Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Technical Report
  • Published:

A high-efficiency AAV for endothelial cell transduction throughout the central nervous system

Abstract

Endothelial cells have a crucial role in nervous system function and mounting evidence points to endothelial impairment as a major contributor to a wide range of neurological diseases; however, tools to genetically interrogate these cells in vivo remain limited. Here, we describe AAV-BI30, a capsid that specifically and efficiently transduces endothelial cells throughout the central nervous system. At relatively low systemic doses, this vector transduces the majority of arterial, capillary and venous endothelial cells in the brain, retina and spinal cord vasculature of adult C57BL/6 mice. Furthermore, we show that AAV-BI30 robustly transduces endothelial cells in multiple mouse strains and rats in vivo and human brain microvascular endothelial cells in vitro. Finally, we demonstrate the capacity of AAV-BI30 to achieve efficient and endothelial-specific Cre-mediated gene manipulation in the central nervous system. This combination of attributes makes AAV-BI30 well suited to address outstanding research questions in neurovascular biology and aid the development of therapeutics to remediate endothelial dysfunction in disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: AAV-BI30 enables efficient transduction of brain endothelial cells across species.
Fig. 2: AAV-BI30 efficiently transduces brain endothelial cells across the arterio–venous axis.
Fig. 3: AAV-BI30 targets endothelial cells throughout the retina and spinal cord vasculature.
Fig. 4: AAV-BI30 can be leveraged to achieve efficient endothelial-specific genetic manipulation.

Similar content being viewed by others

Data availability

Statistical data used to generate the graphs presented throughout the manuscript are provided in the form of source data and supplementary statistical data files. The kiCAP-AAV-BI30, NLS-GFP-miR122-WPRE-pA and pAAV-CAG-Cre-miR122-WPRE-pA plasmids generated in this study have been deposited in the Addgene repository under identification numbers 183749, 183775 and 183776, respectively.

References

  1. Deverman, B. E. et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 34, 204–209 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Körbelin, J. et al. A brain microvasculature endothelial cell-specific viral vector with the potential to treat neurovascular and neurological diseases. EMBO Mol. Med. 8, 609–625 (2016).

    PubMed  PubMed Central  Google Scholar 

  3. Tervo, D. G. R. et al. A designer AAV variant permits efficient retrograde access to projection neurons. Neuron 92, 372–382 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Hanlon, K. S. et al. Selection of an efficient AAV vector for robust CNS transgene expression. Mol. Ther. Methods Clin. Dev. 15, 320–332 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kumar, S. R. et al. Multiplexed Cre-dependent selection yields systemic AAVs for Targeting distinct brain cell types. Nat. Methods 17, 541–550 (2020).

    PubMed Central  Google Scholar 

  7. Nonnenmacher, M. et al. Rapid evolution of blood–brain-barrier-penetrating AAV capsids by RNA-driven biopanning. Mol. Ther. Methods Clin. Dev. 20, 366–378 (2020).

    PubMed  PubMed Central  Google Scholar 

  8. Sweeney, M. D., Kisler, K., Montagne, A., Toga, A. W. & Zlokovic, B. V. The role of brain vasculature in neurodegenerative disorders. Nat. Neurosci. 21, 1318–1331 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Mastorakos, P. & McGavern, D. The anatomy and immunology of vasculature in the central nervous system. Sci. Immunol. 4, eaav0492 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

    CAS  PubMed  Google Scholar 

  11. Chen, B. R., Kozberg, M. G., Bouchard, M. B., Shaik, M. A. & Hillman, E. M. A critical role for the vascular endothelium in functional neurovascular coupling in the brain. J. Am. Heart Assoc. 3, e000787 (2014).

    PubMed  PubMed Central  Google Scholar 

  12. Longden, T. A. et al. Capillary K+-sensing initiates retrograde hyperpolarization to increase local cerebral blood flow. Nat. Neurosci. 20, 717–726 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Chow, B. W. et al. Caveolae in CNS Arterioles mediate neurovascular coupling. Nature 579, 106–110 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ben-Zvi, A. et al. Mfsd2a is critical for the formation and function of the blood–brain barrier. Nature 509, 507–511 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Andreone, B. J. et al. Blood–brain barrier permeability is regulated by lipid transport-dependent suppression of caveolae-mediated transcytosis. Neuron 94, 581–594 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Chow, B. W. & Gu, C. Gradual suppression of transcytosis governs functional blood–retinal barrier formation. Neuron 93, 1325–1333 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kerfoot, S. M. & Kubes, P. Overlapping roles of P-selectin and α4 integrin to recruit leukocytes to the central nervous system in experimental autoimmune encephalomyelitis. J. Immunol. 169, 1000–1006 (2002).

    CAS  PubMed  Google Scholar 

  18. Piccio, L. et al. Molecular mechanisms involved in lymphocyte recruitment in inflamed brain microvessels: critical roles for P-selectin glycoprotein ligand-1 and heterotrimeric G(i)-linked receptors. J. Immunol. 168, 1940–1949 (2002).

    CAS  PubMed  Google Scholar 

  19. Tan, C. et al. Endothelium-derived semaphorin 3g regulates hippocampal synaptic structure and plasticity via neuropilin-2/plexinA4. Neuron 101, 920–937 (2019).

    CAS  PubMed  Google Scholar 

  20. Santisteban, M. M. et al. Endothelium-macrophage crosstalk mediates blood–brain barrier dysfunction in hypertension. Hypertension 76, 795–807 (2020).

    CAS  PubMed  Google Scholar 

  21. Liu, X. X. et al. Endothelial Cdk5 deficit leads to the development of spontaneous epilepsy through CXCL1/CXCR2-mediated reactive astrogliosis. J. Exp. Med. https://doi.org/10.1084/jem.20180992 (2020).

  22. Dogbevia, G., Grasshoff, H., Othman, A., Penno, A. & Schwaninger, M. Brain endothelial-specific gene therapy improves experimental Sandhoff disease. J. Cereb. Blood Flow Metab. 40, 1338–1350 (2020).

    CAS  PubMed  Google Scholar 

  23. Chen, D. Y. et al. Endothelium-derived semaphorin 3G attenuates ischemic retinopathy by coordinating β-catenin-dependent vascular remodeling. J. Clin. Invest. 131, e135296 (2021).

    CAS  PubMed Central  Google Scholar 

  24. Nikolakopoulou, A. M. et al. Endothelial LRP1 protects against neurodegeneration by blocking cyclophilin A. J. Exp. Med. 218, e20202207 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Cui, Y. et al. Brain endothelial PTEN/AKT/NEDD4-2/MFSD2A axis regulates blood–brain barrier permeability. Cell Rep. 36, 109327 (2021).

    CAS  PubMed  Google Scholar 

  26. Song, X. et al. Genome editing with AAV-BR1-CRISPR in postnatal mouse brain endothelial cells. Int. J. Biol. Sci. 18, 652–660 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Tabebordbar, M. et al. Directed evolution of a family of AAV capsid variants enabling potent muscle-directed gene delivery across species. Cell 184, 4919–4938 (2021).

    CAS  PubMed  Google Scholar 

  28. Huang, Q. et al. Delivering genes across the blood–brain barrier: LY6A, a novel cellular receptor for AAV-PHP.B capsids. PLoS ONE 14, e0225206 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hordeaux, J. et al. The GPI-linked protein LY6A drives AAV-PHP.B transport across the blood–brain barrier. Mol. Ther. 27, 912–921 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Lagos-Quintana, M. et al. Identification of tissue-specific microRNAs from mouse. Curr. Biol. 12, 735–739 (2002).

    CAS  PubMed  Google Scholar 

  31. Suzuki, T. et al. miR-122a-regulated expression of a suicide gene prevents hepatotoxicity without altering antitumor effects in suicide gene therapy. Mol. Ther. 16, 1719–1726 (2008).

    CAS  PubMed  Google Scholar 

  32. Xie, J. et al. MicroRNA-regulated, systemically delivered rAAV9: a step closer to CNS-restricted transgene expression. Mol. Ther. 19, 526–535 (2011).

    CAS  PubMed  Google Scholar 

  33. Geisler, A. et al. microRNA122-regulated transgene expression increases specificity of cardiac gene transfer upon intravenous delivery of AAV9 vectors. Gene Ther. 18, 199–209 (2011).

    CAS  PubMed  Google Scholar 

  34. Hordeaux, J. et al. The neurotropic properties of AAV-PHP.B are limited to C57BL/6J mice. Mol. Ther. 26, 664–668 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Matsuzaki, Y. et al. Intravenous administration of the adeno-associated virus-PHP.B capsid fails to upregulate transduction efficiency in the marmoset brain. Neurosci. Lett. 665, 182–188 (2018).

    CAS  PubMed  Google Scholar 

  36. dela Paz, N. G. & D’Amore, P. A. Arterial versus venous endothelial cells. Cell Tissue Res. 335, 5–16 (2008).

    Google Scholar 

  37. Shah, A. V., Birdsey, G. M. & Randi, A. M. Regulation of endothelial homeostasis, vascular development and angiogenesis by the transcription factor ERG. Vasc. Pharmacol. 86, 3–13 (2016).

    CAS  Google Scholar 

  38. Graßhoff, H. et al. Short regulatory DNA sequences to target brain endothelial cells for gene therapy. J. Cereb. Blood Flow Metab. https://doi.org/10.1177/0271678X211039617 (2021).

  39. Hill, R. A. et al. Regional blood flow in the normal and ischemic brain is controlled by arteriolar smooth muscle cell contractility and not by capillary pericytes. Neuron 87, 95–110 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Bartanusz, V., Jezova, D., Alajajian, B. & Digicaylioglu, M. The blood–spinal cord barrier: morphology and clinical implications. Ann. Neurol. 70, 194–206 (2011).

    PubMed  Google Scholar 

  41. Stahl, A. et al. The mouse retina as an angiogenesis model. Invest. Ophthalmol. Vis. Sci. 51, 2813–2826 (2010).

    PubMed  PubMed Central  Google Scholar 

  42. Newman, E. A. Functional hyperemia and mechanisms of neurovascular coupling in the retinal vasculature. J. Cereb. Blood Flow Metab. 33, 1685–1695 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Hobson, B. & Denekamp, J. Endothelial proliferation in tumours and normal tissues: continuous labelling studies. Br. J. Cancer 49, 405–413 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    CAS  PubMed  Google Scholar 

  45. Razani, B. et al. Caveolin-1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J. Biol. Chem. 276, 38121–38138 (2001).

    CAS  PubMed  Google Scholar 

  46. Asterholm, I. W., Mundy, D. I., Weng, J., Anderson, R. G. W. & Scherer, P. E. Altered mitochondrial function and metabolic inflexibility associated with loss of caveolin-1. Cell Metab. 15, 171–185 (2012).

    PubMed  PubMed Central  Google Scholar 

  47. Nitta, T. et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J. Cell Biol. 161, 653–660 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Stenman, J. M. et al. Canonical Wnt signaling regulates organ-specific assembly and dizfferentiation of CNS vasculature. Science 322, 1247–1250 (2008).

    CAS  PubMed  Google Scholar 

  49. Liebner, S. et al. Wnt/β-catenin signaling controls development of the blood–brain barrier. J. Cell Biol. 183, 409–417 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Daneman, R. et al. Wnt/β-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc. Natl Acad. Sci. USA 106, 641–646 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Tran, K. A. et al. Endothelial β-catenin signaling is required for maintaining adult blood–brain barrier integrity and central nervous system homeostasis. Circulation 133, 177–186 (2016).

    CAS  PubMed  Google Scholar 

  52. Kalucka, J. et al. Single-cell transcriptome atlas of murine endothelial cells. Cell 180, 764–779 (2020).

    CAS  PubMed  Google Scholar 

  53. Wang, Y. et al. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 465, 483–486 (2010).

    CAS  PubMed  Google Scholar 

  54. Kisanuki, Y. Y. et al. Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo. Dev. Biol. 230, 230–242 (2001).

    CAS  PubMed  Google Scholar 

  55. Pu, W. et al. Mfsd2a+ hepatocytes repopulate the liver during injury and regeneration. Nat. Commun. 7, 13369 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Ridder, D. A. et al. TAK1 in brain endothelial cells mediates fever and lethargy. J. Exp. Med. 208, 2615–2623 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Sawada, Y. et al. High transgene expression by lentiviral vectors causes maldevelopment of Purkinje cells in vivo. Cerebellum 9, 291–302 (2010).

    CAS  PubMed  Google Scholar 

  58. Khabou, H., Cordeau, C., Pacot, L., Fisson, S. & Dalkara, D. Dosage thresholds and influence of transgene cassette in adeno-associated virus-related toxicity. Hum. Gene Ther. 29, 1235–1241 (2018).

    CAS  PubMed  Google Scholar 

  59. Goertsen, D. et al. AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat. Neurosci. https://doi.org/10.1038/s41593-021-00969-4 (2021).

  60. Ehling, M., Adams, S., Benedito, R. & Adams, R. H. Notch controls retinal blood vessel maturation and quiescence. Development 140, 3051–3061 (2013).

    CAS  PubMed  Google Scholar 

  61. Batista, A. R. et al. Ly6a differential expression in blood–brain barrier is responsible for strain specific central nervous system transduction profile of AAV-PHP.B. Hum. Gene Ther. 31, 90–102 (2020).

    CAS  PubMed  Google Scholar 

  62. Tabebordbar, M. et al. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science 351, 407–411 (2016).

    CAS  PubMed  Google Scholar 

  63. Levy, J. M. et al. Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nat. Biomed. Eng. 4, 97–110 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Challis, R. C. et al. Systemic AAV vectors for widespread and targeted gene delivery in rodents. Nat. Protoc. 14, 379–414 (2019).

    CAS  PubMed  Google Scholar 

  65. McQuin, C. et al. CellProfiler 3.0: next-generation image processing for biology. PLoS Biol. 16, e2005970 (2018).

    PubMed  PubMed Central  Google Scholar 

  66. Goldey, G. J. et al. Removable cranial windows for long-term imaging in awake mice. Nat. Protoc. 9, 2515–2538 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. DiMattia, M. A. et al. Structural insight into the unique properties of adeno-associated virus serotype 9. J. Virol. 86, 6947–6958 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Gu and Deverman laboratories for their insightful feedback throughout the study and their comments on the manuscript during drafting. This research was supported by a National Science Foundation Graduate Research Fellowship (grant no. DGE1745303) (T.K.), an HMS Mahoney Postdoctoral Fellowship (L.K.), an award from the National Institutes of Health (NIH) Common Fund and the National Institute of Neurological Disorders and Stroke through the Somatic Cell Genome Engineering Consortium (UG3NS111689) (B.D.), a Brain Initiative award funded through the National Institute of Mental Health (UG3MH120096) (B.D.), the Stanley Center for Psychiatric Research (B.D.), a Fidelity Biosciences Research Initiative (C.G.), an Allen Distinguished Investigator Award (C.G.), an AHA-Allen Initiative in Brain Health and Cognitive Impairment Award (C.G.), NIH R35NS116820 (C.G), NIH RF1DA048786 (C.G.) and NIH R01 HL153261 (C.G.). The research of C.G. was supported in part by a Faculty Scholar grant from the Howard Hughes Medical Institute. C.G. is an investigator of the Howard Hughes Medical Institute. Imaging was in part performed in the Neurobiology Imaging Facility at Harvard Medical School. This facility is supported in part by the HMS/BCH Center for Neuroscience Research as part of an NINDS P30 Core Center grant (NS072030).

Author information

Authors and Affiliations

Authors

Contributions

T.K., K.C., C.G. and B.D. conceived the study and interpreted the results. T.K., K.C., L.K., Q.H., J.W., Q.Z., T.B., I.T., S.P., Y.C. and D.R. performed experiments. V.K. and A.C. assisted with data processing. T.K., K.C., L.K., J.W., Y.C. and B.D. prepared the figures. T.K. wrote the manuscript with input from K.C., B.D. and C.G. and editing from L.K. and Y.C. C.G. and B.D. supervised the project. All authors discussed the experiments and read and approved the manuscript.

Corresponding authors

Correspondence to Chenghua Gu or Benjamin E. Deverman.

Ethics declarations

Competing interests

K.C., Q.H. and B.D. are inventors on a provisional patent application filed by the Broad Institute (applicant). The specific aspects of the manuscript covered include modified AAV vectors and methods of making and using the same. B.D. is a scientific founder and scientific advisor of Apertura Gene Therapy. B.D. receives research funding from Apertura Gene Therapy, which was used to generate some of the data in this manuscript. B.D. is on the scientific advisory board of Tevard Biosciences. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Extended Data Fig. 1 AAV-BI30 is more efficient at transducing hCMEC/D3 cells than AAV9 across a wide range of doses.

hCMEC/D3 cells were grown to confluence in a 24-well plate format. AAV9 or AAV-BI30 carrying a CAGNLS-GFP-miR122-WPRE genome was applied to cells at 0, 500, 1,000, 5,000, 10,000, or 50,000 vg/cell. 4 days post-treatment the cells were analyzed for GFP expression via flow cytometry. The profiles show data from a single sample from each condition but are representative of n = 2 replicates.

Extended Data Fig. 2 Comparison of the tropism of AAV-BI30 and AAV9.

AAV-BI30 or AAV9 vectors carrying a CAG-NLS-GFP-WPRE genome were intravenously administered to adult C57BL/6 mice at 1 × 1011 vg/animal and transduction was assessed after 7 days. Panels show endogenous NLS-GFP transgene expression in organs throughout the body. Scale bar shown is 200 μm. Images are representative of n = 3 animals per group.

Extended Data Fig. 3 Characterization of the peripheral tropism of AAV-BI30.

AAV-BI30:CAG-NLS-GFP-miR122-WPRE was intravenously administered to adult C57BL/6 mice at 1 × 1011 vg/animal and transduction was assessed after three weeks. Representative images of AAV-BI30 transduction throughout the periphery; high-zoom colocalization of GFP with endothelial markers is shown in the rightmost column. With the notable exception of lung, AAV-BI30 rarely transduced endothelial cells in the microvasculature of peripheral organs – a striking contrast to efficient transduction seen throughout the CNS vasculature. Note residual NLS-GFP expression in hepatocytes of the liver persisting in the presence of miR122 repeats in the viral genome, illustrating AAV-BI30’s potent transduction of this cell type. Transduction observed in kidney glomeruli was non-endothelial; GFP+ cells are most likely mesangial cells. Relatively strong endothelial transduction in the interlobular vessels of the renal medulla and the aorta suggest that AAV-BI30 may achieve transduction of large-diameter arteries and veins throughout the systemic circulation. Scale bars are as follows: 100 μm in fourth column from left, 15 μm in rightmost column, and 25 μm in aorta panel. Images are representative of n = 3 animals.

Extended Data Fig. 4 AAV-BI30 transduces endothelial cells throughout the BALB/cJ and rat brain.

AAV-BI30:CAG-NLS-GFP-miR122-WPRE was intravenously administered at 1 × 1011 vg/animal (BALB/cJ; top) or 1.42 × 1013 vg/kg (rat; bottom). Transduction was assessed after three (BALB/cJ) or four (rat) weeks. Images demonstrate endothelial expression of NLS-GFP transgene throughout brains of each species. Scale bars shown are 200 μm (BALB/cJ third row from left) or 100 μm (BALB/cJ rightmost row & rat). Images are representative of n = 3 BALB/cJ mice, n = 1 rat.

Extended Data Fig. 5 Endothelial transduction by AAV-BI30 is consistent across brain regions.

AAV-BI30:CAG-NLS-GFP-miR122-WPRE was intravenously administered to adult C57BL/6 mice at 1 × 1011 vg/animal and transduction was assessed after three weeks. Images demonstrate high endothelial expression of NLS-GFP transgene throughout the brain. Region-specific endothelial transduction efficiency was as follows: 86 ± 6% in cortex, 81 ± 5% in hippocampus, 85 ± 3% in thalamus, 83 ± 2% in cerebellum (mean ± s.e.m.; n = 3 animals). Compare to 84 ± 4% efficiency measured across entire brain. Scale bars are as follows: third row from left 100 μm; rightmost row 50 μm.

Source Data

Extended Data Fig. 6 The transduction profile of AAV-BI30 within the brain is highly endothelial-specific.

AAV-BI30 or AAV-BR1 vectors carrying a CAG-NLS-GFP-miR122-WPRE genome were intravenously administered to adult C57BL/6 mice at 1 × 1011 vg/animal and transduction was assessed after three weeks. Left: Representative images of rare instances of neuronal and astrocytic transduction observed following AAV-BI30 administration at the 1 × 1011 vg/animal dose; cell types of interest are demarcated with red arrowheads. Scale bar shown is 50 μm. Right: Quantification of AAV transduction in non-endothelial (GFP+ ERG-) cells per mm2 of cortex using 18 μm sagittal sections of brain. An average of 0.6 ± 0.3 and 10.8 ± 3.0 cells / mm2 (mean ± s.e.m.; n = 3 animals per group) were identified in AAV-BI30 and AAV-BR1 administered cohorts, respectively. Consistent with previous reports, neurons constituted the majority of non-endothelial cells transduced by AAV-BR1. The data presented were compared using an unpaired, two-tailed t-test (t4 = 3.37; P = 0.0281).

Source Data

Extended Data Fig. 7 AAV-BI30 efficiently transduces the brain’s largest arteries.

AAV-BI30:CAG-NLS-GFP-miR122-WPRE was intravenously administered to an adult C57BL/6 mouse at 5 × 1011 vg/animal and transduction was assessed after 3 weeks. Robust, endothelial-specific transduction was observed throughout the cerebral arteries, Circle of Willis, and the head of the basilar artery. Scale bar shown is 100 μm. Images are representative of n = 1 mouse.

Extended Data Fig. 8 AAV-BI30-mediated gene transfer enables long-term transgene expression in CNS endothelial cells.

AAV-BI30:CAG-NLS-GFP-miR122-WPRE was intravenously administered to adult C57BL/6 mice at 1 × 1011 vg/animal and transduction was assessed after approximately 5 months (152 days). Vascular counterstains displayed are as follows: ICAM2 in brain parenchyma and spinal cord; isolectin in retina; and ERG in pia vasculature. High-zoom colocalization with GFP shown in rightmost column. Scale bars are as follows: middle row 200 μm; rightmost row 50 μm. Images are representative of n = 3 mice.

Extended Data Fig. 9 AAV-BI30-mediated Cre delivery achieves efficient endothelial-specific gene deletion.

A 1 × 1011 vg/animal dose of AAV-BI30:CAG-Cre-miR122-WPRE or saline was intravenously administered to adult Cav1fl/fl mice, and Caveolin-1 protein levels were assessed after four weeks. Representative images of the brain microvasculature show strongly reduced endothelial Caveolin-1 protein levels in AAV-BI30 injected animals. Note lack of Caveolin-1 loss in the smooth muscle cell layer of arterioles, demarcated with red arrowheads. High-zoom colocalization of Caveolin-1 and ICAM2 shown in rightmost column. Scale bars are as follows: second column from right 100 μm; rightmost column 50 μm. Images are representative of n = 3 mice per group.

Extended Data Fig. 10 Comparison of engineered AAV capsids with enhanced CNS endothelial cell transduction.

The heptameric peptides used to generate the listed capsids were uniformly inserted within the surface exposed hypervariable region VIII of the AAV sequence67. Specifically, heptamers are inserted between amino acids 587 & 588 in AAV-PPS and 588 & 589 in AAVs BI30, BR1, PHP.B, PHP.V1, and PHP.V2. AAV-PHP.eB shares AAV-PHP.B’s TLAVPFK insert and additionally contains an AQ to DG mutation at amino acids 587-588 (bracketed sequence).

Supplementary information

Supplementary Information

Supplementary Figs. 1–6

Reporting Summary

Supplementary Data 1

Statistical data for figures in Supplementary Information

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Krolak, T., Chan, K.Y., Kaplan, L. et al. A high-efficiency AAV for endothelial cell transduction throughout the central nervous system. Nat Cardiovasc Res 1, 389–400 (2022). https://doi.org/10.1038/s44161-022-00046-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-022-00046-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing