Abstract
Background:
Comorbid anxiety and depression are common and are associated with greater disease burden than either alone. Our recent efforts have identified an association between gut microbiota dysfunction and severity of anxiety and depression. In this follow-up, we applied Differential Co-Expression Analysis (DiffCoEx) to identify potential gut microbiota biomarker(s) candidates of treatment resistance among psychiatric inpatients.
Methods:
In a sample of convenience, 100 psychiatric inpatients provided clinical data at admission and discharge; fecal samples were collected early during the hospitalization. Whole genome shotgun sequencing methods were used to process samples. DiffCoEx was used to identify clusters of microbial features significantly different based on treatment resistance status. Once overlapping features were identified, a knowledge-mining tool was used to review the literature using a list of microbial species/pathways and a select number of medical subject headlines (MeSH) terms relevant for depression, anxiety, and brain-gut-axis dysregulation. Network analysis used overlapping features to identify microbial interactions that could impact treatment resistance.
Results:
DiffCoEx analyzed 10,403 bacterial features: 43/44 microbial features associated with depression treatment resistance overlapped with 43/114 microbial features associated with anxiety treatment resistance. Network analysis resulted in 8 biological interactions between 16 bacterial species. Clostridium perfringens evidenced the highest connection strength (0.95). Erysipelotrichaceae bacterium 6_1_45 has been most widely examined, is associated with inflammation and dysbiosis, but has not been associated with depression or anxiety.
Conclusion:
DiffCoEx potentially identified gut bacteria biomarker candidates of depression and anxiety treatment-resistance. Future efforts in psychiatric microbiology should examine the mechanistic relationship of identified pro-inflammatory species, potentially contributing to a biomarker-based algorithm for treatment resistance.
Keywords: Depression, Anxiety, Metagenomics Gastrointestinal microbiome, Biomarkers, Brain-gut Axis
1. Introduction
Anxiety and depressive disorders are among the most common psychiatric illnesses globally. In a 12- month period, 10–20% of all adult primary care visits occur during an anxiety or depressive episode, and 50% of those patients will have a comorbid disorder (Hirschfeld, 2001). Comorbidity is often the rule rather than the exception (Kessler et al., 1994). Approximately two-thirds of patients with depression have a comorbid anxiety disorder and one-third or more of patients with panic disorder or GAD also have depression (Clayton, 1990; Gorman, 1996; Klerman, 1990). STAR*D demonstrated symptom comorbidity with 53% of patients with MDD having anxiety and anxious depression (Fava et al., 2004; Kalin, 2020). Phenomenologically, both are regarded as internalizing disorders with shared symptoms that include: negative affect, impaired emotion regulation, psychomotor disturbance, and neuroticism (Sindermann et al., 2021). Neuroimaging studies consistently implicate structural and functional alterations in the orbitofrontal cortex/middle frontal cortex and in prefrontal limbic pathways across both anxiety and depression (Kalin, 2020; Sindermann et al., 2021; Van Ameringen, 2021). Individuals with co-occurring anxiety and depression have more severe symptoms and poorer prognosis than individuals with either disorder alone (Gorman, 1996; Van Ameringen, 2021). Genetic studies indicate substantial shared variance across depression and anxiety (Kendler et al., 2007). Generalized anxiety disorder (GAD) and major depressive disorder (MDD) are leading contributors to global disability, accounting for one-third to one-half of the global economic burden of mental illness (Franklin et al., 2017; Sindermann et al., 2021; Wittchen, 2002). Comorbidity is also associated with greater social and functional impairment, reduced quality of life, and increased risk for suicide(Van Ameringen, 2021; Zhou et al., 2017). In the last decade, there has been emerging evidence indicating a strong correlation between anxiety/depression and the gut microbiome (Hai-yin Jiang et al., 2018).
The gut microbiome is a complex community that functions to maintain host physiology. Gut microbiota can influence the brain, via the bidirectional communication of the Gut-Brain Axis (GBA) – the central nervous system (CNS), the intestines, and the gut microbiota (Rutsch et al., 2020). This bidirectional communication network has many roles that are essential for homeostasis and physiological functioning, including the modulation of the immune system (Barko et al., 2018). Under normal conditions, the gut microbiota defends the host against pathogens by stimulating the immune system, leading to low-level inflammation (Galland, 2014; Wu and Wu, 2012). Conversely, the composition of the gut microbiota can be influenced by the presence of chronic inflammation present in a series of diseases (Ni et al., 2017; Wang et al., 2020). Studies have shown that psychiatric symptom-atology, such as those found in depression and anxiety, are associated with an imbalance (dysbiosis) in the gut microbial community (Dickerson et al., 2017; Evans et al., 2017; Janssen, 2016; Haiyin Jiang et al., 2015). Dysbiosis of the gut microbiome can be a result of numerous factors including genetics, diet, and exposure to stress. Due to the chronic low-level inflammation, often present in a dysbiotic state (Rutsch et al., 2020), a gut microbial community in dysbiosis can lead to increased permeability of the intestines and the blood-brain barrier, both of which are associated with various psychiatric disorders (Kelly et al., 2015). Previous studies have identified an association between key microbial taxa and severity of depression and anxiety (A. Madan et al., 2020; Valles-Colomer et al., 2019). This identification has led to a significant push to identify bacterial taxa that can be used as predictive biomarkers for various psychiatric disorders (Lv et al., 2017; Misiak et al., 2020). However, the complex nature of the gut microbiome makes it difficult to understand whether the association of depression and anxiety with individual bacterial species is independent or reflective of a broader compositional shift of clusters of bacteria.
Despite the complex nature of the gut microbiome, there exist several strategies to analyze such data to identify microbiome biomarkers relevant to psychiatric conditions. Homeostasis in healthy, complex systems, such as full transcriptome or metabolomes, is maintained by preserving numerous molecular programs that involve yet unappreciated interactions. Conversely, transition to a disease state or exposure to external stressors changes in lock-step complex molecular networks. An unbiased and agnostic methodology to comprehensively identify such changes is Differential Co-Expression Analysis (DiffCoEx). Initially developed to identify gene markers for cancer datasets, DiffCoEx has been used to identify biomarkers for contrasting pairs of clinical conditions in multiple fields. For instance, DiffCoEx has been used to identify non-oncogene addiction (NOA) genes for 15 different cancer types (Hjaltelin et al., 2019), to recognize differing genes between bronchiolitis and emphysema phenotypes of Chronic Obstructive Pulmonary Disease (COPD) (Qin et al., 2019), as well as to investigate the impact of temperature on the metabolome of Drosophila melanogaster (Hariharan et al., 2014). As a proven method to identify pattern changes caused by pairs of contrasting clinical and experimental conditions (Tesson et al., 2010), DiffCoEx analysis can be particularly helpful in identifying bacterial biomarkers that differentiate between psychiatric treatment outcomes based on fecal samples of patients. In this study, we applied DiffCoEx network analysis to identify potential gut microbiota biomarker of depression and anxiety treatment resistance in a cohort of psychiatric inpatients.
2. Methods
2.1. Participants & setting
From January 1, 2015 to December 31, 2016, one hundred (N = 100) adult psychiatric inpatients at a freestanding, nonprofit facility in the Southwestern United States provided self-collected fecal swabs shortly after admission. Patients at the facility received individualized psychiatric and medical care over a prolonged length of stay (average length of stay = 49.7 ± 14.5 days). Psychiatric care included an individualized combination of comprehensive medical evaluation; optimization of psychotropic pharmaceutical drugs (Madan et al., 2015); personal, group, and skills-based psychotherapy; psychoeducation; and family work. All psychiatric and medical mediations occurred within the context of 24-h nursing supervision and a therapeutic milieu. The prolonged length of stay is anomalous for inpatient psychiatric care in the United States but have been established as necessary to achieve the demonstrated progress and remission from anxiety and depression symptomatology (Fowler et al., 2017) as well as multiple cross-cutting clinical features of psychopathology (Allen et al., 2017).
2.2. Clinical procedures
Data were gathered as part of the hospital’s Adult Outcomes Project to evaluate treatment response (Allen et al., 2009; Fowler et al., 2013) as well as a broader overlapping study aimed at determining biomarkers of psychiatric illness and treatment response (Alok Madan et al., 2017; Sharp et al., 2016). All baseline psychiatric measures were collected within 72 h of admission. The primary outcome assessments (including depression and anxiety severity) were repeated every 14 days during the hospitalization as well as at discharge. A hospital-wide web survey on laptops was used to conduct all assessments. Given pharmacologic agents’ potential to alter the gut microbiota, data related to drugs administered at the hospital were extracted from the internal pharmacy database. A finite number of medication classes were chosen in an a priori manner to minimize the possibility of spurious findings. The medication classes were antibiotics, probiotics, antidepressants, anti-psychotics, and opioids.
2.3. Clinical measures
Demographics, history of psychiatric care, and health-related information were assessed through a standardized patient information survey (Allen et al., 2009; Fowler et al., 2013). Research adaptations of the Structured Clinical Interview for DSM-IV Disorders (SCID-I/II) were employed to diagnose psychiatric disorders, such as personality disorders. Researchers at the master’s level conducted the SCID-I (First et al., 1997) and SCID-II (First et al., 2002) interviews. History of suicidality was measured using the Columbia Suicide Severity Rating Scale (CSSRS) (Madan et al., 2016; Posner et al., 2011). Trauma history was assessed with the Stressful Life Events Screening Questionnaire (SLESQ) (Allen et al., 2015; Goodman et al., 1998). The primary patient-reported outcome measures utilized in this study were the Patient Health Questionnaire – Generalized Anxiety Disorder screener (GAD-7) (Löwe et al., 2008) and the Patient Health Questionnaire – 9 (PHQ-9) (Kroenke and Spitzer, 2002); they were used to evaluate depression and anxiety severity, respectively. These measures also were selected as primary outcome measures in light of our recent report (Fowler et al., 2017) that demonstrated these measures’ sensitivity to alter among patients with SMI receiving multimodal inpatient treatment at the study psychiatric facility. Finally, the Acceptance and Action Questionnaire-II (AAQ-II) (Bond et al., 2011) was selected for these analyses given previous assessments that have demonstrated the AAQ-II to abate depression and anxiety treatment response in the patient cohort (Fowler et al., 2017). All the previously mentioned measures have sound psychometric properties and are commonly used in clinical and research settings.
2.3.1. Microbiota-related procedures
Self-collected fecal swabs from study participants shortly after admission to the hospital (20.1 ± 12.8 days) were initially stored in a 80 °C freezer. Bacterial genomic DNA then were extracted from fecal samples using the MO BIO PowerSoil DNA Isolation Kit. The 16S rDNA V4 region was PCR-amplified and sequenced using an Illumina MiSeq system. The amplification primers used (515F-806R) contain MiSeq adapters and single-end barcodes supporting sample pooling and PCR products sequencing. The 16S rRNA pipeline uses both phylogenetic and alignment-based methods. Read pairs were first demultiplexed using unique molecular barcodes, then merged using USEARCH v7.0.1090. The UPARSE algorithm was employed to cluster 16S rDNA sequences into Observed Operational Taxonomic Units (OTUs) at the 97% similarity cutoff value. Detected OTUs were further annotated by mapping to an optimized SILVA Database version that contained only the 16S v4 region. Abundance was quantified by mapping demultiplexed reads to OTUs. Using a minimum rarefaction depth of 4815, no samples were omitted due to insufficient reads. The final OTU table was then used for alpha-diversity, beta-diversity, and phylogenetic analysis.
Genomic bacterial DNA (gDNA) extraction methods were refined for Whole Genome Shotgun Sequencing (WGS) to improve the bacterial DNA yield and reduce the background amplification (The Human Microbiome Project Consortium, 2012a, 2012b). Paired-end sequencing reads were filtered for low quality sequences and for Illumina phix sequences. Subsequently, Illumina adapters were removed using bbduk (BBMap version 37.58; Bushnell et al., 2017); resulting sequences were mapped to the human hg38 reference database using bowtie2 v.2.3.4.3 (Langmead and Salzberg, 2012) completely and with high-stringency to remove sequence-level host contamination. Taxonomic profiles were inferred using MetaPhlAn2 (Segata et al., 2012), and functional profiling of the microbial community was done using HUMAnN2 (Franzosa et al., 2018). The final output files were represented in biom ormat (McDonald et al., 2012).
2.4. DiffCoEx and network analysis
DiffCoEx was used to identify modules of microbial features, whose abundance differed significantly due to treatment resistance states of depression and anxiety (Tesson et al., 2010). Following the construction of the dissimilarity matrices based on pairwise correlations of bacterial and pathway abundance data, DiffCoEx used hierarchical clustering to identify clusters of microbial features that were significantly different based on presence of treatment resistance as indicated by distinct co-expression patterns. We incorporated permutation testing in the DiffCoEx algorithm to assess the significance of within and module-to-module co-expression changes associated with each pair of differing phenotypes. More specifically, for each module and between each pair of modules of microbial features, dispersion values were calculated using 1000 permutations to assign P-values for within-module, and module-to-module differentiation. For each pair, only those modules with within-module dispersion values smaller than 0.05 (or < 50/1000) were selected.
DiffCoEx algorithm’s outputs revealed an overwhelming amount of overlap between microbial biomarkers identified for depression treatment resistance and anxiety treatment resistance. Thus, common outputs of both phenotypes were used for further network analyses. Using hierarchical clustering within the WGCNA framework (Langfelder and Horvath, 2008), we generated an undirected, weighted correlation network of these biomarkers. With a weight cut-off of 0.3, we visualized the network using the Cytoscape (Shannon et al., 2003) software.
2.5. Pubmed text-based knowledge search
To extract manuscripts that contained species and MESH terms chosen in an a prior manner, we used the PubTator Central text mining platform (Wei et al., 2019) to search PubMed and PMC databases (through February 17, 2022). The search was not limited to a publication timeframe, however, reviews, meta-analyses, and publications not written in English were excluded.
2.6. Ethics
The present study adheres to the guidelines and ethical principles outlined in the Declaration of Helsinki. Participants provided written informed consent following a full explanation of all procedures. Through informed consent, study participants attested to the voluntary nature of their participation. Study design was approved by the Institutional Review Board (IRB) at Baylor College of Medicine.
3. Results
3.1. Patient characteristics
In this study, the final cohort (n = 100) consisted primarily of young, Caucasian adults. At admission, the majority were unemployed, never married, and many had significant trauma histories, as well as some degree of suicidal ideation. Individuals presented with multiple Axis I disorders, and many additionally had high levels of functional impairment and service utilization consistent with current definitions of severe mental illness (SMI) (Ronald C Kessler et al., 2010). Most individuals in this cohort had extensive histories of outpatient and inpatient psychiatric treatment before this study (Table 1). At discharge, 54% of individuals had a PHQ-9 score of >5, signifying treatment resistant depression, and 43% of individuals had treatment resistant anxiety, with a GAD-7 score of >5. A small population of individuals exclusively had treatment resistant depression (5%), or exclusively had treatment resistant anxiety (3%).
Table 1.
Mean (SD) | N (%) | |
---|---|---|
Age, years | 37.0 (13.8) | |
Sex, female | 47 (47.0) | |
Ethnicity, White | 90 (90.0) | |
Single/Never Married | 41 (41.0) | |
Some college or greater | 70 (70.0) | |
Unemployed (past 30 days) | 51 (51.0) | |
Total Axis I Disorders | 2.7 (1.4) | |
Total Axis II Disorders | 0.6 (0.8) | |
Suicidal ideation, past month | 65 (65.0) | |
Suicide attempt(s), lifetime | 0.9 (1.7) | 35 (35.0) |
Substance Use Disorders | 50 (50.0) | |
Major Depressive Disorders | 74 (74.0) | |
Bipolar Spectrum | 14 (14.0) | |
Anxiety Spectrum | 72 (72.0) | |
Psychotic Spectrum | 2 (2.0) | |
Borderline Personality Disorder | 18 (18.0) | |
Traumatic life event(s), lifetime | 2.4 (2.2) | 81 (81.0) |
Any Personality Disorder | 38 (38.0) | |
Outpatient therapists (lifetime) | 3.8 (2.6) | |
Psychopharmacologists (lifetime) | 2.9 (2.2) | |
Hospitalizations for acute psychiatric care (lifetime) | 1.0 (1.4) | |
Hospitalizations for extended psychiatric care (lifetime) | 1.0 (2.0) |
3.2. Modules of bacterial features are differentially co-expressed between depression treatment outcomes
We used DiffCoEx to detect patterns of differential co-expression between the depression treatment responder group (n = 55 patients) and depression treatment resistant group (n = 45 patients). Of the 10,403 bacterial features analyzed by DiffCoEx, 105 were assigned to one of three modules, which were arbitrarily assigned colors (Fig. 1A). Features in all three modules identified in DiffCoEx show significant differential co-expression in the binary variable (depression treatment resistance = 1 vs 0) (dispersion statistics p value<0.05). The turquoise module, containing 44 bacterial features, indicated a stronger unidirectional differential correlation in microbial relative abundance associated with treatment response; specifically, the 44 features were strongly correlated only in the depression treatment resistant patient group. A less distinctly unidirectional correlation difference was seen in the blue and red modules, containing 42 and 19 bacterial features respectively; interestingly, for both modules the depression treatment resistant group showed a loss of correlation compared to the depression treatment response group.
3.3. Modules of bacterial features are differentially co-expressed between anxiety treatment outcomes
We used DiffCoEx to detect patterns of differential co-expression between the anxiety treatment responder group (n = 54 patients) and anxiety treatment resistant group (n = 46 patients). DiffCoEx analysis revealed 157 out of 10,403 bacterial features that were assigned to one of two modules (Fig. 1B). The turquoise module, containing 114 bacterial features, indicated an increased correlation of microbial relative abundance in patients that had treatment resistant anxiety (Anxiety_treatment resistance = 1). A less distinctly unidirectional correlation difference was seen in the blue module, containing 43 bacterial features, with a higher correlation of the 43 features in the anxiety treatment responder patient group.
3.3.1. Detection of a commonly induced microbial features modules in depression and anxiety treatment resistance
Due to the high comorbidity between depression and anxiety, we determined common and concordant microbial features associated with both anxiety and depression treatment resistance. We focused on common features in the turquoise modules of depression and anxiety inferred by DiffCoEx due to their concordant direction of correlation difference. 43 out of the 44 microbial features that were associated with treatment resistant depression overlapped with 43 out 114 microbial features associated with treatment resistant anxiety. Interestingly, the same 43 microbial features showed decreased co-expression in both the depression treatment resistant and the anxiety treatment resistant groups. The integration of differential co-expression results for these two comorbidities has enabled us to prioritize 43/10,403 microbial features we want to investigate in the future.
3.4. Literature search of microbial features
Once the overlapping microbial features between treatment resistance in both depression and anxiety were identified, we employed a knowledge-mining tool, Pubtator, to determine the relevance and importance of our findings. Specifically, Pubtator was used to comprehensively explores PubMed abstracts and PMC full-text articles using the 43 significant features associated with resistance in both anxiety and depression. We searched for articles mentioning any of the 43 features and a list of previously determined MESH terms associated with dysbiosis, inflammation, short chain fatty acids, and with different presentations of depression and anxiety. Conducting text mining allows us to corroborate some of our findings to present DiffCoEx as an effective tool for biomarker identification. The results of the Pubator search revealed inflammation, as well as dysbiosis, were the most annotated mesh terms. Our literature search results further showed that 42% of the microbial features identified with our methods have >15 annotations related to any mesh terms, corroborating our findings. The remaining 58% microbial features that had fewer than 15 annotations related to any mesh term are potentially newly identified biomarker candidates for psychiatric disorders, pointing to a direction for future research.
3.5. Network analysis
Cytoscape (Shannon et al., 2003), a software used to visually build and analyze biological networks, in conjunction with the weighted gene co-expression network analysis (Langfelder and Horvath, 2008), was used to identify the biological interactions in the gut microbiome that could impact depression and treatment outcomes. The overlapping 43 microbial features previously identified in the DiffCoEx analysis were qualified to be included in the network analysis. At a feature correlation weight cutoff of 0.3, eight network connections were identified (Fig. 2). Of those connections, three of them had a weight >0.7, indicating high connection strength. Clostridium perfringens and Prevotella bivia had the strongest connection strength (0.95). Facklamia hominis and Anaeroccocus prevoti (0.79) had the second strongest connection, with a connection strength of 0.79, andErysipelotrichaceae bacterium 6_1_45 and Clostridium innocuum had the third strongest connection, with a connection strength of 0.71. Supporting data for the microbial features network analysis are shown in Table 2.
Table 2.
Network Edge | Weight |
---|---|
Clostridium perfringens: Prevotella bivia | 0.95 |
Anaerococcus hydrogenalis: Facklamia hominis | 0.79 |
Clostridium innocuum: Erysipelotrichaceae bacterium_6_1_45 | 0.71 |
Marvinbryantia formatexigens: Synergistes_sp_3_1_syn1 | 0.51 |
Facklamia hominis: Prevotella bergensis | 0.38 |
Anaerococcus hydrogenalis: Anaerococcus prevotii | 0.38 |
Anaerococcus hydrogenalis: Prevotella bergensis | 0.31 |
Anaerococcus prevotii: Facklamia hominis | 0.31 |
The results of the literature search were further truncated to only highlight the microbial features that had a connection strength of at least 0.3 (Table 3 and Table 4).
Table 3.
Bacterial Species | Dysbiosis | Inflammation | Short Chain Fatty Acids | Animal Models & Depression | In vitro & Depression | Research Subjects & depression | Dysbiosis & Depression | Inflammation & Depression | Short Chain Fatty Acids & Depression | In vitro & Anxiety Disorders | Animal Models & Anxiety Disorders | Research Subjects & Anxiety Disorders | Dysbiosis & Anxiety disorders | Inflammation & Anxiety Disorders | Short Chain Fatty Acids & Anxiety Disorders |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Enterobacteriaceae bacterium_9_2_54FAA | 627 | 6350 | 362 | 35 | 157 | 0 | 7 | 31 | 5 | 0 | 0 | 0 | 1 | 2 | 3 |
Bacteroides sp3_1_40A | 723 | 1177 | 495 | 5 | 5 | 0 | 9 | 8 | 4 | 0 | 0 | 0 | 1 | 0 | 1 |
Klebsiella pneumoniae | 41 | 548 | 13 | 2 | 5 | 0 | 0 | 4 | 0 | 0 | 0 | 0 | 0 | 0 | |
Lachnospiraceae bacterium_4_1_37FAA | 312 | 323 | 203 | 2 | 1 | 0 | 10 | 6 | 4 | 0 | 0 | 0 | 1 | 0 | 0 |
Lachnospiraceae bacterium_5_1_57FAA | 312 | 323 | 203 | 2 | 1 | 0 | 10 | 6 | 4 | 0 | 0 | 0 | 1 | 0 | 0 |
Lachnospiraceae bacterium_6_1_63FAA | 312 | 323 | 202 | 2 | 1 | 0 | 10 | 6 | 4 | 0 | 0 | 0 | 1 | 0 | 0 |
Fusobacterium nucleatum | 103 | 370 | 14 | 0 | 0 | 0 | 0 | 1 | 1 | 0 | 0 | 0 | 0 | 0 | 0 |
Streptococcus agalactiae | 15 | 256 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Clostridium perfringens | 33 | 153 | 63 | 2 | 6 | 0 | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Erysipelotrichaceae bacterium_3_153 | 70 | 67 | 29 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Erysipelotrichaceae bacterium_6_145 | 70 | 67 | 29 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Enterococcus faecium | 27 | 77 | 14 | 0 | 1 | 0 | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Enterobacter cloacae | 7 | 41 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Peptostreptococcus anaerobius | 7 | 17 | 3 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Enterobacter aerogenes | 1 | 17 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Prevotella bivia | 8 | 14 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Citrobacter freundii | 1 | 13 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Acidaminococcus sp_D21 | 9 | 10 | 5 | 0 | 0 | 0 | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Bifidobacterium catenulatum | 4 | 4 | 2 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Parabacteroides goldsteinii | 3 | 10 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Clostridium scindens | 4 | 4 | 2 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Coprococcus eutactus | 3 | 3 | 2 | 0 | 0 | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Synergistes sp_3_1_syn1 | 1 | 2 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Blautia hydrogenotrophica | 0 | 1 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Catenibacterium mitsuokai | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Clostridium innocuum | 1 | 1 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Facklamia hominis | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Lachnospiraceae bacterium_1_1_57FAA | 1 | 1 | 1 | 0 | 0 | 0 | 1 | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 |
Marvinbryantia formatexigens | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Solibacillus silvestris | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Anaerococcus hydrogenalis | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Anaerococcus prevotii | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Bacteroides clarus | 1 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Eremococcus coleocola | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Prevotella bergensis | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Pseudomonas fragi | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Table 4.
Bacterial Pathways | Dysbiosis | Inflammation | Short Chain Fatty Acids | Animal Models & Depression | In vitro & Depression | Research Subjects & depression | Dysbiosis & Depression | Inflammation & Depression | Short Chain Fatty Acids & Depression | In vitro & Anxiety Disorders | Animal Models & Anxiety Disorders | Research Subjects & Anxiety Disorders | Dysbiosis & Anxiety disorders | Inflammation & Anxiety Disorders | Short Chain Fatty Acids & Anxiety Disorders |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
l-lysine biosynthesis I | 22 | 1691 | 39 | 12 | 92 | 0 | 1 | 8 | 0 | 0 | 1 | 0 | 0 | 0 | 1 |
Pentose phosphate pathway | 7 | 175 | 8 | 1 | 6 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
aerobic respiration I (cytochromec) | 0 | 66 | 0 | 4 | 4 | 0 | 0 | 1 | 0 | 0 | 0 | 0 | 0 | 0 | 1 |
Rubisco shunt | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
L-1,2-propanediol degradation | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
superpathway of acetyl-CoA biosynthesis | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
superpathway of purine nucleotide salvage | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
4. Discussion
The present study identified co-expression patterns of gut bacteria and pathways associated with depression and anxiety treatment resistance. As hypothesized there was a strong and concordant directional co-expression pattern for both. 43 out of the 44 bacterial features associated with treatment resistant depression were also associated with treatment resistant anxiety. A comprehensive search of the existing literature using inputs associated with microbial features, psychiatric features, and broadly brain-gut-axis dysregulation features identified taxa and pathways previously implicated in brain-gut dysfunction as well novel features that are uniquely associated with the study sample. Moreover, key biological interactions among bacterial species from the overlapping features were discovered through the construction of an undirected, weighted correlation network analysis. Findings from this study narrow the scope of bacterial features associated with brain-gut dysfunction from >10,000 to fewer than 50 – features that may enable more focused research efforts in the future.
Notable taxa from this study to highlight include: Erysipelotrichaceae bacterium 6_1_45 (most related to existing literature) as well as Clostridium perfringens and Prevotella bivia (which demonstrated the highest connection strength from our network analysis). These taxa are associated with inflammation, globally as well as specifically related to histotoxic and intestinal infections (Freedman et al., 2015; Li et al., 2013; Shrestha et al., 2018) and pelvic inflammatory disease and bacterial vaginosis (Aroutcheva et al., 2008; Gilbert et al., 2019; Mikamo et al., 2014). The role of these taxa individually and interactively in the context of anxiety and depression should be explored in greater detail in future studies. Dysbiosis is causally linked to atypical immune responses, which are often accompanied by an upregulated production of inflammatory cytokines (Schirmer et al., 2016). Previous studies have found an association between an increase in pro-inflammatory cytokines and the pathophysiology of depression and anxiety (Alshammari et al., 2020; Carlessi et al., 2021; Dantzer et al., 2008; Nobis et al., 2020; Schirmer et al., 2016). Notably, toxins present in Clostridium perfringens can cause a dramatic increase in cytokine production, leading to cytokine cascades during sepsis (Hifumi, 2020; Takehara et al., 2019). Thus, examining the mechanistic relationship of these inflammatory species in depression and anxiety is a promising future direction.
Our work quantifies and identifies disruption in the correlation of different microbiota genera. As such, it poses multiple interesting questions: which mechanisms are responsible for such disruptions and how can one potentially intervene? To strengthen a positive correlation between two genera that may be lacking or limited among patients with treatment resistance, a potential intervention would be probiotic supplementation with both genera, informed by preclinical studies in animal models of depression and/or anxiety. Additionally, future efforts may build upon models that use host transcriptomes. LINCS/Connectivity Map (Subramanian et al., 2017) is one such approach that represents a fertile resource to determine potential mechanisms at the level of gene suppression or over-expression, or to stratify over 2300 chemical compounds based on their positive or negative correlation with a novel gene signature. Similar well-powered resources have not been developed for microbiome disruptions. However, with the continuing commoditization of 16S sequencing, and lowering prices for WGS metagenome sequencing, we expect that such resources will eventually be developed and can be used to inform mechanistic studies.
A notable strength of this study is our analytic approach: DiffCoEx provides a more nuanced bioinformatic approach relative to other techniques used to analyze microbiota data. Differential abundance analysis of metagenomics data that employ standard analytics such as ANOVA and Kruskal-Wallis, and subsequent feature selection and machine learning methods including LefSe, RandomForest, Support Vector Machines, are extensively employed in the current literature (e.g., Fung et al., 2021; Estaki et al., 2020) - including our previous reports (Saulnier et al., 2011; Riehle et al., 2012; Aagaard et al., 2012; Madan et al., 2020; Thompson et al., 2021; El Saie et al., 2022). The focus of this study, however, was to explore the network-based analysis of gut microbiome in conjunction with key clinical responses to psychiatric care. The aforementioned approaches do not allow for network-based analysis. A seminal network analysis methodology is weighted correlation network analysis (WGCNA); it has been used extensively in microbiome analysis, in disease models spanning cancer (e.g., Park et al., 2019; Vernocchi et al., 2020) to dietary interventions in humans and farm animals (e.g., Murga-Garrido et al., 2021; Xie et al., 2021). An alternative differential co-expression method to DiffCoEx is Differential Gene Correlation Analysis (DGCA), which identifies and reports differential co-expression at the level of individual pairs of features. DGCA has been successfully employed in analysis of skin (Meisel et al., 2018) and gut (Zou et al., 2022) microbiomes. Unlike WGCNA, which finds modules of correlated features, and DGCA, which identifies differential correlation at the level of individual pairs, DiffCoEx uniquely identifies modules of microbiome species differentially co-expressed; DiffCoEx also provides a robust statistical framework for the final modules reported, using permutation testing.
The relatively large number of donors and the systematic assessment of treatment response in a controlled physical environment are strengths of the present study. Nonetheless, several limitations must be acknowledged and should be addressed in future efforts. The study sample includes psychiatric inpatients with considerable comorbidity, including personality disorders that may be associated with health behaviors (e.g., substance misuse) that influence the structure and function of the gut microbiota. A more diagnostically homogenous patient population may have discordant findings. Though the study sample had a restricted number of dietary options during their hospitalization, they were free to choose from a menu of options for meals during their hospitalization. This variability in dietary intake was not systematically tracked and may have confounded the observed data. Future research should better account for diet, given that there is increasing evidence of the influence of diet on depression and anxiety severity and response to treatment (Adan et al., 2019). The longitudinal measures of treatment outcome were exclusively patient reported with significant potential for under/overreporting of distress. Additionally, results from this study are based on a single fecal sample collected early in treatment. Future research should include serial collection of additionally performance-based outcome measures and fecal samples and to allow for an examination of potential changes (or lack thereof) in the gut microbiota in response to treatment.
5. Conclusion
In conclusion, a novel, data-reduction analytic strategy was used to examine microbial features for the identification of unique bacterial co-expression patterns associated with treatment outcomes for depression, treatment outcomes for anxiety, and common and concordant features associated with both depression and anxiety treatment resistance. This study only begins to examine the co-abundant expression patterns of gut bacteria and pathways in subjects with depression and anxiety. Further research is needed to advance our understanding of the mechanistic role of the gut microbiome in the development, maintenance, and treatment response of psychiatric disorders. This study advances the nascent literature of the complex interdependence between the gut and neural functioning, wherein greater interdisciplinary study of psychiatric microbiology integrates fields of psychiatry and microbiology as well as advanced analytics. Psychiatric microbiology promises a more nuanced understanding of interdependent human functions with the possibility of directly and indirectly manipulating bacterial structure and function to treat psychiatric symptomatology in the future.
Acknowledgments
This research was partially supported by Houston Methodist Foundation, The Menninger Clinic Foundation, Baylor College of Medicine’s Alkek Center for Metagenomics and Microbiome Research. AM is the John S. Dunn Foundation Distinguished Centennial Clinical Academic Scholar in Behavioral Health at Houston Methodist. BLW is the C. James and Carole Walter Looke Presidential Distinguished Centennial at Houston Methodist. FC and CC were partially supported by The Cancer Prevention Institute of Texas (CPRIT) [RP170005, RP210227], NIH/NCI P30 shared resource grant [CA125123], NIH/NIEHS center grants [P30 ES030285] and [P42 ES027725], and NIH/NIMHD [P50MD015496]. Study sponsors were not involved in any aspect of the research. The authors have no other conflicts of interest to report.
Footnotes
Ethical statement
The present study adheres to the guidelines and ethical principles outlined in the Declaration of Helsinki. Participants provided written informed consent following a full explanation of all procedures. Through informed consent, study participants attested to the voluntary nature of their participation. Study design was approved by the Institutional Review Board (IRB) at Baylor College of Medicine.
CRediT authorship contribution statement
Dominique S. Thompson: Methodology, Formal analysis, Writing – original draft, Writing – review & editing, Visualization. Chenlian Fu: Software, Formal analysis, Writing – review & editing, Visualization. Tanmay Gandhi: Software, Formal analysis, Visualization. J. Christopher Fowler: Investigation, Resources, Data curation, Writing – review & editing, Project administration, Supervision, Funding acquisition. B. Christopher Frueh: Investigation, Resources, Data curation, Writing – review & editing, Project administration, Supervision, Funding acquisition. Benjamin L. Weinstein: Investigation, Resources, Data curation, Writing – review & editing, Project administration, Supervision, Funding acquisition. Joseph Petrosino: Resources, Data curation, Writing – review & editing, Supervision. Julia K. Hadden: Data curation, Writing – original draft, Writing – review & editing. Marianne Carlson: Data curation, Writing – original draft, Writing – review & editing. Cristian Coarfa: Conceptualization, Methodology, Validation, Formal analysis, Writing – original draft, Writing – review & editing, Visualization, Supervision. Alok Madan: Conceptualization, Methodology, Investigation, Resources, Data curation, Writing – review & editing, Project administration, Supervision, Funding acquisition.
Declaration of Competing Interest
The authors have no other conflicts of interest to report.
References
- Aagaard K, Riehle K, Ma J, Segata N, Mistretta TA, Coarfa C, Raza S, Rosenbaum S, Van den Veyver I, Milosavljevic A, Gevers D, Huttenhower C, Petrosino J, Versalovic J, 2012. A metagenomic approach to characterization of the vaginal microbiome signature in pregnancy. PLoS One 7 (6), e36466. 10.1371/journal.pone.0036466 (Epub 2012 Jun 13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adan RAH, van der Beek EM, Buitelaar JK, Cryan JF, Hebebrand J, Higgs S, Schellekens H, Dickson SL, 2019. Nutritional psychiatry: towards improving mental health by what you eat. Eur. Neuropsychopharmacol 29 (12), 1321‒1332. 10.1016/j.euroneuro.2019.10.011. [DOI] [PubMed] [Google Scholar]
- Allen JG, Frueh BC, Ellis TE, Latini DM, Mahoney JS, Oldham JM, Sharp C, Wallin L, 2009. Integrating outcomes assessment and research into clinical care in inpatient adult psychiatric treatment. Bull. Menn. Clin 73 (4), 259‒295. 10.1521/bumc.2009.73.4.259. [DOI] [PubMed] [Google Scholar]
- Allen JG, Madan A, Fowler JC, 2015. Reliability and validity of the stressful life events screening questionnaire among inpatients with severe neuropsychiatric illness. Bull. Menn. Clin 79 (3), 187‒202. 10.1521/bumc.2015.79.3.187. [DOI] [PubMed] [Google Scholar]
- Allen JG, Christopher Fowler J, Madan A, Ellis TE, Oldham JM, Christopher Frueh B, 2017. Discovering the impact of psychotherapeutic hospital treatment for adults with serious mental illness. Bull. Menn. Clin 81 (1), 1‒38. 10.1521/bumc.2017.81.1.1. [DOI] [PubMed] [Google Scholar]
- Alshammari MA, Khan MR, Majid Mahmood H, Alshehri AO, Alasmari FF, Alqahtani FM, Alasmari AF, Alsharari SD, Alhossan A, Ahmad SF, Nadeem A, Alshammari TK, 2020. Systemic TNF-α blockade attenuates anxiety and depressive-like behaviors in db/db mice through downregulation of inflammatory signaling in peripheral immune cells. Saudi Pharmaceutical Journal : SPJ : The Official Publication of the Saudi Pharmaceutical Society 28 (5), 621‒629. 10.1016/j.jsps.2020.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aroutcheva A, Ling Z, Faro S, 2008. Prevotella bivia as a source of lipopolysaccharide in the vagina. Anaerobe 14 (5), 256‒260. 10.1016/j.anaerobe.2008.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barko PC, McMichael MA, Swanson KS, Williams DA, 2018. The gastrointestinal microbiome: a review. J. Vet. Intern. Med 32 (1), 9‒25. 10.1111/jvim.14875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bond FW, Hayes SC, Baer RA, Carpenter KM, Guenole N, Orcutt HK, Waltz T, Zettle RD, 2011. Preliminary psychometric properties of the acceptance and action questionnaire-II: a revised measure of psychological inflexibility and experiential avoidance. Behav. Ther 42 (4), 676‒688. 10.1016/j.beth.2011.03.007. [DOI] [PubMed] [Google Scholar]
- Bushnell B, Rood J, Singer E, 2017. BBMerge – Accurate paired shotgun read merging via overlap. PLoS ONE 12 (10), e0185056. 10.1371/journal.pone.0185056Capuco. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlessi AS, Borba LA, Zugno AI, Quevedo J, Réus GZ, 2021. Gut microbiota–brain axis in depression: the role of neuroinflammation. Eur. J. Neurosci 53 (1), 222‒235. 10.1111/ejn.14631. [DOI] [PubMed] [Google Scholar]
- Clayton P, 1990. The comorbidity factor: establishing the primary diagnosis in patients with mixed symptoms of anxiety and depression. J. Clin. Psychiatry 51 (Suppl), 35–39. https://www.semanticscholar.org/paper/The-comorbidity-factor%3A-establishing-the-primary-in-Clayton/d92c5ceb094e6a3ca96db054a2e13a5b2ca97bf1. [PubMed] [Google Scholar]
- Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW, 2008. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat. Rev. Neurosci 9 (1), 46–56. 10.1038/nrn2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dickerson F, Severance E, Yolken R, 2017. The microbiome, immunity, and schizophrenia and bipolar disorder. Brain Behav. Immun 62, 46–52. 10.1016/j.bbi.2016.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El Saie A, Fu C, Grimm SL, Robertson MJ, Hoffman K, Putluri V, Ambati CSR, Putluri N, Shivanna B, Coarfa C, Pammi M, 2022. Mar 25. Metabolome and microbiome multi-omics integration from a murine lung inflammation model of bronchopulmonary dysplasia. Pediatr. Res 10.1038/s41390-022-02002-1.Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Estaki M, Jiang L, Bokulich NA, McDonald D, González A, Kosciolek T, Martino C, Zhu Q, Birmingham A, Vázquez-Baeza Y, Dillon MR, Bolyen E, Caporaso JG, Knight R, 2020. QIIME 2 enables comprehensive end-to-end analysis of diverse microbiome data and comparative studies with publicly available data. Curr. Protoc. Bioinformatics 70, e100. 10.1002/cpbi.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans SJ, Bassis CM, Hein R, Assari S, Flowers SA, Kelly MB, Young VB, Ellingrod VE, McInnis MG, 2017. The gut microbiome composition associates with bipolar disorder and illness severity. J. Psychiatr. Res 87, 23–29. 10.1016/j.jpsychires.2016.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fava M, Alpert JE, Carmin CN, Wisniewski SR, Trivedi MH, Biggs MM, Shores-Wilson K, Morgan D, Schwartz T, Balasubramani GK, Rush AJ, 2004. Clinical correlates and symptom patterns of anxious depression among patients with major depressive disorder in STAR*D. Psychol. Med 34 (7), 1299‒1308. 10.1017/s0033291704002612. [DOI] [PubMed] [Google Scholar]
- First M, Gibbon M, Spitzer R, Benjamin L, 1997. 2Structured Clinical Interview for DSM-IV® Axis II Personality Disorders SCID-II. American Psychiatric Press. [Google Scholar]
- First M, Spitzer R, Gibbon M, Williams J, 2002. Structured Clinical Interview for DSM-IV-TR Axis I Disorders, Research Version, Patient Edition. New York State Psychiatric Institute. [Google Scholar]
- Fowler JC, Allen JG, Oldham JM, Frueh BC, 2013. Exposure to interpersonal trauma, attachment insecurity, and depression severity. J. Affect. Disord 149 (1–3), 313‒318. 10.1016/j.jad.2013.01.045. [DOI] [PubMed] [Google Scholar]
- Fowler JC, Clapp JD, Madan A, Allen JG, Frueh BC, Oldham JM, 2017. An open effectiveness trial of a multimodal inpatient treatment for depression and anxiety among adults with serious mental illness. Psychiatry 80 (1), 42–54. 10.1080/00332747.2016.1196072. [DOI] [PubMed] [Google Scholar]
- Franklin JC, Ribeiro JD, Fox KR, Bentley KH, Kleiman EM, Huang X, Musacchio KM, Jaroszewski AC, Chang BP, Nock MK, 2017. Risk factors for suicidal thoughts and behaviors: a meta-analysis of 50 years of research. Psychol. Bull 143 (2), 187‒232. 10.1037/bul0000084. [DOI] [PubMed] [Google Scholar]
- Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M, Weingart G, Lipson KS, Knight R, Caporaso JG, Segata N, Huttenhower C, 2018. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15 (11), 962‒968. 10.1038/s41592-018-0176-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Freedman JC, Theoret JR, Wisniewski JA, Uzal FA, Rood JI, McClane BA, 2015. Clostridium perfringens type A-E toxin plasmids. Res. Microbiol 166 (4), 264‒279. 10.1016/j.resmic.2014.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fung C, Rusling M, Lampeter T, Love C, Karim A, Bongiorno C, Yuan L-L, 2021. Automation of QIIME2 metagenomic analysis platform. Current Protocols 1, e254. 10.1002/cpz1.254. [DOI] [PubMed] [Google Scholar]
- Galland L, 2014. The gut microbiome and the brain. J. Med. Food 17 (12), 1261‒1272. 10.1089/jmf.2014.7000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilbert NM, Lewis WG, Li G, Sojka DK, Lubin JB, Lewis AL, 2019. Gardnerella vaginalis and Prevotella bivia trigger distinct and overlapping phenotypes in a mouse model of bacterial vaginosis. J. Infect. Dis 220 (7), 1099‒1108. 10.1093/infdis/jiy704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodman LA, Corcoran C, Turner K, Yuan N, Green BL, 1998. Assessing traumatic event exposure: general issues and preliminary findings for the stressful life events screening questionnaire. J. Trauma. Stress 11 (3), 521‒542. 10.1023/A:1024456713321. [DOI] [PubMed] [Google Scholar]
- Gorman JM, 1996. Comorbid depression and anxiety spectrum disorders. Depression and Anxiety 4 (4), 160‒168. [DOI] [PubMed] [Google Scholar]
- Hariharan R, Hoffman JM, Thomas AS, Soltow QA, Jones DP, Promislow D, 2014. Invariance and plasticity in the drosophila melanogastermetabolomic network in response to temperature. BMC Syst. Biol 8 (1), 139. 10.1186/s12918-014-0139-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hifumi T, 2020. Spontaneous non-traumatic Clostridium perfringens Sepsis. Jpn. J. Infect. Dis 73 (3), 177‒180. 10.7883/yoken.JJID.2019.382. [DOI] [PubMed] [Google Scholar]
- Hirschfeld RMA, 2001. The comorbidity of major depression and anxiety disorders: recognition and Management in Primary Care. Primary Care Companion to the Journal of Clinical Psychiatry 3 (6), 244‒254. 10.4088/pcc.v03n0609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hjaltelin JX, Izarzugaza JMG, Jensen LJ, Russo F, Westergaard D, Brunak S, 2019. Identification of hyper-rewired genomic stress non-oncogene addiction genes across 15 cancer types. Npj Systems Biology and Applications 5 (1), 27. 10.1038/s41540-019-0104-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janssen B, 2016. Placental tissue as a molecular ‘footprint’ of in utero exposure to fine particles: Set up of a birth cohort. http://hdl.handle.net/1942/21514.
- Jiang Haiyin, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J, Li L, Ruan B, 2015. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun 48, 186‒194. 10.1016/j.bbi.2015.03.016. [DOI] [PubMed] [Google Scholar]
- Jiang Hai-yin, Zhang X, Yu Z, Zhang Z, Deng M, Zhao J, Ruan B, 2018. Altered gut microbiota profile in patients with generalized anxiety disorder. J. Psychiatr. Res 104, 130‒136. 10.1016/J.JPSYCHIRES.2018.07.007. [DOI] [PubMed] [Google Scholar]
- Kalin NH, 2020. The critical relationship between anxiety and depression. Am. J. Psychiatry 177 (5), 365‒367. 10.1176/appi.ajp.2020.20030305. [DOI] [PubMed] [Google Scholar]
- Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP, 2015. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci 9, 392. 10.3389/fncel.2015.00392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kendler KS, Gardner CO, Gatz M, Pedersen NL, 2007. The sources of co-morbidity between major depression and generalized anxiety disorder in a Swedish national twin sample. Psychol. Med 37 (3), 453‒462. 10.1017/S0033291706009135. [DOI] [PubMed] [Google Scholar]
- Kessler RC, McGonagle KA, Zhao S, Nelson CB, Hughes M, Eshleman S, Wittchen HU, Kendler KS, 1994. Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbidity Survey. Arch. Gen. Psychiatry 51 (1), 8‒19. 10.1001/archpsyc.1994.03950010008002. [DOI] [PubMed] [Google Scholar]
- Kessler Ronald C., Green JG, Gruber MJ, Sampson NA, Bromet E, Cuitan M, Furukawa TA, Gureje O, Hinkov H, Hu C-Y, Lara C, Lee S, Mneimneh Z, Myer L, Oakley-Browne M, Posada-Villa J, Sagar R, Viana MC, Zaslavsky AM, 2010. Screening for serious mental illness in the general population with the K6 screening scale: results from the WHO world mental health (WMH) survey initiative. Int. J. Methods Psychiatr. Res 19 (S1), 4‒22. 10.1002/mpr.310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klerman GL, 1990. Depression and panic anxiety: the effect of depressive co-morbidity on response to drug treatment of patients with panic disorder and agoraphobia. J. Psychiatr. Res 24 (Suppl. 2), 27‒41. 10.1016/0022-3956(90)90033-m. [DOI] [PubMed] [Google Scholar]
- Kroenke K, Spitzer RL, 2002. The PHQ-9: a new depression diagnostic and severity measure. Psychiatr. Ann 32 (9), 509‒515. 10.3928/0048-5713-20020901-06. [DOI] [Google Scholar]
- Langfelder P, Horvath S, 2008. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559. 10.1186/1471-2105-9-559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langmead B, Salzberg SL, 2012. Fast gapped-read alignment with bowtie 2. Nat. Methods 9 (4), 357‒359. 10.1038/nmeth.1923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Adams V, Bannam TL, Miyamoto K, Garcia JP, Uzal FA, Rood JI, McClane BA, 2013. Toxin plasmids of Clostridium perfringens. Microbiology and Molecular Biology Reviews : MMBR 77 (2), 208‒233. 10.1128/MMBR.00062-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Löwe B, Decker O, Müller S, Brähler E, Schellberg D, Herzog W, Herzberg PY, 2008. Validation and standardization of the generalized anxiety disorder screener (GAD-7) in the general population. Med. Care 46 (3), 266‒274. 10.1097/MLR.0b013e318160d093 [DOI] [PubMed] [Google Scholar]
- Lv F, Chen S, Wang L, Jiang R, Tian H, Li J, Yao Y, Zhuo C, 2017. The role of microbiota in the pathogenesis of schizophrenia and major depressive disorder and the possibility of targeting microbiota as a treatment option. Oncotarget 8 (59), 100899‒100907. 10.18632/oncotarget.21284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madan Alok, Walker CR, Weinstein B, Fowler JC, 2015. Pharmacogenomics in practice: a case report of personalized inpatient psychiatric care. Pharmacogenomics 16 (5), 433‒439. 10.2217/pgs.15.9. [DOI] [PubMed] [Google Scholar]
- Madan Alok, Frueh BC, Allen JG, Ellis TE, Rufino KA, Oldham JM, Fowler JC, 2016. Psychometric reevaluation of the columbia-suicide severity rating scale: findings from a prospective, inpatient cohort of severely mentally ill adults. J. Clin. Psychiatry 77 (7), e867‒e873. 10.4088/JCP.15m10069. [DOI] [PubMed] [Google Scholar]
- Madan Alok, Fowler JC, Patriquin MA, Salas R, Baldwin PR, Velasquez KM, Viswanath H, Molfese DL, Sharp C, Allen JG, Hardesty S, Oldham JM, Frueh BC, 2017. A novel approach to identifying a neuroimaging biomarker for patients with serious mental illness. The Journal of Neuropsychiatry and Clinical Neurosciences 29 (3), 275‒283. 10.1176/appi.neuropsych.16090174. [DOI] [PubMed] [Google Scholar]
- Madan A, Thompson D, Fowler JC, Ajami NJ, Salas R, Frueh BC, Bradshaw MR, Weinstein BL, Oldham JM, Petrosino JF, 2020. The gut microbiota is associated with psychiatric symptom severity and treatment outcome among individuals with serious mental illness. J. Affect. Disord 264, 98‒106. 10.1016/j.jad.2019.12.020. [DOI] [PubMed] [Google Scholar]
- McDonald D, Clemente JC, Kuczynski J, Rideout JR, Stombaugh J, Wendel D, Wilke A,Huse S, Hufnagle J, Meyer F, Knight R, Caporaso JG, 2012. The Biological Observation Matrix (BIOM) format or: how I learned to stop worrying and love the ome-ome. GigaScience 1 (1). 10.1186/2047-217X-1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meisel JS, Sfyroera G, Bartow-McKenney C, Gimblet C, Bugayev J, Horwinski J, Kim B, Brestoff JR, Tyldsley AS, Zheng Q, Hodkinson BP, Artis D, Grice EA, 2018. Commensal microbiota modulate gene expression in the skin. Microbiome. 6 (1), 20. 10.1186/s40168-018-0404-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mikamo H, Iwasaku K, Yamagishi Y, Matsumizu M, Nagashima M, 2014. Efficacy and safety of intravenous azithromycin followed by oral azithromycin for the treatment of acute pelvic inflammatory disease and perihepatitis in Japanese women. Journal of Infection and Chemotherapy : Official Journal of the Japan Society of Chemotherapy 20 (7), 429‒435. 10.1016/j.jiac.2014.04.001. [DOI] [PubMed] [Google Scholar]
- Misiak B, Łoniewski I, Marlicz W, Frydecka D, Szulc A, Rudzki L, Samochowiec J, 2020. The HPA axis dysregulation in severe mental illness: can we shift the blame to gut microbiota? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 102. 10.1016/J.PNPBP.2020.109951. [DOI] [PubMed] [Google Scholar]
- Murga-Garrido SM, Hong Q, Cross TL, Hutchison ER, Han J, Thomas SP, Vivas EI, Denu J, Ceschin DG, Tang ZZ, Rey FE, 2021. Gut microbiome variation modulates the effects of dietary fiber on host metabolism. Microbiome. 9 (1), 117. 10.1186/s40168-021-01061-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ni J, Wu GD, Albenberg L, Tomov VT, 2017. Gut microbiota and IBD: causation or correlation? Nat. Rev. Gastroenterol. Hepatol 14 (10), 573‒584. 10.1038/nrgastro.2017.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nobis A, Zalewski D, Waszkiewicz N, 2020. Peripheral markers of depression. Journal of. Clinical Medicine 9 (12). 10.3390/jcm9123793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park CH, Lee JG, Lee AR, Eun CS, Han DS, 2019. Network construction of gastric microbiome and organization of microbial modules associated with gastric carcinogenesis. Sci. Rep 9 (1), 12444. 10.1038/s41598-019-48925-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posner K, Brown GK, Stanley B, Brent DA, Yershova KV, Oquendo MA, Currier GW, Melvin GA, Greenhill L, Shen S, Mann JJ, 2011. The Columbia-suicide severity rating scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am. J. Psychiatry 168 (12), 1266‒1277. 10.1176/appi.ajp.2011.10111704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin J, Yang T, Zeng N, Wan C, Gao L, Li X, Chen L, Shen Y, Wen F, 2019. Differential coexpression networks in bronchiolitis and emphysema phenotypes reveal heterogeneous mechanisms of chronic obstructive pulmonary disease. J. Cell. Mol. Med 23 (10), 6989‒6999. 10.1111/jcmm.14585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riehle K, Coarfa C, Jackson A, Ma J, Tandon A, Paithankar S, Raghuraman S, Mistretta TA, Saulnier D, Raza S, Diaz MA, Shulman R, Aagaard K, Versalovic J, Milosavljevic A, 2012. The Genboree Microbiome Toolset and the analysis of 16S rRNA microbial sequences. BMC Bioinformatics 13 (Suppl. 13), S11. 10.1186/1471-2105-13-S13-S11 (Epub 2012 Aug 24). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rutsch A, Kantsjo JB, Ronchi F, Tay TL, Wekerle H, Rojas OL, 2020. The gut-brain Axis: how microbiota and host Inflammasome influence brain physiology and pathology. Article 11, 1. 10.3389/fimmu.2020.604179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saulnier DM, Riehle K, Mistretta TA, Diaz MA, Mandal D, Raza S, Weidler EM, Qin X, Coarfa C, Milosavljevic A, Petrosino JF, Highlander S, Gibbs R, Lynch SV, Shulman RJ, Versalovic J, 2011. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology 141 (5), 1782‒1791. 10.1053/j.gastro.2011.06.072 (Epub 2011 Jul 8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, Kurilshikov A, Fu J, Joosten LAB, Zhernakova A, Huttenhower C, Wijmenga C, Netea MG, Xavier RJ, 2016. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell 167 (4), 1125‒1136.e8. 10.1016/j.cell.2016.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Segata N, Waldron L, Ballarini A, Narasimhan V, Jousson O, Huttenhower C, 2012. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat. Methods 9 (8), 811–814. 10.1038/nmeth.2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13 (11), 2498‒2504. 10.1101/gr.1239303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharp C, Fowler JC, Salas R, Nielsen D, Allen J, Oldham J, Kosten T, Mathew S, Madan A, Frueh BC, Fonagy P, 2016. Operationalizing NIMH research domain criteria (RDoC) in naturalistic clinical settings. Bull. Menn. Clin 80 (3), 187–212. 10.1521/bumc.2016.80.3.187. [DOI] [PubMed] [Google Scholar]
- Shrestha A, Uzal FA, McClane BA, 2018. Enterotoxic Clostridia: Clostridium perfringens enteric diseases. Microbiology. Spectrum 6 (5). 10.1128/microbiolspec.GPP3-0003-2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sindermann L, Redlich R, Opel N, Böhnlein J, Dannlowski U, Leehr EJ, 2021. Systematic transdiagnostic review of magnetic-resonance imaging results: depression, anxiety disorders and their co-occurrence. J. Psychiatr. Res 142, 226–239. 10.1016/j.jpsychires.2021.07.022. [DOI] [PubMed] [Google Scholar]
- Subramanian A, Narayan R, Corsello SM, Peck DD, Natoli TE, Lu X, Gould J, Davis JF, Tubelli AA, Asiedu JK, Lahr DL, Hirschman JE, Liu Z, Donahue M, Julian B, Khan M, Wadden D, Smith IC, Lam D, Liberzon A, Toder C, Bagul M, Orzechowski M, Enache OM, Piccioni F, Johnson SA, Lyons NJ, Berger AH, Shamji AF, Brooks AN, Vrcic A, Flynn C, Rosains J, Takeda DY, Hu R, Davison D, Lamb J, Ardlie K, Hogstrom L, Greenside P, Gray NS, Clemons PA, Silver S, Wu X, Zhao WN, Read-Button W, Wu X, Haggarty SJ, Ronco LV, Boehm JS, Schreiber SL, Doench JG, Bittker JA, Root DE, Wong B, Golub TR, 2017. A next generation connectivity map: L1000 platform and the First 1,000,000 profiles. Cell. 171 (6), 1437–1452.e17. 10.1016/j.cell.2017.10.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takehara M, Seike S, Sonobe Y, Bandou H, Yokoyama S, Takagishi T, Miyamoto K, Kobayashi K, Nagahama M, 2019. Clostridium perfringens α-toxin impairs granulocyte colony-stimulating factor receptor-mediated granulocyte production while triggering septic shock. Communications Biology 2, 45. 10.1038/s42003-019-0280-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tesson BM, Breitling R, Jansen RC, 2010. DiffCoEx: a simple and sensitive method to find differentially coexpressed gene modules. BMC Bioinformatics 11 (1), 497. 10.1186/1471-2105-11-497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- The Human Microbiome Project Consortium, 2012a. Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214. 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- The Human Microbiome Project Consortium, 2012b. A framework for human microbiome research. Nature 486, 215–221. 10.1038/nature11209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson DS, Fowler JC, Bradshaw MR, Frueh BF, Weinstein BL, Petrosino J, Hadden JK, Madan A, 2021. Is the gut microbiota associated with suicidality? Non-significant finding among a large cohort of psychiatrically hospitalized individuals with serious mental illness. J Affect Disord Rep 6, 100266. 10.1016/j.jadr.2021.100266. [DOI] [Google Scholar]
- Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang J, Tito RY, Schiweck C, Kurilshikov A, Joossens M, Wijmenga C, Claes S, Van Oudenhove L, Zhernakova A, Vieira-Silva S, Raes J, 2019. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol 4 (4), 623–632. 10.1038/s41564-018-0337-x. [DOI] [PubMed] [Google Scholar]
- Van Ameringen M, 2021. Comorbid anxiety and depression in adults, epidemiology, clinical manifestations, and diagnosis. UpToDate. https://www.uptodate.com/contents/comorbid-anxiety-and-depression-in-adults-epidemiology-clinical-manifestations-and-diagnosis.
- Vernocchi P, Gili T, Conte F, Del Chierico F, Conta G, Miccheli A, Botticelli A, Paci P, Caldarelli G, Nuti M, Marchetti P, Putignani L, 2020. Network analysis of gut microbiome and metabolome to discover microbiota-linked biomarkers in patients affected by non-small cell lung Cancer. Int. J. Mol. Sci 21 (22), 8730. 10.3390/ijms21228730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J, Chen W-D, Wang Y-D, 2020. The relationship between gut microbiota and inflammatory diseases: the role of macrophages. Front. Microbiol 11, 1065. 10.3389/fmicb.2020.01065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei C-H, Allot A, Leaman R, Lu Z, 2019. PubTator central: automated concept annotation for biomedical full text articles. Nucleic Acids Res. 47 (W1), W587–W593. 10.1093/nar/gkz389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wittchen H-U, 2002. Generalized anxiety disorder: prevalence, burden, and cost to society. Depression and Anxiety 16 (4), 162–171. 10.1002/da.10065. [DOI] [PubMed] [Google Scholar]
- Wu H-J, Wu E, 2012. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 3 (1), 4‒14. 10.4161/gmic.19320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie F, Xu L, Wang Y, Mao S, 2021. Metagenomic sequencing reveals that high-grain feeding alters the composition and metabolism of cecal microbiota and induces cecal mucosal injury in sheep. mSystems 6 (5), e0091521. 10.1128/mSystems.00915-21(Epub 2021 Oct 5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Y, Cao Z, Yang M, Xi X, Guo Y, Fang M, Cheng L, Du Y, 2017. Comorbid generalized anxiety disorder and its association with quality of life in patients with major depressive disorder. Sci. Rep 7, 40511. 10.1038/srep40511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou A, Nadeau K, Xiong X, Wang PW, Copeland JK, Lee JY, Pierre JS, Ty M, Taj B, Brumell JH, Guttman DS, Sharif S, Korver D, Parkinson J, 2022. Systematic profiling of the chicken gut microbiome reveals dietary supplementation with antibiotics alters expression of multiple microbial pathways with minimal impact on community structure. Microbiome. 10 (1), 127. 10.1186/s40168-022-01319-7. [DOI] [PMC free article] [PubMed] [Google Scholar]