Ethics statement
All analyses performed in this manuscript were carried out in accordance with the Declaration of Helsinki and were approved by the Ethics Committee of the National Center for Child Health and Development, Tokyo, Japan (#518, genetic analysis; #686, experiments using human iPS cells). Written informed consent from the patients or their parents was obtained from all members. All animal protocols were approved by the Animal Care and Use Committee of the National Research Institute for Child Health and Development, Tokyo, Japan. Generation and all experiments of knockout mice were conducted in accordance with the approved animal protocols (#A2015-005-C03).
Subjects
Seven patients of the family visited our hospital and were interviewed about the eye and systemic symptoms of family members. Seven patients underwent comprehensive eye examinations, including slit lamp examination, ocular tonometry, fundus examination and visual acuity testing.Swept source optical coherence tomography was measured using DRI OCT Triton (TOPCON, Japan), and electroretinogram was measured using RETeval (Welch Allen, LKC Technologies, USA) in accordance with International Society of Clinical Electrophysiology of Vision (ISCEV) protocol. For genomic analysis of the seven patients, DNA samples were obtained from peripheral leukocytes. In addition, DNA samples were obtained from peripheral leukocytes or saliva of two unaffected family members (IV-1, and IV-6). Of these, three patients (IV-2, IV-3, and IV-4) were subjected to systematic molecular analyses using array-based CGH and NGS. The three patients (IV-2, IV-3, and IV-4), the remaining four patients (II-1, III-1, III-2, and IV-5), and two unaffected individuals (IV-1 and IV-6) underwent Sanger sequencing for the SNVs detected in IV-2, IV-3, and IV-4.
Molecular genetic analyses of MOMS
CNVs in the genome were analyzed by CGH using a catalog human array (4 × 180 k format; Agilent Technologies, Palo Alto, CA, USA). The Database of Genomic Variants39 was used to exclude benign CNVs. SNVs were screened by NGS using the SureSelect All Exome v5 kit (Agilent Technologies). The library was sequenced as 100-bp paired-end reads using a HiSeq 2000 sequencer (Illumina, San Diego, CA, USA). Sequence data were analyzed by using Avadis NGS 1.3.1 (DNA Chip Research, Yokohama, Japan) and SAMtools 0.1.17 software40. The population frequency of identified SNVs was assessed using dbSNP41, the 1000 Genomes Browser, and the Human Genetic Variation Browser42. Functional consequences of SNVs were predicted by using Polyphen229, SIFT, and MutationTaster230. SNVs with low signal quality scores were excluded from further analyses. Since MOMS appears to be an autosomal dominant disease with a relatively high penetrance, we searched for SNVs meeting the following criteria: (i) those present in a heterozygous state in all the three subjects examined; (ii) those accounting for less than 0.1% of the general population; and (iii) those absent in the control samples obtained from eight unaffected Japanese individuals. Then, we selected SNVs that affect protein sequence or a splice site. Tissue expression patterns of the affected genes were examined using the GTEx portal. The probability of being loss-of-function intolerant was assessed by using the data reported by Lek et al31. The results of NGS were validated by Sanger sequencing. Furthermore, we performed Sanger sequencing for seven individuals of this family, three affected individuals and four unaffected individuals, to examine whether the detected SNV co-segregated with MOMS.
Generation of E130309D02Rik exon 2 and exon 6 knockout mice
Plasmids containing human codon-optimized Cas9 (hCas9) and single-guide RNA (sgRNA) cloning vector were gifted from George Church (Addgene plasmid #41815 and #41824, respectively)43. A plasmid containing inactive Cas9 (dCas9) fused with the FokI DNA nuclease domain (FokI-dCas9; fCas9) was a gift from David Liu (Addgene plasmid #42970). Specific sgRNA sequences were designed using CRISPRdirect44. sgRNAs for exon 6 and exon 2 of INTS15, hCas9 mRNA, and FokI-dCas9 mRNA were prepared as previously described45, 46. Three sgRNA target sequences were cloned into sgRNA cloning vectors by inverse PCR using primers pairs INTS15 exon 6_sg1-F / INTS15 exon 6_sg1-R, INTS15 exon 6_sg2-F / INTS15exon 6_sg2-R, and INTS15exon 2_sg-F / INTS15 exon 2_sg-R (Supplementary Data Set 2). In vitro transcription was performed using the mMESSAGE mMACHINE T7 Transcription Kit (Ambion, Austin, TX, USA). PCR products amplified using T7-added primers (T7-INTS15 exon 6_sg1 for INTS15 exon 6_sg1, T7- INTS15 exon 6_sg2 for INTS15 exon 6_sg2, and T7- INTS15 exon 2_sg for INTS15 exon 2_sg; Supplementary Data Set 2) were used as templates. Transcribed RNAs were purified using the MEGAclear RNA Kit (Ambion).
Superovulated F1 hybrid of C57BL/6 × DBA/2 (BDF1) female mice were crossed with BDF1 male mice. The fertilized mouse zygotes were collected in M2 medium. Microinjection was carried out as previously described47. Essentially, RNAs and ssODNs were mixed just before microinjection into the cytoplasm or pro-nuclei of zygotes, and the injected embryos were incubated at 37ºC until they were transferred into pseudopregnant females at the two-cell stage. The concentration of injected RNAs was always kept at 500 ng/ml in total. For the sgRNA/hCas9 condition, sgRNA and hCas9 mRNA were mixed at a 1:1 ratio, and thus the final concentration was 250 ng/ml each, and for the two sgRNAs/FokI-dCas9 condition, the sgRNAs and FokI-dCas9 mRNA were mixed at a 1:1:1 ratio, and thus the final concentration was 167 ng/ml each. The final concentration of injected ssODNs was 100 ng/ml. All mice were obtained from Sankyo Labo Service (Tokyo, Japan).
Genomic DNA was extracted from the fingers of day 4 to 7 neonates using Kaneka Easy DNA Extraction Kit version 2 (Kaneka #KN-T 110005). For direct sequence analysis, the genomic regions around gRNA target sites were PCR amplified using the primers shown in Supplementary information Dataset S7. The obtained PCR products were treated by ExoSAP-IT Express PCR Product Cleanup Regent (Thermo Fisher Scientific #75001) and sequenced directly using the BigDye Terminator v3.1 Cycle Sequencing Kit (Thermo Fisher Scientific #4337454). To confirm the sequence of the mutated alleles, PCR products were also analyzed by 2% (for Exon2F / Exon2R products) or 2.5% (for Exon6F / Exon6R products) agarose gel electrophoresis and were cloned using the TOPO TA Cloning Kit (Thermo Fischer Scientific #K4575J10). After PCR amplification of the insert using M13 forward and reverse primers, positive clones were subjected to sequence analysis using T7 forward and M13 reverse primers. To detect CRISPR/Cas9-mediated INTS15 exon 2 knockout, a 136-bp fragment including the gRNA target site was amplified from genomic DNA using the primer set INTS15 exon 2 forward and INTS15 exon 2 reverse indicated in Extended Data Tab. 1 and electrophoresed on Extra PAGE One Precast Gel (Nacalai Tesque #13076-14) to detect 7-bp deletion.
Mice at the age of 1 or 2 months were examined with slit-lamp biomicroscopy and ophthalmoscopy. When abnormalities were detected, mice were sacrificed by cervical dislocation, and organs including the eye, brain, heart, lung, intestine, and kidney were examined under a stereoscopic microscope.
Collection and genotyping of blastocysts
Four-week-old INTS15exon 2+/- female mice were superovulated with 5 IU of pregnant mare serum gonadotropin (Folligon; Intervet, Bendigo East, Australia) administered intraperitoneally, followed 48 hours later by 5 IU of human chorionic gonadotropin (Chorulon, Intervet), and were mated with INTS15exon 2+/- male mice of the same strain overnight. Mice were sacrificed by cervical dislocation, and preimplantation embryos were collected. Noon of the day when a vaginal plug was detected was designated embryonic day 0.5 (E0.5). E3.5 blastocysts were isolated by flushing uteri with M2 medium.Genotyping was performed with direct PCR. Individual blastocysts were separated on dishes containing M2 medium and transferred into tubes containing 1 μl of sterile water, to which 3 μl of lysis buffer (0.05% SDS and 0.035 N NaOH) was added. After incubation for 5 min at 95 °C, lysates were subjected to direct PCR in a total volume of 50 μl with LA Taq polymerase (TaKaRa Bio) using the primers described in Supplementary Data Set 7.
RNA in situ hybridization
Whole-mount in situ hybridization was performed as previously described48. A gene-specific fragment of E130309D02Rik was amplified from mouse chondrocyte cDNA by PCR using the following primers: forward, 5′-TTGAGACCTTCGACGTCAATGAG-3′ and reverse, 5′-TCTGGCCACGAGTGGCC-3′.
Paraffin section preparation, hematoxylin and eosin staining and immunohistochemistry
After sacrificing mice by cervical dislocation, eyes were enucleated, fixed in SUPER FIX (KURABO #KY-500), rinsed in water, dehydrated in a graded series of alcohol, and embedded in paraffin, as described previously11. Each block was serially sectioned at 3 μm in thickness. Deparaffinated sections were stained with hematoxylin and eosin. For the immunoenzyme method, avidin-biotin complex methods were performed using VECTASTAIN ABC kit (Vector laboratories #PK-6100) according to the instructions, and observed by using an IX71 inverted microscope (Olympus).
Immunofluorescence
Specimens were fixed with 4% paraformaldehyde (pH 7.0) for 20 min at room temperature and subjected to immunofluorescence staining as described previously35. The antibodies used in this study and dilution rates are listed in Supplementary Data Set 8.
Plasmids
Human INTS15 (NM_024067) ORF clone with C-terminal Myc-DKK tag (Origene #RC219786; pCMV C7orf26-Flag) and negative control GFP expressing clones (TaKaRa Bio #Z2470N; pAcGFP1-C1) were purchased. INTS15 shRNA clone tagged with GFP (GeneCopoeia #HSH018872-CU6; pU6-shINTS15-GFP) and negative control shRNA clone (GeneCopoeia #CSHCTR001-CU6; pU6- non-specific shRNA-GFP) were also purchased. These expression vectors were prepared using EndoFree Plasmid Maxi Kit (QIAGEN #12362).
Cell culture
HeLa cells were obtained from ATCC, and 293T cells was purchased from TaKaRa Bio. HeLa cells were grown in DMEM medium (Thermo Fisher Scientific #11965092) supplemented with 10% FBS and 1% Pen Strep, whereas 293T cells were cultured in DMEM medium with 10% FBS, 1×GlutaMAX Supplement (Thermo Fisher Scientific #35050061), and 1% Pen Strep. Neonatal normal human dermal fibroblasts NHDF (Lonza #CC-2509) was purchased and cultured using Fibroblast Growth Medium-2 (Lonza #CC-3132). Human iPSCs (HPS0007_409B2 cell passage 29) were obtained from RIKEN BioResource Center (Tsukuba, Japan) and were maintained on Laminin-511 E8-coated 6-well plates in StemFit AK02N (Ajinomoto, Tokyo, Japan) for feeder-free culture. For passaging, hiPSCs were detached by treatment with TrypLE Express (Thermo Fisher Scientific) at 37°C for 8 min using a cell scraper. After collection by centrifugation, cells were suspended in medium containing 10 µM Y-276 and seeded at 10,000 cells per well.
Knockdown of INTS15 by siRNA and shRNA
siRNA specific for INTS15 (ON-TARGETplus Human INTS15 (79034) siRNA SMARTpool, #L-016351-02) and negative control siRNA (ON-TARGETplus Non-targeting Pool, #D-001810-10; NS siRNA) were purchased from Dharmacon. Seeded in 24-well plates 2 days before transfection, HeLa cells at a cell confluency of 50–70% were transfected with 50 µL of Opti-MEM I medium containing 5 pmol of siRNA using Lipofectamine RNAiMAX Transfection Reagent (Thermo Fisher Scientific #13778150) or with 50 µL of Opti-MEM I medium containing 500 ng of pU6-shINTS15-GFP or pU6-non-specific shRNA-GFP using Lipofectamine 3000 Transfection Reagent (Thermo Fisher Scientific #L3000001) in accordance with the manufacturer’s instructions. Cells were harvested or fixed at different time points after transfection for subsequent analysis.
Apoptosis assay
HeLa cells were transfected with pU6-shINTS15-GFP or pU6-non-specific shRNA-GFP, cultured in 96-well plates or 8-well glass chambers, and fixed 36 h after transfection. After two washes with PBS containing 0.1% Tween 20, fixed cells were incubated with M30 CytoDEATH antibody solution (1:50) (Roche #12140322001) for 60 min at room temperature. Cells were washed twice and incubated with goat anti-mouse IgG (H+L) secondary antibody and Alexa Fluor Plus 555 solution (1:500) (Thermo Fisher Scientific #A32727) for 30 min at room temperature. After mounting with ProLongGold Antifade Reagent with DAPI (Thermo Fisher Scientific #A32727), M30-positive cells were observed with a DeltaVision Elite microscope (CORNES Technologies).
Immunoprecipitation and immunoblot analysis
293T cells at a confluency of 70–80% in 10 cm dishes were transfected with 1.5 mL of Opti-MEM I medium containing 15 µg of pINTS15-myc-flag using Lipofectamine 3000 Transfection Reagent (Thermo Fisher Scientific #L3000001). After 72 h culture, protein was extracted from the cells using CelLytic M Cell Lysis Reagent (Sigma #C2978) containing protease inhibitor cocktail (Roche #04693116001) in accordance with the manufacturer’s instructions. Immunoprecipitation was performed using EZview Red ANTI-FLAG M2 Affinity Gel (Sigma #F2426) and FLAG Immunoprecipitation Kit (Sigma #FLAGIPT1). Affinity gel was suspended with 1% BSA washing buffer and rotated for 30 min at room temperature. After four washes with washing buffer, the affinity gel was suspended with the cell lysate and rotated for 16 h at 4 oC. After four washes with washing buffer, Flag-tagged INTS15 was eluted from the affinity gel with 3×Flag peptide solution. After concentrating the volume using Amicon Ultra-0.5 (Millipore #UFC5003), protein amounts were determined using the Qubit Protein Assay (Thermo Fisher Scientific #Q33211). Approximately 5 μl of extracts and 1 μl of clarified eluates were boiled for 5 min at 95oC, separated on an 7.5% acrylamide SDS gel, and transferred to PVDF membranes (Bio-Rad #1620174). Membranes were blocked with Blocking One (Nacalai Tesque #03953), incubated for 20 min at room temperature and washed with TBST. The membranes were incubated with primary antibodies against INTS4 and INTS5 (Supplementary Data Set 8) at 4oC overnight, washed with TBST, and incubated for 1 h at room temperature with HRP-conjugated secondary antibodies (Supplementary Data Set 7)diluted in Blocking One. Immunoreactive bands were visualized with SuperSignal West Femto Chemiluminescent Substrate (Thermo Fisher Scientific #34095). For coimmunoprecipitation analysis of endogenous proteins, anti-INTS5 (Supplementary Data Set 8) was immobilized to protein A agarose beads and then incubated with nuclear extract prepared from HeLa cells. After extensive washing, bound material was eluted with Laemmli sample buffer for immunoblot analysis.
BN-PAGE
BN-PAGE was performed as described previously49. Nuclear fraction was extracted from adult mouse brain by NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Scientific #78833) according to the manufacturer's protocol. Approximately 20 µg solubilized proteins were then resolved on BN-PAGE (4–12%), which was followed by Western blot analysis. Molecular weights were determined using the NativeMark Unstained Protein Standard (Life Technologies).
RNA-seq analysis
Poly(A)+ mRNA fractions were isolated from HeLa/INTS15 siRNA cells and negative control HeLa/NS siRNA cells 48 or 72 h after transfection, and whole transcriptome analysis was performed by using HiSeq 2500 (Illumina) to obtain 20 million reads per sample. Cells were treated for 6 h before RNA extraction with 30 µg/mL cycloheximide (Sigma #C7698). Paired-end reads were mapped to the human genome GRCh38.p13. Differential splicing events between the two conditions were analyzed by using the software Multivariate Analysis of Transcript Splicing50. Heatmaps were created using log2 (FPKM) of each transcript. Transcripts were categorized based on GO terms such as brain development and eye development. Transcripts with FPKM of less than 1.0 throughout the samples were excluded from further analysis.
Splicing assays
To confirm intron retention, exon skipping, and alternative splicing events in the transcripts obtained from HeLa/INTS15 siRNA cells and HeLa/NS siRNA cells, primers sets (Supplementary Data Set 7) were designed so that they amplify splice variants in question simultaneously, and semi-quantitative RT-PCR products were analyzed on agarose gels.
Knockdown of INTS15 in iPS-derived retinal and brain tissues
Human cortical and retinal tissues were induced from human iPS cells as described previously35. iPS-derived embryoid bodies with optic vesicles, which are primordia of the brain and retina, were adhered to 24-well plates 22 days after the start of neuronal differentiation. After 48 h, culture medium was replaced with Accell siRNA delivery media (Dharmacon B-005000-100) containing 100 µM Accell Human siRNA (Dharmacon C7orf26, E-016351-00-0005; Non-targeting, D-001910-01) and then cultured for 3 days. Retinal and brain tissues were dissected from colonies for subsequent qRT-PCR analysis.
qRT-PCR analysis of INTS15 knockdown in HeLa cells
Total RNA was extracted from HeLa/INTS15 siRNA and HeLa/NS siRNA cells 48-72 h after transfection using RNeasy Mini Kit (QIAGEN #74136), and its concentration determined using a NanoDrop LITE (Thermo Fisher Scientific). Expression of mRNAs or snRNAs in each RNA sample was determined by two step qRT-PCR using StepOnePlus Real-Time PCR System (Applied Biosystems) as previously described35. CT values were normalized to the expression levels of HPRT1 for mRNA and 7SK for snRNA, and relative amounts of target RNAs were calculated using the 2–ΔΔCT method. The primers used in this study are listed in Supplementary Data Set 7.
Molecular phylogenetic tree
Amino acid sequences of INTS15 of various species were obtained from the Ensembl database. The phylogenetic tree was constructed using the neighbor-joining method and ClustalX software. The reliability of the tree was estimated using the bootstrap method and 10,000 replications.
Microarray analysis
Total RNA extracted from siRNA-treated HeLa cells (50 ng) was used to prepare Cy3-labeled target cRNA with the Low Input Quick Amp Labeling Kit (Agilent Technologies). The resultant cRNA (600 ng) was hybridized to Sureprint G3 Human Gene Expression Microarray 8 × 60 K v3 (Agilent Technologies), and the signal intensities were scanned according to the manufacturer’s protocol. The data were analyzed using GeneSpring GX software (Agilent Technologies). 75 percentile normalization was chosen as the normalized method, and only the probes that were “detected” in at least one sample in the control/target cell samples were used for further analysis. The number of probes used in the analysis was 27,865. The differentially expressed genes were selected using the criteria of |log2 (fold change)| ≥ 1. The functional annotation chart for downregulated genes was obtained using DAVID v6.551.
Statistics
Statistical analysis was performed using Microsoft Excel statistics software. All values are expressed as mean ± SD. Unpaired two-tailed t-tests were used to compare groups. P values less than 0.05 were considered statistically significant; individual P values are indicated by asterisks in graphs (*, P < 0.05).