut microbiome compared to non-athlete female controls, potentially resulting from the unique physiological and dietary demands of high-level endurance cycling. Fourteen female cyclists and thirteen matched controls provided fecal samples during a period of reduced training (off-season cycling). The samples were analyzed using 16S rRNA gene sequencing and short-chain fatty acid (SCFA) quantification. The results revealed significant differences in microbiome composition. The cyclists showed a higher abundance of Bacteroidota (72.7% vs. 15.3%) and a lower abundance of Firmicutes (22.1% vs. 62.5%) compared to the controls, along with reduced alpha-diversity (Shannon index, p < 0.05). Fiber-fermenting families such as Lachnospiraceae and Ruminococcaceae were depleted, consistent with a carbohydrate-focused and relatively low-fiber diet. Interestingly, fecal SCFA levels did not differ, suggesting functional adaptation of the microbiome. These findings indicate that the elite female cyclists may have developed a “performance-adapted” gut microbiome. However, due to the cross-sectional design, causality cannot be established, and the long-term health implications remain uncertain. Full article
Show Figures

Figure 1

22 pages, 2098 KB  
Review
Mammary Gland Microbiota in Benign Breast Diseases
by Nikita I. Ukraincev, Maria I. Kashutina, Larisa I. Kasatkina, Adkhamzhon B. Abduraimov and Yury V. Zhernov
Int. J. Mol. Sci. 2025, 26(20), 9951; https://doi.org/10.3390/ijms26209951 (registering DOI) - 13 Oct 2025
Abstract
The human microbiome is a critical factor in health and disease. While its association with breast cancer (BC) has been increasingly studied, this review provides a dedicated synthesis of the microbiota’s role in benign breast diseases (BBDs)—a common yet microbiologically overlooked spectrum of [...] Read more.
The human microbiome is a critical factor in health and disease. While its association with breast cancer (BC) has been increasingly studied, this review provides a dedicated synthesis of the microbiota’s role in benign breast diseases (BBDs)—a common yet microbiologically overlooked spectrum of conditions. The primary aim of this work is to consolidate the current understanding of the composition, origins, and functional mechanisms of the mammary gland (MG) microbiota specifically in the context of BBD and to evaluate its potential for novel diagnostic and therapeutic targets. We detail the distinct MG microbiota, formed via exogenous (e.g., cutaneous, translocation) and endogenous (e.g., enteromammary, lymphohematogenous) pathways, and its interaction with the host through estrogen metabolism, immunomodulation, and epigenetic modifications. This narrative review reveals unique dysbiotic patterns in BBD, characterized by distinct microbial signatures, such as the enrichment of Corynebacterium kroppenstedtii in granulomatous mastitis and the presence of Staphylococcus aureus in fibroadenomas and lactational mastitis. Furthermore, specific gut microbial profiles are identified in BBD patients, including an increased abundance of genera such as Clostridium and Faecalibacterium, alongside a decrease in Collinsella and Alistipes compared to healthy controls. These specific taxa represent compelling candidates for diagnostic biomarkers. We conclude that microbial dysbiosis is a significant component of BBD pathogenesis. A paradigm shift toward multi-omics approaches and mechanistic studies is now essential to translate these associations into clinical applications. Understanding the BBD-specific microbiome holds the promise of revolutionizing patient care through microbiota-based diagnostics for differentiating benign subtypes and novel, personalized therapeutic strategies aimed at restoring microbial homeostasis. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

9 pages, 411 KB  
Protocol
Changes in Gut Microbiome According to Probiotic Intake in Rectal Cancer Patients Undergoing Diverting Stoma Repair: Study Protocol
by Hyeung-min Park, Jaram Lee, Soo Young Lee, Chang Hyun Kim and Hyeong Rok Kim
J. Clin. Med. 2025, 14(20), 7190; https://doi.org/10.3390/jcm14207190 (registering DOI) - 12 Oct 2025
Viewed by 56
Abstract
Background: The gut microbiome is crucial in sustaining intestinal balance and general health. Following rectal cancer surgery, the creation of a diverting stoma to protect the anastomosis results in a defunctioned colon, leading to dysbiosis. The effect of probiotic intake on gut [...] Read more.
Background: The gut microbiome is crucial in sustaining intestinal balance and general health. Following rectal cancer surgery, the creation of a diverting stoma to protect the anastomosis results in a defunctioned colon, leading to dysbiosis. The effect of probiotic intake on gut dysbiosis following ileostomy repair remains uncertain. Thus, this study aims to determine the changes in gut microbiota based on the intake of probiotics after diverting stoma repair. Methods: This single-center, parallel, prospective pilot study will include patients with primary rectal cancer planning to undergo a diverting stoma during rectal cancer surgery. The study will comprise 20 patients, with 10 patients receiving synbiotics after stoma repair and 10 patients not receiving probiotics. The primary endpoint is the change in the gut microbiota of the resting colon based on the intake of probiotics, assessed through fecal testing at the following time points: before bowel resection, immediately after diverting stoma repair, and 3 weeks after diverting stoma repair. Changes in gut microbiota will be evaluated using alpha- and beta-diversity analyses based on 16S rRNA sequencing of fecal samples. Discussion: This study is the first prospective cohort trial investigating changes in the gut microbiota of the resting colon based on oral probiotic administration in patients undergoing diverting stoma repair. This trial is anticipated to clarify the impact of probiotic intake in these patients. Trial registration: Clinical Research Information Service (CRIS) of the Republic of Korea, KCT0008392, Registered on 27 April 2023. Full article
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)
Show Figures

Figure 1

25 pages, 1363 KB  
Review
Guardians in the Gut: Mechanistic Insights into a Hidden Ally Against Triple-Negative Breast Cancer
by Kayla Jaye, Muhammad A. Alsherbiny, Dennis Chang, Chun-Guang Li and Deep Jyoti Bhuyan
Cancers 2025, 17(19), 3248; https://doi.org/10.3390/cancers17193248 - 7 Oct 2025
Viewed by 332
Abstract
The gut microbiome possesses a diverse range of biological properties that play a role in maintaining host health and preventing disease. Gut microbial metabolites, including short-chain fatty acids, natural purine nucleosides, ellagic acid derivatives, and tryptophan metabolites, have been observed to have complex [...] Read more.
The gut microbiome possesses a diverse range of biological properties that play a role in maintaining host health and preventing disease. Gut microbial metabolites, including short-chain fatty acids, natural purine nucleosides, ellagic acid derivatives, and tryptophan metabolites, have been observed to have complex and multifaceted roles in the gut and in wider body systems in the management of disease, including cancer. Triple-negative breast cancer is the most aggressive subtype of breast cancer, with restricted treatment options and poor prognoses. Recently, preclinical research has investigated the antiproliferative potential of gut microbial metabolites against this type of breast cancer with promising results. However, little is understood about the mechanisms of action and molecular pathways driving this antiproliferative potential. Understanding the complex mechanisms of action of gut microbial metabolites on triple-negative breast cancer will be instrumental in the investigation of the combined administration with standard chemotherapeutic drugs. To date, there is a paucity of research studies investigating the potential synergistic interactions between gut microbial metabolites and standard chemotherapeutic drugs. The identification of synergistic potential between these compounds may provide alternate and more effective therapeutic options in the treatment and management of triple-negative breast cancer. Further investigation into the mechanistic action of gut microbial metabolites against this breast cancer subtype may support the administration of more cost-effective treatment options for breast cancer, with an aim to reduce side effects associated with standard treatments. Additionally, future research will aim to identify more potent metabolite–drug combinations in the mitigation of triple-negative breast cancer progression and metastasis. Full article
(This article belongs to the Special Issue Gut Microbiome, Diet and Cancer Risk)
Show Figures

Graphical abstract

32 pages, 9212 KB  
Article
Liver-Targeted Scutellarin Nanoemulsion Alleviates Fibrosis with Ancillary Modulation of the Gut–Liver Microbiota
by Haoyang Yu, Xia Niu, Bingyu Niu, Peng Lei, Ning Xu, Sitong Yang, Quanyong Yu, Guiling Li and Lulu Wang
Int. J. Mol. Sci. 2025, 26(19), 9746; https://doi.org/10.3390/ijms26199746 - 7 Oct 2025
Viewed by 181
Abstract
Liver fibrosis, a progressive condition with limited pharmacotherapies, poses a global health challenge. Scutellarin (SCU), a flavonoid derived from Erigeron breviscapus, has demonstrated anti-fibrotic activity and modulates gut microbiota. Emerging evidence suggests that SCU may also influence the hepatic microbiome. However, its [...] Read more.
Liver fibrosis, a progressive condition with limited pharmacotherapies, poses a global health challenge. Scutellarin (SCU), a flavonoid derived from Erigeron breviscapus, has demonstrated anti-fibrotic activity and modulates gut microbiota. Emerging evidence suggests that SCU may also influence the hepatic microbiome. However, its clinical utility is constrained by poor water solubility and low oral bioavailability. Here, we developed an SCU-loaded nanoemulsion (SCE) to enhance solubility and liver-targeted delivery. In vitro, SCE increased SCU uptake in hepatic stellate cells (HSCs) and significantly inhibited TGF-β1-induced fibrogenesis. In a bile duct ligation (BDL) mouse model, oral administration of SCE improved hepatic SCU accumulation and produced superior anti-fibrotic efficacy. SCE treatment attenuated fibrosis and collagen deposition in the liver and improved liver function markers. Mechanistic investigations using 16S rRNA sequencing revealed that SCU treatment was associated with beneficial microbiota changes, although its main therapeutic effects were achieved through enhanced hepatic targeting. Notably, the SCE formulation was well-tolerated, showing no significant toxicity in vitro or in vivo. In conclusion, the SCU-loaded nanoemulsion achieved enhanced hepatic delivery of SCU and exerted potent anti-fibrotic effects via multiple mechanisms, including direct suppression of fibrogenesis and ancillary modulation of the gut–liver microbiome, offering a promising therapeutic strategy for liver fibrosis. Full article
(This article belongs to the Special Issue Natural Products in Drug Discovery and Development)
Show Figures

Figure 1

19 pages, 515 KB  
Review
The Role of Environmental Exposures in Pediatric Asthma Pathogenesis: A Contemporary Narrative Review
by Luca Pecoraro, Anna Gloria Lanzilotti, Marta De Musso, Elisabetta Di Muri, Fernanda Tramacere, Emiliano Altavilla and Flavia Indrio
Children 2025, 12(10), 1327; https://doi.org/10.3390/children12101327 - 2 Oct 2025
Viewed by 417
Abstract
Over several decades, childhood asthma has emerged as a significant global public health concern, with the highest prevalence reported in industrialized countries. The rapid rise in asthma incidence and loss of control when the diagnosis is established can be related to environmental and [...] Read more.
Over several decades, childhood asthma has emerged as a significant global public health concern, with the highest prevalence reported in industrialized countries. The rapid rise in asthma incidence and loss of control when the diagnosis is established can be related to environmental and lifestyle changes, especially during early infancy. Current evidence indicates a potential link to an imbalance in immune system responses, influenced by tobacco smoke, traffic-related air pollution, outdoor and indoor allergens, gut microbiome, viral infection, obesity, sedentary lifestyle and dietary patterns. This narrative review aims to explore the landscape of contemporary environmental risk factors for childhood asthma, with a focus on their interplay and the relative importance. Full article
(This article belongs to the Special Issue Pulmonary Function in Children with Respiratory Symptoms)
Show Figures

Figure 1

59 pages, 4148 KB  
Review
Diet and Lifestyle Interventions in Metabolic Dysfunction-Associated Fatty Liver Disease: A Comprehensive Review
by Muhammad Y. Sheikh, Muhammad F. Younus, Annie Shergill and Muhammad N. Hasan
Int. J. Mol. Sci. 2025, 26(19), 9625; https://doi.org/10.3390/ijms26199625 - 2 Oct 2025
Viewed by 304
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH), have become the leading causes of chronic liver disease worldwide, with increasing rates of cirrhosis, hepatocellular carcinoma, and cardiovascular complications. Pathogenesis involves a complex interplay of dietary excess, sedentary [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH), have become the leading causes of chronic liver disease worldwide, with increasing rates of cirrhosis, hepatocellular carcinoma, and cardiovascular complications. Pathogenesis involves a complex interplay of dietary excess, sedentary lifestyle, insulin resistance, adipose tissue dysfunction, and alterations in the gut microbiome, which collectively lead to hepatocellular stress, inflammation, and fibrogenesis. Despite ongoing advances in pharmacotherapy, lifestyle intervention remains the cornerstone of management. Evidence shows that sustained weight loss of ≥5% reduces hepatic steatosis, ≥7% improves necroinflammation, and ≥10% stabilizes or reverses fibrosis. Dietary strategies, including Mediterranean-style patterns, high-protein approaches, and intermittent fasting, have been shown to be effective in improving insulin sensitivity and reducing intrahepatic triglycerides. Exercise interventions, focusing on both aerobic fitness and resistance training, enhance metabolic flexibility and combat sarcopenia, thereby improving hepatic and systemic outcomes. Equally important are behavioral support, digital health tools, and multidisciplinary approaches that enhance adherence and address barriers such as socioeconomic disparities, limited access, and patient engagement issues. Personalized nutrition plans, integrating physical activity, and ongoing support for behavioral change are essential for long-term disease management. This review synthesizes current evidence on the roles of macronutrients, micronutrients, dietary quality, physical activity, and adjunctive behavioral strategies in managing MASLD. By translating mechanistic insights into practical, evidence-based recommendations, we aim to provide clinicians, dietitians, and exercise professionals with effective frameworks to slow disease progression and improve outcomes across diverse patient populations. Full article
(This article belongs to the Special Issue Lifestyle Interventions in MASLD: Molecular and Clinical Perspectives)
Show Figures

Figure 1

25 pages, 1077 KB  
Review
Gut Permeability and Microbiota in Parkinson’s Disease: Mechanistic Insights and Experimental Therapeutic Strategies
by Yicheng Liang, Yuhang Zhao, Alessio Fasano and Chien-Wen Su
Int. J. Mol. Sci. 2025, 26(19), 9593; https://doi.org/10.3390/ijms26199593 - 1 Oct 2025
Viewed by 269
Abstract
Globally, Parkinson’s disease (PD) is the neurodegenerative condition with the most rapidly increasing prevalence, and a growing body of evidence associates its pathology with impairments in the gut–brain axis. Traditionally viewed as a disease marked by the loss of dopaminergic neurons, emerging evidence [...] Read more.
Globally, Parkinson’s disease (PD) is the neurodegenerative condition with the most rapidly increasing prevalence, and a growing body of evidence associates its pathology with impairments in the gut–brain axis. Traditionally viewed as a disease marked by the loss of dopaminergic neurons, emerging evidence emphasizes that chronic neuroinflammation is a driver of neurodegeneration, with gut-originating inflammation playing a crucial role. Increased intestinal permeability, often called “leaky gut,” allows harmful substances, toxins, and misfolded α-synuclein into the systemic circulation, potentially exacerbating neuroinflammation and spreading α-synuclein pathology to the brain through the vagus nerve or compromised blood–brain barrier (BBB). This review synthesizes current insights into the relationship between gut health and PD, emphasizing the importance of gut permeability in disrupting intestinal barrier function. This paper highlights innovative therapeutic approaches, particularly personalized therapies involving gut microbiome engineering, as promising strategies for restoring gut integrity and improving neurological outcomes. Modulating specific gut bacteria to enhance the synthesis of certain metabolites, notably short-chain fatty acids (SCFAs), represents a promising strategy for reducing inflammatory responses and decelerating neurodegeneration in Parkinson’s disease. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

25 pages, 1871 KB  
Review
Microbiota-Derived Extracellular Vesicles as Potential Mediators of Gut–Brain Communication in Traumatic Brain Injury: Mechanisms, Biomarkers, and Therapeutic Implications
by Tarek Benameur, Abeir Hasan, Hind Toufig, Maria Antonietta Panaro, Francesca Martina Filannino and Chiara Porro
Biomolecules 2025, 15(10), 1398; https://doi.org/10.3390/biom15101398 - 30 Sep 2025
Viewed by 333
Abstract
Traumatic brain injury (TBI) remains a major global health problem, contributing significantly to morbidity and mortality worldwide. Despite advances in understanding its complex pathophysiology, current therapeutic strategies are insufficient in addressing the long-term cognitive, emotional, and neurological impairments. While the primary mechanical injury [...] Read more.
Traumatic brain injury (TBI) remains a major global health problem, contributing significantly to morbidity and mortality worldwide. Despite advances in understanding its complex pathophysiology, current therapeutic strategies are insufficient in addressing the long-term cognitive, emotional, and neurological impairments. While the primary mechanical injury is immediate and unavoidable, the secondary phase involves a cascade of biological processes leading to neuroinflammation, blood–brain barrier (BBB) disruption, and systemic immune activation. The heterogeneity of patient responses underscores the urgent need for reliable biomarkers and targeted interventions. Emerging evidence highlights the gut–brain axis as a critical modulator of the secondary phase, with microbiota-derived extracellular vesicles (MEVs) representing a promising avenue for both diagnosis and therapy. MEVs can cross the intestinal barrier and BBB, carrying biomolecules that influence neuronal survival, synaptic plasticity, and inflammatory signaling. These properties make MEVs promising biomarkers for early detection, severity classification, and prognosis in TBI, while also offering therapeutic potential through modulation of neuroinflammation and promotion of neural repair. MEV-based strategies could enable tailored interventions based on the individual’s microbiome profile, immune status, and injury characteristics. The integration of multi-omics with artificial intelligence is expected to fully unlock the diagnostic and therapeutic potential of MEVs. These approaches can identify molecular subtypes, predict outcomes, and facilitate real-time clinical decision-making. By bridging microbiology, neuroscience, and precision medicine, MEVs hold transformative potential to advance TBI diagnosis, monitoring, and treatment. This review also identifies key research gaps and proposes future directions for MEVs in precision diagnostics and gut microbiota-based therapeutics in neurotrauma care. Full article
Show Figures

Figure 1

12 pages, 2307 KB  
Perspective
All That Glitters Ain’t Gold: The Myths and Scientific Realities About the Gut Microbiota
by Priyankar Dey
Nutrients 2025, 17(19), 3121; https://doi.org/10.3390/nu17193121 - 30 Sep 2025
Viewed by 1335
Abstract
Gut microbial modulation through diet is central to human health and disease. Despite tremendous effort in understanding the impact of nutrients and drugs on the gut microbiota, and attempts to develop dietary strategies that facilitate gut-beneficial effects, several erroneous gut microbiota-associated concepts remain [...] Read more.
Gut microbial modulation through diet is central to human health and disease. Despite tremendous effort in understanding the impact of nutrients and drugs on the gut microbiota, and attempts to develop dietary strategies that facilitate gut-beneficial effects, several erroneous gut microbiota-associated concepts remain prevalent in popular belief. This article discusses widespread misconceptions about the gut microbiota, contrasting them with contemporary scientific facts. In this article, ten prevalent myths, including the obsolete 10:1 bacteria-to-human-cell ratio, the reductive categorization of microbes as ‘good’ or ‘bad’, and the discredited universal biomarker status of the Firmicutes/Bacteroidetes ratio in relation to metabolic diseases, have been debunked. Essential facts highlighting the context-dependency of the microbiome, considerable inter-individual heterogeneity, and dynamic reactivity to dietary changes are discussed. This questions the assumptions that increased diversity always signifies health, that probiotics are intrinsically safe, that fecal microbiota transplantation is a universal remedy, or that leaky gut syndrome constitutes a clearly defined diagnosis. It is highlighted that eubiosis and dysbiosis do not possess uniform criteria, and microbiome–drug interactions are extremely individualized. The gut microbiota operates as a dynamic, adaptive ecosystem, necessitating sophisticated, evidence-based methodologies for study and therapeutic application, transcending simplistic misconceptions in favor of tailored insights and therapies. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

19 pages, 4603 KB  
Article
Impact of Different Microbial Biostimulants and Salt Stress on the Endophytome of the Edible Part of Lettuce and Tomato Plants
by José M. Mulet, Patricia Benito, Marina Celdrán, Lynne Yenush and Rosa Porcel
Foods 2025, 14(19), 3366; https://doi.org/10.3390/foods14193366 - 29 Sep 2025
Viewed by 379
Abstract
The human gut microbiota plays a critical role in health throughout life. While fruits and vegetables are well-known sources of nutrients and prebiotics, recent studies suggest they may also contribute viable microorganisms to the gut microbiome, particularly when consumed raw. However, the impact [...] Read more.
The human gut microbiota plays a critical role in health throughout life. While fruits and vegetables are well-known sources of nutrients and prebiotics, recent studies suggest they may also contribute viable microorganisms to the gut microbiome, particularly when consumed raw. However, the impact of agricultural practices—such as the use of microbial biostimulants or exposure to salt stress—on the composition of the edible plant microbiome remains poorly understood. In this study, we performed a comprehensive metataxonomic analysis of the endophytic microbiome in the edible tissues (leaves or fruits) of lettuce (Lactuca sativa) and tomato (Solanum lycopersicum), cultivated under standard conditions with or without microbial biostimulants and salt stress. Our results show that microbial biostimulants—Priestia megaterium (PGPB) and Rhizophagus irregularis (AMF)—as well as moderate salt stress, significantly reshape the composition and diversity of endophytes in both crops. Notably, the PGPB and NaCl treatments enhanced the abundance of bacterial genera such as Pantoea, Stenotrophomonas, and Massilia, which are associated with plant health and may have probiotic potential. Salt stress also increased alpha-diversity indices and favored the presence of Firmicutes and Bacteroidota, phyla commonly linked to a healthy human gut microbiome. Agronomic inputs used in organic and conventional farming, such as microbial biostimulants or controlled salt exposure, may represent novel strategies to enhance the microbial quality of fresh produce and promote gut microbial diversity through diet. Full article
(This article belongs to the Topic Microbes and Their Products for Sustainable Human Life)
Show Figures

Graphical abstract

15 pages, 1930 KB  
Article
Assessing Probiotic Efficacy: Short-Term Impact on Canine Gut Microbiota Using an In Vitro Colonic Fermentation Model
by Achraf Adib Lesaux, Jonas Ghyselinck, Cindy Duysburgh, Massimo Marzorati, Jonna E. B. Koper and Jake Burlet
Pets 2025, 2(4), 33; https://doi.org/10.3390/pets2040033 - 28 Sep 2025
Viewed by 417
Abstract
In dogs, gut microbiome dysbiosis is associated with several health conditions, including gastrointestinal disease. Probiotic supplementation can support a balanced gut microbiome. This study assessed the impact of a probiotic containing a mixture of Lacticaseibacillus casei, Limosilactobacillus fermentum, Levilactobacillus brevis, [...] Read more.
In dogs, gut microbiome dysbiosis is associated with several health conditions, including gastrointestinal disease. Probiotic supplementation can support a balanced gut microbiome. This study assessed the impact of a probiotic containing a mixture of Lacticaseibacillus casei, Limosilactobacillus fermentum, Levilactobacillus brevis, and Enterococcus faecium on the gut microbiota of six dogs using short-term colonic simulations. Two groups were included, i.e., blank versus supplementation with the test product, and incubated for 48 h. Probiotic-supplemented reactors had significantly greater fermentative activity compared with the blank, as shown by lower pH levels and higher gas pressure after 6 h, 24 h, and 48 h of incubation (p < 0.05 for all). Saccharolytic fermentation also increased, with a significantly higher level of acetate at 24 h and propionate at 6 h, 24 h, and 48 h with the test product versus blank (p < 0.05 for all). There was no significant effect of the test product on alpha-diversity, but beta-diversity analysis revealed a clear separation in the microbial community composition between the test product and blank. Eight bacterial taxa were enriched with test product supplementation, including the probiotic test strains as well as Megamonas and Bacteroides species. This study, using in vitro short-term colon simulations with six canine donors, provides insights into the probiotic characteristics of the test product. Full article
Show Figures

Graphical abstract

Back to TopTop