Animal Husbandry
All animal experiments were carried out according to UK Home Office regulations in line with the Animals (Scientific Procedures) Act of 1986 under personal and project licenses registered with the UK Home Office. Mice were maintained in-house under standard housing conditions (12-h light/dark cycles), 24 hr access to water and standard chow diet. All mice were of a C57BL6 background (male, 20-30 g, 6-8 weeks of age). Experimenters were blinded to experimental groups during scoring and quantifications. Sample sizes were chosen in accordance with similar previously published experiments.
Intermittent Fasting
Body weight-matched 6-8 week old male C57BL/6 mice were randomly assigned to intermittent fasting (IF), or ad libitum (control) treatment groups. The IF group did not have access to food (fasting) during the first 24 h and then every second day after that (e.g. fasting during 0–24 h, 48–72 h, 96–120 h) with ad libitum access to food on the alternating days (24–48 h, 72–96 h, 120–144 h). Pre-weighed food was provided in the food hopper of their home cage at 9:30 am (unless a fasting day for the IF groups), and leftover food was weighed 24 h later.
Sciatic nerve crush (SNC) surgery
Mice were anesthetized with isoflurane (5% induction, 2% maintenance), shaved on the hind limbs and lower back, sterilised with iodine, and an ophthalmic solution was applied to the eyes to prevent drying. An incision was made on the skin and the biceps femoris and the gluteus superficialis were opened by blunt dissection and the sciatic nerve was exposed using a surgical hook. The sciatic nerve crush was performed orthogonally for 20 seconds (45 seconds for reinnervation experiment) using a 5 mm surgery forceps (91150-20 Inox-Electronic). The crush was performed at approximately 20 mm distally from the sciatic DRG.
Faecal transplantation
Fresh faeces were collected every day from the cage of 5 healthy C57BL/6 mice, approximately 200 mg/ mouse. A fresh cage was used each day.. 1g of faeces was then homogenized in 10 ml of PBS for 30 seconds at 3000 rpm at 4°C. Thereafter, the supernatant was transferred to a new tube and centrifuged for 5 min at 12000 rpm at 4°C. The pellet was resuspended in 2.5 ml PBS and 500 µl gavaged to each mouse.
Antibiotic administration
Vancomycin (Sigma, V2002) was administered through the drinking water (50mg/kg/day; 214.27 mg/l), which was replenished every second day.
Indole-3-propionate treatment
Indole-3-propionic acid (Sigma, 57400) was diluted at 0.5 mg/200 µl in sterile PBS. Mice were treated with 20 mg/kg/day via gavage or IP injection.
Clostridium sporogenes recolonization
WT and fldC (mutant for (R)-phenyllactyl-CoA dehydratase beta subunit of the phenyllactate dehydratase complex [fldC]) Clostridium sporogenes were cultured overnight in trypticase yeast extract medium anaerobically. Bacterial cultures were mixed with glycerol to reach the final concentrations of: WT 6.01E+06 CFU/ml and C.s. fldC mutant 5.46E+06 CFU/ml in hermetically sealed glass vials. Mice were pre-treated with vancomycin in drinking water for 3 days. C.s. WT or C.s. were transplanted to the cecum via oral gavage for 10 consecutive days at 1E+06 CFU/day.
Monoclonal Antibody injection
Preceding and following nerve injury, monoclonal antibodies; mouse αLy6G (Bioxcell, Clone 1A8); mouse αIFNγ (Bioxcell, clone XMG1.1); rat IgG2a isotype control (Bioxcell, 2A3); rat αIgG1 (Bioxcell, clone HRPN); were administered at 200µg/day for 10 consecutive days via i.p. injection. Monoclonal antibodies anti-CXCR2 (R&D Systems, clone # 242216) and anti-IgG2A (R&D Systems, clone # 54447) were administered for 3 consecutive days post nerve injury at 20 µg/day via i.p. injection.
Serum and cecum preparation
Blood was extracted via heart punctuation and collected into a covered test tube. The blood was allowed to clot for a minimum of 30 min at room temperature and centrifuged at 2000 g for 10 min at 4 °C. The supernatant was then collected into a fresh tube and stored at -20 °C until use.
Cecum content was removed from the cecum using sterile forceps into a tube and snap-frozen in liquid nitrogen immediately.
Gas chromatography – Mass Spectrometry (GC-MS) untargeted metabolomics
Serum samples (100 μl) were prepared as follows: I) samples were spiked with 10 μL internal standard solution (myristic acid-d27, 750mg/ml), II) 850 μL of ice cold methanol were added, followed by centrifugation for 20 min (4o C, 16000 g), III) 750 μL of supernatants were transferred to silanized dark 2mL autosampler vials and were evaporated to dryness in a rotational vacuum concentrator (45o C, 20 mbar, 2 h), IV) 50 μL of methoxyamine solution (2% in pyridine) were added and the samples were incubated overnight at room temperature and finally, V) 100 μL of N-methyl-trimethylsilyl-trifluoroacetamide (MSTFA) +1% trimethylchlorosilane (TMCS) solution were added, the samples were incubated at 60 ºC for 1 h and were transferred to dark autosampler vials with 250 μL silanized inserts. The samples were analysed in an Agilent 7890B-5977B Inert Plus GC-MS system. 2 μL of each sample were injected in a split inlet (1:10 ratio). The chromatographic column was an Agilent ZORBAX DB5- MS (30 m X 250 µm X 0.25 µm + 10m Duraguard). The temperature gradient was 37.5 min long and the mass analyser was operated in full scan mode between 50-600 m/z. The detailed instrumental conditions are described elsewhere (Agilent G1676AA Fiehn GC/MS Metabolomics RTL Library, User Guide, Agilent Technologies, https://www.agilent.com/cs/library/usermanuals/Public/G1676-90001_Fiehn.pdf). Quality Control (QC) samples were created by pooling equal amounts of every serum sample of the study and were analysed interspaced in the analytical run. Study samples were randomized before sample preparation. Peak deconvolution, alignment and annotation and were performed with the use of the Fiehn library via the software packages AMDIS (NIST), Mass Profiler Pro and Unknowns (Agilent technologies) in the pooled QC samples. Peak picking was performed with the GAVIN package (A software complement to AMDIS for processing GC-MS metabolomics data. doi: 10.1016/j.ab.2011.04.009)48. Non-reproducible (CV>30% in QC samples) and contaminated (blank > 20% of the mean QC levels) metabolic features were removed from the dataset.
This data set was then used to build orthogonal partial least-squares-discriminant analysis (OPLS-DA) (IF vs AL) or partial least-squares-discriminant analysis (PLS-DA) (IF, IF-V, AL, AL-V), focusing on the differences among the experimental groups. The OPLS algorithm derives from the partial least-squares (PLS) regression method49. The method explains the maximum separation between class samples Y (n dummy variables for n classes) by using the GC-MS data X. Here the ropls R package (http://bioconductor.org/packages/release/bioc/html/ropls.html) was used, which implements the PCA, PLS-DA and OPLS-DA approaches50. It includes R2 and Q2 goodness-of-fit and goodness-of-prediction statistics, permutation tests, as well as scores, loadings, predictions, diagnostics, outliers graphics50. R2X describes the percentage of predictive and orthogonal variation in X that is explained by the full model. R2Y describes the total sum of variation in Y explained by the model. Q2 describes the predictive performance of the model calculated by full cross validation.
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)
Materials
Indole-3-propionic acid (Aldrich 1 g) as standard and indole-3-propionic-2,2-d2 acid (0.05 g, C.D.N. Isotopes) as internal standard (IS) were used. Acetonitrile, methanol and formic acid were of ULC-MS grade was supplied from Bio-Lab. Water with resistivity 18.2 MΩ was obtained using Direct 3-Q UV system (Millipore). Standard curve was built using concentration range of 3-indole propionic acid 0.01-12 µg/mL, with final concentration of IS 100 ng/mL.
Extract preparation
Plasma (30 µL) and IS (10uL, 1 ug/mL) was incubated 10 min, then 500 µL of methanol was added. The mixture was shaken at 10°C for 30 min (ThermoMixer C, Eppendorf), and centrifuged at 21,000 g for 10 min. Collected supernatant was evaporated in speedvac and then in lyophilizer. Before LC–MC analysis, the obtained residue was re-suspended in 100 µL of 20%-aq methanol, centrifuged twice at 21,000 g for 5 min to remove insoluble material. The soluble part was placed to insert of LC-MS vial. Some samples were diluted 1/20 with 20%-aq methanol.
LC-MS analysis
The LC–MS/MS instrument consisted of an Acquity I-class UPLC system (Waters) and Xevo TQ-S triple quadrupole mass spectrometer (Waters) equipped with an electrospray ion source and operated in positive ion mode. MassLynx and TargetLynx software (version 4.1, Waters) were applied for the acquisition and analysis of data. Chromatographic separation was done on a 100 mm × 2.1 mm internal diameter, 1.7-μm UPLC BEH C18 column equipped with 50 mm × 2.1 mm internal diameter, 1.7-μm UPLC BEH C18 pre-column (both Waters Acquity) with mobile phases A (0.01% formic acid) and B (0.01% formic acid in acetonitrile) at a flow rate of 0.6 ml min−1 and column temperature 60°C. A gradient was used as follows: 0–5 min a linear increase from 20 to 25 % B, then increase to 95 % B during 0.1 min, 5.1–7 min held at 95 % B, then back to 20 % B in 0.1 min, and equilibration at 20 % B for 1.9 min, providing total run time of 9 min. Samples kept at 8 °C were automatically injected in a volume of 1 µl. MS parameters: capillary voltage - 1kV, cone 0 V, Source temperature -150°C, Desolvation temperature - 550°C, Desolvation gas - 110 L/Hr, Cone gas - 200 L/Hr, Nebulizer - 7.0 Bar, Collision gas 0.10 mL/min. MRM transitions (collision energy, eV): 190>55.0 (17), 190>130.1 (12), 190>172.1 (9) for 3-indole propionic acid, and 192.1>56.0 (12), 192.1>130.1 (14), 190>174.1 (14) for IS.
16S rDNA amplicon sequencing
Cecum content was collected from mice and immediately frozen in liquid nitrogen. Samples were stored at -80 °C. Faecal DNA was extracted using Fast DNA spin kit (MP Biomedicals). Libraries were prepared using the 16S Metagenomic Sequencing Library Preparation protocol for the Illumina MiSeq System. In brief, PCR primers (16S Amplicon PCR Forward Primer = 5' TCGTCGGCAGCGTCAGATGTGTATAAGAGACAGCCTACGGGNGGCWGCAG; 16S Amplicon PCR Reverse Primer = 5' GTCTCGTGGGCTCGGAGATGTGTATAAGAGACAGGACTACHVGGGTATCTAATCC) directed at the V3/V4 region of bacterial rRNA genes were used to generate libraries and libraries were validated by the Agilent 2100 Bioanalyzer. Sequencing was performed with 250 bp paired-end reads on Illumina Miseq2500 (Imperial BRC Genomics Facility).
16S rRNA amplicon sequencing data analysis
Only single-end R1 sequences were analysed. Data processing was carried out in R statistical environment version 3.5.2 according to the DADA2 pipeline51 as follows; primers were trimmed from de-multiplexed sequences, amplicon sequence variants (ASVs) were generated using DADA2 package version 1.10.1, and taxonomic assignment was performed using the SILVA rRNA database version 132 [https://www.arb-silva.de/]. ASVs with no phylum assigned were excluded from analyses to yield a final number of n = 906 annotated ASVs. Alpha and beta diversity analyses were carried out using phyloseq package version 1.26.152. Alpha diversity (Shannon Index) was calculated using raw counts53. Groups were compared using Mann Whitney U statistical tests. For beta diversity analyses, data was normalised by log-transformation with a pseudo count of +1. Differential abundance of groups were compared in the DESeq2 package version 1.22.254 using the Wald test with Benjamini and Hochberg adjustments for multiple testing (p value set as 0.05). Alpha diversity is a measure of diversity or “microbial variation” within a (single) sample while beta diversity is a measure of diversity across (several) samples.
RNA sequencing
DRG were collected from animals that had undergone 10 days of IPA or PBS treatment and/or followed by sciatic nerve crush. Sciatic DRG were extracted 72h after sciatic nerve crush or from naïve animals (surgeries were performed as described above) and collected into RNAlater. DRG were crushed with RNase free micro pestle and RNA was then immediately extracted using RNAeasy kit (Qiagen), according to manufacturer’s guidelines. Residual DNA contamination was removed by treating the spin column with 40 units of RNase-free DNase I (Qiagen) for 15 min at 23 °C prior to RNA elution. RNA concentrations and purity were verified for each sample following elution with the Agilent 2100 Bioanalyzer (Agilent). RNA with RIN factors above 8.5 were used for library preparation. cDNA libraries for each sample were generated by the Imperial BRC Genomics Facility using the TruSeq Sample Preparation Kit A (Illumina, San Diego CA) and sequenced using Illumina HiSeq 4000 (PE 2x75 bp) sequencing. Gene ontology (GO) was performed on differentially expressed genes with DAVID 6.8 (Database for Annotation, Visualization, and Integrated Discovery (http://david.abcc.ncifcrf.gov/)). Differentially expressed genes were selected using a threshold of P<0.05 and |1.5|<FC (fold change) or no FC cut-off. To identify IPA-dependent specific differentially expressed genes following SNC, we compared IPA-SNCvsPBS and PBS-SNCvsPBS groups and selected the uniquely up- or downregulated genes in the IPA-SNCvsPBS group.
Sciatic nerve regeneration
24 or 72 hours following the surgery, sciatic nerves were dissected and post-fixed in 4% PFA, incubated at 4°C for 1h and transferred into 30 % sucrose for at least 3 days. Subsequently, the tissue was embedded and frozen in Tissue-Tek OCT and maintained at -80°C until cut into 11 µm sagittal sections. Tissue sections were immunostained for SCG10 (1:1000, rabbit, Novus) a marker for regenerating axons. The crush site was identified by deformation of the nerve and disruption of axons coinciding with highest SCG-10 intensity. The SCG-10 intensity was measured in 500 µm intervals along the length of the nerve distal to sciatic nerve crush site. The intensity was normalised to the SCG-10 intensity before the crush site and plotted as fold-change. 4-6 sections per animal were analysed and imaged with a HWF1 - Zeiss Axio Observer with a Hamamatsu Flash 4.0 fast camera using 10x magnification.
Dorsal Root Ganglia (DRG) cell culture.
Glass coverslips were coated with 0.1 mg/ml PDL, washed and coated with mouse Laminin 2ug/ml (Millipore) for 1-2 hours each previous to the start of the experiment. Sciatic DRG from adult animals were dissected and collected in Hanks balanced salt solution (HBSS) on ice. The DRG were transferred into a digest solution (5mg/ml Dispase II (Sigma), 2.5 mg/ml Collagenase Type II (Worthington) and incubated in a 37 °C water bath for 45 min, with occasional shaking for 30 seconds . Thereafter, the DRG were washed and manually dissociated with a 1ml pipette in media containing 10 % heat inactivated FBS (Invitrogen) and 1x B27 (Invitrogen) in F12:DMEM (Invitrogen). Pipetting was continued until DRG were fully dissociated and no clumps could be observed. Next, the cell suspension was spun down at 1000 rpm for 4 min and resuspended in culture media containing 1x B27 and Penicillin/Streptomycin in F12 : DMEM. 3500 cells were plated on each coverslip (laminin and PDL coated) and maintained in a humidified culture chamber with 5% CO2 at 37 °C, for 12 hours before fixed with 4% PFA and immunostained.
Following cell plating, DRG were treated with indole-3-propionic acid (1 mM, 10 mM or 100 mM, Sigma 57400) or IFNg (5 ng/ml, 100 ng/ml, Peprotech 250530). For IFNγ-dependent ex-vivo DRG outgrowth, animals were injected intraperitoneally with 10 µg IFNγ/mouse 48 hours before DRG cell culture.
Immunohistochemistry
Immunohistochemistry on tissue sections was performed according to standard procedures. Tissue sections were rehydrated with PBS and blocked and permeabilized for 1 hour with either 8% bovine serum albumin (Sigma Aldrich) or 10% normal goat serum (Abcam) containing 0.3% PBS-TritonX-100. Secondly, the sections were incubated with anti-SCG10 (1:1000, Novus), anit-SOX10 (1:1000, Abcam), anti-CD68 (1:1000, Abcam), anti-Ly6G (1:500, BioxCell, clone 1A8), anti-CD8 (1:100, ThermoFisher, 14-0081-82), anti-Tuj1 (1:500, Novus, NB100-1612) anti-CD68 (1:200, Abcam, ab125212), anti-B220 (1:100, Biolegend, 103228), anti-PGP9.5 (1:200, Proteintech, 14730-1-AP), CD4 (1:100, BD Pharmingen, Clone RM4-5), NK1.1 (1:2000, Bioxcell, BE0038), Ly6G-FITC (1:100, Biolegend), CXCR2 (1:50, R&D Systems) at room temperature over-night. The sections were washed three times with PBS, followed by incubation with Alexa Fluor conjugated goat secondary antibodies for 1 hour. All tissue sections were counterstained with DAPI (Molecular Probes) and cover slipped with moviol.
Immunocytochemistry (ICC)
Plated cells were fixed by incubation with cold 4 % PFA for 15 min. Thereafter, they were blocked and permeabilized for 1 hour with 0.3 % TX100 in PBS containing 2 % BSA. The primary antibody staining was performed using anti-βIII Tubulin (1:1000, mouse, Promega) in 0.1 % TX100 in PBS containing 2 % BSA, which O/N incubation at RT. The goat secondary antibody (Alexa) was diluted in 0.1 % TX100 in PBS containing 2 % BSA and cells were incubated for 1 hour. All cells were counterstained with DAPI.
FACS
Cells were isolated from spleens or lymph nodes by mashing using a 70 µm cell strainer and a pestle and transferred into Media (RPMI media 1640 [Thermo Fisher scientific] + 2% Foetal calf serum + 1x Penicillin/Streptomycin + 1x HEPES [Thermo Fisher scientific]). Spleen cells were centrifuged at 1200 rpm for 6 min and resuspended in 1ml of Red blood cell lysis buffer Hybrid Max (Sigma) for 5 min. Next, 9 ml of media was added and centrifuged at 1200 rpm for 6 min. Spleen cells or lymph node (LN) cells were resuspended in Media and plated at 1x106-2x106 cells per well in a 96-well plate. The plate was centrifuged at 2000 rpm for 1 min and 50 µl of blocking stain (1:50 dilution Rat Serum [Sigma], 1:50 dilution TruStain FcX™ [anti-mouse CD16/32] [Biolegend] in FACS buffer [PBS supplemented with with 5% FCS, 2mM EDTA and 0.09% NaAzide]) was added. Cells were blocked for 30 min at 4 °C and subsequently washed three times with 140 µl of FACS buffer. Antibodies were added and cells were incubated for 30 mins in the dark. Next, cells were washed another 3 times and analysed on a BD LSR Fortessa X-20 (BD Biosciences, 4 laser).
Marker (clone)
|
Colour
|
Dilution
|
Company
|
CD45 (30-F11)
|
V450
|
1:100
|
BD Biosciences
|
CD11b (M 1/70)
|
BV421
|
1:200
|
Biolegend
|
CD11c (3.9)
|
PECy7
|
1:200
|
Biolegend
|
Ly6G (1A8)
|
BV711
|
1:200
|
Biolegend
|
Ly6C (HK1.4
|
FITC
|
1:200
|
Biolegend
|
LIVE/DEAD™
|
Aqua (405 nm)
|
1:100
|
Thermo Fisher Scientific
|
CD115
|
PE
|
1:50
|
BD Biosciences
|
Microscopy
Photomicrographs were taken with a Nikon Eclipse TE2000 microscope with an optiMOS scMOS camera using 10x or 20x resolution Zeiss Axio Observer with a Hamamatsu Flash 4.0 fast camera using 10x or 20x magnification. Confocal images were taken with a Leica TCS SP8 II confocal microscope at 40X or 60X magnification and processed with the LAS-AM Leica software (Leica).
Image Analysis for IHC and ICC
Image analysis was conducted using ImageJ (Fiji) software. All analysis was performed by the same experimenter who was blinded to the experimental groups. DRG images were taken using a Nikon Eclipse TE2000 microscope with an optiMOS scMOS camera at either 10x or 20x magnification. Images were analysed by counting the number of cells per area, measuring the intensity against the background or calculating percentage of cells with positive staining.
For neurite length analysis between 15 and 20 images were taken per coverslip and analysed using NeuronJ plugin for Image J software (Image J). All analyses were performed in blind. Approximately 45-60 cells were analysed per animal and condition.
Statistical analysis
Results are graphed as mean ± SEM. Statistical analysis was carried out using GraphPad Prism 7. Normally distributed data was evaluated using a two-tailed unpaired Student’s t-test or a one-way ANOVA when experiments contained more than two groups. Dunnett multiple comparisons test or multiple comparison testing corrected by FDR with Benjamini and Hochberg were applied when appropriate. The two-way ANOVA, Tukey’s or Sidak’s test, was applied when two independent variables on one dependent variable were assessed. A threshold level of significance was set at P<0.05. Significance levels were defined as follows: * P<0.05; ** P<0.01; *** P<0.001; **** p<0.0001. All data analysis was performed blind to the experimental group.