Non-invasive stem cell therapy in a rat model for retinal degeneration and vascular pathology

Shaomei Wang, Bin Lu, Sergei Girman, Jie Duan, Trevor McFarland, Qing-shuo Zhang, Markus Grompe, Grazyna Adamus, Binoy Appukuttan, Raymond Lund, Shaomei Wang, Bin Lu, Sergei Girman, Jie Duan, Trevor McFarland, Qing-shuo Zhang, Markus Grompe, Grazyna Adamus, Binoy Appukuttan, Raymond Lund

Abstract

Background: Retinitis pigmentosa (RP) is characterized by progressive night blindness, visual field loss, altered vascular permeability and loss of central vision. Currently there is no effective treatment available except gene replacement therapy has shown promise in a few patients with specific gene defects. There is an urgent need to develop therapies that offer generic neuro-and vascular-protective effects with non-invasive intervention. Here we explored the potential of systemic administration of pluripotent bone marrow-derived mesenchymal stem cells (MSCs) to rescue vision and associated vascular pathology in the Royal College Surgeons (RCS) rat, a well-established animal model for RP.

Methodology/principal findings: Animals received syngeneic MSCs (1x10(6) cells) by tail vein at an age before major photoreceptor loss.

Principal results: both rod and cone photoreceptors were preserved (5-6 cells thick) at the time when control animal has a single layer of photoreceptors remained; Visual function was significantly preserved compared with controls as determined by visual acuity and luminance threshold recording from the superior colliculus; The number of pathological vascular complexes (abnormal vessels associated with migrating pigment epithelium cells) and area of vascular leakage that would ordinarily develop were dramatically reduced; Semi-quantitative RT-PCR analysis indicated there was upregulation of growth factors and immunohistochemistry revealed that there was an increase in neurotrophic factors within eyes of animals that received MSCs.

Conclusions/significance: These results underscore the potential application of MSCs in treating retinal degeneration. The advantages of this non-invasive cell-based therapy are: cells are easily isolated and can be expanded in large quantity for autologous graft; hypoimmunogenic nature as allogeneic donors; less controversial in nature than other stem cells; can be readministered with minor discomfort. Therefore, MSCs may prove to be the ideal cell source for auto-cell therapy for retinal degeneration and other ocular vascular diseases.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Rod and cone protection.
Figure 1. Rod and cone protection.
A. Retinal sections stained with cresyl violet indicate substantial preservation of photoreceptors across the retina in MSC treated eyes at P90, while in control eyes (sham injected (B) and untreated (C)): only a single layer of photoreceptors remained. A1, A2 &A3: higher power images showing preservation of photoreceptors from the insets A1, A2 &A3 in A. D, E&F: confocal images showing rhodopsin (green in D) and cone arrestin (red in E) positive staining at P90 in MSC treated retina, while in sham injected retina, cone arrestin staining was dramatically reduced (F). All sections were counterstained with DAPI (blue) (scale bars equal 50 µm).
Figure 2. Preservation of visual function.
Figure 2. Preservation of visual function.
A. Visual acuity tested by Optomotor response. Unrestrained animals were placed on a platform, where they tracked the grating with reflexive head movements. The acuity threshold was quantified by increasing the spatial frequency of the grating. RCS rats received MSCs and medium injection via tail vein at P30 and tested at P90. Visual acuity was significantly better in MSC treated eyes compared with controls (P

Figure 3. Vascular protection.

A–F: Retinal whole…

Figure 3. Vascular protection.

A–F: Retinal whole mount was stained with NADPH-diaphorase: A. typical vascular…

Figure 3. Vascular protection.
A–F: Retinal whole mount was stained with NADPH-diaphorase: A. typical vascular pathology in the eye at P90 in untreated RCS rat: vascular complexes (abnormal vessels associated with RPE cells) were mainly located around the optic nerve disc (arrows) and spread peripheral with age. B. vascular complexes in the middle to peripheral retina (arrows). C. high power image showing vascular complexes (arrows) from B. D. RCS retina treated with MSCs at P90: the vascular complexes were dramatically reduced around the optic nerve disc. E. two vascular complexes (arrows) in the middle field of the retina. F. high power image from E showing vascular complexes (arrow). G–L. animal was perfused with FITC-dextran, whole mount was prepared: G. typical vascular leakage, mainly around the optic disc in untreated eye at P90. H–K. high power images from G showing vascular leakage (arrows in H) and abnormal vessels (arrows in I–K). L. MSC treated retina, the vascular leakage around the optic nerve disc was greatly reduced. M&N. high power images from L showing much reduced leakage (arrows in M) and small abnormal vessels (arrow in N) (Scale bars equal 250 µm for A, D, G &L; 100 µm for F).

Figure 4. Upregulation of trophic factors.

A.…

Figure 4. Upregulation of trophic factors.

A. Semi-quantitative RT-PCR for CNTF, bFGF, BDNF and beta…

Figure 4. Upregulation of trophic factors.
A. Semi-quantitative RT-PCR for CNTF, bFGF, BDNF and beta actin. Lane 1: RNA isolated from MSC prior to injection; Lane 2–4: RNA isolated from retinas treated with MSC; Lane 5–7: RNA isolated from non-treated control retinas. B. Densitometry analysis of CNTF, BDNF and bFGF in treated versus untreated samples. Beta actin was used to normalize the data for comparison. Level of CNTF and BDNF in the treated retinas were significantly higher than non-treated controls (p

Figure 5. Distribution of MSCs.

A. phase…

Figure 5. Distribution of MSCs.

A. phase contrast microphotograph of bone marrow derived mesenchymal stem…

Figure 5. Distribution of MSCs.
A. phase contrast microphotograph of bone marrow derived mesenchymal stem cells at passage 2. B. MSCs were preincubated with PKH26 before intravenous injection. C. PKH26 labeled MSCs in the retina two weeks after intravenous injection (arrows); blood vessels were perfused with FITC-dextran (green). D–F. showing PKH26 labeled MSCs in the retinal section (D, arrows pointing PKH26 labeled MSCs; double arrows indicating background staining in debris zone); sections counterstained with DAPI (E); F. merged image from D&E showing PKH26 labeled MSCs counterstained with DAPI (scale bar equals 100 µm).
Similar articles
Cited by
References
    1. Farrar GJ, Kenna PF, Humphries P. On the genetics of retinitis pigmentosa and on mutation-independent approaches to therapeutic intervention. EMBO J. 2002;21:857–864. - PMC - PubMed
    1. Berson EL. Retinitis pigmentosa. The Friedenwald Lecture. Invest Ophthalmol Vis Sci. 1993;34:1659–1676. - PubMed
    1. Hartong DT, Berson EL, Dryja TP. Retinitis pigmentosa. Lancet. 2006;368:1795–1809. - PubMed
    1. Daiger SP, Bowne SJ, Sullivan LS. Perspective on genes and mutations causing retinitis pigmentosa. Arch Ophthalmol. 2007;125:151–158. - PMC - PubMed
    1. Li ZY, Possin DE, Milam AH. Histopathology of bone spicule pigmentation in retinitis pigmentosa. Ophthalmology. 1995;102:805–816. - PubMed
Show all 57 references
Publication types
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3. Vascular protection.
Figure 3. Vascular protection.
A–F: Retinal whole mount was stained with NADPH-diaphorase: A. typical vascular pathology in the eye at P90 in untreated RCS rat: vascular complexes (abnormal vessels associated with RPE cells) were mainly located around the optic nerve disc (arrows) and spread peripheral with age. B. vascular complexes in the middle to peripheral retina (arrows). C. high power image showing vascular complexes (arrows) from B. D. RCS retina treated with MSCs at P90: the vascular complexes were dramatically reduced around the optic nerve disc. E. two vascular complexes (arrows) in the middle field of the retina. F. high power image from E showing vascular complexes (arrow). G–L. animal was perfused with FITC-dextran, whole mount was prepared: G. typical vascular leakage, mainly around the optic disc in untreated eye at P90. H–K. high power images from G showing vascular leakage (arrows in H) and abnormal vessels (arrows in I–K). L. MSC treated retina, the vascular leakage around the optic nerve disc was greatly reduced. M&N. high power images from L showing much reduced leakage (arrows in M) and small abnormal vessels (arrow in N) (Scale bars equal 250 µm for A, D, G &L; 100 µm for F).
Figure 4. Upregulation of trophic factors.
Figure 4. Upregulation of trophic factors.
A. Semi-quantitative RT-PCR for CNTF, bFGF, BDNF and beta actin. Lane 1: RNA isolated from MSC prior to injection; Lane 2–4: RNA isolated from retinas treated with MSC; Lane 5–7: RNA isolated from non-treated control retinas. B. Densitometry analysis of CNTF, BDNF and bFGF in treated versus untreated samples. Beta actin was used to normalize the data for comparison. Level of CNTF and BDNF in the treated retinas were significantly higher than non-treated controls (p

Figure 5. Distribution of MSCs.

A. phase…

Figure 5. Distribution of MSCs.

A. phase contrast microphotograph of bone marrow derived mesenchymal stem…

Figure 5. Distribution of MSCs.
A. phase contrast microphotograph of bone marrow derived mesenchymal stem cells at passage 2. B. MSCs were preincubated with PKH26 before intravenous injection. C. PKH26 labeled MSCs in the retina two weeks after intravenous injection (arrows); blood vessels were perfused with FITC-dextran (green). D–F. showing PKH26 labeled MSCs in the retinal section (D, arrows pointing PKH26 labeled MSCs; double arrows indicating background staining in debris zone); sections counterstained with DAPI (E); F. merged image from D&E showing PKH26 labeled MSCs counterstained with DAPI (scale bar equals 100 µm).
Figure 5. Distribution of MSCs.
Figure 5. Distribution of MSCs.
A. phase contrast microphotograph of bone marrow derived mesenchymal stem cells at passage 2. B. MSCs were preincubated with PKH26 before intravenous injection. C. PKH26 labeled MSCs in the retina two weeks after intravenous injection (arrows); blood vessels were perfused with FITC-dextran (green). D–F. showing PKH26 labeled MSCs in the retinal section (D, arrows pointing PKH26 labeled MSCs; double arrows indicating background staining in debris zone); sections counterstained with DAPI (E); F. merged image from D&E showing PKH26 labeled MSCs counterstained with DAPI (scale bar equals 100 µm).

References

    1. Farrar GJ, Kenna PF, Humphries P. On the genetics of retinitis pigmentosa and on mutation-independent approaches to therapeutic intervention. EMBO J. 2002;21:857–864.
    1. Berson EL. Retinitis pigmentosa. The Friedenwald Lecture. Invest Ophthalmol Vis Sci. 1993;34:1659–1676.
    1. Hartong DT, Berson EL, Dryja TP. Retinitis pigmentosa. Lancet. 2006;368:1795–1809.
    1. Daiger SP, Bowne SJ, Sullivan LS. Perspective on genes and mutations causing retinitis pigmentosa. Arch Ophthalmol. 2007;125:151–158.
    1. Li ZY, Possin DE, Milam AH. Histopathology of bone spicule pigmentation in retinitis pigmentosa. Ophthalmology. 1995;102:805–816.
    1. Milam AH, Li ZY. Retinal pathology in retinitis pigmentosa: Considerations for therapy. 1995. Plenum Press, New York.
    1. Santos A, Humayun MS, de Juan E, Jr, Greenburg RJ, Marsh MJ, et al. Preservation of the inner retina in retinitis pigmentosa. A morphometric analysis. Arch Ophthalmol. 1997;115:511–515.
    1. Ali RR, Sarra GM, Stephens C, Alwis MD, Bainbridge JW, et al. Restoration of photoreceptor ultrastructure and function in retinal degeneration slow mice by gene therapy. Nat Genet. 2000;25:306–310.
    1. Acland GM, Aguirre GD, Bennett J, Aleman TS, Cideciyan AV, et al. Long-term restoration of rod and cone vision by single dose rAAV-mediated gene transfer to the retina in a canine model of childhood blindness. Mol Ther. 2005;12:1072–1082.
    1. Alexander JJ, Umino Y, Everhart D, Chang B, Min SH, et al. Restoration of cone vision in a mouse model of achromatopsia. Nat Med. 2007;13:685–687.
    1. Bainbridge JW, Tan MH, Ali RR. Gene therapy progress and prospects: the eye. Gene Ther. 2006;13:1191–1197.
    1. Hauswirth WW, Aleman TS, Kaushal S, Cideciyan AV, Schwartz SB, 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. 2008;19:979–990.
    1. Williams DS. Usher syndrome: animal models, retinal function of Usher proteins, and prospects for gene therapy. Vision Res. 2008;48:433–441.
    1. Frasson M, Sahel JA, Fabre M, Simonutti M, Dreyfus H, et al. Retinitis pigmentosa: rod photoreceptor rescue by a calcium-channel blocker in the rd mouse. Nat Med. 1999;5:1183–1187.
    1. Tao W, Wen R, Goddard MB, Sherman SD, O'Rourke PJ, et al. Encapsulated cell-based delivery of CNTF reduces photoreceptor degeneration in animal models of retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2002;43:3292–3298.
    1. Li T, Sandberg MA, Pawlyk BS, Rosner B, Hayes KC, et al. Effect of vitamin A supplementation on rhodopsin mutants threonine-17 --> methionine and proline-347 --> serine in transgenic mice and in cell cultures. Proc Natl Acad Sci U S A. 1998;95:11933–11938.
    1. Lund RD, Adamson P, Sauve Y, Keegan DJ, Girman SV, et al. Subretinal transplantation of genetically modified human cell lines attenuates loss of visual function in dystrophic rats. Proc Natl Acad Sci USA. 2001;98:9942–9947.
    1. Lu B, Malcuit C, Wang S, Girman S, Francis P, et al. Long-term Safety and Function of RPE from Human Embryonic Stem Cells in Preclinical Models of Macular Degeneration. Stem Cells 2009
    1. Wang S, Girman S, Lu B, Bischoff N, Holmes T, et al. Long-term vision rescue by human neural progenitors in a rat model of photoreceptor degeneration. Invest Ophthalmol Vis Sci. 2008;49:3201–3206.
    1. Caplan AI. Why are MSCs therapeutic? New data: new insight. J Pathol. 2009;217:318–324.
    1. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98:1076–1084.
    1. Fukuda K. Application of mesenchymal stem cells for the regeneration of cardiomyocyte and its use for cell transplantation therapy. Hum Cell. 2003;16:83–94.
    1. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147.
    1. D'Cruz PM, Yasumura D, Weir J, Matthes MT, Abderrahim H, et al. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum Mol Genet. 2000;9:645–651.
    1. Chaitin MH, Hall MO. Defective ingestion of rod outer segments by cultured dystrophic rat pigment epithelial cells. Invest Ophthalmol Vis Sci. 1983;24:812–820.
    1. Dowling JE, Sidman RL. Inherited retinal dystrophy in the rat. J Cell Biol. 1962;14:73–107.
    1. Gal A, Li Y, Thompson DA, Weir J, Orth U, et al. Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nat Genet. 2000;26:270–271.
    1. Lund RD, Wang S, Klimanskaya I, Holmes T, Ramos-Kelsey R, et al. Human embryonic stem cell-derived cells rescue visual function in dystrophic RCS rats. Cloning Stem Cells. 2006;8:189–199.
    1. Girman SV, Wang S, Lund RD. Time course of deterioration of rod and cone function in RCS rat and the effects of subretinal cell grafting: a light- and dark-adaptation study. Vision Res. 2005;45:343–354.
    1. Wang S, Villegas-Perez MP, Holmes T, Lawrence JM, Vidal-Sanz M, et al. Evolving neurovascular relationships in the RCS rat with age. Curr Eye Res. 2003;27:183–196.
    1. Smith TC, Lee L. Age related macular degeneration - new developments in treatment. Aust Fam Physician. 2007;36:359–361.
    1. Dorrell M, Uusitalo-Jarvinen H, Aguilar E, Friedlander M. Ocular neovascularization: basic mechanisms and therapeutic advances. Surv Ophthalmol. 2007;52(Suppl 1):S3–19.
    1. Fong DS, Aiello L, Gardner TW, King GL, Blankenship G, et al. Retinopathy in diabetes. Diabetes Care. 2004;27(Suppl 1):S84–87.
    1. Smith LE. Pathogenesis of retinopathy of prematurity. Acta Paediatr. 2002;Suppl 91:26–28.
    1. Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE. 2008;3:e1886.
    1. Bringmann A, Pannicke T, Grosche J, Francke M, Wiedemann P, et al. Muller cells in the healthy and diseased retina. Prog Retin Eye Res. 2006;25:397–424.
    1. Steinberg RH. Survival factors in retinal degenerations. Current Opinion in Neurobiology. 1994;4:515–524.
    1. Cayouette M, Behn D, Sendtner M, Lachapelle P, Gravel C. Intraocular gene transfer of ciliary neurotrophic factor prevents death and increases responsiveness of rod photoreceptors in the retinal degeneration slow mouse. J Neurosci. 1998;18:9282–9293.
    1. LaVail MM, Unoki K, Yasumura D, Matthes MT, Yancopoulos GD, et al. Multiple growth factors, cytokines, and neurotrophins rescue photoreceptors from the damaging effects of constant light. Proc Natl Acad Sci U S A. 1992;89:11249–11253.
    1. Hauk TG, Leibinger M, Muller A, Andreadaki A, Knippschild U, et al. Stimulation of axon regeneration in the mature optic nerve by intravitreal application of the toll-like receptor 2 agonist Pam3Cys. Invest Ophthalmol Vis Sci. 2010;51:459–464.
    1. MacDonald IM, Sauve Y, Sieving PA. Preventing blindness in retinal disease: ciliary neurotrophic factor intraocular implants. Can J Ophthalmol. 2007;42:399–402.
    1. Sasahara M, Otani A, Oishi A, Kojima H, Yodoi Y, et al. Activation of bone marrow-derived microglia promotes photoreceptor survival in inherited retinal degeneration. Am J Pathol. 2008;172:1693–1703.
    1. Wang S, Lu B, Wood P, Lund RD. Grafting of ARPE-19 and Schwann Cells to the Subretinal Space in RCS Rats. Invest Ophthalmol Vis Sci. 2005;46:2552–2560.
    1. Wang S, Lu B, Lund RD. Morphological changes in the Royal College of Surgeons rat retina during photoreceptor degeneration and after cell-based therapy. J Comp Neurol. 2005;491:400–417.
    1. Inoue Y, Iriyama A, Ueno S, Takahashi H, Kondo M, et al. Subretinal transplantation of bone marrow mesenchymal stem cells delays retinal degeneration in the RCS rat model of retinal degeneration. Exp Eye Res. 2007;85:234–241.
    1. Arnhold S, Heiduschka P, Klein H, Absenger Y, Basnaoglu S, et al. Adenovirally transduced bone marrow stromal cells differentiate into pigment epithelial cells and induce rescue effects in RCS rats. Invest Ophthalmol Vis Sci. 2006;47:4121–4129.
    1. Zacharek A, Chen J, Cui X, Li A, Li Y, et al. Angiopoietin1/Tie2 and VEGF/Flk1 induced by MSC treatment amplifies angiogenesis and vascular stabilization after stroke. J Cereb Blood Flow Metab. 2007;27:1684–1691.
    1. Guo Y, Graham-Evans B, Broxmeyer HE. Murine embryonic stem cells secrete cytokines/growth modulators that enhance cell survival/anti-apoptosis and stimulate colony formation of murine hematopoietic progenitor cells. Stem Cells. 2006;24:850–856.
    1. Guo Y, Hangoc G, Bian H, Pelus LM, Broxmeyer HE. SDF-1/CXCL12 enhances survival and chemotaxis of murine embryonic stem cells and production of primitive and definitive hematopoietic progenitor cells. Stem Cells. 2005;23:1324–1332.
    1. Hatch HM, Zheng D, Jorgensen ML, Petersen BE. SDF-1alpha/CXCR4: a mechanism for hepatic oval cell activation and bone marrow stem cell recruitment to the injured liver of rats. Cloning Stem Cells. 2002;4:339–351.
    1. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, et al. Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood. 2005;106:419–427.
    1. Wynn RF, Hart CA, Corradi-Perini C, O'Neill L, Evans CA, et al. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood. 2004;104:2643–2645.
    1. Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM, et al. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells. 2006;24:1030–1041.
    1. Shi M, Li J, Liao L, Chen B, Li B, et al. Regulation of CXCR4 expression in human mesenchymal stem cells by cytokine treatment: role in homing efficiency in NOD/SCID mice. Haematologica. 2007;92:897–904.
    1. Neuhuber B, Swanger SA, Howard L, Mackay A, Fischer I. Effects of plating density and culture time on bone marrow stromal cell characteristics. Exp Hematol. 2008;36:1176–1185.
    1. Lennon DP, Caplan AI. I&solation of rat marrow-derived mesenchymal stem cells. Exp Hematol. 2006;34:1606–1607.
    1. Prusky GT, Alam NM, Beekman S, Douglas RM. Rapid quantification of adult and developing mouse spatial vision using a virtual optomotor system. Invest Ophthalmol Vis Sci. 2004;45:4611–4616.

Source: PubMed

3
Iratkozz fel