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
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
remove_circle_outline
remove_circle_outline

Search Results (4,919)

Search Parameters:
Keywords = gut-microbiome

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 930 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
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)
26 pages, 1714 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
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
20 pages, 5778 KB  
Article
Therapeutic Modulation of the Gut Microbiome by Supplementation with Probiotics (SCI Microbiome Mix) in Adults with Functional Bowel Disorders: A Randomized, Double-Blind, Placebo-Controlled Trial
by Won Yeong Bang, Jin Seok Moon, Hayoung Kim, Han Bin Lee, Donggyu Kim, Minhye Shin, Young Hoon Jung, Jongbeom Shin and Jungwoo Yang
Microorganisms 2025, 13(10), 2283; https://doi.org/10.3390/microorganisms13102283 - 30 Sep 2025
Abstract
Functional bowel disorders (FBDs) are chronic gastrointestinal conditions characterized by recurrent symptoms associated with gut microbiota dysbiosis. Although accumulating evidence suggests that probiotics can improve symptoms in patients with FBD, the underlying mechanisms remain to be fully elucidated. In this randomized, double-blind, placebo-controlled [...] Read more.
Functional bowel disorders (FBDs) are chronic gastrointestinal conditions characterized by recurrent symptoms associated with gut microbiota dysbiosis. Although accumulating evidence suggests that probiotics can improve symptoms in patients with FBD, the underlying mechanisms remain to be fully elucidated. In this randomized, double-blind, placebo-controlled clinical trial, 38 adults meeting the Rome IV diagnostic criteria of functional constipation (FC) and functional diarrhea (FD) received either a multi-strain probiotic complex or placebo for 8 weeks. Clinical outcomes were evaluated using the Irritable Bowel Syndrome Severity Scoring System (IBS-SSS), bowel habits questionnaire, and IBS Quality of Life (IBS-QoL) instrument. Fecal samples were collected at baseline and at week 8 for gut microbiota profiling via 16S rRNA gene sequencing and metabolomic analysis using gas chromatography–mass spectrometry. Probiotic supplementation significantly reduced the severity of abdominal bloating and its interference with quality of life, and improved the body image domain of the IBS-QoL. Beta diversity analysis showed significant temporal shifts in the probiotic group, while 16S rRNA sequencing revealed an increased relative abundance of Faecalibacterium prausnitzii and Blautia stercoris. Fecal metabolomic analysis further indicated elevated levels of metabolites implicated in the gut–brain axis. Multi-strain probiotic supplementation alleviated gastrointestinal symptoms and improved aspects of psychosocial well-being in adults with FBDs, potentially through modulation of the human gut microbiome. Full article
(This article belongs to the Section Gut Microbiota)
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
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

21 pages, 10082 KB  
Article
Ulvan-Na, an Ulvan Subjected to Na+ Cation Exchange, Improves Intestinal Barrier Function in Age-Related Leaky Gut
by Yuka Maejima, Yuki Morioka, Yusei Sato, Masanori Hiraoka, Ayumu Onda and Takushi Namba
Mar. Drugs 2025, 23(10), 390; https://doi.org/10.3390/md23100390 - 30 Sep 2025
Abstract
The global increase in life expectancy underscores the need to promote healthy aging, particularly by addressing age-related leaky gut syndrome, which contributes to systemic inflammation and chronic disease. This study focused on the sustainable production and functional development of Ulva meridionalis, a [...] Read more.
The global increase in life expectancy underscores the need to promote healthy aging, particularly by addressing age-related leaky gut syndrome, which contributes to systemic inflammation and chronic disease. This study focused on the sustainable production and functional development of Ulva meridionalis, a fast-growing seaweed, to improve gut health and mitigate the effects of aging. Using land-based aquaculture, a scalable cultivation system for U. meridionalis was established, and its polysaccharide, ulvan, was extracted. Ion exchange treatment enhanced the functionality of ulvan to produce ulvan-Na, which contains high levels of Na+ and conveys superior anti-aging properties. Ulvan-Na restored intestinal barrier integrity in aged mice by reducing serum LPS levels and increasing claudin-1 expression. Ulvan-Na modulated the gut microbiota, increasing beneficial bacteria such as Clostridiales vadin BB60 and suppressing inflammatory bacteria such as Turicibacter. The mechanism was clarified whereby ulvan-Na activates β-catenin to enhance claudin-1 expression. These findings highlight ulvan-Na as a bioactive compound that ameliorates age-related intestinal dysfunction while demonstrating the feasibility of sustainable U. meridionalis production for functional food innovation and environmental conservation. Full article
Show Figures

Figure 1

15 pages, 944 KB  
Review
TMAO and Cardiovascular Disease: Exploring Its Potential as a Biomarker
by Octavian Amaritei, Oana Laura Mierlan, Ciprian Adrian Dinu, Iulia Chiscop, Madalina Nicoleta Matei, Cristian Gutu and Gabriela Gurau
Medicina 2025, 61(10), 1767; https://doi.org/10.3390/medicina61101767 - 30 Sep 2025
Abstract
Gut microbiota has increasingly been shown to exert effects beyond the gastrointestinal tract, some of which are mediated through its metabolites, such as trimethylamine N-oxide (TMAO)—a compound converted by gut bacteria from dietary choline found predominantly in animal products that is associated with [...] Read more.
Gut microbiota has increasingly been shown to exert effects beyond the gastrointestinal tract, some of which are mediated through its metabolites, such as trimethylamine N-oxide (TMAO)—a compound converted by gut bacteria from dietary choline found predominantly in animal products that is associated with cardiovascular disease (CVD). However, a significant gap persists in human clinical trials assessing its potential causal role. This narrative review aims to present the current understanding of the gut microbiome, TMAO, and their relationship with CVD, while proposing future directions that may support the use of TMAO as a biomarker and guide potential interventions to reduce its harmful impact. Both animal and human studies have demonstrated a link between TMAO and CVD, with animal studies also indicating a causal effect—showing increased cardiovascular risk following TMAO administration and reduced risk when TMAO is eliminated. While direct extrapolation from animal models to humans is limited due to biological differences, these findings offer a foundation for the development of well-designed clinical trials in human populations. Although direct approaches to target TMAO—such as trimethylamine (TMA) lyase inhibitors and antisense oligonucleotide (ASO) therapy—have shown promising results in animal studies, they have yet to be investigated in human trials, leaving indirect strategies such as dietary changes and probiotics as the only currently available options. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

18 pages, 957 KB  
Review
Unveiling the Microbiome’s Role in Hidradenitis Suppurativa: A Comprehensive Review of Pathogenetic Mechanisms
by Catarina Queirós, Carmen Lisboa and Sofia Magina
Int. J. Mol. Sci. 2025, 26(19), 9542; https://doi.org/10.3390/ijms26199542 - 30 Sep 2025
Abstract
Hidradenitis suppurativa (HS) is a chronic, recurrent, and highly debilitating inflammatory disorder of the pilosebaceous unit. Its pathogenesis is considered multifactorial, involving genetic, environmental, hormonal, lifestyle, and microbiome-related factors. The microbiota, defined as the collection of microorganisms, their genomes, and their interactions within [...] Read more.
Hidradenitis suppurativa (HS) is a chronic, recurrent, and highly debilitating inflammatory disorder of the pilosebaceous unit. Its pathogenesis is considered multifactorial, involving genetic, environmental, hormonal, lifestyle, and microbiome-related factors. The microbiota, defined as the collection of microorganisms, their genomes, and their interactions within a given environment, colonizes multiple sites of the healthy human body, which include the skin and gut, where it contributes to the maintenance of homeostasis. In HS, both skin and gut microbiota exhibit disruptions in composition and diversity, a state referred to as dysbiosis. Alterations in the expression of antimicrobial peptides in HS further implicate the microbiome in disease pathophysiology. In addition, chronic inflammation, bacterial biofilm formation, and dysbiosis are thought to contribute to the severity and recurrence of HS. Although the precise role of dysbiosis in HS pathogenesis remains unclear, several studies have demonstrated a reduction in cutaneous microbial diversity in HS patients, distinguished by an increased abundance of anaerobic and opportunistic bacteria and a reduction in commensal species. The intestinal microbiome has been even less thoroughly investigated, but available evidence suggests decreased overall diversity and richness, with enrichment of pro-inflammatory and depletion of anti-inflammatory bacterial taxa. This review aims to provide an overview of the current knowledge regarding the role of the microbiome in HS, with the goal of informing the direction of future research, including the potential utility of the microbiome as a biomarker for diagnosis and severity stratification in HS. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

24 pages, 763 KB  
Review
Methylphenidate and Its Impact on Redox Balance and Behavior
by George Jîtcă, Ingrid Evelin Mehelean, Ana Natalia Maier and Carmen-Maria Jîtcă
J. Xenobiot. 2025, 15(5), 157; https://doi.org/10.3390/jox15050157 - 30 Sep 2025
Abstract
Methylphenidate (MPH) and its active enantiomer, dexmethylphenidate, are widely prescribed as first-line therapies for attention deficit hyperactivity disorder (ADHD), yet their increasing non-medical use highlights significant clinical and toxicological challenges. MPH blocks dopamine (DAT) and norepinephrine (NET) transporters, thereby elevating synaptic catecholamine levels. [...] Read more.
Methylphenidate (MPH) and its active enantiomer, dexmethylphenidate, are widely prescribed as first-line therapies for attention deficit hyperactivity disorder (ADHD), yet their increasing non-medical use highlights significant clinical and toxicological challenges. MPH blocks dopamine (DAT) and norepinephrine (NET) transporters, thereby elevating synaptic catecholamine levels. While this underpins therapeutic efficacy, prolonged or abusive exposure has been associated with mitochondrial impairment, disrupted bioenergetics, and excessive reactive oxygen species (ROS) production, which collectively contribute to neuronal stress and long-term neurotoxicity. Growing evidence suggests that the gut–brain axis may critically influence MPH outcomes: diet-induced shifts in microbiome composition appear to regulate oxidative stress, neuroinflammation, and drug metabolism, opening potential avenues for dietary or probiotic interventions. From a forensic perspective, the detection and monitoring of MPH misuse require advanced methodologies, including enantioselective LC–MS/MS and analysis of alternative matrices such as hair or oral fluids, which enable retrospective exposure assessment and improves abuse surveillance. Despite its established therapeutic profile, MPH remains a compound with a narrow balance between clinical benefit and toxicological risk. Future directions should prioritize longitudinal human studies, biomarker identification for abuse monitoring, and the development of mitochondria-targeted therapies to minimize adverse outcomes and enhance safety in long-term treatment. Full article
Show Figures

Figure 1

12 pages, 486 KB  
Article
Research on the Influence of Enterobius vermicularis on the Composition and Quality of the Intestinal Microbiota, and the Susceptibility to Co-Infections
by Eleonora Kaneva, Rumen Harizanov, Maria Pavlova, Desislava Velcheva, Nina Tsvetkova, Aleksandra Ivanova, Mihaela Videnova, Raina Borisova, Ivailo Alexiev and Reneta Dimitrova
Microbiol. Res. 2025, 16(10), 215; https://doi.org/10.3390/microbiolres16100215 - 30 Sep 2025
Abstract
The present study examined the presence of concomitant intestinal parasites and bacteria in individuals infected with Enterobius vermicularis in Bulgaria, and analyzed its effects on the intestinal microbiome and the risk of co-infection. Fecal samples from people with and without (control group) enterobiasis [...] Read more.
The present study examined the presence of concomitant intestinal parasites and bacteria in individuals infected with Enterobius vermicularis in Bulgaria, and analyzed its effects on the intestinal microbiome and the risk of co-infection. Fecal samples from people with and without (control group) enterobiasis were tested for the presence of concomitant bacterial infection. The results were compared to find out about the intestinal microbiome in these groups. The microbiological examination of the control group showed that 8.3% had only conditionally pathogenic flora. However, in individuals with enterobiasis, 56% had normal intestinal bacterial flora, but 46% had disturbed microbiota: 7% were carriers of pathogenic intestinal bacteria and 24% had opportunistic pathogens. The most prevalent were Klebsiella pneumoniae (49%), followed by Enterobacter spp. (22%), and Proteus mirabilis (12.2%). Our study is the first in the country to investigate the impact of E. vermicularis infection on the composition and quality of the gut microbiome, as well as the potential for co-infections with pathogenic gut bacteria. Although our findings are preliminary, they suggest that this nematode may significantly contribute to a predisposition for dysbiosis or the onset of secondary bacterial infections. Full article
Show Figures

Figure 1

19 pages, 300 KB  
Article
Mechanistic Insights into the Physiological and Meat Quality Responses of Broiler Chickens Fed Incremental Turmeric Rhizome Meal
by Uchenna Nonyelum Okonkwo, Christiaan Jacobus Smit and Chidozie Freedom Egbu
Animals 2025, 15(19), 2849; https://doi.org/10.3390/ani15192849 - 29 Sep 2025
Abstract
Natural products, such as turmeric rhizome meal (TRM), may hold the key to a sustainable solution to antimicrobial resistance rise and antibiotic prohibition in food-producing animals. This study evaluated the effects of dietary TRM at 0 (CON), 0.3 (TRM3), 0.6 (TRM6), and 0.9 [...] Read more.
Natural products, such as turmeric rhizome meal (TRM), may hold the key to a sustainable solution to antimicrobial resistance rise and antibiotic prohibition in food-producing animals. This study evaluated the effects of dietary TRM at 0 (CON), 0.3 (TRM3), 0.6 (TRM6), and 0.9 g/kg (TRM9) on growth, nutrient digestibility, immunity, gut function, nutrient transport biomarkers, microbiome, and meat quality in 280 one-day-old male Ross 308 chicks over a 42-day feeding trial. Birds fed TRM indicated higher body weight gain and lower feed conversion ratio (p < 0.05). The TRM groups promoted higher (p = 0.001) serum immunoglobulin Y, immunoglobulin M, and interleukin-10 compared to the CON. Birds fed CON had higher interleukin-2 (p = 0.025), interleukin-6 (p = 0.027), and TNF-α (p = 0.008) levels compared to the TRM groups. Lactobacillus counts in jejunal villi and crypts were higher in the TRM groups than in the CON (p < 0.05). Dietary TRM increased electrogenic glucose and lysine transport, accompanied by up-regulation of claudin-5, zonula occludens 1, and mucin-2 expression (p < 0.05). In breast muscle, TRM fortification reduced malondialdehyde levels (p < 0.05) while increasing long-chain polyunsaturated fatty acids (p < 0.05). Thus, TRM is a potent, residue-free phytobiotic alternative to conventional antibiotic growth promoters in poultry systems. Full article
(This article belongs to the Section Poultry)
17 pages, 1676 KB  
Article
Promoting Re-Epithelialization in Diabetic Foot Wounds Using Integrative Therapeutic Approaches
by Lucia Bubulac, Iuliana-Raluca Gheorghe, Elisabeth Ungureanu, Claudia Florina Bogdan-Andreescu, Cristina-Crenguța Albu, Consuela-Mădălina Gheorghe, Ovidiu Mușat, Irina Anca Eremia, Cristina Aura Panea and Alexandru Burcea
Bioengineering 2025, 12(10), 1053; https://doi.org/10.3390/bioengineering12101053 - 29 Sep 2025
Abstract
Background: Diabetes mellitus is a heterogeneous chronic disease with an increasing global prevalence. In Romania, 11.6% of the population is affected, yet only 6.46% receive treatment. Among diabetic patients, 15–25% develop skin lesions that may progress to ulceration and necrosis, significantly impairing [...] Read more.
Background: Diabetes mellitus is a heterogeneous chronic disease with an increasing global prevalence. In Romania, 11.6% of the population is affected, yet only 6.46% receive treatment. Among diabetic patients, 15–25% develop skin lesions that may progress to ulceration and necrosis, significantly impairing quality of life and increasing the risk of complications. Methods: We conducted a prospective study including 28 patients (14 in the control group and 14 in the intervention group) with type I or II diabetes and chronic ulcers of the calf or foot (>4 cm2). The control group received standard therapy with debridement, dressings, antibiotics when indicated, and local and systemic ozone therapy. The intervention group was treated with an Integrative Therapeutic Protocol combining ozone therapy, pulsed electromagnetic field therapy (PEMF), colon hydrotherapy with probiotic supplementation, and an anti-inflammatory alkaline diet. Wound healing (reduction in ulcer surface area) was the primary endpoint; secondary endpoints included changes in glycemia and inflammatory biomarkers. Results: After 8 weeks, the intervention group achieved 86.2% re-epithelialization versus 58.2% in controls (p < 0.01). Significant improvements were also observed in blood glucose level (−38%), HbA1c (−25%), CRP (−26%), and fibrinogen (−28%) relative to baseline, with differences versus controls reaching statistical significance. Conclusions: The Integrative Therapeutic Protocol accelerated wound healing and improved glycemic and inflammatory profiles compared with ozone therapy alone. Although an alkaline diet was recommended, adherence and its specific contribution were not objectively monitored; therefore, this component should be interpreted with caution. Full article
(This article belongs to the Special Issue Recent Advancements in Wound Healing and Repair)
Show Figures

Graphical abstract

17 pages, 3465 KB  
Article
Longitudinal Gut Microbiome Changes Associated with Transitions from C. difficile Negative to C. difficile Positive on Surveillance Tests
by L. Silvia Munoz-Price, Samantha N. Atkinson, Vy Lam, Blake Buchan, Nathan Ledeboer, Nita H. Salzman and Amy Y. Pan
Microorganisms 2025, 13(10), 2277; https://doi.org/10.3390/microorganisms13102277 - 29 Sep 2025
Abstract
Clostridioides difficile is an obligate anaerobe and is primarily transmitted via the fecal–oral route. Data characterizing the microbiome changes accompanying transitions from non-colonized to C. difficile colonized subjects are currently lacking. In this retrospective cohort study, we examined 16S rRNA gene sequencing data [...] Read more.
Clostridioides difficile is an obligate anaerobe and is primarily transmitted via the fecal–oral route. Data characterizing the microbiome changes accompanying transitions from non-colonized to C. difficile colonized subjects are currently lacking. In this retrospective cohort study, we examined 16S rRNA gene sequencing data in a total of 481 fecal samples belonging to 107 patients. Based on C. difficile status over time, patients were categorized as Negative-to-Positive, Negative Control, and Positive Control. A linear mixed effects model was fitted to investigate the changes in the Shannon α-diversity index over time. Zero-inflated negative binomial/Poisson mixed effects models or generalized linear mixed models with negative binomial/Poisson distribution were used to investigate the changes in taxon counts over time among different groups. A total of 107 patients were eligible for the study. The median number of stool samples per patient was 3 (IQR 2–4). A total of 42 patients transitioned from C. difficile negative to positive (Negative-to-Positive), 47 patients remained negative throughout their tests (Negative Control) and 18 were always C. difficile positive (Positive Control). A significant difference in microbiome composition between the last negative samples and the first positive samples were shown in Negative-to-Positive patients, ANOSIM p = 0.022. In Negative-to-Positive patients, the phylum Pseudomonadota and family Enterobacteriaceae increased significantly in the first positive samples compared to the last negative samples, p = 0.0075 and p = 0.0094, respectively. Within the first 21 days, Actinomycetota decreased significantly over time in the Positive Control group compared to the other two groups (p < 0.001) while Bacillota decreased in both the Negative-to-Positive group and Positive Control. These results demonstrate that the transition from C. difficile negative to C. difficile positive is associated with alterations in gut microbial communities and their compositional patterns over time. Moreover, these changes play an important role in both the emergence and intensification of the gut microbiome dysbiosis in patients who transitioned from C. difficile negative to positive and those who always tested positive. Full article
(This article belongs to the Special Issue The Microbiome in Ecosystems)
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
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

24 pages, 935 KB  
Review
Keystone Species Restoration: Therapeutic Effects of Bifidobacterium infantis and Lactobacillus reuteri on Metabolic Regulation and Gut–Brain Axis Signaling—A Qualitative Systematic Review (QualSR)
by Michael Enwere, Edward Irobi, Adamu Onu, Emmanuel Davies, Gbadebo Ogungbade, Omowunmi Omoniwa, Charles Omale, Mercy Neufeld, Victoria Chime, Ada Ezeogu, Dung-Gwom Pam Stephen, Terkaa Atim and Laurens Holmes
Gastrointest. Disord. 2025, 7(4), 62; https://doi.org/10.3390/gidisord7040062 - 28 Sep 2025
Abstract
Background: The human gut microbiome—a diverse ecosystem of trillions of microorganisms—plays an essential role in metabolic, immune, and neurological regulation. However, modern lifestyle factors such as antibiotic overuse, cesarean delivery, reduced breastfeeding, processed and high-sodium diets, alcohol intake, smoking, and exposure to [...] Read more.
Background: The human gut microbiome—a diverse ecosystem of trillions of microorganisms—plays an essential role in metabolic, immune, and neurological regulation. However, modern lifestyle factors such as antibiotic overuse, cesarean delivery, reduced breastfeeding, processed and high-sodium diets, alcohol intake, smoking, and exposure to environmental toxins (e.g., glyphosate) significantly reduce microbial diversity. Loss of keystone species like Bifidobacterium infantis (B. infantis) and Lactobacillus reuteri (L. reuteri) contributes to gut dysbiosis, which has been implicated in chronic metabolic, autoimmune, cardiovascular, and neurodegenerative conditions. Materials and Methods: This Qualitative Systematic Review (QualSR) synthesized data from over 547 studies involving human participants and standardized microbiome analysis techniques, including 16S rRNA sequencing and metagenomics. Studies were reviewed for microbial composition, immune and metabolic biomarkers, and clinical outcomes related to microbiome restoration strategies. Results: Multiple cohort studies have consistently reported a 40–60% reduction in microbial diversity among Western populations compared to traditional societies, particularly affecting short-chain fatty acid (SCFA)-producing bacteria. Supplementation with B. infantis is associated with a significant reduction in systemic inflammation—including a 50% decrease in C-reactive protein (CRP) and reduced tumor necrosis factor-alpha (TNF-α) levels—alongside increases in regulatory T cells and anti-inflammatory cytokines interleukin-10 (IL-10) and transforming growth factor-beta 1 (TGF-β1). L. reuteri demonstrates immunomodulatory and neurobehavioral benefits in preclinical models, while both probiotics enhance epithelial barrier integrity in a strain- and context-specific manner. In murine colitis, B. infantis increases ZO-1 expression by ~35%, and L. reuteri improves occludin and claudin-1 localization, suggesting that keystone restoration strengthens barrier function through tight-junction modulation. Conclusions: Together, these findings support keystone species restoration with B. infantis and L. reuteri as a promising adjunctive strategy to reduce systemic inflammation, reinforce gut barrier integrity, and modulate gut–brain axis (GBA) signaling, indicating translational potential in metabolic and neuroimmune disorders. Future research should emphasize personalized microbiome profiling, long-term outcomes, and transgenerational effects of early-life microbial disruption. Full article
(This article belongs to the Special Issue Feature Papers in Gastrointestinal Disorders in 2025–2026)
Show Figures

Figure 1

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
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