Donor and host photoreceptors engage in material transfer following transplantation of post-mitotic photoreceptor precursors

R A Pearson, A Gonzalez-Cordero, E L West, J R Ribeiro, N Aghaizu, D Goh, R D Sampson, A Georgiadis, P V Waldron, Y Duran, A Naeem, M Kloc, E Cristante, K Kruczek, K Warre-Cornish, J C Sowden, A J Smith, R R Ali, R A Pearson, A Gonzalez-Cordero, E L West, J R Ribeiro, N Aghaizu, D Goh, R D Sampson, A Georgiadis, P V Waldron, Y Duran, A Naeem, M Kloc, E Cristante, K Kruczek, K Warre-Cornish, J C Sowden, A J Smith, R R Ali

Abstract

Photoreceptor replacement by transplantation is proposed as a treatment for blindness. Transplantation of healthy photoreceptor precursor cells into diseased murine eyes leads to the presence of functional photoreceptors within host retinae that express an array of donor-specific proteins. The resulting improvement in visual function was understood to be due to donor cells integrating within host retinae. Here, however, we show that while integration occurs the majority of donor-reporter-labelled cells in the host arises as a result of material transfer between donor and host photoreceptors. Material transfer does not involve permanent donor-host nuclear or cell-cell fusion, or the uptake of free protein or nucleic acid from the extracellular environment. Instead, RNA and/or protein are exchanged between donor and host cells in vivo. These data require a re-evaluation of the mechanisms underlying rescue by photoreceptor transplantation and raise the possibility of material transfer as a strategy for the treatment of retinal disorders.

Figures

Figure 1. Real-time imaging of transplanted donor…
Figure 1. Real-time imaging of transplanted donor precursor cells migrating into host retinae.
NrlGFP post-mitotic photoreceptor precursor donor cells (green) were transplanted into 12 weeks old Prph2rd2/rd2 mice and explanted retinae were examined by real-time 2-photon fluorescence live imaging 3 days post transplantation (retinae, including donor and host cells, were acutely labelled with Mitotracker Orange CMTMRos (red) before the recording). (a), At the start of the time-lapse recordings, sub-retinally transplanted rod precursor cells were frequently observed at the level of the IPM or apical to the retinal tissue. A dashed line (white) was inserted at the presumptive level of the OLM. (b), Time course of rod precursor cell movement from the IPM into the ONL; penetration through the OLM (white dashed line) is indicated by white arrowheads (see also Supplementary Movie 1). (c,d), High magnification views of time frames shown in (b) depicting migration of the right (c) and left (d) rod precursor cell into the ONL at selected time points. Note that while GFP fluorescence gradually reduced over the imaging period, Mitotracker Orange CMTMRos labelling, which is present in both donor and host cells, persisted in its absence. Scale bars, 10 μm.
Figure 2. Host and donor photoreceptor exchange…
Figure 2. Host and donor photoreceptor exchange fluorescent reporter proteins as assessed by confocal imaging.
NrlGFP post-mitotic photoreceptor precursor donor cells (green) were transplanted into DsRed (red) hosts and examined by confocal microscopy 5-6 weeks post transplantation. Nuclei are labelled with Hoechst (blue). (a, b), histograms showing quantification of the proportion of cells analysed (n=157 cells; N=5 retinae) that expressed GFP alone (GFP+) or GFP and DsRed (GFP+/DsRed+) when measuring inner segments (a) or cell bodies (b). (c), confocal projection image of a representative host retina. Lines represent regions of interest (ROIs) through cells and inner segments shown in (dg). (d), (e) single confocal sections and the respective line plots through two inner segments and (f,g), two cell bodies shown in (c). (h,i), scatter plot (mean±s.d.) and histogram showing the position of GFP+ and GFP+/DsRed+ photoreceptors with respect to the outer limiting membrane (OLM) of host retina. The relative positions of the two populations were significantly different (P=0.0486 two tailed t-test with Welch's correction; P<0.001 2-way ANOVA). (j), confocal projection image showing donor NrlGFP+ cells in SRS of DsRed+ host. Arrows, donor GFP+ cells that are DsRed+. Scale bars 5 μm (c), 3 μm (d) and 10 μm (g).
Figure 3. Host and donor photoreceptors exchange…
Figure 3. Host and donor photoreceptors exchange fluorescent reporter proteins as determined by flow cytometry.
NrlGFP post-mitotic photoreceptor precursor donor cells were transplanted into DsRed hosts and examined by flow cytometry 5-6 weeks post transplantation. (a), box (25–75% percentile) and whiskers (min/max) plot showing median (line) % of GFP+ only and GFP+/DsRed+ photoreceptors within each host retina (N=18 retinae). ***P<0.001, paired t-test. (bd), representative flow cytometry plots for adult (b) wild-type (negative control), (c) DsRed (positive control) and (d) NrlGFP (positive control) retinae. Pink box shows gating for GFP+ cells. (e,f), representative plots from an example of a host retina showing (e) % of total retinal cells that were GFP+ (pink box) and (f) the proportion of these that were GFP+ only (left pink box) or GFP+/DsRed+ (right pink box). (g), plot showing the proportion of CD45+ cells within the GFP+ population shown in (e,f).
Figure 4. Material transfer is not the…
Figure 4. Material transfer is not the result of nuclear fusion.
(ac), representative confocal images of retinal sections from Gnat1−/− and wild-type retinae following transplantation of NrlGFP post-mitotic photoreceptor precursors (green), showing expression of Lamin B (red). GFP+ cells (arrows) within the host ONL only ever presented a single nuclear envelope (red) and nucleus (labelled with Hoechst; blue). Right hand panels show nuclei and Lamin B staining only. Scale bars 10 μm (dg), FISH for the male Y-chromosome. (d), Male and (e), female NrlGFP (green) retinal sections stained for the Y-chromosome (red), which appears as a red dot. Insert, Male GFP+ cells with Y-chromosome staining. No Y-chromosome staining was seen in the female sections (e). (f,g), examples of GFP+ cells within female host retinae 5-6 weeks post-transplantation of male NrlGFP donor cells, showing a GFP+ cell that was positive for Y-chromosome staining (f, insert) and another that was GFP+/Y-chromosome− (g, insert). Right hand panels show GFP and Y-chomosome staining only. Scale bars 25 μm
Figure 5. Material transfer permits robust exchange…
Figure 5. Material transfer permits robust exchange of an array of gene products including those genetically absent from host photoreceptors.
NrlGFP post-mitotic photoreceptor precursor donor cells were transplanted into adult Gnat1−/− hosts and immunostained for rod α-transducin 5-6 weeks post-transplantation (ac). Confocal projection images of GFP+ cells (green) in the ONL of host retinae stained for rod α-transducin (red). Nuclei were stained with Hoechst 33342 (blue). Note that in (b) the right hand panel shows the same image but with increased gain for the blue channel to show the nuclei of the RPE: No donor cells were present in the SRS overlying the GFP+/rod α-transducin+ cells. (d), Examples of GFP−/rod α-transducin+ host cells (arrow) were also seen. (e), Histogram showing the mean percentage (±s.d.) of GFP+ photoreceptors within the host ONL that were rod α-transducin+ (n=138 cells; N=4 retinae). f, rod α-transducin staining was seen in the outer segments of both GFP- and DsRed-reporter-labelled photoreceptors (arrow). Scale bars 10 μm.
Figure 6. Material transfer is specific to…
Figure 6. Material transfer is specific to photoreceptor precursors and does not result from uptake of free proteins from environment.
(ad), confocal projection images of adult retinal sections taken at 48 h, 1 week, 2weeks and 6 weeks post-injection of rEGFP and stained with anti-GFP antibody (green). Insert, Very few GFP+ cells were seen; those that were had normal rod photoreceptor morphology. (e,f) Scatter plots showing the mean (±s.d.) number of GFP+ cells within the host ONL at each time point in stained and unstained serial sections. (g), confocal projection images of retinal sections taken at 6 weeks post-injection of GFP+ fibroblasts. (h), scatter plot showing the number of GFP+ cells within the host ONL. (i), confocal projection images of retinal section taken at 6 weeks post-injection of E11.5 GFP+ retinal progenitor cells (RPCs). (j), scatter plot showing the number of GFP+ cells within the host ONL. For panels (a)-(d) and (g),(h), images from top to bottom show individual and/or combined channels for the same region of interest. Scale bars 50 μm.
Figure 7. Material transfer involves interaction between…
Figure 7. Material transfer involves interaction between donor and host photoreceptors
NrlGFP+/CD73+ (green) and DsRed+/CD73+ (red) donor cells were transplanted into adult wild-type and Gnat1−/− recipients. (ac), confocal projection images showing heterogeneous mixes of DsRed+ (closed arrow) and GFP+ (open arrow) cells within the host ONL. Occasionally, a photoreceptor within the host ONL bore both GFP and DsRed labels (asterisk). Nuclei are labelled with Hoechst (blue). (d,e), confocal projection images showing a similar finding for donor cells in the SRS. The majority expressed either GFP (open arrow) or DsRed (closed arrow), but some expressed both reporters (asterisk). For all panels, images from left to right show individual and/or combined channels for the same region of interest. Scale bars 10 μm.
Figure 8. Material transfer does not require…
Figure 8. Material transfer does not require sustained physical interaction between donor and host cells.
(a), confocal image of GFP+ cells within the host ONL immediately below the donor cell mass. Labelled user segments terminate close to the cell mass. (b), confocal image of GFP+ cells within the host ONL a significant distance away from the cell mass, with no donor cells present in the overlying SRS. (c,d), 3D reconstructions of GFP+ labelled cells in the ONL of (c) wild-type and (d) Gnat1−/− host retinae show no physical interaction with any other GFP+ cell. Scale bars 10 μm.
Figure 9. Transplantation of Cre+ photoreceptor precursors…
Figure 9. Transplantation of Cre+ photoreceptor precursors leads to widespread expression of floxed reporters in host cells.
(a), box (25–75% percentile) and whiskers (min/max) plot showing median (line) number of reporter-labelled photoreceptors in adult dTomatofloxed host retinae following transplantation. **P<0.01 2-way ANOVA. N.B. all controls (uninjected, PBS-injected and final wash) were highly significantly different from both Cre+/GFP+ and TdTom+ populations but are not denoted on graph for clarity). (bd), confocal projection images showing retinal sections after sub-retinal injection of (b) ShH10.CMV.iCre (5 × 1012 viral particles per ml−1), (c) uninjected, (d) PBS-injected, or (e) mice injected with final wash from cell preparation. (f), confocal projection images of GFP+ (green) and tdTomato+ (red) cells within the floxed tdTomato host retinae following transplantation of Crx.GFP+/Cre+ donor cells. Note that there are significantly higher numbers of tdTomato+ cells than GFP+ cells. Inset, example of a GFP+, tdTomato+ cells, surrounded by GFP−/tdTomato+ cells. (g), Immunostaining for CRE (grey) shows robust protein expression in donor cells in SRS and some weaker expression in reporter-labelled cells within host retina. (h), some cells in the SRS cell mass were tdTomato+ (arrow), indicating the potential for bidirectional material transfer. For panels (f) and (g), images from top to bottom show individual and/or combined channels for the same region of interest. (i), tdTomato+ cells were present in areas where donor cell masses were present but no GFP+ cells within the underlying host ONL. (j), few, if any, GFP+ or tdTomato+ cells were seen in retinae where donor cell mass had been rejected (N=2 retinae). Scale bars 10 μm.

References

    1. Bainbridge J. W. et al.. Effect of gene therapy on visual function in Leber's congenital amaurosis. N. Engl. J. Med. 358, 2231–2239 (2008).
    1. Bainbridge J. W. et al.. Long-term effect of gene therapy on Leber's congenital amaurosis. N. Engl. J. Med. 372, 1887–1897 (2015).
    1. Maguire A. M. et al.. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N. Engl. J. Med. 358, 2240–2248 (2008).
    1. MacLaren R. E. et al.. Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial. Lancet 383, 1129–1137 (2014).
    1. Hauswirth W. W. et al.. Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum. Gene. Ther. 19, 979–990 (2008).
    1. Weleber R. G. et al.. Results at 2 years after gene therapy for RPE65-deficient leber congenital amaurosis and severe early-childhood-onset retinal dystrophy. Ophthalmology 123, 1606–1620 (2016).
    1. Pearson R. A. Advances in repairing the degenerate retina by rod photoreceptor transplantation. Biotechnol. Adv. 32, 485–491 (2014).
    1. Pearson R. A., Hippert C., Graca A. B. & Barber A. C. Photoreceptor replacement therapy: challenges presented by the diseased recipient retinal environment. Vis. Neurosci. 31, 333–344 (2014).
    1. MacLaren R. E. et al.. Retinal repair by transplantation of photoreceptor precursors. Nature 444, 203–207 (2006).
    1. Bartsch U. et al.. Retinal cells integrate into the outer nuclear layer and differentiate into mature photoreceptors after subretinal transplantation into adult mice. Exp. Eye. Res. 86, 691–700 (2008).
    1. Eberle D. et al.. Outer segment formation of transplanted photoreceptor precursor cells. PloS ONE 7, e46305 (2012).
    1. Pearson R. A. et al.. Restoration of vision after transplantation of photoreceptors. Nature 485, 99–103 (2012).
    1. Barber A. C. et al.. Repair of the degenerate retina by photoreceptor transplantation. Proc. Natl Acad. Sci. USA 110, 354–359 (2013).
    1. Decembrini S., Koch U., Radtke F., Moulin A. & Arsenijevic Y. Derivation of traceable and transplantable photoreceptors from mouse embryonic stem cells. Stem Cell Rep. 2, 853–865 (2014).
    1. Smiley S. et al.. Establishment of a cone photoreceptor transplantation platform based on a novel cone-GFP reporter mouse line. Sci. Rep. 6, 22867 (2016).
    1. Gust J. & Reh T. A. Adult donor rod photoreceptors integrate into the mature mouse retina. Invest. Ophthalmol. Vis. Sci. 52, 5266–5272 (2011).
    1. Santos-Ferreira T. et al.. Daylight vision repair by cell transplantation. Stem Cells 33, 79–90 (2014).
    1. Lamba D. A., Gust J. & Reh T. A. Transplantation of human embryonic stem cell-derived photoreceptors restores some visual function in Crx-deficient mice. Cell Stem Cell 4, 73–79 (2009).
    1. Gonzalez-Cordero A. et al.. Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat. Biotechnol. 31, 741–747 (2013).
    1. Kemp K., Wilkins A. & Scolding N. Cell fusion in the brain: two cells forward, one cell back. Acta. Neuropathol. 128, 629–638 (2014).
    1. Johansson C. B. et al.. Extensive fusion of haematopoietic cells with Purkinje neurons in response to chronic inflammation. Nat. Cell. Biol. 10, 575–583 (2008).
    1. Weimann J. M., Johansson C. B., Trejo A. & Blau H. M. Stable reprogrammed heterokaryons form spontaneously in Purkinje neurons after bone marrow transplant. Nat. Cell. Biol. 5, 959–966 (2003).
    1. Alvarez-Dolado M. et al.. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature 425, 968–973 (2003).
    1. Harris H. & Watkins J. F. Hybrid cells derived from mouse and man: artificial heterokaryons of mammalian cells from different species. Nature 205, 640–646 (1965).
    1. Cusulin C. et al.. Embryonic stem cell-derived neural stem cells fuse with microglia and mature neurons. Stem Cells 30, 2657–2671 (2012).
    1. Breunig J., Rakic P. & Macklis J. D. Evolving methods for the labelling an mutation of postnatal neuronal precursor cells: a critical review. in Adult Neurogenesis. (eds Gage, F. H., Kempermunn, G., & Song, H.) Published by Cold Spring Hrbour Laboratory Press49–80 (2008).
    1. Warre-Cornish K., Barber A. C., Sowden J. C., Ali R. R. & Pearson R. A. Migration, integration and maturation of photoreceptor precursors following transplantation in the mouse retina. Stem. Cells. Dev. 23, 941–945 (2013).
    1. West E. L. et al.. Pharmacological disruption of the outer limiting membrane leads to increased retinal integration of transplanted photoreceptor precursors. Exp. Eye. Res. 86, 601–611 (2008).
    1. Pearson R. A. et al.. Targeted disruption of outer limiting membrane junctional proteins (Crb1 and ZO-1) increases integration of transplanted photoreceptor precursors into the adult wild-type and degenerating retina. Cell. Transplant. 19, 487–503 (2010).
    1. Akimoto M. et al.. Targeting of GFP to newborn rods by Nrl promoter and temporal expression profiling of flow-sorted photoreceptors. Proc. Natl Acad. Sci. USA 103, 3890–3895 (2006).
    1. Travis G. H., Sutcliffe J. G. & Bok D. The retinal degeneration slow (rds) gene product is a photoreceptor disc membrane-associated glycoprotein. Neuron 6, 61–70 (1991).
    1. Bianchini P. et al.. Live imaging of mammalian retina: rod outer segments are stained by conventional mitochondrial dyes. J. Biomed. Opt. 13, 054017 (2008).
    1. Uckermann O. et al.. Selective staining by vital dyes of Muller glial cells in retinal wholemounts. Glia 45, 59–66 (2004).
    1. Wu Y. et al.. Quantitative determination of spatial protein-protein correlations in fluorescence confocal microscopy. Biophys. J. 98, 493–504 (2010).
    1. Shelton K. R., Egle P. M. & Cochran D. L. Nuclear envelope proteins: identification of lamin B subtypes. Biochem. Biophys. Res. Commun. 103, 975–981 (1981).
    1. Solovei I. et al.. Nuclear architecture of rod photoreceptor cells adapts to vision in mammalian evolution. Cell 137, 356–368 (2009).
    1. West E. L. et al.. Long-term survival of photoreceptors transplanted into the adult murine neural retina requires immune modulation. Stem Cells 28, 1997–2007 (2010).
    1. Li X. et al.. Generation of destabilized green fluorescent protein as a transcription reporter. J. Biol. Chem. 273, 34970–34975 (1998).
    1. Corish P. & Tyler-Smith C. Attenuation of green fluorescent protein half-life in mammalian cells. Protein. Eng. 12, 1035–1040 (1999).
    1. West E. L. et al.. Defining the integration capacity of embryonic stem cell-derived photoreceptor precursors. Stem Cells 30, 1424–1435 (2012).
    1. Lakowski J. et al.. Transplantation of photoreceptor precursors isolated via a cell surface biomarker panel from embryonic stem cell-derived self-forming retina. Stem Cells 33, 2469–2482 (2015).
    1. Klassen H. et al.. Isolation of progenitor cells from GFP-transgenic pigs and transplantation to the retina of allorecipients. Cloning Stem. Cells. 10, 391–402 (2008).
    1. Singhal S. et al.. Chondroitin sulfate proteoglycans and microglia prevent migration and integration of grafted Muller stem cells into degenerating retina. Stem Cells 26, 1074–1082 (2008).
    1. Yao J. et al.. Robust cell integration from co-transplantation of biodegradable MMP2-PLGA microspheres with retinal progenitor cells. Biomaterials 32, 1041–1050 (2011).
    1. West E. L. et al.. Manipulation of the recipient retinal environment by ectopic expression of neurotrophic growth factors can improve transplanted photoreceptor integration and survival. Cell. Transplant. 21, 871–887 (2012).
    1. Nern C. et al.. Fusion of hematopoietic cells with Purkinje neurons does not lead to stable heterokaryon formation under noninvasive conditions. J. Neurosci 29, 3799–3807 (2009).
    1. Prockop D. J. Mitochondria to the rescue. Nat. Med. 18, 653–654 (2012).
    1. Fruhbeis C. et al.. Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 11, e1001604 (2013).
    1. Singh M. S. et al.. Reversal of end-stage retinal degeneration and restoration of visual function by photoreceptor transplantation. Proc. Natl Acad. Sci. USA 110, 1101–1106 (2013).
    1. Reh T. A. Photoreceptor transplantation in late stage retinal degeneration. Invest. Ophthalmol. Vis. Sci. 57, ORSFg1–ORSFg7 (2016).
    1. Calvert P. D. et al.. Phototransduction in transgenic mice after targeted deletion of the rod transducin alpha -subunit. Proc. Natl Acad. Sci. USA 97, 13913–13918 (2000).
    1. Lakowski J. et al.. Effective transplantation of photoreceptor precursor cells selected via cell surface antigen expression. Stem Cells 29, 1391–1404 (2011).
    1. Eberle D., Schubert S., Postel K., Corbeil D. & Ader M. Increased integration of transplanted CD73-positive photoreceptor precursors into adult mouse retina. Invest. Ophthalmol. Vis. Sci. 52, 6462–6471 (2011).
    1. Klimczak R. R., Koerber J. T., Dalkara D., Flannery J. G. & Schaffer D. V. A novel adeno-associated viral variant for efficient and selective intravitreal transduction of rat Muller cells. PloS ONE 4, e7467 (2009).
    1. Nishiguchi K. M. et al.. Gene therapy restores vision in rd1 mice after removal of a confounding mutation in Gpr179. Nat. Commun. 6, 6006 (2015).
    1. Mayginnes J. P. et al.. Quantitation of encapsidated recombinant adeno-associated virus DNA in crude cell lysates and tissue culture medium by quantitative, real-time PCR. J. Virol. Methods 137, 193–204 (2006).

Source: PubMed

3
구독하다