Cells Secreting GDNF and Retinal Degeneration in Rodent Models of Als
Cells Secreting GDNF and Retinal Degeneration in Rodent Models of Als
Cells Secreting GDNF and Retinal Degeneration in Rodent Models of Als
Article
SUMMARY
Human induced pluripotent stem cells (iPSCs) are a renewable cell source that can be differentiated into neural progenitor cells (iNPCs)
and transduced with glial cell line-derived neurotrophic factor (iNPC-GDNFs). The goal of the current study is to characterize iNPC-
GDNFs and test their therapeutic potential and safety. Single-nuclei RNA-seq show iNPC-GDNFs express NPC markers. iNPC-GDNFs
delivered into the subretinal space of the Royal College of Surgeons rodent model of retinal degeneration preserve photoreceptors and
visual function. Additionally, iNPC-GDNF transplants in the spinal cord of SOD1G93A amyotrophic lateral sclerosis (ALS) rats preserve
motor neurons. Finally, iNPC-GDNF transplants in the spinal cord of athymic nude rats survive and produce GDNF for 9 months,
with no signs of tumor formation or continual cell proliferation. iNPC-GDNFs survive long-term, are safe, and provide neuroprotection
in models of both retinal degeneration and ALS, indicating their potential as a combined cell and gene therapy for various neurodegen-
erative diseases.
Stem Cell Reports j Vol. 18 j 1629–1642 j August 8, 2023 j ª 2023 The Authors. 1629
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
phase I/IIa safety trial (clinical trials.gov: NCT02943850). (Shelley et al., 2014; Svendsen et al., 1998), transduced
The primary endpoint of safety at 1 year was met, with with lentivirus to stably express GDNF (Capowski et al.,
no negative effect of the transplant on motor function, 2007; Shelley et al., 2014), then propagated and banked un-
and grafts survived and produced GDNF for up to der cGMP as the CNS10-NPC-GDNF clinical cell lot, and
42 months post-transplantation (Baloh et al., 2022). fNPC-GDNFs were banked as a research-grade cell lot
CNS10-NPC-GDNF is now being delivered to the motor used in this study. To produce iPSC-derived NPCs similar
cortex of ALS patients in a phase I/IIa clinical trial to these fNPCs, a new and substantially shorter protocol
(NCT05306457). Finally, a current phase I/IIa clinical trial was developed. An iPSC line was generated by nucleo-
is transplanting CNS10-NPC into the subretinal space of fecting healthy PBMCs with nonintegrating oriP/EBNA1
RP patients (NCT04284293). plasmids, which allowed for episomal expression of reprog-
Fetal-derived NPCs and their derivatives are a promising ramming factors (Barrett et al., 2014). Using dual SMAD
treatment for Parkinson’s disease, ALS, and RP, as well as inhibition (Chambers et al., 2009), a monolayer of neuroe-
other neurological conditions such as Huntington’s dis- pithelial progenitors was efficiently generated, which were
ease, stroke, and dementia (Andres et al., 2011; Goldberg then collected and transitioned into neurospheres in the
et al., 2017; McBride et al., 2004). However, this product presence of epidermal growth factor (EGF), fibroblast
has some limitations, including low fetal tissue availabil- growth factor 2 (FGF2), and leukemia inhibitory factor
ity that can hinder large scale-up, thus restricting phase (LIF) (Figure 1A). iNPC neurospheres were expanded by me-
III trials and commercialization. Our lab and others have chanical passage using a tissue chopper or mesh chopper.
shown that induced pluripotent stem cells (iPSCs) provide After passage 16, iNPCs were dissociated to single cells
a renewable, scalable, and safe cell source for deriving po- and lentivirally infected to stably express GDNF. Trans-
tential cell products (Rosati et al., 2018; Sareen et al., duced cells reformed as iNPC-GDNF neurospheres that
2014; Tsai et al., 2015). Peripheral blood mononuclear were used in the current studies.
cells (PBMCs) from an individual’s blood sample can be To demonstrate consistency in the production process,
reprogrammed into iPSCs, which proliferate in culture, two independent batches of iNPC-GDNFs were produced.
differentiate into multiple tissue types, and can be cryo- G-band karyotyping revealed that one batch remained
preserved and thawed (Barrett et al., 2014). We have karyotypically normal, while the other batch acquired a
demonstrated that human iPSCs cultured in specified me- karyotypic anomaly not present in the originating iPSC
dia could generate neural-specific cultures that were prop- line with nearly 100% trisomy of chromosome 12, termed
agated as spheres (termed EZ spheres), which could iNPC-GDNF-T12 (Figure S1A). To avoid complications that
engraft and differentiate into astrocytes in the rodent spi- could arise from karayotypic abnormality, subsequent
nal cord and retina (Ebert et al., 2013; Sareen et al., 2014; studies were performed with the karyotypically normal
Tsai et al., 2015). iNPC-GDNF batch, which remained normal up to 19 pas-
Here, we developed and tested an iPSC-based therapy as sages. To identify any potential remaining pluripotent cells
an alternative to fetal-derived products. We generated in the differentiated batches, pluripotency genes POU5F1
iPSC-derived NPCs (termed iNPCs), which were transduced (OCT4), NANOG, SOX2, and KLF4 were plotted from sin-
to express GDNF (iNPC-GDNF), characterized, and gle-nuclei RNA sequencing (snRNA-seq) data (Figure S1B).
compared with fNPC-GDNFs. iNPC-GDNFs were trans- While individual cells expressed some of these markers at
planted into the subretinal space of RCS rats where they low levels, no cell expressed all four or any combination
protected photoreceptors and vision, the spinal cord of of OCT4, NANOG, and KLF4 (Figure S1C). As expected,
SOD1 rats where they protected motor neurons, and the SOX2 was expressed throughout most cells (Figure S1B),
spinal cord of nude rats where they showed long-term sur- as it is also a NPC marker (Zhang and Cui, 2014).
vival and safety. Based on safety and efficacy, iNPC-GDNFs snRNA-seq was performed to assess the similarity be-
can be pursued as a promising combined cell and gene ther- tween iNPC-GDNF and fNPC-GDNF neurosphere cultures.
apy for multiple neurodegenerative diseases. Following batch normalization, UMAP (uniform manifold
approximation and projection) was applied to identify
clusters of cell types in an unbiased manner, which identi-
RESULTS fied 11 separate clusters (Figure 1B, Table S1). When split by
sample source, fNPC-GDNFs were found in all clusters
iNPC-GDNFs and derived astrocytes resemble fNPC- except for 2 and 4, which were almost exclusively iNPC-
GDNFs and derived astrocytes GDNFs, while clusters 8 and 10 were entirely fNPC-GDNFs
NPCs derived from a single human fetal cortex were main- (Figure 1C, Table S1). Importantly, the overlap of a popula-
tained as free-floating spheres (termed neurospheres), tion of iNPC-GDNF clusters in most fNPC-GDNF clusters
expanded by mechanical passage using a tissue chopper demonstrates the remarkable similarity in some cell
B C
D F
D E F G
Figure 2. iNPC-GDNFs are neuroprotective in the RCS rat model of retinal degeneration
(A) Schematic of experiment RCS rats (n = 13) that received subretinal injection of iNPC-GDNFs at postnatal day (P) 21–23. The fellow eye
received either balanced salt solution injection (sham, n = 4) or no treatment (n = 9).
(B and C) Visual function was evaluated by (B) OKR and (C) ERG, which showed significant preservation in cell-treated compared with
control animals (****p < 0.0001, **p < 0.01, *p < 0.05, one-way ANOVA with Tukey’s HSD, ns: not significant).
(D and E) Cresyl violet-stained retinal images show photoreceptor preservation in (D) iNPC-GDNF-treated retina with potential grafted cells
(arrowheads), compared with (E) sham. d1, d2, e1, and e2 are high-power images from areas in boxed outlines.
(F) Percent of retina with at least two layers of photoreceptors in iNPC-GDNF-treated vs. sham (***p < 0.001 via unpaired t test with
Welch’s correction).
(G) Staining with human nuclear marker MAB1281 shows extensive distribution of grafted iNPC-GDNFs; g1 and g2 are high-power images
from areas in boxed outlines.
(H) Synaptophysin and cone arrestin staining show cones with segments and pedicles (arrowheads) were preserved in cell-treated retina
compared with controls.
(I) GFAP staining shows reactive Müller glia in controls and cell-treated retina, GFAP labels iNPC-GDNFs, and host Müller glia (stars, arrows
point to Müller glial end feet). Scale bars represent (D, E, and G) 500 mm and (H and I) 75 mm. INL, inner nuclear layer; IPL, inner plexiform
layer; ONL, outer nuclear layer. d1, e1, and g1 indicate regions near injection site, d2, e2, and g2 indicate regions distal from injection site.
See also Figure S2.
E F
G H
D E
To examine effects of long-term iNPC-GDNF transplants (Figure 4D). While microglia number varied across animals,
on spinal cord inflammation, immunohistochemistry was there was no significant increase in microglia surrounding
performed for human cells and the microglial marker IBA-1 the SC121+ graft compared with the contralateral spinal
This work was supported in part by grants from the California Capowski, E.E., Schneider, B.L., Ebert, A.D., Seehus, C.R., Szulc, J.,
Institute for Regenerative Medicine (CIRM) DR2A-05320 to CNS Zufferey, R., Aebischer, P., and Svendsen, C.N. (2007). Lentiviral
and CIRM-EDUC2-12638 to SR and the Board of Governors vector-mediated genetic modification of human neural progenitor
Regenerative Medicine Institute. We thank the Cedars-Sinai Ge- cells for ex vivo gene therapy. J. Neurosci. Methods 163, 338–349.
nomics Core for assistance with snRNA-seq, the Cedars-Sinai Cy- Chambers, S.M., Fasano, C.A., Papapetrou, E.P., Tomishima, M., Sa-
togenetics Core for karyotype analysis, and Jovita Dimas-Harms delain, M., and Studer, L. (2009). Highly efficient neural conver-
and Jake Inzalaco for immunohistochemistry assistance. Sche- sion of human ES and iPS cells by dual inhibition of SMAD
matics were created using biorender.com signaling. Nat. Biotechnol. 27, 275–280.
Das, M.M., Avalos, P., Suezaki, P., Godoy, M., Garcia, L., Chang,
CONFLICT OF INTERESTS C.D., Vit, J.-P., Shelley, B., Gowing, G., and Svendsen, C.N.
(2016). Human neural progenitors differentiate into astrocytes
The authors declare no competing financial interests. Intellectual
and protect motor neurons in aging rats. Exp. Neurol. 280, 41–49.
protection-patent ‘‘Cortical Neural progenitor cells from iPSCs’’
No. 62/924,523 was filed October 22, 2019. Draper, J.S., Smith, K., Gokhale, P., Moore, H.D., Maltby, E., John-
son, J., Meisner, L., Zwaka, T.P., Thomson, J.A., and Andrews, P.W.
Received: November 7, 2022 (2004). Recurrent gain of chromosomes 17q and 12 in cultured hu-
Revised: March 24, 2023 man embryonic stem cells. Nat. Biotechnol. 22, 53–54.
Accepted: March 27, 2023 Ebert, A.D., Shelley, B.C., Hurley, A.M., Onorati, M., Castiglioni, V.,
Published: April 20, 2023 Patitucci, T.N., Svendsen, S.P., Mattis, V.B., McGivern, J.V., Schwab,
A.J., et al. (2013). EZ spheres: a stable and expandable culture sys-
tem for the generation of pre-rosette multipotent stem cells from
REFERENCES human ESCs and iPSCs. Stem Cell Res. 10, 417–427.
Akhtar, A.A., Gowing, G., Kobritz, N., Savinoff, S.E., Garcia, L., Gamm, D.M., Wang, S., Lu, B., Girman, S., Holmes, T., Bischoff, N.,
Saxon, D., Cho, N., Kim, G., Tom, C.M., Park, H., et al. (2018). Shearer, R.L., Sauvé, Y., Capowski, E., Svendsen, C.N., and Lund,
Inducible expression of GDNF in transplanted iPSC-derived neural R.D. (2007). Protection of visual functions by human neural pro-
progenitor cells. Stem Cell Rep. 10, 1696–1704. genitors in a rat model of retinal disease. PLoS One 2, e338.