Human cell lines and culture conditions
SV40-immortalized human fetal lung fibroblast cells MRC5_VA were maintained in high glucose DMEM medium (Thermo Fisher Scientific, 11965118) supplemented with 10% v/v fetal bovine serum (PAN-Biotech, P30-3302) and 5% penicillin/streptomycin (Thermo Fisher Scientific, 15140122) at 37°C with 5% CO2 and routinely passaged 2–3 times a week. Cell lines were confirmed to be mycoplasma-free.
Animals
Animal experiments undertaken in this study were with approval of the Guangzhou Medical University Animal Care and Use Committee. All mice were maintained under specific pathogen-free conditions in individually ventilated cages at 19–23°C with light from 07:00 to 19:00.
CRISPR/Cas9-mediated genome engineering was conducted to generate a Zfas1−/− knockout C57BL/6 mouse. Cas9 vector and guide-RNA (target sequence: gRNA-A, 5’-GGGGTGGACCTCTGTACTATAGG-3’; gRNA-B, 5’-ACCATGCTCAAGTTGCTGCAAGG-3’) for mouse Zfas1 3’UTR were generated by in vitro transcription, followed by injected into fertilized eggs for knockout mouse production. The positive founders were genotyped by PCR and DNA sequencing analysis. The sequences of the primers used for genotyping were depicted in Table S11.
Acute effects in the skin of shaven Zfas1−/− mice following exposure to UV-B at dose of 500 J/m2/day during 4 consecutive days were conducted by killing three mice per genotype 3 days after the last treatment. Skin samples were routinely processed (hematoxylin-eosin staining) for histopathologic examination as described before(Xu et al., 2019).
Western blotting
For whole cell extracts, cells pellets were lysed in NP-40 lysis buffer [50 mM Tris-HCl pH 7.5, 500 mM NaCl, 2 mM EDTA, 0.5% (v/v) NP-40, 0.5 mM DTT, and Protease Inhibitor Cocktail (made in house)]. Protein samples were resolved by 10% or 15% SDS-PAGE gels and transferred to PVDF membrane (GE healthcare Life Sciences, 10600023), followed by blocking in 5% (w/v) skimmed milk in TBST (50 mM Tris HCl pH 7.6, 150 mM NaCl, 0.1% (v/v) Tween 20) for 1 h at room temperature and incubated with primary antibody (in 5% (w/v) skimmed milk in TBST) overnight at 4°C. Antibody against tubulin or GAPDH served as loading controls. Membranes were washed three times in TBST, followed by incubation with 1:1000 diluted HRP-conjugated secondary antibodies in 5% (w/v) skimmed milk in TBST. After extensive washing with TBST, the proteins were visualized using Chemiluminescent Substrate ECL reagent (Thermo Fisher Scientific, 34075).
CPD and γH2A.X immunofluorescence
Cells were maintained on coverslip and treated with UV-C (10 J/m2) and recovered for the indicated period. After fixing in 4% paraformaldehyde for 15 min, cells were permeabilized in PBS-T (PBS plus 0.3% (v/v) Triton X-100), and the DNA was denatured in 2 M HCl for 5 min (for CPD immunofluorescence). Nonspecific binding was blocked in PBS-T (PBS plus 0.3% (v/v) Triton X-100) with 10% skimmed milk. The anti-CPD antibody (Cosmo Bio, CAC-NM-DND-001) or anti-γH2A.X antibody (Abcam, ab22551) was then applied in blocking solution (1:1000) and incubated at 4°C overnight. Secondary anti-mouse antibody conjugated to Alexa-fluor-488 was added to the coverslip for 1 h at room temperature. The coverslips were mounted onto slides using DAPI (Vector Laboratories, Inc. Peterborough, UK) and imaged as previously described(Liu et al., 2022).
Reverse transcriptase quantitative PCR
Total RNA was extracted using the RNeasy kit (QIAGEN, 74104) for nascent and mature RNA, following the instructions of the manufacturer including an on-column DNase digestion (QIAGEN, 79254). Reverse transcription was performed using PrimeScript™ RT Reagent Kit with gDNA Eraser (Takara, RR047A). cDNA was amplified using iQ SYBR green Supermix (Bio-Rad, 1708880) with 30 cycles of 15 s denaturation at 94°C, 15 s annealing at 60°C, and 20 s extensions at 72°C. Primer sequences are listed in Table S11. Unless otherwise noted, reference gene (GAPDH) normalized RNA expression was compared between variable (ex. UV-treated) and control samples using the Livak equation described before(Livak and Schmittgen, 2001).
Cell synchronization and cell cycle profile analysis
For all G1/S synchronization with thymidine, a double thymidine block was used as follows: cells were incubated for 14 h with 2 mM thymidine (Sigma-Aldrich, T1895), released for 9 h in fresh growth medium supplemented with 24 µM deoxycytidine (Sigma-Aldrich, D3897) after washing out the thymidine, and then blocked again with 2 mM thymidine for 14 h to arrest all cells at the beginning of S-phase. Cell cycle arrest was subsequently released with two washes of thymidine-free medium. After release, cells were treated with UV-C irradiation (10 J/m2) and harvested at the indicated time intervals.
Cell cycle analysis was carried out by flow cytometry. Briefly, cells were seeded onto 10-cm culture dishes, treated as indicated, collected, fixed, and stained with 25 µg/mL propidium iodide (Sigma-Aldrich, 81845) and 10 µg/mL RNAse A (Sigma-Aldrich, RNASEA-RO) in PBS for 30 mins at 37°C, acquired on a FACSCalibur flow cytometer instrument and analyzed using FACStation and FlowJo software (BD Biosciences). A minimum of ten thousand events were analyzed for each sample.
RNA-seq library construction and sequencing
G1/S-phase-synchronized MRC5_VA cells were either left untreated or treated with 10 J/m2 of UV-C irradiation prior to release, followed by recovery for the indicated time periods from 3 to 96 hr. Total RNA was extracted and purified using RNeasy Mini Kit (QIAGEN, 74104) and analyzed on a 2100 Bioanalyzer (Agilent Technologies). All samples had an RIN (RNA Integrity Number) value of greater than 9.6. Subsequently, ribosome RNA was removed from the purified total RNA using Epicentre Ribo-Zero™ rRNA Removal Kit (Epicentre, MRZH11124), and the remaining RNA was used for library preparation using TruSeq Stranded Total RNA Sample Prep Kit (Illumina, 20020597). Libraries were quantified fluorometrically using Qubit® dsDNA HS Assay Kits (Thermo Fisher Scientific, Q32851) on a Qubit 2.0 Fluorometer (Thermo Fisher Scientific). All libraries (> 2 nM/µl) were sequenced on an Illumina NovaSeq 6000 (PE150 run) according to the manufacturer’s instruction to a depth of 220–340 million reads on average.
The sequence data were deposited in the NCBI Gene Expression Omnibus (GEO), with BioProject accession number, GSE239617.
Processing, analysis and graphic display of RNA-seq data
Raw reads were pre-processed with sequence grooming tool FASTQC followed by sequence alignment using HISAT2(Kim et al., 2015). Transcript levels were quantified as fragments per kilobase of transcript per million mapped reads (FPKM) generated by TopHat/Cufflinks(Kim et al., 2013). SAMtools(Li et al., 2009) were used to convert sam files to bam files in order to make raw data visible on IGV (Integrative Genomics Viewer) or UCSC Genome Browser. Transcript levels were converted to the log-space by taking the logarithm to the base 2. R studio (ggplot2 and gplots packages) was used to run custom R scripts to perform box plots, principal component analysis (PCA), dendrograms, and heatmaps. Samples were analyzed through DESeq2(Varet et al., 2016) to obtain log2 fold change and its respective p value. Differentially expressed transcripts have been identified on these transformed values by using the criteria of log2 (Fold Change) ≥ 1 and padj < 0.05. Gene ontology analysis of DEGs (differentially expressed genes) was performed using clusterProfiler(Yu et al., 2012).
Identification of novel lncRNAs from RNA-seq
The known noncoding RNAs expressed in at least one sample were identified by blasting the transcripts against the NONCODE v6.0 database(Zhao et al., 2021) using the following selection criteria: identity > 0.9, coverage > 0.8, and E-value < 105. These transcripts were named as the ID number in the NONCODE v6.0 database.
Based on the features of lncRNA, a series of stringent screening conditions were established to identify novel lncRNAs from the RNA-seq data: (1) Cuffmerge was used to merge the transcripts which were obtained by splicing and remove transcripts with uncertain directions and < 200 bp in length; (2) Cuffcompare was used to filter out transcripts that overlap with the database annotation exon region; (3) Cuffquant was applied to calculate the expression level of each transcript and transcripts with FPKM ≥ 0.2 were selected; (4) Four analysis tools, including CNCI (Coding-Non-Coding-Index, v2), CPC (encoding potential calculator, 0.9-r2), Pfam Scan (v1.3) and phyloCSF were used to predict the coding potential of the transcripts.
ZFAS1 cloning and sequencing analysis (RACE)
3’ and 5’ RACE was performed using the SMARTer® RACE 5’/3’ Kit (Takara, 634858). RNA was extracted from either untreated or 10 J/m2 UV-C exposed MRC5_VA cells, and RACE was performed according to the standard manufacturer’s protocol. Primers used for RACE assay were depicted in Table S11.
Plasmid construction
For construction of the ZFAS1-FLAG plasmids, the predicted ORF of ZFAS1, full length of ZFAS1, and the CDS of ACTB gene were cloned into the Sgfl and Mlul sites of the eukaryotic expression vector pCMV6-Entry with N-terminal start codon ATG and C-terminal Flag tag. Primers used for the cloning were depicted in Table S11.
RNA fluorescence in situ hybridization (FISH)
To detect ZFAS1 RNA, FISH analysis of MRC5_VA cells maintained on sterile glass coverslips was performed with a fluorescence in situ hybridization kit (RiboBio, C10910). Briefly, cells were rinsed in PBS and then fixed in 3.7% formaldehyde plus 10% acetic acid in PBS (pH 7.4) for 15 min at room temperature. Cells were subsequently permeabilized in PBS containing 0.2%-0.5% Triton X-100 and 5 mM vanadyl ribonucleoside complex (10 mM) (NEB, S1402) on ice for 5 min, washed in PBS 3 x 10 min and rinsed once in 2 x SSC buffer (Thermo Fisher Scientific, AM9770). Hybridization was carried out using oligodeoxynucleotide probes for ZFAS1, U6, and 18S (RiboBio) in a moist chamber at 37°C for 12–16 hours while protected from light according to the protocol for adherent mammalian cell lines. The next day, cells were counterstained with DAPI. Images were acquired on a laser scanning confocal microscope (Leica, TCS SP8).
Chromatin/RNA immunoprecipitation combined with quantitative PCR (ChIP-qPCR/RIP-qPCR)
MRC5_VA cells were plated onto 15-cm dishes at a density of 5 x 106 cells per dish and treated with 10 J/m2 UV-C exposure. After 3 hr of treatment, chromatin immunoprecipitation (ChIP) assays were carried out as described before(Wang et al., 2014). For each ChIP assay, the following antibodies (2 µg) were used: RNA polymerase II (Abcam, ab26721), H3K4me3 (Abcam, ab8580), H3K27ac (Abcam, ab4729), or immunoglobulin G control (Abcam, ab172730). ChIP enriched DNA and input DNA were subjected to qRT-PCR analysis with SYBR Green Supermix (Bio-Rad, 1708880). Enrichment by ChIP assay on the specific genomic regions was assessed relative to the immunoglobulin G control.
For RIP assay, SUPERase-In™ RNase inhibitor (1000 U/ml, Ambion, AM2694) and protease inhibitor were added into cell lysis buffer, and Ribonucleoside Vanadyl Complex (10 mM, NEB, S1402) was added into washing buffer. Antibodies specific to human NME1 (Proteintech, 11086-2-AP), SSRP1 (BioLegend, 609701) and XRCC5 (Proteintech, 16389-1-AP) were applied in the assay.
The primer sets were listed in Table S11.
6mA-IP-qPCR
Genomic DNA was isolated from untreated or UV-irradiated MRC5_VA cells and treated with RNase to remove RNA contamination. The DNA sample was then sonicated to produce 200–400 bp fragments. The fragmented DNA was incubated with specific anti-6mA antibody (Synaptic Systems, 202003) in immunoprecipitation buffer (2.7 mM KCl, 2 mM potassium phosphate, 137 mM NaCl, 10 mM Sodium phosphate, 0.05% Triton X-100, pH 7.4) for 2 h at 4°C. The mixture was then immunoprecipitated by Protein A/G Plus-Agarose (Santa Cruz, sc-2003) that was pre-blocked with bovine serum albumin (BSA) at 4°C for 2 h. After extensive washing, the bound DNA was eluted from the beads in elution buffer (50 mM Tris-HCl, pH 8.0, 1% SDS, 10 mM EDTA) at 65°C, treated with proteinase K and purified using QIAquick PCR Purification Kit (Qiagen, 28104). Fold-enrichment of each fragment was determined by quantitative real-time PCR. The primers used for 6mA-IP-qPCR assays were depicted in Table S11.
Clonogenic assay
Cultured MRC5_VA cells depleted with ZFAS1 or overexpressing ZFAS1 were treated with Trypsin/EDTA, counted and seeded onto 6-well plates at a low density (200–500 cells/well) for colony formation. Cells were incubated overnight followed by UV-C irradiation under the dose of 2, 5 and 10 J/m2. Colonies were allowed to form over 14–21-day period and were stained with crystal violet and counted.
ZFAS1 knockdown and overexpression
The target sequences of ZFAS1, shared by both short and long transcript variants, were 5’- GCAGACATCTACAACCTTC − 3’ for shZFAS1-A, 5’- ATGGATTTTGGAAGAGGGA − 3’ for shZFAS1-B, and 5’- GAGTTTAAAAGGCTGTGCC − 3’ for shZFAS1-C (shZFAS1-C was used in the following functional assays). These sequences were cloned into lentiviral shRNA expression hCMV-SMARTvector.
For overexpression assays in MRC5_VA cells, the Lipofectamine 3000 (Invitrogen) was used according to the manufacturer’s protocol. The following plasmids were used: pCMV6-Entry-human ZFAS1 and pCMV6-Entry.
Alkaline comet assay
Comet assay was performed according to ENZO comet assay kit (Enzo life sciences, ADI-900-166) protocol for alkaline comet assay of adherent cells. Samples were visualized using a confocal microscope (Leica, TCS SP8) with 40 x magnification. Comet parameters were analyzed automatically with image acquisition and analysis software package CaspLab.
EdU staining
Wild-type or ZFAS1-depleted MRC5_VA cells were cultured on coverslips and washed with PBS once, followed by irradiation with 10 J/m2 UV-C. After UV-C treatment, cells were immediately incubated with serum-free fresh medium supplemented with 10 µM EdU for 2 h. Cells were then washed with PBS once, followed by fixation with 3.7% formaldehyde for 15 minutes and permeabilization with PBS containing 0.5% Triton X-100 for 20 minutes. After extensive washing with PBS, cells were blocked with 10% FBS in PBS for 30 min. Incorporated EdU was detected by fluorescent-azide coupling reaction (Click-iT™ EdU cell proliferation kit, Invitrogen, C10337). Photograms of the cells were captured with a confocal microscope (Leica, TCS SP8). Captured images were processed and analyzed with ImageJ Software. At least 200 non-S-phase cells were randomly selected per sample. Data points presented in the text are the averages of intensities.
5’ Ethynyl Uridine Staining
EU staining to detect newly synthesized RNA was performed according to the manufacturer’s instructions (Click-iT™ RNA imaging Kits, Invitrogen, C10329). The wild-type or ZFAS1-depleted MRC5_VA cells were exposed to 10 J/m2 UV-C irradiation and incubated for the indicated period of time. Media was replaced with fresh media containing 0.75 mM 5’ Ethynyl Uridine (EU) and cells were incubated for another 2 hrs. EU-containing media was then removed and cells were fixed in PBS buffered formaldehyde (3.7%) for 45 mins at room temperature, washed once with PBS and followed by permeabilization with 0.5% Triton X-100 diluted in PBS for 30 mins. Cells were washed once with PBS and then Alexa Fluor 488 Azide fluorophores were covalently attached to EU-containing RNA by click reaction for 1 hr at room temperature. Cells were then counterstained and mounted with mounting medium containing DAPI. Automated image acquisition of at least 5 fields per well was performed (Leica, TCS SP8).
Chromatin immunoprecipitation and NGS sequencing (ChIP-Seq)
RNAPII-CTD ChIP-Seq was performed under following conditions: the wild-type and ZFAS1-depleted MRC5_VA cells were UV irradiated (10 J/m2 UV-C), followed by incubation for designated time intervals (3 hr, 6 hr, and 12 hr). Cells were harvested by trypsin-treatment and fixed in suspension with formaldehyde (1% final concentration) for 15 min at room temperature, with rotation. The crosslinking reaction was quenched with glycine (125 mM final concentration) for 5 min. Cells were then washed twice with ice-cold 1 x PBS, suspended in 1 ml of ChIP cell lysis buffer (5 mM HEPES pH 8.0, 85 mM KCl, 0.5% NP-40, and protease inhibitors) and incubated on ice for 5 min. Nuclei were pelleted by centrifugation at 3,900 x g for 5 min at 4°C, followed by suspension in ChIP nuclear lysis buffer (50 mM Tris-HCl pH 8.1, 10 mM EDTA pH 8.0, 1% SDS, and protease inhibitors) and incubated for 5 min on ice. Nuclear lysate was sheared by using an ice-water bath-embedded Bioruptor sonication system at high power, 30 sec on, 30 sec off mode for 5–10 min. The size of the sheared DNA was checked by 2% agarose gel electrophoresis to be between 300–500 base pairs (bp). Sonicated chromatin was cleared by centrifugation at 20,000 x g for 15 min at 4°C. Before the immune-precipitation, chromatin was diluted 1:5 with ChIP dilution buffer (0.01% SDS, 1.15 Triton X-100, 1.2 mM EDTA pH 8.0, 16.7 mM Tris-HCl pH 8.1, 167 mM NaCl, and protease inhibitors). 1 µg of anti-RPB1 phospho-Ser5-CTD (RPB1-Ser5, clone 3E8, Millipore, 04-1572-I), phospho-Ser2-CTD (RPB1-Ser2, clone 3E10, Millipore, 04-1571), or rat IgG (Invitrogen), was bound to 15 µl of Protein A/G agarose beads (Invitrogen) in 200 µl 5% BSA in 1 x PBS for 1 hr, before being washed twice with 500 µl of the same buffer. The sonicated chromatin was incubated with the antibody-conjugated beads overnight at 4°C with gentle rotation. Beads were washed twice with 1 ml of each of the following buffers: ChIP low salt buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl pH 8.1, 150 mM NaCl); ChIP high salt buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl pH 8.1, 500 mM NaCl); and ChIP LiCl buffer (10 mM Tris-HCl pH 8.0, 250 mM LiCl, 1% NP-40, 1% deoxycholic acid, and 1 mM EDTA). Beads were washed once with 1 ml of TE buffer (pH 8.0) and centrifuged for 5 min at 14,000 x g before removing the buffer. Beads were finally suspended in 40 µl Elution buffer (50 mM Tris-HCl pH 8.0, 10 mM EDTA, 1% SDS) and incubated at 65°C for 15 min. The eluted ChIP material was incubated at 65°C overnight to reverse the crosslinking with an additional 90 µl of 1% SDS in 1 x TE buffer and 1 µl of 10 mg/ml RNase A. In parallel, the input sample was also RNase treated and reverse crosslinked overnight at 65°C. Proteinase K (100 µg) and Glycogen (20 µg) were added to the eluted ChIP material and incubated for 2 hr at 37°C, and DNA was extracted by column purification (MinElute, QIAGEN, 28006). 1 ng of ChIPed DNA samples were submitted for further manipulation by standard ChIP-Seq library preparation techniques (NEBNext® Ultra™ DNA Library prep kit, NEB, #E7103) and sequenced under Illumina NovaSeq 6000 system, resulting in the production of 150 bp paired-end reads.
Alignment of the ChIP-seq reads was performed as follows: low-quality sequence reads and adapters were filtered out by Trimmomatic (v3.36)(Bolger et al., 2014). The trimmed reads were then aligned to the human reference genome (hg38) with the Burrows-Wheeler Aligner (BWA-v0.7.12-r1039)(Li, 2014). Duplicate reads were removed from the aligned reads by Biobambam2 (v2.0.72)(Tischler and Leonard, 2014). The mapped reads were visualized with the Integrated Genome Viewer (IGV-v2.3.90)(Thorvaldsdóttir et al., 2013). Further analysis was conducted using Bioconductor(Gentleman et al., 2004).
The sequence data were deposited in the NCBI Gene Expression Omnibus (GEO), with BioProject accession number, GSE239617.
ChIP-Seq TSS profiles
From the aligned ChIP-Seq reads, heatmaps and average density profiles were generated to draw spatial distributions of RPB1 along gene bodies under different conditions. Read densities at genomic regions (TSSs) of interest were extracted by SeqMINER 1.3.3(Ye et al., 2011). Heatmaps were generated directly in the software from matrixes of binned read densities for all considered individual items.
For determination of fold changes profiles, the number of reads per bin for a given sample were divided by the number of reads in the indicated control sample and expressed as Log2 FC.
Promoter Escape Indexes
As described before(Chen et al., 2015), promoter escape indexes were calculated by taking the average coverage in rpm in the gene body (ranged from 101 bp to 2 kb downstream of TSS or 101 bp downstream of TSS to TTS for genes larger or smaller than 2 kb, respectively) divided by the average coverage on the promoter-proximal region.
RPB1 Peak Calling
Peaks were called against a rat IgG control using MACS v1.4.2(Zhang et al., 2008). All the peaks were listed in Table S7.
ChIRP-seq/ChIRP-MS
ChIRP was performed using biotinylated probes against ZFAS1 according to previously described methods(Chu et al., 2011). See Table S11 for probe sequences. Independent even and odd probe pools were used to ensure ZFAS1-specific retrieval. High-throughput sequencing libraries were constructed from ChIRPed DNA according to ChIP-seq protocol as described above, and sequenced on Illumina NovaSeq 6000 system with read length of 150bp. Raw reads were uniquely mapped to reference genome (hg38) using Bowtie(Langmead and Salzberg, 2012).
ZFAS1 peak calling, motif finding, and GO term analysis were conducted as described previously(Chu et al., 2011). The sequence data were deposited in the NCBI Gene Expression Omnibus (GEO), with BioProject accession number, GSE239617.
For ChIRP-MS, protein elution, extraction protein sample preparation for MS were conducted as described before(Chu et al., 2015). See Table S11 for ZFAS1 ChIRP probe design.
Behavioral studies
All behavioral studies were conducted during the light period. Mice were habituated to the procedure room for 1 hr before each test. In order to recover, mice were given three days between tests. Between subjects, all behavior apparatuses were sanitized with 70% ethanol solution and dried prior to introducing the next subject. All behavioral tests were performed with 8-week-old mice. Data are presented as means ± SD.
Open field test
Exploratory behavior in a novel environment was assessed by a 30 min session in an open field chamber (45cmL x 45cmW x 30cmH). Mice were placed in the middle of the central area and then recording was started. Locomotor activity (distance travelled and velocity) in the center/periphery of the arena were recorded by using a video camera and analyzed using the EthoVisione XT software (Noldus).
Morris water maze test
Mice were placed in a circular pool (120 cm in diameter and 50 cm height) containing opacified water. On the first day of training, before the first trial, mice were placed on the platform for 30 sec, followed by a 30-sec practice of swimming and three practice climbs onto the platform. The mouse was then placed into the water facing the wall of the pool and allowed to search for the platform. The trial ended when the mice climbed onto the platform or when a maximum of 60 sec elapsed. At the end of each trial, the mouse was allowed to rest on the platform for 60 sec. Then mice were immediately placed into the water again and released to swim and find the platform from a different start location. Four consecutive trials were administered for each animal. On day 5, the evacuation platform was removed and probing was performed.
Isolation of primary mouse lung fibroblasts
Newborn mice were anesthetized with aether, the whole lung was removed. Approximately 1 cm2 fragment of the tissue was excised and placed immediately in sterile PBS to avoid drying. The tissue was cut into 1 mm pieces, transferred into a sterile 30 ml beaker containing 10 ml of DMEM/F12, 0.14 Wunsch units/ml liberase blendzyme 3 and 1 x antibiotic/antimycotic, incubated at 37°C for 30–90 minutes with stirring. After digestion, the tissue fragments were rinsed 3 times with 10 ml of warm DMEM/F12 media containing 15% FBS and 1 x antibiotic/antimycotic, and then centrifuged at 524 x g. The pellet was resuspended in 10 ml of warm DMEM/F12 media with 15% FBS, 1 x antibiotic/antimycotic. The tissue pieces were broken down by pipetting with maximum force. Another 30 ml of DMEM/F12 media with 15% FBS, 1 x antibiotic/antimycotic was added and the solution was centrifuged twice at 524 x g to ensure removal of Liberase. The cell pellets were resuspended in 10 ml of DMEM/F12 media with 15% FBS, 1 x antibiotic/antimycotic and transferred to a 10 cm tissue culture dish and placed in a tissue culture incubator at 37°C, 5% CO2.
Immunohistochemistry
Mice were anesthetized with aether, and subjected to cardiac perfusion with saline, followed by a 10% formalin flush. Tissues (kidney or brain) were removed and sectioned into 3 mm slices before transfer into formalin. Tissues were fixed in 10% formalin for a minimum of 48 h at room temperature and then subjected to paraffin embedding schedule as follow: 70% Ethanol, two changes,1 h each; 80% Ethanol, one change, 1 h; 95% Ethanol, one change, 1 h; 100% Ethanol, three changes, 1.5 h each; Xylene, three changes, 1.5 h each; Paraffin wax (58°C-60°C), two changes, 2 h each. After the paraffin wax cooled down and solidified, the paraffin blocks were trimmed and cut at 5 mm and placed in water bath at about 40°C-45°C. Sections were mounted into slides and air-dried for 30 minutes, then baked in 45°C oven overnight. Before deparaffinization, slides were baked in 65°C oven for 2 h. Slides were placed in a rack and subjected to deparaffinization and dehydration in the following washes: Xylene, two changes, 3 min each; Xylene 1:1 with 100% ethanol, one change, 3 min; 100% ethanol, two changes, 3 min each; 95% ethanol, one change, 3 min; 70% ethanol, one change, 3 min; 50% ethanol, one change, 3 min. Slides were kept in the tap water until ready to perform antigen retrieval. Slides were placed to a boil in antigen retrieval buffer (10 mM Sodium Citrate, 0.05% Tween20, pH 6.0), then maintained at a sub-boiling temperature for 10 min. Slides were cooled down in running tap water for 5 min. For H&E staining, slides were stained in hematoxylin for 3–5 min before antigen retrieval, and then washed in running tap water until sections blue for 5 min or less. Slides were differentiated in 1% acid alcohol (1% HCl in 70% alcohol) for 5 min and then washed in running tap water until the sections were again blue by dipping in an alkaline solution (ammonia water) followed by tap water wash. Slides were stained in 1% Eosin for 10 min and washed in tap water for 1–5 min. Slides were dehydrated through 95% alcohol, 2 changes of absolute alcohol, 5 min each, and cleared in 2 changes of Xylene, 5 min each. Finally, slides were mounted in mounting media. For cell-specific immunohistochemical staining, slides were blocked in blocking solution (1 X PBS-T containing 10% FBS, 1% BSA and 0.3% Triton X-100) for 1 h at room temperature after antigen retrieval. Slides were incubated in primary antibodies (diluted in 1 X PBS-T containing 1% BSA) overnight at 4°C. Slides were washed three times in PBS-T and incubated in fluorophore-conjugated secondary antibody diluted in PBS-T, 1% BSA for 1 h at room temperature. Slides were washed three times in PBS-T and mounted with mounting medium with DAPI.
Quantification and statistical analysis
A two-tailed Student’s t-Test was used for analysis of statistical significance, with a P < 0.05 considered significant. For mouse studies, we determined that a sample size of at least 3–6 animals per genotype per sex was required.