Study design
This cross-sectional study consisted of two steps: the first samples from human volunteers with or without COVID-19 were analyzed, and the second human feces from these volunteers were transplanted into germ-free mice (Fig.1). Initially, post-COVID and health volunteers (controls) were recruited, followed by the collection of feces and blood samples. Collections were performed from 1 to 4 months after infection (on average 2 months). The volunteers answered a survey consisting of clinical symptoms, medications used, and lifestyle questions. Samples were collected between October 2020 - April 2021 in Belo Horizonte metropolitan area in the state of Minas Gerais, Brazil. Fresh feces samples from the subjects were immediately used for Fecal Microbiota Transplantation (FMT) into Germ-Free mice (6-8 weeks old).
To investigate the effects of feces from post-COVID subjects on the susceptibility to pulmonary infection caused by a antimicrobial resistant bacteria, germ-free mice were infected with the bacteria Klebsiella pneumoniae afterfeces transfers from post-COVID and controls subjects. Animals wererandomly divided ten days after FMT into four groups: (1) Controls + Vehicle, (2) Controls + K. pneumoniae B31, (3) post-COVID + Vehicle and (4) post-COVID + K. pneumoniae B31. The infection with K. pneumoniae B31 was performed intratracheally. Subsequent analyses were performed 48 hours after infection. To evaluate the therapeutic effects of a probiotic bacterium, Bifidobacterium longum 51A, on post-COVID microbiota, mice were randomly divided into four groups: (1) Controls + Vehicle, (2) Controls + B longum 51A, (3) post-COVID + Vehicle, and (4) post-COVID + B. longum 51A. The B. longum 51A was administered through oral gavage every 48 hours for 12 days, until the end of the experiment and subsequent analysis.
Study subjects and sample collection
A total of 72 post-COVID subjects (1-4 months after SARS-CoV-2 infection) and 59 controls volunteers (SARS-CoV-2 negative test) were included in this study see the sample workflow in Additional File 1: FigS1.
Post-COVID subjects underwent SARS-CoV-2 testing (i.e., a nucleic acid amplification test [NAAT] or an antigen test) to confirm the infection. Those who tested positive for SARS-CoV-2 and did not show any symptoms related to COVID-19 were classified as asymptomatic. Clinical spectrum of disease severity was classified according to the NHI COVID-19 Treatment Guidelines [34]. Control volunteers were included in the study if they never had symptoms related to COVID-19 and not tested positive for SARS-CoV-2. To ensure, serological tests were performed in all the volunteer serum samples to verify the presence or absence of SARS-CoV-2 antigens. The volunteers were instructed to self-collect the fecal sample in a sterile container and immediately store it in a home refrigerator (4oC) within a maximum of 10 hours. The volunteers' sociodemographic characteristics, lifestyle, and health statuses are shown in Additional File 2: Table S1. All volunteers were aged between 15-60 years old. A survey was applied to investigate factors that influence the health and intestinal microbiota status of the subjects, such as comorbidities, eating habits, and antibiotics use at least 4 months before the application of the survey and sample collection. The parameters used to verify the eating habits of the volunteers are available in Additional File 3: Table S2. The exclusion criteria established in the present study were volunteers not having been vaccinated for SARS-CoV-2 and if they tested positive in the serological test performed on the day of sample collection.
Laboratory animals
Male and female Germ-Free Swiss/NIH mice derived from a GF nucleus (Taconic Farms, Germantown, USA), with ~8-weeks-old were used. They were maintained in flexible plastic isolators (Standard Safety Equipment Co., Pallatine, USA) using classical gnotobiology techniques [35] at the Gnotobiology Laboratory of the Federal University of Minas Gerais (UFMG), Minas Gerais, Brazil. Also, male and female C57BL/6J mice, aging ~8 weeks old, obtained from the UFMG animal facility, were kept in plastic cages in a room with controlled conditions (26°C, 12h light/dark cycle) with steam sterilized food (NuvilabⓇ, Nuvital, Brazil) and water ad libitum. All mouse procedures were performed in accordance with guidelines from the Guide for the Care and Use of Laboratory Animals of the Brazilian National Council of Animal Experimentation (http://www.cobea.org.br/) and Brazilian Federal Law 11.794 (October 8, 2008). The animal study was reviewed and approved by The Institutional Committee for Animal Ethics of the Federal University of Minas Gerais (protocol nº CEUA/UFMG 281/2020 and 55/2021).
Human Fecal Microbiota Transplantation to GF mice
Fresh fecal samples were self-collected by volunteers, were weighed and resuspended in 0.9% sterile saline (NaCl) solution (100mg for each 1 mL of solution), homogenized, and kept for 10 minutes at 4 °C to big particles precipitate. For FMT, a single 100 μL aliquot of the supernatant was used for oral gavage of the GF mice. To assure stable human microbiota into GF mice after FMT 10 days have waited until the beginning of the experiments [36].
Klebsiella pneumoniae intratracheal infection
The bacterium used in the intratracheal pulmonary infection experiments was K. pneumoniae B31, a clinical isolate with a multidrug-resistance profile [37], gently provided by Prof. Vasco Ariston from the Laboratory of Cellular and Molecular Genetics at ICB/UFMG. For the procedure, animals were anesthetized intraperitoneally (i.p.) with 80 mg/kg ketamine and 10 mg/kg xylazine i.p., the trachea was exposed through a skin incision, and 25μL of the suspension containing 1×106 CFU/mL of K. pneumoniae B31, or sterile saline for mock controls, was injected with a 26-gauge needle [38]. After infection, the skin was sutured, and the mice were monitored for 2 days and then euthanized as described in the experimental design.
Mouse Hepatitis Virus-3 intranasal infection and virus quantification
The β-coronavirus mouse hepatitis virus MHV-3 was provided by Prof. Vivian Vasconcelos Costa and Prof. Mauro Martins Teixeira from the Center for Research and Development of Drugs at the Department of Morphology, UFMG. For intranasal infection, C57BL/6J mice were lightly anesthetized with 80 mg/kg ketamine and 10 mg/kg xylazine i.p., and received an intranasal inoculation of 30μL of a suspension containing 3×101 PFU of MHV-3 [39], or sterile saline for mock controls. Then, mice were monitored and euthanized 10 days after infection. MHV-3 was propagated in L929 cells, and viral titration was performed, as described previously [39]. RNA was extracted with kit QIAamp® Viral RNA for viral brain quantification, and the tissues were macerated in the first step with the lysis buffer. cDNA was synthesized using the kit iScript™ gDNA Clear cDNA Synthesis Kit (BIO-RAD), and qPCR assay was performed using Fast SYBR™ Green Master Mix (Applied Biosystems™). Primer sequences are described in Supplementary Information (Additional File 4: Table S3). The standard was obtained by extracting RNA from a known amount of PFU from MHV-3. Results of viral quantification were expressed in Arbitrary Units.
Treatment with Bifidobacterium longum 51A probiotic
The probiotic bacterium B. longum 51A, a strain isolated from the feces samples of healthy children [40] was provided by Prof. Flaviano S. Martins from the Laboratory of Biotherapeutic Agents at ICB/UFMG. B. longum 51A was cultured in De Man, Rogosa, and Sharpe (MRS) broth (Acumedia, USA) supplemented with 0.5% L-cysteine (Synth, Brazil) under an atmosphere containing 85% N2, 10% H2 and 5% CO2, during 48h at 37°C in an anaerobic chamber (Forma Scientific, Marietta, GA, USA). Mice received by oral gavage, a single 100μL dose of suspension containing 1.0 x 109 CFU/mL, or sterile saline [38]. HM mice and C57BL/6 were treated every 48h, during 12 and 10 days, respectively.
Mouse behavioral tests
Novel object recognition and novel object location tests
Tests were performed in a 30 (w) x 30 (d) x 45 (h) cm arena. Before training, each animal was submitted to a 5 min habituation session the previous day, in which they were allowed to freely explore the empty arena. Training consisted of a 5 min session during which animals were placed at the center of the arena in the presence of two identical objects and the time spent exploring each object was recorded. Thirty minutes after training, animals were again placed in the arena for the test session, when one of the two objects used in the training session was replaced by a new one in the object recognition paradigm or moved to a new location in the new object location paradigm, and the time spent exploring familiar and novel objects (or object at the novel location) was measured [41]. The arena and objects were cleaned thoroughly between trials with 70% ethanol to eliminate olfactory cues. Test objects were made of plastic [approximately 3 cm (w) x 3 cm (d) x 4cm (h)] and, during behavioral sessions were fixed to the arena floor using tape to prevent displacement caused by exploratory activity of the animals. Preliminary tests showed that none of the objects used in our experiments evoked innate preference by animals. Results were expressed as a percentage of time exploring each object or location (old or new) in relation to the total exploration time during the test session.
Bronchoalveolar lavage cell counts
After mice euthanasia, bronchoalveolar lavage (BAL) was collected by inserting 1mL of phosphate-buffered saline using 20-gauge catheter connected to a 1mL syringe into the lungs. The 1mL aliquot was collected, centrifuged, and resuspended in 100µL of sterile saline, and the total leukocytes were quantified by Neubauer chamber counting. In addition, differential counts were obtained from cytospin preparations [38].
Measuring SARS-CoV-2 load in the human feces
RNA extraction from feces samples was adapted from previously published protocols [42]. Briefly, fecal samples were diluted 1:5 (w:v) in guanidine, homogenized, and clarified by centrifugation at 4,000g for 20 min at 4°C. Viral RNA was purified using the QIAamp Viral RNA Mini Kit following the manufacturer's instructions, with a final elution volume of 50 µL.
Real-Time quantitative PCR (RT-qPCR) for SARS-CoV-2 test in feces samples and viral load quantification was performed on reactions with a final volume of 20 µL, using the one-step RT-qPCR Master Mix according to the CDC USA protocol [43]. The primers used for qPCR in the N1 and N2 regions of the nucleocapsid gene were sourced from Integrated DNA Technologies (cat. no. 10006770) that were manufactured using the U.S. CDC sequences and QC qualified under a U.S. CDC Emergency Use Authorization. For analysis, we adopted the amplification values of viral targets N1 or N2 with a threshold cycle (Ct) below 40.0 was considered positive for SARS-CoV-2, and above 40 as indeterminate or undetectable. This protocol was adapted from CDC 2019-Novel Coronavirus (2019-nCoV) Real-Time RT-PCR Diagnostic Panel Instructions. We used feces samples spiked with inactivated SARS-CoV-2 (stock titer 6.7 x 106 PFU/mL) as positive controls at different dilutions.
Inflammatory mediators’ analysis by RT-qPCR in lung, intestine, and brain tissue from mice
Total RNA of the lung, intestine, and hippocampus from controls and post-COVID HM GF mice was extracted using TRIzol™ reagent (Thermo Scientific). cDNAs were prepared in a 20-µL final reverse transcription reaction and subjected to qPCR using the kit Power SYBR Green Master Mix (Applied Biosystems), following the manufacturer’s instructions, in the QuantStudio™ 7 Flex real-time PCR system platform (Applied Biosystems). The primer sequences used are described in Supplementary Information (Additional file 4: Table S3). All RT-qPCRs showed good amplification quality, and gene expression changes were determined with the 2−ΔCt method using Ribosomal protein L32 or normalization.
ELISA for measurements of Intestinal fatty acid-binding protein (I‑FABP) and cytokines
ELISA techniques were used to quantify the concentrations of I-FABP as described before [44]. IL-10, TNF-a and IL-1b concentrations were determined using IL-10 ELISA kits (R&D Systems, USA) according to the manufacturer’s instructions. The analyses were performed in duplicate.
Short-chain fatty acids (SCFAs) measurement
The SCFAs measurements were performed as previously described, with adaptations [45]. Feces samples were suspended in 1% phosphoric acid (1:6 – W: V) (Merck, USA), vortexed, and centrifuged at 20,000g for 30 min at 4°C. The supernatant was filtered through a cellulose acetate membrane (0.22 µm), and 10 µL were injected directly into HPLC, using an ionic exchange resin column 300×7.8 mm (Sigma, Germany) with a Micro-Guard cation H+ cartridge (Sigma, Germany) and detector set at 210 nm. The flow rate was 0.5 mL/min until 35 minutes of chromatographic run, being changed to 0.7 mL/min until the end of the run. The column temperature was 30ºC. For serum samples, the SCFAs measurements were performed as previously described, with adaptations [46]. Serum samples were suspended in formic acid 1 mol.L-1, and solution of the internal standard, 2-ethyl-butyric acid 1 mol.L-1 (Sigma, Germany) was added, in proportions 5:5:1 respectively. Then, the suspension was vortexed and centrifuged at 12,000 g for 30 min at 4°C. After, 2 µL of the supernatant were injected into Gas Chromatograph–FID (Agilent, USA), using HP-FFAP column 19091F-105 (Agilent, USA), 50m×0.20mm×0.33 µm, and the detector was set in 240°C. The chromatographic conditions were 60°C for 0.5 minutes, heating at 8°C.min-1 to 180°C for 1 minute. New heating rate from 20 °C.min-1 to 240 °C for 7 minutes. The total run time was 26.5 minutes. Seven-point external calibration curves were adopted to quantify fecal and serum samples, using analytical grade SCFA (Sigma, Germany) as standards.
Histopathology and immunohistochemistry analysis
Intestine and lung tissue from HM GF mice were collected and processed. The inflammatory score was performed with hematoxylin and eosin (H&E) stained slides and evaluated airway, vascular and parenchymal inflammation [47,48]. For immunohistochemistry analysis, lung tissue slides from controls, and post-COVID HM GF mice were immune-stained as described before [49]. Briefly, the slides were incubated with primary anti-α-actin antibodies (human, 1:500) (DAKO, USA) overnight at 4ºC. Then, the primary antibodies were detected using an anti-mouse/anti-rabbit detection system (Novolink Polymer Detection System; Leica Biosystems, Newcastle Upon Tyne, UK) according to the manufacturer’s instructions. The sections were counterstained in diluted Harris Hematoxylin solution and permanently mounted with Entellan (Merck, USA). For the morphometric analysis, images (20X objective) were acquired from α-actin immunolabeled to quantify the muscle layer of the lung section using the Image J1.52 program (NIH, USA). All the analyzes were examined under a light microscope by a pathologist who was blinded to the experiment.
Cultivable fecal microbiota quantification
Fresh fecal samples were weighed, homogenized in sterile 0.9% saline (100mg for each 1 mL of saline), and serially diluted (1:10). Subsequently, different dilutions were plated on different selective media (Merck, USA) for quantification of total aerobic (blood agar in aerobiosis) and anaerobic bacteria (blood agar supplemented with hemin (5 µg/mL) and menadione (1 µg/mL) in anaerobiosis), Enterobacteriaceae (MacConkey agar in aerobiosis), Staphylococcus (Salt Mannitol agar in aerobiosis), Bacteroides (Bacteroides Bile Esculin agar in anaerobiosis), Acid Lactic Bacteria (De Man, Rogosa and Sharpe in aerobiosis) and Bifidobacterium and anaerobic Lactic Acid Bacteria(De Man, Rogosa and Sharpe in anaerobiosis) populations. Aerobic plates were incubated for 24h at 37°C under standard conditions. Anaerobic plates were incubated in an anaerobic chamber (Forma Scientific, Marietta, GA, USA) under an atmosphere containing 85% N2, 10% H2, and 5% CO2 at 37°C for up to 72h. After incubation, the colonies were counted, and data were expressed as the log10 of colony-forming units (CFU) per gram of feces.
Quantification of Enterobacteriaceae on bronchoalveolar lavage
The BAL samples were directly plated on MacConkey agar, incubated in aerobic conditions, and kept under the same culture conditions mentioned above. After incubation, the colonies were counted, and data were expressed as the log10 of CFU per mL of BAL.
Enterobacteriaceae antimicrobial resistance test and identification
After fecal microbiota cultivation, individual Enterobacteriaceae colonies with different morphologies were isolated from MacConkey medium (Sigma, Germany) plates. Pure colonies were suspended in sterile 0.9% saline at a 1.5x108 CFU/mL concentration according to the 0.5 McFarland standard. Then, a sterile swab was soaked in the bacterium solution and inoculated by spreading on Mueller Hinton Agar plates (140x15mm) (Merck, USA). After 15 minutes, a dispenser (Thermo Scientific™ Remel™) with 12 discs (Thermo Scientific™ Oxoid™) referring to β-lactam (amoxicillin-clavulanic acid, cephalosporin, ertapenem, meropenem, imipenem), aminoglycosides (amikacin, streptomycin, gentamicin), quinolones (ciprofloxacin, levofloxacin, norfloxacin), sulfonamide and folate inhibitors (sulfamethoxazole-trimethoprim) and macrolides (azithromycin) antibiotics were added to the inoculated plates. The plates were incubated at 37°C for 24h. The presence or absence of bacterial growth inhibition zones was observed and measured to determine the resistance profile in: sensitive, intermediate, or resistant, according to the CLSI M100Ed31 guidelines. The resistance phenotype was determined according to the number of antimicrobials classes in which each strain presented resistance, being resistant (1-2 antimicrobials) or multidrug-resistant (≥3 antimicrobials). Identification of Enterobacteriaceae strains was performed by Matrix Associated Laser Desorption-Ionization - Time of Flight (MALDI-TOF), using the FlexControl MicroFlex LT mass spectrometer (Brunker Daltonics) as described before [50]. Before identification, calibration was done with the bacteria Escherichia coli DH5α test standard (Brunker Daltonics). For the construction of the network, data from bacteria that showed resistance to ≥2 antimicrobials classes were used. Nodes were classified as the isolated bacteria and edges as shared AMR phenotype. The network was non-directional, and the nodes color was determined according to the resistance phenotypeof the antimicrobial classes. Node size was established according to the number of antimicrobial classes to which each bacterium was resistant. Edge weight was defined according to the number of resistance classesthat the bacteria shared with each other. Network co-occurrence analysis, visualization, and property measurements were performed using the platform Gephi [51].
LC-MS/MS Analysis
Hippocampus samples were mechanically lysed with an ultrasonication probe in protein extraction buffer (100 mM Tris-HCl, 1 mM EDTA, 150 mM NaCl, 1% Triton-X) and protease inhibitors cocktail (Protease Inhibitor Cocktail, SIGMA). 20 μg of protein were submitted to the FASP protocol for tryptic digestion [52]. Digested peptides were then resuspended in 0.1% formic acid and fractionated on an ACQUITY MClass System (Waters Corporation). 1 μg of digested samples were individually loaded onto a Symmetry C18 5 μm, 180 μm × 20 mm precolumn (Waters Corp.) and subsequently separated in a 120 min reversed phase gradient at 300 nL/min (linear gradient, 3–55% ACN over 90 min) using an HSS T3 C18 1.8 μm, 75 μm × 150 mm nanoscale and LC column (Waters Corp.) maintained at 30 °C. For the gradient elution, water-formic acid (99.9/0.1, v/v) was used as eluent A and acetonitrile formic acid (99.9/0.1, v/v) as eluent B. Separated peptides were analyzed in a Synapt G2-Si mass spectrometer directly coupled to the UPLC system. Mass spectrometry was conducted using data-independent acquisition (DIA) in expression mode (MSe), switching between low (4 eV) and high (25–60 eV) collision energies on the gas cell, using a scan time of 1.0s per function over 50–2000 m/z. All spectra were acquired in ion mobility mode (wave velocity: 1.000m/s and a transfer wave velocity: 75m/s). A reference lock mass ([Glu1]-Fibrinopeptide B Standard, Waters Corp.) has been employed for online calibration. Experiments were performed in technical triplicates. LC-MS/MS data were processed for qualitative and quantitative analysis using the software Progenesis (Waters Corp.). Protein identification was obtained using Mus musculus database (UniProt KB/Swiss-Prot Protein reviewed). Search parameters were set as: automatic tolerance for precursor ions and product ions, minimum 1 fragment ions matched per peptide, minimum 3 fragment ions matched per protein, minimum 1 unique peptide matched per protein, 2 missed cleavage, cysteine carbamidomethylating as fixed modification, and oxidation of methionine as variable modifications, false discovery rate (FDR) of the identification algorithm < 1%. Label free quantitative analysis was obtained using the relative abundance intensity integrated in Progenesis software, using all peptides identified for normalization. Filtered tables were generated to exclude proteins with no statistical significance according to ANOVA ≥ 0.05. In silico systems biology was performed using DAVID Bioinformatics Resources [53].
Fecal DNA extraction and sequencing
Feces samples self-collected by the volunteers were stored in a freezer at −70 °C. DNA extractions were performed using a QIAamp DNA Stool Mini Kit (Qiagen, USA) according to the manufacturer’s instructions. Then, DNA quality was checked by 1% (w/v) agarose gel electrophoresis and quantified by NanoDrop™ 2000/2000c Spectrophotometer (Thermo Scientific™). DNA was used as a template in PCR amplicon targeting the V3 and V4 hypervariable regions of the bacterial 16S rRNA gene. The Illumina 16S Metagenomic Sequencing Library Preparation protocol was used to prepare the 16S metagenomics library and sequencing (Illumina, USA). PCR amplification protocols, adapter primers, index sequences, and PCR clean-up process were performed as described in the protocol [54].The 16S library was quantified by Qubit dsDNA HS Assay Kit (Invitrogen, USA) and checked with a 2100 Bioanalyzer Instrument (Agilent Technologies, USA). The sample pool (4 nM) library was diluted further final concentration of 8 pM and added to 20% (v/v) of 8 pM PhiX DNA (Illumina, USA), following Illumina guidelines. Sequencing was performed using the Miseq reagent kit v3 (600 cycles) in the Illumina MiSeq platform and 2×300 bp (MSC v2.4) according to the manufacturer’s instructions (Illumina, USA).
Sequence data processing, inferring gut microbiota composition, and statistical analysis
Processing of metagenomic sequencing data was performed using QIIME2 pipeline [55]. First, sequenced reads were denoised with DADA2 and then processed by VSEARCH [56] to filter eventual chimeras and perform de novo clustering of valid sequences into OTUs requiring 97% sequence similarity. Next, MAFFT Fasttree was applied to conduct phylogenetic analysis based on OTUs. α- and β-diversity were analyzed using the core-metrics-phylogenetic method built-in on QIIME2: for the α-diversity, were calculated Shannon’s diversity index and Chao1; for β-diversity, were calculated Bray-Curtis and weighted UniFrac distances. OTUs were taxonomically classified using Naive Bayes classifiers trained with Silva v. 138, setting 97% of sequence similarity for full-length OTUs [57]. Differential abundance was calculated using ANCOM [57]. The metagenomic libraries produced in this work were deposited in the NCBI SRA database under project number PRJNA843134.
Statistical analysis
Statistical analyses and graphs were done using GraphPad Prism7 (GraphPad Software, La Jolla, CA, United States). Data are shown as mean ± standard deviation (SD). Data that presented normal distributions (p > 0.05 by Shapiro–Wilk tests) were evaluated by Student’s t-test, one-way and two-way analysis of variance (ANOVA), depending on the experimental design. Following significant ANOVAs, we performed a post-hoc test according to the coefficient of variation (CV): Tukey (CV ≤ 15%), Student’s Newman-Keuls (CV entre 15 - 30%) e Duncan (CV >30%). Data from object recognition and location tests were expressed as exploration percentage and analyzed by one-sample t-tests to determine whether the percentage of the exploration differed from the chance performance of 50%. The differences were considered significant when p < 0.05. Outliers were only excluded following Grubbs' test and indicated in figure legends.