JARB Journal of Animal Reproduction and Biotehnology

OPEN ACCESS pISSN: 2671-4639
eISSN: 2671-4663

Article Search

Original Article

Article Original Article
Split Viewer

Journal of Animal Reproduction and Biotechnology 2024; 39(4): 254-266

Published online December 31, 2024

https://doi.org/10.12750/JARB.39.4.254

Copyright © The Korean Society of Animal Reproduction and Biotechnology.

AAV2 serotype demonstrates the highest transduction efficiency in porcine lung-derived cells

Won Seok Ju #, Seokho Kim #,*, Areum Choi , Jae-Yeong Lee , Haesun Lee , Jingu No , Seunghoon Lee , Keonbong Oh and Jae Gyu Yoo

Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea

Correspondence to: Seokho Kim
E-mail: skim97@korea.kr

#These authors contributed equally to this work.

Received: October 31, 2024; Revised: December 10, 2024; Accepted: December 11, 2024

This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Background: The ability of adeno-associated viruses (AAVs) to transduce various cell types with minimal immune responses renders them prominent vectors for gene editing (GE), with different AAV serotypes exhibiting distinct transduction efficiencies due to their specific cellular tropism. However, detailed molecular processes of AAV infection and penetration, as well as the optimal serotype for specific purposes, remain poorly understood. Porcine models are widely used in research benefitting both human and livestock due to anatomical and physiological similarities to humans.
Methods: Transduction efficiencies of 18 AAV serotypes (AAV1–9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8) were evaluated in immortalized porcine lung epithelial cells (pLCsImt) and pulmonary alveolar macrophages 3D4/31 (PAMs 3D4/31).
Results: We found AAV2, DJ, and 2.7m8 to be the most effective in both cell types. The highest enhanced green fluorescent protein expression of 52.46 ± 2.4% in pLCsImt and 64.08 ± 2.4% in PAMs 3D4/31 was observed for AAV2, while negligible transduction was observed for AAV4, rh10, DJ, PHP.eB, PHP.S, and 2-retro. AAV-DJ showed superior transduction efficiency in PK-15, as compared to AAV2 and 2.7m8. Results emphasize the cell type-specific nature of AAV serotype transduction efficiencies. Notably, AAV2 was most effective in both lung and macrophage cells, whereas AAV-DJ was more effective in renal cells.
Conclusions: Our findings suggest that AAV2 was identified as the most efficient serotype for transducing pLCsImt and PAMs 3D4/31, compare to the PK-15 cells. Understanding cell type-specific preferences of AAV serotypes offer crucial insight for tailoring AAV vectors to specific tissue and optimizing genome editing strategies, with potential implications for the advancement of personalized medicine and development of treatments for human and livestock.

Keywords: adeno-associated virus 2 serotype, lung epithelial cells, porcine, pulmonary alveolar macrophages

Pigs are one of the most important livestock animals globally, and their economic value extends beyond being a source of meat for human consumption to include considerable application in the biotechnology industry (Thornton, 2010). Recently, pigs have been utilized as model organisms in in vitro or in vivo gene editing (GE) research, and studies on livestock disease resistance (Whitworth et al., 2022). Among the numerous diseases affecting pigs, porcine reproductive and respiratory syndrome (PRRS), stands out as a major concern, both economically and in terms of animal health (Whitworth et al., 2022). PRRS is generally caused by the PRRS virus (PRRSV), which primarily targets the respiratory and reproductive system in swine. The virus infects pigs via respiratory transmission, entering host cells through specific receptors, including CD163, a scavenger receptor expressed on pulmonary alveolar macrophages (PAMs) (Calvert et al., 2007). Upon binding to CD163, the virus enters the cell, where it replicates and subsequently spreads, triggering a complex immune response that often leads to persistent infection (Van Gorp et al., 2008). Therefore, studies on PRRS using GE strategies are essential for enhancing disease resistance in pigs.

Over the last decade, substantial progress has been made in using GE technologies, including the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system, to enhance desirable traits in pigs—such as disease resistance and reproductive performance—and to produce transgenic and knockout models (Gaj et al., 2013). CRISPR/Cas9 system has revolutionized the GE tool, providing a precise and efficient method to edit targeted genomic sites, making it possible to introduce favorable traits while eliminating unwanted characteristics (Singh and Ali, 2021). This technology has been instrumental in enhancing disease resistance, such as developing pig resistant to PRRS using CD163 gene deletion, that has a devastating economic impact on the swine industry (Whitworth et al., 2016). Moreover, intriguingly, pigs that express Cas9 endonuclease on their own are being generated to overcome the limitations of traditional transgenic animal generation processes (Wang et al., 2017). This advancement offers the advantage of directly verifying the function of various genes in living pigs. Such technological progress is expected to drastically reduce the time and cost required to identify and validate genes suitable for producing various transgenic pigs.

In addition to CRISPR/Cas9, adeno-associated viruses (AAVs) have become effective tools for delivering key CRISPR/Cas9 components, such as the Cas9 protein and single guide RNA, in pigs (Senís et al., 2014). AAVs, which are small, non-enveloped DNA viruses from the Parvoviridae family, are advantageous due to their low immunogenicity and their ability to target specific tissues, making them ideal for in vivo GE applications (Asmamaw Mengstie, 2022). AAVs are classified into distinct serotypes based on differences in their capsid proteins, including virus proteins viral protein 1 (VP1), VP2, and VP3. These capsid proteins are crucial in defining viral tropism, as they determine the interactions between AAVs and their target cells, thereby influencing tissue and cell specificity (Gao et al., 2004; Zincarelli et al., 2008; Mingozzi and High, 2011).

Several studies have aimed to identify tissue-specific AAV serotypes in pigs. For instance, AAV2H22 has successfully transduced porcine airway epithelial cells in vivo (Steines et al., 2016; Cooney et al., 2019). AAV4 is associated with respiratory diseases, such as cystic fibrosis, in pigs (Chen et al., 2021). AAV1 has been reported to be involved in efficient gene transfer to pig myocardium (Su et al., 2008). Additionally, in a study on transgene expression in porcine hepatocytes, AAVrh20 was identified as one of the most efficient capsids, demonstrating high transduction efficiency (Willimann et al., 2024). Furthermore, AAV2.8 effectively delivered RNAi-therapeutics targeting vascular endothelial growth factor A to reduce choroidal neovascularization (Haldrup et al., 2024).

However, AAV serotypes related to lung tissue-derived cells have not been fully screened in pigs. Based on tropisms and infection mechanisms of PRRS virus within the respiratory system (Calvert et al., 2007; Zhang et al., 2022), we compared the transduction efficiencies of various AAV serotypes in PAMs and porcine lung epithelial cells (pLCs) derived from the Cas9-expressing pig. For both types of porcine respiratory cells, our findings revealed extremely high tropism for the AAV2 serotype. This research aims to identify optimal vectors that can advance pulmonary GE strategies and immune modulation.

Cell culture

The pLCs utilized in this study were isolated from a Cas9-expressing pig (Kim et al., 2024); a breed derived from native Korean Black pigs (Cho et al., 2019) and maintained under 5% CO2 in PneumaCultTM-Ex Plus basal medium (StemCell Technologies Inc., Vancouver, BC, Canada, #05041) supplemented with 50× PneumaCultTM Ex-Plus supplement (STEMCELL Technologies Inc., Vancouver, BC, Canada, #05042) in a humidified incubator at 37℃.

Porcine alveolar macrophages 3D4/31 (PAMs 3D4/31) was obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA, #CRL-2844) and maintained under 5% CO2 in Roswell Park Memorial Institute 1640 (RPMI-1640) medium (+ L-Glutamine, + 25 mM HEPES) (Caisson Labs, Smithfield, UT, USA, #RPL09) supplemented with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA, #16000-044) and 1× penicillin-streptomycin (PS; Gibco, Grand Island, NY, USA, #10378-016) in a humidified incubator at 37℃. Dissociation of the pLCs and 3D4/31 PAMs from the culture ware was performed in the incubator using 0.25% trypsin-EDTA (Gibco, Grand Island, NY, USA, #25200-056) over 5 min, with the resulting cells washed once with phosphate-buffered saline without calcium magnesium (PBS; Caisson Labs, Smithfield, UT, USA, #PBL01).

Porcine kidney-15 (PK-15) cells and porcine ear fibroblasts were maintained under 5% CO2 in Dulbecco’s Modified Eagle’s Medium (DMEM) (Gibco, Grand Island, NY, USA, #10569-010) supplemented with 15% FBS, 1% minimum essential medium non-essential amino acids (MEM NEAA; Gibco, Grand Island, NY, USA, #11140-050), 1× Antibiotic-Antimycotic (Gibco, Grand Island, NY, USA, #15240-096), 0.1× Low Serum Growth Supplement (LSGS; Gibco, Grand Island, NY, USA, #S-003-10), and 0.001× beta-mercaptoethanol in a humidified incubator at 37℃.

Immortalization of pLCs using lentivirus particles

Immortalization was achieved by infecting primary pLCs at passage 4 according to the manufacturer’s instructions. Prior to infection, 0.2 mL of lentivirus suspension was placed in each well and diluted with complete medium containing hTERT-pCMV Hygro vector (GeneCopoeia Inc., Rockville, MD, USA, #LP-728-100) and polybrene transfection reagent (EMD Millipore Corp., Burlington, MA, USA, #TR-1003-G) to obtain a final concentration of 5 μg/mL. The original culture medium containing the pLCs was then removed and the virus-containing medium added to the wells. The control was produced by adding 0.2 mL of complete medium with polybrene to pLCs. Cells were then maintained under 5% CO2 in the humidified incubator at 37℃ for 18 h and the medium, with or without virus, was aspirated and replaced with fresh complete medium (0.5 mL) without polybrene and left for 24 h. To select stably transduced pLCs, the medium was replaced with fresh complete medium containing 150 μg/mL Hygromycin B (Gibco, Grand Island, NY, USA, #10687010) every 3 days. Cells were then split at a 1:10 ratio by trypsinization and re-seeded onto culture dishes until hygromycin-resistant colonies became visible. Immortalized pLCs (pLCsImt) were harvested at passage 10 and analyzed for the transient expression of human telomerase reverse transcriptase (hTERT) using real-time quantitative polymerase chain reaction (qPCR). Primary pLCs from passage 4 were used as internal hTERT gene controls. The primers used are listed in Table 1.

Table 1 . Primers used for real-time quantitative PCR analysis

SymbolGene namePrimer sequence (5’ → 3’)Accession no.Tm (℃)Product size (bp)
eGFPEnhanced green fluorescent proteinFw: AAG CAG AAG AAC GGC ATC AAX96418.155.597
Rv: GGG GGT GTT CTG CTG GTA GT61.0
hTERTHuman telomerase reverse transcriptaseFw: GCC GAG ACC AAG CAC TTC CTC GAC TNM_001193376.363.8111
Rv: GCA ACT TGC TCC AGA CAC TCT TCC G61.5
KRT5Keratin 5Fw: GCA GAT TGA GTG GAG AAG GTG TTXM_021067000.158.067
Rv: CCA GAG GAG AGG GTG TTT GTG59.1
FOXJ1Forkhead box protein 1Fw: CGC CAC AAC CTG TCT CTG AAXM_003357959.458.868
Rv: CCC TTG CCC GGC TCA T60.5
SYPSynaptophysinFw: CCT CAT CGG CTG AAT TCT TGGXM_003135078.554.761
Rv: GCC CCC ATG GAG TAG AGG AA60.4
Scgb1a1Secretoglobin family 1A member 1Fw: CAG AGG TCT GCC CGA GCT TXM_003353832.361.660
Rv: TGG CAA GTG TGC CCT TGA58.7
PDPNPodoplaninFw: TGC CAG TTC TGC TCT TCG TTTXM_005665017.357.966
Rv: CCG TGC TGG CTC CTT CTG60.8
SFTPBSurfactant protein-BFw: ACG CCC CAA CCG ATG ACNM_001102679.160.154
Rv: TGA GGA TGC TGG CGA TGT C59.0
GAPDHGlyceraldehyde-3-phosphate dehydrogenaseFw: ATG TTC CAG TAT GAT TCC ACC CAF01707955.2132
Rv: ACC AGC ATC ACC CCA TTT G56.8

bp, base pair; eGFP, enhanced green fluorescent protein; FOXJ1, forkhead box protein 1; Fw, forward; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; hTERT, human telomerase reverse transcriptase; KRT5, keratin 5; PDPN, podoplanin; Rv, reverse; Scgb1a1, secretoglobin family 1A member 1; SFTPB, surfactant protein-B; SYP, synaptophysin.



Single cell sorting by FACS

All procedures were conducted by the manufacturer’s instructions. Briefly, whole pLCsImt were index-sorted on an InvitrogenTM Bigfoot Spectral Cell Sorter (Thermo Fisher Scientific Inc., Waltham, MA, USA, #PL00304) operated by users experienced in single cell technologies. The instrument was used in a setup with a 70 μm nozzle and regular quality checks were done by running a BigfootTM Calibration Beads (Thermo Fisher Scientific Inc., Waltham, MA, USA, #PL00287). Best drop delay was determined using the BigfootTM Calibration Beads (Thermo Fisher Scientific Inc.) just before each experiments. Cells were hydrodynamically focused within a fluidic stream where single cells become encapsulated in individual droplets. To optimize the positioning of single sorted cells in the center of each well of the 96-well plate, “Run Image Alignment” was confirmed before every sorted plate. Next, single cells were sorted directly into 96-well clear bottom TC Surface plate (Thermo Fisher Scientific Inc., Waltham, MA, USA, #165306) pre-filled with PneumaCultTM-Ex Plus basal medium supplemented with 50× PneumaCultTM Ex-Plus supplement (1 cell per well). In each plate, three wells were left without sorted pLCsImt as the negative controls. The single-cell sorted pLCImt was maintained immediately after FACS sorting under 5% CO2 in a humidified incubator at 37℃. Single clones including #2, 3, 5, 6, 8, 10, 11, 16, 18, 19, 20, C2, C4, D1, and D4 were used for analyzing the expression level of the epithelial specific markers.

AAVs and in vitro transduction

All AAV serotypes (including AAV1-9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8) were purchased from VectorBuilder (VectorBuilder Inc., Chicago, IL, USA, #PANEL-AAVSP01-28), containing the enhanced green fluorescent protein (eGFP) reporter gene-driven cytomegalovirus promoter.

For in vitro transduction, pLCsImt and PAMs 3D4/31 were seeded in 24-well plates (Thermo Fisher Scientific Inc., Waltham, MA, USA, #142475) with the appropriate growth medium under 5% CO2 in the humidified incubator, which was maintained at 37℃. The medium was aspirated when the cells reached approximately 80% confluence, and transduction performed. To maximize the transduction efficiency, 250 μL of the AAV-containing growth medium was added to cells with the designated multiplicity of infection (MOI). The 24-well plate was then gently swirled to mix and cover the cells and the results incubated under 5% CO2 in the humidified incubator at 37℃ for 18 h. The AAV-containing medium was then safely removed, replaced with 500 μL fresh growth medium, and the results incubated overnight in the incubator.

Transduction efficiency

To elucidate the transduction efficiency, pLCsImt and PAMs 3D4/31 were seeded at a concentration of 5.0 × 104 cells per well in 24-well plates and incubated with 20K MOI of AAVs for 18 h. The AAV-containing medium was then replaced with fresh complete medium. Three days post-transduction, pLCsImt and PAMs 3D4/31 cells were stained with 1 μg/mL of Hoechst 33342 (Invitrogen Inc., Waltham, MA, USA, #H3570) in the growth medium for 5 min at room temperature. The staining medium was then removed and the cells washed once with DPBS containing 1× PS to remove any residual Hoechst dye. The eGFP expression levels were observed in both cell types three days following AAV transduction. A laser-scanning confocal microscope (Nikon AX, Nikon, Tokyo, Japan) was then used to visualize and photograph three random microscopic fields, with the blue-fluorescent nuclei followed by the green and red fluorescent signals. The images obtained were analyzed using ImageJ software and the total cell numbers elucidated by counting the blue-fluorescent nuclei, which was performed independently by two co-workers to ensure accuracy. The blue fluorescent images were then merged with the green (eGFP-positive cells) and red fluorescent images (tdTomato; Cas9) (Kim et al., 2024), and the transduction efficiency of the AAVs expressed as the percentage of green fluorescent cells relative to the total cell count. Data were presented as mean ± standard error of the mean.

Real-time qPCR

To assess the expression level of the eGFP mRNA (transduction), total RNA was isolated from pLCsImt and PAMs 3D4/31 five days following transduction of the AAVs using TRIzolTM Regeant (Invitrogen Inc.) and the Direct-zolTM RNA MiniPrep kit (Zymo Research Corp.) according to the manufacturer’s instructions. To further determine expression levels of epithelial specific markers such as keratin 5, forkhead box protein 1, synaptophysin, secretoglobin family 1A member 1, podoplanin, and surfactant protein-B, total RNA was isolated from single-cell sorted pLCsImt. Whole pLCsImt and porcine ear fibroblasts were used as positive and negative controls, respectively.

The quality and concentration of the extracted RNA were assessed using a NanoDrop® ND-1000 spectrophotometer (NanoDrop Technologies Inc., Wilmington, DE, USA) and the RNA stored at -80℃ until use. Synthesis of cDNA from the extracted total RNA (100 ng) was performed using the High Capacity cDNA Reverse Transcription kit (Applied Biosystems Inc., Waltham, MA, USA, #4368814), with reverse transcription performed at 37℃ for 120 min according to the manufacturer’s instructions. Specific primers were designed using Primer Express 3.01 software (Applied Biosystems Inc., Waltham, MA, USA). Details of the primer information can be seen in Table 1. Real-time qPCR was performed using the SYBR Green Master mix (Applied Biosystems Inc., Foster City, CA, USA, #4309155) and a StepOnePlus Real-Time PCR system (Applied Biosystems Inc., Foster City, CA, USA) under the following thermal-cycling procedure: 95℃ for 10 min, 40 cycles at 95℃ for 15 s, and 60℃ for 1 min (1.6℃/s ramp rate) according to the manufacturer’s instructions (Applied Biosystems Inc., Foster City, CA, USA). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal control. Gene expression was quantified via relative quantification (RQ) using the using ΔΔCt method and the data presented as the RQ. All experiments were performed at least in triplicate.

Statistical analysis

All data were analyzed for statistical significance using one-way ANOVA followed by post hoc Tukey’s HSD multiple range test for three or more groups using Statistical Products and Service Solutions (SPSS; International Business Machines SPSS Corp., Armonk, NY, USA) software (ver. 27). Error bars in the data for eGFP mRNA expression and the percentage of eGFP-positive cells represent the minimum and maximum RQ expression levels around the mean. Different letters denote significant differences between the groups. Student’s t-test was performed to compare the differences between two independent groups, allowing analysis of the hTERT expression levels. A statistical p-value of < 0.05 was considered significant in this study. All experiments were performed in triplicate.

AAV serotypes show different transduction efficiencies in immortalized porcine lung epithelial cells

The pLCsImt utilized in this study were generated via a meticulous process which involved the isolation and culture of primary pLCs (Supplementary Fig. 1A, left panel) from porcine lung tissue that intrinsically expresses Cas9 followed by immortalization via transduction with the hTERT gene. The resulting pLCsImt (Supplementary Fig. 1A, middle panel) exhibited high expression of the hTERT gene at passage 10 or beyond. Before the transduction of AAV serotypes in pLCsImt, we analyzed the expression related to the 6 epithelial specific markers (keratin 5, forkhead box protein 1, synaptophysin, secretoglobin family 1A member 1, podoplanin, and surfactant protein-B) in single-cell sorted pLCsImt whether they exhibit the characteristics of epithelial cells. As shown in Supplementary Fig. 2, while individual pLCImt sorted as single cell displayed relative differences in the expression of genes, it was confirmed that pLCsImt exhibit distinct epithelial cell-specific characteristics, compared to porcine ear fibroblasts. However, we used the whole pLCsImt, not the single-cell sorted pLCsImt, to investigate the AAV serotype transduction efficiency in pLCsImt.

The transduction efficiency of the 18 studied AAV serotypes (AAV1-9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8) on the pLCsImt with distinct capsid sequences was systematically compared, for which cells were infected with each AAV serotype and the visual transduction efficiency assessed three days post-infection using fluorescence microscopy. As shown in Fig. 1A, highly eGFP expression appeared three days post-infection for AAV2, AAV-DJ, and AAV2.7m8 as compared to the control, with weak green fluorescence observed for AAV1, AAV3, AAV5, AAV6, AAV6.2, AAV7, AAV8, and AAV9. AAV2-QuadYF was weakly expressed as compared to AAV2, AAV-DJ, and AAV2.7m8; however, no green fluorescence was observed in pLCsImt infected with AAV4, AAV-rh10, AAV-DJ/8, AAV-PHP.eB, AAV-PHP.S, and AAV2-retro three days post-infection. The highest percentage of 52.46 ± 2.4% was observed for the AAV2 serotype in terms of eGFP expression in the pLCsImt, with all other serotypes demonstrating much lower results in comparison, such as AAV-DJ at 22.11 ± 3.73% and AAV2.7m8 at 18.31 ± 2.14% (Fig. 1B).

Figure 1. Transduction efficiency of AAV serotypes containing eGFP on day 3 post-infection in pLCsImt. (A) Representative eGFP (green) fluorescence images in pLCsImt three days after transduction of various AAV vectors (AAV1–9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8). Individual AAV serotypes were infected at MOIs of 20 K for 18 h. The mock was not treated with the AAV vector and served as the negative control. Nuclei were stained with Hoechst 33342 (blue). Scale bars indicate 250 μm. (B) Quantification of eGFP-positive cells in different AAV serotype-infected pLCsImt. Data are presented as a mean ± standard error. Two independent co-workers counted. Different letters denote significant differences within each group, a-dp < 0.05 (n = 3). (C) Comparison with the mRNA expression level of eGFP in AAV serotype infected-pLCsImt. Relative quantification (RQ) of the gene expression was calculated using the ΔΔCt method, with data presented as RQ. Error bars represent the RQ of the minimum and maximum expression levels around the mean. All of quantification were normalized to the internal housekeeping gene GAPDH. Different letters denote significant differences within each group, a-kp < 0.05. All experiments were performed in triplicate. AAV, adeno-associated virus; eGFP, enhanced green fluorescence protein; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; MOI, multiplicity of infection; pLCsImt, immortalized porcine lung epithelial cells; RQ, relative quantification.

To further ascertain the transduction efficiency of the individual AAV serotypes, the mRNA expression levels of eGFP (all AAV vectors used in this study were eGFP-tagged) were genetically assessed using the pLCsImt (Fig. 1C). As seen in Fig. 1C, compared to the control group, the AAV2 serotype exhibited 27,215.58-fold higher eGFP expression levels, and although AAV-DJ (6,557.42-fold) and AAV2.7m8 (7,908.83-fold) showed high eGFP expression in pLCsImt, the AAV2 serotype demonstrated the highest transduction efficiency of the 18 serotypes evaluated. These results indicate that AAV2 is highly efficient for the transduction of pLCsImt and that AAV serotypes are cell type-specific.

AAV serotypes show comparable transduction efficiency in PAMs 3D4/31 to pLCsImt

To evaluate and compare the PAMs 3D4/31 (Supplementary Fig. 1A, right panel), the cells were transduced with each AAV serotype. The results were then compared with those observed for pLCsImt. Similar to pLCsImt, PAMs 3D4/31 were infected with all 18 of the AAV serotypes and the transduction efficiency evaluated. As shown in Fig. 2A, the results showed high eGFP expression for AAV2, AAV-DJ, and AAV2.7m8 as compared to the control, with weak green fluorescence observed in other serotypes, such as AAV1, AAV6, AAV6.2, and AAV2-QuadYF, and almost no eGFP expression for AAV3 and AAV5. No green fluorescence was observed in PAMs 3D4/31 infected with AAV4, AAV7, AAV8, AAV9, AAV-rh10, AAV-DJ/8, AAV-PHP.eB, AAV-PHP.S, and AAV2-retro. In particular, quantification of the eGFP expression for the AAV2 and AAV-DJ serotypes in PAMs 3D4/31 showed higher percentages of 64.08 ± 2.4% and 52.81 ± 4.73% as compared to the other AAV serotypes (Fig. 2B).

Figure 2. Transduction efficiency of individual AAV serotypes containing eGFP on day 3 post-infection in PAMs 3D4/31. (A) Representative eGFP (green) fluorescence images in PAMs 3D4/31 three days after transduction of various AAV vectors. Individual AAV serotypes were infected at MOIs of 20 K for 18 h. The mock was not treated with the AAV vector and served as the negative control. Nuclei were stained with Hoechst 33342 (blue). Scale bars indicate 250 μm. (B) Quantification of eGFP-positive cells in different AAV serotype-infected PAMs 3D4/31. Data were presented as a mean ± standard error. Two independent co-workers counted. Different letters denote significant differences within each group, a-fp < 0.05 (n = 3). (C) Comparison with the mRNA expression level of eGFP in AAV serotype infected-PAMs 3D4/31. RQ of the gene expression was calculated using the ΔΔCt method, with data presented as RQ. Error bars represent the RQ for the minimum and maximum expression levels around the mean. All of quantification were normalized to the internal housekeeping gene GAPDH. Different letters denote significant differences within each group, a-ip < 0.05. All experiments were performed in triplicate. PAMs 3D4/31, pulmonary alveolar macrophages 3D4/31.

To further determine the transduction efficiency of the various AAV serotypes, the mRNA expression levels of eGFP were also genetically assessed in PAMs 3D4/31 (Fig. 2C). As shown in Fig. 2C, compared with the control, the AAV2 serotype exhibited 1,951.98-fold higher eGFP expression levels, with the AAV-DJ serotype (1,921.74-fold) showing marginally lower eGFP expression than the AAV2 serotype, and the next highest eGFP expression levels exhibited by AAV2-QuadYF (1,341.23-fold) and AAV2.7m8 (680.99-fold). Similar to pLCsImt, AAV2 demonstrated the highest transduction efficiency for PAMs 3D4/31.

Transduction efficiency of individual AAV serotypes in porcine-derived cell types

Comprehensive analysis of the serotype transduction efficiencies for the various AAV serotypes in pLCsImt and PAMs 3D4/31 unequivocally demonstrated the highest transduction efficiency for the AAV2 serotype across both cell types (Fig. 3A, i-vi), with the AAV-DJ and AAV2.7m8 serotypes manifested sequentially lower yet substantial transduction efficiencies. In contrast, analogous transduction experiments in PK-15 cells resulted in a disparate serotype preference (Supplementary Fig. 3A and 3B), with AAV-DJ emerging as the serotype with paramount transduction efficiency, surpassing both AAV2 and AAV2.7m8. Notably, AAV2, which has previously shown superior efficacy in lung cells (Fig. 3A, i and iv), exhibited the lowest transduction efficiency among the three serotypes in renal cells. These findings indicate a marked cellular specificity for the AAV serotype transduction efficiencies, which underscores the differential affinity and transduction potential of the distinct AAV serotypes in various cellular environments. Specifically, the data showed pronounced tropism for AAV2 in both pLCsImt and PAMs 3D4/31, while the AAV-DJ serotype demonstrated a superior propensity for transduction in porcine renal cells. This cellular specificity is indicative of inherent differences in the cellular receptor availability, intracellular trafficking mechanisms, and nuclear entry pathways among the cell types examined (Fig. 3B).

Figure 3. Comparison of distinct AAV serotypes in vitro transduction efficiency in porcine. (A) Representative images showing significant eGFP expression (green) in both the pLCsImt and PAMs 3D4/31 transduced with AAV2, AAV-DJ, and AAV2.7m8 serotypes. Scale bars indicate 250 μm. (B) Schematic illustration demonstrating the cellular specificity of the various AAV serotypes assessed in this study. Individual AAV serotypes show different cellular tropism in the porcine model.

Genetically engineered pigs hold significant promise for application in both agriculture and biotechnological research. Nevertheless, their production remains challenging, costly, and time-intensive, primarily due to the lack of authentic germline-competent pluripotent stem cells (Fan and Lai, 2013). Due to these limitations, living Cas9-expressing GE pigs attracts considerable interest and substantially facilitate in vivo functional investigation of genes related disease resistance, such as PRRS, in agricultural and livestock industry (Whitworth et al., 2016; Wang et al., 2017).

AAV vectors have been rigorously investigated for their potential use in GE and biotechnological researches (Bijlani et al., 2022). Owing to their versatility, non-pathogenic characteristics, remarkable efficiency in gene delivery, and sustained persistence, AAVs have been projected to be extensively utilized across a broad range of fields (Asmamaw Mengstie, 2022; Issa et al., 2023), indicating their higher value. To date, 12 distinct AAV serotypes and over 100 natural variants have been identified (Balakrishnan and Jayandharan, 2014). Numerous AAV serotypes and variants exist, each exhibiting distinct transduction efficiencies depending on the specific cell or tissue type (Srivastava, 2016; Wiley et al., 2018; Issa et al., 2023). Several researches showed the importance of selecting the optimal AAV serotype for efficient gene delivery, particularly when targeting specific tissue including the lung (Liu et al., 2020; Yoon et al., 2021). Therefore, selecting an appropriate AAV serotype is crucial for effective AAV-based gene delivery. Our analysis of AAV recombinant serotypes using the porcine lung cells-derived from the Cas9-expressing pig (Kim et al., 2024) showed that AAV2 has the higher and superior tropism for pLCsImt. In addition to pLCsImt, AAV2 showed substantially higher transduction efficiency in PAMs 3D4/31. These findings indicate the specificity and superior efficiency of AAV2 across both cell types compared to other serotypes tested, thereby underscoring the importance of selecting the optimal AAV serotype for efficient gene delivery.

The AAV2 is capable of infiltrating and transducing various cellular phenotypes including lung across interspecies boundaries (Halbert et al., 2000; Chen et al., 2005). AAV2 has reported that it has also remarkable efficiency in neurons, hepatocytes, and mesenchymal stromal cells compared to other serotypes, such as serotypes 1, 3, 4, 5, 6, and 8 (Chng et al., 2007; Stender et al., 2007; Srivastava, 2016; Logan et al., 2017). In our study, AAV2 showed the highest transduction efficiency in both pLCsImt and PAMs 3D4/31, with eGFP expression levels of 52.46 ± 2.4% in pLCsImt and 64.08 ± 2.4% in PAMs 3D4/31 compared the others. The results firmly underscore its potential as a promising vector for the effective gene delivery in porcine lung tissues. Meanwhile, AAV-DJ and AAV2.7m8 also exhibited significant transduction capabilities in both cell types in this study, although their efficiencies were lower than that of AAV2. These results are consistent with Lisowski et al. (2015), who reported broad tropism for AAV-DJ across various tissues. AAV2.7m8, a variant of AAV2 modified to enhance its tropism (Bennett et al., 2020), also performed well, suggesting that engineered variants such as AAV2.7m8 hold promise for applications where high transduction efficiency is specifically required (Mendell et al., 2021). Interestingly, AAV-DJ exhibited relatively higher transduction efficiency than both AAV2 and AAV2.7m8 in PK-15 cells, indicating that the sensitivity of AAV varies in a cell-type-specific manner (Grimm et al., 2008; Issa et al., 2023).

In contrast, the other serotypes such as AAV4, AAV-rh10, AAV-DJ/8, AAV-PHP.eB, AAV-PHP.S, and AAV2-retro exhibited minimal to no transduction in both lung cell types in this study. In case of AAV4, although it has been reported potential as a vector for lung tissue-specific gene delivery (Issa et al., 2023), it has been reported to exhibit low overall expression levels in murine lung tissue (Zincarelli et al., 2008). In fact, AAV4 has primarily showed higher transduction efficiency in retinal cells in rodent, canine, and non-human primate (Weber et al., 2003). Furthermore, AAV-DJ/8 has been reported that it showed diverse tropisms and proclivity for liver, spleen, and neural cells (Grimm et al., 2008). In case of AAV-PHP.eB and -PHP.S, they display a predilection for the central nervous system (Chan et al., 2017). Similar with AAV-PHP.eB and -PHP.S, AAV2-retro demonstrates specific tropism for spinal neurons (Wang et al., 2018). Previous studies have also highlighted the variable efficiency of these serotypes in different tissues. These findings underscore the importance of appropriate AAV serotype selection according to target cell types once more (Su et al., 2008; Haldrup et al., 2024).

In this way, the transduction efficiency observed in tissues does not always translate reliably to isolated cell types due to the factors such as the local microenvironment, receptor expression levels, and intracellular tracking pathways, which can vary between tissues and isolated cells (Zengel et al., 2023). While our study only focused on the transduction efficiencies in Cas9-expressing pig-derived lung cells (Kim et al., 2024), further validation using isolated and purified cell types could enhance our understanding of species- and cell type-specific transduction mechanisms of AAVs (Qi et al., 2013).

However, one of the limitations of this study is the lack of assessment of the effect of pre-existing neutralizing antibodies (NAbs) on the infection efficiency of the tested AAV serotypes. Neutralizing antibodies are immune system proteins that specifically recognize and bind to viral particles, effectively preventing them from infecting cells. Pre-existing NAbs can significantly hinder the efficacy of AAV-mediated gene delivery, as they can neutralize the viral vector before it reaches the target cells (Kotterman et al., 2015; Gorovits et al., 2020). Studies have shown that NAb prevalence against AAV2 is notably high in both humans and pigs, potentially limiting its effectiveness in clinical applications (Calcedo et al., 2011; Dai et al., 2022). NAbs can vary significantly across populations and species, leading to variability in vector efficacy. This variation complicates the translation of preclinical findings to clinical settings and impacts applications in livestock health and productivity. Therefore, future research should include a comprehensive evaluation of NAbs to better predict and enhance the effectiveness of AAV-based gene therapies in both human and livestock contexts. Specifically, understanding the role of NAbs in livestock is crucial to ensure the success of gene delivery methods aimed at improving productivity, disease resistance, and overall animal health. Strategies to circumvent the challenges posed by NAbs could include engineering novel capsid variants that evade recognition by existing antibodies, using empty capsids as decoys to absorb NAbs, or employing immunosuppressive regimens to temporarily reduce antibody levels during vector administration.

Our study focuses on the application of in vivo GE technologies at enhancing disease resistance, such as PRRS, in pigs. To achieve this, we utilized the lung cells derived from Cas9-expressing pig models to identify the optimal and efficient AAV vector carrying CD163 guide RNA. In this process, we found that AAV2 exhibited considerably higher transduction efficiency in lung epithelial cells. Despite the many promising suggestions for AAV delivery systems into the lung, there are still challenges that require practical consideration. These findings from our attempt would help in applying AAV-based in vivo GE for PRRS in the future. The current study is meaningful in that AAVs with high transduction efficiencies for transgene delivery into the lung in Cas9-expressing pig were compared and determined among various AAV serotypes.

Our data indicate that AAV2 was identified as the most efficient serotype for transducing pLCsImt and PAMs 3D4/31, highlighting its potential for pulmonary GE application. Pigs have been used as a useful model and important resources in livestock industry as well as biotechnological research (Bähr and Wolf, 2012; Hryhorowicz et al., 2017; Wells et al., 2017; Clauss et al., 2019). Besides, ensuring the safety of GE is crucial, particularly minimizing immune responses and off-target effects on organs by selecting the efficient vector in AAV-mediated GE application. These findings emphasize the importance of serotype selection in gene delivery and provide a foundation for further research aimed at optimizing AAV vectors for in vivo GE application in livestock industry. The demonstrated cellular specificity of AAV transduction also calls for continued investigation of the factors influencing vector performance in different cellular contexts, paving the way for tailored GE approaches.

This study was supported by 2024 the RDA Fellowship Program of the National Institute of Animal Science, Rural Development Administration, Republic of Korea.

Conceptualization, W.S.J. and S.K.; methodology, W.S.J. and S.K.; investigation, W.S.J., A.C., and S.K.; data curation, W.S.J., A.C., and S.K.; writing-original draft preparation, W.S.J. and S.K.; writing-review and editing, W.S.J., J-Y.L., H.L., J.N., S.L., K.O., J.G.Y., and S.K.; project administration, S.K.; funding acquisition, S.K.

This research was supported by the Cooperative Research Program for Agriculture Science and Technology Development (Project No. PJ015712) of Rural Development Administration, Republic of Korea.

  1. Asmamaw Mengstie M. 2022. Viral vectors for the in vivo delivery of CRISPR components: advances and challenges. Front. Bioeng. Biotechnol. 12;10:895713.
    Pubmed KoreaMed CrossRef
  2. Balakrishnan B and Jayandharan GR. 2014. Basic biology of adeno-associated virus (AAV) vectors used in gene therapy. Curr. Gene Ther. 14:86-100.
    Pubmed CrossRef
  3. Bennett A, Keravala A, Makal V, Kurian J, Belbellaa B, Aeran R, Tseng YS, Sousa D, Spear J, Agbandje-McKenna M. 2020. Structure comparison of the chimeric AAV2. 7m8 vector with parental AAV2. J. Struct. Biol. 209:107433.
    Pubmed KoreaMed CrossRef
  4. Bijlani S, Pang KM, Sivanandam V, Chatterjee S. 2022. The role of recombinant AAV in precise genome editing. Front. Genome Ed. 3:799722.
    Pubmed KoreaMed CrossRef
  5. Bähr A and Wolf E. 2012. Domestic animal models for biomedical research. Reprod. Domest. Anim. 47 Suppl 4:59-71.
    Pubmed CrossRef
  6. Calcedo R, Morizono H, Wang L, McCarter R, He J, Jones D, Wilson JM. 2011. Adeno-associated virus antibody profiles in newborns, children, and adolescents. Clin. Vaccine. Immunol. 18:1586-1588.
    Pubmed KoreaMed CrossRef
  7. Calvert JG, Slade DE, Shields SL, Jolie R, Mannan RM, Welch SK. 2007. CD163 expression confers susceptibility to porcine reproductive and respiratory syndrome viruses. J. Virol. 81:7371-7379.
    Pubmed KoreaMed CrossRef
  8. Chan KY, Jang MJ, Yoo BB, Greenbaum A, Ravi N, Wu WL, Sánchez-Guardado L, Lois C, Mazmanian SK, Gradinaru V. 2017. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20:1172-1179.
    Pubmed KoreaMed CrossRef
  9. Chen CL, Jensen RL, Schnepp BC, Connell MJ, Shell R, Sferra TJ, Bartlett JS, Johnson PR. 2005. Molecular characterization of adeno-associated viruses infecting children. J. Virol. 79:14781-14792.
    Pubmed KoreaMed CrossRef
  10. Chen OG, Mather SE, Brommel CM, Hamilton BA, Ehler A, Villacreses R, Girgis RE, Abou Alaiwa M, Stoltz DA, Li X. 2021. Transduction of pig small airway epithelial cells and distal lung progenitor cells by AAV4. Cells 10:1014.
    Pubmed KoreaMed CrossRef
  11. Chng K, Larsen SR, Zhou S, Wright JF, Rasko JE. 2007. Specific adeno-associated virus serotypes facilitate efficient gene transfer into human and non-human primate mesenchymal stromal cells. J. Gene Med. 9:22-32.
    Pubmed CrossRef
  12. Cho IC, Park HB, Ahn JS, Han SH, Lee JB, Lim HT, Yoo CK, Jung EJ, Kim DH, Sun WS, Ramayo-Caldas Y, Kim SG, Kang YJ, Kim YK, Shin HS, Seong PN, Hwang IS, Park BY, Hwang S, Lee SS, Ryu YC, Lee JH, Ko MS, Lee K, Andersson G, Lee JW. 2019. A functional regulatory variant of MYH3 influences muscle fiber-type composition and intramuscular fat content in pigs. PLoS Genet 15:e1008279.
    Pubmed KoreaMed CrossRef
  13. Clauss S, Bleyer C, Schüttler D, Tomsits P, Renner S, Klymiuk N, Wakili R, Massberg S, Kääb S. 2019. Animal models of arrhythmia: classic electrophysiology to genetically modified large animals. Nat. Rev. Cardiol. 16:457-475.
    Pubmed CrossRef
  14. Cooney AL, Thornell IM, Singh BK, Shah VS, Stoltz DA, McCray PB Jr, Sinn PL. 2019. A novel AAV-mediated gene delivery system corrects CFTR function in pigs. Am. J. Respir. Cell Mol. Biol. 61:747-754.
    Pubmed KoreaMed CrossRef
  15. Dai Y, Kavita U, Lampen MH, Gielen S, Banks G, Levesque P, Kozhich A, Pillutla R, Zhang Y, Adam L. 2022. Prevalence of pre-existing neutralizing antibodies against adeno-associated virus serotypes 1, 2, 5, 6, 8, and 9 in sera of different pig strains. Hum. Gene Ther. 33:451-459.
    Pubmed CrossRef
  16. Fan N and Lai L. 2013. Genetically modified pig models for human diseases. J. Genet. Genomics 40:67-73.
    Pubmed CrossRef
  17. Gaj T, Barbas CF 3rd. 2013. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends. Biotechnol. 31:397-405.
    Pubmed KoreaMed CrossRef
  18. Gao G, Vandenberghe LH, Alvira MR, Lu Y, Calcedo R, Wilson JM. 2004. Clades of Adeno-associated viruses are widely disseminated in human tissues. J. Virol. 78:6381-6388.
    Pubmed KoreaMed CrossRef
  19. Gorovits B, Fiscella M, Havert M, Koren E, Long B, Purushothama S. 2020. Recommendations for the development of cell-based anti-viral vector neutralizing antibody assays. AAPS J. 22:24. (Erratum published 2020, AAPS. J. 22:42).
    Pubmed CrossRef
  20. Grimm D, Lee JS, Wang L, Desai T, Akache B, Kay MA. 2008. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 82:5887-5911.
    Pubmed KoreaMed CrossRef
  21. Halbert CL, Rutledge EA, Allen JM, Miller AD. 2000. Repeat transduction in the mouse lung by using adeno-associated virus vectors with different serotypes. J. Virol. 74:1524-1532.
    Pubmed KoreaMed CrossRef
  22. Haldrup SH, Fabian-Jessing BK, Jakobsen TS, Lindholm AB, Adsersen RL, Aagaard L, Bek T, Corydon TJ. 2024. Subretinal AAV delivery of RNAi-therapeutics targeting VEGFA reduces choroidal neovascularization in a large animal model. Mol. Ther. Methods Clin. Dev. 32:101242.
    Pubmed KoreaMed CrossRef
  23. Hryhorowicz M, Zeyland J, Lipiński D. 2017. Genetically modified pigs as organ donors for xenotransplantation. Mol. Biotechnol. 59:435-444.
    Pubmed KoreaMed CrossRef
  24. Issa SS, Shaimardanova AA, Rizvanov AA. 2023. Various AAV serotypes and their applications in gene therapy: an overview. Cells 12:785.
    Pubmed KoreaMed CrossRef
  25. Kim S, No JG, Lee S, Choi A, Lee JY, Hyung N, Ju WS, Lee JY, Kwak TU, Lee P, Park MR, Byun SJ, Lee H, Oh KB, Yoo JG. 2024. In vitro gene editing using primary cells derived from Cas9-expressing pigs. J. Anim. Sci. Technol. . https://doi.org/10.5187/jast.2024.e77.
    CrossRef
  26. Kotterman MA, Yin L, Strazzeri JM, Flannery JG, Schaffer DV. 2015. Antibody neutralization poses a barrier to intravitreal adeno-associated viral vector gene delivery to non-human primates. Gene Ther. 22:116-126.
    Pubmed KoreaMed CrossRef
  27. Lisowski L, Alexander IE. 2015. Adeno-associated virus serotypes for gene therapeutics. Curr. Opin. Pharmacol. 24:59-67.
    Pubmed CrossRef
  28. Liu Z, Klose K, Neuber S, Jiang M, Stamm C. 2020. Comparative analysis of adeno-associated virus serotypes for gene transfer in organotypic heart slices. J. Transl. Med. 18:437.
    Pubmed KoreaMed CrossRef
  29. Logan GJ, Dane AP, Hallwirth CV, Smyth CM, Wilkie EE, Amaya AK, Zhu E, Khandekar N, Ginn SL, Liao SHY, Cunningham SC, Sasaki N, Cabanes-Creus M, Tam PPL, Russell DW, Alexander IE. 2017. Identification of liver-specific enhancer-promoter activity in the 3' untranslated region of the wild-type AAV2 genome. Nat. Genet. 49:1267-1273.
    Pubmed CrossRef
  30. Mendell JR, Al-Zaidy SA, Rodino-Klapac LR, Goodspeed K, Gray SJ, Kay CN, Boye SL, Boye SE, George LA, Salabarria S, Corti M, Tremblay JP. 2021. Current clinical applications of in vivo gene therapy with AAVs. Mol. Ther. 29:464-488.
    Pubmed KoreaMed CrossRef
  31. Mingozzi F and High KA. 2011. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat. Rev. Genet. 12:341-355. (Erratum published 2011, Nat. Rev. Genet. 12:515).
    Pubmed CrossRef
  32. Qi YF, Li QH, Shenoy V, Zingler M, Jun JY, Verma A, Raizada MK. 2013. Comparison of the transduction efficiency of tyrosine-mutant adeno-associated virus serotype vectors in kidney. Clin. Exp. Pharmacol. Physiol. 40:53-55.
    Pubmed KoreaMed CrossRef
  33. Senís E, Fatouros C, Große S, Wiedtke E, Niopek D, Mueller AK, Grimm D. 2014. CRISPR/Cas9-mediated genome engineering: an adeno-associated viral (AAV) vector toolbox. Biotechnol. J. 9:1402-1412.
    Pubmed CrossRef
  34. Singh P and Ali SA. 2021. Impact of CRISPR-Cas9-based genome engineering in farm animals. Vet. Sci. 8:122.
    Pubmed KoreaMed CrossRef
  35. Srivastava A. 2016. In vivo tissue-tropism of adeno-associated viral vectors. Curr. Opin. Virol. 21:75-80.
    Pubmed KoreaMed CrossRef
  36. Steines B, Dickey DD, Bergen J, Excoffon KJ, Weinstein JR, Li X, Yan Z, Abou Alaiwa MH, Shah VS, Bouzek DC, Powers LS, Gansemer ND, Ostedgaard LS, Engelhardt JF, Stoltz DA, Welsh MJ, Sinn PL, Zabner J. 2016. CFTR gene transfer with AAV improves early cystic fibrosis pig phenotypes. JCI Insight 1:e88728.
    Pubmed KoreaMed CrossRef
  37. Stender S, Murphy M, O'Brien T, Stengaard C, Ulrich-Vinther M, Barry F. 2007. Adeno-associated viral vector transduction of human mesenchymal stem cells. Eur. Cell Mater. 13:93-99. discussion 99.
    Pubmed CrossRef
  38. Su H, Yeghiazarians Y, Lee A, Huang Y, Arakawa-Hoyt J, Ye J, Orcino G, Kan YW. 2008. AAV serotype 1 mediates more efficient gene transfer to pig myocardium than AAV serotype 2 and plasmid. J. Gene Med. 10:33-41.
    Pubmed CrossRef
  39. Thornton PK. 2010. Livestock production: recent trends, future prospects. Philos. Trans. R Soc. Lond. B Biol. Sci. 365:2853-2867.
    Pubmed KoreaMed CrossRef
  40. Van Gorp H, Van Breedam W, Nauwynck HJ. 2008. Sialoadhesin and CD163 join forces during entry of the porcine reproductive and respiratory syndrome virus. J. Gen. Virol. 89(Pt 12:2943-2953.
    Pubmed CrossRef
  41. Wang K, Jin Q, Ruan D, Yang Y, Liu Q, Wu H, Zhou Z, Ouyang Z, Liu Z, Zhao Y, Zhao B, Zhang Q, Peng J, Lai C, Fan N, Liang Y, Lan T, Li N, Wang X, Wang X, Fan Y, Doevendans PA, Sluijter JPG, Liu P, Lai L. 2017. Cre-dependent Cas9-expressing pigs enable efficient in vivo genome editing. Genome Res. 27:2061-2071.
    Pubmed KoreaMed CrossRef
  42. Wang Z, Maunze B, Wang Y, Blackmore MG. 2018. Global connectivity and function of descending spinal input revealed by 3D microscopy and retrograde transduction. J. Neurosci. 38:10566-10581.
    Pubmed KoreaMed CrossRef
  43. Weber M, Rabinowitz J, Provost N, Conrath H, Folliot S, Briot D, Chérel Y, Chenuaud P, Samulski J, Rolling F. 2003. Recombinant adeno-associated virus serotype 4 mediates unique and exclusive long-term transduction of retinal pigmented epithelium in rat, dog, and nonhuman primate after subretinal delivery. Mol. Ther. 7:774-781.
    Pubmed CrossRef
  44. Wells KD, Bardot R, Whitworth KM, Trible BR, Fang Y, Mileham A, Kerrigan MA, Samuel MS, Rowland RRR. 2017. Replacement of porcine CD163 scavenger receptor cysteine-rich domain 5 with a CD163-like homolog confers resistance of pigs to genotype 1 but not genotype 2 porcine reproductive and respiratory syndrome virus. J. Virol. 91:e01521-16.
    Pubmed KoreaMed CrossRef
  45. Whitworth KM, Green JA, Redel BK, Geisert RD, Lee K, Telugu BP, Prather RS. 2022. Improvements in pig agriculture through gene editing. CABI Agric. Biosci. 3:41.
    Pubmed KoreaMed CrossRef
  46. Whitworth KM, Rowland RR, Ewen CL, Trible BR, Kerrigan MA, Cino-Ozuna AG, Samuel MS, Lightner JE, McLaren DG, Mileham AJ, Prather RS. 2016. Gene-edited pigs are protected from porcine reproductive and respiratory syndrome virus. Nat. Biotechnol. 34:20-22.
    Pubmed CrossRef
  47. Wiley LA, Burnight ER, Kaalberg EE, Jiao C, Riker MJ, Halder JA, Luse MA, Han IC, Russell SR, Sohn EH, Stone EM, Mullins RF. 2018. Assessment of adeno-associated virus serotype tropism in human retinal explants. Hum. Gene Ther. 29:424-436.
    Pubmed KoreaMed CrossRef
  48. Willimann M, Tiyaboonchai A, Adachi K, Li B, Waldburger L, Nakai H, Thöny B. 2024. AAV capsid screening for translational pig research using a mouse xenograft liver model. bioRxiv. . https://doi.org/10.1101/2024.05.29.596409.
    CrossRef
  49. Yoon DS, Lee KM, Cho S, Ko EA, Kim J, Jung S, Shim JH, Gao G, Lee JW. 2021. Cellular and tissue selectivity of AAV serotypes for gene delivery to chondrocytes and cartilage. Int. J. Med. Sci. 18:3353-3360.
    Pubmed KoreaMed CrossRef
  50. Zengel J, Wang YX, Seo JW, Ning K, Hamilton JN, Wu B, Raie M, Holbrook C, Su S, Clements DR, Pillay S, Puschnik AS, Winslow MM, Idoyaga J, Nagamine CM, Sun Y, Mahajan VB, Ferrara KW, Carette JE. 2023. Hardwiring tissue-specific AAV transduction in mice through engineered receptor expression. Nat. Methods. 20:1070-1081.
    Pubmed KoreaMed CrossRef
  51. Zhang J, Wang J, Zhang X, Zhao C, Zhou S, Du C, Tan Y, Shi K. 2022. Transcriptome profiling identifies immune response genes against porcine reproductive and respiratory syndrome virus and Haemophilus parasuis co-infection in the lungs of piglets. J. Vet. Sci. 23:e2.
    CrossRef
  52. Zincarelli C, Soltys S, Rabinowitz JE. 2008. Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Mol. Ther. 16:1073-1080.
    Pubmed CrossRef

Article

Original Article

Journal of Animal Reproduction and Biotechnology 2024; 39(4): 254-266

Published online December 31, 2024 https://doi.org/10.12750/JARB.39.4.254

Copyright © The Korean Society of Animal Reproduction and Biotechnology.

AAV2 serotype demonstrates the highest transduction efficiency in porcine lung-derived cells

Won Seok Ju #, Seokho Kim #,*, Areum Choi , Jae-Yeong Lee , Haesun Lee , Jingu No , Seunghoon Lee , Keonbong Oh and Jae Gyu Yoo

Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea

Correspondence to:Seokho Kim
E-mail: skim97@korea.kr

#These authors contributed equally to this work.

Received: October 31, 2024; Revised: December 10, 2024; Accepted: December 11, 2024

This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background: The ability of adeno-associated viruses (AAVs) to transduce various cell types with minimal immune responses renders them prominent vectors for gene editing (GE), with different AAV serotypes exhibiting distinct transduction efficiencies due to their specific cellular tropism. However, detailed molecular processes of AAV infection and penetration, as well as the optimal serotype for specific purposes, remain poorly understood. Porcine models are widely used in research benefitting both human and livestock due to anatomical and physiological similarities to humans.
Methods: Transduction efficiencies of 18 AAV serotypes (AAV1–9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8) were evaluated in immortalized porcine lung epithelial cells (pLCsImt) and pulmonary alveolar macrophages 3D4/31 (PAMs 3D4/31).
Results: We found AAV2, DJ, and 2.7m8 to be the most effective in both cell types. The highest enhanced green fluorescent protein expression of 52.46 ± 2.4% in pLCsImt and 64.08 ± 2.4% in PAMs 3D4/31 was observed for AAV2, while negligible transduction was observed for AAV4, rh10, DJ, PHP.eB, PHP.S, and 2-retro. AAV-DJ showed superior transduction efficiency in PK-15, as compared to AAV2 and 2.7m8. Results emphasize the cell type-specific nature of AAV serotype transduction efficiencies. Notably, AAV2 was most effective in both lung and macrophage cells, whereas AAV-DJ was more effective in renal cells.
Conclusions: Our findings suggest that AAV2 was identified as the most efficient serotype for transducing pLCsImt and PAMs 3D4/31, compare to the PK-15 cells. Understanding cell type-specific preferences of AAV serotypes offer crucial insight for tailoring AAV vectors to specific tissue and optimizing genome editing strategies, with potential implications for the advancement of personalized medicine and development of treatments for human and livestock.

Keywords: adeno-associated virus 2 serotype, lung epithelial cells, porcine, pulmonary alveolar macrophages

INTRODUCTION

Pigs are one of the most important livestock animals globally, and their economic value extends beyond being a source of meat for human consumption to include considerable application in the biotechnology industry (Thornton, 2010). Recently, pigs have been utilized as model organisms in in vitro or in vivo gene editing (GE) research, and studies on livestock disease resistance (Whitworth et al., 2022). Among the numerous diseases affecting pigs, porcine reproductive and respiratory syndrome (PRRS), stands out as a major concern, both economically and in terms of animal health (Whitworth et al., 2022). PRRS is generally caused by the PRRS virus (PRRSV), which primarily targets the respiratory and reproductive system in swine. The virus infects pigs via respiratory transmission, entering host cells through specific receptors, including CD163, a scavenger receptor expressed on pulmonary alveolar macrophages (PAMs) (Calvert et al., 2007). Upon binding to CD163, the virus enters the cell, where it replicates and subsequently spreads, triggering a complex immune response that often leads to persistent infection (Van Gorp et al., 2008). Therefore, studies on PRRS using GE strategies are essential for enhancing disease resistance in pigs.

Over the last decade, substantial progress has been made in using GE technologies, including the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system, to enhance desirable traits in pigs—such as disease resistance and reproductive performance—and to produce transgenic and knockout models (Gaj et al., 2013). CRISPR/Cas9 system has revolutionized the GE tool, providing a precise and efficient method to edit targeted genomic sites, making it possible to introduce favorable traits while eliminating unwanted characteristics (Singh and Ali, 2021). This technology has been instrumental in enhancing disease resistance, such as developing pig resistant to PRRS using CD163 gene deletion, that has a devastating economic impact on the swine industry (Whitworth et al., 2016). Moreover, intriguingly, pigs that express Cas9 endonuclease on their own are being generated to overcome the limitations of traditional transgenic animal generation processes (Wang et al., 2017). This advancement offers the advantage of directly verifying the function of various genes in living pigs. Such technological progress is expected to drastically reduce the time and cost required to identify and validate genes suitable for producing various transgenic pigs.

In addition to CRISPR/Cas9, adeno-associated viruses (AAVs) have become effective tools for delivering key CRISPR/Cas9 components, such as the Cas9 protein and single guide RNA, in pigs (Senís et al., 2014). AAVs, which are small, non-enveloped DNA viruses from the Parvoviridae family, are advantageous due to their low immunogenicity and their ability to target specific tissues, making them ideal for in vivo GE applications (Asmamaw Mengstie, 2022). AAVs are classified into distinct serotypes based on differences in their capsid proteins, including virus proteins viral protein 1 (VP1), VP2, and VP3. These capsid proteins are crucial in defining viral tropism, as they determine the interactions between AAVs and their target cells, thereby influencing tissue and cell specificity (Gao et al., 2004; Zincarelli et al., 2008; Mingozzi and High, 2011).

Several studies have aimed to identify tissue-specific AAV serotypes in pigs. For instance, AAV2H22 has successfully transduced porcine airway epithelial cells in vivo (Steines et al., 2016; Cooney et al., 2019). AAV4 is associated with respiratory diseases, such as cystic fibrosis, in pigs (Chen et al., 2021). AAV1 has been reported to be involved in efficient gene transfer to pig myocardium (Su et al., 2008). Additionally, in a study on transgene expression in porcine hepatocytes, AAVrh20 was identified as one of the most efficient capsids, demonstrating high transduction efficiency (Willimann et al., 2024). Furthermore, AAV2.8 effectively delivered RNAi-therapeutics targeting vascular endothelial growth factor A to reduce choroidal neovascularization (Haldrup et al., 2024).

However, AAV serotypes related to lung tissue-derived cells have not been fully screened in pigs. Based on tropisms and infection mechanisms of PRRS virus within the respiratory system (Calvert et al., 2007; Zhang et al., 2022), we compared the transduction efficiencies of various AAV serotypes in PAMs and porcine lung epithelial cells (pLCs) derived from the Cas9-expressing pig. For both types of porcine respiratory cells, our findings revealed extremely high tropism for the AAV2 serotype. This research aims to identify optimal vectors that can advance pulmonary GE strategies and immune modulation.

MATERIALS AND METHODS

Cell culture

The pLCs utilized in this study were isolated from a Cas9-expressing pig (Kim et al., 2024); a breed derived from native Korean Black pigs (Cho et al., 2019) and maintained under 5% CO2 in PneumaCultTM-Ex Plus basal medium (StemCell Technologies Inc., Vancouver, BC, Canada, #05041) supplemented with 50× PneumaCultTM Ex-Plus supplement (STEMCELL Technologies Inc., Vancouver, BC, Canada, #05042) in a humidified incubator at 37℃.

Porcine alveolar macrophages 3D4/31 (PAMs 3D4/31) was obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA, #CRL-2844) and maintained under 5% CO2 in Roswell Park Memorial Institute 1640 (RPMI-1640) medium (+ L-Glutamine, + 25 mM HEPES) (Caisson Labs, Smithfield, UT, USA, #RPL09) supplemented with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA, #16000-044) and 1× penicillin-streptomycin (PS; Gibco, Grand Island, NY, USA, #10378-016) in a humidified incubator at 37℃. Dissociation of the pLCs and 3D4/31 PAMs from the culture ware was performed in the incubator using 0.25% trypsin-EDTA (Gibco, Grand Island, NY, USA, #25200-056) over 5 min, with the resulting cells washed once with phosphate-buffered saline without calcium magnesium (PBS; Caisson Labs, Smithfield, UT, USA, #PBL01).

Porcine kidney-15 (PK-15) cells and porcine ear fibroblasts were maintained under 5% CO2 in Dulbecco’s Modified Eagle’s Medium (DMEM) (Gibco, Grand Island, NY, USA, #10569-010) supplemented with 15% FBS, 1% minimum essential medium non-essential amino acids (MEM NEAA; Gibco, Grand Island, NY, USA, #11140-050), 1× Antibiotic-Antimycotic (Gibco, Grand Island, NY, USA, #15240-096), 0.1× Low Serum Growth Supplement (LSGS; Gibco, Grand Island, NY, USA, #S-003-10), and 0.001× beta-mercaptoethanol in a humidified incubator at 37℃.

Immortalization of pLCs using lentivirus particles

Immortalization was achieved by infecting primary pLCs at passage 4 according to the manufacturer’s instructions. Prior to infection, 0.2 mL of lentivirus suspension was placed in each well and diluted with complete medium containing hTERT-pCMV Hygro vector (GeneCopoeia Inc., Rockville, MD, USA, #LP-728-100) and polybrene transfection reagent (EMD Millipore Corp., Burlington, MA, USA, #TR-1003-G) to obtain a final concentration of 5 μg/mL. The original culture medium containing the pLCs was then removed and the virus-containing medium added to the wells. The control was produced by adding 0.2 mL of complete medium with polybrene to pLCs. Cells were then maintained under 5% CO2 in the humidified incubator at 37℃ for 18 h and the medium, with or without virus, was aspirated and replaced with fresh complete medium (0.5 mL) without polybrene and left for 24 h. To select stably transduced pLCs, the medium was replaced with fresh complete medium containing 150 μg/mL Hygromycin B (Gibco, Grand Island, NY, USA, #10687010) every 3 days. Cells were then split at a 1:10 ratio by trypsinization and re-seeded onto culture dishes until hygromycin-resistant colonies became visible. Immortalized pLCs (pLCsImt) were harvested at passage 10 and analyzed for the transient expression of human telomerase reverse transcriptase (hTERT) using real-time quantitative polymerase chain reaction (qPCR). Primary pLCs from passage 4 were used as internal hTERT gene controls. The primers used are listed in Table 1.

Table 1. Primers used for real-time quantitative PCR analysis.

SymbolGene namePrimer sequence (5’ → 3’)Accession no.Tm (℃)Product size (bp)
eGFPEnhanced green fluorescent proteinFw: AAG CAG AAG AAC GGC ATC AAX96418.155.597
Rv: GGG GGT GTT CTG CTG GTA GT61.0
hTERTHuman telomerase reverse transcriptaseFw: GCC GAG ACC AAG CAC TTC CTC GAC TNM_001193376.363.8111
Rv: GCA ACT TGC TCC AGA CAC TCT TCC G61.5
KRT5Keratin 5Fw: GCA GAT TGA GTG GAG AAG GTG TTXM_021067000.158.067
Rv: CCA GAG GAG AGG GTG TTT GTG59.1
FOXJ1Forkhead box protein 1Fw: CGC CAC AAC CTG TCT CTG AAXM_003357959.458.868
Rv: CCC TTG CCC GGC TCA T60.5
SYPSynaptophysinFw: CCT CAT CGG CTG AAT TCT TGGXM_003135078.554.761
Rv: GCC CCC ATG GAG TAG AGG AA60.4
Scgb1a1Secretoglobin family 1A member 1Fw: CAG AGG TCT GCC CGA GCT TXM_003353832.361.660
Rv: TGG CAA GTG TGC CCT TGA58.7
PDPNPodoplaninFw: TGC CAG TTC TGC TCT TCG TTTXM_005665017.357.966
Rv: CCG TGC TGG CTC CTT CTG60.8
SFTPBSurfactant protein-BFw: ACG CCC CAA CCG ATG ACNM_001102679.160.154
Rv: TGA GGA TGC TGG CGA TGT C59.0
GAPDHGlyceraldehyde-3-phosphate dehydrogenaseFw: ATG TTC CAG TAT GAT TCC ACC CAF01707955.2132
Rv: ACC AGC ATC ACC CCA TTT G56.8

bp, base pair; eGFP, enhanced green fluorescent protein; FOXJ1, forkhead box protein 1; Fw, forward; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; hTERT, human telomerase reverse transcriptase; KRT5, keratin 5; PDPN, podoplanin; Rv, reverse; Scgb1a1, secretoglobin family 1A member 1; SFTPB, surfactant protein-B; SYP, synaptophysin..



Single cell sorting by FACS

All procedures were conducted by the manufacturer’s instructions. Briefly, whole pLCsImt were index-sorted on an InvitrogenTM Bigfoot Spectral Cell Sorter (Thermo Fisher Scientific Inc., Waltham, MA, USA, #PL00304) operated by users experienced in single cell technologies. The instrument was used in a setup with a 70 μm nozzle and regular quality checks were done by running a BigfootTM Calibration Beads (Thermo Fisher Scientific Inc., Waltham, MA, USA, #PL00287). Best drop delay was determined using the BigfootTM Calibration Beads (Thermo Fisher Scientific Inc.) just before each experiments. Cells were hydrodynamically focused within a fluidic stream where single cells become encapsulated in individual droplets. To optimize the positioning of single sorted cells in the center of each well of the 96-well plate, “Run Image Alignment” was confirmed before every sorted plate. Next, single cells were sorted directly into 96-well clear bottom TC Surface plate (Thermo Fisher Scientific Inc., Waltham, MA, USA, #165306) pre-filled with PneumaCultTM-Ex Plus basal medium supplemented with 50× PneumaCultTM Ex-Plus supplement (1 cell per well). In each plate, three wells were left without sorted pLCsImt as the negative controls. The single-cell sorted pLCImt was maintained immediately after FACS sorting under 5% CO2 in a humidified incubator at 37℃. Single clones including #2, 3, 5, 6, 8, 10, 11, 16, 18, 19, 20, C2, C4, D1, and D4 were used for analyzing the expression level of the epithelial specific markers.

AAVs and in vitro transduction

All AAV serotypes (including AAV1-9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8) were purchased from VectorBuilder (VectorBuilder Inc., Chicago, IL, USA, #PANEL-AAVSP01-28), containing the enhanced green fluorescent protein (eGFP) reporter gene-driven cytomegalovirus promoter.

For in vitro transduction, pLCsImt and PAMs 3D4/31 were seeded in 24-well plates (Thermo Fisher Scientific Inc., Waltham, MA, USA, #142475) with the appropriate growth medium under 5% CO2 in the humidified incubator, which was maintained at 37℃. The medium was aspirated when the cells reached approximately 80% confluence, and transduction performed. To maximize the transduction efficiency, 250 μL of the AAV-containing growth medium was added to cells with the designated multiplicity of infection (MOI). The 24-well plate was then gently swirled to mix and cover the cells and the results incubated under 5% CO2 in the humidified incubator at 37℃ for 18 h. The AAV-containing medium was then safely removed, replaced with 500 μL fresh growth medium, and the results incubated overnight in the incubator.

Transduction efficiency

To elucidate the transduction efficiency, pLCsImt and PAMs 3D4/31 were seeded at a concentration of 5.0 × 104 cells per well in 24-well plates and incubated with 20K MOI of AAVs for 18 h. The AAV-containing medium was then replaced with fresh complete medium. Three days post-transduction, pLCsImt and PAMs 3D4/31 cells were stained with 1 μg/mL of Hoechst 33342 (Invitrogen Inc., Waltham, MA, USA, #H3570) in the growth medium for 5 min at room temperature. The staining medium was then removed and the cells washed once with DPBS containing 1× PS to remove any residual Hoechst dye. The eGFP expression levels were observed in both cell types three days following AAV transduction. A laser-scanning confocal microscope (Nikon AX, Nikon, Tokyo, Japan) was then used to visualize and photograph three random microscopic fields, with the blue-fluorescent nuclei followed by the green and red fluorescent signals. The images obtained were analyzed using ImageJ software and the total cell numbers elucidated by counting the blue-fluorescent nuclei, which was performed independently by two co-workers to ensure accuracy. The blue fluorescent images were then merged with the green (eGFP-positive cells) and red fluorescent images (tdTomato; Cas9) (Kim et al., 2024), and the transduction efficiency of the AAVs expressed as the percentage of green fluorescent cells relative to the total cell count. Data were presented as mean ± standard error of the mean.

Real-time qPCR

To assess the expression level of the eGFP mRNA (transduction), total RNA was isolated from pLCsImt and PAMs 3D4/31 five days following transduction of the AAVs using TRIzolTM Regeant (Invitrogen Inc.) and the Direct-zolTM RNA MiniPrep kit (Zymo Research Corp.) according to the manufacturer’s instructions. To further determine expression levels of epithelial specific markers such as keratin 5, forkhead box protein 1, synaptophysin, secretoglobin family 1A member 1, podoplanin, and surfactant protein-B, total RNA was isolated from single-cell sorted pLCsImt. Whole pLCsImt and porcine ear fibroblasts were used as positive and negative controls, respectively.

The quality and concentration of the extracted RNA were assessed using a NanoDrop® ND-1000 spectrophotometer (NanoDrop Technologies Inc., Wilmington, DE, USA) and the RNA stored at -80℃ until use. Synthesis of cDNA from the extracted total RNA (100 ng) was performed using the High Capacity cDNA Reverse Transcription kit (Applied Biosystems Inc., Waltham, MA, USA, #4368814), with reverse transcription performed at 37℃ for 120 min according to the manufacturer’s instructions. Specific primers were designed using Primer Express 3.01 software (Applied Biosystems Inc., Waltham, MA, USA). Details of the primer information can be seen in Table 1. Real-time qPCR was performed using the SYBR Green Master mix (Applied Biosystems Inc., Foster City, CA, USA, #4309155) and a StepOnePlus Real-Time PCR system (Applied Biosystems Inc., Foster City, CA, USA) under the following thermal-cycling procedure: 95℃ for 10 min, 40 cycles at 95℃ for 15 s, and 60℃ for 1 min (1.6℃/s ramp rate) according to the manufacturer’s instructions (Applied Biosystems Inc., Foster City, CA, USA). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal control. Gene expression was quantified via relative quantification (RQ) using the using ΔΔCt method and the data presented as the RQ. All experiments were performed at least in triplicate.

Statistical analysis

All data were analyzed for statistical significance using one-way ANOVA followed by post hoc Tukey’s HSD multiple range test for three or more groups using Statistical Products and Service Solutions (SPSS; International Business Machines SPSS Corp., Armonk, NY, USA) software (ver. 27). Error bars in the data for eGFP mRNA expression and the percentage of eGFP-positive cells represent the minimum and maximum RQ expression levels around the mean. Different letters denote significant differences between the groups. Student’s t-test was performed to compare the differences between two independent groups, allowing analysis of the hTERT expression levels. A statistical p-value of < 0.05 was considered significant in this study. All experiments were performed in triplicate.

RESULTS

AAV serotypes show different transduction efficiencies in immortalized porcine lung epithelial cells

The pLCsImt utilized in this study were generated via a meticulous process which involved the isolation and culture of primary pLCs (Supplementary Fig. 1A, left panel) from porcine lung tissue that intrinsically expresses Cas9 followed by immortalization via transduction with the hTERT gene. The resulting pLCsImt (Supplementary Fig. 1A, middle panel) exhibited high expression of the hTERT gene at passage 10 or beyond. Before the transduction of AAV serotypes in pLCsImt, we analyzed the expression related to the 6 epithelial specific markers (keratin 5, forkhead box protein 1, synaptophysin, secretoglobin family 1A member 1, podoplanin, and surfactant protein-B) in single-cell sorted pLCsImt whether they exhibit the characteristics of epithelial cells. As shown in Supplementary Fig. 2, while individual pLCImt sorted as single cell displayed relative differences in the expression of genes, it was confirmed that pLCsImt exhibit distinct epithelial cell-specific characteristics, compared to porcine ear fibroblasts. However, we used the whole pLCsImt, not the single-cell sorted pLCsImt, to investigate the AAV serotype transduction efficiency in pLCsImt.

The transduction efficiency of the 18 studied AAV serotypes (AAV1-9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8) on the pLCsImt with distinct capsid sequences was systematically compared, for which cells were infected with each AAV serotype and the visual transduction efficiency assessed three days post-infection using fluorescence microscopy. As shown in Fig. 1A, highly eGFP expression appeared three days post-infection for AAV2, AAV-DJ, and AAV2.7m8 as compared to the control, with weak green fluorescence observed for AAV1, AAV3, AAV5, AAV6, AAV6.2, AAV7, AAV8, and AAV9. AAV2-QuadYF was weakly expressed as compared to AAV2, AAV-DJ, and AAV2.7m8; however, no green fluorescence was observed in pLCsImt infected with AAV4, AAV-rh10, AAV-DJ/8, AAV-PHP.eB, AAV-PHP.S, and AAV2-retro three days post-infection. The highest percentage of 52.46 ± 2.4% was observed for the AAV2 serotype in terms of eGFP expression in the pLCsImt, with all other serotypes demonstrating much lower results in comparison, such as AAV-DJ at 22.11 ± 3.73% and AAV2.7m8 at 18.31 ± 2.14% (Fig. 1B).

Figure 1.Transduction efficiency of AAV serotypes containing eGFP on day 3 post-infection in pLCsImt. (A) Representative eGFP (green) fluorescence images in pLCsImt three days after transduction of various AAV vectors (AAV1–9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8). Individual AAV serotypes were infected at MOIs of 20 K for 18 h. The mock was not treated with the AAV vector and served as the negative control. Nuclei were stained with Hoechst 33342 (blue). Scale bars indicate 250 μm. (B) Quantification of eGFP-positive cells in different AAV serotype-infected pLCsImt. Data are presented as a mean ± standard error. Two independent co-workers counted. Different letters denote significant differences within each group, a-dp < 0.05 (n = 3). (C) Comparison with the mRNA expression level of eGFP in AAV serotype infected-pLCsImt. Relative quantification (RQ) of the gene expression was calculated using the ΔΔCt method, with data presented as RQ. Error bars represent the RQ of the minimum and maximum expression levels around the mean. All of quantification were normalized to the internal housekeeping gene GAPDH. Different letters denote significant differences within each group, a-kp < 0.05. All experiments were performed in triplicate. AAV, adeno-associated virus; eGFP, enhanced green fluorescence protein; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; MOI, multiplicity of infection; pLCsImt, immortalized porcine lung epithelial cells; RQ, relative quantification.

To further ascertain the transduction efficiency of the individual AAV serotypes, the mRNA expression levels of eGFP (all AAV vectors used in this study were eGFP-tagged) were genetically assessed using the pLCsImt (Fig. 1C). As seen in Fig. 1C, compared to the control group, the AAV2 serotype exhibited 27,215.58-fold higher eGFP expression levels, and although AAV-DJ (6,557.42-fold) and AAV2.7m8 (7,908.83-fold) showed high eGFP expression in pLCsImt, the AAV2 serotype demonstrated the highest transduction efficiency of the 18 serotypes evaluated. These results indicate that AAV2 is highly efficient for the transduction of pLCsImt and that AAV serotypes are cell type-specific.

AAV serotypes show comparable transduction efficiency in PAMs 3D4/31 to pLCsImt

To evaluate and compare the PAMs 3D4/31 (Supplementary Fig. 1A, right panel), the cells were transduced with each AAV serotype. The results were then compared with those observed for pLCsImt. Similar to pLCsImt, PAMs 3D4/31 were infected with all 18 of the AAV serotypes and the transduction efficiency evaluated. As shown in Fig. 2A, the results showed high eGFP expression for AAV2, AAV-DJ, and AAV2.7m8 as compared to the control, with weak green fluorescence observed in other serotypes, such as AAV1, AAV6, AAV6.2, and AAV2-QuadYF, and almost no eGFP expression for AAV3 and AAV5. No green fluorescence was observed in PAMs 3D4/31 infected with AAV4, AAV7, AAV8, AAV9, AAV-rh10, AAV-DJ/8, AAV-PHP.eB, AAV-PHP.S, and AAV2-retro. In particular, quantification of the eGFP expression for the AAV2 and AAV-DJ serotypes in PAMs 3D4/31 showed higher percentages of 64.08 ± 2.4% and 52.81 ± 4.73% as compared to the other AAV serotypes (Fig. 2B).

Figure 2.Transduction efficiency of individual AAV serotypes containing eGFP on day 3 post-infection in PAMs 3D4/31. (A) Representative eGFP (green) fluorescence images in PAMs 3D4/31 three days after transduction of various AAV vectors. Individual AAV serotypes were infected at MOIs of 20 K for 18 h. The mock was not treated with the AAV vector and served as the negative control. Nuclei were stained with Hoechst 33342 (blue). Scale bars indicate 250 μm. (B) Quantification of eGFP-positive cells in different AAV serotype-infected PAMs 3D4/31. Data were presented as a mean ± standard error. Two independent co-workers counted. Different letters denote significant differences within each group, a-fp < 0.05 (n = 3). (C) Comparison with the mRNA expression level of eGFP in AAV serotype infected-PAMs 3D4/31. RQ of the gene expression was calculated using the ΔΔCt method, with data presented as RQ. Error bars represent the RQ for the minimum and maximum expression levels around the mean. All of quantification were normalized to the internal housekeeping gene GAPDH. Different letters denote significant differences within each group, a-ip < 0.05. All experiments were performed in triplicate. PAMs 3D4/31, pulmonary alveolar macrophages 3D4/31.

To further determine the transduction efficiency of the various AAV serotypes, the mRNA expression levels of eGFP were also genetically assessed in PAMs 3D4/31 (Fig. 2C). As shown in Fig. 2C, compared with the control, the AAV2 serotype exhibited 1,951.98-fold higher eGFP expression levels, with the AAV-DJ serotype (1,921.74-fold) showing marginally lower eGFP expression than the AAV2 serotype, and the next highest eGFP expression levels exhibited by AAV2-QuadYF (1,341.23-fold) and AAV2.7m8 (680.99-fold). Similar to pLCsImt, AAV2 demonstrated the highest transduction efficiency for PAMs 3D4/31.

Transduction efficiency of individual AAV serotypes in porcine-derived cell types

Comprehensive analysis of the serotype transduction efficiencies for the various AAV serotypes in pLCsImt and PAMs 3D4/31 unequivocally demonstrated the highest transduction efficiency for the AAV2 serotype across both cell types (Fig. 3A, i-vi), with the AAV-DJ and AAV2.7m8 serotypes manifested sequentially lower yet substantial transduction efficiencies. In contrast, analogous transduction experiments in PK-15 cells resulted in a disparate serotype preference (Supplementary Fig. 3A and 3B), with AAV-DJ emerging as the serotype with paramount transduction efficiency, surpassing both AAV2 and AAV2.7m8. Notably, AAV2, which has previously shown superior efficacy in lung cells (Fig. 3A, i and iv), exhibited the lowest transduction efficiency among the three serotypes in renal cells. These findings indicate a marked cellular specificity for the AAV serotype transduction efficiencies, which underscores the differential affinity and transduction potential of the distinct AAV serotypes in various cellular environments. Specifically, the data showed pronounced tropism for AAV2 in both pLCsImt and PAMs 3D4/31, while the AAV-DJ serotype demonstrated a superior propensity for transduction in porcine renal cells. This cellular specificity is indicative of inherent differences in the cellular receptor availability, intracellular trafficking mechanisms, and nuclear entry pathways among the cell types examined (Fig. 3B).

Figure 3.Comparison of distinct AAV serotypes in vitro transduction efficiency in porcine. (A) Representative images showing significant eGFP expression (green) in both the pLCsImt and PAMs 3D4/31 transduced with AAV2, AAV-DJ, and AAV2.7m8 serotypes. Scale bars indicate 250 μm. (B) Schematic illustration demonstrating the cellular specificity of the various AAV serotypes assessed in this study. Individual AAV serotypes show different cellular tropism in the porcine model.

DISCUSSION

Genetically engineered pigs hold significant promise for application in both agriculture and biotechnological research. Nevertheless, their production remains challenging, costly, and time-intensive, primarily due to the lack of authentic germline-competent pluripotent stem cells (Fan and Lai, 2013). Due to these limitations, living Cas9-expressing GE pigs attracts considerable interest and substantially facilitate in vivo functional investigation of genes related disease resistance, such as PRRS, in agricultural and livestock industry (Whitworth et al., 2016; Wang et al., 2017).

AAV vectors have been rigorously investigated for their potential use in GE and biotechnological researches (Bijlani et al., 2022). Owing to their versatility, non-pathogenic characteristics, remarkable efficiency in gene delivery, and sustained persistence, AAVs have been projected to be extensively utilized across a broad range of fields (Asmamaw Mengstie, 2022; Issa et al., 2023), indicating their higher value. To date, 12 distinct AAV serotypes and over 100 natural variants have been identified (Balakrishnan and Jayandharan, 2014). Numerous AAV serotypes and variants exist, each exhibiting distinct transduction efficiencies depending on the specific cell or tissue type (Srivastava, 2016; Wiley et al., 2018; Issa et al., 2023). Several researches showed the importance of selecting the optimal AAV serotype for efficient gene delivery, particularly when targeting specific tissue including the lung (Liu et al., 2020; Yoon et al., 2021). Therefore, selecting an appropriate AAV serotype is crucial for effective AAV-based gene delivery. Our analysis of AAV recombinant serotypes using the porcine lung cells-derived from the Cas9-expressing pig (Kim et al., 2024) showed that AAV2 has the higher and superior tropism for pLCsImt. In addition to pLCsImt, AAV2 showed substantially higher transduction efficiency in PAMs 3D4/31. These findings indicate the specificity and superior efficiency of AAV2 across both cell types compared to other serotypes tested, thereby underscoring the importance of selecting the optimal AAV serotype for efficient gene delivery.

The AAV2 is capable of infiltrating and transducing various cellular phenotypes including lung across interspecies boundaries (Halbert et al., 2000; Chen et al., 2005). AAV2 has reported that it has also remarkable efficiency in neurons, hepatocytes, and mesenchymal stromal cells compared to other serotypes, such as serotypes 1, 3, 4, 5, 6, and 8 (Chng et al., 2007; Stender et al., 2007; Srivastava, 2016; Logan et al., 2017). In our study, AAV2 showed the highest transduction efficiency in both pLCsImt and PAMs 3D4/31, with eGFP expression levels of 52.46 ± 2.4% in pLCsImt and 64.08 ± 2.4% in PAMs 3D4/31 compared the others. The results firmly underscore its potential as a promising vector for the effective gene delivery in porcine lung tissues. Meanwhile, AAV-DJ and AAV2.7m8 also exhibited significant transduction capabilities in both cell types in this study, although their efficiencies were lower than that of AAV2. These results are consistent with Lisowski et al. (2015), who reported broad tropism for AAV-DJ across various tissues. AAV2.7m8, a variant of AAV2 modified to enhance its tropism (Bennett et al., 2020), also performed well, suggesting that engineered variants such as AAV2.7m8 hold promise for applications where high transduction efficiency is specifically required (Mendell et al., 2021). Interestingly, AAV-DJ exhibited relatively higher transduction efficiency than both AAV2 and AAV2.7m8 in PK-15 cells, indicating that the sensitivity of AAV varies in a cell-type-specific manner (Grimm et al., 2008; Issa et al., 2023).

In contrast, the other serotypes such as AAV4, AAV-rh10, AAV-DJ/8, AAV-PHP.eB, AAV-PHP.S, and AAV2-retro exhibited minimal to no transduction in both lung cell types in this study. In case of AAV4, although it has been reported potential as a vector for lung tissue-specific gene delivery (Issa et al., 2023), it has been reported to exhibit low overall expression levels in murine lung tissue (Zincarelli et al., 2008). In fact, AAV4 has primarily showed higher transduction efficiency in retinal cells in rodent, canine, and non-human primate (Weber et al., 2003). Furthermore, AAV-DJ/8 has been reported that it showed diverse tropisms and proclivity for liver, spleen, and neural cells (Grimm et al., 2008). In case of AAV-PHP.eB and -PHP.S, they display a predilection for the central nervous system (Chan et al., 2017). Similar with AAV-PHP.eB and -PHP.S, AAV2-retro demonstrates specific tropism for spinal neurons (Wang et al., 2018). Previous studies have also highlighted the variable efficiency of these serotypes in different tissues. These findings underscore the importance of appropriate AAV serotype selection according to target cell types once more (Su et al., 2008; Haldrup et al., 2024).

In this way, the transduction efficiency observed in tissues does not always translate reliably to isolated cell types due to the factors such as the local microenvironment, receptor expression levels, and intracellular tracking pathways, which can vary between tissues and isolated cells (Zengel et al., 2023). While our study only focused on the transduction efficiencies in Cas9-expressing pig-derived lung cells (Kim et al., 2024), further validation using isolated and purified cell types could enhance our understanding of species- and cell type-specific transduction mechanisms of AAVs (Qi et al., 2013).

However, one of the limitations of this study is the lack of assessment of the effect of pre-existing neutralizing antibodies (NAbs) on the infection efficiency of the tested AAV serotypes. Neutralizing antibodies are immune system proteins that specifically recognize and bind to viral particles, effectively preventing them from infecting cells. Pre-existing NAbs can significantly hinder the efficacy of AAV-mediated gene delivery, as they can neutralize the viral vector before it reaches the target cells (Kotterman et al., 2015; Gorovits et al., 2020). Studies have shown that NAb prevalence against AAV2 is notably high in both humans and pigs, potentially limiting its effectiveness in clinical applications (Calcedo et al., 2011; Dai et al., 2022). NAbs can vary significantly across populations and species, leading to variability in vector efficacy. This variation complicates the translation of preclinical findings to clinical settings and impacts applications in livestock health and productivity. Therefore, future research should include a comprehensive evaluation of NAbs to better predict and enhance the effectiveness of AAV-based gene therapies in both human and livestock contexts. Specifically, understanding the role of NAbs in livestock is crucial to ensure the success of gene delivery methods aimed at improving productivity, disease resistance, and overall animal health. Strategies to circumvent the challenges posed by NAbs could include engineering novel capsid variants that evade recognition by existing antibodies, using empty capsids as decoys to absorb NAbs, or employing immunosuppressive regimens to temporarily reduce antibody levels during vector administration.

Our study focuses on the application of in vivo GE technologies at enhancing disease resistance, such as PRRS, in pigs. To achieve this, we utilized the lung cells derived from Cas9-expressing pig models to identify the optimal and efficient AAV vector carrying CD163 guide RNA. In this process, we found that AAV2 exhibited considerably higher transduction efficiency in lung epithelial cells. Despite the many promising suggestions for AAV delivery systems into the lung, there are still challenges that require practical consideration. These findings from our attempt would help in applying AAV-based in vivo GE for PRRS in the future. The current study is meaningful in that AAVs with high transduction efficiencies for transgene delivery into the lung in Cas9-expressing pig were compared and determined among various AAV serotypes.

CONCLUSION

Our data indicate that AAV2 was identified as the most efficient serotype for transducing pLCsImt and PAMs 3D4/31, highlighting its potential for pulmonary GE application. Pigs have been used as a useful model and important resources in livestock industry as well as biotechnological research (Bähr and Wolf, 2012; Hryhorowicz et al., 2017; Wells et al., 2017; Clauss et al., 2019). Besides, ensuring the safety of GE is crucial, particularly minimizing immune responses and off-target effects on organs by selecting the efficient vector in AAV-mediated GE application. These findings emphasize the importance of serotype selection in gene delivery and provide a foundation for further research aimed at optimizing AAV vectors for in vivo GE application in livestock industry. The demonstrated cellular specificity of AAV transduction also calls for continued investigation of the factors influencing vector performance in different cellular contexts, paving the way for tailored GE approaches.

SUPPLEMENTARY MATERIALS

Supplementary material can be found via https://doi.org/10.12750/JARB.39.4.254

Acknowledgements

This study was supported by 2024 the RDA Fellowship Program of the National Institute of Animal Science, Rural Development Administration, Republic of Korea.

Author Contributions

Conceptualization, W.S.J. and S.K.; methodology, W.S.J. and S.K.; investigation, W.S.J., A.C., and S.K.; data curation, W.S.J., A.C., and S.K.; writing-original draft preparation, W.S.J. and S.K.; writing-review and editing, W.S.J., J-Y.L., H.L., J.N., S.L., K.O., J.G.Y., and S.K.; project administration, S.K.; funding acquisition, S.K.

Funding

This research was supported by the Cooperative Research Program for Agriculture Science and Technology Development (Project No. PJ015712) of Rural Development Administration, Republic of Korea.

Ethical Approval

Not applicable.

Consent to Participate

Not applicable.

Consent to Publish

Not applicable.

Availability of Data and Materials

Not applicable.

Conflicts of Interest

No potential conflict of interest relevant to this article was reported.

Fig 1.

Figure 1.Transduction efficiency of AAV serotypes containing eGFP on day 3 post-infection in pLCsImt. (A) Representative eGFP (green) fluorescence images in pLCsImt three days after transduction of various AAV vectors (AAV1–9, 6.2, rh10, DJ, DJ/8, PHP.eB, PHP.S, 2-retro, 2-QuadYF, and 2.7m8). Individual AAV serotypes were infected at MOIs of 20 K for 18 h. The mock was not treated with the AAV vector and served as the negative control. Nuclei were stained with Hoechst 33342 (blue). Scale bars indicate 250 μm. (B) Quantification of eGFP-positive cells in different AAV serotype-infected pLCsImt. Data are presented as a mean ± standard error. Two independent co-workers counted. Different letters denote significant differences within each group, a-dp < 0.05 (n = 3). (C) Comparison with the mRNA expression level of eGFP in AAV serotype infected-pLCsImt. Relative quantification (RQ) of the gene expression was calculated using the ΔΔCt method, with data presented as RQ. Error bars represent the RQ of the minimum and maximum expression levels around the mean. All of quantification were normalized to the internal housekeeping gene GAPDH. Different letters denote significant differences within each group, a-kp < 0.05. All experiments were performed in triplicate. AAV, adeno-associated virus; eGFP, enhanced green fluorescence protein; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; MOI, multiplicity of infection; pLCsImt, immortalized porcine lung epithelial cells; RQ, relative quantification.
Journal of Animal Reproduction and Biotechnology 2024; 39: 254-266https://doi.org/10.12750/JARB.39.4.254

Fig 2.

Figure 2.Transduction efficiency of individual AAV serotypes containing eGFP on day 3 post-infection in PAMs 3D4/31. (A) Representative eGFP (green) fluorescence images in PAMs 3D4/31 three days after transduction of various AAV vectors. Individual AAV serotypes were infected at MOIs of 20 K for 18 h. The mock was not treated with the AAV vector and served as the negative control. Nuclei were stained with Hoechst 33342 (blue). Scale bars indicate 250 μm. (B) Quantification of eGFP-positive cells in different AAV serotype-infected PAMs 3D4/31. Data were presented as a mean ± standard error. Two independent co-workers counted. Different letters denote significant differences within each group, a-fp < 0.05 (n = 3). (C) Comparison with the mRNA expression level of eGFP in AAV serotype infected-PAMs 3D4/31. RQ of the gene expression was calculated using the ΔΔCt method, with data presented as RQ. Error bars represent the RQ for the minimum and maximum expression levels around the mean. All of quantification were normalized to the internal housekeeping gene GAPDH. Different letters denote significant differences within each group, a-ip < 0.05. All experiments were performed in triplicate. PAMs 3D4/31, pulmonary alveolar macrophages 3D4/31.
Journal of Animal Reproduction and Biotechnology 2024; 39: 254-266https://doi.org/10.12750/JARB.39.4.254

Fig 3.

Figure 3.Comparison of distinct AAV serotypes in vitro transduction efficiency in porcine. (A) Representative images showing significant eGFP expression (green) in both the pLCsImt and PAMs 3D4/31 transduced with AAV2, AAV-DJ, and AAV2.7m8 serotypes. Scale bars indicate 250 μm. (B) Schematic illustration demonstrating the cellular specificity of the various AAV serotypes assessed in this study. Individual AAV serotypes show different cellular tropism in the porcine model.
Journal of Animal Reproduction and Biotechnology 2024; 39: 254-266https://doi.org/10.12750/JARB.39.4.254

Table 1 . Primers used for real-time quantitative PCR analysis.

SymbolGene namePrimer sequence (5’ → 3’)Accession no.Tm (℃)Product size (bp)
eGFPEnhanced green fluorescent proteinFw: AAG CAG AAG AAC GGC ATC AAX96418.155.597
Rv: GGG GGT GTT CTG CTG GTA GT61.0
hTERTHuman telomerase reverse transcriptaseFw: GCC GAG ACC AAG CAC TTC CTC GAC TNM_001193376.363.8111
Rv: GCA ACT TGC TCC AGA CAC TCT TCC G61.5
KRT5Keratin 5Fw: GCA GAT TGA GTG GAG AAG GTG TTXM_021067000.158.067
Rv: CCA GAG GAG AGG GTG TTT GTG59.1
FOXJ1Forkhead box protein 1Fw: CGC CAC AAC CTG TCT CTG AAXM_003357959.458.868
Rv: CCC TTG CCC GGC TCA T60.5
SYPSynaptophysinFw: CCT CAT CGG CTG AAT TCT TGGXM_003135078.554.761
Rv: GCC CCC ATG GAG TAG AGG AA60.4
Scgb1a1Secretoglobin family 1A member 1Fw: CAG AGG TCT GCC CGA GCT TXM_003353832.361.660
Rv: TGG CAA GTG TGC CCT TGA58.7
PDPNPodoplaninFw: TGC CAG TTC TGC TCT TCG TTTXM_005665017.357.966
Rv: CCG TGC TGG CTC CTT CTG60.8
SFTPBSurfactant protein-BFw: ACG CCC CAA CCG ATG ACNM_001102679.160.154
Rv: TGA GGA TGC TGG CGA TGT C59.0
GAPDHGlyceraldehyde-3-phosphate dehydrogenaseFw: ATG TTC CAG TAT GAT TCC ACC CAF01707955.2132
Rv: ACC AGC ATC ACC CCA TTT G56.8

bp, base pair; eGFP, enhanced green fluorescent protein; FOXJ1, forkhead box protein 1; Fw, forward; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; hTERT, human telomerase reverse transcriptase; KRT5, keratin 5; PDPN, podoplanin; Rv, reverse; Scgb1a1, secretoglobin family 1A member 1; SFTPB, surfactant protein-B; SYP, synaptophysin..


References

  1. Asmamaw Mengstie M. 2022. Viral vectors for the in vivo delivery of CRISPR components: advances and challenges. Front. Bioeng. Biotechnol. 12;10:895713.
    Pubmed KoreaMed CrossRef
  2. Balakrishnan B and Jayandharan GR. 2014. Basic biology of adeno-associated virus (AAV) vectors used in gene therapy. Curr. Gene Ther. 14:86-100.
    Pubmed CrossRef
  3. Bennett A, Keravala A, Makal V, Kurian J, Belbellaa B, Aeran R, Tseng YS, Sousa D, Spear J, Agbandje-McKenna M. 2020. Structure comparison of the chimeric AAV2. 7m8 vector with parental AAV2. J. Struct. Biol. 209:107433.
    Pubmed KoreaMed CrossRef
  4. Bijlani S, Pang KM, Sivanandam V, Chatterjee S. 2022. The role of recombinant AAV in precise genome editing. Front. Genome Ed. 3:799722.
    Pubmed KoreaMed CrossRef
  5. Bähr A and Wolf E. 2012. Domestic animal models for biomedical research. Reprod. Domest. Anim. 47 Suppl 4:59-71.
    Pubmed CrossRef
  6. Calcedo R, Morizono H, Wang L, McCarter R, He J, Jones D, Wilson JM. 2011. Adeno-associated virus antibody profiles in newborns, children, and adolescents. Clin. Vaccine. Immunol. 18:1586-1588.
    Pubmed KoreaMed CrossRef
  7. Calvert JG, Slade DE, Shields SL, Jolie R, Mannan RM, Welch SK. 2007. CD163 expression confers susceptibility to porcine reproductive and respiratory syndrome viruses. J. Virol. 81:7371-7379.
    Pubmed KoreaMed CrossRef
  8. Chan KY, Jang MJ, Yoo BB, Greenbaum A, Ravi N, Wu WL, Sánchez-Guardado L, Lois C, Mazmanian SK, Gradinaru V. 2017. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20:1172-1179.
    Pubmed KoreaMed CrossRef
  9. Chen CL, Jensen RL, Schnepp BC, Connell MJ, Shell R, Sferra TJ, Bartlett JS, Johnson PR. 2005. Molecular characterization of adeno-associated viruses infecting children. J. Virol. 79:14781-14792.
    Pubmed KoreaMed CrossRef
  10. Chen OG, Mather SE, Brommel CM, Hamilton BA, Ehler A, Villacreses R, Girgis RE, Abou Alaiwa M, Stoltz DA, Li X. 2021. Transduction of pig small airway epithelial cells and distal lung progenitor cells by AAV4. Cells 10:1014.
    Pubmed KoreaMed CrossRef
  11. Chng K, Larsen SR, Zhou S, Wright JF, Rasko JE. 2007. Specific adeno-associated virus serotypes facilitate efficient gene transfer into human and non-human primate mesenchymal stromal cells. J. Gene Med. 9:22-32.
    Pubmed CrossRef
  12. Cho IC, Park HB, Ahn JS, Han SH, Lee JB, Lim HT, Yoo CK, Jung EJ, Kim DH, Sun WS, Ramayo-Caldas Y, Kim SG, Kang YJ, Kim YK, Shin HS, Seong PN, Hwang IS, Park BY, Hwang S, Lee SS, Ryu YC, Lee JH, Ko MS, Lee K, Andersson G, Lee JW. 2019. A functional regulatory variant of MYH3 influences muscle fiber-type composition and intramuscular fat content in pigs. PLoS Genet 15:e1008279.
    Pubmed KoreaMed CrossRef
  13. Clauss S, Bleyer C, Schüttler D, Tomsits P, Renner S, Klymiuk N, Wakili R, Massberg S, Kääb S. 2019. Animal models of arrhythmia: classic electrophysiology to genetically modified large animals. Nat. Rev. Cardiol. 16:457-475.
    Pubmed CrossRef
  14. Cooney AL, Thornell IM, Singh BK, Shah VS, Stoltz DA, McCray PB Jr, Sinn PL. 2019. A novel AAV-mediated gene delivery system corrects CFTR function in pigs. Am. J. Respir. Cell Mol. Biol. 61:747-754.
    Pubmed KoreaMed CrossRef
  15. Dai Y, Kavita U, Lampen MH, Gielen S, Banks G, Levesque P, Kozhich A, Pillutla R, Zhang Y, Adam L. 2022. Prevalence of pre-existing neutralizing antibodies against adeno-associated virus serotypes 1, 2, 5, 6, 8, and 9 in sera of different pig strains. Hum. Gene Ther. 33:451-459.
    Pubmed CrossRef
  16. Fan N and Lai L. 2013. Genetically modified pig models for human diseases. J. Genet. Genomics 40:67-73.
    Pubmed CrossRef
  17. Gaj T, Barbas CF 3rd. 2013. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends. Biotechnol. 31:397-405.
    Pubmed KoreaMed CrossRef
  18. Gao G, Vandenberghe LH, Alvira MR, Lu Y, Calcedo R, Wilson JM. 2004. Clades of Adeno-associated viruses are widely disseminated in human tissues. J. Virol. 78:6381-6388.
    Pubmed KoreaMed CrossRef
  19. Gorovits B, Fiscella M, Havert M, Koren E, Long B, Purushothama S. 2020. Recommendations for the development of cell-based anti-viral vector neutralizing antibody assays. AAPS J. 22:24. (Erratum published 2020, AAPS. J. 22:42).
    Pubmed CrossRef
  20. Grimm D, Lee JS, Wang L, Desai T, Akache B, Kay MA. 2008. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 82:5887-5911.
    Pubmed KoreaMed CrossRef
  21. Halbert CL, Rutledge EA, Allen JM, Miller AD. 2000. Repeat transduction in the mouse lung by using adeno-associated virus vectors with different serotypes. J. Virol. 74:1524-1532.
    Pubmed KoreaMed CrossRef
  22. Haldrup SH, Fabian-Jessing BK, Jakobsen TS, Lindholm AB, Adsersen RL, Aagaard L, Bek T, Corydon TJ. 2024. Subretinal AAV delivery of RNAi-therapeutics targeting VEGFA reduces choroidal neovascularization in a large animal model. Mol. Ther. Methods Clin. Dev. 32:101242.
    Pubmed KoreaMed CrossRef
  23. Hryhorowicz M, Zeyland J, Lipiński D. 2017. Genetically modified pigs as organ donors for xenotransplantation. Mol. Biotechnol. 59:435-444.
    Pubmed KoreaMed CrossRef
  24. Issa SS, Shaimardanova AA, Rizvanov AA. 2023. Various AAV serotypes and their applications in gene therapy: an overview. Cells 12:785.
    Pubmed KoreaMed CrossRef
  25. Kim S, No JG, Lee S, Choi A, Lee JY, Hyung N, Ju WS, Lee JY, Kwak TU, Lee P, Park MR, Byun SJ, Lee H, Oh KB, Yoo JG. 2024. In vitro gene editing using primary cells derived from Cas9-expressing pigs. J. Anim. Sci. Technol. . https://doi.org/10.5187/jast.2024.e77.
    CrossRef
  26. Kotterman MA, Yin L, Strazzeri JM, Flannery JG, Schaffer DV. 2015. Antibody neutralization poses a barrier to intravitreal adeno-associated viral vector gene delivery to non-human primates. Gene Ther. 22:116-126.
    Pubmed KoreaMed CrossRef
  27. Lisowski L, Alexander IE. 2015. Adeno-associated virus serotypes for gene therapeutics. Curr. Opin. Pharmacol. 24:59-67.
    Pubmed CrossRef
  28. Liu Z, Klose K, Neuber S, Jiang M, Stamm C. 2020. Comparative analysis of adeno-associated virus serotypes for gene transfer in organotypic heart slices. J. Transl. Med. 18:437.
    Pubmed KoreaMed CrossRef
  29. Logan GJ, Dane AP, Hallwirth CV, Smyth CM, Wilkie EE, Amaya AK, Zhu E, Khandekar N, Ginn SL, Liao SHY, Cunningham SC, Sasaki N, Cabanes-Creus M, Tam PPL, Russell DW, Alexander IE. 2017. Identification of liver-specific enhancer-promoter activity in the 3' untranslated region of the wild-type AAV2 genome. Nat. Genet. 49:1267-1273.
    Pubmed CrossRef
  30. Mendell JR, Al-Zaidy SA, Rodino-Klapac LR, Goodspeed K, Gray SJ, Kay CN, Boye SL, Boye SE, George LA, Salabarria S, Corti M, Tremblay JP. 2021. Current clinical applications of in vivo gene therapy with AAVs. Mol. Ther. 29:464-488.
    Pubmed KoreaMed CrossRef
  31. Mingozzi F and High KA. 2011. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat. Rev. Genet. 12:341-355. (Erratum published 2011, Nat. Rev. Genet. 12:515).
    Pubmed CrossRef
  32. Qi YF, Li QH, Shenoy V, Zingler M, Jun JY, Verma A, Raizada MK. 2013. Comparison of the transduction efficiency of tyrosine-mutant adeno-associated virus serotype vectors in kidney. Clin. Exp. Pharmacol. Physiol. 40:53-55.
    Pubmed KoreaMed CrossRef
  33. Senís E, Fatouros C, Große S, Wiedtke E, Niopek D, Mueller AK, Grimm D. 2014. CRISPR/Cas9-mediated genome engineering: an adeno-associated viral (AAV) vector toolbox. Biotechnol. J. 9:1402-1412.
    Pubmed CrossRef
  34. Singh P and Ali SA. 2021. Impact of CRISPR-Cas9-based genome engineering in farm animals. Vet. Sci. 8:122.
    Pubmed KoreaMed CrossRef
  35. Srivastava A. 2016. In vivo tissue-tropism of adeno-associated viral vectors. Curr. Opin. Virol. 21:75-80.
    Pubmed KoreaMed CrossRef
  36. Steines B, Dickey DD, Bergen J, Excoffon KJ, Weinstein JR, Li X, Yan Z, Abou Alaiwa MH, Shah VS, Bouzek DC, Powers LS, Gansemer ND, Ostedgaard LS, Engelhardt JF, Stoltz DA, Welsh MJ, Sinn PL, Zabner J. 2016. CFTR gene transfer with AAV improves early cystic fibrosis pig phenotypes. JCI Insight 1:e88728.
    Pubmed KoreaMed CrossRef
  37. Stender S, Murphy M, O'Brien T, Stengaard C, Ulrich-Vinther M, Barry F. 2007. Adeno-associated viral vector transduction of human mesenchymal stem cells. Eur. Cell Mater. 13:93-99. discussion 99.
    Pubmed CrossRef
  38. Su H, Yeghiazarians Y, Lee A, Huang Y, Arakawa-Hoyt J, Ye J, Orcino G, Kan YW. 2008. AAV serotype 1 mediates more efficient gene transfer to pig myocardium than AAV serotype 2 and plasmid. J. Gene Med. 10:33-41.
    Pubmed CrossRef
  39. Thornton PK. 2010. Livestock production: recent trends, future prospects. Philos. Trans. R Soc. Lond. B Biol. Sci. 365:2853-2867.
    Pubmed KoreaMed CrossRef
  40. Van Gorp H, Van Breedam W, Nauwynck HJ. 2008. Sialoadhesin and CD163 join forces during entry of the porcine reproductive and respiratory syndrome virus. J. Gen. Virol. 89(Pt 12:2943-2953.
    Pubmed CrossRef
  41. Wang K, Jin Q, Ruan D, Yang Y, Liu Q, Wu H, Zhou Z, Ouyang Z, Liu Z, Zhao Y, Zhao B, Zhang Q, Peng J, Lai C, Fan N, Liang Y, Lan T, Li N, Wang X, Wang X, Fan Y, Doevendans PA, Sluijter JPG, Liu P, Lai L. 2017. Cre-dependent Cas9-expressing pigs enable efficient in vivo genome editing. Genome Res. 27:2061-2071.
    Pubmed KoreaMed CrossRef
  42. Wang Z, Maunze B, Wang Y, Blackmore MG. 2018. Global connectivity and function of descending spinal input revealed by 3D microscopy and retrograde transduction. J. Neurosci. 38:10566-10581.
    Pubmed KoreaMed CrossRef
  43. Weber M, Rabinowitz J, Provost N, Conrath H, Folliot S, Briot D, Chérel Y, Chenuaud P, Samulski J, Rolling F. 2003. Recombinant adeno-associated virus serotype 4 mediates unique and exclusive long-term transduction of retinal pigmented epithelium in rat, dog, and nonhuman primate after subretinal delivery. Mol. Ther. 7:774-781.
    Pubmed CrossRef
  44. Wells KD, Bardot R, Whitworth KM, Trible BR, Fang Y, Mileham A, Kerrigan MA, Samuel MS, Rowland RRR. 2017. Replacement of porcine CD163 scavenger receptor cysteine-rich domain 5 with a CD163-like homolog confers resistance of pigs to genotype 1 but not genotype 2 porcine reproductive and respiratory syndrome virus. J. Virol. 91:e01521-16.
    Pubmed KoreaMed CrossRef
  45. Whitworth KM, Green JA, Redel BK, Geisert RD, Lee K, Telugu BP, Prather RS. 2022. Improvements in pig agriculture through gene editing. CABI Agric. Biosci. 3:41.
    Pubmed KoreaMed CrossRef
  46. Whitworth KM, Rowland RR, Ewen CL, Trible BR, Kerrigan MA, Cino-Ozuna AG, Samuel MS, Lightner JE, McLaren DG, Mileham AJ, Prather RS. 2016. Gene-edited pigs are protected from porcine reproductive and respiratory syndrome virus. Nat. Biotechnol. 34:20-22.
    Pubmed CrossRef
  47. Wiley LA, Burnight ER, Kaalberg EE, Jiao C, Riker MJ, Halder JA, Luse MA, Han IC, Russell SR, Sohn EH, Stone EM, Mullins RF. 2018. Assessment of adeno-associated virus serotype tropism in human retinal explants. Hum. Gene Ther. 29:424-436.
    Pubmed KoreaMed CrossRef
  48. Willimann M, Tiyaboonchai A, Adachi K, Li B, Waldburger L, Nakai H, Thöny B. 2024. AAV capsid screening for translational pig research using a mouse xenograft liver model. bioRxiv. . https://doi.org/10.1101/2024.05.29.596409.
    CrossRef
  49. Yoon DS, Lee KM, Cho S, Ko EA, Kim J, Jung S, Shim JH, Gao G, Lee JW. 2021. Cellular and tissue selectivity of AAV serotypes for gene delivery to chondrocytes and cartilage. Int. J. Med. Sci. 18:3353-3360.
    Pubmed KoreaMed CrossRef
  50. Zengel J, Wang YX, Seo JW, Ning K, Hamilton JN, Wu B, Raie M, Holbrook C, Su S, Clements DR, Pillay S, Puschnik AS, Winslow MM, Idoyaga J, Nagamine CM, Sun Y, Mahajan VB, Ferrara KW, Carette JE. 2023. Hardwiring tissue-specific AAV transduction in mice through engineered receptor expression. Nat. Methods. 20:1070-1081.
    Pubmed KoreaMed CrossRef
  51. Zhang J, Wang J, Zhang X, Zhao C, Zhou S, Du C, Tan Y, Shi K. 2022. Transcriptome profiling identifies immune response genes against porcine reproductive and respiratory syndrome virus and Haemophilus parasuis co-infection in the lungs of piglets. J. Vet. Sci. 23:e2.
    CrossRef
  52. Zincarelli C, Soltys S, Rabinowitz JE. 2008. Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Mol. Ther. 16:1073-1080.
    Pubmed CrossRef

JARB Journal of Animal Reproduction and Biotehnology

qr code

OPEN ACCESS pISSN: 2671-4639
eISSN: 2671-4663