Gut Microbes

Gut Microbes

Published by Taylor & Francis

Online ISSN: 1949-0984

·

Print ISSN: 1949-0976

Journal websiteAuthor guidelines

Top-read articles

66 reads in the past 30 days

Beneficial role of lactobacillus in intestines, generating beneficial compounds responsible for anti-pathogenic activities and regulation of immune cells, acting as a regulator of tight junction.
Illustration of probiotic to target host. (a) Sources of probiotic to target area (b) Role of probiotic between healthy and disease person, relating to many other disorders, here we showed the main one (c) Special example of Lactobacillus, mechanism of action in intestine.
Ideal properties for any bacteria to become probiotic.
Lactobacillus probiotics have a wide variety of positive impacts on the human body, which contribute to some of the health benefits they offer. Its primary purpose is to enhance the functioning of the body as well as the metabolic processes of the body. Most of the portion has been highlighted here.
Probiotic significance of Lactobacillus strains: a comprehensive review on health impacts, research gaps, and future prospects

November 2024

·

302 Reads

·

18 Citations

·

·

·

[...]

·

Download

57 reads in the past 30 days

Integrative metagenomics and metabolomics reveal age-associated gut microbiota and metabolite alterations in a hamster model of COVID-19

April 2025

·

57 Reads

Aims and scope


Gut Microbes publishes research on intestinal microbiota, and its role in gastrointestinal, liver, cardiac disease, cancer, and irritable and inflammatory bowel conditions.

  • The intestinal microbiota plays a pivotal role in human physiology.
  • Characterizing its structure and function has implications for health and disease, impacting nutrition and obesity, brain function, allergic responses, immunity, inflammatory bowel disease, irritable bowel syndrome, cancer development, cardiac disease, liver disease, and others.
  • Gut Microbes provides a platform for presenting and discussing cutting-edge research on all aspects of microorganisms populating the intestine.
  • The journal focuses in particular on mechanistic and cause and effect studies.
  • Gut Microbes brings together a multidisciplinary community of scientists working in the areas of: Profiling the intestinal microbiota, Gastrointestinal disease: mechanisms, host defense, diagnosis, epidemiology, Host-pathogen interactions including enteric disease and bacterial pathogenesis, Quorum sensing and toxicity, Probiotics and prebiotics, Novel treatments and clinical trials, Role of intestinal microbiota and…

For a full list of the subject areas this journal covers, please visit the journal website.

Recent articles


Delivery mode, birth order, and sex impact neonatal microbial colonization
  • Article

April 2025

·

1 Read

Katherine M Kennedy

·

Andreas Plagemann

·

Julia Sommer

·

[...]

·

Deborah M Sloboda


E. coli genetically modified for purine nucleobase release promotes butyrate generation and colonic wound healing during DSS insult
  • Article
  • Full-text available

April 2025

·

4 Reads

The gut microbiota transforms energy stored as undigestible carbohydrates into a remarkable number of metabolites that fuel intestinal bacterial communities and the host tissue. Colonic epithelial cells at the microbiota–host interface depend upon such microbiota-derived metabolites (MDMs) to satisfy their energy requisite. Microbial dysbiosis eliciting MDM loss contributes to barrier dysfunction and mucosal disease. Recent work has identified a role for microbiota-sourced purines (MSPs), notably hypoxanthine, as an MDM salvaged by the colonic epithelium for nucleotide biogenesis and energy balance. Here, we investigated the role of MSPs in mice during disease-modeled colonic energetic stress using a strain of E. coli genetically modified for enhanced purine nucleobase release (E. coli Mutant). E. coli Mutant colonization protected against DSS-induced tissue damage and permeability while promoting proliferation for wound healing. Metabolite and metagenomic analyses suggested a colonic butyrate-purine nucleobase metabolic axis, wherein the E. coli Mutant provided purine substrate for Clostridia butyrate production and host purine salvage, altogether supplying the host substrate for efficient nucleotide biogenesis and energy balance.



Urease in acetogenic Lachnospiraceae drives urea carbon salvage in SCFA pools

April 2025

·

4 Reads

The gut microbiota produces short-chain fatty acids (SCFA) and acidifies the proximal colon which inhibits enteric pathogens. However, for many microbiota constituents, how they themselves resist these stresses is unknown. The anaerobic Lachnospiraceae family, which includes the acetogenic genus Blautia, produce SCFA, are genomically diverse, and vary in their capacity to acidify culture media. Here, we investigated how Lachnospiraceae tolerate pH stress and found that subunits of urease were associated with acidification in a random forest model. Urease cleaves urea into ammonia and carbon dioxide, however the role of urease in the physiology of Lachnospiraceae is unknown. We demonstrate that urease-encoding Blautia show urea-dependent changes in SCFA production, acidification, growth, and, strikingly, urease encoding Blautia directly incorporate the carbon from urea into SCFAs. In contrast, ureolytic Klebsiella pneumoniae or Proteus mirabilis do not show the same urea-dependency or carbon salvage. In agreement, the combination of urease and acetogenesis functions is rare in gut taxa. We find that Lachnospiraceae urease and acetogenesis genes can be co-expressed in healthy individuals and colonization of mice with a ureolytic Blautia reduces urea availability in colon contents demonstrating Blautia urease activity in vivo. In human and mouse microbial communities, the acetogenic recycling of urea carbon into acetate by Blautia leads to the incorporation of urea carbon into butyrate indicating carbon salvage into broader metabolite pools. Altogether, this shows that urea plays a central role in the physiology of health-associated Lachnospiraceae which use urea in a distinct manner that is different from that of ureolytic pathogens.


Genetic variation in gut microbe as a key regulator of host social behavior in C. elegans

April 2025

·

2 Reads

Gut microbiota have been shown to influence the social behaviors of their hosts, while variations in host genetics can affect the composition of the microbiome. Nonetheless, the degree to which genetic variations in microbial populations impact host behavior, as well as any potential transgenerational effects, remains inadequately understood. Utilizing C. elegans as a model organism, we identified 77 strains of E. coli from a total of 3,983 mutants that significantly enhanced aggregation behavior through various neurobehavioral pathways. This discovery underscores a collaborative regulatory mechanism between microbial genetics and host behavior. Notably, we observed that some mutant bacteria might affect social behavior via the mitochondrial pathway. Additionally, the modulation of social behavior has been identified as a heritable trait in offspring. Our results provide a novel perspective on the regulatory role of microbial genetic variation in host behavior, which may have significant implications for human studies and the development of genetically engineered probiotics aimed at enhancing well-being across generations.


(a) Schema for randomized, double-blinded, placebo-controlled trial of microbiota transplant therapy (MTT) for participants with mild-to-moderate ulcerative colitis. Participants were administered placebo capsules or encapsulated MTT from a healthy donor daily for 8 weeks. Stool samples for metagenomic sequencing were collected from participants at baseline (week 0), and weeks 4, 8, and 12. (b) A visual depiction of pre-treatment (yellow), donor (blue), and post-treatment (green) strains as a Venn Diagram. While the engrafted donor strains constitute the focus of our manuscript, further information on MTT impacts can also be found in determining the likelihood of ambiguous strains being engrafted or persistent. (c) A visual depiction of the MAGEnTa pipeline. We begin with using metaSpades and Bowtie 2 to construct metagenome assembled genomes (MAGs) databases from the pre-mtt and donor samples. We then align the post-MTT sample with these databases to determine which reads match the donor and the pre-mtt (baseline) samples. Ambiguous reads matching both databases are evaluated by a Bayesian estimation to assign the likely source. Read source outputs are combined into proportions, assigning each read to engrafted, persistent, or novel/unknown (unmapped). This figure was created in BioRender.com. DB (database), MAGs (metagenome assembled genomes), MTT (microbiota transplant therapy).
(a) Creation of 15 simulated datasets totaling 165 samples. For each of 15 donor and pre-mtt pairs in a CDI study, we obtain 11 samples comprised of different percentages of the donor and pre-microbiota transplant therapy (MTT) material. All samples are an even depth of 10 million reads. (b) From the 50% donor and 50% pre-mtt simulated samples, we also simulated the inclusion of an external, “contaminate” source. For this simulation, our external source is another healthy donor unrelated to the original donor and pre-MTT pair. This simulation seeks to represent other microbes introduced to the patient by external means such as food or environment. (c) Engraftment resulting from 15 simulations, displaying simulated (x-axis) vs. observed (y-axis), separated and colored by alignment identity. The expected outcome would be an x=y line as shown in gray, which the observed results closely follow. (d) The recall error of the algorithm per identity with the 50% donor and 50% pre samples, showing the false negative rate (type II error) per simulation, grouped by identity. Less stringent identity has a slightly lower type II error rate. (e) The accuracy error when 20% of the simulated 50% donor sample comes from a contaminate source in the form of an unrelated donor sample. The more stringent the identity, the fewer false positives (type I error).
(a) Boxplot showing the percent of donor engraftment per group at each week. The greater spread in the intervention group is likely due to the variances in success of engraftment following the encapsulated MTT course. (b) Agreement between methods per sample. Whole-genome sequencing (WGS) MAGEnTa pipeline results are on the x-axis and 16S rRNA gene SourceTracker results on the y-axis. The intervention samples (blue) have high agreement (Pearson correlation, p < 0.001), and placebo samples have no agreement (Pearson correlation, p = 0.45); however, poor placebo sample agreement may be poor due to the low engraftment observed. (c) Percentage of donor engraftment for each patient and its matched placebo donor. Donor engraftment in the microbiota transplant therapy (MTT) group is significantly higher at weeks eight and 12 compared to placebo. Three donors were used and participants received donor material from only one donor. MTT is shown in blue and placebo in pink. Some participants are missing samples (predominantly from week 4), but the intervention of daily oral MTT began after collecting the baseline sample at week 0 and ran for the first 8 weeks, collecting samples every 4 weeks. (d) MetaBinner results from binning engrafted metagenome assembled genomes (MAGs) shown as a percentage of engrafted reads for weeks 4, 8, and 12 samples receiving material from Donor A. The top 10 MAG bins are distinctly colored to show relative abundance of reads aligning with these bins across all engrafted reads.
Metagenomic source tracking after microbiota transplant therapy

April 2025

·

28 Reads

Reliable engraftment assessment of donor microbial communities and individual strains is an essential component of characterizing the pharmacokinetics of microbiota transplant therapies (MTTs). Recent methods for measuring donor engraftment use whole-genome sequencing and reference databases or metagenome-assembled genomes (MAGs) to track individual bacterial strains but lack the ability to disambiguate DNA that matches both donor and patient microbiota. Here, we describe a new, cost-efficient analytic pipeline, MAGEnTa, which compares post-MTT samples to a database comprised MAGs derived directly from donor and pre-treatment metagenomic data, without relying on an external database. The pipeline uses Bayesian statistics to determine the likely sources of ambiguous reads that align with both the donor and pre-treatment samples. MAGEnTa recovers engrafted strains with minimal type II error in a simulated dataset and is robust to shallow sequencing depths in a downsampled dataset. Applying MAGEnTa to a dataset from a recent MTT clinical trial for ulcerative colitis, we found the results to be consistent with 16S rRNA gene SourceTracker analysis but with added MAG-level specificity. MAGEnTa is a powerful tool to study community and strain engraftment dynamics in the development of MTT-based treatments that can be integrated into frameworks for functional and taxonomic analysis.


Time-restricted feeding protects against septic liver injury by reshaping gut microbiota and metabolite 3-hydroxybutyrate

April 2025

·

8 Reads

Liver injury is an independent risk factor for multiple organ dysfunction and high mortality in patients with sepsis. However, the pathological mechanisms and therapeutic strategies for sepsis-associated liver injury have not been fully elucidated. Time-restricted feeding (TRF) is a promising dietary regime, but its role in septic liver injury remains unknown. Using 16S rRNA gene sequencing, Q200 targeted metabolomics, transcriptomics, germ-free mice, Hmgcs2/Lpin1 gene knockout mice, and Aml12 cells experiments, we revealed that TRF can mitigate septic liver injury by modulating the gut microbiota, particularly by increasing Lactobacillus murinus (L. murinus) abundance, which was significantly reduced in septic mice. Further study revealed that live L. murinus could markedly elevate serum levels of metabolite 3-hydroxybutyrate (3-HB) and alleviate sepsis-related injury, while the knockout of the key enzyme for 3-HB synthesis (3-hydroxy-3-methylglutaryl-CoA synthase 2, Hmgcs2) in the liver negated this protective effect. Additionally, serum 3-HB levels were significantly positively correlated with L. murinus abundance and negatively correlated with liver injury indicators in septic patients, demonstrating a strong predictive value for septic liver injury (AUC = 0.8429). Mechanistically, 3-HB significantly inhibited hepatocyte ferroptosis by activating the PI3K/AKT/mTOR/LPIN1 pathway, reducing ACSL4, MDA, LPO, and Fe²⁺ levels. This study demonstrates that TRF reduces septic liver injury by modulating gut microbiota to increase L. murinus, which elevates 3-HB to activate PI3K/AKT/mTOR/LPIN1 and inhibit hepatocyte ferroptosis. Overall, this study elucidates the protective mechanism of TRF against septic liver injury and identifies 3-HB as a potential therapeutic target and predictive biomarker, thereby providing new insights into the clinical management and diagnosis of septic liver injury.


Fusobacterium nucleatum exacerbates colitis via STAT3 activation induced by Acetyl-CoA accumulation

April 2025

·

3 Reads

Fusobacterium nucleatum (F. nucleatum) has emerged as a potential contributor to ulcerative colitis (UC) pathogenesis, although the specific mechanisms remain incompletely understood. This study demonstrates that F. nucleatum promotes colitis by disrupting intestinal barrier integrity, inducing apoptosis in epithelial cells, and modulating inflammatory pathways. Furthermore, we demonstrate that F. nucleatum promotes STAT3 acetylation at K685, followed by phosphorylation at Y705, thereby enhancing its transcriptional activity and exacerbating colitis severity. Additionally, F. nucleatum-mediated upregulation of acetyl-CoA levels is responsible for STAT3 acetylation, linking metabolic processes to UC pathophysiology. Pharmacological inhibition of acetyl-CoA production effectively mitigates F. nucleatum-induced colitis in experimental models, suggesting potential therapeutic strategies targeting these pathways. These findings unveil a novel regulatory pathway in F. nucleatum-associated UC progression and offer new insights for future UC prevention and treatment.


Environmental factors influence epigenetic modifications through gut microbiota and its metabolites in IBD.
Mechanisms of epigenetic modifications by SCFAs in IBD.
Mechanism of folate-induced DNAm in IBD.
Polyphenols influence ncRNA in IBD.
Impact of bile acids on histone modifications in IBD.
Gut microbiota and epigenetic inheritance: implications for the development of IBD

Inflammatory bowel disease (IBD), including Crohn’s disease and ulcerative colitis, is considered significant global health concerns worldwide. Many studies have demonstrated that environmental and dietary factors influence the gut microbiota, which in turn orchestrates the host immune responses. These interactions are also involved in complex metabolic processes that contribute to the pathogenesis of IBD. Furthermore, recent studies in genomics and metabolomics have unveiled the intricate relationship between microbial influencers and host epigenetics. The dynamics of gut microbiota and its metabolites intricately align with DNA methylation, histone methylation, lactylation, glycosylation, and non-coding RNAs, which are key players in epigenetics. Here, we summarize and discuss the complex interplay among gut microbiota, epigenetics, and environmental and dietary factors, and their impact on the pathogenesis of IBD. Furthermore, we highlight the importance of multi-omics technologies in dissecting the host-microbe interactions in IBD, potentially offering a framework for developing effective treatment strategies.


The GM and joint health are interconnected, known as the “gut-joint axis”. Under the gut-joint axis model, extracellular vesicles secreted by the GM may play a significant role in this axis. BEVs are not only small membrane-enclosed structures rich in bacterial-derived molecules but also natural nanoparticles. BEVs carry various oa-related cargoes, including membrane-associated cargo, cytoplasmic cargo, and genetic cargo. Natural BEVs and tailored engineered BEVs hold significant importance and promise in the treatment of OA. In summary, BEVs represent a new bridge in the gut-joint axis. The figure was created with https://app.bioreunder.com/.
The biogenesis, classification and composition of bacterial extracellular vesicles. The G⁻ bacteria can produce OMVs through blebbing of the outer membrane and can also generate OIMVs and EOMVs through explosive cell lysis. G⁺ bacteria produce CMVs through the process of bubbling cell death. BEVs contain various contents, including nucleic acids (DNA, RNA), proteins, and metabolites. IM: inner membrane, OM: outer membrane, PG: peptidoglycan, LPS: Lipopolysaccharide. The figure was created with https://app.Biore under.com/.
of the role of bacterial extracellular vesicles in the gut-joint axis. The type and state of bacteria determine the functionality of BEVs. BEVs secreted by a dysregulated microbiota, or pathogenic bacteria may be a cause of OA, while BEVs secreted by probiotics could also be a solution for OA. In OA, BEVs can interact directly or indirectly with intestinal epithelial cells and immune cells, participating in the maintenance of the intestinal barrier and the regulation of anti-inflammatory responses. ZO-1: zonula occludens-1, RIPK: receptor-interacting protein kinase, tnf-α: tumor necrosis factor alpha, TJs: tight junction proteins, MCP-1: monocyte chemoattractant protein-1, YB-1: Y-box binding protein 1. The figure was created with https://app.bioreunder.com/.
The engineering approaches for isolating and modifying BEVs. (a) Schematic diagram of the separation and purification procedures for several common EVs. Ultracentrifugation utilizes extremely high rotational speeds to generate gravitational forces that precipitate vesicles out of solution. Density gradient centrifugation involves creating a density gradient within the centrifuge tube, allowing vesicles of different densities to settle at distinct layers during centrifugation. Ultrafiltration uses membrane filters with specific molecular weight cut-offs. Pressure is applied to force the liquid through the membrane, retaining and concentrating vesicles larger than the pore size. Affinity isolation employs specific antibodies or other ligands that bind to surface markers on vesicles, enabling selective capture of target vesicles. Size-exclusion chromatography separates vesicles based on their size in a gel column, where larger molecules are eluted first, followed by smaller particles, suitable for separating vesicles irrespective of their density. Precipitation involves using chemical reagents such as polyethylene glycol (PEG) to alter solution conditions, causing vesicles to aggregate and precipitate. (b) A separation method applicable to the vast majority of bacteria, divided into three steps: (1) removal of bacteria and debris (2) removal of non-bev proteins and concentration (3) isolation and purification. (c) Engineering modification methods for BEVs, mainly divided into pre-isolation modification and post-isolation modification. That is, one involves the modification of donor bacteria responsible for BEV production, and the other involves direct surface modification of BEVs. The figure was created with https://app.bioreunder.com/.
An overview of the application of BEV-based OA diagnosis and treatment. BEVs can serve as laboratory indicators for the early diagnosis of OA patients. Analysis of changes in the secretion and transport of BEVs can reflect the state of the GM and its contribution to disease. BEVs can offer new therapeutic strategies as treatment targets. This includes inhibiting BEVs produced as a result of dysbiosis, supplementing with BEVs derived from probiotics, and constructing novel biocompatible materials combined with EVs to directly target joints, thereby promoting cartilage repair and reducing inflammation. BEVs can enhance existing treatment methods by achieving targeted drug delivery through techniques such as genetic engineering and electroporation. https://app.bioreunder.com/
Bacterial extracellular vesicles in osteoarthritis: a new bridge of the gut-joint axis

Osteoarthritis (OA) is a degenerative joint disease primarily characterized by cartilage degeneration. Increasing evidence suggests that there is an interplay between the gut microbiota (GM) and joint health, known as the gut-joint axis. In recent years, with the advancement of bacterial extracellular vesicles (BEVs) research, its role in the gut-joint axis has attracted increasing attention. BEVs are phospholipid bilayer nanocarriers with sizes ranging from 20 to 400 nm, which can deliver various bioactive molecules to modulate the intestinal and joint microenvironments. Due to nanoscale structure, low toxicity, high drug loading capacity, good biocompatibility, easy modification, and industrialization, BEVs are promising for OA treatment. Here, we reviewed the research status of BEVs including biogenies, classification, structures, and composition, and summarized the cargo within BEVs that is associated with OA. The application of natural BEVs in the treatment of OA is the core, and the separation, purification, and modification strategies for engineered BEVs are summarized. Finally, we provide an overview and prospects of the role of BEVs in the clinical diagnosis and treatment of OA. We hope that a comprehensive understanding of BEVs will provide new solutions for OA treatment.


Overview of the gut–brain axis. Gut microbes release neurotransmitters and QSMs that travel through the vagus nerve and influence the CNS. In the event of gut dysbiosis, neurodegenerative disorders may arise as a result of the stress response, immunological response, and neuroinflammation. Created with BioRender.com.
Cross-kingdom signaling in the GBA. The gut microbiota releases several neuromodulators, such as neurotransmitters, oligopeptides, and other metabolites, interacting with epithelial and immune cells. Specialized cells lining the intestinal epithelium maintain intestinal homeostasis, form a mucus barrier, and facilitate the passage of material through the intestines.⁴⁵ neurons also recognize neurotransmitters, which travel through the vagus nerve to the brain. Gut dysbiosis resulting from imbalanced gut microbial populations leads to immunological effects and neuroinflammation, possibly resulting in neurodegenerative and psychiatric disorders. Created with BioRender.com.
Dynamic alterations in the GBM. Shifts in different microbiota populations resulting from gut dysbiosis can enhance present risk factors that increase the likelihood of developing MCI/AD pathology. Created with BioRender.com.
Neurological impact of different diets. Western diets (left) were primarily composed of processed carbohydrates and saturated fats that may impair the BBB and increase the likelihood of neuroinflammation and gut dysbiosis, leading to worsening of neurodegeneration. In contrast, the Mediterranean diet (right) primarily consists of essential fibers, nutrients, and minerals that help maintain the integrity of the BBB and restore gut dysbiosis, thereby preventing neuroinflammation that leads to neurological impairment—created with BioRender.com.
Microbial diversity and fitness in the gut–brain axis: influences on developmental risk for Alzheimer’s disease

The gut–brain axis (GBA) denotes the dynamic and bidirectional communication system that connects the gastrointestinal tract and the central nervous system (CNS). This review explored this axis, focusing on the role of microbial diversity and fitness in maintaining gastrointestinal health and preventing neurodegeneration, particularly in Alzheimer’s disease (AD). Gut dysbiosis, characterized by the imbalance in populations of beneficial and harmful bacteria, has been associated with increased systemic inflammation, neuroinflammation, and the progression of AD through pathogenic mechanisms involving amyloid deposition, tauopathy, and increased blood–brain barrier (BBB) permeability. Emerging evidence highlighted the therapeutic potential of probiotics, dietary interventions, and intermittent fasting in restoring microbial balance, reducing inflammation, and minimizing neurodegenerative risks. Probiotics and synbiotics are promising in helping improve cognitive function and metabolic health, while dietary patterns like the Mediterranean diet were linked to decreased neuroinflammation and enhanced gut–brain communication. Despite significant advancement, further research is needed to elucidate the specific microbial strains, metabolites, and mechanisms influencing brain health. Future studies employing longitudinal designs and advanced omics technologies are essential to developing targeted microbiome-based therapies for managing AD-related disorders.


Protection against experimental necrotizing enterocolitis by fecal filtrate transfer requires an active donor virome

April 2025

·

10 Reads

Necrotizing enterocolitis (NEC) remains a frequent catastrophic disease in preterm infants, and fecal filtrate transfer (FFT) has emerged as a promising prophylactic therapy. This study explored the role of virome viability for the protective effect of FFT. Using ultraviolet (UV) irradiation, we established a viral inactivation protocol and administered FFT, UV-inactivated FFT (iFFT) or sterile saline orally to preterm piglets at risk for experimental NEC. The gut pathology and barrier properties were assessed, while the microbiome was explored by 16S rRNA amplicon and metavirome sequencing. Like in prior studies, FFT reduced NEC severity and intestinal inflammation, while these effects were absent in the iFFT group. Unexpectedly, piglets receiving FFT exhibited mild side effects in the form of early-onset diarrhea. The FFT also converged the gut colonization by increased viral heterogeneity and a reduced abundance of pathobionts like Clostridium perfringens and Escherichia. In contrast, the gut microbiome of iFFT recipients diverged from both FFT and the controls. These findings highlight the clear distinction between the ability of active and inactivate viromes to modulate gut microbiota and decrease pathology. The efficacy of FFT may be driven by active bacteriophages, and loss of virome activity could have consequences for the treatment efficacy.


Establishment and characterization of inducible tet-on gGT H. pylori strains (a) Graphic representation of the tet-on gGT system. The ggt promoter was replaced by the tet inducible promoter tetO1 (PtetO1). TetR was introduced in the H. pylori genome and is controlled by a flaA promoter (PFlaA). TetR binds to tetO1 blocking expression of gGT. The inducer anhydrotetracycline (ATc) binds to tetR resulting in disassociation of the repressor and activation of ggt expression. (b) gGT-activity of conditional gGT mutants. Bacteria were cultured in BHI/10% FCS in the presence of 100 ng/ml ATc for 24 h. H. pylori g:O1 t has tetO1 and tetR integrated into its genome. In H. pylori g:O1 C only the ggt promoter was replaced by tetO1, n = 3–6. Bars represent the mean of three to six independent experiments as represented by single dots. Error bars represent the standard deviation. n: number of independent experiments. Mann-Whitney U-test. *p < 0.05.
H. pylori gGT contributes to initial colonization and persistence colony forming units (CFU) in the stomach (mg) of C57BL/6 mice after infection with PMSS1 wt, ∆ggt or g:O1 C. Mice were inoculated with 2 × 10⁸ bacteria twice and were sacrificed after a defined amount of time (1 week −6 months) after the first infection. Colonization was assessed by plating stomach homogenates. The detection limit is indicated by a dotted line. Each dot represents one individual mouse. PMSS1 wt (n = 15–27), PMSS1 ∆ggt (n = 8–32), PMSS1 g:O1 C (n = 12), n: number of mice. Mann-Whitney U-test. ****p < 0.01.
H. pylori gGT contributes to acid survival supporting stomach colonization (a) Survival of H. pylori in McIlvaine buffer (pH 3) supplemented with 8 mm urea, n = 3. The detection limit of the assay is indicated by a dotted line. (b) Correlation of survival of clinical isolates after 2.5 h with gGT-activity. Survival was determined in McIlvaine buffer (pH 3) supplemented with 8 mm urea. Each dot represents the mean survival of one clinical isolate, n = 3. (c) Survival of H. pylori in fasted state simulated gastric fluid (FaSSGF) supplemented with 8 mm urea. The detection limit of the assay is indicated by a dotted line., n = 4. (d) Growth of H. pylori in Brucella broth supplemented with 20% FCS at pH 5 normalized to the growth in Brucella broth at pH 7. Growth was quantified spectrophotometrically after 30 h. n = 3. (e) Urease activity after exposure of H. pylori to pH 5. Strains were grown in BHI with 10% FCS, resuspended for 2 h in McIlvaine buffer (pH 5) and lysed for the detection of urease activity. Data was normalized to the mean activity of H. pylori wt or tet-on H. pylori g:O1 t. n = 3. (f) Correlation of urease- and gGT-activity of clinical isolates. Each dot represents the mean activity of an isolate. n = 3. (g, h) Colony forming units (CFU) in the stomach (mg) of C57BL/6 mice after infection with PMSS1 wt or ∆ggt. Mice were inoculated with 2 × 10⁸ bacteria. Colonization was assessed by plating stomach homogenates. The detection limit is indicated by a dotted line. Each dot represents one individual mouse. (g) CFU/mg after 1 week of infection. Two groups of mice received sodium bicarbonate (NaHCO3) before the infection to elevate the pH in the stomach. (h) CFU/mg in different stomach sections (F: forestomach, S1–S3: stomach sections) after infection for 2 weeks. Colonization was normalized to the mean CFU/mg across sections S1–S3 for each mouse. n: number of independent experiments. Pearson’s correlation (b, f); Mann-Whitney U-test (c, g, h); ANOVA (d, e) *p < 0.05, **p < 0.01.
H. pylori gGT promotes survival in iron-restricted environments (a) Log2 transformed fold change of proteins potentially involved in metal acquisition comparing the proteome of PMSS1 wt and ∆ggt. The proteomes of three single clones isolated after 1 week, 1 month, and 3 months of infection from different mice were determined. Significant differences (-log10(p-value) > 1.3 and log2(fold change) ≥ I1I) are marked with gray boxes. Missing data points are colored white and are marked with a cross. (b) Schematic representation of the poly(a)tract in the fecA3 upstream region between positions − 181 and − 163 before the translation start. The number of adenine bases in the tract was quantified for all re-isolates (RI) and for the aliquot used for infection (BI) (c) Growth of H. pylori in iron-restricted medium. Deferoxamine mesylate (DFO) was added to the culture in different concentrations (20 µm, 50 µm, and 75 µm). Growth was monitored spectrophotometrically and was normalized to the growth without supplementation. Bars represent the mean of three to four independent experiments as indicated by dots. Error bars represent the standard deviation. (d) Correlation of growth in iron-limited conditions with gGT-activity. Each dot represents the mean growth of one clinical isolate as measured in three independent experiments. Clinical isolates were grown in the presence of 50 µm DFO and growth data were normalized to the growth without supplementation. (e) Reduction of iron(iii)citrate to Fe²⁺ in the presence of varying concentrations of Cys-Gly or GSH. As a positive control (+) 1 M ascorbic acid was added to fully reduce the available iron. As a negative control (-) Fe²⁺ concentrations were measured in a Fe(III) citrate solution. (f) Uptake of iron(iii)citrate into H. pylori in the presence and absence of 2 mm GSH or Cys-Gly. Students t-test (a, c, e, f). ANOVA (c); Pearson’s correlation (d). *p < 0.05.
H. pylori’s motility and adhesion capacity is increased in the absence of gGT during infection (a) Log2 transformed fold change of flagellar proteins and adhesion factors comparing the proteome of PMSS1 wt and ∆ggt. The proteomes of three single clones isolated after 1 week, 1 month, and 3 months of infection from different mice were determined. Significant differences (-log10(p-value) > 1.3 and log2(fold change) ≥ I1I) are marked with gray boxes. Missing data points are colored white and are marked with a cross. Values out of the range of the heat map are colored gray. (b) Schematic representation of the genomic organization of the hpaA locus as determined by whole genome sequencing. ins: insertion, del: deletion. ORF: open reading frame, BI: before infection, RI: re-isolate (c,d) Flagellar motility of H. pylori strains in the presence and absence of gGT. Soft agar assays on Brucella/10% FCS plates were performed. 100 ng/ml ATc was added to the plates to switch on gGT-activity in inducible strains. gGT-activity was not induced in the pre-culture prepared for the experiment. Motility of deletion strains and inducible strains was determined (c) as well as motility before (BI) and after infection (AI) (D). Bars represent the mean of 8 to 14 independent experiments as indicated by single dots (C) or the mean motility of 4 to 5 isolates as measured in three independent experiments (d). (e) Correlation of flagellar motility of clinical isolates with gGT-activity. Each dot represents the mean motility of an isolate as determined by 3 independent experiments (f) Adhesion capacity of H. pylori before infection (BI) and after infection (AI) to AGS cells. Bars represent the mean of three independent experiments. MFI is the median fluorescence intensity on the PE channel. Error bars represent the standard deviation. Student’s t-test (a, c, f). ANOVA (d), Pearson’s correlation (e). **p < 0.01, ****p < 0.0001.
Helicobacter pylori γ-glutamyltransferase is linked to proteomic adaptions important for colonization

April 2025

·

55 Reads

Helicobacter pylori γ-glutamyltransferase (gGT) is a virulence factor that promotes bacterial colonization and immune tolerance. Although some studies addressed potential functional mechanisms, the supportive role of gGT for in vivo colonization remains unclear. Additionally, it is unknown how different gGT expression levels may lead to compensatory mechanisms ensuring infection and persistence. Hence, it is crucial to unravel the in vivo function of gGT. We assessed acid survival under conditions mimicking the human gastric fluid and elevated the pH in the murine stomach prior to H. pylori infection to link gGT-mediated acid resistance to colonization. By comparing proteomes of gGT-proficient and -deficient isolates before and after infecting mice, we investigated proteomic adaptations of gGT-deficient bacteria during infection. Our data indicate that gGT is crucial to sustain urease activity in acidic environments, thereby supporting survival and successful colonization. Absence of gGT triggers expression of proteins involved in the nitrogen and iron metabolism and boosts the expression of adhesins and flagellar proteins during infection, resulting in increased motility and adhesion capacity. In summary, gGT-dependent mechanisms confer a growth advantage to the bacterium in the gastric environment, which renders gGT a valuable target for the development of new treatments against H. pylori infection.


Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding

April 2025

·

6 Reads

Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.


Lessons learned from FMT studies and strategic approach for precision microbiome therapy for IBD.
Key features of host-microbe interactions underlying the premise of microbial-based therapy for IBD.
Precision microbiota therapy for IBD: premise and promise

Inflammatory Bowel Disease (IBD) is a spectrum of chronic inflammatory diseases of the intestine that includes subtypes of ulcerative colitis (UC) and Crohn’s Disease (CD) and currently has no cure. While IBD results from a complex interplay between genetic, environmental, and immunological factors, sequencing advances over the last 10–15 years revealed signature changes in gut microbiota that contribute to the pathogenesis of IBD. These findings highlight IBD as a disease target for microbiome-based therapies, with the potential to treat the underlying microbial pathogenesis and provide adjuvant therapy to the emerging spectrum of advanced therapies for IBD. Building on the success of fecal microbiota transplantation (FMT) for Clostridioides difficile infection, therapies targeting gut microbiota have emerged as promising approaches for treating IBD; however, unique aspects of IBD pathogenesis highlight the need for more precision in the approach to microbiome therapeutics that leverage aspects of recipient and donor selection, diet and xenobiotics, and strain-specific interactions to enhance the efficacy and safety of IBD therapy. This review focuses on both pre-clinical and clinical studies that support the premise for microbial therapeutics for IBD and aims to provide a framework for the development of precision microbiome therapeutics to optimize clinical outcomes for patients with IBD.


3'-Sialyllactose and B. infantis synergistically alleviate gut inflammation and barrier dysfunction by enriching cross-feeding bacteria for short-chain fatty acid biosynthesis

April 2025

·

16 Reads

Ulcerative colitis (UC) poses significant threats to human health and quality of life worldwide, as it is a chronic inflammatory bowel disease. 3'-sialyllactose (3'−SL) is a key functional component of milk oligosaccharides. This study systematically evaluates the prebiotic effects of 3'-SL and its therapeutic potential in combination with Bifidobacterium infantis (B. infantis) for UC. The findings reveal that 3'-SL and B. infantis synergistically mitigate intestinal inflammation and barrier dysfunction by promoting the production of short-chain fatty acids (SCFAs) through cross-feeding mechanisms among gut microbiota. Individually, 3'-SL, B. infantis, and the synbiotic treatment all effectively alleviated UC symptoms, including reduced weight loss, improved disease activity scores, and prevention of colon shortening. Histopathological and immunofluorescence analyses further demonstrated that the synbiotic treatment significantly ameliorated colonic injury, enhanced barrier function, restored goblet cell counts, increased glycoprotein content in crypt goblet cells, and upregulated the expression of tight junction proteins (ZO-1, occludin, and claudin-1). Notably, the synbiotic treatment outperformed the individual components by better restoring gut microbiota balance, elevating SCFA levels, and modulating serum cytokine profiles, thereby reducing inflammation. These findings provide mechanistic insights into the protective effects of the synbiotic and underscore its therapeutic potential for UC and other intestinal inflammatory disorders.


Raffinose-metabolizing bacteria impair radiation-associated hematopoietic recovery via the bile acid/FXR/NF-κB signaling pathway

April 2025

·

11 Reads

Radiation-associated hematopoietic recovery (RAHR) is critical for mitigating lethal complications of acute radiation syndrome (ARS), yet therapeutic strategies remain limited. Through integrated multi-omics analysis of a total body irradiation (TBI) mouse model, we identify Bacteroides acidifaciens-dominated gut microbiota as key mediators of RAHR impairment. 16S ribosomal rRNA sequencing revealed TBI-induced dysbiosis characterized by Bacteroidaceae enrichment, while functional metagenomics identified raffinose metabolism as the most significantly perturbed pathway. Notably, raffinose supplementation (10% w/v) recapitulated radiation-induced microbiota shifts and delayed bone marrow recovery. Fecal microbiota transplantation (FMT) revealed a causative role for raffinose-metabolizing microbiota, particularly Bacteroides acidifaciens, in delaying RAHR progression. Mechanistically, B. acidifaciens-mediated bile acid deconjugation activated FXR, subsequently suppressing NF-κB-dependent hematopoietic recovery. Therapeutic FXR inhibition via ursodeoxycholic acid (UDCA) had been shown to be a viable method for rescuing RAHR. Our results delineated a microbiome-bile acid-FXR axis as a master regulator of post-irradiation hematopoiesis. Targeting B. acidifaciens or its metabolic derivatives could represent a translatable strategy to mitigate radiation-induced hematopoietic injury.


Reutericyclin, a specialized metabolite of Limosilactobacillus reuteri, mitigates risperidone-induced weight gain in mice

April 2025

·

14 Reads

The role of xenobiotic disruption of microbiota, corresponding dysbiosis, and potential links to host metabolic diseases are of critical importance. In this study, we used a widely prescribed antipsychotic drug, risperidone, known to influence weight gain in humans, to induce weight gain in C57BL/6J female mice. We hypothesized that microbes essential for maintaining gut homeostasis and energy balance would be depleted following treatment with risperidone, leading to enhanced weight gain relative to controls. Thus, we performed metagenomic analyses on stool samples to identify microbes that were excluded in risperidone-treated animals but remained present in controls. We identified multiple taxa including Limosilactobacillus reuteri as a candidate for further study. Oral supplementation with L. reuteri protected against risperidone-induced weight gain (RIWG) and was dependent on cellular production of a specialized metabolite, reutericyclin. Further, synthetic reutericyclin was sufficient to mitigate RIWG. Both synthetic reutericyclin and L. reuteri restored energy balance in the presence of risperidone to mitigate excess weight gain and induce shifts in the microbiome associated with leanness. In total, our results identify reutericyclin production by L. reuteri as a potential probiotic to restore energy balance induced by risperidone and to promote leanness.


Microbial competition mechanisms that contribute to colonization resistance can be disrupted by many routine exposures and potentially restored by strain-aware microbiota interventions. In some patients, particularly with high healthcare burden, these disrupting exposures recur or progress (noted by circular arrow), which likely contributes to further impairments in restricting colonization with antibiotic-resistant strains. Basic science and translational studies highlighted in this review suggest interventions that may be used to exploit fitness costs of antibiotic resistant strains and enhance colonization resistance. From this perspective, supporting optimal prescribing of antibiotics and other disruptive medications and dietary fiber supplementation can be viewed as microbiota interventions. The ideal microbiota intervention(s) would be feasible to administer as often as indicated (also noted by circular arrow).
Leveraging strain competition to address antimicrobial resistance with microbiota therapies

The enteric microbiota is an established reservoir for multidrug-resistant organisms that present urgent clinical and public health threats. Observational data and small interventional studies suggest that microbiome interventions, such as fecal microbiota products and characterized live biotherapeutic bacterial strains, could be an effective antibiotic-sparing prevention approach to address these threats. However, bacterial colonization is a complex ecological phenomenon that remains understudied in the context of the human gut. Antibiotic resistance is one among many adaptative strategies that impact long-term colonization. Here we review and synthesize evidence of how bacterial competition and differential fitness in the context of the gut present opportunities to improve mechanistic understanding of colonization resistance, therapeutic development, patient care, and ultimately public health.


Gut microbiota shift in Ghanaian individuals along the migration axis: the RODAM-Pros cohort

April 2025

·

28 Reads

Migration is associated with a substantial change in environmental exposures and health outcomes. We aimed to investigate the shift in gut microbiota composition and the associations with cardiometabolic outcomes in the RODAM-Pros cohort spanning multiple research sites across continents. We determined gut microbiota composition of 1,177 Ghanaian participants in rural Ghana, urban Ghana, and Amsterdam, the Netherlands, using 16S rRNA sequencing. We observed a clear gradient in gut microbiota composition and alpha and beta diversity from rural Ghana to urban Ghana, to Amsterdam. We used pairwise XGBoost machine learning classification models to identify which microbes were most distinct between locations in prevalence and abundance. The associations between these microbes and the locations could partly be explained by differences in confounders such as dietary intake. Groups of microbes that emerged or disappeared along the migration axis were associated with cardiometabolic outcomes, including higher body mass index, higher HbA1c and higher diastolic blood pressure. Concluding, we identified associations between a shift in gut microbiota composition and cardiometabolic risk along the migration axis, underscoring the relevance of gut health in the context of migration-associated adverse health outcomes.


Cross-feeding-based rational design of a probiotic combination of Bacterides xylanisolvens and Clostridium butyricum therapy for metabolic diseases

April 2025

·

40 Reads

The human gut microbiota has gained interest as an environmental factor that contributes to health or disease. The development of next-generation live biotherapeutic products (LBPs) is a promising strategy to modulate the gut microbiota and improve human health. In this study, we identified a novel cross-feeding interaction between Bacteroides xylanisolvens and Clostridium butyricum and developed their combination into a novel LBP for treating metabolic syndrome. Using in-silico analysis and in vitro experiments, we demonstrated that B. xylanisolvens supported the growth and butyrate production of C. butyricum by supplying folate, while C. butyricum reciprocated by providing pABA for folate biosynthesis. Animal gavage experiments showed that the two-strain combination LBP exhibited superior therapeutic efficacy against metabolic disorders in high-fat diet-induced obese (DIO) mice compared to either single-strain treatment. Further omics-based analyses revealed that the single-strain treatments exhibited distinct taxonomic preferences in modulating the gut microbiota, whereas the combination LBP achieved more balanced modulation to preserve taxonomic diversity to a greater extent, thereby enhancing the stability and resilience of the gut microbiome. Moreover, the two-strain combinations more effectively restored gut microbial functions by reducing disease-associated pathways and opportunistic pathogen abundance. This work demonstrates the development of new LBP therapy for metabolic diseases from cross-feeding microbial pairs which exerted better self-stability and robust efficacy in complex intestinal environments compared to conventional single-strain LBPs.


Pathways responsible for the biosynthesis of short-chain fatty acids (SCFAs) derived from indigestible dietary fiber and carbohydrate fermentation by gut microbiota. The primary SCFAs include acetate, generated through the wood–Ljungdahl pathway or from acetyl-CoA; butyrate, formed from two acetyl-CoA molecules; and propionate, produced from phosphoenolpyruvate via either the acrylate or succinate pathway or through propanediol pathway.
SCFAs primarily act on target cells through three mechanisms. (i) The first mechanism involves the binding of SCFAs to GPCRs on the cell membrane, such as GPR109A, GPR43, and GPR41. This binding can inhibit downstream pathways, including PLC, MAPK, Nf-κB, and others. (ii) SCFAs can enter the cell with the help of transporters on the cell membrane, then move into the cell nucleus where they inhibit HDAC and activate HAT. This results in increased histone acetylation, the gradual loosening of dense chromosomes, and ultimately, increased gene (LHR, XIAP or IDO-1) expression. (iii) Another mechanism of action is that SCFAs enter the cell with the help of AhR, then move to the nucleus. Nuclear receptors, such as AhR and ARNT, can bind to DNA, which suppresses gene expression involved in detoxification, metabolism, cell development, and the essential role of cellular sensors for xenobiotics, coordinating the body response.
The intestinal microbiota contributes to the epigenetic regulation of colorectal cancer by producing SCFAs, which serve as both substrates and regulators that influence chromatin-modifying enzymes. The mechanisms by which this occurs is the inhibition of histone deacetylase activity, leading to chromatin alterations typically linked to the upregulation of target gene expression in a variety of different cancer cell lines.
The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention

Cancer is a long-term illness that involves an imbalance in cellular and immune functions. It can be caused by a range of factors, including exposure to environmental carcinogens, poor diet, infections, and genetic alterations. Maintaining a healthy gut microbiome is crucial for overall health, and short-chain fatty acids (SCFAs) produced by gut microbiota play a vital role in this process. Recent research has established that alterations in the gut microbiome led to decreased production of SCFA’s in lumen of the colon, which associated with changes in the intestinal epithelial barrier function, and immunity, are closely linked to colorectal cancer (CRC) development and its progression. SCFAs influence cancer progression by modifying epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNA functions thereby affecting tumor initiation and metastasis. This suggests that restoring SCFA levels in colon through microbiota modulation could serve as an innovative strategy for CRC prevention and treatment. This review highlights the critical relationship between gut microbiota and CRC, emphasizing the potential of targeting SCFAs to enhance gut health and reduce CRC risk.


Exploring the mechanism of intestinal bacterial translocation after severe acute pancreatitis: the role of Toll-like receptor 5

April 2025

·

17 Reads

Severe acute pancreatitis (SAP)-induced intestinal bacterial translocation and enterogenic infection are among the leading causes of mortality in patients. However, the mechanisms by which SAP disrupted the intestinal barrier and led to bacterial translocation remained unclear. Therefore, we employed multi-omics analysis including microbiome, metabolome, epigenome, transcriptome, and mass cytometry (CyTOF) to identify potential targets, followed by functional validation using transgenic mice. The integrated multi-omics analysis primarily indicated overgrowth of intestinal flagellated bacteria, upregulation of intestinal Toll-like receptor 5 (TLR5) and acute inflammatory response, and increased infiltration of intestinal high-expressing TLR5 lamina propria dendritic cells (TLR5hi LPDC) after SAP. Subsequently, intestinal flagellin-TLR5 signaling was activated after SAP. Intestinal barrier disruption, bacterial translocation, and helper T cells (Th) differentiation imbalance caused by SAP were alleviated in TLR5 knocked out (Tlr5−/−) or conditionally knocked out on LPDC (Tlr5ΔDC) mice. However, TLR5 conditional knockout on intestinal epithelial cells (Tlr5ΔIEC) failed to improve SAP-induced bacterial translocation. Moreover, depletion of LPDC and regulatory T cells (Treg) ameliorated bacterial translocation after SAP. Our findings identify TLR5 on LPDC as a potential novel target for preventing or treating intestinal bacterial translocation caused by SAP.


Research workflow and data collection statistics.
Exploration of optimal data preprocessing methods.
Analysis of the impact of four data preprocessing steps on model AUC.
Comparison of batch effect removal methods and algorithms and summary of optimal machine learning methods for gut microbiome-based model construction.
Best practices for developing microbiome-based disease diagnostic classifiers through machine learning

April 2025

·

10 Reads

The human gut microbiome, crucial in various diseases, can be utilized to develop diagnostic models through machine learning (ML). The specific tools and parameters used in model construction such as data preprocessing, batch effect removal and modeling algorithms can impact model performance and generalizability. To establish an generally applicable workflow, we divided the ML process into three above-mentioned steps and optimized each sequentially using 83 gut microbiome cohorts across 20 diseases. We tested a total of 156 tool-parameter-algorithm combinations and benchmarked them according to internal- and external- AUCs. At the data preprocessing step, we identified four data preprocessing methods that performed well for regression-type algorithms and one method that excelled for non-regression-type algorithms. At the batch effect removal step, we identified the “ComBat” function from the sva R package as an effective batch effect removal method and compared the performance of various algorithms. Finally, at the ML algorithm selection step, we found that Ridge and Random Forest ranked the best. Our optimized work flow performed similarly comparing with previous exhaustive methods for disease-specific optimizations, thus is generally applicable and can provide a comprehensive guideline for constructing diagnostic models for a range of diseases, potentially serving as a powerful tool for future medical diagnostics.


Global changes in disease burdens of infants and children with urbanization. (a) Changes in total prevalence (per 100,000) of 104 non-communicable diseases (NCDs) among children under 5 years across 203 countries between 1990 and 2021. (b) Changes in total prevalence (per 100,000) of neonatal disorders among infants under 1 year old across 203 countries between 1990 and 2021.
Mechanisms by which prenatal urbanized diets affect maternal-fetal crosstalk. Effects of healthy (left) and urbanized (right) prenatal diets on fetal development are illustrated. Urbanized dietary patterns significantly alter gut microbiota composition and metabolic profiles, potentially disrupting the host’s immune and metabolic systems. These microbiota dysbiosis, metabolic abnormalities, and immune dysfunctions resulting from urbanized diets may transmit from the mother to offspring via placental transfer, further adversely affecting offspring health. Human and animal studies were distinguished by using solid and dashed lines, respectively. SCFAs, short-chain fatty acids; TMA, trimethylamine; TMAO, trimethylamine N-oxide; 5-AVAB, 5-aminovaleric acid betaine; IPA, 3-indolepropionic acid; 5-HT, serotonin; LPS, lipopolysaccharides; MMCs, maternal microchimeric cells; GPCRs, G protein-coupled receptors; IgG, immunoglobulin G; AHR, aryl hydrocarbon receptor; IL-22, interleukin-22; ILC3, intestinal group 3 innate lymphoid cells; tnf-a, tumor necrosis factor-alpha; CX3CL1, chemokine C-X3-C motif ligand 1; NK cell, natural killer cell.
Mechanisms by which urbanized postnatal diets affect the maternal-infant crosstalk. Effects of healthy (left) and urbanized (right) postnatal diets on neonatal development are illustrated. Urbanized diets alter the composition of HMOs, immune cells, microbiome (bacteria, fungi, and viruses), and metabolites in breast milk, which can be transferred to offspring, thus influencing the assembly of the early-life gut microbiome and the development of the immune system. Human and animal studies were distinguished by using solid and dashed lines, respectively. SCFAs, short-chain fatty acids; TMA, trimethylamine; TMAO, trimethylamine N-oxide; HMOs, human milk oligosaccharides; 5-AVAB, 5-aminovaleric acid betaine; ILA, indole-3-lactic acid; 12,13-diHOME, 12,13-dihydroxy-9Z-octadecenoic acid; sIgA, secretory immunoglobulin A; AHR, aryl hydrocarbon receptor; IL-6, interleukin-6; TNF-α, tumor necrosis factor-alpha; NEC, necrotizing enterocolitis.
A proposed framework for the development of microbiome-directed precision diet. The framework consists of a three-step strategy: 1) identify specific molecules in the blood, or breast milk of mothers to predict the long-term health trajectories of offspring; 2) establish a comprehensive database that captures information on human and gut microbial metabolism and their links to hundreds of diseases and nutritional data, aiming to predict host-diet-microbiome interactions and their metabolic output; 3) predict disease risks in their offspring by measuring pregnant women’s metabolic profiles and customizing dietary and nutrition strategies to modulate key metabolite levels according to each pregnant woman ’s unique gut microbiome composition and function. Continuous optimization of algorithms and databases is essential to improve the ability to predict the host and microbial metabolic output of a defined diet, ultimately maximizing maternal nutritional benefits and enhancing offspring health.
Maternal consumption of urbanized diet compromises early-life health in association with gut microbiota

Urbanization has significantly transformed dietary habits worldwide, contributing to a globally increased burden of non-communicable diseases and altered gut microbiota landscape. However, it is often overlooked that the adverse effects of these dietary changes can be transmitted from the mother to offspring during early developmental stages, subsequently influencing the predisposition to various diseases later in life. This review aims to delineate the detrimental effects of maternal urban-lifestyle diet (urbanized diet) on early-life health and gut microbiota assembly, provide mechanistic insights on how urbanized diet mediates mother-to-offspring transfer of bioactive substances in both intrauterine and extrauterine and thus affects fetal and neonatal development. Moreover, we also further propose a framework for developing microbiome-targeted precision nutrition and diet strategies specifically for pregnant and lactating women. The establishment of such knowledge can help develop proactive preventive measures from the beginning of life, ultimately reducing the long-term risk of disease and improving public health outcomes.


Journal metrics


12.2 (2023)

Journal Impact Factor™


21%

Acceptance rate


18.2 (2023)

CiteScore™


5 days

Submission to first decision


2.069 (2023)

SNIP


3.075 (2023)

SJR

Editors