Scores of anxiety and depression are associated with changes in the composition of the oral microbiota and related metabolites
We applied a multi-omics approach to reveal molecular mechanisms underlying the microbiome-oral-brain axis interplay in anxiety and depression. We first assessed the relationships of oral bacterial composition and anxiety and depression using the Hamilton Anxiety Scale (HAMA) [13] and seventeen items version of Hamilton Depression Scale (HAMD-17) [14] in patients with health (n=70, HAMD: 0-7, HAMA < 7) and anxiety and depression (ANDP, n=87, HAMD > 7, HAMA > 20). Pan/Core species analysis is used to describe the changes in total species and core species with the increase of sample size. The results showed that the number of total ASV increased and then gradually flattened out and the number of shared ASV decreased and then gradually flattened out, suggesting that the sequencing sample size is sufficient (Figure S1A). There were no significant differences in the oral microbial community richness (Sobs, Chaos and Ace index) and diversity (Shannon index) between the two groups (Figure S1B). Then, the principal co-ordinates analysis (PCoA) was applied to reveal the overall bacterial phenotypes of beta-diversity. PCoA showed oral microbiota structure between health and anxiety and depression patients were distinct (Fig. 1A). Typing analysis on genus level showed that two distinct clusters: type I (g_Streptococcus) and type II (g_Pseudomonas), in which type II focus on the oral microbiomes sample of anxiety and depression patients (Fig. 1B). The composition of oral microbiota differed between the two groups (Fig. 1C, Figure S1C), Pseudomonas was significantly increased, while Leptotrichia and Solobacterium were significantly decreased in ANDP group compared with health group (Fig. 1D). Circos maps of communities show the relationship between samples and species, reflecting the proportion of dominant species in each group. Largely, Circos figures demonstrated that the dominant species in ANDP group is genus Pseudomonas (Fig. 1E). Compositional analysis by LefSe revealed that oral swabs of healthy controls were enriched with the genus Leptotrichia and Solobacterium, family Erysipelotrichaceae and ASV130 and so on, and ASV384 was predominant microbiota in ANDP group (Fig. 1F).
To reveal metabolic phenotypes related to the oral microbiome in anxiety and depression, we performed metabolic profiling of the saliva from health and anxiety and depression patients. The Orthogonal Partial Least Squares Discriminant Analysis (OPLS-DA) score plot showed distinct clusters of the saliva metabolites between the ANDP and health control groups (Fig. 1G), suggesting that the metabolic patterns were reprogrammed in ANDP patients. The Venn diagrams displayed the difference between each group, exhibiting specific 17 metabolites in ANDP and 21 metabolites in the health control group, respectively (Fig. 1H). A total of 87 significantly changed metabolites between the two groups samples were identified of which 24 showed a downward trend, and 63 showed an increasing trend (Fig. 1I). Briefly, top 32 metabolites (VIP > 2, p < 0.05) were exhibited (Figure S1D), including Eicosapentaenoic Acid, amino acids, bile acid, steroids, fatty acids and other classified metabolites. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that 20 altered metabolites-associated metabolic pathways, including primary bile acid biosynthesis, glycine, serine and threonine metabolism, fatty acid biosynthesis and tryptophan metabolism (Fig. 1J) were associated with mental disorders [7, 19-22].
Next, we explored the potential correlations of these differential oral microbiota and differential saliva metabolites. Overall, the results showed that there is a correlation with differential oral microbiota and metabolites of which Pseudomonas was positively correlated with 12-Ketodeoxycholic acid and negatively correlated with Eicosapentaenoic Acid (Fig. 1K), implying that the pathogenic oral microbial-derived metabolites induced by anxiety and depression may affect the occurrence and development of anxiety and depression. Taken together, these data suggest that mental disorders were accompanied by oral microbial dysbiosis and altered metabolites.
Differential oral and intestinal microbiota disorders in CRS mice
Behavioral tests were performed after 4 weeks of CRS model establishment. The immobility time for the FST and TST was remarkably lengthened and the center motion distance for the OFT significantly decreased in CRS mice as compared with control mice, suggesting obvious anxiety- and depression-like behaviors (Fig. 2A). Next, we analyzed the microbiota of oral swab and stool samples from mice with CRS and controls using 16S rRNA sequencing. The α-diversity values including species richness (Sobs, Chaos and Ace index) and species diversity (Shannon index) were compared between the CRS and control groups. There were no significant differences in the oral and gut microbial community richness and diversity between the two groups (Figure S2A, B). PCoA showed that oral and gut microbiota structure the two groups all could be distinguished at the ASV level (Fig. 2B, C). The composition of oral and gut microbiota differed between the CRS and control groups (Fig. 2D, E). We found that Pseudomonas, Pasteurellaceae and Muribacter was higher, while Streptococcus was reduced in CRS group compared with control group at the genus level for oral microbiota (Fig. 2F). For gut microbiota, the relative abundance of Muribaculaceae and Saccharimonas was enriched in the CRS group relative to the control groups, whereas Lachnospiraceae and Desulfovibrionaceae was lower than control groups (Fig. 2G). LEfSe analysis showed that, compared to control groups, CRS group was characterized by enriched ASV belonging to the Phylum Proteobacteria and Actinobacteriota and the families Pasteurellaceae and Nocardiaceae for oral microbiota (Fig. 2H, I). For gut microbiota, LEfSe analysis showed that CRS group was characterized by enriched ASV belonging to the Phylum Bacteroidota and the families Muribaculaceae compared to control groups, and depleted ASV belonging to the Phylum Firmicutes and Desulfobacterota and the family Desulfovibrionaceae and Lachnospiraceae (Fig. 2J, K). Together, these results indicate that CRS induced oral and gut microbiota disorders, and have significantly different microbial signatures in oral and gut, suggesting that differential shaping effect of oral and intestinal microbiota on depression.
Saliva microbiota transplantation in germ-free mice recapitulate the alteration of oral microbiota in CRS exposed conventional mice
To confirm the direct role of altered oral microbiota induced by CRS on depression, we transferred oral swabs from CRS mice and controls to GF mice, referred to as “GF-Stress group” and “GF-control group” respectively. Then, we performed 16S rRNA sequencing on oral swabs in GF mice. Similar to the conventional mice model, there were no found significant differences in the oral microbial community richness (Sobs, Chaos and Ace index) and diversity (Shannon index) between the GF-Stress group and GF-control group (Figure S3A). PCoA showed that oral microbiota structure the two groups could be separated completely at the ASV level (Fig. 3A). At the genus level, the Venn diagrams displayed that specific 183 genera in GF-Stress group and 25 genera in GF-control group, and shared 45 genera (Fig. 3B). The composition of oral microbiota differed between the two groups (Fig. 3C, Figure S3B). Clearly, the dominant species in GF-Stress group is genus Pseudomonas and Lactobacillus (Fig. 3D). We further tested consistence of microbiota alterations between the two mice models. And we found that bacteria with increased abundance in CRS-exposed mice compared with control mice were consistently increased in GF-Stress mice compared with GF-Control mice, whereas bacteria with decreased abundance in CRS-exposed mice were consistently reduced in GF-Stress mice compared with GF-Control mice (Fig. 3E). Together, these results indicate that the transplantation of oral swabs from CRS mice could induces oral microbiota alterations, and oral swabs microbiota transplantation in GF mice recapitulate the alteration of oral microbiota in CRS exposed conventional mice.
An emotional impairment is transferred to germ-free mice through the oral microbiota
To investigate whether oral microbiome works in the progression of depression independent of gut microbes, we inoculated respectively oral microbiota from mice with health and well-established CRS model into GF mice (Fig. 3F). GF mice colonized with oral microbiome from CRS model developed more obvious anxiety- and depression-like behaviors than GF mice that inoculated with oral microbiome from control mice (Fig. 3G-I). Specifically, the immobility time for the FST (Fig. 3G) and TST (Fig. 3H) was obviously increased and the center motion distance for the OFT (Fig. 3I) markedly shortened in CRS mice as compared with control mice. These results indicate that the presence of oral microbiome contributes to the development of emotional impairment.
CRS-altered oral microbiota induces microbial-derived metabolites alteration in germ-free mice
To determine alterations in serum metabolites after oral microbiota transplantation from CRS mice, we performed liquid chromatography tandem mass spectrometry (LC-MS/MS) on serum in GF mice from GF-Stress group and GF-control group. The Principal Component Analysis (PCA) and OPLS-DA score plot showed a complete separation of the serum metabolites between the GF-Stress group and GF-control group (Fig. 4A). The Venn diagrams displayed the difference between each group, exhibiting specific 7 metabolites in GF-Stress group (Fig. 4B). A total of 201 significantly changed metabolites between the two groups samples were identified of which 68 showed a downward trend, and 133 showed an increasing trend (Fig. 4C). Top 30 metabolites were exhibited (Fig. 6D), including Eicosapentaenoic Acid, amino acids, bile acid, steroids, fatty acids and other classified metabolites. Importantly, Eicosapentaenoic Acid was significantly decreased in GF-Stress group compared with GF-control group (Fig. 4D). KEGG enrichment analysis showed that 20 altered metabolites-associated metabolic pathways, including Glycine, serine and threonine metabolism, Steroid hormone biosynthesis, Butanoate metabolism, Fatty acid biosynthesis, primary bile acid biosynthesis, Alanine, aspartate and glutamate metabolism and GABAergic synapse (Fig. 4E). These data indicate that oral microbiota induced by CRS promotes the alteration of serum metabolites in GF mice.
The potential relationships between serum metabolites and differential oral microbiota induced by CRS were evaluated. In general, Procrustes analysis showed significant correlation between differential oral microbiota and serum metabolites (Fig. 4F). In particular, Eicosapentaenoic Acid was negatively correlated with Pseudomonas, Romboutsia, Pasteurellaceae and Muribacter, but positively correlated with Streptococcus, Turicibacter and Rodentibacter. 4-Phenyl-3-buten-2-ol, 7-Ketocholesterol and 12-Ketodeoxycholic acid was positively correlated with Pseudomonas, Rhodococcus and Muribacter, but negatively correlated with Turicibacter and Streptococcus (Fig. 4G). Together, these results indicate that oral microbiota from CRS models causes oral microbial-derived metabolites alteration in GF mice, implying that the pathogenic oral microbial-derived metabolites could enter the blood circulation system and play an important role in psychiatric illnesses.
Oral microbiota and microbial-derived metabolites induced by CRS could directly impact brain function in germ-free mice
The blood-brain barrier controls substance transport between the blood and the brain, and its permeability is reflected tight junction proteins [23]. We assessed the mRNA expression of ZO-1, occludin, and claudin-1 by RT-PCR in hypothalamus and hippocampus regions of GF-Stress group and GF-control group. Significantly lower expression of ZO-1, occludin, and claudin-1 was observed in GF-Stress group compared with GF-control group (Fig. 5A). Western blot and immunofluorescence staining used to detect the protein expression of ZO-1 in the frontal cortex, and the results were consistent with RT-PCR (Fig. 5B, C). Moreover, the frontal cortex in the GF-Stress group displayed lower levels of 5-hydroxytryptamine (5-HT) and higher levels of norepinephrine (Fig. 5D), implying the alteration of neurotransmitter. These results indicate that oral microbiota from CRS models causes the increasing of blood-brain barrier permeability and neurotransmitter imbalance in GF mice, which shows the presence of oral-brain axis in depression.
To further clarify the potential roles of the microbiota-oral-brain axis, correlations between the representative values of behavioral changes, levels of neurotransmitter in the frontal cortex and changes in oral microbial-derived metabolites were assessed. As depicted in Fig. 4F and G, the oral microbiota that significantly differed between groups were highly correlated with the expression of serum metabolites. Additionally, microbial-derived metabolites were significantly correlated with neurotransmitter in the frontal cortex and depression-like behavioral changes (Fig. 5E). Together, these results indicate that oral microbiota can exert significant roles in mental disorders by oral- brain axis.
Ectopic colonization of oral microbiome induced by CRS in the intestine impaired gut barrier function in germ-free mice
Studies showed that oral bacteria do not colonize the distal gut in a healthy state [24]. In this study, PCoA showed that the microbiota compositions of the saliva were clearly separated from that of the stool in the CRS groups (Fig. 6A). However, we also found the same bacteria in the saliva and stool samples in CRS states (Fig. 6B), suggesting that we may warrant further investigation for an underlying pathology. To gain the underlying mechanisms of mental disorders induced by oral microflora from CRS mice, we performed 16S rRNA sequencing on stool in GF mice from GF-Stress group and GF-control group. The data showed that the α-diversity values including gut microbial species richness (Sobs, Chaos and Ace index) and species diversity (Shannon index) were significant differences between the GF-Stress group and GF-control group (Fig. 6C). PCoA showed that gut microbiota structure the two groups could be separated completely at the ASV level (Fig. 6D). The composition of gut microbiota differed between the two groups (Fig. 6E). From the gut community Circos diagram, we observed that the dominant genera in GF-Stress group were Muribaculaceae, Lactobacillus, Alistipes, Bacteroides, Erysipelatoclostridium, Helicobacter, Turicibacter and Monoglobus (Fig. 6F). Among them, the abundance of Alistipes, Erysipelatoclostridium, Helicobacter, Turicibacter and Monoglobus was significantly increased in GF-Stress group (Fig. 6G). LEfSe analysis showed that GF-Stress group was characterized by enriched ASV belonging to the Phylum Bacteroidota and Campilobacterota and the families Rikenellaceae and Helicobacteraceae, compared to GF-control group, and depleted ASV belonging to the Phylum Firmicutes, Bacteroidota and Desulfobacterota and the family Erysipelotrichaceae, Prevotellaceae and Desulfovibrionaceae (Fig. 6H). These data suggest that oral microbiota induced by CRS colonized the intestinal tract and induced gut microbiota dysbiosis.
Hematoxylin and eosin (HE) staining from colon revealed that the epithelium layer was loose, the structure of intestinal crypts was abnormal, the goblet cells were reduced and infiltration of inflammatory cells in GF-Stress group compared with the GF-control group (Fig. 6I). This data indicates that the intestinal morphological characteristics were altered in GF-Stress group, and thus the intestinal function may also be altered by oral microbiota. Next, we studied the effects of oral microbiota on the mRNA and protein levels of intestinal mucosa tight binding protein genes (ZO-1, occludin and claudin-1). The RT-PCR results demonstrated that, compared to GF-control group, the mRNA expression levels of ZO-1, occludin and claudin-1 were significantly downregulated in GF-Stress group (Fig. 6J). The colon tissue immunofluorescence results demonstrated that the protein expression of ZO-1 was markedly reduced in GF-Stress group compared to GF-control group (Fig. 6K). These data indicate that oral microbiota induced by CRS harm the gut barrier function and increase intestinal permeability in GF mice.
Pseudomonas aeruginosa intervention and Eicosapentaenoic Acid supplementation change depression-like behavior association with microbial translocation in antibiotic-treated mice
To strengthen the effect of the microbiota and microbial-derived metabolites that significantly differed between groups on anxiety and depression, we also used antibiotic-treated mice that were orally treated with a neomycin, metronidazole, vancomycin and ampicillin in the drinking water. The Pseudomonas aeruginosa (PA) and Eicosapentaenoic Acid (EPA) was administrated to antibiotic-treated mice by oral gavage lasting 3 weeks. Then, the behavioral phenotypes were evaluated (Fig. 7A). Compared with CRS controls, the immobility time for the FST and TST was obviously reduced and the center motion distance for the OFT markedly extended in the mice treated with EPA (Fig. 7B-D), suggesting EPA supplementation confers protection against depression-like states in mice. Conversely, PA intervention exacerbated anxiety- and depression-like states (Fig. 7E-G). Overall, these results suggest that oral microbiota and microbial-derived metabolites affect anxiety- and depression-like states in antibiotic-treated mice.