Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
BMC Psychiatry ; 24(1): 334, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698338

ABSTRACT

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.


Subject(s)
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
2.
J Affect Disord ; 356: 664-671, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38615845

ABSTRACT

OBJECTIVE: Most patients with major depressive disorder (MDD) have somatic symptoms, but little studies pay attention in the microbial-inflammatory mechanisms of these somatic symptoms. Our study aimed to investigate alterations in gut microbiota and its correlation with inflammatory marker levels and somatic symptoms in first-episode treatment-naive MDD. METHODS: Subjects contained 160 MDD patients and 101 healthy controls (HCs). MDD patients were divided into MDD with somatic symptoms group (MDDS) and MDD without somatic symptoms group (MDDN) based on Somatic Self-rating Scale (SSS). 16S ribosomal RNA sequencing were performed to analyze the composition of the fecal microbiota. The inflammatory factors were measured using enzyme linked immunosorbent assay (ELISA). Correlation among the altered gut microbiota, inflammatory factor and severity of clinical symptoms were analysized. RESULTS: Relative to HCs, MDD patients had higher levels of high-sensitivity C-reactive protein (hs-CRP) as well as disordered α-diversity and ß-diversity of gut microbiota. Linear discriminant effect size (LEfSe) analysis showed that MDD patients had higher proportions of Bifidobacterium, Blautia, Haemophilus and lower proportions of Bacteroides, Faecalibacterium, Roseburia, Dialister, Sutterella, Parabacteroides, Bordetella, and Phascolarctobacterium from the genus aspect. Furthermore, correlation analysis showed Bacteroides and Roseburia had negative correlations with the hs-CRP, HAMD-24, the total and factor scores of SSS in all participants. Further, compared with MDDN, the Pielous evenness was higher in MDDS. Random Forest (RF) analysis showed 20 most important genera discriminating MDD-S and MDDN, HCs. The ROC analysis showed that the AUC was 0.90 and 0.81 combining these genera respectively. CONCLUSION: Our study manifested MDD patients showed disordered gut microbiota and elevated hs-CRP levels, and altered gut microbiota was closely associated with hs-CRP, depressive symptoms, and somatic symptoms.


Subject(s)
C-Reactive Protein , Depressive Disorder, Major , Feces , Gastrointestinal Microbiome , Humans , Depressive Disorder, Major/microbiology , Depressive Disorder, Major/blood , Female , Male , Adult , C-Reactive Protein/analysis , Feces/microbiology , Middle Aged , Medically Unexplained Symptoms , RNA, Ribosomal, 16S/genetics , Case-Control Studies , Young Adult
3.
Transl Psychiatry ; 13(1): 137, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37117202

ABSTRACT

Disturbed gut microbiota is a potential factor in the pathogenesis of major depressive disorder (MDD), yet whether gut microbiota dysbiosis is associated with the severity of MDD remains unclear. Here, we performed shotgun metagenomic profiling of cross-sectional stool samples from MDD (n = 138) and healthy controls (n = 155). The patients with MDD were divided into three groups according to Hamilton Depression Rating Scale 17 (HAMD-17), including mild (n = 24), moderate (n = 72) and severe (n = 42) individuals, respectively. We found that microbial diversity was closely related to the severity of MDD. Compared to HCs, the abundance of Bacteroides was significantly increased in both moderate and severe MDD, while Ruminococcus and Eubacterium depleted mainly in severe group. In addition, we identified 99 bacteria species specific to severity of depression. Furthermore, a panel of microbiota marker comprising of 37 bacteria species enabled to effectively distinguish MDD patients with different severity. Together, we identified different perturbation patterns of gut microbiota in mild-to-severe depression, and identified potential diagnostic and therapeutic targets.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , Microbiota , Humans , Depressive Disorder, Major/microbiology , Cross-Sectional Studies , Feces/microbiology , Bacteria
4.
Nutrients ; 15(6)2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36986112

ABSTRACT

Probiotics are currently the subject of intensive research pursuits and also represent a multi-billion-dollar global industry given their vast potential to improve human health. In addition, mental health represents a key domain of healthcare, which currently has limited, adverse-effect prone treatment options, and probiotics may hold the potential to be a novel, customizable treatment for depression. Clinical depression is a common, potentially debilitating condition that may be amenable to a precision psychiatry-based approach utilizing probiotics. Although our understanding has not yet reached a sufficient level, this could be a therapeutic approach that can be tailored for specific individuals with their own unique set of characteristics and health issues. Scientifically, the use of probiotics as a treatment for depression has a valid basis rooted in the microbiota-gut-brain axis (MGBA) mechanisms, which play a role in the pathophysiology of depression. In theory, probiotics appear to be ideal as adjunct therapeutics for major depressive disorder (MDD) and as stand-alone therapeutics for mild MDD and may potentially revolutionize the treatment of depressive disorders. Although there is a wide range of probiotics and an almost limitless range of therapeutic combinations, this review aims to narrow the focus to the most widely commercialized and studied strains, namely Lactobacillus and Bifidobacterium, and to bring together the arguments for their usage in patients with major depressive disorder (MDD). Clinicians, scientists, and industrialists are critical stakeholders in exploring this groundbreaking concept.


Subject(s)
Depressive Disorder, Major , Probiotics , Humans , Depression/drug therapy , Depressive Disorder, Major/therapy , Depressive Disorder, Major/microbiology , Mental Health , Probiotics/therapeutic use , Bifidobacterium
5.
Sci Rep ; 12(1): 20977, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36470908

ABSTRACT

Microbiota-gut-brain axis signaling plays a pivotal role in mood disorders. The communication between the host and the gut microbiota may involve complex regulatory networks. Previous evidence showed that host-fecal microRNAs (miRNAs) interactions partly shaped gut microbiota composition. We hypothesized that some miRNAs are correlated with specific bacteria in the fecal samples in patients with major depressive disorder (MDD), and these miRNAs would show enrichment in pathways associated with MDD. MDD patients and healthy controls were recruited to collect fecal samples. We performed 16S ribosome RNA sequence using the Illumina MiSeq sequencers and analysis of 798 fecal miRNAs using the nCounter Human-v2 miRNA Panel in 20 subjects. We calculated the Spearman correlation coefficient for bacteria abundance and miRNA expressions, and analyzed the predicted miRNA pathways by enrichment analysis with false-discovery correction (FDR). A total of 270 genera and 798 miRNAs were detected in the fecal samples. Seven genera (Anaerostipes, Bacteroides, Bifidobacterium, Clostridium, Collinsella, Dialister, and Roseburia) had fold changes greater than one and were present in over 90% of all fecal samples. In particular, Bacteroides and Dialister significantly differed between the MDD and control groups (p-value < 0.05). The correlation coefficients between the seven genera and miRNAs in patients with MDD showed 48 pairs of positive correlations and 36 negative correlations (p-value < 0.01). For miRNA predicted functions, there were 57 predicted pathways with a p-value < 0.001, including MDD-associated pathways, axon guidance, circadian rhythm, dopaminergic synapse, focal adhesion, long-term potentiation, and neurotrophin signaling pathway. In the current pilot study, our findings suggest specific genera highly correlated with the predicted miRNA functions, which might provide clues for the interaction between host factors and gut microbiota via the microbiota-gut-brain axis. Follow-up studies with larger sample sizes and refined experimental design are essential to dissect the roles between gut microbiota and miRNAs for depression.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , MicroRNAs , Humans , Gastrointestinal Microbiome/genetics , Depressive Disorder, Major/genetics , Depressive Disorder, Major/microbiology , MicroRNAs/genetics , Pilot Projects , Feces/microbiology , Bacteria/genetics , Bacteroides/genetics , Clostridiales/genetics , Veillonellaceae/genetics
6.
Front Cell Infect Microbiol ; 12: 907239, 2022.
Article in English | MEDLINE | ID: mdl-35899051

ABSTRACT

Objective: Increasing evidence shows a close relationship between gut microbiota and major depressive disorder (MDD), but the specific mechanisms remain unknown. This study was conducted to explore differential gut microbiota compositions related to the severity of MDD. Methods: Healthy controls (HC) (n = 131) and MDD patients (n = 130) were included. MDD patients with Hamilton Depression Rating Scale (HDRS) score <25 and ≥25 were assigned into moderate (n = 72) and severe (n = 58) MDD groups, respectively. Univariate and multivariate analyses were used to analyze the gut microbiota compositions at the genus level. Results: Thirty-six and 27 differential genera were identified in moderate and severe MDD patients, respectively. The differential genera in moderate and severe MDD patients mainly belonged to three (Firmicutes, Actinobacteriota, and Bacteroidota) and two phyla (Firmicutes and Bacteroidota), respectively. One specific covarying network from phylum Actinobacteriota was identified in moderate MDD patients. In addition, five genera (Collinsella, Eggerthella, Alistipes, Faecalibacterium, and Flavonifractor) from the shared differential genera by two MDD groups had a fair efficacy in diagnosing MDD from HC (AUC = 0.786). Conclusions: Our results were helpful for further exploring the role of gut microbiota in the pathogenesis of depression and developing objective diagnostic methods for MDD.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , Bacteria , Depressive Disorder, Major/microbiology , Humans
7.
Nat Genet ; 54(2): 134-142, 2022 02.
Article in English | MEDLINE | ID: mdl-35115689

ABSTRACT

Human genetic variation affects the gut microbiota through a complex combination of environmental and host factors. Here we characterize genetic variations associated with microbial abundances in a single large-scale population-based cohort of 5,959 genotyped individuals with matched gut microbial metagenomes, and dietary and health records (prevalent and follow-up). We identified 567 independent SNP-taxon associations. Variants at the LCT locus associated with Bifidobacterium and other taxa, but they differed according to dairy intake. Furthermore, levels of Faecalicatena lactaris associated with ABO, and suggested preferential utilization of secreted blood antigens as energy source in the gut. Enterococcus faecalis levels associated with variants in the MED13L locus, which has been linked to colorectal cancer. Mendelian randomization analysis indicated a potential causal effect of Morganella on major depressive disorder, consistent with observational incident disease analysis. Overall, we identify and characterize the intricate nature of host-microbiota interactions and their association with disease.


Subject(s)
Diet , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Genetic Variation , Host Microbial Interactions , Polymorphism, Single Nucleotide , ABO Blood-Group System/genetics , Bifidobacterium/physiology , Clostridiales/physiology , Cohort Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/microbiology , Dietary Fiber , Enterococcus faecalis/physiology , Gastrointestinal Microbiome/genetics , Genome-Wide Association Study , Humans , Lactase/genetics , Mediator Complex/genetics , Mendelian Randomization Analysis , Metagenome , Morganella/physiology
8.
Nutrients ; 13(11)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34835987

ABSTRACT

Recent studies have suggested that gut-brain axis may be one of the mechanisms of major depression disorder (MDD). The current study aimed to investigate the effects of Lactobacillus plantarum PS128 (PS128) on psychophysiology in patients with MDD. We recruited 11 patients with MDD and gave them PS128 for 8 weeks. We compared depression symptoms, serum markers of inflammation and gut permeability, and gut microbiota before and after 8-week intervention and also explored the correlations among symptoms, biomarkers, and gut microbiota. After 8-week PS128 intervention, scores of Hamilton Depression Rating Scale-17 and Depression and Somatic symptoms Scale significantly decreased. Serum levels of high sensitivity c-reactive protein, interluekin-6, and tumor necrosis factor-α, zonulin and intestinal fatty acid binding protein, and the composition of gut microbiota did not significantly change after 8-week PS128 intervention. However, we found changes of some genera were correlated with changes of symptoms and biomarkers. In conclusion, this is an open trial with small sample size and has several limitations. The results need to be verified by randomized, double-blind, placebo-controlled trial with larger sample size.


Subject(s)
Depressive Disorder, Major/microbiology , Depressive Disorder, Major/psychology , Lactobacillus plantarum/physiology , Adult , Aged , Biodiversity , Biomarkers/blood , Depressive Disorder, Major/blood , Gastrointestinal Microbiome , Humans , Middle Aged , Phylogeny , Psychophysiology , Surveys and Questionnaires , Young Adult
9.
J Nerv Ment Dis ; 209(9): 691-692, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34448737

ABSTRACT

ABSTRACT: Burgeoning body of evidence from neuroscience is pouring in highlighting a potential association between gut microbiota with the pathophysiology of depression and anxiety. Manipulation of gut microbiota may be then useful to decode this role and to provide novel therapeutics for major depressive disorder (MDD), developing microbiota-related biomarkers to stratify patients at risk and to delineate more homogeneous biotypes of MDD.


Subject(s)
Depressive Disorder, Major/microbiology , Gastrointestinal Microbiome , Host Microbial Interactions , Animals , Anxiety/microbiology , Biomarkers , Depression/microbiology , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/diet therapy , Humans , Mice , Prebiotics , Probiotics/therapeutic use , Rats
10.
Sci Rep ; 11(1): 14918, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290352

ABSTRACT

The alterations in the gut microbiota have been reported to be correlated with the development of depression. The purpose of this study was to investigate the changes of intestinal microbiota in depressed patients after antidepressant treatment. We recruited 30 MDD patients (MDD group) and 30 healthy controls (control group). The MDD group received individualized treatment with escitalopram at a maximum dose of 20 mg/day. After depressive symptoms improved to a HAMD scale score > 50%, a fecal sample was collected again and used as the follow-up group. The differences of gut microbiota between patients and controls, the characteristics of gut microbiota under treatment and the potential differences in metabolic functions were thus investigated. The Firmicutes/Bacteroidetes ratio was significantly different within three groups, and the ratio of follow-up group was significantly lower than those of the other two groups. Alpha diversity was significantly higher in MDD group than those of the other groups, and the alpha diversity was not significantly different between control and follow-up groups. The beta diversity of some patients resembled participants in the control group. The metabolic function of gut microbiota after treatment was still different from that of the control group. This study suggests that the intestinal flora of depressed patients has a tendency to return to normal under escitalopram treatment.


Subject(s)
Citalopram/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/microbiology , Gastrointestinal Microbiome/drug effects , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Citalopram/administration & dosage , Citalopram/pharmacology , Depressive Disorder, Major/etiology , Female , Humans , Male , Middle Aged , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/pharmacology
11.
Nutrients ; 13(5)2021 May 20.
Article in English | MEDLINE | ID: mdl-34065187

ABSTRACT

The field of probiotic has been exponentially expanding over the recent decades with a more therapeutic-centered research. Probiotics mediated microbiota modulation within the microbiota-gut-brain axis (MGBA) have been proven to be beneficial in various health domains through pre-clinical and clinical studies. In the context of mental health, although probiotic research is still in its infancy stage, the promising role and potential of probiotics in various mental disorders demonstrated via in-vivo and in-vitro studies have laid a strong foundation for translating preclinical models to humans. The exploration of the therapeutic role and potential of probiotics in major depressive disorder (MDD) is an extremely noteworthy field of research. The possible etio-pathological mechanisms of depression involving inflammation, neurotransmitters, the hypothalamic-pituitary-adrenal (HPA) axis and epigenetic mechanisms potentially benefit from probiotic intervention. Probiotics, both as an adjunct to antidepressants or a stand-alone intervention, have a beneficial role and potential in mitigating anti-depressive effects, and confers some advantages compared to conventional treatments of depression using anti-depressants.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/microbiology , Depressive Disorder, Major/therapy , Probiotics/therapeutic use , Epigenesis, Genetic , Humans , Hypothalamo-Hypophyseal System/microbiology , Inflammation
12.
J Adv Res ; 30: 27-38, 2021 05.
Article in English | MEDLINE | ID: mdl-34026284

ABSTRACT

Introduction: Major depressive disorder is caused by gene-environment interactions, and the host microbiome has been recognized as an important environmental factor. However, the underlying mechanisms of the host-microbiota interactions that lead to depression are complex and remain poorly understood. Objectives: The present study aimed to explore the possible mechanisms underlying gut microbiota dysbiosis-induced depressive-like behaviors. Methods: We used high-performance liquid chromatography-tandem mass spectrometry to analyze alterations in the hippocampal lysine acetylome and succinylome in male mice that had received gut microbiota from fecal samples of either patients with major depressive disorder or healthy controls. This was followed by bioinformatic analyses. Results: A total of 315 acetylation sites on 223 proteins and 624 succinylation sites on 494 proteins were differentially expressed in the gut microbiota-dysbiosis mice. The significantly acetylated proteins were primarily associated with carbon metabolism disruption and gene transcription suppression, while the synaptic vesicle cycle and protein translation were the most significantly altered functions for succinylated proteins. Additionally, our findings suggest that gut microbiota dysbiosis disturbs mitochondria-mediated biological processes and the MAPK signaling pathway through crosstalk between acetylation and succinylation on relevant proteins. Conclusions: This is the first study to demonstrate modifications in acetylation and succinylation in gut microbiota-dysbiosis mice. Our findings provide new avenues for exploring the pathogenesis of gut microbiota dysbiosis-related depression, and highlight potential targets for depression treatment.


Subject(s)
Depressive Disorder, Major/metabolism , Dysbiosis/metabolism , Gastrointestinal Microbiome , Hippocampus/metabolism , Lysine/metabolism , Proteome/metabolism , Acetylation , Animals , Chromatography, High Pressure Liquid/methods , Depression/metabolism , Depression/microbiology , Depressive Disorder, Major/microbiology , Dysbiosis/microbiology , Fecal Microbiota Transplantation/methods , Gene-Environment Interaction , Humans , Male , Mice , Protein Processing, Post-Translational , Succinic Acid/metabolism , Tandem Mass Spectrometry/methods
13.
J Psychiatry Neurosci ; 46(3): E358-E368, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34008933

ABSTRACT

Background: The microbiota interacts with the brain through the gut-brain axis, and a distinct dysbiosis may lead to major depressive episodes. Bacteria can pass through the gut barrier and be found in the blood. Using a multiomic approach, we investigated whether a distinct blood microbiome and metabolome was associated with major depressive episodes, and how it was modulated by treatment. Methods: In this case-control multiomic study, we analyzed the blood microbiome composition, inferred bacterial functions and metabolomic profile of 56 patients experiencing a current major depressive episode and 56 matched healthy controls, before and after treatment, using 16S rDNA sequencing and liquid chromatography coupled to tandem mass spectrometry. Results: The baseline blood microbiome in patients with a major depressive episode was distinct from that of healthy controls (patients with a major depressive episode had a higher proportion of Janthinobacterium and lower levels of Neisseria) and changed after antidepressant treatment. Predicted microbiome functions confirmed by metabolomic profiling showed that patients who were experiencing a major depressive episode had alterations in the cyanoamino acid pathway at baseline. High baseline levels of Firmicutes and low proportions of Bosea and Tetrasphaera were associated with response to antidepressant treatment. Based on inferred baseline metagenomic profiles, bacterial pathways that were significantly associated with treatment response were related to xenobiotics, amino acids, and lipid and carbohydrate metabolism, including tryptophan and drug metabolism. Metabolomic analyses showed that plasma tryptophan levels are independently associated with response to antidepressant treatment. Limitations: Our study has some limitations, including a lack of information on blood microbiome origin and the lack of a validation cohort to confirm our results. Conclusion: Patients with depression have a distinct blood microbiome and metabolomic signature that changes after treatment. Dysbiosis could be a new therapeutic target and prognostic tool for the treatment of patients who are experiencing a major depressive episode.


Subject(s)
Antidepressive Agents/therapeutic use , Blood/microbiology , Brain-Gut Axis/drug effects , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/microbiology , Dysbiosis/microbiology , Metabolome/drug effects , Microbiota/drug effects , Adult , Antidepressive Agents/pharmacology , Bacteria/classification , Bacteria/drug effects , Blood/drug effects , Carbohydrate Metabolism/drug effects , Case-Control Studies , Depressive Disorder, Major/blood , Depressive Disorder, Major/complications , Dysbiosis/blood , Dysbiosis/complications , Dysbiosis/metabolism , Female , Gastrointestinal Microbiome/drug effects , Humans , Lipid Metabolism/drug effects , Male
14.
Schizophr Res ; 234: 51-57, 2021 08.
Article in English | MEDLINE | ID: mdl-32334937

ABSTRACT

It is being increasingly recognized that human mucosal surfaces are not sterile but are colonized with microorganisms collectively known as the microbiome. The microbiome can alter brain functioning in humans and animals by way of a series of interactions operative in the brain-immune-gut interactome. We characterized the oropharyngeal microbiome in 316 individuals, including 121 with schizophrenia, 62 with mania, 48 with major depressive disorder, and 85 controls without a psychiatric disorder. We found that the oropharyngeal microflora of individuals with schizophrenia and individuals with mania differed from controls in composition and abundance as measured by the weighted UniFrac distance (both p < .003 adjusted for covariates and multiple comparisons). This measure in individuals with major depressive disorder did not differ from that of controls. We also identified five bacterial taxa which differed among the diagnostic groups. Three of the taxa, Neisseria subflava, Weeksellaceae, and Prevotella, were decreased in individuals with schizophrenia or mania as compared to controls, while Streptococci was increased in these groups. One taxa, Schlegelella, was only found in individuals with mania. Neisseria subflava was also positively associated with cognitive functioning as measured by the Repeatable Battery for the Assessment of Neuropsychological Status. There were no taxa significantly altered in individuals with major depression. Individuals with schizophrenia and mania have altered compositions of the oropharyngeal microbiome. An understanding of the biology of the microbiome and its effect on the brain might lead to new insights into the pathogenesis, and ultimately, the prevention and treatment of these disorders.


Subject(s)
Depressive Disorder, Major , Mania , Microbiota , Oropharynx/microbiology , Schizophrenia , Comamonadaceae , Depressive Disorder, Major/microbiology , Flavobacteriaceae , Humans , Mania/microbiology , Neisseria , Prevotella , Schizophrenia/microbiology , Streptococcus
15.
Nutrients ; 14(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35010912

ABSTRACT

A growing number of studies in rodents indicate a connection between the intestinal microbiota and the brain, but comprehensive human data is scarce. Here, we systematically reviewed human studies examining the connection between the intestinal microbiota and major depressive and bipolar disorder. In this review we discuss various changes in bacterial abundance, particularly on low taxonomic levels, in terms of a connection with the pathophysiology of major depressive and bipolar disorder, their use as a diagnostic and treatment response parameter, their health-promoting potential, as well as novel adjunctive treatment options. The diversity of the intestinal microbiota is mostly decreased in depressed subjects. A consistent elevation of phylum Actinobacteria, family Bifidobacteriaceae, and genus Bacteroides, and a reduction of family Ruminococcaceae, genus Faecalibacterium, and genus Roseburia was reported. Probiotics containing Bifidobacterium and/or Lactobacillus spp. seemed to improve depressive symptoms, and novel approaches with different probiotics and synbiotics showed promising results. Comparing twin studies, we report here that already with an elevated risk of developing depression, microbial changes towards a "depression-like" microbiota were found. Overall, these findings highlight the importance of the microbiota and the necessity for a better understanding of its changes contributing to depressive symptoms, potentially leading to new approaches to alleviate depressive symptoms via alterations of the gut microbiota.


Subject(s)
Bipolar Disorder/microbiology , Depressive Disorder, Major/microbiology , Gastrointestinal Microbiome , Adult , Animals , Bacteroides , Bifidobacterium , Bipolar Disorder/physiopathology , Bipolar Disorder/therapy , Brain-Gut Axis , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/therapy , Faecalibacterium , Female , Humans , Lactobacillus , Male , Middle Aged , Probiotics/therapeutic use , Synbiotics/administration & dosage , Young Adult
16.
Mol Neurobiol ; 57(10): 4269-4295, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32700250

ABSTRACT

There is robust evidence that major depression (MDD) is accompanied by a low-grade activation of the immune-inflammatory response system, which is involved in the pathophysiology of this disorder. It is also becoming apparent that glia cells are in reciprocal communication with neurons, orchestrate various neuromodulatory, homeostatic, metabolic, and immune mechanisms, and have a crucial role in neuroinflammatory mechanisms in MDD. Those cells mediate the central nervous system (CNS) response to systemic inflammation and psychological stress, but at the same time, they may be an origin of the inflammatory response in the CNS. The sources of activation of the inflammatory response in MDD are immense; however, in recent years, it is becoming increasingly evident that the gastrointestinal tract with gut-associated lymphoid tissue (GALT) and increased intestinal permeability to bacterial LPS and food-derived antigens contribute to activation of low-grade inflammatory response with subsequent psychiatric manifestations. Furthermore, an excessive permeability to gut-derived antigenic material may lead to subsequent autoimmunities which are also known to be comorbid with MDD. In this review, we discuss fascinating interactions between the gastrointestinal tract, increased intestinal permeability, intestinal microbiota, and glia-neuron cross talk, and their roles in the pathogenesis of the inflammatory hypothesis of MDD. To emphasize those crucial intercommunications for the brain functions, we propose the term of microbiota-gut-immune-glia (MGIG) axis.


Subject(s)
Depressive Disorder, Major/immunology , Depressive Disorder, Major/microbiology , Gastrointestinal Microbiome/immunology , Neuroglia/pathology , Animals , Autoimmunity , Depressive Disorder, Major/pathology , Humans , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neurons/pathology
17.
Mol Psychiatry ; 25(11): 2759-2772, 2020 11.
Article in English | MEDLINE | ID: mdl-32332994

ABSTRACT

The gut microbiota are being called the human "second brain," as they play a key role in the regulation of the central nervous system (CNS). Recent findings provide strong evidence for the presence of bidirectional communication networks between the gut microbiota and the CNS, and such crosstalk has been correlated with alterations in major depressive disorder (MDD) and other psychiatric disorders. Further, germ-free animal models have been used to investigate the effect of the microbiota on MDD and other psychiatric disorders, which have greatly expanded our knowledge of the role of the microbiota in the etiology of MDD and promoted causality studies of this psychiatric disorder and others as well. In this review, we first introduce the methodological approaches used for microbiota research and then provide an overview of current research progress on the modulatory function and composition of the gut microbiota in MDD and the therapeutic effect of probiotics that has been gained using data from human studies as well as animal experiments. Future research should focus on identification and characterization of specific bacterial strains involved in MDD with the hope of applying these findings in the prevention and treatment of MDD.


Subject(s)
Depression/microbiology , Depressive Disorder, Major/microbiology , Gastrointestinal Microbiome , Animals , Depression/diet therapy , Depressive Disorder, Major/diet therapy , Germ-Free Life , Humans , Probiotics/therapeutic use
19.
Nutrients ; 12(4)2020 04 08.
Article in English | MEDLINE | ID: mdl-32276499

ABSTRACT

BACKGROUND AND SIGNIFICANCE: There is a need to develop new hypothesis-driven treatment for both both major depression (MD) and schizophrenia in which the risk of depression is 5 times higher than the general population. Major depression has been also associated with poor illness outcomes including pain, metabolic disturbances, and less adherence. Conventional antidepressants are partly effective, and 44% of the subjects remain unremitted under treatment. Improving MD treatment efficacy is thus needed to improve the SZ prognosis. Microbiota-orientated treatments are currently one of the most promising tracks. METHOD: This work is a systematic review synthetizing data of arguments to develop microbiota-orientated treatments (including fecal microbiota transplantation (FMT)) in major depression and schizophrenia. RESULTS: The effectiveness of probiotic administration in MD constitutes a strong evidence for developing microbiota-orientated treatments. Probiotics have yielded medium-to-large significant effects on depressive symptoms, but it is still unclear if the effect is maintained following probiotic discontinuation. Several factors may limit MD improvement when using probiotics, including the small number of bacterial strains administered in probiotic complementary agents, as well as the presence of a disturbed gut microbiota that probably limits the probiotics' impact. FMT is a safe technique enabling to improve microbiota in several gut disorders. The benefit/risk ratio of FMT has been discussed and has been recently improved by capsule administration. CONCLUSION: Cleaning up the gut microbiota by transplanting a totally new human gut microbiota in one shot, which is referred to as FMT, is likely to strongly improve the efficacy of microbiota-orientated treatments in MD and schizophrenia and maintain the effect over time. This hypothesis should be tested in future clinical trials.


Subject(s)
Biological Therapy/methods , Depressive Disorder, Major/microbiology , Depressive Disorder, Major/therapy , Schizophrenia/microbiology , Schizophrenia/therapy , Adult , Fecal Microbiota Transplantation , Female , Humans , Male , Microbiota , Middle Aged , Probiotics/therapeutic use , Treatment Outcome , Young Adult
20.
Harv Rev Psychiatry ; 28(1): 26-39, 2020.
Article in English | MEDLINE | ID: mdl-31913980

ABSTRACT

Microorganisms can be found in virtually any environment. In humans, the largest collection of microorganisms is found in the gut ecosystem. The adult gut microbiome consists of more genes than its human host and typically spans more than 60 genera from across the taxonomic tree. In addition, the gut contains the largest number of neurons in the body, after the brain. In recent years, it has become clear that the gut microbiome is in communication with the brain, through the gut-brain axis. A growing body of literature shows that the gut microbiome plays a shaping role in a variety of psychiatric disorders, including major depressive disorder (MDD). In this review, the interplay between the microbiome and MDD is discussed in three facets. First, we discuss factors that affect the onset/development of MDD that also greatly impinge on the composition of the gut microbiota-especially diet and stressful life events. We then examine the interplay between the microbiota and MDD. We examine evidence suggesting that the microbiota is altered in MDD, and we discuss why the microbiota should be considered during MDD treatment. Finally, we look toward the future and examine how the microbiota might become a therapeutic target for MDD. This review is intended to introduce those familiar with the neurological and psychiatric aspects of MDD to the microbiome and its potential role in the disorder. Although research is in its very early days, with much yet to be the understood, the microbiome is offering new avenues for developing potentially novel strategies for managing MDD.


Subject(s)
Depressive Disorder, Major/metabolism , Depressive Disorder, Major/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Animals , Brain/metabolism , Brain/microbiology , Depressive Disorder, Major/diet therapy , Gastrointestinal Tract/physiology , Humans , Probiotics/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...