Summary

Toxicity Screens in Human Retinal Organoids for Pharmaceutical Discovery

Published: March 04, 2021
doi:

Summary

Here we present a step-by-step protocol to generate mature human retinal organoids and utilize them in a photoreceptor toxicity assay to identify pharmaceutical candidates for the age-related retinal degenerative disease macular telangiectasia type 2 (MacTel).

Abstract

Organoids provide a promising platform to study disease mechanism and treatments, directly in the context of human tissue with the versatility and throughput of cell culture. Mature human retinal organoids are utilized to screen potential pharmaceutical treatments for the age-related retinal degenerative disease macular telangiectasia type 2 (MacTel).

We have recently shown that MacTel can be caused by elevated levels of an atypical lipid species, deoxysphingolipids (deoxySLs). These lipids are toxic to the retina and may drive the photoreceptor loss that occurs in MacTel patients. To screen drugs for their ability to prevent deoxySL photoreceptor toxicity, we generated human retinal organoids from a non-MacTel induced pluripotent stem cell (iPSC) line and matured them to a post-mitotic age where they develop all of the neuronal lineage-derived cells of the retina, including functionally mature photoreceptors. The retinal organoids were treated with a deoxySL metabolite and apoptosis was measured within the photoreceptor layer using immunohistochemistry. Using this toxicity model, pharmacological compounds that prevent deoxySL-induced photoreceptor death were screened. Using a targeted candidate approach, we determined that fenofibrate, a drug commonly prescribed for the treatment of high cholesterol and triglycerides, can also prevent deoxySL toxicity in the cells of the retina.

The toxicity screen successfully identified an FDA-approved drug that can prevent photoreceptor death. This is a directly actionable finding owing to the highly disease-relevant model tested. This platform can be easily modified to test any number of metabolic stressors and potential pharmacological interventions for future treatment discovery in retinal diseases.

Introduction

Modeling human disease in cell culture and animal models has provided invaluable tools for the discovery, modification, and validation of pharmacologic therapeutics, allowing them to advance from candidate drug to approved therapy. Although a combination of in vitro and non-human in vivo models has long been a critical component of the drug development pipeline they frequently fail to predict the clinical performance of novel drug candidates1. There is a clear need for the development of technologies that bridge the gap between simplistic human cellular monocultures and clinical trials. Recent technological advances in self-organized three-dimensional tissue cultures, organoids, have improved their fidelity to the tissues they model making them promising tools in the preclinical drug development pipeline2.

A major advantage of human cell culture over non-human in vivo models is the ability to replicate the specific intricacies of human metabolism which can vary considerably even between higher order vertebrates such as humans and mice3. However, this specificity can be overshadowed by a loss in tissue complexity; such is the case for retinal tissue where multiple cell types are intricately interwoven and have a unique symbiotic metabolic interplay between cellular subtypes that cannot be replicated in a monoculture4. Human organoids, which provide a facsimile of complex human tissues with the accessibility and scalability of cell culture, have the potential to overcome the deficiencies of these disease modeling platforms.

Retinal organoids derived from stem cells have proven to be particularly faithful in modeling the complex tissue of the human neural retina5. This has made the retinal organoid model a promising technology for the study and treatment of retinal disease6,7. To date much of the disease modeling in retinal organoids has focused on monogenic retinal diseases where retinal organoids are derived from iPSC lines with disease-causing genetic variants7. These are generally highly penetrant mutations that manifest as developmental phenotypes. Less work has been effectively done on aging diseases where genetic mutations and environmental stressors impact tissue that has developed normally. Neurodegenerative diseases of aging can have complex genetic inheritance and contributions from environmental stressors that are inherently difficult to model using short-term cell cultures. However, in many cases these complex diseases can coalesce on common cellular or metabolic stressors that, when tested on a fully developed human tissue, can provide powerful insights into neurodegenerative diseases of aging8.

The late-onset macular degenerative disease, macular telangiectasia type II (MacTel), is a great example of a genetically complex neurodegenerative disease that coalesces on a common metabolic defect. MacTel is an uncommon retinal degenerative disease of aging that results in photoreceptor and Müller glia loss in the macula, leading to a progressive loss in central vision9,10,11,12,13. In MacTel, an undetermined, possibly multifactorial, genetic inheritance drives a common reduction in circulating serine in patients, resulting in an increase in a neurotoxic lipid species called deoxysphingolipids (deoxySL)14,15. To prove that accumulation of deoxySL is toxic to the retina and to validate potential pharmaceutical therapeutics, we developed this protocol to assay photoreceptor toxicity in human retinal organoids14.

Here we outline a specific protocol for differentiating human retinal organoids, establishing a toxicity and rescue assay using organoids, and quantifying outcomes. We provide a successful example where we determine the tissue-specific toxicity of a suspected disease-causing agent, deoxySL, and validate the use of a safe generic drug, fenofibrate, for the potential treatment of deoxySL-induced retinal toxicity. Previous work has shown that fenofibrate can increase the degradation of deoxySL and lower circulating deoxySL in patients, however, its efficacy in reducing deoxySL-induced retinal toxicity has not been tested16,17. Although we present a specific example, this protocol can be utilized to evaluate the effect of any number of metabolic/environmental stressors and potential therapeutic drugs on retinal tissue.

Protocol

1. Thawing, passaging, and expanding iPSCs/ESCs NOTE: For all cell culturing steps, use best practices to maintain a sterile cell culture. Coat a 6-well cell culture plate with basement membrane matrix medium. To prepare 1x of this medium, follow product specifications or dilute 75 µL cold matrix medium with 9 mL of DMEM/F12. Add 1.5 mL of freshly prepared 1x medium per well in a 6-well plate. Incubate at 37 °C for 30 min. Aspirate off the basement …

Representative Results

Retinal organoids were generated from a non-MacTel control iPSC line. After organoids reached 26 weeks in culture they were selected and split into experimental groups. Organoids were treated with varying concentrations of deoxySA to determine if deoxySA is toxic to photoreceptors. Four concentrations of deoxySA were tested, from 0 to 1 µM (Figure 2) and organoids were treated for 8 days, with media changes every other day. Cell death in response to deoxySA is concentration-dependent an…

Discussion

Differentiation protocol variations
Since the invention of self-forming optic cups by Yoshiki Sasai's group20, many labs have developed protocols to generate retinal organoids that can vary at almost every step5,18,19,21. An exhaustive list of protocols can be found in Capowski et al.22. The differentiation protocol we p…

Disclosures

The authors have nothing to disclose.

Acknowledgements

Supported by the Lowy Medical Research Institute. We would like to thank the Lowy family for their support of the MacTel project. We would like to thank Mari Gantner, Mike Dorrell, and Lea Scheppke for their intellectual input and assistance preparing the manuscript.

Materials

0.5M EDTA Invitrogen 15575020
125mL Erlenmeyer Flasks VWR 89095-258
1-deoxysphinganine Avanti 860493
B27 Supplement, minus vitamin A Gibco 12587010
Beaver 6900 Mini-Blade Beaver-Visitec BEAVER6900
D-(+)-Sucrose VWR 97061-432
DAPI Thermo-fisher D1306
Dispase II, powder Gibco 17105041
DMEM, high glucose, pyruvate Gibco 11995073
DMEM/F12 Gibco 11330
Donkey anti-rabbit Ig-G, Alexa Fluor plus 555 Thermo-fisher A32794
donkey serum Sigma D9663-10ML
FBS, Heat Inactivated Corning 45001-108
Fenofibrate Sigma F6020
Glutamax Gibco 35050061
Heparin Stemcell Technologies 7980
In Situ Cell Death Detection Kit, Fluorescin Sigma 11684795910
Matrigel, growth factor reduced Corning 356230
MEM Non-Essential Amino Acids Solution Gibco 11140050
mTeSR 1 Stemcell Technologies 85850
N2 Supplement Gibco 17502048
Penicillin-Streptomycin Gibco 15140122
Pierce 16% Formaldehyde Thermo-fisher 28906
Rabbit anti-Recoverin antibody Millipore AB5585
Sodium Citrate Sigma W302600
Steriflip Sterile Disposable Vacuum Filter Units MilliporeSigma SE1M179M6
Taurine Sigma T0625
Tissue Plus- O.C.T. compound Fisher Scientific 23-730-571
Tissue-Tek Cryomold EMS 62534-10
Triton X-100 Sigma X100
Tween-20 Sigma P1379
Ultra-Low Attachment 6 well Plates Corning 29443-030
Ultra-Low Attachment 75cm2 U-Flask Corning 3814
Vacuum Filtration System VWR 10040-436
Vectashield-mounting medium vector Labs H-1000
wax pen-ImmEdge vector Labs H-4000
Y-27632 Dihydrochloride (Rock inhibitor) Sigma Y0503

References

  1. Waring, M. J., et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nature Review Drug Discovery. 14 (7), 475-486 (2015).
  2. Khalil, A. S., Jaenisch, R., Mooney, D. J. Engineered tissues and strategies to overcome challenges in drug development. Advances in Drug Delivery Reviews. 158, 116-139 (2020).
  3. Demetrius, L. Of mice and men. When it comes to studying ageing and the means to slow it down, mice are not just small humans. EMBO Reports. , 39-44 (2005).
  4. Lindsay, K. J., et al. Pyruvate kinase and aspartate-glutamate carrier distributions reveal key metabolic links between neurons and glia in retina. Proceedings of the National Academy of Sciences U. S. A. 111 (43), 15579-15584 (2014).
  5. Cowan, C. S., et al. Cell types of the human retina and its organoids at single-cell resolution. Cell. 182 (6), 1623-1640 (2020).
  6. Kruczek, K., Swaroop, A. Pluripotent stem cell-derived retinal organoids for disease modeling and development of therapies. Stem Cells. 38 (10), 1206-1215 (2020).
  7. Sinha, D., Phillips, J., Phillips, M. J., Gamm, D. M. Mimicking retinal development and disease with human pluripotent stem cells. Investigative Ophthalmology and Visual Science. 57 (5), 1-9 (2016).
  8. Gan, L., Cookson, M. R., Petrucelli, L., La Spada, A. R. Converging pathways in neurodegeneration, from genetics to mechanisms. Nature Neuroscience. 21 (10), 1300-1309 (2018).
  9. Aung, K. Z., Wickremasinghe, S. S., Makeyeva, G., Robman, L., Guymer, R. H. The prevalence estimates of macular telangiectasia type 2: the Melbourne collaborative cohort study. Retina. 30 (3), 473-478 (2010).
  10. Chew, E. Y., et al. Effect of ciliary neurotrophic factor on retinal neurodegeneration in patients with macular telangiectasia type 2: A randomized clinical trial. Ophthalmology. 126 (4), 540-549 (2019).
  11. Gass, J. D., Blodi, B. A. Idiopathic juxtafoveolar retinal telangiectasis. Update of classification and follow-up study. Ophthalmology. 100 (10), 1536-1546 (1993).
  12. Klein, R., et al. The prevalence of macular telangiectasia type 2 in the Beaver Dam eye study. American Journal of Ophthalmology. 150 (1), 55-62 (2010).
  13. Powner, M. B., et al. Loss of Muller’s cells and photoreceptors in macular telangiectasia type 2. Ophthalmology. 120 (11), 2344-2352 (2013).
  14. Gantner, M. L., et al. Serine and lipid metabolism in macular disease and peripheral neuropathy. New England Journal of Medicine. 381 (15), 1422-1433 (2019).
  15. Scerri, T. S., et al. Genome-wide analyses identify common variants associated with macular telangiectasia type 2. Nature Genetics. 49 (4), 559-567 (2017).
  16. Alecu, I., et al. Cytotoxic 1-deoxysphingolipids are metabolized by a cytochrome P450-dependent pathway. Journal of Lipid Research. 58 (1), 60-71 (2017).
  17. Othman, A., et al. Fenofibrate lowers atypical sphingolipids in plasma of dyslipidemic patients: A novel approach for treating diabetic neuropathy. Journal of Clinical Lipidology. 9 (4), 568-575 (2015).
  18. Ohlemacher, S. K., Iglesias, C. L., Sridhar, A., Gamm, D. M., Meyer, J. S. Generation of highly enriched populations of optic vesicle-like retinal cells from human pluripotent stem cells. Curr Protoc Stem Cell Biol. 32, 1-20 (2015).
  19. Zhong, X., et al. Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nature Communication. 5, 4047 (2014).
  20. Eiraku, M., et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 472 (7341), 51-56 (2011).
  21. Wahlin, K. J., et al. Photoreceptor outer segment-like structures in long-term 3D retinas from human pluripotent stem cells. Science Reports. 7 (1), 766 (2017).
  22. Capowski, E. E., et al. Reproducibility and staging of 3D human retinal organoids across multiple pluripotent stem cell lines. Development. 146 (1), (2019).
  23. Luo, Z., et al. An optimized system for effective derivation of three-dimensional retinal tissue via Wnt signaling regulation. Stem Cells. 36 (11), 1709-1722 (2018).
  24. Ovando-Roche, P., et al. Use of bioreactors for culturing human retinal organoids improves photoreceptor yields. Stem Cell Research Therapy. 9 (1), 156 (2018).
check_url/62269?article_type=t

Play Video

Cite This Article
Eade, K., Giles, S., Harkins-Perry, S., Friedlander, M. Toxicity Screens in Human Retinal Organoids for Pharmaceutical Discovery. J. Vis. Exp. (169), e62269, doi:10.3791/62269 (2021).

View Video