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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (434)

Search Parameters:
Keywords = trimethylamine N-Oxide

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 1084 KB  
Review
Nutritional Modulation of the Gut–Kidney Axis
by Razvan George Bogdan, Felicia Gabriela Gligor, Paula Anderco, Livia Mirela Popa, Adriana Popescu, Vlad Adam Bloanca, Elisa Leonte, Mihai Iliescu Glaja, Zorin Petrisor Crainiceanu and Cristian Ichim
Nutrients 2026, 18(2), 263; https://doi.org/10.3390/nu18020263 - 14 Jan 2026
Viewed by 34
Abstract
Background: Chronic kidney disease (CKD) represents a state of persistent, sterile low-grade inflammation in which sustained innate immune activation accelerates renal decline and cardiovascular complications. Diet-induced gut dysbiosis and intestinal barrier dysfunction lower mucosal immune tolerance, promote metabolic endotoxemia, and position the gut [...] Read more.
Background: Chronic kidney disease (CKD) represents a state of persistent, sterile low-grade inflammation in which sustained innate immune activation accelerates renal decline and cardiovascular complications. Diet-induced gut dysbiosis and intestinal barrier dysfunction lower mucosal immune tolerance, promote metabolic endotoxemia, and position the gut as an upstream modulator of systemic inflammatory signaling along the gut–kidney axis. Scope: Most studies address microbiota-derived metabolites, food-derived bioactive peptides, or omega-3 fatty acids separately. This review integrates evidence across these domains and examines their convergent actions on epithelial barrier integrity, immune polarization, oxidative-inflammatory stress, and inflammasome-dependent pathways relevant to CKD progression. Key mechanisms: CKD-associated dysbiosis is characterized by reduced short-chain fatty acid (SCFA) production and increased generation and accumulation of uremic toxins and co-metabolites, including indoxyl sulfate, p-cresyl sulfate, trimethylamine N-oxide, and altered bile acids. Reduced SCFA availability weakens tight junction-dependent barrier function and regulatory immune programs, favoring Th17-skewed inflammation and endotoxin translocation. Bioactive peptides modulate inflammatory mediator networks and barrier-related pathways through effects on NF-κB/MAPK signaling and redox balance, while omega-3 fatty acids and specialized pro-resolving mediators support resolution-phase immune responses. Across these modalities, shared control points include barrier integrity, metabolic endotoxemia, oxidative stress, and NLRP3 inflammasome activation. Conclusions: Although evidence remains heterogeneous and largely preclinical, combined nutritional modulation targeting these convergent pathways may offer greater immunomodulatory benefit than isolated interventions. Future multi-omics-guided, factorial trials are required to define responder phenotypes and translate precision immunonutrition strategies into clinical CKD care. Full article
Show Figures

Figure 1

52 pages, 4367 KB  
Review
The Microbiome–Neurodegeneration Interface: Mechanisms, Evidence, and Future Directions
by Lilia Böckels, Daniel Alexa, Dorin Cristian Antal, Cristina Gațcan, Cosmin Alecu, Kristina Kacani, Raul Andrei Crețu, Emanuel Andrei Piseru, Robert Valentin Bîlcu and Dan Iulian Cuciureanu
Cells 2026, 15(2), 135; https://doi.org/10.3390/cells15020135 - 12 Jan 2026
Viewed by 453
Abstract
The gut microbiota has emerged as a central regulator of the gut–brain axis, profoundly influencing neural, immune, and metabolic homeostasis. Increasing evidence indicates that disturbances in microbial composition and function contribute to the onset and progression of neurodegenerative diseases (NDs) through mechanisms involving [...] Read more.
The gut microbiota has emerged as a central regulator of the gut–brain axis, profoundly influencing neural, immune, and metabolic homeostasis. Increasing evidence indicates that disturbances in microbial composition and function contribute to the onset and progression of neurodegenerative diseases (NDs) through mechanisms involving neuroinflammation, oxidative stress, and impaired neurotransmission. Gut dysbiosis is characterized by a loss of microbial diversity, a reduction in beneficial commensals, and an enrichment of pro-inflammatory taxa. These shifts alter intestinal permeability and systemic immune tone, allowing microbial metabolites and immune mediators to affect central nervous system (CNS) integrity. Metabolites such as short-chain fatty acids (SCFAs), tryptophan derivatives, lipopolysaccharides (LPS), and trimethylamine N-oxide (TMAO) modulate blood–brain barrier (BBB) function, microglial activation, and neurotransmitter synthesis, linking intestinal imbalance to neuronal dysfunction and cognitive decline. Disruption of this gut–brain communication network promotes chronic inflammation and metabolic dysregulation, key features of neurodegenerative pathology. SCFA-producing and tryptophan-metabolizing bacteria appear to exert neuroprotective effects by modulating immune responses, epigenetic regulation, and neuronal resilience. The aim of this work was to comprehensively explore the current evidence on the bidirectional communication between the gut microbiota and the CNS, with a focus on identifying the principal molecular, immune, and metabolic mechanisms supported by the strongest and most consistent data. By integrating findings from recent human studies, this review sought to clarify how microbial composition and function influence neurochemical balance, immune activation, and BBB integrity, ultimately contributing to the onset and progression of neurodegenerative processes. Collectively, these findings position the gut microbiota as a dynamic interface between the enteric and CNS, capable of influencing neurodegenerative processes through immune and metabolic signaling. Full article
Show Figures

Figure 1

20 pages, 317 KB  
Review
Diet, Physical Exercise, and Gut Microbiota Modulation in Metabolic Syndrome: A Narrative Review
by Ana Onu, Andrei Tutu, Daniela-Marilena Trofin, Ilie Onu, Anca-Irina Galaction, Cristiana Amalia Onita, Daniel-Andrei Iordan and Daniela-Viorelia Matei
Life 2026, 16(1), 98; https://doi.org/10.3390/life16010098 - 10 Jan 2026
Viewed by 193
Abstract
Background: Metabolic syndrome (MetS) is a multifactorial condition characterized by insulin resistance, dyslipidemia, hypertension, and central obesity, and is strongly influenced by lifestyle factors. Growing evidence highlights the gut microbiota as a key mediator linking diet and physical exercise to cardiometabolic health. Objective: [...] Read more.
Background: Metabolic syndrome (MetS) is a multifactorial condition characterized by insulin resistance, dyslipidemia, hypertension, and central obesity, and is strongly influenced by lifestyle factors. Growing evidence highlights the gut microbiota as a key mediator linking diet and physical exercise to cardiometabolic health. Objective: This narrative review aims to qualitatively synthesize current evidence on the effects of physical exercise and major dietary patterns including the Mediterranean diet (MedDiet), Dietary Approaches to Stop Hypertension (DASH), and ketogenic/very-low-calorie ketogenic diets (KD/VLCKD) on gut microbiota composition and function, and their implications for metabolic health in MetS. Methods: A qualitative narrative synthesis of experimental, observational, and interventional human and animal studies was performed. The reviewed literature examined associations between structured physical exercise or dietary interventions and changes in gut microbiota diversity, key bacterial taxa, microbial metabolites, and cardiometabolic outcomes. Considerable heterogeneity across studies was noted, including differences in populations, intervention duration and intensity, dietary composition, and microbiota assessment methodologies. Results: Across human interventional studies, moderate-intensity physical exercise was most consistently associated with increased gut microbial diversity and enrichment of short-chain fatty acid (SCFA)-producing taxa, contributing to improved insulin sensitivity and reduced inflammation. MedDiet and DASH were generally linked to favorable microbiota profiles, including increased abundance of Faecalibacterium prausnitzii, Akkermansia muciniphila, and Bifidobacterium, alongside reductions in pro-inflammatory metabolites such as lipopolysaccharides and trimethylamine N-oxide. In contrast, KD and VLCKD were associated with rapid weight loss and glycemic improvements but frequently accompanied by reductions in SCFA-producing bacteria, depletion of Bifidobacterium, and markers of impaired gut barrier integrity, raising concerns regarding long-term microbiota resilience. Conclusions: Lifestyle-based interventions exert diet- and exercise-specific effects on the gut microbiota–metabolism axis. While MedDiet, DASH, and regular moderate physical activity appear to promote sustainable microbiota-mediated cardiometabolic benefits, ketogenic approaches require careful personalization, limited duration, and medical supervision. These findings support the integration of dietary quality, exercise prescription, and individual microbiota responsiveness into translational lifestyle strategies for MetS prevention and management. Full article
19 pages, 3913 KB  
Article
Gut Microbiota-Derived Metabolite and Heart Failure with Reduced Ejection Fraction (HFrEF): Elevated Trimethylamine N-Oxide (TMAO) as a Potential Biomarker
by Sheh Wen Kuan, Wei Leik Ng, Alexander Loch, Kek Heng Chua, Kim-Kee Tan and Boon Pin Kee
Int. J. Mol. Sci. 2026, 27(2), 703; https://doi.org/10.3390/ijms27020703 - 9 Jan 2026
Viewed by 184
Abstract
Gut-derived metabolites, particularly trimethylamine N-oxide (TMAO), have been implicated in the pathophysiology of heart failure (HF). This study investigated the associations between TMAO, cardiac function, and clinical parameters to evaluate TMAO’s potential as a biomarker for heart failure with reduced ejection fraction (HFrEF). [...] Read more.
Gut-derived metabolites, particularly trimethylamine N-oxide (TMAO), have been implicated in the pathophysiology of heart failure (HF). This study investigated the associations between TMAO, cardiac function, and clinical parameters to evaluate TMAO’s potential as a biomarker for heart failure with reduced ejection fraction (HFrEF). Forty HFrEF patients and forty-one matched healthy controls were recruited for serum TMAO quantification using enzyme-linked immunosorbent assay (ELISA). Associations were examined using Spearman correlation and regression models. TMAO levels were significantly elevated in HFrEF patients (3.64 µM [IQR 3.00–4.31]) compared with controls (1.22 µM [IQR 0.92–2.36]) (p < 0.05). Elevated TMAO correlated with impaired cardiac structural and functional parameters, as well as lower serum albumin. Multinomial regression revealed that both TMAO (OR 1.83, 95% CI 1.04–3.23, p = 0.036; OR 2.05, 95% CI 1.18–3.57, p = 0.010, respectively) and albumin (OR 0.56, 95% CI 0.36–0.89, p = 0.015; OR 0.61, 95% CI 0.39–0.93, p = 0.022, respectively) were independently associated with HFrEF severity, showing significant correlations in both mildly (EF 30–40%) and moderately (20–30%) reduced EF groups. Receiver operating characteristic (ROC) analyses showed that TMAO had good discriminative ability for HFrEF (AUC = 0.853), and it improved when combined with clinical covariates (AUC = 0.967), supporting its role as a potential biomarker. These findings support integrating this gut-derived metabolite and nutritional marker into HFrEF risk stratification frameworks. Full article
Show Figures

Figure 1

22 pages, 861 KB  
Review
Gut-Kidney Axis: Unraveling the Role of the Microbiome in Chronic Kidney Disease
by Mihai Rusu, Cristian Ichim, Paula Anderco, Andreea Pălăștea and Adrian Boicean
Biomedicines 2026, 14(1), 109; https://doi.org/10.3390/biomedicines14010109 - 6 Jan 2026
Viewed by 301
Abstract
Chronic kidney disease (CKD), which affects over 850 million individuals globally, is increasingly regarded as a systemic condition in which the gut microbiota represents a key pathogenic node. This review provides an integrated overview of mechanistic, translational and clinical data implicating the gut–kidney [...] Read more.
Chronic kidney disease (CKD), which affects over 850 million individuals globally, is increasingly regarded as a systemic condition in which the gut microbiota represents a key pathogenic node. This review provides an integrated overview of mechanistic, translational and clinical data implicating the gut–kidney axis in CKD. The CKD-associated microbiota displays a characteristic dysbiosis, marked by depletion of short-chain fatty acid–producing commensals, overgrowth of proteolytic and urease-expressing taxa and disruption of epithelial barrier integrity. These disturbances favor the generation and systemic accumulation of gut-derived uremic toxins, most notably indoxyl sulfate, p-cresyl sulfate, indole-3-acetic acid and trimethylamine-N-oxide, which promote endothelial dysfunction, vascular calcification, fibrosis and chronic inflammation, thereby hastening renal function loss and heightening cardiovascular risk. Microbiome-directed interventions, including dietary modification, prebiotics, probiotics, synbiotics, intestinal dialysis, fecal microbiota transplantation, gut-acting sorbents and nephroprotective phytochemicals, are summarized with emphasis on their effects on uremic toxin burden and clinical surrogates. System-level implications of the gut–kidney axis for cardiovascular disease, immunosenescence and sarcopenia are discussed, together with future priorities for integrating multi-omics profiling and precision microbiome-based strategies into nephrology practice. Full article
(This article belongs to the Section Microbiology in Human Health and Disease)
Show Figures

Figure 1

13 pages, 254 KB  
Article
Intestinal Permeability Biomarkers for Predicting Cardiometabolic Risk in Type 2 Diabetes Mellitus
by Nursel Dal, Saniye Bilici, Sirin Akin and Perim Fatma Turker
Nutrients 2026, 18(1), 167; https://doi.org/10.3390/nu18010167 - 4 Jan 2026
Viewed by 375
Abstract
Background: Diabetes can increase cardiovascular risk (CVR) through hyperglycemia and intestinal damage. The purpose of this study is to evaluate several intestinal permeability biomarkers in predicting CVR in patients with type 2 diabetes mellitus (T2DM). Methods: This study was conducted in 2024 with [...] Read more.
Background: Diabetes can increase cardiovascular risk (CVR) through hyperglycemia and intestinal damage. The purpose of this study is to evaluate several intestinal permeability biomarkers in predicting CVR in patients with type 2 diabetes mellitus (T2DM). Methods: This study was conducted in 2024 with a total of 70 patients with T2DM, aged 19–64 years (32.9% men, 67.1% women). Socio-demographic data and health status were collected; Framingham Risk Score (FRS), anthropometric measures, and serum parameters (glucose, HbA1c, lipids, CRP, TNF-α, IL-6, trimetilamine-N-oxide (TMAO), zonulin, intestinal fatty acid binding protein (I-FABP)) were evaluated, and visceral adiposity index (VAI) and plasma atherogenic index (PAI) were calculated. Results: The mean age of patients (n = 70) was 55.0 ± 7.55 years. According to FRS, 18.5% of individuals were determined to be at medium–high CVR; a positive correlation was found between BMI, waist–height ratio, body fat ratio, VAI value, and FRS total score (p < 0.05). Serum TMAO, zonulin, and I-FABP levels did not differ between low-risk and medium–high-risk patients (p > 0.05). Serum TMAO, zonulin, and I-FABP levels were positively correlated with TNF-α and IL-6 levels, and serum TMAO and I-FABP levels were positively correlated with triglyceride levels (p < 0.05). Moreover, serum zonulin and I-FABP levels were positively correlated with PAI (p < 0.05). Conclusions: Abdominal obesity and intestinal permeability may affect inflammatory processes and blood lipids in patients with T2DM. Further studies with large samples are needed to examine dietary factors related to the relationship between intestinal permeability and cardiometabolic risk. Full article
(This article belongs to the Special Issue Diet, Gut Health, and Clinical Nutrition)
14 pages, 2395 KB  
Article
Systemic Metabolomic Remodeling in Pressure Overload-Induced Heart Failure Indicates Modulation of a Gut–Liver–Heart Axis by the Adiponectin Receptor Agonist ALY688
by Yubin Lei, Benjie Li, Tori Gosse, Sungji Cho, Hye Kyoung Sung, Jiarui Chen and Gary Sweeney
Metabolites 2026, 16(1), 38; https://doi.org/10.3390/metabo16010038 - 1 Jan 2026
Viewed by 255
Abstract
Background/Objectives: Numerous studies have documented cardioprotective effects of adiponectin in animal models of cardiometabolic disease (CMD). Adiponectin receptor agonist ALY688 has demonstrated functional significance against pressure overload-induced cardiac remodeling events in a mouse model of heart failure with reduced ejection fraction (HFrEF), potentially [...] Read more.
Background/Objectives: Numerous studies have documented cardioprotective effects of adiponectin in animal models of cardiometabolic disease (CMD). Adiponectin receptor agonist ALY688 has demonstrated functional significance against pressure overload-induced cardiac remodeling events in a mouse model of heart failure with reduced ejection fraction (HFrEF), potentially through modulation of the systemic metabolome. However, the specific metabolites and their pathophysiological contribution to cardioprotection in cardiac hypertrophy or heart failure remain unclear. This study aimed to characterize systemic metabolic alterations across five tissues in HFrEF and determine how ALY688 modifies these pathways to mediate cardioprotection in the transverse aortic constriction (TAC) model. Methods: Targeted metabolic profiling was performed on heart, liver, muscle, epididymal white adipose tissue (eWAT), and serum collected five weeks post-surgery from wild-type male C57BL/6 mice. Mice underwent either Sham or TAC-induced left ventricular pressure overload, with or without daily subcutaneous ALY688 administration. Metabolites were quantified using liquid chromatography–tandem mass spectrometry (LC–MS/MS) and statistically analyzed at the tissue level. Results: Consistent with pathological cardiac remodeling, the comprehensive metabolomic analysis revealed that TAC induced widespread disruption of systemic metabolic homeostasis. ALY688 treatment significantly modified several key metabolite classes, including triglycerides (TGs) and glycosylceramides (HexCer). Notably, ALY688 also altered multiple gut-derived metabolites, including trimethylamine N-oxide (TMAO), 5-aminovaleric acid (5-AVA), and glycodeoxycholic acid (GDCA), highlighting a potential gut–liver–heart axis mediating its cardioprotective effects. Conclusions: These findings demonstrate that ALY688 mitigates TAC-induced metabolic dysregulation across multiple tissues. The identified metabolic signatures suggest that ALY688 exerts cardioprotective effects, at least in part, through restoration of systemic metabolic homeostasis and engagement of a gut–liver–heart metabolic axis. These results provide mechanistic insight into adiponectin receptor agonism and support further exploration of ALY688 as a potential therapeutic strategy for HFrEF. Full article
(This article belongs to the Special Issue Metabolomics in Respiratory, Cardiovascular and Metabolic Disorders)
Show Figures

Figure 1

33 pages, 1431 KB  
Review
Microbiota-Driven Immune Dysregulation Along the Gut–Lung–Vascular Axis in Asthma and Atherosclerosis
by Elena-Larisa Zimbru, Răzvan-Ionuț Zimbru, Florina-Maria Bojin, Sorin Dan Chiriac, Laura Haidar, Minodora Andor, Gabriela Tănasie, Carmen Tatu, Marius Georgescu, Cristina Uța, Camelia-Felicia Bănărescu, Sabine Groza and Carmen Panaitescu
Biomedicines 2026, 14(1), 73; https://doi.org/10.3390/biomedicines14010073 - 29 Dec 2025
Viewed by 398
Abstract
Background: Asthma and atherosclerosis frequently coexist in clinical populations and share convergent immunometabolic pathways amplified by gut microbial dysbiosis. We propose the gut–lung–vascular axis as a unifying mechanistic framework connecting epithelial and endothelial inflammation providing a foundation for understanding shared inflammatory mechanisms beyond [...] Read more.
Background: Asthma and atherosclerosis frequently coexist in clinical populations and share convergent immunometabolic pathways amplified by gut microbial dysbiosis. We propose the gut–lung–vascular axis as a unifying mechanistic framework connecting epithelial and endothelial inflammation providing a foundation for understanding shared inflammatory mechanisms beyond tissue-specific disease boundaries. Methods: A targeted narrative review systematically appraised clinical, experimental and multi-omics studies published over the last five years to delineate microbiota-driven pathways relevant to asthma and atherosclerosis. Particular emphasis was placed on specific microbial taxa, metabolite profiles and immunometabolic networks that connect gut dysbiosis with respiratory and cardiovascular dysfunction. Results: Across human and experimental cohorts, dysbiosis marked by depletion of short-chain fatty acids (SCFAs) producing taxa (Faecalibacterium, Roseburia, Bacteroides) and enrichment of pathobionts (Proteobacteria, Haemophilus, Moraxella, Streptococcus) promotes epithelial and endothelial barrier dysfunction, amplifying Th2/Th17-skewed inflammation and endothelial injury. Key metabolites, including SCFAs, trimethylamine N-oxide (TMAO), secondary bile acids (BA), indole/tryptophan derivatives and lipopolysaccharides (LPS), serve as molecular connectors linking gut, airway and vascular inflammation. Microbial signatures and metabolomic patterns hold emerging diagnostic and therapeutic potential, and several drug classes (e.g., statins, corticosteroids, proton-pump inhibitors (PPIs)) further modulate host–microbiota interactions. Conclusions: Shared microbial taxa and metabolite signatures in asthma and atherosclerosis support microbiota-mediated immune dysregulation along the gut–lung–vascular axis as a common pathogenic framework. Microbial and metabolite profiling may enable improved risk stratification and precise, microbiota-targeted therapies. Integrating microbiome-informed diagnostics and personalized interventions could help reduce systemic inflammation and the burden of these overlapping inflammatory diseases. Full article
Show Figures

Figure 1

29 pages, 2272 KB  
Review
The Dual Role of Gut Microbiota and Their Metabolites in Hepatocellular Carcinoma: A Context-Dependent Framework
by Shuyu Zuo, Junhui Ma, Xue Li, Zhengyang Fan, Xiao Li, Yingen Luo and Lei Su
Microorganisms 2026, 14(1), 73; https://doi.org/10.3390/microorganisms14010073 - 29 Dec 2025
Viewed by 442
Abstract
Hepatocellular carcinoma (HCC) is a global health threat, and gut microbiota play a pivotal role in its pathogenesis through the gut–liver axis. However, the literature contains divergent or opposing findings: key microbial metabolites, such as secondary bile acids and indole derivatives, exhibit potent [...] Read more.
Hepatocellular carcinoma (HCC) is a global health threat, and gut microbiota play a pivotal role in its pathogenesis through the gut–liver axis. However, the literature contains divergent or opposing findings: key microbial metabolites, such as secondary bile acids and indole derivatives, exhibit potent pro- and anti-tumorigenic activities across different studies, hindering a unified understanding of their veritable roles. To resolve this ambiguity, this review proposes a unifying “context dependency” framework. We posit that the functions of gut microbiota and their metabolites are not fixed but are dynamically determined by the host’s physiological and pathological “context,” defined here as the integrated dynamic background shaped by local metabolite concentrations, host immune status, specific receptor expression, and tumor microenvironment (TME) features. This framework is systematically substantiated through an analysis of the dichotomous effects of major microbial metabolites, including bile acids (BAs), short-chain fatty acids (SCFAs), trimethylamine N-oxide (TMAO), and indole derivatives. We further elucidate that the key “contextual factors” governing these functional outcomes include the TME, host immune status, metabolite concentration gradients, and the activation patterns of specific signaling pathways (e.g., farnesoid X receptor/takeda G protein-coupled receptor 5, aryl hydrocarbon receptor). This novel framework not only provides a theoretical foundation for integrating existing paradoxical findings but also paves the way for the future development of context-specific precision diagnostic biomarkers and targeted microbial intervention therapies for HCC. Full article
Show Figures

Figure 1

23 pages, 2214 KB  
Review
Role of Gut Microbiome in Oncogenesis and Oncotherapies
by Renuka Sri Sai Peddireddi, Sai Kiran Kuchana, Rohith Kode, Saketh Khammammettu, Aishwarya Koppanatham, Supriya Mattigiri, Harshavardhan Gobburi and Suresh K. Alahari
Cancers 2026, 18(1), 99; https://doi.org/10.3390/cancers18010099 - 29 Dec 2025
Viewed by 518
Abstract
The gut microbiome has emerged as a key regulator of human health, influencing not only metabolism and immunity but also the development and treatment of cancer. Mounting evidence suggests that microbial dysbiosis contributes to oncogenesis by driving chronic inflammation, producing genotoxic metabolites, altering [...] Read more.
The gut microbiome has emerged as a key regulator of human health, influencing not only metabolism and immunity but also the development and treatment of cancer. Mounting evidence suggests that microbial dysbiosis contributes to oncogenesis by driving chronic inflammation, producing genotoxic metabolites, altering bile acid metabolism, and disrupting epithelial barrier integrity. At the same time, the gut microbiome significantly modulates the host response to oncotherapies including chemotherapy, radiotherapy, and especially immunotherapy, where microbial diversity and specific taxa determine treatment efficacy and toxicity. This review synthesizes current evidence on the role of the gut microbiome in both oncogenesis and oncotherapies, focusing on thirteen cancers with the strongest and most clinically relevant microbiome associations, colorectal cancer, gastric cancer, hepatocellular carcinoma, gallbladder cancer, esophageal cancer, pancreatic cancer, oral squamous cell carcinoma, cervical cancer, prostate cancer, breast cancer, lung cancer, brain cancer, and melanoma. These cancers were selected based on robust mechanistic data linking microbial alterations to tumor initiation, progression, and therapy modulation, as well as their global health burden and translational potential. In addition, we have provided mechanistic insights or clinical correlations between the microbiome and cancer outcomes. Across cancers, common microbial mechanisms included pro-inflammatory signaling (e.g., NF-κB and STAT3 pathways), DNA damage from bacterial toxins (e.g., colibactin, nitrosating species), and metabolite-driven tumor promotion (e.g., secondary bile acids, trimethylamine N-oxide). Conversely, beneficial commensals such as Faecalibacterium prausnitzii and Akkermansia muciniphila supported antitumor immunity and improved responses to immune checkpoint inhibitors. In conclusion, the gut microbiome functions as both a driver of malignancy and a modifiable determinant of therapeutic success. Integrating microbiome profiling and modulation strategies such as dietary interventions, probiotics, and fecal microbiota transplantation into oncology practice may pave the way for personalized and more effective cancer care. Full article
Show Figures

Figure 1

23 pages, 1990 KB  
Article
CXCL1, RANTES, IFN-γ, and TMAO as Differential Biomarkers Associated with Cognitive Change After an Anti-Inflammatory Diet in Children with ASD and Neurotypical Peers
by Luisa Fernanda Méndez-Ramírez, Miguel Andrés Meñaca-Puentes, Luisa Matilde Salamanca-Duque, Marysol Valencia-Buitrago, Andrés Felipe Ruiz-Pulecio, Carlos Alberto Ruiz-Villa, Diana María Trejos-Gallego, Juan Carlos Carmona-Hernández, Sandra Bibiana Campuzano-Castro, Marcela Orjuela-Rodríguez, Vanessa Martínez-Díaz, Jessica Triviño-Valencia and Carlos Andrés Naranjo-Galvis
Med. Sci. 2026, 14(1), 11; https://doi.org/10.3390/medsci14010011 - 26 Dec 2025
Viewed by 240
Abstract
Background/Objective: Neuroimmune and metabolic dysregulation have been increasingly implicated in the cognitive heterogeneity of autism spectrum disorder (ASD). However, it remains unclear whether anti-inflammatory diets engage distinct biological and cognitive pathways in autistic and neurotypical children. This study examined whether a 12-week [...] Read more.
Background/Objective: Neuroimmune and metabolic dysregulation have been increasingly implicated in the cognitive heterogeneity of autism spectrum disorder (ASD). However, it remains unclear whether anti-inflammatory diets engage distinct biological and cognitive pathways in autistic and neurotypical children. This study examined whether a 12-week anti-inflammatory dietary protocol produces group-specific neuroimmune–metabolic signatures and cognitive responses in autistic children, neurotypical children receiving the same diet, and untreated neurotypical controls. Methods: Twenty-two children (11 with ASD, six a on neurotypical diet [NT-diet], and five neurotypical controls [NT-control]) completed pre–post assessments of plasma IFN-γ, CXCL1, RANTES (CCL5), trimethylamine-N-oxide (TMAO), and an extensive ENI-2/WISC-IV neuropsychological battery. Linear mixed-effects models were used to test the Time × Group effects on biomarkers and cognitive domains, adjusting for age, sex, and baseline TMAO. Bayesian estimation quantified individual changes (posterior means, 95% credible intervals, and posterior probabilities). Immune–cognitive coupling was explored using Δ–Δ correlation matrices, network metrics (node strength, degree centrality), exploratory mediation models, and responder (≥0.5 SD domain improvement) versus non-responder analyses. Results: In ASD, the diet induced robust reductions in IFN-γ, RANTES, CXCL1, and TMAO, with decisive Bayesian evidence for IFN-γ and RANTES suppression (posterior P(δ < 0) > 0.99). These shifts were selectively associated with gains in verbal learning, semantic fluency, verbal reasoning, attention, and visuoconstructive abilities, whereas working memory and executive flexibility changes were heterogeneous, revealing executive vulnerability in individuals with smaller TMAO reductions. NT-diet children showed modest but consistent improvements in visuospatial processing, attention, and processing speed, with minimal biomarker changes; NT controls remained biologically and cognitively stable. Network analyses in ASD revealed a dense chemokine-anchored architecture with CXCL1 and RANTES as central hubs linking biomarker reductions to improvements in fluency, memory, attention, and executive flexibility. ΔTMAO predicted changes in executive flexibility only in ASD (explaining >50% of the variance), functioning as a metabolic node of executive susceptibility. Responders displayed larger coordinated decreases in all biomarkers and broader cognitive gains compared to non-responders. Conclusions: A structured anti-inflammatory diet elicits an ASD-specific, coordinated neuroimmune–metabolic response in which suppression of CXCL1 and RANTES and modulation of TMAO are tightly coupled with selective improvements in verbal, attentional, and executive domains. Neurotypical children exhibit modest metabolism-linked cognitive benefits and minimal immune modulation. These findings support a precision-nutrition framework in ASD, emphasizing baseline immunometabolic profiling and network-level biomarkers (CXCL1, RANTES, TMAO) to stratify responders and design combinatorial interventions targeting neuroimmune–metabolic pathways. Full article
(This article belongs to the Section Translational Medicine)
Show Figures

Figure 1

18 pages, 1596 KB  
Review
Interplay Among Gut Microbiota-Derived TMAO, Autonomic Nervous System Dysfunction, and Heart Failure Progression
by Laura Calvillo, Emilio Vanoli, Fulvio Ferrara and Eugenio Caradonna
Int. J. Mol. Sci. 2026, 27(1), 203; https://doi.org/10.3390/ijms27010203 - 24 Dec 2025
Viewed by 390
Abstract
The gut microbiota is crucial for metabolic homeostasis and cardiovascular health. Dysbiosis triggers a gut–brain–heart axis dysfunction: vagal signaling promotes neuroinflammation and cerebral damage, which in turn impairs cardiac function. This bidirectional cycle is further exacerbated by reduced cerebral perfusion. Trimethylamine-N-oxide (TMAO), a [...] Read more.
The gut microbiota is crucial for metabolic homeostasis and cardiovascular health. Dysbiosis triggers a gut–brain–heart axis dysfunction: vagal signaling promotes neuroinflammation and cerebral damage, which in turn impairs cardiac function. This bidirectional cycle is further exacerbated by reduced cerebral perfusion. Trimethylamine-N-oxide (TMAO), a metabolite of dietary choline and L-carnitine, acts as a primary mediator in this network. Elevated TMAO levels—resulting from bacterial conversion and hepatic oxidation—are linked to atherosclerosis and heart failure. Mechanistically, TMAO activates the NLRP3 inflammasome, inhibits the SIRT3-SOD2 pathway, and promotes platelet hyperreactivity. Furthermore, it modulates the autonomic nervous system, enhancing sympathetic activity and cardiac arrhythmias. Clinical evidence suggests TMAO is a potent predictor of mortality in HF. While current HF therapies focus on end-organ response (beta-blockers) or humoral pathways (ACE inhibitors), directly targeting the microbiota and TMAO offers a novel therapeutic frontier. Integrating TMAO assessment into risk models and utilizing advanced in vitro gut–brain models will be essential for developing personalized, groundbreaking cardiovascular interventions. Within this framework, the main aim of the present review is to describe how cardiac autonomic control can be directly modulated by the microbiota and its byproducts like TMAO. This latter is a leading target candidate for novel HF prevention and therapy interventions. Full article
Show Figures

Figure 1

14 pages, 577 KB  
Article
Serum Trimethylamine N-Oxide Levels Are Associated with Peripheral Artery Disease in Patients with Type 2 Diabetes Mellitus
by Cing-Yu Liang, Jer-Chuan Li, Chin-Hung Liu, Du-An Wu and Bang-Gee Hsu
Medicina 2025, 61(12), 2243; https://doi.org/10.3390/medicina61122243 - 18 Dec 2025
Viewed by 372
Abstract
Background and Objectives: Peripheral arterial disease (PAD), frequently observed in individuals with type 2 diabetes mellitus (T2DM), is associated with diminished life quality, increased cardiovascular risk, and higher mortality rates. Similarly, trimethylamine N-oxide (TMAO), a uremic toxin produced by gut microbiota, has [...] Read more.
Background and Objectives: Peripheral arterial disease (PAD), frequently observed in individuals with type 2 diabetes mellitus (T2DM), is associated with diminished life quality, increased cardiovascular risk, and higher mortality rates. Similarly, trimethylamine N-oxide (TMAO), a uremic toxin produced by gut microbiota, has been linked to hypertension, cardiovascular disease, and increased overall mortality. In this study, we aimed to investigate whether serum TMAO levels are related to PAD in T2DM cases. Materials and Methods: In this cross-sectional investigation performed at one medical center, 120 patients with type 2 diabetes mellitus (T2DM) were included. High-performance liquid chromatography–mass spectrometry and an automated oscillometric device were used to measure serum TMAO levels and ankle–brachial index (ABI) values, respectively. Individuals exhibiting an ABI of less than 0.9 were classified as belonging to the low-ABI group. Results: Of the 120 participants, 23 (19.2%) had low ABI. Compared with the normal-ABI group, the low-ABI group was older (p = 0.017) and exhibited higher levels of urine albumin-to-creatinine ratio (UACR, p < 0.001), C-reactive protein (CRP, p < 0.001), and TMAO (p < 0.001). After adjusting for age, UACR, and CRP, multivariable logistic regression analysis identified serum TMAO concentration as an independent predictor of PAD in T2DM patients (odds ratio [OR]: 1.051; 95% confidence interval [CI]: 1.017–1.086; p = 0.003). In Spearman’s rank correlation analyses, log-transformed left ABI (log-left ABI, p = 0.017) and log-right ABI (p = 0.001) negatively correlated with log-TMAO. In patients with T2DM, the predictive performance of serum TMAO levels for PAD yielded an area under the receiver operating characteristic (ROC) curve of 0.812 (95% CI: 0.701–0.923; p < 0.001). Conclusions: Among individuals with T2DM, higher serum TMAO levels were associated with lower left and right ABI values and an increased likelihood of PAD. Full article
Show Figures

Figure 1

14 pages, 873 KB  
Article
Correlations Between Trimethylamine-N-Oxide, Megalin, Lysine and Markers of Tubular Damage in Chronic Kidney Disease
by Stefania Kapetanaki, Samira Salihovic, Ashok Kumar Kumawat, Ziad A. Massy, Katarina Persson, Peter Barany, Peter Stenvinkel, Marie Evans and Isak Demirel
Toxins 2025, 17(12), 592; https://doi.org/10.3390/toxins17120592 - 11 Dec 2025
Viewed by 494
Abstract
Trimethylamine-N-oxide (TMAO), a gut microbiota-derived dietary metabolite, is linked to progression of chronic kidney disease (CKD). Megalin, a renal proximal tubule receptor crucial for albumin reabsorption, also plays a role in CKD. However, the relationship between them is not well explored. The aim [...] Read more.
Trimethylamine-N-oxide (TMAO), a gut microbiota-derived dietary metabolite, is linked to progression of chronic kidney disease (CKD). Megalin, a renal proximal tubule receptor crucial for albumin reabsorption, also plays a role in CKD. However, the relationship between them is not well explored. The aim of this study was to investigate if there are any correlations between the levels of TMAO, megalin, lysine and markers of tubular damage in CKD. Urinary metabolites (TMAO, choline, L-carnitine, betaine, lysine) and tubular markers (megalin, albumin, EGF, MCP-1) were quantified by LC-MS/MS and ELISA. Associations were evaluated using analysis of covariance (ANCOVA) adjusted for age and diabetes, with false discovery rate correction. Compared with controls, CKD patients showed higher urinary choline (FDR < 0.001), betaine (FDR = 0.007), lysine (FDR = 0.005), and soluble megalin (FDR < 0.001) but lower EGF and EGF/MCP-1 ratio (both FDR < 0.001). Correlation analyses revealed that serum TMAO was positively associated with soluble megalin and negatively with EGF/MCP-1 ratio. Choline, L-carnitine, and betaine were positively correlated with megalin. This cross-sectional study identifies associations between urinary metabolites, megalin, and tubular injury markers in advanced CKD. Although causality cannot be inferred, the results point to a potential metabolic–tubular link that should be explored in future longitudinal and mechanistic studies. Full article
Show Figures

Figure 1

20 pages, 1470 KB  
Article
Osmolytes vs. Anabolic Reserves: Contrasting Gonadal Metabolomes in Two Sympatric Mediterranean Sea Urchins
by Estela Carbonell-Garzón, Ricardo Ibanco-Cañete, Pablo Sanchez-Jerez and Frutos C. Marhuenda Egea
Metabolites 2025, 15(12), 787; https://doi.org/10.3390/metabo15120787 - 10 Dec 2025
Viewed by 352
Abstract
Background an Objectives: The Mediterranean sea urchins Paracentrotus lividus and Arbacia lixula co-occur on shallow rocky reefs but display contrasting ecological and physiological traits. We compared their gonadal metabolomes to identify species-specific metabolic strategies. Methods: High-resolution magic angle spinning nuclear magnetic resonance (HR-MAS [...] Read more.
Background an Objectives: The Mediterranean sea urchins Paracentrotus lividus and Arbacia lixula co-occur on shallow rocky reefs but display contrasting ecological and physiological traits. We compared their gonadal metabolomes to identify species-specific metabolic strategies. Methods: High-resolution magic angle spinning nuclear magnetic resonance (HR-MAS NMR) spectroscopy to intact gonadal tissues, combining multivariate chemometric modelling with targeted integration, boxplot-based univariate analysis and pathway analysis. Results:A. lixula showed an osmolyte- and redox-oriented phenotype with elevated betaine, taurine, sarcosine, trimethylamine (TMA), trimethylamine N-oxide (TMAO), carnitine, creatine, malonate, methylmalonate, uridine and xanthine. In contrast, P. lividus exhibited an amino-acid-enriched anabolic profile dominated by lysine, glycine and glutamine, together with higher levels of formaldehyde, methanol and 3-carboxypropyl-trimethylammonium. Pathway analysis indicated that A. lixula metabolites mapped onto glycine/serine–threonine metabolism and the folate-linked one-carbon pool, whereas P. lividus metabolites were enriched in glyoxylate/dicarboxylate, nitrogen and amino-acid pathways. These contrasting osmolyte–C1 versus nitrogen–amino-acid strategies are compatible with species-specific host–microbiota metabolic interactions inferred from published microbiome data. Conclusions: Overall, our results support a framework in which A. lixula adopts a resilience-oriented osmolyte strategy and P. lividus an efficiency-oriented anabolic strategy, highlighting HR-MAS NMR metabolomics as a powerful approach to investigate adaptive biochemical diversity in marine invertebrates. Full article
(This article belongs to the Collection Advances in Metabolomics)
Show Figures

Figure 1

Back to TopTop