Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.943
1.
Carbohydr Polym ; 339: 122284, 2024 Sep 01.
Article En | MEDLINE | ID: mdl-38823935

Interactions between human gut microbiota and dietary fibres (DF) are influenced by the complexity and diversity of both individual microbiota and sources of DF. Based on 480 in vitro fermentations, a full factorial experiment was performed with six faecal inocula representing two enterotypes and three DF sources with nanometer, micrometer, and millimeter length-scales (apple pectin, apple cell walls and apple particles) at two concentrations. Increasing DF size reduced substrate disappearance and fermentation rates but not biomass growth. Concentrated DF enhanced butyrate production and lactate cross-feeding. Enterotype differentiated final microbial compositions but not biomass or fermentation metabolite profiles. Individual donor microbiota differences did not influence DF type or concentration effects but were manifested in the promotion of different functional microbes within each population with the capacity to degrade the DF substrates. Overall, consistent effects (independent of donor microbiota variation) of DF type and concentration on kinetics of substrate degradation, microbial biomass production, gas kinetics and metabolite profiles were found, which can form the basis for informed design of DF for desired rates/sites and consequences of gut fermentation. These results add further evidence to the concept that, despite variations between individuals, the human gut microbiota represents a community with conserved emergent properties.


Dietary Fiber , Feces , Fermentation , Gastrointestinal Microbiome , Pectins , Pectins/metabolism , Dietary Fiber/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Feces/microbiology , Malus/metabolism , Adult , Male , Female , Bacteria/metabolism , Bacteria/classification , Biomass
2.
FASEB J ; 38(11): e23648, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38822661

Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.


Behavior, Animal , Germ-Free Life , Serotonin , Animals , Serotonin/metabolism , Mice , Male , Gastrointestinal Microbiome/physiology , Brain/metabolism , Tryptophan Hydroxylase/metabolism , Tryptophan Hydroxylase/genetics , Anxiety/metabolism , Anxiety/microbiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Colon/metabolism , Colon/microbiology
3.
Arch Dermatol Res ; 316(6): 315, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38822918

Hypertrophic scarring is a fibro-proliferative disorder caused by abnormal cutaneous wound healing. Circulating metabolites and the gut microbiome may be involved in the formation of these scars, but high-quality evidence of causality is lacking. To assess whether circulating metabolites and the gut microbiome contain genetically predicted modifiable risk factors for hypertrophic scar formation. Two-sample Mendelian randomization (MR) was performed using MR-Egger, inverse-variance weighting (IVW), Mendelian Randomization Pleiotropy RESidual Sum and Outlier, maximum likelihood, and weighted median methods. Based on the genome-wide significance level, genetically predicted uridine (P = 0.015, odds ratio [OR] = 1903.514, 95% confidence interval [CI] 4.280-846,616.433) and isovalerylcarnitine (P = 0.039, OR = 7.765, 95% CI 1.106-54.512) were positively correlated with hypertrophic scar risk, while N-acetylalanine (P = 0.013, OR = 7.98E-10, 95% CI 5.19E-17-0.012) and glycochenodeoxycholate (P = 0.021, OR = 0.021 95% CI 0.003-0.628) were negatively correlated. Gastranaerophilales and two unknown gut microbe species (P = 0.031, OR = 0.378, 95% CI 0.156-0.914) were associated with an decreased risk of hypertrophic scarring. Circulating metabolites and gut microbiome components may have either positive or negative causal effects on hypertrophic scar formation. The study provides new insights into strategies for diagnosing and limiting hypertrophic scarring.


Cicatrix, Hypertrophic , Gastrointestinal Microbiome , Mendelian Randomization Analysis , Humans , Gastrointestinal Microbiome/physiology , Cicatrix, Hypertrophic/microbiology , Cicatrix, Hypertrophic/blood , Cicatrix, Hypertrophic/etiology , Risk Factors , Genome-Wide Association Study , Polymorphism, Single Nucleotide
5.
NPJ Syst Biol Appl ; 10(1): 56, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802371

Despite significant advances in reconstructing genome-scale metabolic networks, the understanding of cellular metabolism remains incomplete for many organisms. A promising approach for elucidating cellular metabolism is analysing the full scope of enzyme promiscuity, which exploits the capacity of enzymes to bind to non-annotated substrates and generate novel reactions. To guide time-consuming costly experimentation, different computational methods have been proposed for exploring enzyme promiscuity. One relevant algorithm is PROXIMAL, which strongly relies on KEGG to define generic reaction rules and link specific molecular substructures with associated chemical transformations. Here, we present a completely new pipeline, PROXIMAL2, which overcomes the dependency on KEGG data. In addition, PROXIMAL2 introduces two relevant improvements with respect to the former version: i) correct treatment of multi-step reactions and ii) tracking of electric charges in the transformations. We compare PROXIMAL and PROXIMAL2 in recovering annotated products from substrates in KEGG reactions, finding a highly significant improvement in the level of accuracy. We then applied PROXIMAL2 to predict degradation reactions of phenolic compounds in the human gut microbiota. The results were compared to RetroPath RL, a different and relevant enzyme promiscuity method. We found a significant overlap between these two methods but also complementary results, which open new research directions into this relevant question in nutrition.


Algorithms , Computational Biology , Gastrointestinal Microbiome , Metabolic Networks and Pathways , Phenols , Gastrointestinal Microbiome/physiology , Humans , Phenols/metabolism , Computational Biology/methods
6.
Adv Microb Physiol ; 84: 83-133, 2024.
Article En | MEDLINE | ID: mdl-38821635

The gut microbiota is increasingly recognised as a key player in influencing human health and changes in the gut microbiota have been strongly linked with many non-communicable conditions in humans such as type 2 diabetes, obesity and cardiovascular disease. However, characterising the molecular mechanisms that underpin these associations remains an important challenge for researchers. The gut microbiota is a complex microbial community that acts as a metabolic interface to transform ingested food (and other xenobiotics) into metabolites that are detected in the host faeces, urine and blood. Many of these metabolites are only produced by microbes and there is accumulating evidence to suggest that these microbe-specific metabolites do act as effectors to influence human physiology. For example, the gut microbiota can digest dietary complex polysaccharides (such as fibre) into short-chain fatty acids (SCFA) such as acetate, propionate and butyrate that have a pervasive role in host physiology from nutrition to immune function. In this review we will outline our current understanding of the role of some key microbial metabolites, such as SCFA, indole and bile acids, in human health. Whilst many studies linking microbial metabolites with human health are correlative we will try to highlight examples where genetic evidence is available to support a specific role for a microbial metabolite in host health and well-being.


Bile Acids and Salts , Fatty Acids, Volatile , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Fatty Acids, Volatile/metabolism , Bile Acids and Salts/metabolism , Indoles/metabolism , Host Microbial Interactions , Bacteria/metabolism , Bacteria/genetics , Animals
7.
PeerJ ; 12: e17385, 2024.
Article En | MEDLINE | ID: mdl-38818452

Background: Koalas, an Australian arboreal marsupial, depend on eucalypt tree leaves for their diet. They selectively consume only a few of the hundreds of available eucalypt species. Since the koala gut microbiome is essential for the digestion and detoxification of eucalypts, their individual differences in the gut microbiome may lead to variations in their eucalypt selection and eucalypt metabolic capacity. However, research focusing on the relationship between the gut microbiome and differences in food preferences is very limited. We aimed to determine whether individual and regional differences exist in the gut microbiome of koalas as well as the mechanism by which these differences influence eucalypt selection. Methods: Foraging data were collected from six koalas and a total of 62 feces were collected from 15 koalas of two zoos in Japan. The mitochondrial phylogenetic analysis was conducted to estimate the mitochondrial maternal origin of each koala. In addition, the 16S-based gut microbiome of 15 koalas was analyzed to determine the composition and diversity of each koala's gut microbiome. We used these data to investigate the relationship among mitochondrial maternal origin, gut microbiome and eucalypt diet selection. Results and Discussion: This research revealed that diversity and composition of the gut microbiome and that eucalypt diet selection of koalas differs among regions. We also revealed that the gut microbiome alpha diversity was correlated with foraging diversity in koalas. These individual and regional differences would result from vertical (maternal) transmission of the gut microbiome and represent an intraspecific variation in koala foraging strategies. Further, we demonstrated that certain gut bacteria were strongly correlated with both mitochondrial maternal origin and eucalypt foraging patterns. Bacteria found to be associated with mitochondrial maternal origin included bacteria involved in fiber digestion and degradation of secondary metabolites, such as the families Rikenellaceae and Synergistaceae. These bacteria may cause differences in metabolic capacity between individual and regional koalas and influence their eucalypt selection. Conclusion: We showed that the characteristics (composition and diversity) of the gut microbiome and eucalypt diet selection of koalas differ by individuals and regional origins as we expected. In addition, some gut bacteria that could influence eucalypt foraging of koalas showed the relationships with both mitochondrial maternal origin and eucalypt foraging pattern. These differences in the gut microbiome between regional origins may make a difference in eucalypt selection. Given the importance of the gut microbiome to koalas foraging on eucalypts and their strong symbiotic relationship, future studies should focus on the symbiotic relationship and coevolution between koalas and the gut microbiome to understand individual and regional differences in eucalypt diet selection by koalas.


Eucalyptus , Gastrointestinal Microbiome , Phascolarctidae , Animals , Gastrointestinal Microbiome/physiology , Gastrointestinal Microbiome/genetics , Phascolarctidae/microbiology , Eucalyptus/microbiology , Female , Diet/veterinary , Feces/microbiology , Food Preferences , Phylogeny , Male , Japan , Maternal Inheritance/genetics
8.
Cell Host Microbe ; 32(5): 623-624, 2024 May 08.
Article En | MEDLINE | ID: mdl-38723597

Common nutrients in our diet often affect our health through unexpected mechanisms. In a recent issue of Nature, Scott et al. show gut microbes convert dietary tryptophan into metabolites activating intestinal dopamine receptors, which can block attachment of bacterial pathogens to host cells.


Dopamine , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Dopamine/metabolism , Humans , Receptors, Dopamine/metabolism , Animals , Tryptophan/metabolism , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism , Bacteria/metabolism , Host-Pathogen Interactions , Bacterial Adhesion
9.
Cell Host Microbe ; 32(5): 630-632, 2024 May 08.
Article En | MEDLINE | ID: mdl-38723600

The gut microbiota has the capacity to metabolize food-derived molecules. In this issue of Cell Host & Microbe, Li et al. explore how some bacterial species of the gut microbiota can deplete amino acids in the gut lumen, modulating the amino acid landscape and energy metabolism of the host.


Amino Acids , Energy Metabolism , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Amino Acids/metabolism , Humans , Bacteria/metabolism , Bacteria/genetics , Animals , Host Microbial Interactions , Gastrointestinal Tract/microbiology
10.
Cell Host Microbe ; 32(5): 639-650, 2024 May 08.
Article En | MEDLINE | ID: mdl-38723604

There is rapidly growing awareness of microbiome assembly and function in early-life gut health. Although many factors, such as antibiotic use and highly processed diets, impinge on this process, most research has focused on people residing in high-income countries. However, much of the world's population lives in low- and middle-income countries (LMICs), where, in addition to erratic antibiotic use and suboptimal diets, these groups experience unique challenges. Indeed, many children in LMICs are infected with intestinal helminths. Although helminth infections are strongly associated with diverse developmental co-morbidities and induce profound microbiome changes, few studies have directly examined whether intersecting pathways between these components of the holobiont shape health outcomes in early life. Here, we summarize microbial colonization within the first years of human life, how helminth-mediated changes to the gut microbiome may affect postnatal growth, and why more research on this relationship may improve health across the lifespan.


Gastrointestinal Microbiome , Helminthiasis , Helminths , Gastrointestinal Microbiome/physiology , Humans , Helminths/physiology , Animals , Infant , Intestinal Diseases, Parasitic
11.
Medicine (Baltimore) ; 103(19): e38088, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728472

Microbiota modulation, the intentional change in the structure and function of the microbial community, is an emerging trajectory that holds the promise to mitigate an infinite number of health issues. The present review illustrates the underlying principles of microbiota modulation and the various applications of this fundamental process to human health, healthcare management, and pharmacologic interventions. Different strategies, directing on dietary interventions, fecal microbiota transplantation, treatment with antibiotics, bacteriophages, microbiome engineering, and modulation of the immune system, are described in detail. This therapeutic implication is reflected in clinical applications to gastrointestinal disorders and immune-mediated diseases for microbiota-modulating agents. In addition to this, the review outlines the challenges of translating researched outcomes into clinical practice to consider safety and provides insights into future research directions of this rapidly developing area.


Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome/physiology , Anti-Bacterial Agents/therapeutic use , Probiotics/therapeutic use , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/microbiology
12.
Gut Microbes ; 16(1): 2350784, 2024.
Article En | MEDLINE | ID: mdl-38727219

The gut microbiota constitutes a vast ecological system within the human body, forming a mutually interdependent entity with the host. In recent years, advancements in molecular biology technologies have provided a clearer understanding of the role of the gut microbiota. They not only influence the local immune status and metabolic functions of the host's intestinal tract but also impact the functional transformation of hematopoietic stem cells (HSCs) through the gut-blood axis. In this review, we will discuss the role of the gut microbiota in influencing hematopoiesis. We analyze the interactions between HSCs and other cellular components, with a particular emphasis on the direct functional regulation of HSCs by the gut microbiota and their indirect influence through cellular components in the bone marrow microenvironment. Additionally, we propose potential control targets for signaling pathways triggered by the gut microbiota to regulate hematopoietic function, filling crucial knowledge gaps in the development of this research field.


Gastrointestinal Microbiome , Hematopoiesis , Hematopoietic Stem Cells , Hematopoiesis/physiology , Gastrointestinal Microbiome/physiology , Humans , Hematopoietic Stem Cells/microbiology , Animals , Signal Transduction , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Gastrointestinal Tract/microbiology , Bone Marrow/microbiology , Bone Marrow/physiology
13.
Nat Commun ; 15(1): 4276, 2024 May 20.
Article En | MEDLINE | ID: mdl-38769296

Alterations in gut microbiota composition are suggested to contribute to cardiometabolic diseases, in part by producing bioactive molecules. Some of the metabolites are produced by very low abundant bacterial taxa, which largely have been neglected due to limits of detection. However, the concentration of microbially produced metabolites from these taxa can still reach high levels and have substantial impact on host physiology. To explore this concept, we focused on the generation of secondary bile acids by 7α-dehydroxylating bacteria and demonstrated that addition of a very low abundant bacteria to a community can change the metabolic output dramatically. We show that Clostridium scindens converts cholic acid into the secondary bile acid deoxycholic acid (DCA) very efficiently even though the abundance of C. scindens is low, but still detectable by digital droplet PCR. We also show that colonization of germ-free female mice with a community containing C. scindens induces DCA production and affects host metabolism. Finally, we show that DCA correlates with impaired glucose metabolism and a worsened lipid profile in individuals with type 2 diabetes, which implies that this metabolic pathway may contribute to the development of cardiometabolic disease.


Deoxycholic Acid , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Glucose , Deoxycholic Acid/metabolism , Animals , Gastrointestinal Microbiome/physiology , Female , Glucose/metabolism , Mice , Humans , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/metabolism , Mice, Inbred C57BL , Clostridium/metabolism , Clostridium/genetics , Cholic Acid/metabolism , Male
16.
Vet Q ; 44(1): 1-9, 2024 Dec.
Article En | MEDLINE | ID: mdl-38733121

The gut microbiota (GM) is essential for mammalian health. Although the association between infant GM and breast milk (BM) composition has been well established in humans, such a relationship has not been investigated in horses. Hence, this study was conducted to analyze the GM formation of foals during lactation and determine the presence of low-molecular-weight metabolites in mares' BM and their role in shaping foals' GM. The fecal and BM samples from six pairs of foals and mares were subjected to 16S ribosomal RNA metagenomic and metabolomic analyses, respectively. The composition of foal GM changed during lactation time; hierarchical cluster analysis divided the fetal GM into three groups corresponding to different time points in foal development. The level of most metabolites in milk decreased over time with increasing milk yield, while threonic acid and ascorbic acid increased. Further analyses revealed gut bacteria that correlated with changes in milk metabolites; for instance, there was a positive correlation between Bacteroidaceae in the foal's gut microbiota and serine/glycine in the mother's milk. These findings help improve the rearing environment of lactating horses and establish artificial feeding methods for foals.


Animals, Newborn , Feces , Gastrointestinal Microbiome , Lactation , Milk , RNA, Ribosomal, 16S , Animals , Gastrointestinal Microbiome/physiology , Horses , Female , Milk/chemistry , Milk/microbiology , Feces/microbiology , Feces/chemistry , Animals, Newborn/microbiology , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/analysis
17.
Gut Microbes ; 16(1): 2350173, 2024.
Article En | MEDLINE | ID: mdl-38738780

Although fecal microbiota composition is considered to preserve relevant and representative information for distal colonic content, it is evident that it does not represent microbial communities inhabiting the small intestine. Nevertheless, studies investigating the human small intestinal microbiome and its response to dietary intervention are still scarce. The current study investigated the spatio-temporal dynamics of the small intestinal microbiome within a day and over 20 days, as well as its responses to a 14-day synbiotic or placebo control supplementation in 20 healthy subjects. Microbial composition and metabolome of luminal content of duodenum, jejunum, proximal ileum and feces differed significantly from each other. Additionally, differences in microbiota composition along the small intestine were most pronounced in the morning after overnight fasting, whereas differences in composition were not always measurable around noon or in the afternoon. Although overall small intestinal microbiota composition did not change significantly within 1 day and during 20 days, remarkable, individual-specific temporal dynamics were observed in individual subjects. In response to the synbiotic supplementation, only the microbial diversity in jejunum changed significantly. Increased metabolic activity of probiotic strains during intestinal passage, as assessed by metatranscriptome analysis, was not observed. Nevertheless, synbiotic supplementation led to a short-term spike in the relative abundance of genera included in the product in the small intestine approximately 2 hours post-ingestion. Collectively, small intestinal microbiota are highly dynamic. Ingested probiotic bacteria could lead to a transient spike in the relative abundance of corresponding genera and ASVs, suggesting their passage through the entire gastrointestinal tract. This study was registered to http://www.clinicaltrials.gov, NCT02018900.


Bacteria , Feces , Gastrointestinal Microbiome , Intestine, Small , Synbiotics , Humans , Synbiotics/administration & dosage , Gastrointestinal Microbiome/physiology , Male , Adult , Intestine, Small/microbiology , Intestine, Small/metabolism , Female , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacteria/genetics , Feces/microbiology , Young Adult , Probiotics/administration & dosage , Metabolome , Healthy Volunteers , Spatio-Temporal Analysis
18.
Nutrients ; 16(9)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38732510

Amino acids are essential for normal pregnancy and fetal development. Disruptions in maternal amino acid metabolism have been associated with various adult diseases later in life, a phenomenon referred to as the developmental origins of health and disease (DOHaD). In this review, we examine the recent evidence highlighting the significant impact of amino acids on fetal programming, their influence on the modulation of gut microbiota, and their repercussions on offspring outcomes, particularly in the context of cardiovascular-kidney-metabolic (CKM) syndrome. Furthermore, we delve into experimental studies that have unveiled the protective effects of therapies targeting amino acids. These interventions have demonstrated the potential to reprogram traits associated with CKM in offspring. The discussion encompasses the challenges of translating the findings from animal studies to clinical applications, emphasizing the complexity of this process. Additionally, we propose potential solutions to overcome these challenges. Ultimately, as we move forward, future research endeavors should aim to pinpoint the most effective amino-acid-targeted therapies, determining the optimal dosage and mode of administration. This exploration is essential for maximizing the reprogramming effects, ultimately contributing to the enhancement of cardiovascular-kidney-metabolic health in offspring.


Amino Acids , Cardiovascular Diseases , Fetal Development , Gastrointestinal Microbiome , Kidney , Humans , Pregnancy , Female , Amino Acids/metabolism , Kidney/metabolism , Animals , Gastrointestinal Microbiome/physiology , Prenatal Exposure Delayed Effects , Kidney Diseases , Maternal Nutritional Physiological Phenomena
19.
Biol Res ; 57(1): 23, 2024 May 06.
Article En | MEDLINE | ID: mdl-38705984

Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats.


Anxiety , Brain-Gut Axis , Diet, High-Fat , Gastrointestinal Microbiome , Animals , Male , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Anxiety/microbiology , Brain-Gut Axis/physiology , Rats , Rats, Sprague-Dawley , Obesity/microbiology , Obesity/psychology , Obesity/metabolism , Signal Transduction/physiology , Behavior, Animal/physiology
20.
BMC Psychiatry ; 24(1): 334, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698338

BACKGROUND: This study aimed to explore the gut microbiota and inflammatory factor characteristics in major depressive disorder (MDD) patients with anorexia and to analyze the correlation between gut microbiota and inflammatory factors, anorexia, and HAMD scores. METHODS: 46 MDD patients and 46 healthy controls (HC) were included in the study. The 46 MDD patients were divided into two groups according to whether they had anorexia:20 MDD without anorexia (MDA0 group) and 26 MDD with anorexia (MDA1 group). We used the Hamilton Depression Scale-24 (HAMD-24) to evaluate the depression status of all participants and 16 S ribosomal RNA (16 S rRNA)sequencing to evaluate the composition of the gut microbiota. Inflammatory factors in peripheral blood such as C-reactive protein (CRP) were detected using enzyme-linked immunosorbent assay (ELISA). Spearman's correlation analysis was used to evaluate the correlation between gut microbiota and inflammatory factors, HAMD scores, and anorexia. RESULTS: 1). CRP was significantly higher in the MDA0, MDA1, than HC. 2). An analysis of α-diversity shows: the Simpson and Pielou indices of the HC group are higher than the MDA1 group (P < 0.05). 3). The ß-diversity analysis shows differences in the composition of microbial communities between the MDA0, MDA1, and HC group. 4). A correlation analysis showed that Blautia positively correlated with anorexia, HAMD scores, and CRP level, whereas Faecalibacterium, Bacteroides, Roseburia, and Parabacteroides negatively correlated with anorexia, HAMD scores, and CRP level. 5). The receiver operating characteristic (ROC) curve was drawn using the differential bacterial genera between MDD patients with or without anorexia as biomarkers to identify whether MDD patients were accompanied with anorexia, and its area under curve (AUC) was 0.85. The ROC curve was drawn using the differential bacterial genera between MDD patients with anorexia and healthy controls as biomarkers to diagnose MDD patients with anorexia, with its AUC was 0.97. CONCLUSION: This study suggested that MDD patients with anorexia had a distinct gut microbiota compared to healthy individuals, with higher level of CRP. Blautia was more abundant in MDD patients with anorexia and positively correlated with CRP, HAMD scores, and anorexia. The gut microbiota might have influenced MDD and anorexia through the inflammatory factor CRP.


Anorexia , C-Reactive Protein , Depressive Disorder, Major , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Depressive Disorder, Major/blood , Depressive Disorder, Major/microbiology , Female , Adult , Male , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Anorexia/microbiology , Anorexia/blood , Inflammation/blood , Middle Aged , Case-Control Studies , RNA, Ribosomal, 16S/genetics , Young Adult
...