Ethics Statement
All animal experiments were conducted in an AAALAC International-accredited facility, approved by the Rocky Mountain Laboratories Institutional Care and Use Committee (Protocol number 2021-037-E) and adhered to the guidelines put forth in the Guide for the Care and Use of Laboratory Animals 8th edition, the Animal Welfare Act, United States Department of Agriculture and the United States Public Health Service Policy on the Humane Care and Use of Laboratory Animals. Work with infectious SARS-CoV-2 virus strains under BSL3 conditions was approved by the Institutional Biosafety Committee (IBC) and conducted in BSL-4 facilities. For the removal of specimens from high containment areas virus inactivation of all samples was performed according to IBC-approved standard operating procedures [26].
Viruses
Delta variant (B.1.617.2) hCoV19/USA/MD/HP05647/2021, EPI_ISL_2331496, was kindly provided by Andrew Pekosz (John Hopkins Bloomberg School of Public Health, MD, USA). Delta variant hCoV-19/USA/KY-CDC-2-4242084/2021, EPI_ISL_1823618, was obtained from BEI resources. SARS-CoV-2 WA1 (Lineage A) strain expressing Neon Green (SARS-CoV-2 mNeonGreen) was kindly provided to us through Pei-Yong Shi (Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA) [27].
Virus propagation was performed in Vero E6 cells (ATCC CRL-1586) in DMEM supplemented with 2% fetal bovine serum (FBS), 1 mM L-glutamine, 50 U/mL penicillin, and 50 µg/mL streptomycin (DMEM2). Vero E6 cells were maintained in DMEM supplemented with 10% fetal bovine serum, 1 mM L-glutamine, 50 U/mL penicillin, and 50 µg/mL streptomycin. No mycoplasma and no contaminants were detected. All virus stocks were sequenced; no SNPs compared to the patient sample sequence were detected in the original Delta stocks EPI_ISL_1823618 and EPI_ISL_2331496. Stock EPI_ISL_1823618 was used for the growth curve infection on transfected baby hamster kidney cells and EPI_ISL_2331496 for the in vivo infection to obtain a high enough inoculum titer. A new stock EPI_ISL_2331496 was required for the in vitro infection experiments on primary cell lines: One SNP, nsp14 N410N (wobble), was found compared to the patient sample.
Generation of primary and immortalized cell lines
At necropsy, a sample of uropatagium, lung, and kidney were collected in media (10% FBS DMEM/Ham’s F-12 with primocin, penicillin and streptomycin, amphotericin B, and non-essential amino acids) and placed on ice. Within 4 hours of collection, the lung and kidney were cut into smaller sections and digested in media with Liberase TM (Roche, REF 05401127001) while rotating for 1 hour at room temperature. The digested tissue was centrifuged at 500g for 5 minutes, then transferred to a T25 flask with fresh media. Within 24 hours of collection, a 3 mm section of uropatagium was punch biopsied and washed twice in 1X DPBS. This 3 mm section was cut into smaller sections using a sterile blade and transferred to fresh media with Liberase TM. After incubation at 37oC with 5% CO2 and rotating overnight, the digested uropatagium was centrifuged at 500g for 5 minutes and plated in a T25 flask with 6 mL of fresh media. All cells were confirmed to be mycoplasma negative. Immortalized Jamaican fruit bat kidney cells (AJi) were previously described [28, 29].
Jamaican fruit bat primary cells in vitro infections
Immortalized (AJi) and primary Jamaican fruit bat cells and Vero E6 cells were plated (250,000 cells/mL, 350µL) in 48-well plates and incubated at 37oC with 5% CO2. Sixteen hours after plating, media was removed and cells were inoculated with recombinant SARS-CoV-2 mNeonGreen [27] or SARS-CoV-2 Delta at multiplicity of infection (MOI) 3.0 in the presence or absence of 5.0 µg/mL TPCK trypsin. One hour after inoculation, cells were washed twice with 1X DBPS and given fresh media with or without 5.0 µg/mL TPCK trypsin. The SARS-CoV-2 mNeonGreen infected cells were monitored for fluorescence using the FITC channel of an ECHO Revolve fluorescence microscope. Supernatants and cell lysates were collected in RLT (Qiagen) from SARS-CoV-2 Delta inoculated AjUFi_RML4 cells 48 hours after infection. Supernatants were titrated on Vero-TMPRSII cells. RLT lysates were transferred to ethanol prior to extraction of RNA using the RNEasy extraction kit (Qiagen). Cellular RNA was used to measure intracellular subgenomic (sg)RNA and host genes using qRT-PCR (Table 1). Relative gene expression is presented as ΔΔCT target gene/ΔΔCT HPRT.
Table 1
qRT PCR primers for Jamaican fruit bat immune genes
AJ IFNb F | ACTTCAAGTTTCCCGAGGAGA |
AJ IFNb P | FAM-GCACGGGCTGGAATGAGACCATCATTGA |
AJ IFNb R | GGTCCATCTGCCAACTGAGT |
AJ ISG15 F | CAGAAGGTGGCTGAGCTGAA |
AJ ISG15 P | FAM-TGGCTGAGTTTCCAGGGGAGGCCC |
AJ ISG15 R | CTTGTATTCCTTCAGCTGCGC |
AJ Mx1 F | GTTCTTCATGCTCCGGTCGT |
AJ Mx1 P | FAM-GCCAGAAGCTGAGCAATGCCATGTTGC |
AJ Mx1 R | TTCCTCTTGTCGCTGGTGTC |
AJ OAS1 F | CGTGGTGCAGAGTCACAGAT |
AJ OAS1 P | FAM-CGCGTGATACGGAAACCTCGCTCGC |
AJ OAS1 R | GGGCAGGACATCAAACTCCA |
AJ IL6 F | AACAGCAAGGAGGCACTGAC |
AJ IL6 P | FAM-ACCTGAACCTTCCGAAACTGACAAGAAG |
AJ IL6 R | CAGACCGGTGGTGAGTCTC |
AJ TNFa F | ACTGGCTCAGACCCTTGGAT |
AJ TNFa P | FAM-ACCCCAAGTGACAAGCCTGTTGCCC |
AJ TNFa R | GAGAGCATTGGCCACCTGAT |
AJ HPRT F | AGATGGTGAAGGTCGCAAG |
HPRT P | FAM-ACTTTGTTGGATTTGAAATTCCAGACAAGTTTG |
HPRT R | CCTGAAGTATTCATTATAGTCAAGGG |
Entry assays
The spike encoding sequences for SARS-CoV-2 variants Lineage A, D614G, Alpha, Beta, Delta, and Omicron were truncated by deleting 19 aa at the C-terminus. The S proteins with the 19 aa deletions were previously reported to increase efficiency of incorporation into virions of VSV pseudotypes [30, 31]. These sequences were codon optimized for human cells, then appended with a 5’ Kozak consensus sequence (GCCACC) and 3’ tetra-glycine linker followed by nucleotides encoding a FLAG-tag sequence (DYKDDDDK). The spike sequences were synthesized and cloned into pcDNA3.1+ (GenScript). Pseudotype production was carried out as described previously [28]. Briefly, plates pre-coated with poly-L-lysine (MilliporeSigma) were seeded with 293T cells and transfected the following day with 1,200 ng of empty plasmid and 400 ng of plasmid encoding coronavirus spike or no-spike plasmid control (green fluorescent protein (GFP)). After 24 h, transfected cells were incubated with VSVΔG seed particles pseudotyped with VSV-G as previously described [28, 32]. After one hour of incubation with intermittent shaking at 37˚C, cells were washed four times and incubated with 2 mL DMEM supplemented with 2% FBS, penicillin/streptomycin, and L-glutamine for 48 h. Supernatants were collected, centrifuged at 500g for 5 minutes, aliquoted, and stored at -80˚C.
Human and Jamaican fruit bat angiotensin converting enzyme 2 (ACE2) (Q9BYF1.2 and XM_037157556.1, respectively) were synthesized and cloned into pcDNA3.1+ (GenScript). All DNA constructs were verified by Sanger sequencing (ACGT). Baby hamster kidney (BHK) cells and immortalized Jamaican fruit bat kidney cells were seeded in black 96-well plates and transfected the following day with 100 ng plasmid DNA encoding human or bat ACE2 using either polyethylenimine (Polysciences) for the BHK cells, or Lipofectamine 3000 (ThermoFisher) for the Jamaican fruit bat kidney cells. All downstream experiments were performed 24 h post-transfection. Transfected cells were inoculated with equivalent volumes of pseudotype stocks. Plates were then centrifuged at 1200g at 4˚C for one hour and incubated overnight at 37˚C. 18–20 h post-infection, Bright-Glo luciferase reagent (Promega) was added 1:1 to each well, and luciferase was measured. Relative entry was calculated by normalizing the relative light unit for spike pseudotypes to the plate relative light unit average for the no-spike control.
Growth kinetics on transfected BHK cells
BHK cells were seeded for confluency in 24-well plates and transfected the next day with 100 ng plasmid DNA encoding human or hamster ACE2 or GFP, using polyethylenimine (Polysciences). All downstream experiments were performed 24 h post-transfection. Cells were inoculated with Delta (MOI = 0.01) in DMEM. After 1 h, wells were washed twice with 1 mL DMEM (T0), then incubated in 1 mL DMEM supplemented with 2% FBS. Supernatants were collected at T0, T12, T24, and T48 from unique individual wells.
Bat inoculation
Jamaican fruit bats (Artibeus jamaicensis) from a closed colony were utilized for these studies. Animals were cohoused in same sex groups of up to five animals per cage. The study comprised 30 animals total, 9 males and 21 females, of which 10 were pregnant. Animals were allowed to acclimate to the facility for 5 days. Twenty-one animals were intranasally and orally inoculated with a total of 1.5 x 105 TCID50 of hCoV19/USA/MD/HP05647/2021 (B.1.617.2/Delta, EPI_ISL_2331496), and four animals were inoculated with DMEM as controls via the same routes. On day 1, five animals were cohoused as sentinels (Fig. 2A). Caging scheme is depicted in Supplemental Table 2. All inoculations and subsequent manipulations were performed under isoflurane anesthesia. Six SARS-CoV-2 inoculated bats were euthanized one day post-inoculation (DPI), and five bats were euthanized on days 4 and 7 post-inoculation to assess viral replication in tissue samples. Fourteen inoculated animals, 5 sentinels, and 4 controls were euthanized 28 days post-inoculation for disease course assessment and shedding analysis. Blood was collected at baseline and on 14 and 21 DPI. Bats were weighed, body temperature was obtained via implanted transponder (BMDS IPTT-300), and oropharyngeal and rectal swabs were taken on day 0, 1, 2, 3, 4, 7, 14, 21 and 28. Swabs were collected in 1 mL DMEM with 200 U/mL penicillin and 200 µg/mL streptomycin. For the control group, mock swabs were performed to ensure animals underwent the same anesthesia protocols as infection groups. Bats were observed daily for clinical signs of disease. Necropsies and tissue sampling were performed according to IBC-approved protocols. Ventrodorsal, right, and left lateral thoracic radiographs were taken on day 0 and at euthanasia prior to any other procedures. Radiographs were evaluated and scored for the presence of pulmonary infiltrates by two board-certified clinical veterinarians according to a standard scoring system [33]. Briefly, each lung lobe (upper left, middle left, lower left, upper right, middle right, lower right) was scored individually based on the following criteria: 0 = normal examination; 1 = mild interstitial pulmonary infiltrates; 2 = moderate interstitial pulmonary infiltrates, perhaps with partial cardiac border effacement and small areas of pulmonary consolidation (alveolar patterns and air bronchograms); and 3 = pulmonary consolidation as the primary lung pathology, seen as a progression from grade 2 lung pathology. Thoracic radiograph findings were reported as a single radiograph score for each animal. To obtain this score, the scores assigned to each of the six lung lobes were added together and recorded as the radiograph score for each animal. Scores therefore may range from 0 to 18.
Viral RNA detection
Swabs were collected as described above. 140 µL was used for RNA extraction using the QIAamp Viral RNA Kit (Qiagen) using QIAcube HT automated system (Qiagen) according to the manufacturer's instructions with an elution volume of 150 µL. For tissues, RNA was isolated using the RNeasy Mini kit (Qiagen) according to the manufacturer’s instructions and eluted in 60 µL. Sub-genomic (sg) and genomic (g) viral RNA were detected by qRT-PCR [34, 35]. RNA was tested with TaqMan™ Fast Virus One-Step Master Mix (Applied Biosystems) using QuantStudio 6 or 3 Flex Real-Time PCR System (Applied Biosystems). SARS-CoV-2 standards with known copy numbers were used to construct a standard curve and calculate copy numbers/mL or copy numbers/g.
Virus titration
Viable virus in tissue samples was determined as previously described [15]. In brief, lung tissue samples were weighed, then homogenized in 1 mL of DMEM (2% FBS). Swabs were used undiluted. Vero E6 cells were inoculated with ten-fold serial dilutions of homogenate, incubated for 1 hour at 37°C, and the first two dilutions washed twice with 2% DMEM. For swab samples, cells were inoculated with ten-fold serial dilutions and no wash was performed. After 6 days, cells were scored for cytopathic effect. TCID50/mL was calculated by the Spearman-Karber method.
Serology
Maxisorp plates (Nunc) were coated with 50 ng receptor binding domain (RBD) protein (generated and kindly provided by Florian Krammer) per well. Plates were incubated overnight at 4˚C. Plates were blocked with casein in phosphate buffered saline (PBS) (ThermoFisher) for 1 hour at room temperature. Serum was diluted two-fold in blocking buffer and samples (duplicate) were incubated for 1 hour at room temperature. Secondary horseradish peroxidase (HRP)-conjugated recombinant A/G protein (Invitrogen, lot number WH 324034) diluted 1:10,000 was used for detection and visualized with KPL TMB two-component peroxidase substrate kit (SeraCare, 5120-0047). The reaction was stopped with KPL stop solution (SeraCare) and plates were read at 450nm. Plates were washed 3x with PBS-T (0.1% Tween) in between steps. The threshold for positivity was calculated as the mean plus 3x the standard deviation of negative control bat sera.
Virus neutralization. Heat-inactivated, irradiated sera were two-fold serially diluted in DMEM, and 100 TCID50 of SARS-CoV-2 was added. After 1 hour of incubation at 37˚C and 5% CO2, the virus:serum mixture was added to Vero E6 cells. CPE was scored after 5 days at 37˚C and 5% CO2.
Histopathology
Necropsies and tissue sampling were performed according to IBC-approved protocols. Tissues were fixed for a minimum of 7 days in 10% neutral buffered formalin with 2 changes. Tissues were placed in cassettes and processed with a Sakura VIP-6 Tissue Tek on a 12-hour automated schedule using a graded series of ethanol, xylene, and PureAffin. Prior to staining, embedded tissues were sectioned at 5 µm and dried overnight at 42°C. Using GenScript U864YFA140-4/CB2093 NP-1 (1:1000), specific anti-CoV immunoreactivity was detected using the Vector Laboratories ImPress VR horse anti-rabbit IgG polymer (# MP-6401) as secondary antibody. ACE2 was detected using a primary antibody from R&D Systems (cat#AF933, 1:100 dilution) and a secondary anti-goat IgG polymer (ImmPress, Vector Laboratories cat#MP-7405). TMPRSSII was detected using a primary antibody from Abcam (cat#ab109131, 1:2000 dilution) and a secondary horse anti-rabbit IgG polymer (ImmPress VR, Vector Laboratories cat#MP-6401).
The tissues were stained using the Discovery Ultra automated stainer (Ventana Medical Systems) with a ChromoMap DAB kit Roche Tissue Diagnostics (#760 − 159).
Next-generation sequencing of mRNA
Tissue sections were collected directly into 1 mL of Trizol (Thermofisher Scientific), 200 µL of 1-bromo-3-chloropropane (MilliporeSigma) was added, samples mixed, and centrifuged at 16,000g for 15 min at 4°C. 600 µL of RNA in aqueous phase was collected from each sample and passed through Qiashredder column (Qiagen) at 21,000g for 2 min to homogenize any remaining genomic DNA, then combined with 600 µL of RLT lysis buffer (Qiagen, Valencia, CA) with 1% beta mercaptoethanol (MilliporeSigma), and RNA extracted using Qiagen AllPrep DNA/RNA 96-well system. An additional on-column DNase-1 treatment was performed during RNA extraction. RNA was quantitated by spectrophotometry, and yield ranged from 0.4 to 17.8 µg. Two hundred nanograms of RNA was used as input for polyA pull-out and NGS library preparation following the Illumina Stranded mRNA prep workflow (Illumina). The NGS libraries were prepared, amplified for 15 cycles, AMPureXP bead (Beckman Coulter) purified, assessed on a TapeStation 4200 (Agilent Technologies), and quantified using the Kapa Library Quantification Kit (Illumina). Amplified libraries were normalized, pooled at equal 2nM amounts, and sequenced as 2 X 75 bp reads on the NextSeq instrument using three high output chemistry kits (Illumina). Raw fastq reads were trimmed of Illumina adapter sequences using cutadapt version 1.12, and then trimmed and filtered for quality using the FASTX-Toolkit (Hannon Lab). Remaining reads were aligned to the Artibeus jamaicensis genome assembly version 1.0 using Hisat2 [36]. Reads mapping to genes were counted using htseq-count [37]. Differential expression analysis was performed using the Bioconductor package DESeq2 [38] and data was further analyzed and plotted using ggplot2 (V3.4.0) as part of the tidyverse package (V1.3.2) [39]. Pathway analysis was performed using Ingenuity Pathway Analysis (Qiagen), and gene clustering was performed using Partek Genomics Suite (Partek Inc.).
Phylogenetic tree
To map the RML colony to the most likely Jamaican fruit bat subspecific lineage, we obtained the entire cytochrome b data set for Jamaican fruit bat from Larsen et al. (2007) [40], comprising 176 individuals across all documented subspecies (Simmons 2005), as well as additional Artibeus species and brown fruit-eating bat (Koopmania concolor) to root the phylogeny. In addition, we extracted the cytochrome b locus from the WHU_Ajam2 assembly (https://www.ncbi.nlm.nih.gov/assembly/GCA_014825515.1) as well as the more recently released CSHL_Jam assembly (http://compgen.cshl.edu/bat/). All cytochrome b sequences were then aligned using MAFFT v7.475 [41], and a maximum likelihood phylogeny was generated using RaxML-NG v1.1.0 [42] using the GTR + G substitution model and 1,000 bootstrap replicates; all other parameters were left in default settings. The final phylogeny is rooted on the brown fruit-eating bat for visualization.
Viral metagenome analysis on uninfected Jamaican fruit bat colony
Per-cycle base call (BCL) files were converted to fastq files and demultiplexed using bcl2fastq v2.20.0.244 (Illumina, Inc. San Diego, CA). Raw fastq files were concatenated for 114 samples across twelve tissues (spleen, brain, liver, upper and lower gastrointestinal tract, Peyer’s patches, blood, kidney, lung, mediastinal and mesenteric lymph nodes, and nasal turbinates) from five female and five male bats, and then processed through Metavirs pipeline [43] built in snakemake [44]. Briefly, adapter sequences were trimmed using cutadapt [45], and reads were filtered for host genomes using Bowtie2 [46]. Remaining reads were assembled in parallel using Megahit [47] and Metaspades [48], and contigs were annotated using the Contig Annotation Tool [49] and Kraken2 [50]. Results were visualized using Krona [51].
Metabolome analysis
A 1 cm sample of ileal tissue was collected from each bat for metabolome analysis at the time of necropsy. Liquid chromatography mass spectrometry (LCMS) grade solvents were used for all LCMS methods. Tissue samples were immersed directly in 0.4 mL of methanol and shredded at 30 Hz for 10 minutes using a tissue shredder and one stainless steel bead (Qiagen, 5 mm) per sample. Supernatant was then irradiated at 2 mRad for sample removal from high containment. 0.4 mL of water and 0.4 mL of chloroform were then added to each sample. Samples were shaken for 30 minutes at 4˚C and centrifuged at 16,000xg for 20 minutes to establish layering. 400 µL of the top (aqueous) layer was collected. The aqueous layer was diluted 5x in 50% methanol in water for LCMS injection. A subaliquot of the aqueous layer was taken for O-benzylhydroxylamine derivatization of carboxylic acids and SCFA analysis.
Samples were derivatized with O-benzylhydroxylamine (O-BHA) according to previously established protocols with modifications [52, 53]. A reaction buffer consisting of 1 M pyridine and 0.5 M hydrochloric acid in water was prepared fresh. A volume of 35 µL of the aqueous metabolite extract was sub-aliquoted. 10 µL of 1 M O-BHA in reaction buffer and 10 µL of 1 M 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide in reaction buffer were added to the sample. Samples were shaken at room temperature for 2 hours. The reaction was quenched with 50 µL of 0.1% formic acid for 10 minutes. Derivatized carboxylic acid compounds were extracted via the addition of 400 µL ethyl acetate. Following mixing and centrifugation at 16,000xg for 5 minutes at 4˚C to induce layering, the upper (organic) layer was collected and dried under vacuum. Samples were resuspended in 300 µL of water for LCMS injection.
Tributylamine and all synthetic molecular references were purchased from Millipore-Sigma. LCMS grade water, methanol, isopropanol, and acetic acid were purchased through Fisher Scientific. All samples were separated using a Sciex ExionLC™ AC system and measured using a Sciex 5500 QTRAP® mass spectrometer. Aqueous metabolites were analyzed using a previously established ion pairing method with modification [54]. Quality control samples were injected after every 10 injections. Samples were separated on a Waters™ Atlantis T3 column (100 Å, 3 µm, 3 mm x 100 mm) and eluted using a binary gradient from 5mM tributylamine, 5 mM acetic acid in 2% isopropanol, 5% methanol, 93% water (v/v) to 100% isopropanol over 15 minutes. Two distinct MRM pairs in negative mode were used for each metabolite. Derivatized short chain fatty acid samples were separated with a Waters™ Atlantis dC18 column (100 Å, 3 µm, 3 mm x 100 mm) and eluted using a 6-minute gradient from 5–80% B with buffer A as 0.1% formic acid in water and B as 0.1% formic acid in methanol. Short chain fatty acids and central metabolic carboxylic acids were detected using MRMs from previously established methods, and identity was confirmed by comparison to derivatized standards [52, 53]. All signals were integrated using MultiQuant® Software 3.0.3. Signals with greater than 50% missing values were discarded, and remaining missing values were replaced with the lowest registered signal value. All signals with a QC coefficient of variance greater than 30% were discarded. Metabolites with multiple MRMs were quantified with the higher intensity MRM. Filtered datasets were total sum normalized prior to analysis. Short chain fatty acid datasets were stitched to their corresponding polar metabolite dataset via common signals for lactate. Single and multi-variate analysis was performed in MarkerView® Software 1.3.1.
sPLSDA analyses of metabolomic data were performed in R using the mixOmics [55] package and loadings and variates visualized in GraphPad Prism. Abundances of specific metabolites were visualized and analyzed in GraphPad Prism. Correlations between metabolites and bacterial genera were calculated and visualized in GraphPad Prism.
Statistical Analysis
Significance tests were performed as indicated where appropriate for the data using GraphPad Prism 9. Unless stated otherwise, statistical significance levels were determined as follows: ns = p > 0.05; * = p ≤ 0.05; ** = p ≤ 0.01; *** = p ≤ 0.001; **** = p ≤ 0.0001. Exact nature of tests is stated where appropriate.
Data availability statement
All data are available on request from the corresponding authors or will be uploaded to FigShare and appropriate sequencing repositories. All material requests should be sent to V.J.M., [email protected].