Various organs, including the heart, liver, skeletal muscle, brain and spinal cord, highly efficiently after its systemic administration [24,25,36?8]. The demonstration of broad gene delivery to neurons after systemic scAAV9 injection [24,25] and the therapeutic proof-of-principle of this method in a mouse model of SMA [27?9] have paved the way for the clinical development of intravenous scAAV9 gene therapy for SMA in Europe and the USA. This study provides the first demonstration that scAAV9 can transduce ocular tissues following its intravenous injection in adult mice. One month after the injection of a scAAV9 encoding a reporter gene in eight-week-old mice, 94-09-7 transgene expression was detected in multiple layers of the retina, in the optic nerve and in the ciliary bodies. These findings suggest that scAAV9 may cross the mature blood-eye barrier, which, in adult mammalian eyes, consists of tissue layers separating the neural retina and the transparent refractive media from the circulating blood. Like the BBB, there are two main barrier systems in the eye: one essentially regulating inward movements from the blood into the eye at the level of the ciliarybody (the blood-aqueous barrier), and the other preventing outward movement from the retina into the blood (the bloodretinal barrier) [23]. We found that retinal ganglion cells were the principal cells transduced in the retina after the intravenous injection of scAAV9 in adult mice. These findings suggest that scAAV9 may be delivered to the neural retina either directly from the retinal circulation, by crossing the blood-retinal barrier, or indirectly, entering the aqueous and vitreous humors via the ciliary bodies he structural equivalent of the blood-aqueous barrier?to reach its final destination, the retinal cells. The ciliary processes and the adjacent retinal cells appeared to be strongly transduced after intravenous scAAV9 injection, suggesting that at least some of the vector passed across the tight junctions between the non pigmented cells of the ciliary epithelium. These findings are of particular importance because systemic AAV9-mediated transduction of the retina has previously been reported to be dependent on the age of the animal, with efficient transduction observed only in neonatal or fetal animals [39?2]. Such discrepancies between our data and previous work from several groups may be due to the use in our study of a selfcomplementary genome-based AAV9, or to species- differences in the vector purchase 4EGI-1 tropism. For example, Bostick et al. showed that the systemic injection of single-stranded (ss) AAV9 mediated gene transfer to the inner layer of the retina in neonatal mice, but that systemic ssAAV9 gene transfer was inefficient in adults [39], suggesting the superiority of the scAAV9 versus its single-strandedSystemic scAAV9 Gene Transfer to the RetinaSystemic scAAV9 Gene Transfer to the RetinaFigure 3. Systemic injection of AAV serotype 2 does not lead to transduction of the neural retina. GFP expression in representative cross-sections of the retina of adult mice one month after systemic administration of 2.1012 vg scAAV-GFP of serotype 9 (A ) or serotype 2 (G ) in adult mice (n = 3 per condition). GFP expression was detected in the neural retina in all mice from the serotype 9 treated-group (panel A to F are from three different animals). As expected, the highest transduction efficiency was observed at the level of the RGC layer. In contrast, no GFP expression was detected in th.Various organs, including the heart, liver, skeletal muscle, brain and spinal cord, highly efficiently after its systemic administration [24,25,36?8]. The demonstration of broad gene delivery to neurons after systemic scAAV9 injection [24,25] and the therapeutic proof-of-principle of this method in a mouse model of SMA [27?9] have paved the way for the clinical development of intravenous scAAV9 gene therapy for SMA in Europe and the USA. This study provides the first demonstration that scAAV9 can transduce ocular tissues following its intravenous injection in adult mice. One month after the injection of a scAAV9 encoding a reporter gene in eight-week-old mice, transgene expression was detected in multiple layers of the retina, in the optic nerve and in the ciliary bodies. These findings suggest that scAAV9 may cross the mature blood-eye barrier, which, in adult mammalian eyes, consists of tissue layers separating the neural retina and the transparent refractive media from the circulating blood. Like the BBB, there are two main barrier systems in the eye: one essentially regulating inward movements from the blood into the eye at the level of the ciliarybody (the blood-aqueous barrier), and the other preventing outward movement from the retina into the blood (the bloodretinal barrier) [23]. We found that retinal ganglion cells were the principal cells transduced in the retina after the intravenous injection of scAAV9 in adult mice. These findings suggest that scAAV9 may be delivered to the neural retina either directly from the retinal circulation, by crossing the blood-retinal barrier, or indirectly, entering the aqueous and vitreous humors via the ciliary bodies he structural equivalent of the blood-aqueous barrier?to reach its final destination, the retinal cells. The ciliary processes and the adjacent retinal cells appeared to be strongly transduced after intravenous scAAV9 injection, suggesting that at least some of the vector passed across the tight junctions between the non pigmented cells of the ciliary epithelium. These findings are of particular importance because systemic AAV9-mediated transduction of the retina has previously been reported to be dependent on the age of the animal, with efficient transduction observed only in neonatal or fetal animals [39?2]. Such discrepancies between our data and previous work from several groups may be due to the use in our study of a selfcomplementary genome-based AAV9, or to species- differences in the vector tropism. For example, Bostick et al. showed that the systemic injection of single-stranded (ss) AAV9 mediated gene transfer to the inner layer of the retina in neonatal mice, but that systemic ssAAV9 gene transfer was inefficient in adults [39], suggesting the superiority of the scAAV9 versus its single-strandedSystemic scAAV9 Gene Transfer to the RetinaSystemic scAAV9 Gene Transfer to the RetinaFigure 3. Systemic injection of AAV serotype 2 does not lead to transduction of the neural retina. GFP expression in representative cross-sections of the retina of adult mice one month after systemic administration of 2.1012 vg scAAV-GFP of serotype 9 (A ) or serotype 2 (G ) in adult mice (n = 3 per condition). GFP expression was detected in the neural retina in all mice from the serotype 9 treated-group (panel A to F are from three different animals). As expected, the highest transduction efficiency was observed at the level of the RGC layer. In contrast, no GFP expression was detected in th.