Efficient Gene Transfer to Kidney Mesenchymal Cells Using a Synthetic Adeno-Associated Viral Vector : Journal of the American Society of Nephrology

Journal Logo

Basic Research

Efficient Gene Transfer to Kidney Mesenchymal Cells Using a Synthetic Adeno-Associated Viral Vector

Ikeda, Yoichiro1; Sun, Zhao1; Ru, Xiao2; Vandenberghe, Luk H.2,3,4; Humphreys, Benjamin D.1

Author Information
Journal of the American Society of Nephrology 29(9):p 2287-2297, September 2018. | DOI: 10.1681/ASN.2018040426
  • Free
  • Infographic
  • SDC
  • SDC

Abstract

Kidney pericytes and perivascular fibroblasts are the major progenitor population for myofibroblasts during kidney fibrosis and an important therapeutic target to slow the progression of fibrotic kidney diseases.1 There are currently no antifibrotic drugs approved for the treatment of CKD, which represents a major unmet medical need.

Recombinant adeno-associated virus (AAV) is a nonintegrating, nonenveloped, and replication-deficient parvovirus that has emerged as a powerful tool for gene delivery to mammalian cells. Advantages of AAV-directed gene transfer include stable transduction in nondividing cells, low immune response, and favorable safety profile in humans. Currently, AAV-based gene therapy trials are ongoing for a range of monogenic diseases, including neuromuscular disease, hemophilia, and inherited forms of blindness.2–6 In 2017, the US Food and Drug Administration approved the first gene therapy on the basis of an AAV vector for the treatment of a form of inherited retinal degeneration.7

Broader application of AAV gene therapy is limited by incomplete understanding of viral tropism. Recent efforts to engineer AAV vectors have shown that transduction efficiency can be improved and that tropism can be modified.8 Systemic injection of AAV achieves efficient transduction in several organs but not kidney, irrespective of capsid protein.9–15 Arterial injection of AAV2 transduces the proximal tubules of the kidney but at low efficiency.16–20 Either intravenous or in utero administration of AAV9 has been reported to transduce the proximal tubules, podocytes, and glomerular endothelium,21–24 and retrograde ureteral delivery results in transduction of tubular epithelium.25 Similarly, injection of AAV2 or -8 via the retrograde ureteral or pelvic approach transduces proximal tubule.26,27 Thus, various AAVs have been shown to transduce kidney epithelia, but efficiency is generally low; they may require arterial or ureteral administration, and no protocol to date has demonstrated transduction of kidney mesenchymal cells.

In this study, we systematically evaluated different AAV serotypes for their ability to deliver genetic material to kidney pericytes and fibroblasts, the myofibroblast progenitor populations.28 We report that the synthetic AAVAnc80 efficiently transduces kidney mesenchymal cells, including pericytes, fibroblasts, and mesangial cells. We validate use of Anc80 for gene transfer by knocking out Gli2 from kidney mesenchyme using Anc80-Cre, confirming an antifibrotic effect after chronic obstruction. We implicate the β-catenin pathway in myofibroblast expansion, because knockout of mesenchymal β-catenin expression using the same strategy was also antifibrotic. Finally, we show successful mesenchymal cell transduction in human kidney organoids, suggesting that Anc80 may be useful for targeting this lineage in humans.

Methods

AAV Preparation and Injection

Native AAV-2/2 and alternative serotypes AAV5, -6, -8, and -9 and Anc80 were generated with either the CMV or the synthetic CASI promoter. Transgenes were CMV.eGFP.WPRE, CASI.eGFP.WPRE, or CASI.Cre.WPRE. AAV production and titration were performed by the Gene Transfer Vector Core (http://vector.meei.harvard.edu/) at the Grousbeck Gene Therapy Center at the Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary as described previously.8,29 Briefly, all AAV preparations were made by plasmid cotransfection in human embryonic kidney 293 cells using polyethyleneimine. The packaging plasmid AAV Rep2-CapX (X indicating the serotype) was cotransfected with an AAV2 ITR-flanked transgene and adenohelper constructs. Three days after transfections, cells and media were harvested and underwent lysis and digestion with Benzonase. Viral vector was further purified from the lysate by tangential flow filtration and iodixanol gradient ultracentrifugation before reformulation in PBS.8,29 A final volume of 100 μl per mouse was injected via the retro-orbital venous plexus.

Animal Experiments

All mouse experiments were performed according to the animal experimental guidelines issued by the Animal Care and Use Committee at Washington University. Gli2-flox (JAX #007926), Rosa26tdTomato (JAX #007909), β-catenin–flox (JAX #004152), and C57BL/6J (JAX #000664) mice were purchased from Jackson Laboratories (Bar Harbor, ME). Anesthesia was achieved with continuous evaporated isoflurane (2%) using Anesthesia System RC2 (922100; VetEqip) during surgery. Buprenorphine (0.05 mg/kg), meloxicam (1 mg/kg), and lidocaine (1%) were given subcutaneously to achieve analgesia. Unilateral ureteral obstruction (UUO) surgery was as follows. After flank incision, the right kidney was exposed and freed from the perirenal fat tissue, and the ureter was tied off at the level of the lower pole using two 4.0 silk ties. Wounds were closed by staples. Mice were euthanized 10 days after UUO surgery. UUO surgeries in C57Bl6 wild-type mice were performed at 8 weeks of age using the same technique. For characterization of kidney transduction efficiency and dose, all experiments used three mice, except for the dose of 1012 genome copies (GCs) per mouse, which used n=2 due to cost. For the Gli2 experiments, both groups had n=6. For the β-catenin and lineage-tracing experiments, both groups had n=4.

Tissue Preparation and Histology

Mice were anesthetized with isofluorane (Baxter) and subsequently perfused via the left ventricle with 4°C PBS for 1 minute. For histologic analyses, tissue sections were fixed in 10% formaldehyde for 1 hour, paraffin embedded, cut with a rotating microtome at a thickness of 3 μm, and stained according to routine histologic protocols. For immunofluorescence studies, kidneys were fixed in 4% paraformaldehyde on ice for 1 hour and then incubated in 30% sucrose in PBS at 4°C overnight. OCT-embedded (Sakura Finetek) tissues were cryosectioned into 5-μm sections and mounted on superfrost slides (Fisher Scientific). Sections were washed in 1× PBS, blocked in 10% normal goat serum (Vector Labs), and incubated with primary antibodies (Supplemental Table 1). Secondary antibodies were FITC or Cy3 conjugated (Jackson ImmunoResearch). Sections were then stained with 4′,6-diamidino-2-phenylindole and mounted in Prolong Gold (Life Technologies). All images were obtained by confocal imaging (Nikon C2+).

Real Time PCR Experiments

Tissue was harvested and immediately snap frozen in liquid nitrogen. RNA was extracted according to the manufacturer’s instructions using the RNeasy Mini Kit (Qiagen), and 600 ng of total RNA was reverse transcribed with iScript (BioRad). During the RNA extraction, DNA was removed by a DNAse digestion step (Life Technologies). Quantitative PCRs were carried out with iQ-SYBR Green Supermix (BioRad) and the Applied Biosystems 7300 Teal-Time PCR System. Cycling conditions were 95°C for 3 minutes and then 40 cycles of 95°C for 15 seconds and 60°C for 1 minute followed by one cycle of 95°C for 10 seconds. Glyceraldehyde-3-phosphate dehydrogenase was used as a housekeeping gene. Data were analyzed using the 2−ΔΔct method. Primers are listed in Supplemental Table 2.

Western Blot

Protein was extracted from tissues or cells with RIPA or urea lysis buffer (urea: 8 M) with cOmplete Mini Protease Inhibitor Cocktail (Sigma-Aldrich). Proteins samples were separated in standard SDS polyacrylamide gel, transferred to polyvinylidene difluoride membranes, and incubated with primary antibodies with 5% skim milk or 3% BSA TBST buffer for 2 hours at room temperature or overnight at 4°C followed by the reaction with secondary antibody dissolved in 5% skim milk TBST buffer for 1 hour at room temperature. Goat anti-mouse (P0447), goat anti-rabbit (P0448), or rabbit anti-goat (P0449) Igs conjugated with HRP (DAKO) were used as secondary antibodies. For developing the signal, ECL Plus Western Blotting Substrate (Pierce Biotechnology) or SuperSignal West Femto Maximal Sensitivity Substrate (Pierce Biotechnology) was used according to the manufacturer’s instruction, and the signals were detected using the ChemDoc Detection System (BioRad).

Quantification of Fluorescent Reporter Proteins

Mesangial and cortical interstitial areas were focused to quantify the fraction of fluorescent reporter proteins. At least six different areas in cortex were taken, and percentage of fluorescent protein–positive area per PDGFRβ-positive area was calculated using Nikon NIS elements.

Clonal Analyses

Five-week-old male mice were injected with a low dose of AAV (1010 GCs per mouse) followed by 3 weeks of incubation. UUO surgery was performed, and mice were euthanized 10 days after surgery; 15-μm-thick frozen sections were directly stained with 4′,6-diamidino-2-phenylindole and then analyzed using confocal microscopy. Coherent clusters of tdTomato-positive cells were counted for total cell number by DAPI counterstaining.

Statistical Analyses

Data are presented as mean±SD. Comparison of two groups was performed using unpaired t tests. Paired t tests were used for comparison of repeated measures in the same group. Statistical analyses were performed using GraphPad Prism 5.0c (GraphPad Software Inc., San Diego, CA). A P value of <0.05 was considered significant.

Results

In Vivo Kidney Transduction by AAV Variants

The capsid protein is a major determinant of cellular tropism, and we generated a panel of hybrid AAV vectors using the genome of the approved AAV serotype 2 and capsid proteins from AAV serotypes 5, 6, 8, and 9. We generated an additional AAV using the synthetic capsid protein Anc80, which has been engineered to reduce antigenic epitopes and has been shown to have unique transduction characteristics.8,30–32 All AAV serotypes contained the same CMV.eGFP.WPRE transgene construct. We reasoned that systemic delivery would be most practical for future clinical application, and therefore, all studies were conducted with intravenous administration; unless noted otherwise, we evaluated transduction 3 weeks after injection. Using a dose of 1011 GCs per mouse, we confirmed efficient transduction by AAV2/8 and AAV2/Anc80 of liver, with lower levels in heart, lung, and kidney (Figure 1A, Supplemental Figure 1). AAV2/8 and AAV2/Anc80 were the only AAV serotypes to exhibit kidney transduction, with Anc80 achieving three- to fivefold higher transduction than AAV8. GFP expression was primarily interstitial and enriched in the outer medulla.

fig1
Figure 1.:
High efficiency transduction of kidney stroma by synthetic adeno-associated virus (AAV) Anc80. (A) Only AAV serotypes 2/2 and 2/Anc80 showed any kidney tropism with eGFP expression detected in glomeruli and interstitium. Scale bar, 50 μm. (B) Costaining revealed that interstitial eGFP expression localized to PDGFRβ-positive pericytes and perivascular fibroblasts, and intraglomerular eGFP expression was observed exclusively in WT1-negative and PDGFRβ-positive mesangial cells. Scale bar, 20 μm. (C) AAV2/Anc80 also transduced a small group of epithelial cells adjacent to renin and Nos1-positive juxtaglomerular cells. Scale bar, 20 μm. (D and E) Transduction of glomerulus and interstitium by Anc80 with CMV-eGFP versus CASI-eGFP expression constructs. Scale bar, 20 μm. (F) Superior transduction detected after injection of Anc80-Cre into R26tdTomato reporter mice. Costaining of tdTomato and the endothelial marker endomucin revealed no detectable transduction of endothelium by AAV. (G) High efficincy transduction of mesangium (upper panels) and interstitial pericytes and perivascular fibroblasts (lower panels) revealed by Anc80-Cre injection into R26-tdTomato reporter mice. Scale bar, 100 μm in upper panel; 20 μm in lower panel. (H–J) Quantitation of interstitial and mesangial transduction between CMV and CASI promoters and eGFP or Cre reporters. DAPI, 4′,6-diamidino-2-phenylindole. *P<0.05 for one-way ANOVA within the group; **P<0.01 for one-way ANOVA within the group; # P<0.01 for two-way repeated ANOVA between the groups.

We further characterized Anc80 cellular tropism in the kidney by immunofluorescence staining. GFP expression was limited to interstitial, glomerular, and juxtaglomerular regions. Interstitial GFP-positive cells uniformly expressed PDGFRβ, indicating a pericyte and fibroblast identity (Figure 1B). In the glomerulus, GFP-positive cells were WT1 negative and PDGFRβ positive, consistent with mesangium (Figure 1B). The only epithelial cells transduced were always found in a tubule adjacent to the glomerulus in cells that express the Na+:K+:2Cl cotransporter Solute Carrier Family 12 Member 1, indicating the thick ascending limb. Intriguingly, only a subset of epithelia was transduced, however, and these were always cells adjacent to renin and nitric oxide synthase 1–expressing cells that themselves were not transduced (Figure 1C). These results indicate that AAV Anc80 efficiently transduces a subset of epithelia in the thick ascending limb adjacent to the juxtaglomerular apparatus (JGA).

We next asked whether the synthetic CASI promoter, consisting of a truncated CMV enhancer, chicken β-actin promoter, and UBC enhancer as well as splice donor and splice acceptor sequences, could increase sensitivity to detect transduction compared with the CMV promoter. Although overall kidney cell tropism was unchanged, we observed much better pericyte and fibroblast transduction with the CASI promoter compared with the CMV promoter (18% versus 4% of PDGFRβ-positive cells, respectively) (Figure 1, D and E). By contrast, mesangial cell transduction was somewhat reduced with the CASI promoter. Quantitation of transduction efficiencies between AAV2/2 and AAV2/Anc80 with either CMV or CASI promoters is shown in Figure 1, H and I. eGFP expression in JGA epithelia was detectable within 1 week of injection of AAV2/Anc80, whereas expression in mesangium and interstitium required 2 weeks for detection. Expression in mesangium increased from 2 to 4 weeks, whereas interstitial expression remained constant over this timeframe (Supplemental Figure 2).

Enhanced Sensitivity to Detect Gene Delivery by AAV-Cre

The fact that the CASI promoter increased apparent transduction efficiency compared with the CMV promoter suggested that we might lack adequate sensitivity to accurately measure viral transduction using a GFP reporter system. We reasoned that AAV encoding Cre recombinase in combination with a reporter mouse might provide superior sensitivity. We, therefore, generated AAV driving expression of Cre recombinase and injected 3×1011 GC per mouse into R26-tdTomato reporter mice. We observed very strong expression in all three kidney cell types (pericytes/fibroblasts, mesangial cells, and JGA), and overall transduction efficiency was far superior compared with AAV-GFP (Figure 1, F and G). Importantly, there was no transduction of peritubular endothelial cells in either the medulla (Figure 1F) or the cortex (Supplemental Figure 3). Anc80-Cre could transduce nearly 70% of interstitial pericytes and perivascular fibroblasts and nearly 90% of mesangium (Figure 1J). A dose-response study of AAV2/Anc80-Cre revealed increasing transduction through the highest dose tested (3×1011 GC per mouse) (Supplemental Figure 4).

We next investigated whether AAV causes adverse effects on uninjured kidney and other major organs. Somewhat surprisingly, our studies showed that AAV8 and the CASI.GFP.WPRE cassette (but not the CMV.GFP.WPRE cassette) led to liver toxicity at 1012 GC per mouse, including hepatocyte hypertrophy with nuclear swelling, monocellular infiltration along central veins, and bridging fibrosis (Figure 2A). It was unclear whether this toxicity was related to the vector or the expression of the transgene, but it was somewhat reflective of the previous findings on AAV8 by Wang et al.33 We also observed evidence of spontaneous myofibroblast transition in normal kidneys at the 1012 GC per mouse dose of the same AAV8 and cassette. This was evident by both upregulated expression of αSMA and immunofluorescence and increased mRNA encoding both αSMA and fibronectin (Figure 2, B–D). Of note, kidney histologic abnormalities were not detectable, and no histologic abnormalities were observed in the lungs or heart with any of the AAV serotypes tested (data not shown). It is unclear whether the kidney damage was the primary result of pericyte transduction by the highest dose of AAV8 or alternatively, a secondary effect of liver fibrosis; however, it is likely the latter, because transgene expression levels were very high in liver but lower in kidney. There was no toxicity observed with any of the other AAVs, including Anc80. In particular, we did not observe heart or lung inflammation or fibrosis 8 weeks after injection of Anc80-CASI-Cre dosed at 3×1011 GC per mouse (Supplemental Figure 5). Taken together, these results indicate that, in contrast to AAV8, Anc80 allows efficient gene transfer to kidney pericytes, mesangial cells, and JGA without toxicity.

fig2
Figure 2.:
The adeno-associated virus 2/8 (AAV2/8) exhibits liver and kidney toxicity at the highest dose. (A) AAV2/8 CASI.GFP.WPRE caused bridging fibrosis in liver at a dose of 1012 genome copies per mouse. Scale bar, 100 μM. (B) The same AAV was associated with mild myofibroblast transition at the same dose but not lower doses. Scale bar, 50 μm. (C and D) Myofibroblast differentiation quantitated by real time PCR for αSMA and fibronectin (FN) across AAV serotypes. DAPI, 4′,6-diamidino-2-phenylindole. *P<0.05.

Anc80 Transduces Myofibroblast Progenitors

Any gene therapy approach to treat kidney fibrosis requires transduction of myofibroblast progenitors. We, therefore, formally tested whether the pericytes and perivascular fibroblasts transduced by Anc80 are myofibroblast progenitors. We administered Anc80-CASI-Cre in doses ranging from 1010 to 3×1011 GCs per mouse to R26-tdTomato reporter mice and performed UUO. The lowest dose labeled about 5% of kidney pericytes, whereas 1×1011 GCs per mouse labeled 45% (Figure 3, A and B). In the UUO kidney, 15% of tdTomato-positive interstitial cells were αSMA-positive myofibroblasts at the low dose of AAV-Cre, and 60% were αSMA-positive myofibroblasts at the medium to midhigh dose. Injection of AAV-Cre at the low dose labeled isolated pericyte clones with tdTomato, and after UUO, these clones had expanded. We quantitated clonal expansion, measured an overall 4.7-fold increase in tdTomato-positive interstitial cells after UUO, and observed a right shift in the clone size distribution after UUO (Figure 3C). These results confirm that Anc80 transduces kidney myofibroblast progenitors that undergo proliferative expansion during fibrotic injury.

fig3
Figure 3.:
Anc80-Cre transduces myofibroblast progenitors. (A and B) The indicated doses of Anc80-Cre were injected into R26tdTomato mice 3 weeks before unilateral ureteral obstruction (UUO). At 10 days, interstitial tdTomato-positive cells had differentiated into αSMA-positive myofibroblasts. (C) Quantitation of myofibroblast transduction efficiencies at different doses of Anc80-Cre. (D) Clonal analysis of coherent clones at low-dose transduction shows a right shift in the clone size-frequency graph after UUO. CLK, contralateral kidney; DAPI, 4′,6-diamidino-2-phenylindole; GC, genome copy. *P<0.05.

Pericyte-Specific Knockout of Gli2 by AAV-Cre Ameliorates Kidney Fibrosis

We have previously shown that Gli2 is required for myofibroblast proliferation during kidney fibrosis.34 We next aimed to evaluate the feasibility of gene editing using AAV-Cre. We generated R26tdTomato reporter mice that were either Gli2flox/flox homozygotes or Gli2flox/wt heterozygotes and injected 3×1011 GCs per mouse of AAV-Anc80-CASI-Cre 3 weeks before UUO surgery. On the basis of tdTomato expression, we achieved a transduction efficiency of 60% of cortical pericytes and perivascular fibroblasts and observed no significant histologic abnormalities in the contralateral, uninjured kidney (Figure 4, A and B). The Gli2flox/flox homozygotes had substantially reduced fibrosis compared with the Gli2flox/wt heterozygote controls by a variety of measures. These included reduced trichrome-positive collagen area and αSMA and Col1a1 expression by immunofluorescence (Figure 3, A and B) and reduced protein expression of Col1a1 by immunofluorescence (Figure 4, C and D). There were also reduced levels of total αSMA and fibronectin by Western blot (Figure 4, E–G). Additionally, the Gli2 knockout kidneys had reduced levels of mRNA encoding Gli1, Gli2, αSMA, Col3a1, Col1a1 and fibronectin (Figure 4, H–M). Injected mice from either group did not develop histologic abnormalities in contralateral kidneys, lung, heart, or liver (data not shown). These results indicate that Anc80-Cre can efficiently mediate recombination of floxed alleles other than R26 locus in kidney stroma.

fig4
Figure 4.:
Anc80-Cre confirms a central role for Gli2 in renal fibrosis. (A and B) In total, 3×1011 genome copies per mouse of Anc80-Cre were injected into heterozygous or homozygous Gli2-flox mice. Three weeks later, unilateral ureteral obstruction (UUO) was performed, and kidneys were assessed for fibrosis 10 days later. Trichrome and αSMA staining shows reduced fibrosis in the Gli2 knockout group. (C and D) Reduced Col1a1 staining in Gli2 knockout. Scale bar, 50 μm. (E) Reduced total αSMA and fibronectin (FN) protein levels in Gli2 knockout compared with control. (F and G) Quantitation of bands by densitometry. (H–M) Levels of mRNAs encoding fibrotic readouts and mediators as measured by real time PCR. CLK, contralateral kidney; DAPI, 4′,6-diamidino-2-phenylindole; GAPDH, glyceraldehyde-3-phosphate dehydrogenase. *P<0.05; **P<0.01 compared to CLK kidney. # P<0.05 and ## P<0.01 compared to UUO Gli2 heterozygote.

Pericyte-Specific Gene Targeting of β-Catenin by AAV-Cre Ameliorates Kidney Fibrosis after UUO

Several lines of evidence have implicated canonical Wnt–β-catenin signaling in myofibroblast proliferation and differentiation during kidney fibrogenesis. This has never been tested directly, however, due to a lack of appropriate mouse models. We took advantage of Anc80-Cre tropism to test the hypothesis that canonical β-catenin signaling is necessary for renal fibrogenesis. We injected Anc80-Cre into β-cateninf/f versus β-cateninf/wt mice and subsequently induced fibrosis by UUO. Once again, 3×1011 GCs per mouse was injected 3 weeks before UUO surgery. After 10 days, mice with β-catenin ablation by Anc80-Cre had an even greater reduction in fibrosis than we observed with Gli2 knockout (Figure 5, A and B). We could detect substantially reduced levels of interstitial and total β-catenin protein as well as αSMA and fibronectin (Figure 5, C–E). Importantly, we also saw substantial reductions in a variety of fibrosis readouts as assessed by real time PCR (Figure 5, F–M). These included reductions in β-catenin targets Axin2 and Lef1, indicating successful ablation of canonical β-catenin signaling.

fig5
Figure 5.:
Deletion of β-catenin in kidney stroma by Anc80-Cre is antifibrotic. (A) Heterozygous or homozygous β-catenin–flox mice were injected with 3×1011 genome copies (GCs) per mouse of Anc80-Cre. Three weeks later, unilateral ureteral obstruction (UUO) was performed. Trichrome staining of UUO day 10 kidneys shows a strong reduction in interstitial collagen deposition in the β-catenin knockout mice. (B) Reduced myofibroblast differentiation in homozygous β-catenin knockout mice as reflected by reduced αSMA immunofluorescence. (C) Anc80-Cre reduced β-catenin protein expression in myofibroblasts as assessed by αSMA and β-catenin immunofluorescence. Scale bar, 50 μm. (D) Reduced total protein levels of β-catenin, fibronectin, and αSMA in Col1a1 staining in homozygous β-catenin–flox mice injected with Anc80-Cre compared with heterozygous β-catenin–flox mice. (E–G) Quantitation of band intensity from Western blot. Reduced total αSMA and fibronectin protein levels in Gli2 knockout compared with control. (H–M) Levels of mRNAs encoding fibrotic readouts and mediators as measured by real time PCR. *P<0.05; **P<0.01 compared to CLK. # P<0.05 and ## P<0.01 compared to UUO b-catenin heterozygote.

Analogous Tropism of Anc80 in Human Mesenchymal Cells

Mice may not predict the biology of AAV vectors in primates, and therefore, we next evaluated whether Anc80 is capable of transducing human mesenchymal cells. We first cultured adult human kidney fibroblasts and transduced them with Anc80.CASI.GFP. We observed GFP expression in PDGFRβ+ mesenchymal cells, suggesting a similar tropism of this virus in human as in mouse (Figure 6A). Because primary cells may lose differentiation marker expression in culture, we also asked whether Anc80-GFP could transduce the mesenchymal lineage in human kidney organoids generated from induced pluripotent stem cells. We observed that Anc80.CASI.GFP drove GFP expression exclusively in the interstitium of kidney organoids and that 26% of stromal marker Meis1-positive cells35 and 10% of PDGFRβ-positive cells were GFP positive (Figure 6, B–D). Although we cannot be certain of the identity of interstitial GFP-positive cells that are negative for MEIS1 and PDGFRβ, we speculate that these are mesenchymal lineage progenitors that have not yet fully differentiated, hence their lack of terminal differentiation markers, including MEIS1 and PDGFRβ. Collectively, these observations suggest that Anc80 AAV is capable of transducing the human kidney mesenchymal cell lineage.

fig6
Figure 6.:
Anc80 transduces human kidney mesenchymal cells. (A) Primary culture of adult kidney fibroblasts transduced with Anc80-eGFP. (B) Human kidney organoids exposed to Anc80-eGFP exhibit eGFP epifluorescence 4 days after transduction. (C) Costaining for stromal markers Meis1 and PDGFRβ reveals that Anc80 is capable of transducing these kidney mesenchymal cells. (D) Quantitation of transduction efficiency. DAPI, 4′,6-diamidino-2-phenylindole; GC, genome copy.

Discussion

We report three major findings. We have identified the first AAV serotype, namely Anc80, that efficiently transduces adult kidney mesenchymal cells and myofibroblast progenitors. We also show that Anc80-Cre mediates high-efficiency recombination of two separate floxed alleles, Gli2 and β-catenin, confirming important roles for these signaling pathways in mediating renal fibrosis and validating this approach in mice. Finally, we show that Anc80 also can also deliver genetic material to human mesenchymal cells in culture and in human kidney organoids, providing a proof of principle for the use of AAV-based vectors in gene therapy approaches to treat kidney fibrosis. The mechanistic basis for the enhanced transduction properties of the mesenchymal lineage in adult murine kidney is the subject of future studies, but it may relate to the rapid kinetics of expression after Anc80 infection that may enable transduction of less permissive cell targets.30,32

Our findings have applications for preclinical fibrosis models, because a major limitation of current investigations of stromal biology is the lack of high-efficiency Cre driver lines for the efficient manipulation of gene expression in adult kidney. Although FoxD1-Cre mediates recombination in nearly all kidney stroma, it is only active during nephrogenesis, and the FoxD1-CreERt2 driver is not expressed in adult kidney.36 We have used the Gli1-CreERt2 driver with success,34,37,38 but kidney expression is limited to outer medullary stroma in healthy adult kidney, with very limited expression in cortex. Thus, an important application of our findings is as a novel method for efficient, inducible gene knockout in adult kidney mesenchymal cells throughout cortex and medulla. Our approach carries the added benefit of not requiring breeding of a Cre driver allele onto the background of a floxed allele for a particular gene of interest.

These results confirm and extend important roles for Gli2 and canonical Wnt–β-catenin signaling in kidney fibrogenesis. We had already shown a role for Gli2 in myofibroblast differentiation, and here, we used this knowledge as a positive control to assess whether Anc80-Cre can delete genes in loci other than the R26 locus. Previously, tubular-specific β-catenin ablation was shown to have no antifibrotic effects on injured kidney,39 and recently, it was reported that tubule-derived Wnts are required for fibroblast activation and kidney fibrogenesis.40 However, whether interstitial stroma are the targets of tubule-derived Wnts has not been formally tested because of the lack of an inducible CreERt2 line with expression specific to kidney stroma. Our results, therefore, confirm that interstitial pericytes and fibroblasts do respond to Wnt ligands and that canonical β-catenin signaling is required for myofibroblast differentiation during fibrosis. This result is consistent with our prior demonstration that genetic stabilization of β-catenin in pericytes is sufficient to drive myofibroblast differentiation.41

Translation of these findings as a therapeutic approach to treat CKD could take several forms. AAV could be used, for example, to deliver siRNA targeting either Gli2 or β-catenin. Alternatively, it could deliver genes encoding inhibitors of these pathways, such as hedgehog-interacting protein or the secreted Wnt inhibitor Wnt inhibitory factor 1.42,43 Several hurdles, however, will need to be overcome to bring such a strategy into the clinic. Anc80 exhibits high-efficiency transduction of liver, muscle, retina, and other issues, leading to potential extrarenal toxicities from expression of hedgehog or Wnt pathway inhibitors. One potential solution is continued engineering of AAV tropism to achieve kidney specificity, and recent findings show the feasibility of such efforts.31 Alternatively, use of a kidney mesenchymal cell–specific promoter would limit extrarenal expression. Finally, although AAV supports gene expression for many months, the slowly progressive nature of CKD may present a need for repeated injections, raising the possibility of immunogenicity. Most mammals acquire humoral immunity against AAV capsids early in life, and as a consequence, they mount an immune response against conventional AAVs.8,44–47 Anc80 evokes a weaker immune response, because it is a synthetic virus, but repeated injections could lead to acquired immunity.8,30

In summary, this study evaluated five different conventional AAVs and a synthetic AAV for gene therapy applications in kidney myofibroblast progenitors. We identify Anc80 as a promising candidate for gene therapy applications targeting the kidney mesenchymal cell lineage.

Disclosures

L.H.V. is an inventor on several technologies licensed to pharmaceutical and biotechnology companies, including Anc80 technology, which was licensed to Astellas, Vivet Therapeutics, Lonza Houston, and Selecta Biosciences. L.H.V. receives research funding from and consults for Lonza Houston, Solid, and Selecta Biosciences. L.H.V. is a cofounder and equity holder of GenSight Biologics and Akouos. L.H.V. is also a member of the Scientific Advisory Board of GenSight Biologics, Akouos, and NightStarX. For all other authors, no competing financial interests exist.

Published online ahead of print. Publication date available at www.jasn.org.

This article contains supplemental material online at http://jasn.asnjournals.org/lookup/suppl/doi:10.1681/ASN.2018040426/-/DCSupplemental.

The antirenin antibody was a gift from Ariel R. Gomez.

This work was supported by Giving/Grousbeck (L.H.V.) and National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases grants DK103740 (to B.D.H.), DK107374 (to B.D.H.), DK076169 (to B.D.H.), and DK115255 (to B.D.H.).

References

1. Humphreys BD: Mechanisms of renal fibrosis. Annu Rev Physiol 80: 309–326, 201829068765
2. Bunting S, Zhang L, Xie L, Bullens S, Mahimkar R, Fong S, et al.: Gene therapy with BMN 270 results in therapeutic levels of FVIII in mice and primates and normalization of bleeding in hemophilic mice. Mol Ther 26: 496–509, 201829292164
3. Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, et al.: Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med 377: 1713–1722, 201729091557
4. Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M: Gene therapy comes of age. Science 359: eaan4672, 201829326244
5. George LA, Sullivan SK, Giermasz A, Rasko JEJ, Samelson-Jones BJ, Ducore J, et al.: Hemophilia B gene therapy with a high-specific-activity factor IX variant. N Engl J Med 377: 2215–2227, 201729211678
6. Bennett J, Wellman J, Marshall KA, McCague S, Ashtari M, DiStefano-Pappas J, et al.: Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: A follow-on phase 1 trial. Lancet 388: 661–672, 201627375040
7. Russell S, Bennett J, Wellman JA, Chung DC, Yu ZF, Tillman A, et al.: Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: A randomised, controlled, open-label, phase 3 trial. Lancet 390: 849–860, 201728712537
8. Zinn E, Pacouret S, Khaychuk V, Turunen HT, Carvalho LS, Andres-Mateos E, et al.: In silico reconstruction of the viral evolutionary lineage yields a potent gene therapy vector. Cell Reports 12: 1056–1068, 201526235624
9. Srivastava A: In vivo tissue-tropism of adeno-associated viral vectors. Curr Opin Virol 21: 75–80, 201627596608
10. Zincarelli C, Soltys S, Rengo G, Rabinowitz JE: Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Mol Ther 16: 1073–1080, 2008
11. Wu Z, Asokan A, Samulski RJ: Adeno-associated virus serotypes: Vector toolkit for human gene therapy. Mol Ther 14: 316–327, 2006
12. Qi Y, Liu X, Li H, Shenoy V, Li Q, Hauswirth WW, et al.: Selective tropism of the recombinant adeno-associated virus 9 serotype for rat cardiac tissue. J Gene Med 12: 22–34, 201019830780
13. Inagaki K, Fuess S, Storm TA, Gibson GA, McTiernan CF, Kay MA, et al.: Robust systemic transduction with AAV9 vectors in mice: Efficient global cardiac gene transfer superior to that of AAV8. Mol Ther 14: 45–53, 2006
14. Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, et al.: Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nat Med 10: 828–834, 200415273747
15. Wang Z, Zhu T, Qiao C, Zhou L, Wang B, Zhang J, et al.: Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nat Biotechnol 23: 321–328, 200515735640
16. Takeda S, Takahashi M, Mizukami H, Kobayashi E, Takeuchi K, Hakamata Y, et al.: Successful gene transfer using adeno-associated virus vectors into the kidney: Comparison among adeno-associated virus serotype 1-5 vectors in vitro and in vivo. Nephron, Exp Nephrol 96: e119–e126, 200415122061
17. Qi YF, Li QH, Shenoy V, Zingler M, Jun JY, Verma A, et al.: Comparison of the transduction efficiency of tyrosine-mutant adeno-associated virus serotype vectors in kidney. Clin Exp Pharmacol Physiol 40: 53–55, 201323216315
18. Yan JT, Wang T, Wang DW: Recombinant adeno-associated virus-mediated human kallikrein gene therapy protects against hypertensive target organ injuries through inhibiting cell apoptosis. Acta Pharmacol Sin 30: 1253–1261, 200919684610
19. Benigni A, Tomasoni S, Turka LA, Longaretti L, Zentilin L, Mister M, et al.: Adeno-associated virus-mediated CTLA4Ig gene transfer protects MHC-mismatched renal allografts from chronic rejection. J Am Soc Nephrol 17: 1665–1672, 200616641148
20. Chen S, Agarwal A, Glushakova OY, Jorgensen MS, Salgar SK, Poirier A, et al.: Gene delivery in renal tubular epithelial cells using recombinant adeno-associated viral vectors. J Am Soc Nephrol 14: 947–958, 200312660329
21. Rocca CJ, Ur SN, Harrison F, Cherqui S: rAAV9 combined with renal vein injection is optimal for kidney-targeted gene delivery: Conclusion of a comparative study. Gene Ther 21: 618–628, 201424784447
22. Picconi JL, Muff-Luett MA, Wu D, Bunchman E, Schaefer F, Brophy PD: Kidney-specific expression of GFP by in-utero delivery of pseudotyped adeno-associated virus 9. Mol Ther Methods Clin Dev 1: 14014, 201426015958
23. Schievenbusch S, Strack I, Scheffler M, Nischt R, Coutelle O, Hosel M, et al.: Combined paracrine and endocrine AAV9 mediated expression of hepatocyte growth factor for the treatment of renal fibrosis. Mol Ther 18: 1302–1309, 2010
24. Hillestad ML, Guenzel AJ, Nath KA, Barry MA: A vector-host system to fingerprint virus tropism. Hum Gene Ther 23: 1116–1126, 201222834781
25. Konkalmatt PR, Asico LD, Zhang Y, Yang Y, Drachenberg C, Zheng X, et al.: Renal rescue of dopamine D2 receptor function reverses renal injury and high blood pressure. JCI Insight 1: e85888, 201627358912
26. Chung DC, Fogelgren B, Park KM, Heidenberg J, Zuo X, Huang L, et al.: Adeno-associated virus-mediated gene transfer to renal tubule cells via a retrograde ureteral approach. Nephron Extra 1: 217–223, 201122470395
27. Ito K, Chen J, Khodadadian JJ, Vaughan ED Jr ., Lipkowitz M, Poppas DP, et al.: Adeno-associated viral vector transduction of green fluorescent protein in kidney: Effect of unilateral ureteric obstruction. BJU Int 101: 376–381, 200818184329
28. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, et al.: Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 176: 85–97, 201020008127
29. Lock M, Alvira M, Vandenberghe LH, Samanta A, Toelen J, Debyser Z, et al.: Rapid, simple, and versatile manufacturing of recombinant adeno-associated viral vectors at scale. Hum Gene Ther 21: 1259–1271, 201020497038
30. Landegger LD, Pan B, Askew C, Wassmer SJ, Gluck SD, Galvin A, et al.: A synthetic AAV vector enables safe and efficient gene transfer to the mammalian inner ear. Nat Biotechnol 35: 280–284, 201728165475
31. Carvalho LS, Xiao R, Wassmer SJ, Langsdorf A, Zinn E, Pacouret S, et al.: Synthetic adeno-associated viral vector efficiently targets mouse and nonhuman primate retina in vivo [published online ahead of print March 20, 2018]. Hum Gene Ther doi: 10.1089/hum.2017.15429325457
32. Wang L, Xiao R, Andres-Mateos E, Vandenberghe LH: Single stranded adeno-associated virus achieves efficient gene transfer to anterior segment in the mouse eye. PLoS One 12: e0182473, 201728763501
33. Wang L, Wang H, Bell P, McCarter RJ, He J, Calcedo R, et al.: Systematic evaluation of AAV vectors for liver directed gene transfer in murine models. Mol Ther 18: 118–125, 201019861950
34. Kramann R, Fleig SV, Schneider RK, Fabian SL, DiRocco DP, Maarouf O, et al.: Pharmacological GLI2 inhibition prevents myofibroblast cell-cycle progression and reduces kidney fibrosis. J Clin Invest 125: 2935–2951, 201526193634
35. Chang-Panesso M, Kadyrov FF, Machado FG, Kumar A, Humphreys BD: Meis1 is specifically upregulated in kidney myofibroblasts during aging and injury but is not required for kidney homeostasis or fibrotic response [published online ahead of print March 28, 2018]. Am J Physiol Renal Physiol doi: 10.1152/ajprenal.00030.201829592525
36. Kobayashi A, Mugford JW, Krautzberger AM, Naiman N, Liao J, McMahon AP: Identification of a multipotent self-renewing stromal progenitor population during mammalian kidney organogenesis. Stem Cell Reports 3: 650–662, 201425358792
37. Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S, Bondzie PA, et al.: Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 16: 51–66, 201525465115
38. Kramann R, Wongboonsin J, Chang-Panesso M, Machado FG, Humphreys BD: Gli1+ pericyte loss induces capillary rarefaction and proximal tubular injury. J Am Soc Nephrol 28: 776–784, 201727624490
39. Zhou D, Tan RJ, Zhou L, Li Y, Liu Y: Kidney tubular β-catenin signaling controls interstitial fibroblast fate via epithelial-mesenchymal communication. Sci Rep 3: 1878, 201323698793
40. Zhou D, Fu H, Zhang L, Zhang K, Min Y, Xiao L, et al.: Tubule-derived Wnts are required for fibroblast activation and kidney fibrosis. J Am Soc Nephrol 28: 2322–2336, 201728336721
41. DiRocco DP, Kobayashi A, Taketo MM, McMahon AP, Humphreys BD: Wnt4/β-catenin signaling in medullary kidney myofibroblasts. J Am Soc Nephrol 24: 1399–1412, 201323766539
42. Chuang PT, McMahon AP: Vertebrate Hedgehog signalling modulated by induction of a Hedgehog-binding protein. Nature 397: 617–621, 199910050855
43. Kawano Y, Kypta R: Secreted antagonists of the Wnt signalling pathway. J Cell Sci 116: 2627–2634, 200312775774
44. Mingozzi F, Maus MV, Hui DJ, Sabatino DE, Murphy SL, Rasko JE, et al.: CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat Med 13: 419–422, 200717369837
45. Erles K, Sebökovà P, Schlehofer JR: Update on the prevalence of serum antibodies (IgG and IgM) to adeno-associated virus (AAV). J Med Virol 59: 406–411, 199910502275
46. Li C, Narkbunnam N, Samulski RJ, Asokan A, Hu G, Jacobson LJ, et al.; Joint Outcome Study Investigators: Neutralizing antibodies against adeno-associated virus examined prospectively in pediatric patients with hemophilia. Gene Ther 19: 288–294, 201221697954
47. Calcedo R, Morizono H, Wang L, McCarter R, He J, Jones D, et al.: Adeno-associated virus antibody profiles in newborns, children, and adolescents. Clin Vaccine Immunol 18: 1586–1588, 201121775517
Keywords:

virology; chronic kidney disease; gene therapy

Copyright © 2018 by the American Society of Nephrology