INTRODUCTION
Adeno-associated viruses (AAVs) are icosahedral parvoviruses with a 4.7-kb single-stranded DNA genome (
1). The AAV genome contains two alternatively spliced open reading frames:
Rep, which contains genes that facilitate AAV replication and genome packaging, and
Cap, which contains the structural proteins that form the viral capsid. The AAV genome is flanked by inverted terminal repeats (ITRs), which represent the only necessary packaging signal in
cis, thereby allowing AAV to package exogenous DNA sequences cloned between these ITRs (
1,
2).
AAV cellular entry begins with the engagement of glycan attachment factors on the cell surface (
3). Distinct AAV serotypes bind different glycans, such as sialic acid, galactose, or heparan sulfate, and capsid interaction with these diverse carbohydrates plays a key role in determining cell entry and tissue tropism (
4). AAV also requires additional attachment factors, including integrins, as well as the recently identified universal AAV receptor (AAVR; KIAA0319L) for cell entry (
5). The latter, AAVR, was identified using a haploid genetic screen as an essential factor for viral cell entry (
5). Other high-throughput screening studies using siRNA-based libraries have revealed host restriction factors involving the SUMOylation pathway, the U2 snRNP spliceosome, and DNA damage machinery (
6–8). Although Genome-Scale CRISPR Knockout (GeCKO) screens have been utilized to discover host factors enabling or restricting infection by several viruses, including influenza virus and dengue virus, GeCKO screening has not yet been applied to study AAV biology (
9,
10).
In the present study, we employ a CRISPR screen to elucidate novel host restriction factors in AAV transduction. Specifically, we report the discovery of the apical polarity determinant Crumbs 3 (Crb3) as a key restriction factor, and demonstrate that CRISPR knockout (KO) of Crb3 renders cultured hepatocytes more permissive to AAV. Further, we demonstrate that Crb3 enables sequestration of essential glycan attachment factors, but not AAVR from the cell surface. Ablation of Crb3 disrupts tight junction integrity and cell polarity resulting in mislocalization of glycans to the cell surface, allowing viral attachment and entry.
DISCUSSION
Cell polarity and tight-junction integrity in epithelial cells play important roles in cellular entry for many viruses, including hepatitis C virus and adenovirus (
26–29). Tight junctions and polarity restrict hepatitis C virus cellular entry in polarized hepatocytes by facilitating basolateral localization of receptors (
26,
27). Meanwhile, characterization of adenoviral infection of epithelial cultures has revealed that the coxsackie B and adenovirus type 2 and 5 receptor (CAR) localizes to the basolateral membrane in human airway epithelia and that tight-junction disruption was necessary for adenoviral entry (
28,
29).
In this study, we carried out a CRISPR screen to identify novel host restriction factors for AAV transduction. Our results reveal that the molecular mechanism behind restriction of AAV infection in cultured hepatocytes is driven by Crb3, a determinant of apical polarity and tight-junction integrity. Although we only utilized AAV9 in our screen, we observed that Crb3 KO increases transduction with other hepatotropic AAV serotypes such as AAV3B and AAV8. We also observed that overexpression of Crb3 overexpression renders hepatocytes more resistant to AAV transduction. Interrogation of cell polarity and tight junction integrity in Crb3 KO recapitulated previous studies demonstrating the importance of Crb3 in these pathways, with polarity marker E-cadherin showing mislocalization and tight junction components ZO-1/Occludin demonstrating marked disruption (
18).
While hepatocyte polarity has not been well investigated in the context of AAV transduction, polarized human airway epithelia have been shown to be less effectively transduced with AAV2 from the apical membrane due to reduced cellular entry (
30–32). In contrast, AAV5 has been shown to effectively transduce from the apical membrane compared to AAV2, while AAV1 has also been demonstrated to have improved apical transduction over closely related AAV6 (
33,
34). Furthermore, AAVR has recently been shown to localize to the basolateral membrane in human airway epithelia, and AAV5 has been shown to be able to transduce cells independently of AAVR (
12,
35).
With regard to hepatocytes, Crb3 KO did not impact expression or localization of AAVR but demonstrated marked overexpression of galactosylated glycans on the cell surface, which are known to be cognate attachment factors for AAV9 (
11). As visualized in
Fig. 6C and
D, the AAV9 galactose footprint (yellow) is localized at the 3-fold axis of symmetry. The AAV2i8g9 capsid was derived from an AAV2 parental capsid with AAV8 residues engrafted to ablate heparan sulfate binding (red), as well as the galactose footprint described earlier from AAV9 (yellow) (
Fig. 6E and
F). Both of these divergent capsids with galactose footprints demonstrated augmented binding in the context of Crb3 KO. These observations suggest that the increased binding can be specifically attributed to capsid recognition of relocalized galactosylated glycans on the surfaces of Crb3 KO cells.
The dichotomy in hepatocyte architecture in cell culture
in vitro and organ systems
in vivo is well documented (
36,
37). We postulate that the architecture of cultured hepatocytes limits binding of some AAV capsids by restricting access to attachment factors located intracellularly or on the basolateral membrane. Disrupting the apical polarity determinant Crb3 may facilitate enhanced binding and transduction by augmenting cell surface localization of galactosylated glycans. In contrast, hepatocytes in the liver
in vivo have an apical membrane that faces the bile canniculus, while the basolateral side faces blood flow (
36). It is tempting to speculate that this presentation of glycan attachment factors on the luminal side of hepatocytes (in conjunction with other host factors, e.g., serum proteins) enables efficient hepatocyte binding and liver transduction commonly observed with AAV vectors.
It is also noteworthy to mention that our study might inform further studies evaluating AAV transcytosis, since AAV capsids appear to transit across cellular barriers in a serotype and cell type specific manner (
32). For example, while AAV2 transduces tissue culture cells with high efficiency, AAV2 fails to transcytose across CaCo-2 (colon carcinoma) cells and MDCK (Madin-Darby kidney) cells of epithelial origin (
32). Notably, AAV5 effectively crossed Caco-2 barrier epithelial monolayers; while competitive inhibition of AAV5 engagement with sialic acid reduced transduction, it did not ablate trancytosis, suggesting that AAV5 may utilize a distinct cellular entry pathway in crossing tissue layers (
32). Previous work in our lab suggests that sialic acid binding affinity of AAV1 can be modulated by engrafting a minimal footprint of amino acid residues derived from AAVrh.10, which, in turn, can profoundly impact the ability to traverse the vascular endothelium/blood-brain barrier (
38,
39). Together, these data suggest that glycan receptor engagement and polarized receptor localization could play integral roles in determining AAV transduction versus transcellular transport profiles
in vitro and vector biodistribution
in vivo. Although outside the scope of the present study, the exact mechanism by which Crumbs 3 might modulate cell surface glycan presentation remains to be determined. Specifically, additional mechanistic studies determining whether Crb3 facilitates increased expression or relocalization of glycan attachment factors on the surface of different cell types, as well as correlation with tissue architectures
in vivo, are warranted. Nevertheless, it is plausible that this paradigm is relevant in the case of epithelial and endothelial barriers to AAV transport in general.
MATERIALS AND METHODS
Reagents.
Antibodies were as follows: mouse anti-actin (Abcam, ab3280), mouse anti-KIAA0319L(AAVR) (Abcam, ab105385), mouse anti-E-cadherin (BD Biosciences, 610182), mouse anti-occludin (Thermo Fisher, 33-1500), and rabbit anti-ZO-1 (Thermo Fisher, 61-7300). Propidium iodide (P4170) was purchased from Sigma-Aldrich, while Zombie Violet (423113) was obtained from BioLegend. Erythrina Cristigalli Lectin (ECL; FL-1141) was purchased from Vector Laboratories. Oligonucleotide qPCR primers and sgRNAs were purchased from Integrated DNA Technologies (Coralville, IA), with sequences listed in
Table 1. The Crb3 qRT-PCR primer set has been reported earlier (
40).
Cell lines.
Human hepatocarcinoma (Huh7) cells were obtained from the UNC Lineberger Tissue Culture Facility. HEK293 were obtained from the UNC Vector Core. Cells were maintained in Dulbecco modified Eagle medium supplemented with 10% fetal bovine serum (FBS) and 100 U/ml penicillin-streptomycin in 5% CO2 at 37°C.
Recombinant virus production.
Recombinant AAV vectors packaging a chicken β-actin (CBA) promoter-driven firefly luciferase cassette and self-complementary AAV (scAAV) vectors packaging a hybrid CBA (CBh) promoter driving GFP were generated using triple plasmid transfection in HEK293 cells as described previously (
23). Viral titers were obtained as previously indicated by quantitative PCR with primers against to ITRs using a Roche LightCycler 480 (Roche Applied Sciences, Pleasanton, CA) (
23).
For cloning of recombinant lentiviral cassettes, guides were annealed, phosphorylated, and ligated into BsmBI-digested lentiCRISPRv2, which was a gift from Feng Zhang (Addgene, plasmid 52961), and Crb3 cDNA was cloned into pLX304 for overexpression via Gateway Cloning, which was a gift from David Root (Addgene, plasmid 25890). Recombinant lentivirus packaging guides against Cldn15, Crb3, or Scr control guides, as well as CMV-driven overexpression of Crb3, was produced via triple plasmid transfection with psPax2 and VSVG glycoprotein for pseudotyping in HEK293 cells, as previously described (
13).
Generation of CRISPR library.
The human GeCKOv2 CRISPR knockout pooled library was a gift from Feng Zhang (Addgene, 1000000048). These cassettes were used to produce recombinant lentivirus packaging puromycin resistance, Cas9, and sgRNA constructs, as described above and previously (
13,
41). Huh7 cells were transduced with this recombinant lentivirus such that only one in three cells was infected to ensure that cells would only have a single knockout and then subjected to puromycin selection as described above.
FACS and flow cytometry.
For viable cell sorting, cells were washed twice with ice cold 1× phosphate-buffered saline (PBS), resuspended in 2% FBS plus PBS, and filtered to remove clumps. The cells were then stained for viability with propidium iodide (in the case of GFP-sorted populations) or Zombie Violet (in the case of tdTomato sorted populations). The cells were analyzed and sorted by using a FACSAria II (Becton Dickinson). For flow cytometric analysis, the cells were prepared as described above, fixed with paraformaldehyde, and filtered to remove clumps prior to analysis on a CyAn ADP (Beckman Coulter).
High-throughput sequencing and analysis.
Genomic DNA extracted from pre- and postselection CRISPR libraries were subjected to PCR-based amplification of guide sequences and indexing as previously described (
41). Libraries were sequenced with the MiSeq platform (Illumina). Guides were identified and enrichment was quantified with MaGeCK (
14), and guide enrichment data were plotted using R.
Luciferase assays.
Huh7 cells were counted and seeded overnight at equal density (3e4 cells/well) on 24-well plates. At 24 h postransduction, cells were harvested in passive lysis buffer and lysate combined with luciferin substrate from Promega (Madison, WI). Luciferase signal was then quantified by a VictorX plate reader from Perkin-Elmer (Waltham, MA).
Binding assays.
Cells were seeded overnight, prechilled at 4°C for 30 min, and incubated with rAAV-CBA-luciferase at 4°C for 1 h, followed by three washes with ice-cold 1× PBS to remove unbound virions. Then, 300 μl of ddH2O was added to each well, and the cells were subjected to three freeze-thaw cycles prior to extraction of total genomic DNA using an IBI Mini-Genomic DNA kit (IBI, Dubuque, IA). Quantification of viral genomes per cell was determined via qPCR of DNA samples with primers against the luciferase transgene and the host laminin gene.
Confocal microscopy.
Cells were seeded on slide covers in 24-well plates overnight and then fixed with 4% paraformaldehyde and permeabilized with 0.1% Triton X-100. After 30 min of blocking with 5% normal goat serum, the cells were stained with primaries, washed three times with PBS, and stained with fluorescent secondaries and DAPI (4′,6′-diamidino-2-phenylindole). After being mounted in Prolong Diamond (Invitrogen), the specimens were imaged by using a Zeiss 710 scanning confocal microscope.
Lectin staining.
Lectin staining was performed as previously described (
11). Briefly, the cells were prechilled for 30 min at 4°C and then incubated with FITC-conjugated ECL for 90 min. After three washes with ice-cold 1× PBS, the cells were imaged on EVOS, and the images were quantified with ImageJ (
42).
Structural modeling.
AAV9 and AAV2 viral protein (VP) structural coordinates were obtained from the RCSB Protein Data Bank (PDB codes
3UX1 and
1LP3) (
43,
44). Homology modeling of the AAV2i8g9 VP3 monomer was performed using the crystal structure of AAV2 VP3 as a template via the SWISS-MODEL protein structure modeling server, and structural coordinates were mapped with WinCoot (
45,
46). Three-dimensional trimer models of AAV9 and AAV2i8g9 were created using the oligomer generator function in VIPERdb-Virus Particle ExploreR2 (
47). Structural models were visualized using PyMOL. The AAV9 galactose footprint (AAV9 VP1 numbering D-271, N-272, Y-446, N-470, and W-503), as well as the engrafted galactose footprint on 2i8g9, are highlighted in yellow, while the i8 motif on 2i8g9 (AAV8 VP1 numbering 588-QQNTAP-593) is shown in blue (
11,
24,
48).
Statistical analysis.
Data represented as mean values plus and minus the SEM with n ≥ 3. A two-tailed unpaired Student t test was calculated with GraphPad Prism version 6. P values of <0.05 were considered significant. Asterisks are used to denote P values (*, P < 0.05; **, P < 0.01; ***, P < 0.005).
ACKNOWLEDGMENTS
We thank the UNC Chapel Hill High-throughput Sequencing Facility, Flow Cytometry Core and Microscopy Services Laboratory, as well as the Duke University Light Microscopy Core Facility.
V.J.M. and A.A. conceived the study, designed experiments and wrote the manuscript. V.J.M. carried out screening and viral infection experiments as well as bioinformatics analysis and transduction assays and analyzed the data. T.O.T., J.A.Y., and M.P. carried out clonal cell generation and assisted with transduction assays, as well as Western blots. S.M.-T. provided methodology and resources. A.A. provided conceptualization, writing, supervision, and funding acquisition.