Abstract
Rationale
Mycobacterium vaccae (NCTC 11659) is an environmental saprophytic bacterium with anti-inflammatory, immunoregulatory, and stress resilience properties. Previous studies have shown that whole, heat-killed preparations of M. vaccae prevent allergic airway inflammation in a murine model of allergic asthma. Recent studies also demonstrate that immunization with M. vaccae prevents stress-induced exaggeration of proinflammatory cytokine secretion from mesenteric lymph node cells stimulated ex vivo, prevents stress-induced exaggeration of chemically induced colitis in a model of inflammatory bowel disease, and prevents stress-induced anxiety-like defensive behavioral responses. Furthermore, immunization with M. vaccae induces anti-inflammatory responses in the brain and prevents stress-induced exaggeration of microglial priming. However, the molecular mechanisms underlying anti-inflammatory effects of M. vaccae are not known.
Objectives
Our objective was to identify and characterize novel anti-inflammatory molecules from M. vaccae NCTC 11659.
Methods
We have purified and identified a unique anti-inflammatory triglyceride, 1,2,3-tri [Z-10-hexadecenoyl] glycerol, from M. vaccae and evaluated its effects in freshly isolated murine peritoneal macrophages.
Results
The free fatty acid form of 1,2,3-tri [Z-10-hexadecenoyl] glycerol, 10(Z)-hexadecenoic acid, decreased lipopolysaccharide-stimulated secretion of the proinflammatory cytokine IL-6 ex vivo. Meanwhile, next-generation RNA sequencing revealed that pretreatment with 10(Z)-hexadecenoic acid upregulated genes associated with peroxisome proliferator-activated receptor alpha (PPARα) signaling in lipopolysaccharide-stimulated macrophages, in association with a broad transcriptional repression of inflammatory markers. We confirmed using luciferase-based transfection assays that 10(Z)-hexadecenoic acid activated PPARα signaling, but not PPARγ, PPARδ, or retinoic acid receptor (RAR) α signaling. The effects of 10(Z)-hexadecenoic acid on lipopolysaccharide-stimulated secretion of IL-6 were prevented by PPARα antagonists and absent in PPARα-deficient mice.
Conclusion
Future studies should evaluate the effects of 10(Z)-hexadecenoic acid on stress-induced exaggeration of peripheral inflammatory signaling, central neuroinflammatory signaling, and anxiety- and fear-related defensive behavioral responses.
Keywords: 10(Z)-hexadecenoic acid, Bacteria, Inflammation, Interleukin 6, Lipid, Macrophage, Mycobacteria, PPAR, RNA-seq, vaccae
Introduction
The global prevalence of anxiety disorders has been estimated to be 7.3%, ranging from 5.3% in African cultures to 10.4% in Euro/Anglo cultures (Baxter et al. 2013). According to the Diagnostic and Statistical Manual of Mental Disorders (5th ed.) (DSM-5; American Psychiatric Association 2013), anxiety disorders include those that share features of excessive fear and anxiety and related behavioral disturbances, such as generalized anxiety disorder, panic disorder, social anxiety disorder (social phobia), and specific phobia (American Psychiatric Association 2013). Posttraumatic stress disorder (PTSD), although formerly classified as an anxiety disorder, is classified as a trauma- and stressor-related disorder (American Psychiatric Association 2013). Collectively, anxiety and trauma-related disorders are complex and multifactorial, and their differentiation and management are complicated by phenotypic heterogeneity. The etiology and pathophysiology of these disorders are thought to involve interactions among the genome, epigenome, and environment (Nugent et al. 2011). Recently, investigation of the etiology and pathophysiology of psychiatric and neurological diseases has expanded to include a potential role of the microbiota–gut–brain (MGB) axis (Forsythe et al. 2010; Cryan and Dinan 2012, 2015; Leclercq et al. 2016). Of particular interest, evidence from preclinical and clinical studies suggests that exaggerated inflammation, which in some cases may be secondary to dysregulation of the microbiome, may be a risk factor for the development of trauma- and stressor-related disorders (for review, see Langgartner et al. 2018). These studies raise the question of whether or not microbial-based interventions with anti-inflammatory or immunoregulatory properties may have value in the prevention or treatment of trauma- and stressor-related disorders.
Evidence suggests that some common pathogenic and non-pathogenic microorganisms, to which humans have been exposed throughout evolution, drive anti-inflammatory and immunoregulatory mechanisms that inhibit inappropriate immune responses by the host (Rook and Rosa Brunet 2002; Rook 2009, 2010; Okada et al. 2010). Throughout human evolution, the interactions between these ancestral microorganisms, which we have collectively referred to as “old friends,” and the innate immune system promoted immunoregulation. These “old friends” included microorganisms that (1) were part of host physiology (human microbiota); (2) were harmless but inevitably contaminating air, food, and water (environmental microbiota); or (3) led to severe host tissue damage when attacked by the host immune system (e.g., helminthic parasites) (Rook 2013; Blaser 2017).
“Old friends” are thought to suppress host inflammation through a variety of mechanisms, including the induction of specific subsets of antigen-presenting cells such as macrophages and dendritic cells (DCs) and modulation of innate immunity (Le Bert et al. 2011; Garn et al. 2016; Lowry et al. 2016). In their absence, the host may develop inappropriate immune responses to allergens, self-antigens, or gut microbiota. It has been hypothesized that increases in allergies, auto-immune diseases, inflammatory bowel diseases, and psychiatric disorders in modern living conditions may be due, in part, to decreased exposure to “old friends” (Rook 2010; Lyte and Cryan 2014; Bloomfield et al. 2016; Lowry et al. 2016; Stamper et al. 2016). In parallel, individuals with a diagnosis of PTSD have a higher risk of development of any autoimmune disease, relative to those with other psychiatric disorders, or relative to those with no psychiatric disorder (O’Donovan et al. 2015), suggesting that impaired immuno-regulation or inappropriate inflammation may confer risk for development of both autoimmune conditions and PTSD. The saprophytic mycobacterium, Mycobacterium vaccae (National Collection of Type Cultures (NCTC) 11659), has shown encouraging therapeutic potential in diseases of inflammation and immunodysregulation (Gutzwiller et al. 2007; Rook et al. 2007) and has shown immunoregulatory and stress-protective effects in murine models (Zuany-Amorim et al. 2002; Adams et al. 2004; Lowry et al. 2007; Reber et al. 2016; Fox et al. 2017; Frank et al. 2018). Mycobacteria are abundant in municipal water supplies (Gebert et al. 2018) and are a normal component of the healthy human microbiome of the oral cavity (buccal mucosa and dental plaque) and upper respiratory tract (nostrils and oropharynx) and, therefore, are considered part of the microbiome of the upper airways (Macovei et al. 2015).
The identification of specific microbially derived molecules with anti-inflammatory or immunoregulatory properties may provide novel therapeutic avenues for the treatment of diseases of immunodysregulation or trauma- and stressor-related disorders where exaggerated inflammation is thought to be a risk factor (Lowry et al. 2016; Langgartner et al. 2018). We have previously shown that treatment with a heat-killed preparation of the saprophytic mycobacterium, M. vaccae, prevents murine allergic pulmonary inflammation by inducing CD4+CD45RBlow Tregs (Zuany-Amorim et al. 2002). These cells are allergen-specific and, upon passive transfer, can protect recipient allergic mice from airway inflammation by significantly reducing eosinophilia in the lungs. In addition, treatment with M. vaccae induces a population of pulmonary CD11c+ antigen-presenting cells, which are characterized by increased expression of IL-10, transforming growth factor beta (TGFβ) and interferon α (IFNα) (Adams et al. 2004). Furthermore, at least in vitro, priming of human DCs with M. vaccae induces strong inhibition of Th2 responses (Le Bert et al. 2011).
Meanwhile, we have shown that immunization of mice with M. vaccae promotes a more proactive response to a chronic psychosocial stressor, prevents stress-induced colitis, prevents stress-induced exaggeration of chemically induced colitis in a model of inflammatory bowel disease, and attenuates anxiety-like defensive behavioral responses (Reber et al. 2016). Consistent with these findings, immunization with M. vaccae prevents stress-induced exaggeration of interferon gamma and IL-6 secretion from freshly isolated mesenteric lymph node cells stimulated with anti-CD3 antibody ex vivo. Importantly, preimmunization with M. vaccae, in stressed mice, resulted in a two orders of magnitude increase in IL-10 secretion from mesenteric lymph node cells stimulated ex vivo. However, until now, specific constituents of M. vaccae that suppress inflammation in macrophages in the periphery or central nervous system have not been identified.
Through a screening process of M. vaccae NCTC 11659 lipid extracts, a single triglyceride, 1,2,3-tri [Z-10-hexadecenoyl] glycerol, was identified with potential immunotherapeutic benefits (Rosa Brunet and Rook 2008). The lipid was demonstrated to prevent allergic airway inflammation, and the lipid recapitulated the therapeutic effects of whole heat-killed M. vaccae. The protective phenotype was characterized by increased IL-10, decreased IL-5, and reduced infiltration of eosinophils and macrophages in bronchoalveolar lavage fluid (Rosa Brunet and Rook 2008). It was also shown that the efficacy of the triglyceride was not dependent on the glycerol structure, as the synthetic, constituent free fatty acid, 10(Z)-hexadecenoic acid, was sufficient to suppress pulmonary airway inflammation. The mechanism through which this long-chain, monounsaturated fatty acid was capable of limiting symptoms of inflammation is unknown, but it is explored here in a model of macrophage activation.
Notably, it is relatively rare in nature for an organism to naturally produce a fatty acid that is unsaturated at the C10 position, yet several mycobacteria species—including M. vaccae, can perform that desaturation (Scheuerbrandt and Bloch 1962; Coyle et al. 1992; Böttger et al. 1993; Springer et al. 1993; Suutari and Laakso 1993; Chou et al. 1998; Tay et al. 1998; Pacífico et al. 2018). We successfully synthesized the free fatty acid, 10(Z)-hexadecenoic acid, and using cell-based assays and RNA-seq, revealed that 10(Z)-hexadecenoic acid upregulated genes associated with the peroxisome proliferator-activated receptor (PPAR) signaling pathway and inhibited proinflammatory signaling of activated macrophages ex vivo. Furthermore, studies using cultured cells transfected with lipid-regulated transcription factors revealed that both the monoacylglycerol lipid constituent of M. vaccae and its free fatty acid form selectively increased PPARα signaling. The effects of 10(Z)- hexadecenoic acid to inhibit proinflammatory signaling of activated macrophages ex vivo were prevented by PPARα antagonists and absent in PPARα-deficient mice. This is the first report, to our knowledge, to show that a synthetic M. vaccae-derived lipid acts to induce anti-inflammatory responses in host immune cells by acting as an agonist at host PPARα receptors.
Materials and methods
Animals
Adult male BALB/c mice (BALB/cAnHsd; Cat. No. 047; Harlan, Indianapolis, IN, USA), 6–8 weeks old, were used and housed under standard conditions with food and water available ad libitum. Adult male PPARα−/− (B6;129S4-Pparatm1Gonz/J; Cat. No. 008154; Jackson Laboratories, Bar Harbor, ME, USA) and control mice (C57BL/6J; Cat. No. 000664; Jackson Laboratories), 6–8 weeks old, were used and housed under standard conditions with food and water available ad libitum. Although the C57BL/6J inbred strain is considered an approximate control for the PPARα−/− mice (B6;129S4-Pparatm1Gonz/J; Jackson Laboratories), future studies should ideally compare PPARα−/− mice to wild-type littermates.
All experimental protocols were consistent with the National Institutes of Health Guide for the Care and Use of Laboratory Animals, Eighth Edition (The National Academies Press 2011), and the Institutional Animal Care and Use Committee at the University of Colorado Boulder approved all procedures. This work was covered under CU Boulder IACUC Protocol Numbers 2134–14MAY2018 and 2361–14MAY2018-DT. The research described here was conducted in compliance with The ARRIVE Guidelines: Animal research: reporting of in vivo experiments, originally published in PLOS Biology, June 2010 (Kilkenny and Altman 2010). All possible efforts were made to minimize the number of animals used and their suffering.
Synthesis of 10(Z)-hexadecenoic acid; (10Z) -hexadec-10-enoic acid (CAS No. 2511–97-9)
Unless otherwise noted, reagents were obtained commercially and used without further purification. Dichloromethane (CH2Cl2) was distilled over calcium hydride (CaH2) under a nitrogen atmosphere. Tetrahydrofuran (THF; (CH2)4O) was distilled from sodium-benzophenone under a nitrogen atmosphere. Thin-layer chromatography analysis of reaction mixtures was performed on Dynamic Adsorbents, Inc., silica gel F-254 TLC plates. Flash chromatography was carried out on Zeoprep 60 ECO silica gel. 1H spectra were recorded with a Varian INOVA 500 spectrometer. Compounds were detected by monitoring UV absorbance at 254 nm.
To a 5-mL sealed tube containing 1-heptene (0.50 mL, 3. 55 mmol), methyl 10-undecenoate ( 0. 080 mL, 0.36 mmol), and THF (0.35 mL), a Grubbs Z-selective metathesis catalyst was added (Grubbs Catalyst(R) C633, 2.2 mg, 3.48 μmol, Cat. No. 771082, Sigma-Aldrich, St. Louis, MO, USA). The reaction was stirred at 45 °C for 8 h before cooling to room temperature. The slurry was filtrated through a short plug of silica gel and concentrated. The obtained oil was dissolved in 1.0 mL THF. The solution was cooled to 0 °C, then 9-borabicyclo[3.3.1] nonane (9-BBN) solution in THF (1.28 mL, 0.50 M, 0.64 mmol) was added. After 2 h stirring at 0 °C, the reaction was quenched with 60 μL EtOH, then 1.5 mL pH 7 potassium phosphate buffer and 1.5 mL 30% H2O2. The mixture was stirred at room temperature for 12 h, then extracted with 5 mL EtOAc three times. The combined organic layers were washed with 4 mL saturated Na2S2O3 and 3 mL brine, then dried over Na2SO4, filtered, and concentrated. To the crude oil in 1.0 mL THF was added LiOH monohydrate (38 mg, 0.90 mmol) in 1.0 mL water. After 2 h, the reaction solution was cooled to 0 °C before the addition of 0.91 mL 1.0 N HCl. After being concentrated under reduced pressure, the aqueous solution was saturated with NaCl and extracted with 3 mL dichloromethane three times. The combined organic layers were dried over Na2SO4, filtered, and concentrated. Purification by flash chromatography (2:1:1 hexanes/dichloromethane/diethyl ether) provided (10Z)- hexadec-10-enoic acid (0.022 g, 90%) as a colorless oil. 1H NMR (500 MHz, CDCl3): δ 5.48–5.22 (m, 2H), 2.35 (t, J = 7.5 Hz, 2H), 2.01 (q, J = 6.6 Hz, 4H), 1.63 (p, J = 7.4 Hz, 2H), 1.35–1.15 (m, 16H), 0.88 (t, J = 6.9 Hz, 3H).
Murine peritoneal macrophage isolation and screening
Murine peritoneal macrophages were isolated and cultured as previously described (Zhang et al. 2008) and used to determine the effects of 10 ( Z )-hexadecenoic acid on lipopolysaccharide (LPS)-induced IL-6 secretion. Briefly, mice received a single injection of 3% thioglycollate medium (1 mL, i.p.; Cat. No. 9000–294, VWR, Radnor, PA, USA). Mice were euthanized 96 h later using cervical dislocation, and macrophages were collected in Dulbecco’s phosphate-buffered saline (DPBS; Cat. No. 14190136, Invitrogen, Carlsbad, CA, USA). Cells were centrifuged and resuspended in Dulbecco’s modified Eagle medium/Nutrient Mixture F-12 (DMEM/F-12; Cat. No. 10565018, Invitrogen) supplemented with 10% (v/v) fetal bovine serum (Cat. No. 16000036, Invitrogen) and 1% penicillin/streptomycin (Cat. No. 15140148, Invitrogen). One mouse yielded enough cells for one experimental replicate; 1 × 105 cells/well were allowed to adhere for 1.5 h before being washed with DPBS. 10(Z)- hexadecenoic acid was dissolved in DMEM/F-12 with 0.5% (v/v) dimethyl sulfoxide (Cat. No. D8418, Sigma-Aldrich). The macrophages were incubated with either 10(Z)- hexadecenoic acid (0.4, 4, 20, 100, 500, 1000 μM) or DMEM/F-12 for 1 h before being stimulated with either 1 μg/mL LPS (serotype 0127:B8, Sigma-Aldrich, St. Louis, MO, USA) or DMEM/F-12. Culture supernatants were collected at 6, 12, and 24 h post-stimulation.
Cytokine measurements
Cell culture supernatants (10 μL) from freshly isolated peritoneal macrophages were diluted 1:200, and IL-6 was measured using ELISA (Cat. No. 431304, BioLegend, San Diego, CA, USA). The assay has a minimal detectable concentration of 2 pg/mL IL-6. All samples were measured using duplicate wells in the ELISA.
Cytotoxicity assay
Cytotoxicity was determined using the sulforhodamine B (SRB) colorimetric assay, as previously described (Vichai and Kirtikara 2006). Briefly, without removing the culture media, cells were fixed by adding cold trichloroacetic acid and incubated at 4 °C for 1 h. The plates were washed with slow-running tap water and set out to dry overnight. Then, 0.057% SRB (Cat. No. AC333130050, Fisher, Pittsburgh, PA, USA), solubilized in 10 mM Tris (Cat. No. BP153, Fisher), was added to each well. After 30 min, plates were washed with 1% acetic acid and set out to dry overnight. SRB was measured at 490 nm on a Synergy HT microplate reader (Part Number 7091000, Biotek, Winooski, VT, USA). Cell viability was expressed as the ratio of experimental and control growth.
Ligands
For studies using reporter gene assays following transfection of COS1 cells, rosiglitazone, troglitazone, and WY14643 were obtained from Alexis Biochemicals (San Diego, CA, USA); ATRA and AM580 were obtained from Sigma-Aldrich. In addition, GW9662 was a gift from T.M. Willson (GlaxoSmithKline, Brentford, UK). For experiments using freshly isolated peritoneal macrophages, GW 6471 (Cat. No. 4618), GW 9662 (Cat. No. 1508), GSK 0660 (Cat. No. 3433), WY 14643 (Cat. No. 1312), rosiglitazone (Cat. No. 5325), and GW 0742 (Cat. No. 2229) were obtained from Tocris Bioscience (Bristol, UK).
Transfections and reporter gene assays
Cells were transfected with the following receptor and reporter constructs: Gal4-PPARα-LBD, Gal4-PPARγ-LBD, Gal4-PPARδ-LBD, Gal4-RARα-LBD, pMH100-TK-luc, and pCMX-β-galactosidase (Chen and Evans 1995). All transfection experiments were performed with COS1 cells using polyethylenimine (Sigma-Aldrich) reagent (Szatmari et al. 2006). After 6–8 h of the transfection, the medium was replaced with DMEM medium containing the indicated ligands or vehicle (as control) (Chen and Evans 1995; Benko et al. 2003). Cells were lysed and assayed for reporter expression 18 h after transfection. The luciferase assay system (Promega, Madison, WI, USA) was used as described previously (Nagy et al. 1999). Measurements were carried out with a Wallac Victor-2, multilabel counter. Luciferase activity of each sample was normalized to the β-galactosidase activity.
RNA extraction and library preparation
Total RNA content of 1 × 105 macrophages pretreated for 1 h with 200 μM 10(Z)-hexadecenoic acid (utilizing separate macrophage preparations from n = 3 mice) or vehicle (utilizing separate macrophage preparations from n = 3 mice) and stimulated with 1 μg/mL LPS was extracted using TRI Reagent® (Cat. No. T9424, Sigma-Aldrich) according to the manufacturer’s instructions. The RNA input was quantified on a Qubit™ 3.0 Fluorometer (Cat. No. Q33216, Thermo Fisher, Waltham, MA, USA) to ensure there was sufficient starting material. The RNA sequencing libraries were generated with the NEBNext rRNA Depletion Kit (Cat. No. E6310, New England BioLabs) in order to enrich the samples in mRNA and NEBNext Ultra Directional RNA Library Prep Kit for Illumina (Cat. No. 7240, New England BioLabs). Briefly, mRNA was purified from 100 ng of total RNA, fragmented, and converted to double-stranded cDNA. Barcodes were ligated to the cDNA fragments, and prior to PCR enrichment of the library, the cDNA product was quantified on a Qubit 3.0 Fluorometer (Thermo Fisher). The integrity of the purified oligo libraries was evaluated on an Agilent Bioanalyzer 2100 (Cat. No. G2939BA, Agilent, Santa Clara, CA, USA).
Sequencing
Libraries were sequenced at the Next Generation Sequencing Facility at the University of Colorado Boulder. The libraries were multiplexed and sequenced on an Illumina HiSeq 2000 Sequencing System (Cat. No. SY-401–1001, Illumina, San Diego, CA, USA). For each sample, paired-end 100-bp reads were sequenced using V3 chemistry.
RNA read processing, mapping, and differential expression
Quality analysis of sequencing data was done using FastQC. The adaptors and low-quality raw reads were cut with Trimmomatic (version 0.32) (Bolger et al. 2014). The reads were aligned to the mouse genome, mm10 (University of California, Santa Cruz, CA, USA), using the TopHat2 sequence aligner (version 2.0.6) (Kim et al. 2013). Reads mapping to exon features were counted using HTseq (version 0.6.1) (Anders et al. 2015). The raw reads and count data have been deposited in the GEO database under accession number GSE125930. Differentially expressed genes were identified using the R package, DESeq (version 1.28.0) (Anders and Huber 2010).
Statistical analysis
Data are presented as means ± SEM or means + SEM. Data were subjected to a normality test and one-way analysis of variance (ANOVA); Fisher’s least significant difference (LSD) tests were performed as appropriate. A two-tailed p value ≤ 0.05 was considered significant. ELISA IL-6 data were analyzed using linear mixed effects models using the software package SPSS (version 21.0, SPSS Inc., Chicago, IL, USA). Network visualizations were created in Cytoscape (version 3.5.1) using an enrichment map plug-in (Merico et al. 2011).
Results
10(Z)-hexadecenoic acid decreases LPS-induced secretion of IL-6 in macrophages
To simulate inflammation, freshly isolated mouse peritoneal macrophages were challenged with LPS (1 μg/mL) ex vivo (outlined in Fig. 1). Macrophages that were cultured in the presence of 10(Z)-hexadecenoic acid (0, 0.4, 4, 20, 100, 500, 1000 μM) for 1 h prior to 1 μg/mL LPS stimulation secreted less IL-6 relative to macrophages cultured with media alone prior to LPS stimulation (Fig. 2a–c) (F(1, 111) = 15.20, p < 0.001). This difference was observable as early as 6 h after LPS challenge and was sustained for at least 24 h. We selected the 6-, 12-, and 24-h time points for measurement of IL-6 as previous studies have shown increased IL-6 secretion using LPS-stimulated peritoneal macrophage cultures in mice at these time points, with linear increases in IL-6 up to the 24-h time point (Shacter et al. 1993; Wollenberg et al. 1993; Lin and Tang 2007; Lee et al. 2015; Arteaga Figueroa et al. 2017). The effect also appeared to be concentration and time dependent. Using a constrained logistic model on the relative secretion of IL-6, we estimated the EC50 to be 823, 115, and 190 μM at the 6-, 12-, and 24-h observations, respectively (Fig. 2). Post hoc pairwise comparisons of raw IL-6 values relative to paired media control values at the same time point (n = 3 per group) are presented in Table S1. This time and concentration dependence may indicate that a receptormediated transcriptional change is occurring. In contrast to the effects of 10(Z)-hexadecenoic acid on LPS-induced IL-6 secretion, it had no detectable effect on IL-6 secretion by itself (IL-6 was undetectable in all conditions; Fig. S1). We cannot exclude the possibility, however, that 10(Z)-hexadecenoic acid by itself had effects on IL-6 secretion that were below the limit of detectability of the assay used (i.e., 2 pg/mL). Cell viability was measured to dispel the possibility that senescence or cell death was contributing to reduced IL-6 secretion. Using a high concentration (1 mM) of 10(Z)-hexadecenoic acid, less than 40% of macrophages were viable at most time points. However, macrophages cultured with all other concentrations of 10(Z)-hexadecenoic acid studied (i.e., 10, 50, 125, 250, and 500 μM) were as viable as media controls (Fig. S2).
Fig. 1.

Experimental timeline for ex vivo macrophage stimulation. Abbreviations: FFA, free fatty acid; LPS, lipopolysaccharide
Fig. 2.

Anti-inflammatory effects of 10(Z)-hexadecenoic acid in freshly isolated murine peritoneal macrophages. Freshly isolated murine peritoneal macrophages were incubated for 1 h with synthetic 10(Z)- hexadecenoic acid (0, 0.4, 4, 20, 100, 500, 1000 μM), then challenged with lipopolysaccharide (LPS; 1 μg/mL). Cell supernatants were collected at a 6 h, b 12 h, and c 24 h after LPS challenge. Interleukin (IL) 6 concentrations in the supernatant were determined using enzymelinked immunosorbent assay (ELISA) and reported relative to media-only controls (n = 6 replicates, with each replicate using different freshly isolated peritoneal macrophages; each sample was run using duplicate wells in the ELISA). Data were fit with a logistic function, which was used to estimate the EC50. Data are expressed as mean ± SEM
Treatment with 10(Z)-hexadecenoic acid induces a broad anti-inflammatory transcriptional profile in LPS-stimulated macrophages
To explore the potential effects of 10(Z)-hexadecenoic acid on transcriptional responses in LPS-stimulated macrophages, we used RNA-seq. Murine peritoneal macrophages were incubated with 200 μM 10(Z)-hexadecenoic acid or a media-only control condition for 1 h prior to stimulation with LPS. Using IL-6 as a measure for the suppressive activity of 10(Z)-hexadecenoic acid, we estimated the EC50 at 12 h to be 115 μM. The 200-μM concentration was chosen as it was sufficiently larger than the EC50, but less than a concentration that would affect macrophage viability. After a 12 h exposure to LPS, the RNA was extracted and depleted of rRNA. We selected the 12-h time point for measurement of mRNA using RNA-seq as previous studies have shown increased IL-6 secretion using LPS-stimulated peritoneal macrophage cultures in mice at this time point, as well as the ability to suppress IL-6 mRNA expression at this time point by interfering with a TLR4–MyD88–BLT2–Nox1–ROS–NF-κB pathway leading to IL-6 secretion (Lee et al. 2015). The cDNA libraries were sequenced in a 100-bp paired-end experiment generating 51–63 million reads per sample (Table S2; Fig. S3).
For differential expression, we examined LPS-stimulated macrophages pretreated with either 10(Z)-hexadecenoic acid or vehicle (GSE125930). Differentially expressed transcripts were identified using the R package, DESeq (Anders and Huber 2010). A total of 203 genes were found to be differentially expressed with an FDR-adjusted p < 0.1 (Table S3). Of the 203 differentially expressed genes, 109 were downregulated in the 10(Z)-hexadecenoic acid condition, and 20% of those genes were associated with proinflammatory processes (Table S4). The top 20 differentially expressed genes are reported in Fig. 3a. Consistent with the ex vivo macrophage experiments measuring IL-6 protein with ELISA, the second most significantly differentially expressed transcript was IL-6 (Table S3).
Fig. 3.

Gene networks upregulated following pretreatment with 10(Z)- hexadecenoic acid in LPS-stimulated macrophages suggest anti-inflammatory effects are mediated by PPARα. Murine peritoneal macrophages were treated with either 10(Z)-hexadecenoic acid (200 μM) or vehicle. Following a 12-h period after stimulation with lipopolysaccharide (LPS), total RNA content was measured using RNA-seq. a Heat map of the top 20 differentially expressed transcripts. b, c Genes significantly b upregulated or c downregulated following treatment with 10(Z)-hexadecenoic acid were separately queried on the Database for Annotation, Visualization and Integrated Discovery (DAVID). b The top 5 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enriched for genes upregulated following pretreatment of LPS-stimulated macrophages with 10(Z)-hexadecenoic acid, relative to media pretreated, LPS-stimulated macrophages. c The top 5 KEGG pathways enriched for genes downregulated following pretreatment of LPS-stimulated macrophages with 10(Z)-hexadecenoic acid, relative to media pretreated, LPS-stimulated macrophages. d Pathway analysis using the entire transcriptional data set was performed with Gene Set Enrichment Analysis (GSEA). Pathways enriched for genes upregulated following pretreatment of LPS-stimulated macrophages with 10(Z)-hexadecenoic acid, relative to mediapretreated, LPS-stimulated macrophages, were visualized in a network built by their gene set overlap. The size of the network node represents the number of genes shared between the particular gene set and the transcription data. The weight of network edges represents the degree of gene set overlap. In the largest cluster of pathways enriched in genes upregulated with 10(Z)-hexadecenoic acid, lipid metabolism and peroxisome proliferator-activated receptors (PPARs) were implicated as some of the more salient pathways. Abbreviations: Adamtsl4, thrombospondin repeat-containing protein 1; AMPK, 5′ AMP-activated protein kinase; Ch25h, cholesterol 25-hydroxylase; Cish, cytokine-inducible SH2 containing protein; Ctla2b, cytotoxic T-lymphocyte- associated protein 2-beta; Cyp26b1, cytochrome P450 family 26 subfamily B member 1; Dusp1, dual specificity phosphatase 1; ECM, extracellular matrix; F3, coagulation factor III; Flrt3, fibronectin leucine-rich transmembrane protein 3; Hdc, histidine decarboxylase; Hp, haptoglobin; Il1b, interleukin 1 beta; Il6, interleukin 6; LKB1, liver kinase B1; Mir5105, microRNA 5105; MTOR, mechanistic target of rapamycin kinase; Plbd1, phospholipase B domain containing 1; Plin2, perilipin 2; PPAR, peroxisome proliferator-activated receptor; PPARA, peroxisome proliferator-activated receptor alpha; Ptgs2, prostaglandin- endoperoxide synthase 2; RORA, RAR-related orphan receptor A; TNF, tumor necrosis factor; Tns1, tensin 1; Tsc22d3, Tsc22 domain family member 3; Vnn3, vanin 3
PPARα-regulated genes are associated with 10(Z) -hexadecenoic acid treatment in LPS-stimulated macrophages
To better understand the pathways affected by 10(Z)- hexadecenoic acid treatment, the list of 203 differentially expressed genes was queried against the Database for Annotation, Visualization and Integrated Discovery (DAVID) (Huang et al. 2009). Within the top 40 most significantly enriched KEGG pathways, 34 (i.e., 85%) were related to disease or inflammation (Table S5). In addition, 32 of the top 40 most significantly enriched KEGG pathways (i.e., 80%) were exclusively enriched for genes that were significantly downregulated by treatment with 10(Z)-hexadecenoic acid. Among these most significantly affected pathways, there was a wide scope of immunological context, which included infections, diseases, cytokine signaling, and various inflammatory pathways. The top 5 pathways with genes that were exclusively downregulated by treatment with 10(Z)- hexadecenoic acid are reported in Fig. 3c.
While the majority of pathways with genes affected by 10(Z)-hexadecenoic acid involved genes that were exclusively downregulated by 10(Z)-hexadecenoic acid, some pathways involved genes that were exclusively upregulated by 10(Z)- hexadecenoic acid. Of the top 40 pathways, 7 (i.e., 17.5%) pathways were exclusively enriched for genes that were significantly upregulated by treatment with 10(Z)-hexadecenoic acid. Overall, of 203 genes that were differentially expressed following treatment with 10(Z)-hexadecenoic acid, 93 genes (46%) were upregulated. The pathways with detectable enrichment involved regulation of lipolysis in adipocytes, glycerolipid metabolism, circadian entrainment, PPAR signaling pathway, and extracellular matrix–receptor interaction (Fig. 3b). The top 5 pathways with genes that were exclusively upregulated by treatment with 10(Z)-hexadecenoic acid are reported in Fig. 3b. The PPAR signaling pathway was among the top 5 most-enriched KEGG pathways with genes that were exclusively upregulated by treatment with 10( Z)- hexadecenoic acid (Fig. 3b).
In a secondary analysis, rather than rely on a subset of genes for biological interpretation, we used all expression data in Gene Set Enrichment Analysis (GSEA). We queried all detected transcripts against the KEGG pathways database (c2.cp.kegg.v6.2), and the top 5 enriched gene sets for the 10(Z)-hexadecenoic acid phenotype were as follows: “peroxisome” (KEGG: hsa04146), a main site of fatty acid oxidation via the β-oxidation cycle; “ppar_signaling_pathway” (KEGG: hsa03320); “citrate_cycle_tca_cycle” (KEGG: hsa00020); “fatty_acid_metabolism” (KEGG: hsa00071); and “propanoate_metabolism” (KEGG: hsa00640) (Table S6). Of potential interest, four of these KEGG pathways, “ peroxisome ” (KEGG: h s a0 4146 ), “ ppar_signaling_ pathway” (KEGG: hsa03320), “fatty_acid_metabolism” ( KEGG: hsa00071), and “propanoate_metabolism” (KEGG: hsa00640), were also found to be enriched in livers from 24 h fasted PPARα+/+ relative to PPAR−/− mice, while “peroxisome” (KEGG: hsa04146), “ppar_signaling_pathway” (KEGG: hsa03320), and “fatty_acid_metabolism” (KEGG: hsa00071) were found to be enriched in livers from wild-type mice treated with the PPARα agonist Wy14643, relative to vehicle (Kersten 2014). Together, these studies support a convergence of 10(Z)- hexadecenoic acid effects on PPARα signaling pathways induced by physiological or pharmacological stimuli. Propionate is one of the short-chain fatty acids, which are emerging as key mediators and regulators of host–microbe cross-talk, with a significant impact on host metabolism, including as an energy source (Hoyles et al. 2018). All five gene sets were significant with an unadjusted p value < 0.05 but failed to reach significance using FDR-adjusted p values; nevertheless, the overall pattern is consistent with a modulation of lipid metabolism. In a network visualization of gene set overlap between all detected pathways using GSEA, PPARs and peroxisomal lipid metabolism were prominent vertices (Fig. 3d). We also searched against the collection of transcription factor binding motifs (c3.tft.v6.0), which revealed enrichment for CREB, Gfi1, and PPARα cis-regulatory motifs upstream of the genes upregulated with 10(Z)-hexadecenoic acid treatment in LPS-stimulated macrophages (Table S7). Again, these were nominally significant (i.e., p < 0.05; q > 0.05), but these findings bolster PPARs, and specifically PPARα, as a potential receptor mediating anti-inflammatory effects of 10(Z)- hexadecenoic acid.
Downstream signaling of TLR4 is inhibited with 10(Z) -hexadecenoic acid treatment
NF-κB is one of three major transcription factors down stream of LPS-induced activation of toll-like receptor 4 (TLR4), the other two being IRF3 and AP-1 (Kawasaki and Kawai 2014). Using all expression data in GSEA, we found that pretreatmentwith10(Z)-hexadecenoicacid, relativetotreatmentwith vehicle, prior to LPS stimulation, downregulated signaling pathways downstream from TLR4, such as NF-κB and IRF3, but not AP-1. In a network visualization of the most significant pathways (FDR < 0.1), there is a bifurcation at the “ZHOU_INFLAMMATORY_RESPONSE_LPS” node representing the NF-κB and IRF3 responses (Fig. 4a). Among all nodes in this network, we counted and ranked the occurrence of enriched transcripts. The counts for the highest ranking transcripts were categorized into either NF-κB- regulated responses or IRF-regulated responses (Fig. 4b). We also examined enrichment for transcription factor motifs anddetectedenrichmentfor NF-κB, IRF1, IRF2, and IRF_Q6 amongothers, in transcripts associatedwith the vehicle-treated,LPSstimulatedgroup(Table S8). Alternatively, thiscanbe understood to mean that mRNA transcripts that are located near those transcription factor binding sites are downregulated in 10(Z)-hexadecenoic acid-treated, LPS-stimulated macrophages. To better understand the classification of the IRF_Q6 geneset(N = 242 genes) anddifferentiallyexpressed genes(q < 0.1, N = 203 genes), theywerebothqueriedagainst the Interferome database (Rusinova et al. 2013). There are three types of interferons (IFNs), namely type I (composed of α, β, κ, ε, and ω subtypes), type II (IFNγ), and type III (IFNλ; also called IL-28 and IL-29), which are distinguished by having distinct genetic loci, amino acid sequence homology, and specific cognate receptors (Pestka et al. 2004). This analysis revealed that a vast majority of the differentially expressed genes are regulated by both type I and II interferon responses(Fig. 4c), consistentwiththehypothesisthat 10(Z)- hexadecenoic acid alters TLR4, IRF3, and interferon signaling. Of note, cells infected with Mycobacterium tuberculosis induce type I interferons, including IFNα and IFNβ, which are thought to promote infection with M. tuberculosis (Travar et al. 2016). Using enrichment tools, like DAVID and GSEA, these RNA-seq data suggest that both NF-κB and IRF3 pathways are downregulated in LPS-stimulated macrophages when treated with 10(Z)-hexadecenoic acid.
Fig. 4.

10(Z)-hexadecenoic acid suppresses expression of transcription factors downstream of TLR4. Murine peritoneal macrophages were treated with either 10(Z)-hexadecenoic acid (200 μM) or vehicle for 1 h, then challenged with lipopolysaccharide (LPS; 1 μg/mL). Following a 12-h period after stimulation with LPS, mRNA was measured using RNA-seq. a From the Gene Set Enrichment Analysis (GSEA; c2.all.v6.2), pathways enriched with genes downregulated following pretreatment of LPS-stimulated macrophages with 10(Z)- hexadecenoic acid, relative to media-pretreated, LPS-stimulated macrophages, were visualized in a network built by their gene set overlap. The size of the network node indicates the number of genes shared between the particular gene set and the transcription data from our study. The weight of network edges indicates the degree of gene set overlap between nodes. The color of the node indicates whether the genes in the gene set were downregulated in NF-κB pathways (blue), downregulated in IRF pathways (purple), ambiguously downregulated (gray), or upregulated (red) in the 10(Z)-hexadecenoic acid condition. b Among the leading edges of enriched pathway gene sets, the occurrence of high ranking genes in either the NF-κB-regulated network (blue) or IRF-regulated network (purple) (corresponding to data illustrated in panel a) are reported. c Genes included in the IRF_Q6 gene set (left; i.e., genes having at least one occurrence of the transcription factor binding site V$IRF_Q6 (v7.4 TRANSFAC) in the regions spanning up to 4 kb around their transcription starting sites) and the significant 10(Z)- hexadecenoic acid-dependent differentially expressed genes with q < 0.1 (right) were queried against the Interferome database (v2.0) to identify their association with known interferon responses. The majority of genes in both gene sets are attributed to both type I and type II interferon responses. Abbreviations: CCL2, C-C motif chemokine ligand 2; CXCL1, C-X-C motif chemokine ligand 1; CXCL2, C-X-C motif chemokine ligand 2; IER3, immediate early response 3; IFN, interferon; IFNA, interferon alpha; IFNB1, interferon beta 1; IKK, inhibitor of nuclear factor kappa B kinase; INHBA, inhibin subunit beta A; IL1A, interleukin 1 alpha; IL1B, interleukin 1 beta; IL1R, interleukin 1 receptor; IL6, interleukin 6; JUNB, junB proto-oncogene, AP-1 transcription factor subunit; LPS, lipopolysaccharide; NFKB, nuclear factor kappa B; NFKB1, nuclear factor kappa B subunit 1; PLAUR, plasminogen activator, urokinase receptor; PSMB8, proteasome subunit beta 8; PSMB9, proteasome subunit beta 9; PSMB10, proteasome subunit beta 10; PSME1, proteasome activator subunit 1; PTGS2, prostaglandin-endoperoxide synthase 2; STAT3, signal transducer and activator of transcription 3; TNF, tumor necrosis factor
The anti-inflammatory effects of 10(Z)-hexadecenoic acid are mediated through PPARα
10(Z)-hexadecenoic acid specifically activates PPARα
Fatty acids can modulate inflammation via the activation n of nuclear hormone receptors (Chinetti et al. 2000; Kidani and Bensinger 2012). Therefore, we assessed the nuclear receptor activation capacity of (1) the triacylglycerol (TAG), 1, 2, 3- tri [ Z - 10 - he xa de ce no yl] gly ce rol ; (2 ) t h e monoacylglycerol (MAG), 1 -[ Z −10-hexadeceno yl]glycerol; and (3) the free fatty acid (FFA), 10(Z)- hexadecenoic acid. We conducted reporter gene assays via the transfection of COS1 cells using GAL4-fusion ligand binding domains (LBDs) of various lipid-activated nuclear receptors (PPARα-LBD, PPARγ-LBD, PPARδ-LBD, and RARα-LBD) along with a plasmid carrying MH100-TK- luciferase reporter (Chen and Evans 1995). Transfected cells were incubated with TAG, MAG, or FFA for 18 h, and relative luciferase activity, normalized to β- galactosidase activity, was measured. Each reporter transfection was validated with the respective receptor agonist (PPARα, WY-14643; PPARγ, rosiglitazone (RSG); PPARδ, GW1516; RARα, AM580). Both the MAG and FFA, at concentrations of 80 μM, reliably increased PPARα-, but not PPARγ-, PPARδ-, or RARα-regulated reporter expression (Fig. 5a–d). The triglyceride had no effect (Fig. 5a–d). Together, these results demonstrate that 10(Z)- hexadecenoic acid and its monoacylglycerol form selectively activate the PPARα receptor.
Fig. 5.

Analysis of the effects of M. vaccae-derived lipids on peroxisome proliferator-activated receptor (PPAR) α, PPARγ, PPARδ, and retinoic acid receptor (RAR) α signaling in transfection assays using COS-1 cells. a Relative activity of PPARα following incubation with the 1,2,3-tri [Z- 10-hexadecenoyl] glycerol (PI-70; TAG), monoacylglycerol, 1-[Z-10- hexadecenoyl] glycerol (PI-69; MAG), or 10(Z)-hexadecenoic acid (PI- 71; FFA) for 18 h, expressed as relative luciferase activity, normalized to β-galactosidase activity. b Relative activity of PPARγ. c Relative activity of PPARδ. d Relative activity of RARα. Abbreviations and concentrations: AM580 (RARα-specific agonist, 100 nM); GW1516 (PPARδ agonist, 1 μM); RSG, rosiglitazone (PPARγ agonist, 2.5 μM); troglitazone (PPARγ agonist, 10 μM); WY-14643 (PPARα agonist, 2 μM). Data are representative of two to three replicates per experiment
PPARα is required for anti-inflammatory effects of 10(Z) -hexadecenoic acid
Next, we investigated if this interaction was necessary for inhibiting LPS-stimulated release of IL-6. Agonists and antagonists of each PPAR were used to test if PPARα has a singular role in this process. The agonists and antagonists and their receptor specificities are listed in Table S9. Macrophages were incubated with a single PPAR antagonist for 1 h prior to treatment with either 200 μM 10(Z)-hexadecenoic acid or a PPAR agonist complementary to its respective PPAR antagonist. After another 1 h incubation period, the cells were stimulated with LPS (1 μg/mL), and IL-6 was measured 12 h later. Only with the PPARα antagonist, GW 6471, could the anti-inflammatory effects of 10(Z)-hexadecenoic acid be significantly reversed (Fig. 6a). The effects of the PPARγ and PPARδ antagonists were comparable to media (Fig. 6a). These results suggest a selective interaction between 10(Z)-hexadecenoic acid and PPARα, as the PPARα antagonist, GW 6471, had no effect on macrophage viability (Fig. S4), while it was effective in reversing the anti-inflammatory effects of the PPARα agonist, WY-14643, as measured by IL-6 secretion in LPS-stimulated macrophages (Fig. S5). To further explore the role of PPARα in the anti-inflammatory effects of 10(Z)-hexadecenoic acid, we repeated the assay with freshly isolated peritoneal macrophages from adult male C57BL/6J wild-type and PPARα−/− mice. As expected, 10(Z)-hexadecenoic acid suppressed LPS-stimulated IL-6 in macrophages from wild-type C57BL/6J mice, but this effect was absent in macrophages from PPARα KO mice (Fig. 6b). This indicated a full reversal of the anti-inflammatory effect of 10(Z)-hexadecenoic acid and the necessity of PPARα in mediating the effect.
Fig. 6.

PPARα is required for suppression of LPS-induced inflammation in macrophages. A peroxisome proliferator-activated receptor (PPAR) α, γ, or δ antagonist (GW 6471, GW 9662, GSK 0660, respectively) or vehicle was applied to murine peritoneal macrophages followed by treatment with either 10(Z)-hexadecenoic acid (200 μM), vehicle, or dexamethasone (Dex; 10 μM), then stimulated with lipopolysaccharide (LPS; 1 μg/mL). a After 12 h, interleukin (IL) 6 was measured in the cell supernatant and reported relative to vehicle controls. b The necessity of PPARα was shown in a PPARα knockout (KO) model. Murine peritoneal macrophages from PPARα−/− or WT mice were incubated with either 10(Z)-hexadecenoic acid (50 or 200 μM) or vehicle, then stimulated with LPS (1 μg/mL). #p < 0.05, Fisher’s least significant difference (LSD), relative to cells only treated with 10(Z)-hexadecenoic acid. *p < 0.05 relative to KO. Abbreviations: 10H, 10(Z)-hexadecenoic acid; Dex, dexamethasone; IL-6, interleukin 6; KO, PPARα knockout; WT, wild type
Discussion
Here, we characterized the monounsaturated C16 free fatty acid, 10(Z)-hexadecenoic acid, derived from M. vaccae NCTC 11659, a saprophytic bacterium with anti-inflammatory and immunoregulatory properties that previously has been shown to prevent stress-induced exaggeration of peripheral inflammation and neuroinflammation and to prevent stress-induced exaggeration of anxiety- and fear-related defensive behavioral responses. In addition, we showed that 10(Z)-hexadecenoic acid induced a broad transcriptional repression of inflammatory gene markers (see, for example, Tables S10–11) and suppressed IL-6 secretion from freshly isolated, LPS-stimulated, murine peritoneal macrophages. Furthermore, we showed that both the monoacylated glycerol, 1-[Z-10-hexadecenoyl] glycerol and 10(Z)-hexadecenoic acid, activated PPARα signaling, as measured by transfection assays. Finally, we showed that PPARα antagonists prevented the anti-inflammatory effects of 10(Z)-hexadecenoic acid in macrophages, while the ex vivo effects of the lipid were absent in macrophages isolated from PPARα-deficient mice.
Here, we focused on the effects of 10(Z)-hexadecenoic acid on peritoneal macrophages. Based on a number of lines of evidence, the effects of 10(Z)-hexadecenoic acid actions on peritoneal macrophages may have important implications for CNS immunity and subsequent behavioral outcomes. Intraperitoneal administration of LPS is known to induce priming of hippocampal microglia and worsen CNS outcomes (Cunningham 2005, 2013; Cunningham et al. 2009). Although the mechanisms through which peripheral inflammation signals to the CNS to induce microglial priming and neuroinflammatory responses are not entirely clear, a number of potential signaling mechanisms have been proposed. These include (1) entry of cytokines into the brain at circumventricular organs that have a reduced blood–brain barrier; (2) binding of cytokines to cerebralvascular endothelium, inducingthe secretionofcentral neuroinflammatory mediators; (3) carrier-mediated transport of immune signals into the brain, across the blood–brain barrier; (4) migration of proinflammatory monocytes from the periphery to the CNS; and (5) activation of peripheral afferent nerves, including vagal and nonvagal pathways (Watkins et al. 1995; Maier et al. 1998; Maier 2003; Miller et al. 2010; Miller and Raison 2016).
Together, these data support the hypothesis that bacterially derived 10(Z)-hexadecenoic acid may induce a form of macrophage “inflammation anergy” (i.e., a condition characterized by the absence of the normal immune response to a particular antigen, see, for example, Smythies et al. 2005, 2010) through actions on PPARα. Peroxisome proliferator-activated receptors, PPARα, PPARγ, and PPARδ, are ligand-activated nuclear receptors, each of which acts as a heterodimer with retinoid X receptor (RXR), with potent anti-inflammatory properties, through interference with proinflammatory transcription factor pathways (Chinetti et al. 2003). PPARα−/− mice have increased vulnerability to chemically induced colitis, experimental autoimmune encephalitis (EAE, a model of multiple sclerosis), and experimentally induced allergic asthma, consistent with the hypothesis that endogenous PPARα suppresses inflammatory signaling in these models (for review, Bensinger and Tontonoz 2008). Activation of PPARα in macrophages inhibits the production of proinflammatory response markers, including IL-6, IL-1β, TNF, and inducible nitric oxide synthase (Xu et al. 2005; Paukkeri et al. 2007). Interaction between PPARα and TLR4 signaling has been observed in other endogenous systems, like vascular smooth muscle cells, where responses to activation of TLR4 with LPS are mitigated by a PPARα agonist (Ji et al. 2010). The anti-inflammatory effects were mediated, in part, by a reduction of tissue inhibitor of metalloprotease-1 (TIMP-1), which was also reduced in our study (Table S3). PPARα- mediated inhibition of TLR4 signaling has also been shown in enteric glial cells (Esposito et al. 2014), and a potential downstream target of PPARα-mediated suppression, TRIF, is required for LPS-induced activation of microglia (Burfeind et al. 2018). TRIF KO mice have attenuated expression of Il6, Ccl2, and Cxcl2, which were all also suppressed in our study (Table S3), in the hypothalamus after peripheral LPS stimulation (Burfeind et al. 2018). Furthermore, bacterially derived agonists of PPARs have potential for modulation of hostacquired immunity; PPARs have been found to regulate T- cell survival, activation, and CD4+ T-helper cell differentiation into the Th1, Th2, Th17, and Treg lineages (Choi and Bothwell 2012).
Synthesis of 10(Z)-hexadecenoic acid by mycobacteria may be an example of molecular mimicry of eukaryotic signaling. Endogenous host-derived agonists of PPARα include 16:1 isoforms of palmitoleic acid (Kliewer et al. 1997; Kota et al. 2005), a lipokine released from adipose cells. Palmitoleic acid localizes predominantly to nuclear fractions, consistent with a nuclear mechanism of action in host cells (Foryst-Ludwig et al. 2015), and is potently antiinflammatory (Chan et al. 2015). In addition, the endocannabinoid, palmitoylethanolamide (PEA), acts as an agonist at PPARα (Verme et al. 2005; Guida et al. 2017). Of interest to trauma- and stressor-related psychiatric disorders, PEA induces potent antidepressant-like behavioral responses (Yu et al. 2011) and, through induction of cannabinoid 2 receptors, alters the phenotype of macrophages and microglia (Guida et al. 2017). Recent studies have demonstrated PEA increases the biosynthesis of allopregnanolone, an endocannabinoid, in the spinal cord, brainstem, hippocampus, and amygdala, effects that are associated with faster fear extinction learning and improvement of aggression in socially isolated mice (Sasso et al. 2012; Locci and Pinna 2017; Pinna 2018). Future studies should determine if 10(Z)-hexadecenoic acid is sufficient to induce the enhanced fear extinction learning previously demonstrated using whole, heat-killed M. vaccae (Fox et al. 2017), and to what extent these effects are mediated by PPARα.
Mycobacteria are unique in that they accumulate triacylglycerols as intracellular lipophilic inclusions. For example, Mycobacterium smegmatis accumulates triacylglycerols and the acyl chain composition varies depending on the growth medium (Garton et al. 2002). Monounsaturated fatty acids, C16:1 hexadecenoic acid and C18:1 octadecenoic acid, were found to be high when bacteria were grown in nutrient-rich Middlebrook 7H9 broth, relative to low-nitrogen Youmans’ broth, but highest when bacteria were grown in Youmans’ broth with monounsaturated oleic acid ((9Z)- octadec-9-enoic acid) supplementation. Thus, it is possible that mycobacteria synthesize and store triacylglycerols using environmental fatty acids as substrates, potentially for export to the cell envelope and release. If so, it may be possible to modify the immunoregulatory and anti-inflammatory potential of mycobacteria through modification of growth conditions.
Of potential importance, conjugated linoleic acids are bacterial metabolites. For example, specific members of the genus Lactobacillus, including Lactobacillus reuteri and L. plantarum, as well as bifidobacteria, mediate the conversion of dietary linoleic acid into immunomodulatory conjugated linoleic acids (Coakley et al. 2003; Lee et al. 2003; Ogawa et al. 2005; Kishino et al. 2013). Most of the conjugated linoleic acid produced is located in the extracellular space (~ 98%) (Lee et al. 2003; Roman-Nunez et al. 2007), suggesting that bacterially derived conjugated linoleic acids may be metabolic signaling molecules that modulate the host immune response. These bacterially derived fatty acid metabolites include 10-hydroxy-cis-12- octadecenoic acid (HYA), cis-9,trans-11-linoleic acid, trans-9,cis-11-linoleic acid, and cis-10,trans-12-linoleic acid (Lee et al. 2003; Miyamoto et al. 2015), among many others (Ogawa et al. 2005). Several of these bacterially derived fatty acid metabolites are potent PPARα agonists (IC50 values from 140 to 400 nM) (Moya-Camarena et al. 1999). Perhaps the closest analogue of 10(Z)-hexadecenoic acid identified here is trans-10-octadecenoic acid, produced by L. plantarum from linoleic acid (Kishino et al. 2013) or γ-linolenic acid (Ogawa et al. 2005). Although, to the best of our knowledge, the efficacy of trans-10-octadecenoic acid at PPARα receptors is not known, production of 10(Z)-hexadecenoic acid and diverse conjugated linoleic acids, which then act at host PPARα receptors, may be a general strategy of commensal organisms to suppress host immune responses, and promote symbiotic relationships with the host. Consistent with this hypothesis, macrophages lining the gut mucosa are anergic, characterized by an inability to mount proinflammatory responses, despite avid phagocytic activity (Smythies et al. 2005), while lung airway macrophages are immunoregulatory (Strickland et al. 1996; Soroosh et al. 2013; Duan and Croft 2014). Recent studies have also identified α-linolenic acid-derived bacterial metabolites, 13-hydroxy-9(Z),15(Z)-octadecadienoic acid (13-OH) and 13-oxo-9(Z),15(Z)-octadecadienoic acid (13-oxo), that induce differentiation of anti-inflammatory M2 macrophages through activation of G protein-coupled receptor 40 (GPR40) (Ohue-Kitano et al. 2018). Together, these data support the hypothesis that bacterially derived “postbiotic” compounds, including fatty acid metabolites, have important beneficial effects on the host via diverse host receptor signaling mechanisms.
Although we did not assess the effects of 10(Z)- hexadecenoic acid on DCs or immunoregulation, defined as the balance between regulatory and effector T cells, conjugated linoleic acid suppresses NF-κB signaling and IL-12 production in DCs through IL-10 production (Loscher et al. 2005). Exposure of murine DCs to conjugated linoleic acid suppresses their ability to promote differentiation of naïve T cells into Th1 and/or Th17 cells in vitro following their adoptive transfer in vivo (Draper et al. 2014). Future studies should investigate the effects of 10(Z)-hexadecenoic acid on inflammatory signaling in macrophages, DCs, as well as on T-cell differentiation and function, the potential role of PPARα in these effects, and consequences for stress-induced exaggeration of anxiety- and fear-related behavioral responses.
Overall, our data suggest that chemical mimicry of eukaryotic signaling molecules may be common among environmental bacteria, including mycobacteria (Gebert et al. 2018), that are abundant in host mucosal surfaces (Macovei et al. 2015), and bacterially derived anti-inflammatory lipids have potential as a novel approach to therapeutic intervention in inflammatory disease and stress-related psychiatric disorders, where immunodysregulation and inappropriate inflammation have been identified as risk factors (Rohleder 2014; Langgartner et al. 2018).
Supplementary Material
Acknowledgements
We are grateful to Zachary D. Barger for proofreading the manuscript. We thank the University of Colorado Boulder BioFrontiers Institute Next-Gen Sequencing Core Facility, which performed the Illumina sequencing.
Funding information This work was supported by the National Institute of Mental Health (grant number 1R21MH116263; CAL). Dr. Christopher A. Lowry is supported by the Department of the Navy, Office of Naval Research Multidisciplinary University Research Initiative (MURI) Award (grant number N00014–15-1–2809), Department of Veterans Affairs Office of Research and Development (VA-ORD) RR&D Small Projects in Rehabilitation Research (SPiRE) (I21) (grant number 1 I21 RX002232–01), Colorado Clinical & Translational Sciences Institute (CCTSI) Center for Neuroscience (grant number CNSTT-15–145), the Colorado Department of Public Health and Environment (CDPHE; grant number DCEED-3510), and the Alfred P. Sloan Foundation (grant number, G-2016–7077). Dr. Robin Dowell is supported by NSF Career MCB #1350915.
Abbreviations
- CD
Cluster of differentiation
- CNS
Central nervous system
- DC
Dendritic cell
- DSM-5
Diagnostic and Statistical Manual of Mental Disorders (5th ed.)
- IL
Interleukin
- IFN
Interferon
- IRF
Interferon regulatory factor
- LPS
Lipopolysaccharide
- MGB
Microbiota–gut–brain
- NCTC
National Collection of Type Cultures
- NF-κB
Nuclear factor kappa-light-chain-enhancer of activated B cells
- PEA
Palmitoylethanolamide
- PPAR
Peroxisome proliferator-activated receptor
- PTSD
Posttraumatic stress disorder
- RAR
Retinoic acid receptor
- TGFβ
Transforming growth factor beta
- TLR
Toll-like receptor
- Treg
Regulatory T cell
Footnotes
Compliance with ethical standards
All experimental protocols were consistent with the National Institutes of Health Guide for the Care and Use of Laboratory Animals, Eighth Edition (The National Academies Press 2011), and the Institutional Animal Care and Use Committee at the University of Colorado Boulder approved all procedures. This work was covered under CU Boulder IACUC Protocol Numbers 2134–14MAY2018 and 2361–14MAY2018-DT. The research described here was conducted in compliance with The ARRIVE Guidelines: Animal research: reporting of in vivo experiments, originally published in PLOS Biology, June 2010 (Kilkenny and Altman 2010).
Conflict of interest Christopher A. Lowry serves on the Scientific Advisory Board of Immodulon Therapeutics Ltd. Dr. Robin Dowell is a founder and scientific advisor of Arpeggio Biosciences.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- Adams VC, Hunt JRF, Martinelli R et al. (2004) Mycobacterium vaccae induces a population of pulmonary CD11c+ cells with regulatory potential in allergic mice. Eur J Immunol 34:631–638. 10.1002/eji.200324659 [DOI] [PubMed] [Google Scholar]
- American Psychiatric Association; (2013) Diagnostic and statistical manual of mental disorder (5th ed.). American Psychiatric Association, Arlington, VA [Google Scholar]
- Anders S, Huber W (2010) Differential expression analysis for sequence count data. Genome Biol 11:R106 10.1186/gb-2010-11-10-r106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anders S, Pyl PT, Huber W (2015) HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31:166–169. 10.1093/bioinformatics/btu638 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arteaga Figueroa L, Abarca-Vargas R, García Alanis C, Petricevich VL (2017) Comparison between peritoneal macrophage activation by Bougainvillea xbuttiana extract and LPS and/or interleukins. Biomed Res Int 2017:1–11. 10.1155/2017/4602952 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baxter AJ, Scott KM, Vos T, Whiteford HA (2013) Global prevalence of anxiety disorders: a systematic review and meta-regression. Psychol Med 43:897–910 [DOI] [PubMed] [Google Scholar]
- Benko S, Love JD, Beládi M, Schwabe JWR, Nagy L (2003) Molecular determinants of the balance between co-repressor and co-activator recruitment to the retinoic acid receptor. J Biol Chem 278:43797–43806. 10.1074/jbc.M306199200 [DOI] [PubMed] [Google Scholar]
- Bensinger SJ, Tontonoz P (2008) Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 454:470–477. 10.1038/nature07202 [DOI] [PubMed] [Google Scholar]
- Blaser MJ (2017) The theory of disappearing microbiota and the epidemics of chronic diseases. Nat Rev Immunol 17:461–463 [DOI] [PubMed] [Google Scholar]
- Bloomfield SF, Rook GA, Scott EA et al. (2016) Time to abandon the hygiene hypothesis: new perspectives on allergic disease, the human microbiome, infectious disease prevention and the role of targeted hygiene. Perspect Public Health 136:213–224. 10.1177/1757913916650225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolger AM, Lohse M, Usadel B (2014) Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30:2114–2120. 10.1093/bioinformatics/btu170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Böttger EC, Hirschel B, Coyle MB (1993) Mycobacterium genavense. Int J Syst Bacteriol 43:841–843. 10.1099/00207713-43-4-841 [DOI] [PubMed] [Google Scholar]
- Burfeind KG, Zhu X, Levasseur PR, Michaelis KA, Norgard MA, Marks DL (2018) TRIF is a key inflammatory mediator of acute sickness behavior and cancer cachexia. Brain Behav Immun 73:364–374. 10.1016/j.bbi.2018.05.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan KL, Pillon NJ, Sivaloganathan DM, Costford SR, Liu Z, Théret M, Chazaud B, Klip A (2015) Palmitoleate reverses high fat-induced proinflammatory macrophage polarization via AMP-activated protein kinase (AMPK). J Biol Chem 290:16979–16988. 10.1074/jbc.M115.646992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen JD, Evans RM (1995) A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377:454–457 [DOI] [PubMed] [Google Scholar]
- Chinetti G, Fruchart JC, Staels B (2000) Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm Res 49:497–505 [DOI] [PubMed] [Google Scholar]
- Chinetti G, Fruchart JC, Staels B (2003) Peroxisome proliferator-activated receptors: new targets for the pharmacological modulation of macrophage gene expression and function. Curr Opin Lipidol 14: 459–468. 10.1097/01.mol.0000092630.86399.00 [DOI] [PubMed] [Google Scholar]
- Choi JM, Bothwell ALM (2012) The nuclear receptor PPARs as important regulators of T-cell functions and autoimmune diseases. Mol Cell 33:217–222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chou S, Chedore P, Kasatiya S (1998) Use of gas chromatographic fatty acid and mycolic acid cleavage product determination to differentiate among Mycobacterium genavense, Mycobacterium fortuitum, Mycobacterium simiae, and Mycobacterium tuberculosis. J Clin Microbiol 36:577–579 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coakley M, Ross RP, Nordgren M, Fitzgerald G, Devery R, Stanton C (2003) Conjugated linoleic acid biosynthesis by human-derived Bifidobacterium species. J Appl Microbiol 94:138–145. 10.1046/j.1365-2672.2003.01814.x [DOI] [PubMed] [Google Scholar]
- Committee for the Update of the Guide for the Care and Use of Laboratory Animals, Institute for Laboratory Animal Research, Division on Earth and Life Sciences, National Research Council of the National Academies (2011) Guide for the Care and Use of Laboratory Animals The National Academies Press, Washington D.C. [Google Scholar]
- Coyle MB, Carlson LDC, Wallis CK et al. (1992) Laboratory aspects of Mycobacterium genavense, a proposed species isolated from AIDS patients. J Clin Microbiol 30(12):3206–3212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cryan JF, Dinan TG (2012) Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci 13: 701–712. 10.1038/nrn3346 [DOI] [PubMed] [Google Scholar]
- Cryan JF, Dinan TG (2015) More than a gut feeling: the microbiota regulates neurodevelopment and behavior. Neuropsychopharmacology 40:241–242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunningham C (2005) Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. J Neurosci 25:9275–9284. 10.1523/jneurosci.2614-05.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunningham C (2013) Microglia and neurodegeneration: the role of systemic inflammation. Glia 61:71–90. 10.1002/glia.22350 [DOI] [PubMed] [Google Scholar]
- Cunningham C, Campion S, Lunnon K, Murray CL, Woods JFC, Deacon RMJ, Rawlins JNP, Perry VH (2009) Systemic inflammation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biol Psychiatry 65:304–312. 10.1016/j.biopsych.2008.07.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Draper E, DeCourcey J, Higgins SC, Canavan M, McEvoy F, Lynch M, Keogh B, Reynolds C, Roche HM, Mills KHG, Loscher CE (2014) Conjugated linoleic acid suppresses dendritic cell activation and subsequent Th17 responses. J Nutr Biochem 25:741–749. 10.1016/j.jnutbio.2014.03.004 [DOI] [PubMed] [Google Scholar]
- Duan W, Croft M (2014) Control of regulatory T cells and airway tolerance by lung macrophages and dendritic cells. Ann Am Thorac Soc 11: S306–S313. 10.1513/AnnalsATS.201401-028AW [DOI] [PMC free article] [PubMed] [Google Scholar]
- Esposito G, Capoccia E, Turco F, Palumbo I, Lu J, Steardo A, Cuomo R, Sarnelli G, Steardo L (2014) Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-α activation. Gut 63:1300–1312. 10.1136/gutjnl-2013-305005 [DOI] [PubMed] [Google Scholar]
- Forsythe P, Sudo N, Dinan T, Taylor VH, Bienenstock J (2010) Mood and gut feelings. Brain Behav Immun 24:9–16. 10.1016/j.bbi.2009.05.058 [DOI] [PubMed] [Google Scholar]
- Foryst-Ludwig A, Kreissl MC, Benz V, Brix S, Smeir E, Ban Z, Januszewicz E, Salatzki J, Grune J, Schwanstecher AK, Blumrich A, Schirbel A, Klopfleisch R, Rothe M, Blume K, Halle M, Wolfarth B, Kershaw EE, Kintscher U (2015) Adipose tissue lipolysis promotes exercise-induced cardiac hypertrophy involving the lipokine C16:1n7-palmitoleate. J Biol Chem 290:23603–23615. 10.1074/jbc.M115.645341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fox JH, Hassell JE, Siebler PH et al. (2017) Preimmunization with a heat- killed preparation of Mycobacterium vaccae enhances fear extinction in the fear-potentiated startle paradigm. Brain Behav Immun 66:70–84. 10.1016/j.bbi.2017.08.014 [DOI] [PubMed] [Google Scholar]
- Frank MG, Fonken LK, Dolzani SD, Annis JL, Siebler PH, Schmidt D, Watkins LR, Maier SF, Lowry CA (2018) Immunization with Mycobacterium vaccae induces an anti-inflammatory milieu in the CNS: attenuation of stress-induced microglial priming, alarmins and anxiety-like behavior. Brain Behav Immun 73:352–363. 10.1016/j.bbi.2018.05.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garn H, Bahn S, Baune BT, Binder EB, Bisgaard H, Chatila TA, Chavakis T, Culmsee C, Dannlowski U, Gay S, Gern J, Haahtela T, Kircher T, Müller-Ladner U, Neurath MF, Preissner KT, Reinhardt C, Rook G, Russell S, Schmeck B, Stappenbeck T, Steinhoff U, van Os J, Weiss S, Zemlin M, Renz H (2016) Current concepts in chronic inflammatory diseases: interactions between microbes, cellular metabolism, and inflammation. J Allergy Clin Immunol 138:47–56. 10.1016/j.jaci.2016.02.046 [DOI] [PubMed] [Google Scholar]
- Garton NJ, Christensen H, Minnikin DE et al. (2002) Intracellular lipophilic inclusions of mycobacteria in vitro and in sputum. Microbiology 148:2951–2958. 10.1099/00221287-148-10-2951 [DOI] [PubMed] [Google Scholar]
- Gebert MJ, Delgado-Baquerizo M, Oliverio AM Webster TM, Nichols LM, Honda JR, Chan ED, Adjemian J, Dunn RR, Fierer N (2018) Ecological analyses of mycobacteria in showerhead biofilms and their relevance to human health. MBio 9:e01614–18. 10.1128/mBio.01614-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guida F, Luongo L, Boccella S, Giordano ME, Romano R, Bellini G, Manzo I, Furiano A, Rizzo A, Imperatore R, Iannotti FA, D’Aniello E, Piscitelli F, sca Rossi F, Cristino L, di Marzo V, de Novellis V, Maione S (2017) Palmitoylethanolamide induces microglia changes associated with increased migration and phagocytic activity: involvement of the CB2 receptor. Sci Rep 7:1–11. 10.1038/s41598-017-00342-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gutzwiller MER, Reist M, Peel JE, Seewald W, Brunet LR, Roosje PJ (2007) Intradermal injection of heat-killed Mycobacterium vaccae in dogs with atopic dermatitis: a multicentre pilot study. Vet Dermatol 18:87–93. 10.1111/j.1365-3164.2007.00579.x [DOI] [PubMed] [Google Scholar]
- Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, McArthur S (2018) Microbiome–host systems interactions: protective effects of propionate upon the blood–brain barrier. Microbiome 6:55 10.1186/s40168-018-0439-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang DW, Sherman BT, Lempicki RA et al. (2009) Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37:1–13. 10.1093/nar/gkn923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ji Y, Wang Z, Li Z, Liu J (2010) Modulation of LPS-mediated inflammation by fenofibrate via the TRIF-dependent TLR4 signaling pathway in vascular smooth muscle cells. Cell Physiol Biochem 25:631–640. 10.1159/000315082 [DOI] [PubMed] [Google Scholar]
- Kawasaki T, Kawai T (2014) Toll-like receptor signaling pathways. Front Immunol 5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kersten S (2014) Integrated physiology and systems biology of PPARα. Mol Metab 3:354–371. 10.1016/j.molmet.2014.02.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kidani Y, Bensinger S (2012) Liver X receptor and peroxisome proliferator-activated receptor as integrators of lipid homeostasis and immunity. Immunol Rev 249:72–83. 10.1111/j.1600-065X.2012.01153.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kilkenny C, Altman DG (2010) Improving bioscience research reporting: ARRIVE-ing at a solution. Lab Anim 44:377–378. 10.1258/la.2010.0010021 [DOI] [PubMed] [Google Scholar]
- Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL (2013) TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol 14:R36 10.1186/gb-2013-14-4-r36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kishino S, Takeuchi M, Park S-B, Hirata A, Kitamura N, Kunisawa J, Kiyono H, Iwamoto R, Isobe Y, Arita M, Arai H, Ueda K, Shima J, Takahashi S, Yokozeki K, Shimizu S, Ogawa J (2013) Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci U S A 110: 17808–17813. 10.1073/pnas.1312937110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, Lehmann JM (1997) Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci U S A 94:4318–4323. 10.1073/pnas.94.9.4318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kota BP, Huang THW, Roufogalis BD (2005) An overview on biological mechanisms of PPARs. Pharmacol Res 51:85–94 [DOI] [PubMed] [Google Scholar]
- Langgartner D, Lowry CA, Reber SO (2018) Old friends, immunoregulation, and stress resilience. Pflugers Arch - Eur J Physiol 471:237–269. 10.1007/s00424-018-2228-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Bert N, Chain BM, Rook G, Noursadeghi M (2011) DC priming by M. vaccae inhibits Th2 responses in contrast to specific TLR2 priming and is associated with selective activation of the CREB pathway. PLoS One 6:e18346 10.1371/journal.pone.0018346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leclercq S, Forsythe P, Bienenstock J (2016) Posttraumatic stress disorder: does the gut microbiome hold the key? Can J Psychiatr 61:204–213. 10.1177/0706743716635535 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SO, Hong GW, Oh DK (2003) Bioconversion of linoleic acid into conjugated linoleic acid by immobilized Lactobacillus reuteri. Biotechnol Prog 19:1081–1084. 10.1021/bp0257933 [DOI] [PubMed] [Google Scholar]
- Lee AJ, Cho KJ, Kim JH (2015) MyD88-BLT2-dependent cascade contributes to LPS-induced interleukin-6 production in mouse macrophage. Exp Mol Med 47 10.1038/emm.2015.8 [DOI] [PubMed] [Google Scholar]
- Lin JY, Tang CY (2007) Interleukin-10 administration inhibits TNF-alpha and IL-1beta, but not IL-6, secretion of LPS-stimulated peritoneal macrophages. J Food Drug Anal 15(1):48–54 [Google Scholar]
- Locci A, Pinna G (2017) Neurosteroid biosynthesis down-regulation and changes in GABAA receptor subunit composition: a biomarker axis in stress-induced cognitive and emotional impairment. Br J Pharmacol 174:3226–3241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loscher CE, Draper E, Leavy O, Kelleher D, Mills KHG, Roche HM (2005) Conjugated linoleic acid suppresses NF-κB activation and IL-12 production in dendritic cells through ERK-mediated IL-10 induction. J Immunol 175:4990–4998 [DOI] [PubMed] [Google Scholar]
- Lowry CA, Hollis JH, de Vries A et al. (2007) Identification of an immune-responsive mesolimbocortical serotonergic system: potential role in regulation of emotional behavior. Neuroscience 146:756–772. 10.1016/j.neuroscience.2007.01.067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowry CA, Smith DG, Siebler PH, Schmidt D, Stamper CE, Hassell JE, Yamashita PS, Fox JH, Reber SO, Brenner LA, Hoisington AJ, Postolache TT, Kinney KA, Marciani D, Hernandez M, Hemmings SMJ, Malan-Muller S, Wright KP, Knight R, Raison CL, Rook GAW (2016) The microbiota, immunoregulation, and mental health: implications for public health. Curr Environ Health Rep 3:270–286. 10.1007/s40572-016-0100-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lyte M (2014) Microbial endocrinology and the microbiota-gut-brain axis. Adv Exp Med Biol 817:3–24. 10.1007/978-1-4939-0897-4_1 [DOI] [PubMed] [Google Scholar]
- Macovei L, McCafferty J, Chen T, Teles F, Hasturk H, Paster BJ, Campos-Neto A (2015) The hidden “mycobacteriome” of the human healthy oral cavity and upper respiratory tract. J Oral Microbiol 7:1–11. 10.3402/jom.v7.26094 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maier SF (2003) Bi-directional immune-brain communication: implications for understanding stress, pain, and cognition. Brain Behav Immun 17:69–85 [DOI] [PubMed] [Google Scholar]
- Maier SF, Goehler LE, Fleshner M, Watkins LR (1998) The role of the vagus nerve in cytokine-to-brain communication. Ann N YAcad Sci 840:289–300 [DOI] [PubMed] [Google Scholar]
- Merico D, Isserlin R, Bader GD (2011) Visualizing gene-set enrichment results using the cytoscape plug-in enrichment map. Methods Mol Biol 781:257–277. 10.1007/978-1-61779-276-2_12 [DOI] [PubMed] [Google Scholar]
- Miller AH, Raison CL (2016) The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol 16:22–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller AH, Maletic V, Raison CL (2009) Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol Psychiatry 65:732–41. 10.1016/j.psiq.2010.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyamoto J, Mizukure T, Park SB, Kishino S, Kimura I, Hirano K, Bergamo P, Rossi M, Suzuki T, Arita M, Ogawa J, Tanabe S (2015) A gut microbial metabolite of linoleic acid, 10-hydroxycis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. J Biol Chem 290:2902–2918. 10.1074/jbc.M114.610733 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moya-Camarena SY, Vanden Heuvel JP, Blanchard SG et al. (1999) Conjugated linoleic acid is a potent naturally occurring ligand and activator of PPARα. J Lipid Res 40:1426–1433. 10.1631/jzus.B1200175 [DOI] [PubMed] [Google Scholar]
- Nagy L, Kao HY, Love JD, Li C, Banayo E, Gooch JT, Krishna V, Chatterjee K, Evans RM, Schwabe JWR (1999) Mechanism of corepressor binding and release from nuclear hormone receptors. Genes Dev 13:3209–3216. 10.1101/gad.13.24.3209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nugent NR, Tyrka AR, Carpenter LL, Price LH (2011) Gene- environment interactions: early life stress and risk for depressive and anxiety disorders. Psychopharmacology 214:175–196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Donovan A, Cohen BE, Seal KH, Bertenthal D, Margaretten M, Nishimi K, Neylan TC (2015) Elevated risk for autoimmune disorders in Iraq and Afghanistan veterans with posttraumatic stress disorder. Biol Psychiatry 77:365–374. 10.1016/j.biopsych.2014.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogawa J, Kishino S, Ando A, Sugimoto S, Mihara K, Shimizu S (2005) Production of conjugated fatty acids by lactic acid bacteria. J Biosci Bioeng 100:355–364. 10.1263/jbb.100.355 [DOI] [PubMed] [Google Scholar]
- Ohue-Kitano R, Yasuoka Y, Goto T, Kitamura N, Park SB, Kishino S, Kimura I, Kasubuchi M, Takahashi H, Li Y, Yeh YS, Jheng HF, Iwase M, Tanaka M, Masuda S, Inoue T, Yamakage H, Kusakabe T, Tani F, Shimatsu A, Takahashi N, Ogawa J, Satoh-Asahara N, Kawada T (2018) A-linolenic acid–derived metabolites from gut lactic acid bacteria induce differentiation of anti-inflammatory M2 macrophages through G protein-coupled receptor 40. FASEB J 32: 304–318. 10.1096/fj.201700273R [DOI] [PubMed] [Google Scholar]
- Okada H, Kuhn C, Feillet H, Bach J-F (2010) The “hygiene hypothesis” for autoimmune and allergic diseases: an update. Clin Exp Immunol 160:1–9. 10.1111/j.1365-2249.2010.04139.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pacífico C, Fernandes P, de Carvalho CCCR (2018) Mycobacterial response to organic solvents and possible implications on crossresistance with antimicrobial agents. Front Microbiol 9 10.3389/fmicb.2018.00961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paukkeri EL, Leppänen T, Sareila O, Vuolteenaho K, Kankaanranta H, Moilanen E (2007) PPARα agonists inhibit nitric oxide production by enhancing iNOS degradation in LPS-treated macrophages. Br J Pharmacol 152:1081–1091. 10.1038/sj.bjp.0707477 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pestka S, Krause CD, Walter MR (2004) Interferons, interferon-like cytokines, and their receptors. Immunol Rev 202:8–32 [DOI] [PubMed] [Google Scholar]
- Pinna G (2018) Biomarkers for PTSD at the interface of the endocannabinoid and neurosteroid axis. Front Neurosci 12:482 10.3389/fnins.2018.00482 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reber SO, Siebler PH, Donner NC, Morton JT, Smith DG, Kopelman JM, Lowe KR, Wheeler KJ, Fox JH, Hassell JE Jr, Greenwood BN, Jansch C, Lechner A, Schmidt D, Uschold-Schmidt N, Füchsl AM, Langgartner D, Walker FR, Hale MW, Lopez Perez G, van Treuren W, González A, Halweg-Edwards AL, Fleshner M, Raison CL, Rook GA, Peddada SD, Knight R, Lowry CA ( 2016) Immunization with a heat-killed preparation of the environmental bacterium Mycobacterium vaccae promotes stress resilience in mice. Proc Natl Acad Sci 113:3130–3139. 10.1073/pnas.1600324113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohleder N (2014) Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom Med 76:181–189 [DOI] [PubMed] [Google Scholar]
- Roman-Nunez M, Cuesta-Alonso EP, Gilliland SE (2007) Influence of sodium glycocholate on production of conjugated linoleic acid by cells of Lactobacillus reuteri ATCC 55739. J Food Sci 72:140–143. 10.1111/j.1750-3841.2007.00347.x [DOI] [PubMed] [Google Scholar]
- Rook GAW (2009) Review series on helminths, immune modulation and the hygiene hypothesis: the broader implications of the hygiene hypothesis. Immunology 126:3–11. 10.1111/j.1365-2567.2008.03007.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook GAW (2010) 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: Darwinian medicine and the “hygiene” or “old friends” hypothesis. Clin Exp Immunol 160:70–79. 10.1111/j.1365-2249.2010.04133.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook GA (2013) Regulation of the immune system by biodiversity from the natural environment: an ecosystem service essential to health. Proc Natl Acad Sci 110:18360–18367. 10.1073/pnas.1313731110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook GAW, Rosa Brunet L (2002) Give us this day our daily germs. Biologist (London) 49:145–149 [PubMed] [Google Scholar]
- Rook GAW, Hamelmann E, Rosa Brunet L (2007) Mycobacteria and allergies. Immunobiology 212:461–473. 10.1016/j.imbio.2007.03.003 [DOI] [PubMed] [Google Scholar]
- Rosa Brunet L, Rook G (2008) United States Patent Application No. US 2008/0004341 A1 Retrieved from https://patents.google.com/patent/US20080004341. Accessed 05 May 2019
- Rusinova I, Forster S, Yu S, Kannan A, Masse M, Cumming H, Chapman R, Hertzog PJ (2013) INTERFEROME v2.0: an updated database of annotated interferon-regulated genes. Nucleic Acids Res 41:D1040–D1046. 10.1093/nar/gks1215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sasso O, Russo R, Vitiello S, Raso GM, D’Agostino G, Iacono A, la Rana G, Vallée M, Cuzzocrea S, Piazza PV, Meli R, Calignano A (2012) Implication of allopregnanolone in the antinociceptive effect of N-palmitoylethanolamide in acute or persistent pain. Pain 153:33–41. 10.1016/j.pain.2011.08.010 [DOI] [PubMed] [Google Scholar]
- Scheuerbrandt G, Bloch K (1962) Unsaturated fatty acids in microorganisms. J Biol Chem 237:2064–2069 [PubMed] [Google Scholar]
- Shacter E, Arzadon GK, Williams JA (1993) Stimulation of interleukin-6 and prostaglandin E2 secretion from peritoneal macrophages by polymers of albumin. Blood 82:2853–2864 [PubMed] [Google Scholar]
- Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, Benjamin WH, Orenstein JM, Smith PD (2005) Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest 115:66–75. 10.1172/JCI200519229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smythies LE, Shen R, Bimczok D, Novak L, Clements RH, Eckhoff DE, Bouchard P, George MD, Hu WK, Dandekar S, Smith PD (2010) Inflammation anergy in human intestinal macrophages is due to Smad-induced IκBα expression and NF-κB inactivation. J Biol Chem 285:19593–19604. 10.1074/jbc.M109.069955 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soroosh P, Doherty TA, Duan W, Mehta AK, Choi H, Adams YF, Mikulski Z, Khorram N, Rosenthal P, Broide DH, Croft M (2013) Lung-resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J Exp Med 210:775–788. 10.1084/jem.20121849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Springer B, Kirschner P, Rost-Meyer G et al. (1993) Mycobacterium interjectum, a new species isolated from a patient with chronic lymphadenitis. J Clin Microbiol 31:3083–3089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stamper CE, Hoisington AJ, Gomez OM et al. (2016) The microbiome of the built environment and human behavior: implications for emotional health and well-being in postmodern western societies. Int Rev Neurobiol 131:289–323. 10.1016/bs.irn.2016.07.006 [DOI] [PubMed] [Google Scholar]
- Strickland D, Kees UR, Holt PG (1996) Regulation of T-cell activation in the lung: isolated lung T cells exhibit surface phenotypic characteristics of recent activation including down-modulated T-cell receptors, but are locked into the G0/G1 phase of the cell cycle. Immunology 87:242–249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suutari M, Laakso S (1993) The effect of growth temperature on the fatty acid composition of Mycobacterium phlei . Arch Microbiol 159: 119–123 [DOI] [PubMed] [Google Scholar]
- Szatmari I, Pap A, Rühl R, Ma JX, Illarionov PA, Besra GS, Rajnavolgyi E, Dezso B, Nagy L (2006) PPARgamma controls CD1d expression by turning on retinoic acid synthesis in developing human dendritic cells. J Exp Med 203:2351–2362. 10.1084/jem.20060141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tay STL, Hemond HF, Polz MF et al. (1998) Two new Mycobacterium strains and their role in toluene degradation in a contaminated stream. Appl Environ Microbiol 64:1715–1720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Travar M, Petkovic M, Verhaz A (2016) Type I, II, and III interferons: regulating immunity to Mycobacterium tuberculosis infection. Arch Immunol Ther Exp 64:19–31 [DOI] [PubMed] [Google Scholar]
- Verme JL, Fu J, Astarita G et al. (2005) The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol 67:15–19. 10.1124/mol.104.006353 [DOI] [PubMed] [Google Scholar]
- Vichai V, Kirtikara K (2006) Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc 1:1112–1116. 10.1038/nprot.2006.179 [DOI] [PubMed] [Google Scholar]
- Watkins LR, Maier SF, Goehler LE (1995) Cytokine-to-brain communication: a review & analysis of alternative mechanisms. Life Sci 57: 1011–1026 [DOI] [PubMed] [Google Scholar]
- Wollenberg GK, DeForge LE, Bolgos G, Remick DG (1993) Differential expression of tumor necrosis factor and interleukin-6 by peritoneal macrophages in vivo and in culture. Am J Pathol 143:1121–1130 [PMC free article] [PubMed] [Google Scholar]
- Xu J, Storer PD, Chavis JA, Racke MK, Drew PD (2005) Agonists for the peroxisome proliferator-activated receptor-α and the retinoid X receptor inhibit inflammatory responses of microglia. J Neurosci Res 81:403–411. 10.1002/jnr.20518 [DOI] [PubMed] [Google Scholar]
- Yu HL, Deng XQ, Li YJ, Li YC, Quan ZS, Sun XY (2011) N-palmitoylethanolamide, an endocannabinoid, exhibits antidepressant effects in the forced swim test and the tail suspension test in mice. Pharmacol Rep 63:834–839. 10.1016/S1734-1140(11)70596-5 [DOI] [PubMed] [Google Scholar]
- Zhang X, Goncalves R, Mosser DM (2008) The isolation and characterization of murine macrophages. Curr Protoc Immunol 14 10.1002/0471142735.im1401s83.The [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuany-Amorim C, Sawicka E, Manlius C, le Moine A, Brunet LR, Kemeny DM, Bowen G, Rook G, Walker C (2002) Suppression of airway eosinophilia by killed Mycobacterium vaccae-induced allergen-specific regulatory T-cells. Nat Med 8:625–629. 10.1038/nm0602-625 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
