Recombinant viruses
Genetic manipulations of herpesviruses were conducted using the bacterial chromosome vector (BAC) based lambda red recombineering system as described previously 54. Viral gene deletions, mutations, and insertion of transgenes to the herpesvirus genomes have been verified by PCR and Sanger sequencing. The expression of transgenes has been confirmed by ELISA assays. Supplementary Fig. 1 and Supplementary Fig. 3a-c demonstrate the structures and in vitro growth of recombinant HSV-1 KOS-BAC viruses. Supplementary Fig. 2a and Supplementary Fig. 3d-f illustrate the structures and growth of recombinant VZV-BACs, while Supplementary Fig. 2b,c list the structures of recombinant mCMV-BACs and HCMV-BACs. Human sodium iodide symporter (NIS) and beta subunit of human chorionic gonadotropin (bHCG) were inserted into the HSV-1 and VZV genomes as reporter genes 55.
Construction of recombinant HSV-1s: The eGFP expression cassette was amplified from the pcSV40KeGFP plasmid using primers No. 1-2 followed by the digestion with PciI/ KasI, and was then ligated into the PciI/ KasI treated pBeloBAC11 vector to obtain the BAC vector pSG. The BamHI site in pSG was removed by PCR amplification primers No. 3-4 followed by a blunt-end ligation, the obtained vector was designated as pSG2. To insert pSG2 between UL43 and UL44 of the HSV-1 KOS genome, the left and right homology arms were PCR amplified from KOS genome using two sets of primers, primers No. 5-6 and primers No. 7-8. The two homology arms were then digested with BamHI and HindIII, followed by being ligated into the BamHI/HindIII digested pSG2 vector, the obtained vector was designated as pSG3. The pSG3 vector was linearized with BamHI and then co-transfected with linear KOS genome into Vero cells (1 of linearized pSG3 and 1 of KOS genome were added per 5 105 cells). At 3 days post transfection, GFP+ plaques were picked and transferred to Vero cells. KOS-BAC episome DNA was isolated from the GFP+ cells using the Hirt method as described previously 54 and then transformed into the MegaX DH10B competent bacterial cells. Colonies containing full length KOS genome sequence were selected. Sequence integrity of the extracted KOS-BAC plasmid was confirmed by next generation sequencing (Genewiz Illumina MiSeq, 2 250bp configuration). The KOS-BAC plasmid was further transferred into the SW102 recombineering bacterial strain. To construct the NIS-P2A-bHCG cassette, the NIS and bHCG fragments were PCR amplified from pAAV-NIS plasmid and pAAV-bHCG plasmid using primers No. 9-12, and then were digested with HindIII/AflII and AflII/XbaI respectively, before being ligated into the HindIII/ XbaI digested pcSV40KeGFP vector, obtained plasmid was designated as pcSV40-pCMV-NIS-P2A-bHCG. The pCMV-NIS-P2A-bHCG cassette was amplified using primers No. 13-14 and was digested with SalI/ EcoRI before being ligated into the SalI/ EcoRI digested PL451 plasmid, obtained plasmid was designated as PL4512. The pCMV-NIS-bHCG-FRT-Kan-FRT cassette was amplified from the PL4512 plasmid using primers No. 15-16 and was then inserted between UL10 and UL11 of KOS-BAC within SW102 cells. The KanR sequencing in the KOS-BAC was then removed within the SW105 cells containing the ara-inducible Flpe gene 54, obtained vector was designated as KOS-BAC-NIS-bHCG (R2.3). For deletion of viral genes with only one copy of ORF in R2.3, the pEM7-galK cassette was PCR amplified from the pGalK plasmid with two long primers, each contains 50nt sequence at the 5´ terminal as homology arm. Then the whole ORF of viral gene was replaced by the galK cassette mediated by the homology arms. The galK cassette inserted into viral genome was eventually removed using a 100nt single stranded DNA with the same sequence of homology arms. Positive and negative selections for galK+and galK- colonies were performed as described previously 54. Recombinant viruses with deletion of viral genes (ICP47, UL41, ICP6, or ICP27) were constructed using the primers No. 17-28. RL1(ICP34.5) and RL2(ICP0) were replaced with the galK cassette and the KanR cassette (amplified from plasmid pRSV-rev-Kan) respectively, using primers No. 29-33 and primers No. 34-38. To construct the gB R858H point mutation, the nucleotide G at position 2573 of gB ORF was replaced with the galK cassette, which was then replaced with nucleotide A, primers used were No. 39-41. Transgenes (PEDF, scIL12 (single chain IL12-p70), and rIL15RA/IL15 (IL15Ralpha sushi/IL15)) were cloned from vectors (reagents No. 13, 16, 17, and 18) into the pSelect-zeo-fcy::fur plasmid using primers No. 46-47, and 52-59. PEDF was cloned into the pcSV40KeGFP plasmid using primers No. 64-67. Transgene cassettes were then inserted into the KOS-BAC backbone using primers No. 42-45. Recombinant HSV-1s were reconstituted in the Vero cells, or in Vero-cre cells (for removing the BAC sequence from virus constructs). The KOS construct carrying ICP27 deletion was failed to be reconstituted.
Construction of recombinant VZVs: To generate a VZV capturing BAC vector, two homology arms were amplified from the vOka episomal DNA using primers No. 70-73, and then were cloned into the pSG2 vector using restriction sites BamHI and HindIII, obtained vector was pSG6. The chloramphenicol resistance gene (CmR) cassette in the pSG6 vector was then replaced with the neomycin resistance cassette pPGK-pEM7-NeoR using primers No. 74-77, obtained vector was pSG7. pSG7 was linearized with BamHI and transfected into APRE-19 cells (2 of linearized pSG7 was added per 5 105 cells), at 24h post transfection live VZV virus was added to the transfected cells with multiplicity of infection (MOI)=0.05. At 48h post infection, G418 (Geneticin) was added to cells with final concentration of 800 /ml to enrich the GFP+ infected cells. The circular VZV BAC DNA was extracted from the GFP+ cells and transferred to DH10B competent cells. BAC colonies containing full length VZV genome were selected. VZV-BAC DNA was then transformed into the SW102 cells. Sequence integrity of the VZV-BACs was verified by next generation sequencing (Genewiz Illumina MiSeq, 2×250bp configuration). The pCMV-NIS-P2A-bHCG cassette, pCMV-NIS cassette and pCMV-bHCG cassette were amplified from plasmids pcSV40-pCMV-NIS-P2A-bHCG, pcSV40-pCMV-NIS and pcSV40-pCMV-bHCG using primers No. 78 and 79, and then inserted between ORF60 and ORF61 of vOka genome within SW102 cells. The scIL12 expression cassette was amplified and inserted between ORF60 and ORF61 of vOka genome using primers No. 80 and 82 or used to replace the AmpR sequence of the pcSV40-pCMV-bHCG cassette inserted using primers No. 80 and 81. The Tet-off drug-controllable pRSV-tTA- pTight-scIL12 cassette was amplified from vector pTet-off-mIL12BA-ZeoR and inserted between ORF60 and ORF61 of VZV genome using primers No. 82 and 113. The vOka carrying gBY881F hyperfusogenic mutation was constructed using primers No. 117-119. Recombinant VZVs were reconstituted in the ARPE-19 cells, or in ARPE-19-cre cells (for removing the BAC sequence from virus constructs).
Construction of recombinant mCMVs: To generate a mCMV capturing BAC vector, dTomato expression cassette was amplified using primers No. 94 and 96, and then ligated to the PCR amplified linear SG vector (with primers No. 93 and 95), obtained vector was pET. Left and right homology arms were then amplified from the mCMV Smith genome using primers No. 97-100 and were inserted into the pET vector to construct the pET2 capture vector. pET2 was then linearized with AvrII and co-transfected with mCMV linear genome into NIH-3T3 cells (1 of linearized pET2 and 1 of Smith genome per 5 105 cells). dTomato+ plaques were picked and enriched. Circular mCMV-BAC DNA was extracted and transformed into SW102 cells for genetic manipulations. Sequence integrity of the selected mCMV-BAC plasmid was confirmed by next generation sequencing (Genewiz Illumina MiSeq, 2×250bp configuration). For deletion of viral genes from mCMV-BAC, primers No.101-110 were used to amplify the galK cassette for replacing the viral genes m129-m131, m132-m135, m136-m138, m144, and m02-m16, respectively. The galK cassette within viral genome was then removed using a synthesized single stranded DNA consist of the same sequence of both homology arms. Recombinant mCMVs were reconstituted in NIH3T3-cre cells to remove the BAC sequence.
Construction of recombinant HCMVs: To generate a HCMV capturing BAC vector, left and right homology arms were amplified from the HCMV AD169 genome using primers No. 83-86, and then were cloned into the pSG2 vector, obtained vector was pSG4. pSG4 was linearized with BamHI and co-transfected with the HCMV linear genome (1 of linearized pSG4 and 1 of AD169 genome per 5 105 cells) into MRC-5 cells. GFP+ plaques were picked and enriched. Circular HCMV-BAC DNA was extracted and transformed into SW102 cells for genetic manipulations. Sequence integrity of the HCMV-BAC was confirmed by next generation sequencing (Genewiz Illumina MiSeq, 2 250bp configuration). The UL1-UL20 region and sequences (UL148, UL147A, UL147, UL146, UL145, UL139, UL138, UL136, UL135, UL133, UL148A-D, UL150 and UL150A) within the UL/b' region were found lost in the AD169-BAC, the reference AD169 sequence used for alignments was GenBank: FJ527563.1. To replace the UL131A like sequence in the AD169-BAC with the UL131A ORF from Merlin strain, the AD169 UL131A like sequence was first replaced by the galK cassette which was further replaced with the Merlin UL131 ORF, primers used were No. 87-90.
HSV-1s and CMVs were purified as described previously 56 with modifications. The supernatant of infected cells in forty 100mm dishes was collected when more than 90% of cells showed cytopathic effect (CPE) and was centrifuged at 3000 x g, 4 for 15min to remove large cell debris. Virus in the supernatant was then pelleted down using a Beckman Coulter Optima L-100K Ultracentrifuge with a SW32Ti rotor at 100,000 x g, 4 for 1h. Virus pellets were then incubated in PBS overnight at 4 , before being resuspended and loaded onto a continuous 15%-50% (w/v) sucrose density gradient in SW41 centrifuge tubes. The tubes were then centrifuged at 80,000 x g,4 for 1 h with a SW41 rotor. Virus bands were collected and diluted with PBS in SW41 centrifuge tubes, and were then centrifuged at 100,000 × g, 4 for 1h to pellet viral particles. Finally, the obtained virus pellets were incubated and resuspended in ice-cold PBS, before being aliquoted and stored at -80 . Virus titers were determined in Vero cells (for HSV-1), APRE-19 cells (for VZV), NIH3T3 cells (for mCMV) or MRC-5 cells (for HCMV) with plaque formation assay.
Cell lines
The Vero cells, Vero-cre cells, APRE-19 cells, ARPE-19-cre cells, NIH3T3 cells, NIH3T3-cre cells, MRC-5 cells, CT-2A-luc, B16-F10, and B16-F10-nectin1 were cultured in Dulbecco's Modified Eagle Medium (DMEM) with 10% fetal bovine serum (FBS) and 100 units/ml of penicillin/streptomycin. The EMT6 cells and EMT6-nectin1 cells were cultured in Waymouth's MB 752/1 Medium with 2mM L-glutamine, 100 units/ml of penicillin/streptomycin, and 15% FBS. The CT26 cells and CT26-nectin1 cells were cultured in RPMI-1640 Medium with 10% FBS and 100 units/mL of penicillin/streptomycin. The A20 cells were cultured in RPMI-1640 Medium with 10% FBS, 0.05 mM 2-mercaptoethanol, and 100 units/ml of penicillin/streptomycin. The human ovarian adenocarcinoma SK-OV-3 cells were cultured in McCoy's 5a Medium with 10% fetal bovine serum (FBS) and 100 units/ml of penicillin/streptomycin. The nectin1 stable mouse tumor cell lines were generated with lentivirus transduction followed by puromycin selection and fluorescence-activated cell sorting (FACS). Lentivirus expressing human nectin1 was produced with the lentivirus vector pLV-EF1-nectin1-PGK-puro.
Flow cytometry
The puromycin enriched tumor cells were resuspended in ice cold FACS Buffer (1 mM EDTA, 25 mM HEPES pH 7.0, and 1% BSA in Ca/Mg2+ free Hanks Buffer) (1×106 cells in 1ml buffer per tube), and then were stained with PE anti-human CD111 (Nectin-1) antibody or PE Mouse IgG1, κ Isotype Ctrl antibody together with the Live-or-Dye™ 350/448 for 30 min at room temperature. The nectin1-positive live cell populations were sorted with a BD FACSCanto Flow Cytometer and then were expanded in growth media at 37°C, 5%CO2. To analyze the expression of nectin1 in the expanded stable cells, 1×106 stable cells in 1ml ice cold FACS Buffer was stained with PE anti-human CD111 antibody or PE Mouse IgG1, κ Isotype Ctrl antibody together with the Live-or-Dye™ 350/448 for 30 min at room temperature. Wild type cells were stained parallelly as control. Stained cells were analyzed with a Bio-Rad ZE5 Cell Analyzer.
I-125 uptake assay
Vero cells were plated in six-well plates (5×106 cells per well) and cultured overnight before being infected with HSVs at MOI=0.01. Three replicates of wells were used for each group. Immediately prior to the uptake assay, cells were incubated at 37°C for 1h in 1 ml uptake buffer (Hank’s balanced salt solution (HBSS) containing 10 mM HEPES, pH 7.4). The radioactive substrate Na125I in 0.1 M NaOH was diluted in uptake buffer, and then 5 μl contained ~1,000,000 counts per minute was added to each well. Cells were incubated at 37°C for 50 minutes and then were washed twice with 2 ml ice-cold uptake buffer. Cells were lysed with pre-warmed 1 ml 1M NaOH. Cell lysate was transferred to tubes for gamma counting using a Wallac 1480 gamma counter.
Animal studies
All mice experiments were approved by Mayo Clinic Institutional Animal Care and Use Committee (IACUC) and were performed in compliance with Mayo Clinic IACUC guidelines. For studies using syngeneic immune-competent tumor models, mouse tumor cells suspended in PBS (5×107 cells/ml for A20 model, 1×107 cells/ml for all other models) were subcutaneously implanted to both flanks (100µl per side of flank) of 5~6 weeks old female mice (Balb/c or C57BL/6J). When average tumor diameter reached 5 mm, viruses suspended in PBS were intratumorally (IT) delivered into the tumors on the right flank (50µl per IT dose) or were intravenously (IV) delivered (100µl per IV dose). Totally three doses were given for each mouse at indicated time points. For studies using immune-comprised tumor models, human tumor cells suspended in PBS (5×107 cells/ml) were subcutaneously implanted to the right flanks (100µl per mouse) of 6 weeks old female nude mice. When average tumor diameter reached 5 mm, viruses suspended in PBS were IT delivered into the tumors (50µl per IT dose). The body weight and tumor volumes were monitored three times per week post virus injection. Blood and tumor samples were collected at indicated time points. Animals were euthanatized according to predetermined criteria (weight loss equal to or exceeding 20% of baseline, tumor burden that equals or exceeds 10% of body weight, development of hind limb paralysis or other signs of neurotoxicity, or focal motor deficits). Mice with ulcerated tumors will be monitored daily for body weight, tumor size and status of ulceration. Spleens were collected when animals were euthanatized. With no specific indications, the doses for IT delivery were 1×106 plaque formation units (PFUs) of HSV-1, 1×106 PFUs of HSV-1+1×105 PFUs of CMV, or 1×105 PFUs of CMV in 50ul PBS per injection. When a ten-fold higher dose was injected for each virus, the doses used were: 1×107 PFUs of KOS R30.4, 1×107 PFUs of KOS R30.44, 1×107 PFUs of KOS R30.44+1×107 PFUs of KOS R30.5, 1×107 PFUs of KOS R30.44+1×106 PFUs of mCMV, or 1×107 PFUs of KOS R30.44+1×107 PFUs of KOS R30.5+1×106 PFUs of mCMV in 50ul PBS per IT injection.
RT-qPCR
Tumors were cut into small pieces and crushed on ice. Total RNA was extracted from around 100 mg tissue per sample using the RNeasy Midi Kit according to the manufacturer’s instructions. Power SYBR™ Green RNA-to-CT™ 1-Step Kit was used for semi-quantifying the viral gene expression levels, and 100 ng of total RNA was added for each reaction. Primers used were listed in Supplementary Table 1 primer No. 120-125. Relative viral gene expression levels (fold change) were analyzed using the ∆∆Ct method and presented as mean ± SEM from replicate samples.
Immunohistochemistry analysis
Tumors collected on day 2 post virus injection were fixed in 4% paraformaldehyde (PFA) for 3 days before being dehydrated with 30% sucrose in PBS at 4°C for additional 2 days. The tumor samples were then subjected to cryosectioning with a Leica CM1860 Cryostat, sickness of tumor slices was set at 40µm. Tumor slices were penetrated with 1% Triton X-100 in TBS buffer and stained with 4′,6-diamidino-2-phenylindole (DAPI) and conjugated antibodies (Supplementary Table 1 Reagent #57-66). Images were collected with a Zeiss LSM 780 confocal microscope and analyzed with the software ZEN 3.3. Images with maximum intensity projection were presented.
Cytokine array assays
On 3 dpt, tumors with similar size (100-120 mm3) were collected from each group. On 9 dpt, tumors with the smallest size in each group were collected. Tumors were crushed and homogenized in PBS with protease inhibitors (10 μg/ml Aprotinin, 10 μg/ml Leupeptin, and 10 μg/ml Pepstatin). Triton X-100 was added to the lysate to a final concentration of 1%. Tumor samples were then freezed at -80°C, thawed, and centrifuged at 10,000×g for 5 min to remove cell debris. Cytokine levels in tumor lysate were detected using the Proteome Profiler Mouse XL Cytokine Arrays and Mouse Cytokine Panel A Arrays according to manufacturer’s instructions. Protein concentrations of samples were determined with BCA assay, and 3 mg and 1 mg of total protein were loaded onto the membranes for 3 dpt and 9 dpt tumor lysate samples, respectively. Cytokine levels in supernatant of cultured human macrophages were detected using the Proteome Profiler Human XL Cytokine Array Kit. Cell culture supernatant samples were loaded onto the membranes with 100 µg total protein per sample. Membranes for each experiment were developed with autoradiography on a single film. Pixel densities of dots on developed X-ray films were scanned using the ImageJ software. Signals between membranes were normalized using the pixel density of reference dots on each membrane. Cytokines that showed more than 1.25× or less than 0.75× fold regulation were displayed.
ELISA analyses
ELISA analyses were performed according to the manufacturer's instructions. ELISA kits were listed in Supplementary Table 1.
Culture of human monocyte-derived macrophages
To generate monocyte-derived macrophages, human peripheral blood CD14+ monocytes in monocyte attachment medium were plated to six-well plates (1×106 cells in 1ml medium per well) and cultured at 5% CO2 and 37°C for 1h to let the monocytes attach. Adherent cells were washed with pre-warm monocyte attachment medium for three times and then cultured with the macrophage generation medium (RPMI 1640 with 10% human AB serum, 1μg/ml hIL-4, 1μg/ml hIL-10, and 1μg/ml hM-CSF) for 7 days, medium and cytokines were renewed on day 3. Differentiated macrophages were detached with macrophage detachment solution, counted, and seeded to plates in macrophage generation medium for in vitro analyses. Immunofluorescence staining using the conjugate antibodies against human macrophage markers CD206 and CD163 was performed.
Spleen cell expansion and cytotoxicity assays
Spleens were collected when condition of mice meet predetermined euthanasia criteria or at experimental terminal. Freshly collected spleens were crushed in RPMI-1640 Medium, and tissue debris was removed with 70µm cell strainers. Cell suspension passed through the strainers was centrifuged at 1,800 rpm for 5min. Cells were resuspended and incubated in 5ml RBC Lysis Buffer for 5min before being pelleted down at 1,800 rpm. Spleen cell pellets were washed twice with PBS and then stored at -80°C.
To analyze the cytotoxicity of spleen cells against tumor cells, the isolated spleen cells were first expanded with spleen cell expansion medium (TexMACS Medium with 10%FBS, 60ng/ml mIL2, 1µM 2-mercaptoethanol, and 100 units/ml of penicillin/streptomycin) at 5% CO2 and 37°C until more than 1×108 cells can be obtained for each spleen sample. The expanded spleen cells were then stimulated with target tumor cells in six-well plates. Briefly, tumors cells were seeded to six-well plates (5×105 cells per well) and cultured overnight, then spleen cells were added to the wells (5×106 cells per well) and co-incubated for 4 days, media was renewed every day. On day 4 post co-culture, the suspension cells in each well were collected, counted, and designated as stimulated spleen cells, and the viability of the stimulated spleen cells were determined with trypan blue exclusion. Tumor cells and the stimulated spleen cells were sequentially seeded to 96-well plates (2×104 tumor cells and 2×104 live stimulated spleen cells per well), and were co-cultured in spleen cell expansion medium at 5% CO2 and 37°C. The control wells were loaded only with 2×104 tumor cells per well. At day 4 post co-culture, cell medium was renewed to remove the suspension cells, and cytotoxicity was analyzed with TACS MTT Cell Proliferation Assay Kit.
Elispot assays
IFN-γ elispot assays were conducted using the Mouse IFN-gamma ELISpot Kit as described previously with modifications 36. Spleens were collected when condition of mice meet predetermined euthanasia criteria or at experimental terminal. Spleen cells suspended in RPMI-1640 Medium (supplemented with 10% FBS, 10 ng/ml mIL2, 1µM 2-mercaptoethanol, and 100 units/ml of penicillin/streptomycin) were loaded to the 96-well plates (1×105 or 5×105 live spleen cells per well). Two technical replicates were set for each sample. Antitumor immunity was analyzed by loading live tumor cells (1×105 tumor cells per well) to the spleen cell pre-seeded wells. Antiviral immunity was analyzed by adding UV-inactivated viral particles (1×106 inactivated particles per well) to the spleen cell pre-seeded wells. After tumor cells or viral particles were loaded, the plates were cultured at 5% CO2 and 37°C for 3 days before spots were developed and counted. Phytohaemagglutinin P (PHA-P) (100 µg/ml) was added in the positive control groups. To prepare UV-inactivated HSV-1 and CMV particles, purified and titrated infectious virus stocks were UV-inactivated (120 mJ/CM2, 30min) with a CL-1000 Ultraviolet Crosslinker. Inactivation of virus was confirmed by titration.
Antiviral neutralizing antibody titration for serum samples
Plaque reduction neutralization test (PRNT) was performed to quantify the anti-HSV-1/mCMV neutralizing antibody titers in the mouse serum samples. Briefly, Vero or NIH-3T3 cells were plated to 24-well plates (2×105 cells per well) and cultured overnight to form cell monolayers. Mouse serum samples were heat-inactivated (56°C for 30 min) and 2-fold diluted beginning with a 1:25 dilution (1:25, 1:50, 1:100, 1:200, 1:400, and 1:800), and were added to 96-well V-bottom plates. A total of 60 PFUs of HSV-1 KOS-BAC or mCMV Smith-BAC virus were added per well. The virus-serum mixtures (150 µl per well) were inoculated at 37°C for 1h before being transferred to the Vero or NIH-3T3 monolayers. Virus without serum was added to the virus-only control wells. After 1h’s absorption, the inoculums were removed and the DMEM/agarose overlay medium (containing 10% FBS, 1% low-melting point agarose, 0.375% sodium bicarbonate, and 100 units/ml of penicillin/streptomycin) was added to the cells. The plates were incubated at 5% CO2 and 37°C before GFP or dTomato positive plaques were counted for all groups. PRNT50 was calculated with “R 4.2.2” package using the algorithm described previously 57.
Single cell RNA sequencing of spleen cells
Spleen cells after expansion and stimulation with tumor cells as described above were counted and subjected to 10X Genomics® Chromium™ 3' gene expression RNA-seq provided by Azenta. Samples must pass the minimal QC requirement (more than 70% cell viability after dead cell removal) for library preparation. Target number of cells per sample was 3,000, and target number of reads per cell was 50,000. Data was analyzed by ROSALIND® (https://rosalind.bio/). Quality scores were assessed using FastQC. Cell Ranger was used to align reads to the Mus musculus genome build GRCm38, count UMIs, call cell barcodes, and perform default clustering. Individual sample reads were normalized via Relative Log Expression (RLE) using DESeq2 R library 58, DEseq2 was also used to calculate fold changes and p-values and perform optional covariate correction. The t-distributed stochastic neighbor embedding (t-SNE) Projection of Cells was colored by the cluster identification for each cell-barcode. The axes correspond to the 2-dimensional embedding produced by the t-SNE algorithm. Cells with high variance were removed from this plot.
Statistics
Statistical analyses of data were performed using GraphPad Prism 8 software. Values were presented as mean ± Standard Error of Mean (SEM). All data sets passed Normality test (Shapiro-Wilk test or Kolmogorov-Smirnov test) before being subjected to one-way ANOVA (Dunnett's multiple comparisons test), two-way ANOVA (Tukey's multiple comparisons test or Dunnett's multiple comparisons test), Unpaired t-test (two tailed), or Multiple t-tests to calculate p-values for group comparisons. p < 0.05 was considered significant. ns, not significant, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.