Molecular cloning
All clonings were performed using standard molecular techniques. Fragments for cloning were generated by PCR using Platinum SuperFi II Master Mix (Thermo Fisher Scientific) and appropriate oligonucleotides (IDT DNA), by plasmid digests using standard restriction enzymes (NEB), or synthesized as Gene Fragments (TWIST Biosciences or IDT DNA). Fragment assemblies were performed using the NEBuilder HiFi DNA Assembly Mix (NEB) or Instant Sticky-end Ligase Master Mix (NEB). Assembled fragments were transformed into self-made chemically competent E. coli DH5α cells. Correct clones were identified by plasmid preparation (Monarch Plasmid Miniprep Kit, NEB) and Sanger Sequencing (Azenta) or RCA directly on cells (Microsynth). Subsequently, plasmids were isolated using the Plasmid Maxiprep Kit (QIAGEN) and used for transfection. All cloned sequences of this study are listed in Supplementary Table X.
Plasmid transfection
One day prior transfection, cells were seeded at 3.0x104 cells per well for 96 well plates, 2.2x105 for 24 well plates, 7.5x105 for 6 well plates, and 4.0x106 for 10 cm dishes. Cells were transfected with JetOptimus DNA transfection reagent (Polyplus transfection). For 96 well 75 ng DNA per well were transfected, for 24 well plates 300 ng DNA, for 6 well plates 1 µg, and for 10 cm dishes 5 μg DNA.
Cell culture and cell lines
HEK293T cells were cultivated at 37°C, 5% CO2 in an H2O-saturated atmosphere, and maintained in DMEM (Gibco) supplemented with 10% FBS (Gibco) and 1% penicillin-streptomycin (Gibco). HEK293T Split-Luciferase reporter cell line was generated by Cas9 cleavage at the AAVS1 locus and homology-directed integration of a donor construct containing LgBiT, the C-terminal fragment of Firefly Luciferase, separated by a P2A sequence, and the puromycin resistance gene. Three days after transfection, the cells were selected for two weeks with 2 µg/ml puromycin (Thermo Fisher Scientific). HEK293T cells stably expressing EGFR or IL7Rα were generated by means of amplifying the EGFR sequence from AddGene plasmid #23935 (a gift from William Hahn & David Root), whereas IL7Rα was synthesized (TWIST Biosciences). Both coding sequences were cloned into the AAVS1 knock-in donor plasmid, transfected with AAVS1 targeting Cas9, and selected with 2 µg/ml puromycin. Dual-positive EGFR and IL7Rα receptor cells were generated by cloning IL7Rα into an AAVS1 donor plasmid containing a blasticidin resistance gene, and cells were transfected and selected in 10 µg/ml blasticidin medium (Thermo Fisher Scientific).
Quantification of STV-mediated target RNA release into the cell culture supernatant
Supernatants from the STV-releasing cells were collected and filtered through 0.45 µm PVDF filters (Merck Millipore) after 48 h. RNA was extracted with Monarch Total RNA Miniprep Kit (NEB), and isolated RNA was used as template for RT-qPCR with Luna Universal One-Step RT-qPCR Kit (NEB), along with a primer/FAM-probe set (custom design, Metabion), specific for EGFP mRNA. The reaction was analyzed on a QuantStudio 7 Flex device (Thermo Fisher Scientific).
Integration of diffusion-designed symmetric oligomers into STV design
Previously designed RFdiffusion symmetric oligomers were filtered for successfully assembled oligomers based on size exclusion data10. Additionally, all D2 symmetric oligomers were excluded. The resulting 39 sequences were synthesized (eBlocks, IDT DNA) and cloned as a C-terminal fusion to the additional STV components (PHPLC, SynL, and tdPCP).
Integrating structure-mined membrane binding domains into STV design
The pleckstrin homology domain PDB: 1MAI was used as input structure for structure-based homology search with FoldSeek23,37. 10 sequences from three categories (other species, human, metagenome) were selected based on the highest homology to the input structure. Each sequence was synthesized (IDT DNA) and fused to the N-terminus of the previously identified ideal STV construct, containing SynL, tdPCP, and HE0690.
Sequence and structural alignments of the structure-mined membrane binding domains
The amino acid sequences of the structure-mined MBDs were aligned to that of Rattus norvegicus PHPLCδ, using the residues visible in the X-ray structure (PDB: 1MAI). The alignment was performed with the MAFFT version 7 --add tool38, using default settings (strategy: auto, scoring matrix: BLOSUM62, gap opening penalty = 1.53, offset value = 0.0). For the structural alignment, the structure of the MBD region was extracted from the AlphaFold239 (human and other species MBDs) or ESMFold24 (metagenomic MBDs) prediction of the respective structure-mined proteins containing these MBDs. These structures were aligned, and the RMSD to PDB: 1MAI was calculated using the PyMOL super alignment tool.
Screening of symmetric oligomer and membrane binding proteins for RNA release and uptake
Cells were seeded in 96 well format and transfected with each of the oligomer or membrane STV constructs along with plasmids coding for VSV-G and N-SplitLuc-PP7 (in a 2:1:7 ratio). 24 h post-transfection, 5 µl of supernatant was collected from the transfected cells, mixed with 45 µl PBS, and measured using Nano-Glo HiBiT Lytic Detection System (Promega) at a Centro LB960 device (Berthold Laboratories), using 0.5 sec integration time. 48 h post-transfection, 120 µl of supernatant was collected and filtered through a 0.45 µm PVDF 96 well filter plate (Sigma Aldrich) by centrifugation (1,500 g, 4°C, 20 min). Cleared supernatant was added to a seeded 96 well plate of C-split luciferase reporter cells. 24 h later Nano-Glo Dual-Luciferase Reporter Assay (Promega) was performed on the cells after complete removal of the supernatant. STV uptake was quantified by light emission from the NanoLuc substrate. N-splitLuc-PP7 mRNA uptake and expression were measured by the light emission from Firefly Luciferase substrate.
Validation of STV-mediated transfer of EGFP mRNA by Flow Cytometry
HEK293T producer cells were transfected in 24 well format with plasmids coding for STV constructs, VSV-G, and EGFP-PP7 (2:1:7 ratio). STV-containing supernatant was collected for two consecutive days, filtered through a 0.45 μm PVDF membrane filter, and concentrated 5-10 fold with Lenti-X Concentrator (Takara Bio) in fresh DMEM. 10-20 µl of resuspended STVs were added to a 96 well plate of HEK293T cells. After 24 h, the treated cells were detached using StemPro Accutase (Thermo Fisher Scientific), mixed with FACS buffer (EDTA/BSA), and filtered through cell strainer-containing tubes. Subsequently, samples were gated for living, single cells and EGFP mRNA uptake and expression were analyzed by Flow Cytometry (BD FACSaria III, BD Biosciences). Data analysis was performed using the FlowJo software (BD Biosciences).
Design of additional oligomers with C8 symmetry
Additional oligomers featuring C8 symmetry were generated using the open-source version of RFdiffusion, along with the script provided for symmetric oligomers10. These computations were performed on a single A100 GPU.
Determination of subcellular STV localization
HEK293T cells were transfected with STV constructs containing different membrane-binding domains. 24 h later, cells were fixed with 10% Formalin (Sigma Aldrich) and permeabilized in 1% BSA/0.5% Triton X-100 containing PBS. Permeabilized cells were incubated with primary anti-HA antibody (Sigma Aldrich, cat. H3663), overnight at 4°C. Subsequently, the cells were washed and stained with an Alexa 488 coupled, secondary donkey anti-mouse antibody (Thermo Fisher Scientific, cat. A21202), overnight at 4°C. Stained cells were mounted with ProLong Diamond reagent (Thermo Fisher Scientific) and imaged at an Axio Imager M2 fluorescence microscope (Carl Zeiss).
Characterization of packaging capacity by Flow Cytometry
EGFP-PP7-STVs were produced in 24 well format as previously described. Additionally, producer cells were transfected with mRuby3-PP7 constructs containing random UTR sequences of variable lengths. Concentrated STVs were added to HEK293T target cells. After 24 h, EGFP and mRuby3 expression was quantified using Flow Cytometry, as described previously.
Concentration of STVs by ultracentrifugation for analytical and experimental purposes
Producer cells, seeded in Poly-L-Lysin (Sigma Aldrich) coated 10 cm dishes, were transfected with plasmids coding for STV-C8 components required for the respective experiment. If not specified otherwise, supernatants were collected for three consecutive days and stored until day 3 at 4°C. The collected supernatant was centrifuged for 5 min at 1,000 g and passed through a 0.45 μm PVDF membrane filter. Filtered supernatant was added to a cushion of 20% (w/v) sucrose (Sigma Aldrich) in PBS. Subsequent ultracentrifugation was performed at 26,000 rpm for 2 h and 4°C using a SW28 rotor in an Optima L-60 ultracentrifuge (Beckman Coulter). After the centrifugation, the supernatant and the sucrose solution were removed, and the pellet was resuspended in 50 µl ice-cold 1x PBS (Thermo Fisher Scientific) on an orbital shaker at 150 rpm for 45 min at 4°C. To remove debris, the resuspended pellet was centrifuged at 1,000 g for 5 min at 4°C and stored at -80°C. Following this process, samples were concentrated approximately 300-fold.
Determination of STV purity for downstream analysis
STV-C8 samples were concentrated via ultracentrifugation, and the sample purity was determined by silver staining. Samples were prepared in 2x Laemmli buffer (Sigma Aldrich) for 10 min at 98°C. The SDS-PAGE was run on a 4-15% gradient TGX gel (BioRad) using a 1x Tris/Glycine/SDS running buffer (BioRad) for 60 min at 130 V. Subsequently, the gel was silver stained according to the manufacturer’s description (Serva). A gel was run in parallel with the same samples and blotted onto a nitrocellulose membrane for 60 min, 100 V at 4°C in transfer buffer (Tris/Gylcine-buffer, BioRad). The STV-C8 protein position on the membrane was determined by imaging with Nano-Glo HibiT Blotting system (Promega) in a Fusion SL Vilber machine (Peqlab). The HibiT signal on the membrane was used as a reference to identify STV proteins on the corresponding silver-stained gel.
Visualization of STV-C8 assemblies by cryo-ET
In 10 cm dishes coated with Poly-L-Lysin (Sigma Aldrich), seeded producer cells were transfected with plasmids coding for STV-C8 and EGFP-PP7. 24 h after transfection, the cells were washed with PBS, and serum-free DMEM was added to the cells. After another 24 h, the supernatant was collected and concentrated via ultracentrifugation as described previously. For subsequent cryo-ET analysis, the purified STV-C8 vesicles were diluted to 109 particles/μl in PBS. The samples were applied to holey R 3.5/1 carbon 200 mesh copper grids (Quantifoil), covered with a homemade 3 nm thick continuous carbon film by flotation. The grids were treated by glow discharge (at 4 mA for 10 s), then blotted and cryo-cooled into liquid ethane using a Vitrobot IV (Thermo Fisher Scientific) with the chamber operating at 95% humidity and at 10°C. For each tomogram, tilt-series were automatically acquired using Tomo5 software on a Krios G4 equipped with a cold-FEG operated at 300 kV and equipped with a Falcon IVi camera and a Selectris X energy filter (Thermo Fisher Scientific). A magnification of 81,000x was applied at a pixel size of 1.63 Å. Each tilt was acquired in the EER format and fractionated into 270 frames for a total dose of 2 e/Ų. Each tomogram was composed of 61 tilts, acquired according to a dose symmetric scheme with angles between 60° and -60° and a 2° increment between tilts. Processing was performed using Relion 5 beta3 software40. MotionCor2 was used to apply the gain reference and to align the EER fractionation by groups of 45 frames41. CTF estimation was performed using CTFFIND 4.1, tilt-series alignment was performed using AreTomo242,43. The tomogram was reconstructed using Relion 5 beta3, and denoising was performed using CryoCARE in Relion 5 beta3. Data segmentation was performed with MemBrain-seg for membranes44 and manually in Amira for C8 particles.
Characterization of STV-C8 RNA content
STV-C8 particles were produced and purified as described in the previous section. RNA was isolated from the particles, as well as from corresponding producer cells, using the Monarch Total RNA Miniprep Kit (NEB). Subsequently, Illumina RNA-Seq Library Prep and sequencing with 20 million paired-end reads per sample were performed on a NovaSeq device. Sequencing reads were mapped to the human reference transcriptome using the STAR aligner and differential expression analysis was performed using DESeq2. Library preparation, sequencing, and data analysis were performed by Azenta (Leipzig).
Characterization of STV-C8 protein content
STV-C8 were produced and purified as described in the previous section. Total protein was extracted by lysing the sample with lysis buffer (Preomics, Martinsried) supplemented with cOmplete Protease Inhibitor (Roche). The released protein was quantified using a BCA assay (Thermo Fisher Scientific Scientific). 10 µg of protein per sample was further processed by Filter Aided Sample Preparation (FASP45) and subsequently measured on a QExactive HFx mass spectrometer online coupled to a Ultimate 3000 RSLC (Thermo Fisher Scientific). Data analysis was performed by label-free quantificaton in MaxQuant 2.4.9.0 (MPI, Martinsried46) using a merged database of SwissProt human protein database and the sequences of exogenously expressed proteins. Statistical analysis was performed in Perseus (MPI, Martinsried47).
RNA and protein Gene Set Enrichment Analysis
Significantly enriched or depleted genes (p adjusted < 0.005, log2FC +3/-3) or proteins (-log q < 0.05, log2FC +3/-3) were selected, and a gene set enrichment analysis was performed using gProfiler2 with default options (e111_eg58_p18_30541362).
Benchmarking of EGFP mRNA delivery efficiency of STVs compared to SEND, EPN, and VLP
SEND/MmPeg10 was ordered from Addgene (#174858, a gift from Feng Zhang), and EPN-MCP, VLP-MCP, EGFP-MS2, SEND-EGFP constructs were ordered as synthesis (Twist Bioscience) and cloned into a CAG promoter expression backbone. For each system, the corresponding capsid scaffold and cargo RNA plasmids were co-transfected with VSV-G plasmid in 24 well format. Supernatants were produced for 48 h and concentrated as previously described. Concentrated vehicles were added to a 96 well plate of HEK293T, Vero E6, N2a, and HepG2 cells. 24 h later EGFP expression was analyzed by Flow Cytometry as described earlier.
Benchmarking of STV against LNP characteristics
A plasmid encoding EGFP under the control of the T7 promoter was cloned. The plasmid was linearized via digesting downstream of the stop codon, leaving a 3’UTR of similar length as in the STV cargo plasmid. The reaction was purified (Monarch DNA Cleanup Kit, NEB) and used as template for in vitro transcription (HiScribe T7 Quick High Yield RNA Synthesis Kit, NEB). Subsequently, the RNA was purified (Monarch RNA Cleanup Kit, NEB) and capped with the Vaccinia Capping System (NEB). The reaction was purified again and polyadenylated with E. coli Poly(A) Polymerase (NEB). After a final purification step, the EGFP coding mRNA was diluted to 150 ng/μl in 20 mM citrate buffer (pH 4.0). LNPs were composed of ALC-0315 (Cayman Chemical, cat. 34337), DOPE (Avanti Polar Lipids, cat. 850725), Cholesterol (ChemCruz, cat. sc-202539), and DMG-PEG 2000 (Avanti Polar Lipids, 880151) in the respective ratio (50:10:38.5:1.5). The lipid and RNA solutions were quickly mixed at a 1:3 volume ratio, resulting in a final weight ratio of 40:1. 1 μl of the prepared sample was diluted in 3 ml of PBS in a cuvette (Sarstedt) and analyzed by DLS using a Zetasizer Pro (Malvern Panalytical). EGFP mRNA-containing STV-C8 particles were prepared via ultracentrifugation, as described previously. The absolute STV-C8 protein content was determined by extrapolation from a HiBiT Control Protein (Promega) standard curve. EGFP-mRNA content in STV-C8 was determined by absolute RT-qPCR quantification (Luna Universal One-Step RT-qPCR Kit, NEB) with an in vitro transcribed EGFP mRNA standard and STV-C8 particle number was determined by DLS. Different concentrations of STV-C8(EGFP) and EGFP mRNA containing LNPs were added to HEK293T cells and EGFP expression was analyzed by Flow Cytometry. For the concentration of each vehicle, in which approx. 50% of cells turned EGFP positive, the required amount of EGFP mRNA for both vehicles was calculated and compared.
Analysis of STV-C8-induced interferon signaling
A549-IFN-GFP cells (a gift from Ralf Bartenschlager) that report interferon signaling by GFP expression were transfected with luciferase plasmid DNA as a positive control for interferon stimulation and treated with STV-C8 particles, containing a luciferase mRNA. GFP expression upon treatment was monitored after 24 h at an EVOS imaging device (Thermo Fisher Scientific).
Comparison of LNP and STV-C8-induced cytotoxicity
HEK293T cells were seeded in 96 well plate format and transfected with 50 ng EGFP mRNA containing LNPs and transduced with purified STV-C8 (EGFP) particles. Both particles were used at a concentration that induced EGFP expression in approx. 50% of cells. After 24 h, cells were detached with Trypsin 0.05% (Thermo Fisher Scientific), resuspended in Annexin V binding assay buffer (10 mM HEPES, 140 mM NaCl, and 2.5 mM CaCl2, pH 7.4), and labeled 1:100 with Annexin V-iFluor 680 (Abcam). Subsequently Annexin V staining intensity was quantified using Flow Cytometry.
Establishing cell-type specific STV-C8 by peptide binder engineering
Previouslydesigned EGFRn, EGFRc, and IL-7Rα minibinders30 were exposed on the STV-C8 surface by expressing them as a fusion construct, consisting of a signal peptide, minibinder sequences, and a transmembrane domain, along with STV-C8 components and an LDLR-binding deficient mutant of VSV-G (K63Q, R370Q29). Transfections were performed in 6 well plates with EGFP mRNA cargo, the supernatant was collected for 48 h and concentrated with LentiX concentrator (TakaraBio). 30 μl of concentrated supernatant was transferred to either WT-HEK293T cells or HEK293T cells stably expressing the EGFR or IL-7Rα receptors. 24 h later, the EGFP expression was analyzed by Flow Cytometry as described earlier.
STV-C8 mediated EGFP mRNA delivery into RPE spheroids
Human retinal organoids were differentiated from the hiPSCs-F49B7 cell line, derived from healthy donors, and tested for pluripotency markers as well as germ layer differentiation potential. hiPSC were seeded on 6 well plates coated with Matrigel (Corning) and cultured in mTeSR plus medium (STEMCELL Technologies). The medium was changed every two days. At 70% confluency, iPSCs were passaged in small clumps using 0.5 mM EDTA (0.5 M, pH 8.6, Thermo Fisher Scientific). On day 0, hiPSCs were dissociated as small aggregates using 0.5 mM EDTA. The aggregates were suspended in cold Matrigel (GFR, Corning) and incubated at 37°C for 20 min to allow gelling. hiPSCs/Matrigel aggregates were gently dispersed in the Neural Induction Medium (DMEM/F12+GlutaMax, 1% B27 with Vit A supplement, 0.5% N2 supplement, 0.1 mM 2-Mercaptoethanol, 2 mM GlutaMAX, and 1% penicillin/streptomycin, all from Thermo Fisher Scientific). The aggregates were cultivated in Ultra-Low Adherent 6 well culture plates (Costar, Corning). On day 5, the floating cysts were seeded on Matrigel-coated 6 well plates. On Day 15, the cysts were detached by adding Dispase (0.5 mg/mL in DMEM/F12, STEMCELL Technologies) for 3-4 mins at 37°C, followed by washing with DMEM/F12 medium and growing in the Retinal Differentiation Medium (DMEM/F12 +GlutaMax, 2% B27 without vitamin A, 1% NEAA, and 1% penicillin/streptomycin, all from Thermo Fisher Scientific). On Day 25, the immature retinal organoids were transferred to Retinal Maturation Medium (DMEM/F12 +GlutaMax, 8% FBS, 2% B27 without vitamin A, 1% NEAA, 1% A/A, all from Thermo Fisher Scientific, and 1% 100 mM taurine from Sigma-Aldrich). Half of the medium was changed every 2-3 days, and all organoids were cultured in a humidified incubator at 37°C and 5% CO2 until the end of the experiment. The retinal pigment epithelium (RPE) was developed during the retinal organoid generation as a patch attached to it. On day 200, RPE spheroids were dissected from human retinal organoids. Then, they were sorted into 96-well U Bottom Ultra-Low Attachment plate (Nucleon Sphera, Thermo Scientific). Each well consisted of 3-4 RPE spheroids. RPE spheroids were transduced with 10 µl of STV-C8(EGFP)/VSV-G or STV-C8(EGFP). The RPE spheroids were fixed two days after the treatment and then gradually dehydrated in 10% sucrose at RT, 30% at RT, and 50% overnight at 4°C. The spheroids were embedded in O.C.T (Tissue-Tek O.C.T. compound, Sakura) and immediately frozen at -80°C until solidification. RPE spheroids were sectioned into 10 µm thickness using a cryostat (Leica CM3050 S, Leica Biosystems). Cryosections were rehydrated and incubated in a 5% chemo-blocker solution (Merck) for 30 min, followed by 30 min incubation in 0.3% triton-X. Anti-RPE65 (Proteintech, cat: 17939-1-AP) and anti-GFP (Santa Cruz, cat. sc-101536) primary antibodies were diluted in 5% chemo-blocking solution and incubated overnight at 4°C. The cells were washed three times in PBS. Goat anti-rat Alexa Fluor 488 (Thermo Fisher Scientific) and donkey anti-rabbit Alexa Fluor 555 (Thermo Fisher Scientific) secondary antibodies were diluted in 5% chemo-blocking solution and incubated for 1 h at room temperature. Finally, the sections were washed with PBS and mounted using Fluoroshield with DAPI (Sigma Aldrich). Immunolabeled RPE spheroids were imaged using a Leica TCS SP8 spectral confocal laser scanning microscope (Leica Microsystems).
EGFP delivery into human monocytes
Primary human monocytes (ATCC, cat. CRL-3622) were seeded in 96 well format. 5 µl of concentrated EGFP mRNA containing STVs were added to the cells, and EGFP expression was analyzed by Flow Cytometry 24 h later.
Isolation of primary astroglia from mouse postnatal cortex and Ascl1 mRNA delivery
Primary astrocytes were isolated from the cerebral cortex of postnatal day 5 C57BL/6N mice. The cortex was isolated, cut into small pieces, and mechanically dissociated by vigorous pipetting. Subsequently, the cell suspension was centrifuged for 7 min at 1,300 rpm, the cell pellet was plated in a T25 flask and cultivated for 7-13 days in DMEM/F-12 GlutaMAX, supplemented with 10% FBS, 10% penicillin/streptomycin, 5% horse serum, 4.5% D-(+)-glucose, 2% B27, 10 ng/ml bFGF, 10 ng/ml EGF (all from Thermo Fisher Scientific). Upon reaching 90% confluency, the cells were passaged using 0.05% Trypsin/EDTA (Thermo Fisher Scientific) and approx. 75.000 cells were seeded onto Poly-D-Lysin (Sigma Aldrich) coated glass coverslips. 24 h later, 15 µl of concentrated EGFP or Ascl1-P2A-EGFP containing STV-C8 was added to the cells. After 48 h, cells were fixed in 10% formalin (Sigma Aldrich) and incubated with anti-GFP (Abcam, cat. ab13970) or anti-Mash1 (Abcam, ab211327) primary antibody in PBS containing 1% BSA (Sigma Aldrich) and 0.3% Triton X-100 (Sigma Aldrich) overnight at 4°C. After washing, the cells were stained with Alexa488 coupled donkey anti-chicken (Dianova, cat. 703-546-155) or Alexa594 coupled donkey anti-rabbit (Thermo Fisher Scientific, cat. A21207) secondary antibody for 1-2 h in darkness at room temperature. Subsequently, cells were DAPI stained, coverslips were mounted using Aqua Poly/Mount (Polyscience), and samples were imaged using an Axio Imager M2 fluorescence microscope (Carl Zeiss).
Exon 51 deletion of dystrophin gene in primary porcine fibroblasts
Two Cas9 sgRNA plasmids containing a PP7 motif in the stem-loop of the sgRNA, along with porcine dystrophin targeting spacers, were cloned. The sgRNAs target intron 50 (AGAGTTCCTAAGGTAGAGAG) and intron 51 (ATAAAGATAAGAGCTGGCAG) to delete exon 5115. Additionally, a plasmid coding for NLS and NES fused Cas9, along with a 3’UTR PP7 motif, was cloned. HEK293T producer cells were seeded in poly-Lysin coated 10 cm dishes and co-transfected with Cas9 mRNA and the two sgRNA plasmids (1:1:1 ratio), along with STV-C8 and VSV-G coding plasmids. STV-C8 particles were collected and concentrated via ultracentrifugation, as described before. Pig primary fibroblasts were seeded in a collagen-coated 48 well plate in DMEM medium48, supplemented with 1% NEAA, 10 mM HEPES, 15% FBS (all from Thermo Fisher Scientific) and 2-Mercaptoethanol (Merck). Seeded cells were treated with 20 µl STVs for 72 h. Subsequently, genomic DNA was extracted (Monarch Genomic DNA Purification Kit, NEB), and a 2 kb fragment covering the deleted region was amplified (Primer: CCCATGACATTTACCCTATTATTATCCC and GCTAATGTTCATTTTAAAAAGGAATCTGTC) using Platinum SuperFi II Master Mix (Thermo Fisher Scientific). The PCR product was run on a 1.5% agarose gel and imaged.
Treatment of SARS-CoV-2 infected iPSC-derived human lung cells with STV-delivered Cas13d-NCS
For lung cell differentiation, hiPSCs (ISFi001-A - RRID: CVCL_YT30) were cultured in StemMACS medium (Miltenyi Biotec) on plates coated with Geltrex Reduced Growth Factor (Thermo Fisher Scientific). 70% confluent iPSC colonies were isolated as single cell suspension with Accutase (Thermo Fisher Scientific), 5 min at 37°C, neutralized with StemMACS medium, centrifuged for 3 min at 200 g, room temperature, and 1.0–1.2×106 cells were seeded onto non-adherent 6-well plates (Corning, 3471) in StemMACS medium supplemented with 10 μM Y2763 (Enzo Life Sciences). Differentiation basal medium (DBM) was prepared with DMEM/F12 1:1 GlutaMAX (Thermo Fisher Scientific) supplemented with 1x NEAA (Thermo Fisher Scientific), 0.1% Albumax (Thermo Fisher Scientific), 1× B27 (Thermo Fisher Scientific). Formation of embryonic bodies (EB) was induced by changing the medium to 50% StemMACS medium /50% DBM with 20 ng/ml Activin A (Bio-Techne). Medium was replaced entirely to DBM with 20 ng/ml Activin for 48 h. Definitive endoderm (DE; Days 0 to 5) was induced by plating EBs onto Geltrex Reduced Growth Factor-coated plates at 7 EBs per cm2 of culture surface in DBM supplemented with 150 ng/ml Activin A and 25 ng/ml bone morphogenic protein 4 (BMP4) (Thermo Fisher Scientific) for 5 days with daily medium changes. Anteriorization of DE (Days 6 to 10) was elicited by changing DBM supplements to 50 ng/ml EGF (Invitrogen) and 50 ng/ml bFGF (Thermo Fisher Scientific), 3 μM SB431542 (Miltenyi Biotec) and 10 ng/ml Noggin (Sigma Aldrich) for 5 days with medium changes every day. Lung progenitors giving rise to alveolar epithelial cells type II (Days 10 to 17) were generated by changing the medium to DBM containing 50 ng/ml BMP2 (Thermo Fisher Scientific), 50 ng/ml FGF10 (Peprotech), 50 ng/ml BMP4, 50 ng/ml bFGF, and 50 ng/ml WNT3A (Bio-Techne) for 7 days. Successful differentiation into alveolar epithelial cells was confirmed by expression analysis of ACE2 and SLC34A2 by RT-qPCR (Luna Universal One-Step RT-qPCR, NEB). Additionally, NLS and NES containing Cas13d-NCS33 was cloned into a PP7 motif containing backbone in the 3’UTR, and a PP7 motif was attached 3’ to a crRNA, targeting the SARS-CoV-2 3’UTR region (GUCAUCCAAUUUGAUGGCACCUG). Subsequently, lung progenitor cells were seeded into Geltrex-coated 96-well plates at a density of 2×104 cells/well (Merck) and differentiated for 7 days in differentiation medium. Differentiated lung cells were transduced with 40 μl concentrated STV-C8, containing Cas13d-NCS/SARS-CoV-2 or non-target crRNA. 24 h later, the cells were infected with SARS-CoV-2-GFP (MOI10), and viral replication was monitored for 72 h in an Incucyte S3 live imaging system (Sartorius).
Analysis of STV-C8 inactivation in human blood samples
Peripheral blood mononuclear cells (PBMCs) were isolated by diluting blood 2-4 times the volume of PBS. 35 ml of the diluted blood suspension was carefully layered onto 15 ml of Ficoll (density = 1.077 g/mL) in a Falcon tube and centrifuged without brake at 400 g for 30 min at 20°C. After centrifugation, the upper layer was aspirated, leaving PBMCs at the interphase. The PBMC layer was transferred to a fresh Falcon tube, filled with PBS, and centrifuged again at 300 g for 10 min at 20°C. The resulting cell pellet was resuspended in PBS, and cell counting was performed using Trypan blue staining. For long-term storage, PBMCs were frozen at the density of 1x107 cells/ml in FBS supplemented with 20% DMSO. Blood samples were collected in EDTA-free tubes for the isolation of blood serum. The tubes were gently inverted several times to mix the blood and then allowed to clot at 4°C for 3-4 h. After clotting, the samples were centrifuged at 2500 g for 10 min at room temperature (RT). Using a sterile pipette, the top clear layer (serum) was carefully transferred to new sterile microcentrifuge tubes or storage vials. For long-term storage, aliquoted serum was stored at -80°C. STV-C8(N-Split-Luc) were produced in 24 well plates and collected for 48 h. The collected supernatant was concentrated, using LentiX (TakaraBio) and concentrated as described. 30 μl of concentrated STV-C8 particles were mixed with 30 μl of 1:10 diluted serum, 30 μl of resuspended PBMCs (approximately 3.0x105 cells) or PBS and incubated at 37°C for 60 min. After the incubation period, 50 μl of the STV-C8 with PBMCs or serum mixes were transferred to a 96 well plate of Split-luc reporter cells. The next day, N-Split-Luc RNA expression was analyzed using ONE-GloEX Luciferase (Promega) assay.
Testing of STV-C8 storage conditions
STV-C8(N-Split-Luc) were produced in 6 well format for 2 days, concentrated, using LentiX (TakaraBio), and stored for 7 days at 4°C or -80°C. Subsequently, 50 µl of stored samples were added to Split-Luc reporter cells. The next day, N-Split-Luc RNA expression was analyzed using ONE-GloEX Luciferase (Promega) assay.
Delivering of OpenCRISPR-1 with STV-C8
The coding sequence of OpenCRISPR-1 was ordered (Twist Bioscience) and cloned into a CAG promoter containing expression plasmid. The coding sequence was fused to two NLS and one NES signal, and the PP7 aptamer was added to the 3’UTR. Additionally, a sgRNA containing the PP7 aptamer in the stem-loop region and a spacer targeting the stop codon in eTLR cells (GCUCCCACAACGAAGACUGAC) was cloned49. STV-C8 particles, containing OpenCRISPR-1 or Cas9 and the sgRNA, were produced in 6 well format for 3 days, concentrated, using LentiX (TakaraBio), and 20 µl concentrated particles were added to a 96 well plate of eTLR cells. 3 days later, the cells were imaged at an EVOS imaging device (Thermo Fisher Scientific).
Analysis of mouse whole-body biodistribution of STV-C8 mediated EGFP expression
STV-C8(EGFP) particles were produced in coated 10 cm dishes for 3 days and concentrated by ultracentrifugation, as described before. 50 µl of concentrated STV-C8(EGFP) samples were injected intravenously into four-week-old female Balb/c WT mice. The mice were sacrificed 24 or 72 h after injection and intracardially perfused with heparinized PBS (10 U/ml heparin) and 4% paraformaldehyde (PFA). The skin was removed, and the bodies were fixed in 4% PFA overnight at 4°C. As previously described34, for vDISCO whole-body staining and clearing the following steps were performed: In brief decolorization (25% CUBIC reagent in PBS), decalcification (10% (wt/vol) EDTA in PBS), signal-enhancement with anti-GFP nanobodies (Chromotek, anti-GFP-AF647), dehydration (with tetrahydrofuran), delipidation (with dichloromethane) and refractive-index matching with a mixture of benzyl alcohol and benzyl benzoate (BABB). A Blaze light sheet system (LaVision BioTec) with an axial resolution of 4 µm was used for light sheet imaging. Full-scale mouse body imaging was performed using a 4x magnification objective (Olympus XFLUOR 4x corrected/0.28 NA [WD=10 mm]). High-magnification tile scans were obtained with 22% overlap, and the light-sheet width was reduced to 80%. For the z-step, the size was set to 6 µm, with a time exposure of 40 ms in the background channel (488 nm) and 60 ms in the signal channel (640 nm, 647-boosted GFP signal). A Fiji plugin was used to stitch the raw TIFF files to a full plane. The individual planes were merged into a 3D file format with Imaris converter and visualized by Imaris34.
In vivo treatment of porcine muscle cells to delete exon 51 from the dystrophin gene
Large animal work was approved and ethically monitored by the Bavarian local authority (ROB-55.2-2532.Vet_02-19-39). STV-C8(Cas9/sgRNAs) were produced in coated 10 cm dishes and concentrated by ultracentrifugation as described before. The pig was sedated by intramuscular injection of ketamine and azaperone. For analgesia, fentanyl was applied intravenously. Subsequently, the injection site was shaved and disinfected, and 1 ml of concentrated STV-C8 sample was injected at 1.75 cm depth, using a 22G safety needle, into the right hind leg (M. biceps femoris). The animal was clinically monitored post-injection. After 3 days, the animal was sedated and euthanized by i.v. injection of pentobarbital. Several muscle samples around the injection site, as well as samples from the uninjected back (latissimus dorsi), were prepared, and genomic DNA (Monarch Genomic DNA Purification Kit, NEB) was extracted. A PCR, using Platinum SuperFi II Master Mix (Thermo Fisher Scientific), was performed (Primer: CCCATGACATTTACCCTATTATTATCCC and GCTAATGTTCATTTTAAAAAGGAATCTGTC) to assess the deletion efficiency on an agarose gel by comparing band intensities. The resulting bands at 2 kb (wild type) and 1 kb (genomic deletion) were extracted from the gel and verified by Sanger sequencing (Microsynth). Additionally, the PCR product was sequenced using Oxford Nanopore sequencing (Eurofins Genomics), and the deletion frequency was analyzed using Geneious Prime (Dotmatics).
Statistical Analysis
Statistical tests and graphical representations of the numerical data were performed using GraphPad Prism.