1
|
Harkin J, Peña KH, Gomes C, Hernandez M, Lavekar SS, So K, Lentsch K, Feder EM, Morrow S, Huang KC, Tutrow KD, Morris A, Zhang C, Meyer JS. A highly reproducible and efficient method for retinal organoid differentiation from human pluripotent stem cells. Proc Natl Acad Sci U S A 2024; 121:e2317285121. [PMID: 38870053 PMCID: PMC11194494 DOI: 10.1073/pnas.2317285121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Human pluripotent stem cell (hPSC)-derived retinal organoids are three-dimensional cellular aggregates that differentiate and self-organize to closely mimic the spatial and temporal patterning of the developing human retina. Retinal organoid models serve as reliable tools for studying human retinogenesis, yet limitations in the efficiency and reproducibility of current retinal organoid differentiation protocols have reduced the use of these models for more high-throughput applications such as disease modeling and drug screening. To address these shortcomings, the current study aimed to standardize prior differentiation protocols to yield a highly reproducible and efficient method for generating retinal organoids. Results demonstrated that through regulation of organoid size and shape using quick reaggregation methods, retinal organoids were highly reproducible compared to more traditional methods. Additionally, the timed activation of BMP signaling within developing cells generated pure populations of retinal organoids at 100% efficiency from multiple widely used cell lines, with the default forebrain fate resulting from the inhibition of BMP signaling. Furthermore, given the ability to direct retinal or forebrain fates at complete purity, mRNA-seq analyses were then utilized to identify some of the earliest transcriptional changes that occur during the specification of these two lineages from a common progenitor. These improved methods also yielded retinal organoids with expedited differentiation timelines when compared to traditional methods. Taken together, the results of this study demonstrate the development of a highly reproducible and minimally variable method for generating retinal organoids suitable for analyzing the earliest stages of human retinal cell fate specification.
Collapse
Affiliation(s)
- Jade Harkin
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Kiersten H. Peña
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Melody Hernandez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Sailee S. Lavekar
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Kaman So
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Kelly Lentsch
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Elyse M. Feder
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN46202
| | - Sarah Morrow
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Kang-Chieh Huang
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Kaylee D. Tutrow
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Ann Morris
- Department of Biology, University of Kentucky, Lexington, KY40506
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Jason S. Meyer
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN46202
| |
Collapse
|
2
|
Adams JW, Vinokur A, de Souza JS, Austria C, Guerra BS, Herai RH, Wahlin KJ, Muotri AR. Loss of GTF2I promotes neuronal apoptosis and synaptic reduction in human cellular models of neurodevelopment. Cell Rep 2024; 43:113867. [PMID: 38416640 PMCID: PMC11002531 DOI: 10.1016/j.celrep.2024.113867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024] Open
Abstract
Individuals with Williams syndrome (WS), a neurodevelopmental disorder caused by hemizygous loss of 26-28 genes at 7q11.23, characteristically portray a hypersocial phenotype. Copy-number variations and mutations in one of these genes, GTF2I, are associated with altered sociality and are proposed to underlie hypersociality in WS. However, the contribution of GTF2I to human neurodevelopment remains poorly understood. Here, human cellular models of neurodevelopment, including neural progenitors, neurons, and three-dimensional cortical organoids, are differentiated from CRISPR-Cas9-edited GTF2I-knockout (GTF2I-KO) pluripotent stem cells to investigate the role of GTF2I in human neurodevelopment. GTF2I-KO progenitors exhibit increased proliferation and cell-cycle alterations. Cortical organoids and neurons demonstrate increased cell death and synaptic dysregulation, including synaptic structural dysfunction and decreased electrophysiological activity on a multielectrode array. Our findings suggest that changes in synaptic circuit integrity may be a prominent mediator of the link between alterations in GTF2I and variation in the phenotypic expression of human sociality.
Collapse
Affiliation(s)
- Jason W Adams
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA; Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, CA 92093, USA
| | - Annabelle Vinokur
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA
| | - Janaína S de Souza
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA
| | - Charles Austria
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA
| | - Bruno S Guerra
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA; Experimental Multiuser Laboratory, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil
| | - Roberto H Herai
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA; Experimental Multiuser Laboratory, Pontifícia Universidade Católica do Paraná, Curitiba, PR 80215-901, Brazil
| | - Karl J Wahlin
- Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA; Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
3
|
Soucy JR, Todd L, Kriukov E, Phay M, Malechka VV, Rivera JD, Reh TA, Baranov P. Controlling donor and newborn neuron migration and maturation in the eye through microenvironment engineering. Proc Natl Acad Sci U S A 2023; 120:e2302089120. [PMID: 37931105 PMCID: PMC10655587 DOI: 10.1073/pnas.2302089120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 09/30/2023] [Indexed: 11/08/2023] Open
Abstract
Ongoing cell therapy trials have demonstrated the need for precision control of donor cell behavior within the recipient tissue. We present a methodology to guide stem cell-derived and endogenously regenerated neurons by engineering the microenvironment. Being an "approachable part of the brain," the eye provides a unique opportunity to study neuron fate and function within the central nervous system. Here, we focused on retinal ganglion cells (RGCs)-the neurons in the retina are irreversibly lost in glaucoma and other optic neuropathies but can potentially be replaced through transplantation or reprogramming. One of the significant barriers to successful RGC integration into the existing mature retinal circuitry is cell migration toward their natural position in the retina. Our in silico analysis of the single-cell transcriptome of the developing human retina identified six receptor-ligand candidates, which were tested in functional in vitro assays for their ability to guide human stem cell-derived RGCs. We used our lead molecule, SDF1, to engineer an artificial gradient in the retina, which led to a 2.7-fold increase in donor RGC migration into the ganglion cell layer (GCL) and a 3.3-fold increase in the displacement of newborn RGCs out of the inner nuclear layer. Only donor RGCs that migrated into the GCL were found to express mature RGC markers, indicating the importance of proper structure integration. Together, these results describe an "in silico-in vitro-in vivo" framework for identifying, selecting, and applying soluble ligands to control donor cell function after transplantation.
Collapse
Affiliation(s)
- Jonathan R. Soucy
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Levi Todd
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Emil Kriukov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Monichan Phay
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Volha V. Malechka
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - John Dayron Rivera
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Petr Baranov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| |
Collapse
|
4
|
Bai J, Koos DS, Stepanian K, Fouladian Z, Shayler DWH, Aparicio JG, Fraser SE, Moats RA, Cobrinik D. Episodic live imaging of cone photoreceptor maturation in GNAT2-EGFP retinal organoids. Dis Model Mech 2023; 16:dmm050193. [PMID: 37902188 PMCID: PMC10690052 DOI: 10.1242/dmm.050193] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023] Open
Abstract
Fluorescent reporter pluripotent stem cell-derived retinal organoids are powerful tools to investigate cell type-specific development and disease phenotypes. When combined with live imaging, they enable direct and repeated observation of cell behaviors within a developing retinal tissue. Here, we generated a human cone photoreceptor reporter line by CRISPR/Cas9 genome editing of WTC11-mTagRFPT-LMNB1 human induced pluripotent stem cells (iPSCs) by inserting enhanced green fluorescent protein (EGFP) coding sequences and a 2A self-cleaving peptide at the N-terminus of guanine nucleotide-binding protein subunit alpha transducin 2 (GNAT2). In retinal organoids generated from these iPSCs, the GNAT2-EGFP alleles robustly and exclusively labeled immature and mature cones. Episodic confocal live imaging of hydrogel immobilized retinal organoids allowed tracking of the morphological maturation of individual cones for >18 weeks and revealed inner segment accumulation of mitochondria and growth at 12.2 μm3 per day from day 126 to day 153. Immobilized GNAT2-EGFP cone reporter organoids provide a valuable tool for investigating human cone development and disease.
Collapse
Affiliation(s)
- Jinlun Bai
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - David S. Koos
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Translational Biomedical Imaging Laboratory, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Kayla Stepanian
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Zachary Fouladian
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dominic W. H. Shayler
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jennifer G. Aparicio
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Scott E. Fraser
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Translational Biomedical Imaging Laboratory, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Rex A. Moats
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Translational Biomedical Imaging Laboratory, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - David Cobrinik
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
5
|
Agarwal D, Dash N, Mazo KW, Chopra M, Avila MP, Patel A, Wong RM, Jia C, Do H, Cheng J, Chiang C, Jurlina SL, Roshan M, Perry MW, Rho JM, Broyer R, Lee CD, Weinreb RN, Gavrilovici C, Oesch NW, Welsbie DS, Wahlin KJ. Human retinal ganglion cell neurons generated by synchronous BMP inhibition and transcription factor mediated reprogramming. NPJ Regen Med 2023; 8:55. [PMID: 37773257 PMCID: PMC10541876 DOI: 10.1038/s41536-023-00327-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/31/2023] [Indexed: 10/01/2023] Open
Abstract
In optic neuropathies, including glaucoma, retinal ganglion cells (RGCs) die. Cell transplantation and endogenous regeneration offer strategies for retinal repair, however, developmental programs required for this to succeed are incompletely understood. To address this, we explored cellular reprogramming with transcription factor (TF) regulators of RGC development which were integrated into human pluripotent stem cells (PSCs) as inducible gene cassettes. When the pioneer factor NEUROG2 was combined with RGC-expressed TFs (ATOH7, ISL1, and POU4F2) some conversion was observed and when pre-patterned by BMP inhibition, RGC-like induced neurons (RGC-iNs) were generated with high efficiency in just under a week. These exhibited transcriptional profiles that were reminiscent of RGCs and exhibited electrophysiological properties, including AMPA-mediated synaptic transmission. Additionally, we demonstrated that small molecule inhibitors of DLK/LZK and GCK-IV can block neuronal death in two pharmacological axon injury models. Combining developmental patterning with RGC-specific TFs thus provided valuable insight into strategies for cell replacement and neuroprotection.
Collapse
Affiliation(s)
- Devansh Agarwal
- Shu Chien-Gene Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Nicholas Dash
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Kevin W Mazo
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Manan Chopra
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Maria P Avila
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Amit Patel
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Ryan M Wong
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Cairang Jia
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Hope Do
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Jie Cheng
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Colette Chiang
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Shawna L Jurlina
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Mona Roshan
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Michael W Perry
- Department of Biological Sciences, UC San Diego, La Jolla, CA, USA
| | - Jong M Rho
- Department of Neurosciences, UC San Diego, La Jolla, CA, USA
| | - Risa Broyer
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Cassidy D Lee
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Robert N Weinreb
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | | | - Nicholas W Oesch
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
- Department of Psychology, UC San Diego, La Jolla, CA, USA
| | - Derek S Welsbie
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Karl J Wahlin
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Welsbie D, Zack DJ, Johnson TV. Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium. Mol Neurodegener 2023; 18:64. [PMID: 37735444 PMCID: PMC10514988 DOI: 10.1186/s13024-023-00655-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Erika A Aguzzi
- The Institute of Ophthalmology, University College London, London, England, UK
| | - Julie Cho
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael James Gilhooley
- The Institute of Ophthalmology, University College London, London, England, UK
- Moorfields Eye Hospital, London, England, UK
| | - Casey Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Meher A Saleem
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Alex L Kolodkin
- The Solomon H Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ahmara G Ross
- Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian C Samuels
- Department of Ophthalmology and Visual Sciences, Callahan Eye Hospital, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derek Welsbie
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, CA, USA
| | - Donald J Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA.
| |
Collapse
|
7
|
Lavekar SS, Harkin J, Hernandez M, Gomes C, Patil S, Huang KC, Puntambekar SS, Lamb BT, Meyer JS. Development of a three-dimensional organoid model to explore early retinal phenotypes associated with Alzheimer's disease. Sci Rep 2023; 13:13827. [PMID: 37620502 PMCID: PMC10449801 DOI: 10.1038/s41598-023-40382-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of Aβ plaques and neurofibrillary tangles, resulting in synaptic loss and neurodegeneration. The retina is an extension of the central nervous system within the eye, sharing many structural similarities with the brain, and previous studies have observed AD-related phenotypes within the retina. Three-dimensional retinal organoids differentiated from human pluripotent stem cells (hPSCs) can effectively model some of the earliest manifestations of disease states, yet early AD-associated phenotypes have not yet been examined. Thus, the current study focused upon the differentiation of hPSCs into retinal organoids for the analysis of early AD-associated alterations. Results demonstrated the robust differentiation of retinal organoids from both familial AD and unaffected control cell lines, with familial AD retinal organoids exhibiting a significant increase in the Aβ42:Aβ40 ratio as well as phosphorylated Tau protein, characteristic of AD pathology. Further, transcriptional analyses demonstrated the differential expression of many genes and cellular pathways, including those associated with synaptic dysfunction. Taken together, the current study demonstrates the ability of retinal organoids to serve as a powerful model for the identification of some of the earliest retinal alterations associated with AD.
Collapse
Affiliation(s)
- Sailee S Lavekar
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jade Harkin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melody Hernandez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shruti Patil
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kang-Chieh Huang
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shweta S Puntambekar
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jason S Meyer
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
8
|
Liu YV, Santiago CP, Sogunro A, Konar GJ, Hu MW, McNally MM, Lu YC, Flores-Bellver M, Aparicio-Domingo S, Li KV, Li ZL, Agakishiev D, Hadyniak SE, Hussey KA, Creamer TJ, Orzolek LD, Teng D, Canto-Soler MV, Qian J, Jiang Z, Johnston RJ, Blackshaw S, Singh MS. Single-cell transcriptome analysis of xenotransplanted human retinal organoids defines two migratory cell populations of nonretinal origin. Stem Cell Reports 2023; 18:1138-1154. [PMID: 37163980 DOI: 10.1016/j.stemcr.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 05/12/2023] Open
Abstract
Human retinal organoid transplantation could potentially be a treatment for degenerative retinal diseases. How the recipient retina regulates the survival, maturation, and proliferation of transplanted organoid cells is unknown. We transplanted human retinal organoid-derived cells into photoreceptor-deficient mice and conducted histology and single-cell RNA sequencing alongside time-matched cultured retinal organoids. Unexpectedly, we observed human cells that migrated into all recipient retinal layers and traveled long distances. Using an unbiased approach, we identified these cells as astrocytes and brain/spinal cord-like neural precursors that were absent or rare in stage-matched cultured organoids. In contrast, retinal progenitor-derived rods and cones remained in the subretinal space, maturing more rapidly than those in the cultured controls. These data suggest that recipient microenvironment promotes the maturation of transplanted photoreceptors while inducing or facilitating the survival of migratory cell populations that are not normally derived from retinal progenitors. These findings have important implications for potential cell-based treatments of retinal diseases.
Collapse
Affiliation(s)
- Ying V Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clayton P Santiago
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akin Sogunro
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Gregory J Konar
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ming-Wen Hu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minda M McNally
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu-Chen Lu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miguel Flores-Bellver
- CellSight Ocular Stem Cell and Regeneration Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - Silvia Aparicio-Domingo
- CellSight Ocular Stem Cell and Regeneration Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - Kang V Li
- CellSight Ocular Stem Cell and Regeneration Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - Zhuo-Lin Li
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dzhalal Agakishiev
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah E Hadyniak
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Katarzyna A Hussey
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Tyler J Creamer
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linda D Orzolek
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Teng
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - Jiang Qian
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zheng Jiang
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Robert J Johnston
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA.
| | - Seth Blackshaw
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Bai J, Koos DS, Stepanian K, Fouladian Z, Shayler DWH, Aparicio JG, Fraser SE, Moats RA, Cobrinik D. Episodic live imaging of cone photoreceptor maturation in GNAT2-EGFP retinal organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530518. [PMID: 36909527 PMCID: PMC10002746 DOI: 10.1101/2023.02.28.530518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Fluorescent reporter pluripotent stem cell (PSC) derived retinal organoids are powerful tools to investigate cell type-specific development and disease phenotypes. When combined with live imaging, they enable direct and repeated observation of cell behaviors within a developing retinal tissue. Here, we generated a human cone photoreceptor reporter line by CRISPR/Cas9 genome editing of WTC11-mTagRFPT-LMNB1 human induced pluripotent stem cells (iPSCs) by inserting enhanced green fluorescent protein (EGFP) coding sequences and a 2A self-cleaving peptide at the N-terminus of Guanine Nucleotide-Binding Protein Subunit Alpha Transducin 2 (GNAT2). In retinal organoids generated from these iPSCs, the GNAT2-EGFP allele robustly and exclusively labeled both immature and mature cones starting at culture day 34. Episodic confocal live imaging of hydrogel immobilized retinal organoids allowed tracking of morphological maturation of individual cones for >18 weeks and revealed inner segment accumulation of mitochondria and growth at 12.2 cubic microns per day from day 126 to day 153. Immobilized GNAT2-EGFP cone reporter organoids provide a valuable tool for investigating human cone development and disease.
Collapse
Affiliation(s)
- Jinlun Bai
- The Vision Center, Department of Surgery, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David S. Koos
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Translational Biomedical Imaging Laboratory, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Radiology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Kayla Stepanian
- The Vision Center, Department of Surgery, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Zachary Fouladian
- The Vision Center, Department of Surgery, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dominic W. H. Shayler
- The Vision Center, Department of Surgery, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jennifer G. Aparicio
- The Vision Center, Department of Surgery, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Scott E. Fraser
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Translational Biomedical Imaging Laboratory, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
- Translational Imaging Center, University of Southern California, Los Angeles, CA, USA
| | - Rex A. Moats
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Translational Biomedical Imaging Laboratory, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Radiology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - David Cobrinik
- The Vision Center, Department of Surgery, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Watari K, Yamasaki S, Tu HY, Shikamura M, Kamei T, Adachi H, Tochitani T, Kita Y, Nakamura A, Ueyama K, Ono K, Morinaga C, Matsuyama T, Sho J, Nakamura M, Fujiwara M, Hori Y, Tanabe A, Hirai R, Terai O, Ohno O, Ohara H, Hayama T, Ikeda A, Nukaya D, Matsushita K, Takahashi M, Kishino A, Kimura T, Kawamata S, Mandai M, Kuwahara A. Self-organization, quality control, and preclinical studies of human iPSC-derived retinal sheets for tissue-transplantation therapy. Commun Biol 2023; 6:164. [PMID: 36765170 PMCID: PMC9918541 DOI: 10.1038/s42003-023-04543-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Three-dimensional retinal organoids (3D-retinas) are a promising graft source for transplantation therapy. We previously developed self-organizing culture for 3D-retina generation from human pluripotent stem cells (hPSCs). Here we present a quality control method and preclinical studies for tissue-sheet transplantation. Self-organizing hPSCs differentiated into both retinal and off-target tissues. Gene expression analyses identified the major off-target tissues as eye-related, cortex-like, and spinal cord-like tissues. For quality control, we developed a qPCR-based test in which each hPSC-derived neuroepithelium was dissected into two tissue-sheets: inner-central sheet for transplantation and outer-peripheral sheet for qPCR to ensure retinal tissue selection. During qPCR, tissue-sheets were stored for 3-4 days using a newly developed preservation method. In a rat tumorigenicity study, no transplant-related adverse events were observed. In retinal degeneration model rats, retinal transplants differentiated into mature photoreceptors and exhibited light responses in electrophysiology assays. These results demonstrate our rationale toward self-organizing retinal sheet transplantation therapy.
Collapse
Affiliation(s)
- Kenji Watari
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Suguru Yamasaki
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan ,grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Hung-Ya Tu
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Masayuki Shikamura
- grid.417982.10000 0004 0623 246XResearch & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe 650-0047 Japan
| | - Tatsuya Kamei
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Hideki Adachi
- grid.417741.00000 0004 1797 168XPreclinical Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Konohana-ku, Osaka 554-0022 Japan
| | - Tomoaki Tochitani
- grid.417741.00000 0004 1797 168XPreclinical Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Konohana-ku, Osaka 554-0022 Japan
| | - Yasuyuki Kita
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Aya Nakamura
- grid.417741.00000 0004 1797 168XTechnology Research & Development Division, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Kazuki Ueyama
- grid.417741.00000 0004 1797 168XTechnology Research & Development Division, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Keiichi Ono
- grid.417741.00000 0004 1797 168XTechnology Research & Development Division, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Chikako Morinaga
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan ,grid.7597.c0000000094465255RIKEN Program for Drug Discovery and Medical Technology Platforms, RIKEN Cluster for Science, Technology and Innovation Hub., Saitama, 351-0198 Japan
| | - Take Matsuyama
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Junki Sho
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Miyuki Nakamura
- grid.417982.10000 0004 0623 246XResearch & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe 650-0047 Japan
| | - Masayo Fujiwara
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Yoriko Hori
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Anna Tanabe
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Rina Hirai
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Orie Terai
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Osamu Ohno
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Hidetaka Ohara
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Tetsuya Hayama
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Atsushi Ikeda
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Daiki Nukaya
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Keizo Matsushita
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan ,grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Masayo Takahashi
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Akiyoshi Kishino
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Toru Kimura
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Shin Kawamata
- grid.417982.10000 0004 0623 246XResearch & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe 650-0047 Japan
| | - Michiko Mandai
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan ,grid.7597.c0000000094465255RIKEN Program for Drug Discovery and Medical Technology Platforms, RIKEN Cluster for Science, Technology and Innovation Hub., Saitama, 351-0198 Japan
| | - Atsushi Kuwahara
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
11
|
A Tet-Inducible CRISPR Platform for High-Fidelity Editing of Human Pluripotent Stem Cells. Genes (Basel) 2022; 13:genes13122363. [PMID: 36553630 PMCID: PMC9777998 DOI: 10.3390/genes13122363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/16/2022] Open
Abstract
Pluripotent stem cells (PSCs) offer an exciting resource for probing human biology; however, gene-editing efficiency remains relatively low in many cell types, including stem cells. Gene-editing using the CRISPR-Cas9 system offers an attractive solution that improves upon previous gene-editing approaches; however, like other technologies, off-target mutagenesis remains a concern. High-fidelity Cas9 variants greatly reduce off-target mutagenesis and offer a solution to this problem. To evaluate their utility as part of a cell-based gene-editing platform, human PSC lines were generated with a high-fidelity (HF) tetracycline-inducible engineered Streptococcus pyogenes SpCas9 (HF-iCas9) integrated into the AAVS1 safe harbor locus. By engineering cells with controllable expression of Cas9, we eliminated the need to include a large Cas9-expressing plasmid during cell transfection. Delivery of genetic cargo was further optimized by packaging DNA targeting guide RNAs (gRNAs) and donor fragments into a single plasmid backbone. The potential of homology-directed repair (HDR) based gene knock-in at the CLYBL safe harbor site and endogenous SOX2 and SIX6 genes were demonstrated. Moreover, we used non-homologous end-joining (NHEJ) for gene knockout of disease-relevant alleles. These high-fidelity CRISPR tools and the resulting HF-iCas9 cell lines will facilitate the production of cell-type reporters and mutants across different genetic backgrounds.
Collapse
|
12
|
Agarwal D, Kuhns R, Dimitriou CN, Barlow E, Wahlin KJ, Enke RA. Bulk RNA sequencing analysis of developing human induced pluripotent cell-derived retinal organoids. Sci Data 2022; 9:759. [PMID: 36494376 PMCID: PMC9734101 DOI: 10.1038/s41597-022-01853-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Retinogenesis involves the transformation of the anterior developing brain into organized retinal lamellae coordinated by intricate gene signalling networks. This complex process has been investigated in several model organisms such as birds, fish, mammals and amphibians, yet many facets of retinal development are different in humans and remain unexplored. In this regard, human pluripotent stem cell (hPSC)-derived 3D retinal organoids and Next Generation Sequencing (NGS) have emerged as key technologies that have facilitated the discovery of previously unknown details about cell fate specification and gene regulation in the retina. Here we utilized hPSCs integrated with fluorescent reporter genes (SIX6-p2A-eGFP/CRX-p2A-h2b-mRuby3) to generate retinal organoids and carry out bulk RNA sequencing of samples encompassing the majority of retinogenesis (D0-D280). This data set will serve as a valuable reference for the vision research community to characterize differentially expressed genes in the developing human eye.
Collapse
Affiliation(s)
- Devansh Agarwal
- Viterbi Family Department of Ophthalmology at the Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rian Kuhns
- Department of Biology, James Madison University, Harrisonburg, VA, 22807, USA
| | | | - Emmalyn Barlow
- Department of Biology, James Madison University, Harrisonburg, VA, 22807, USA
| | - Karl J Wahlin
- Viterbi Family Department of Ophthalmology at the Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Ray A Enke
- Department of Biology, James Madison University, Harrisonburg, VA, 22807, USA.
- The Center for Genome & Metagenome Studies, James Madison University Harrisonburg, Harrisonburg, VA, 22807, USA.
| |
Collapse
|
13
|
Benati D, Leung A, Perdigao P, Toulis V, van der Spuy J, Recchia A. Induced Pluripotent Stem Cells and Genome-Editing Tools in Determining Gene Function and Therapy for Inherited Retinal Disorders. Int J Mol Sci 2022; 23:ijms232315276. [PMID: 36499601 PMCID: PMC9735568 DOI: 10.3390/ijms232315276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal disorders (IRDs) affect millions of people worldwide and are a major cause of irreversible blindness. Therapies based on drugs, gene augmentation or transplantation approaches have been widely investigated and proposed. Among gene therapies for retinal degenerative diseases, the fast-evolving genome-editing CRISPR/Cas technology has emerged as a new potential treatment. The CRISPR/Cas system has been developed as a powerful genome-editing tool in ophthalmic studies and has been applied not only to gain proof of principle for gene therapies in vivo, but has also been extensively used in basic research to model diseases-in-a-dish. Indeed, the CRISPR/Cas technology has been exploited to genetically modify human induced pluripotent stem cells (iPSCs) to model retinal disorders in vitro, to test in vitro drugs and therapies and to provide a cell source for autologous transplantation. In this review, we will focus on the technological advances in iPSC-based cellular reprogramming and gene editing technologies to create human in vitro models that accurately recapitulate IRD mechanisms towards the development of treatments for retinal degenerative diseases.
Collapse
Affiliation(s)
- Daniela Benati
- Centre for Regenerative Medicine, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Amy Leung
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Pedro Perdigao
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | | | - Alessandra Recchia
- Centre for Regenerative Medicine, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence: (J.v.d.S.); (A.R.)
| |
Collapse
|
14
|
Jones MK, Agarwal D, Mazo KW, Chopra M, Jurlina SL, Dash N, Xu Q, Ogata AR, Chow M, Hill AD, Kambli NK, Xu G, Sasik R, Birmingham A, Fisch KM, Weinreb RN, Enke RA, Skowronska-Krawczyk D, Wahlin KJ. Chromatin Accessibility and Transcriptional Differences in Human Stem Cell-Derived Early-Stage Retinal Organoids. Cells 2022; 11:3412. [PMID: 36359808 PMCID: PMC9657268 DOI: 10.3390/cells11213412] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 02/08/2023] Open
Abstract
Retinogenesis involves the specification of retinal cell types during early vertebrate development. While model organisms have been critical for determining the role of dynamic chromatin and cell-type specific transcriptional networks during this process, an enhanced understanding of the developing human retina has been more elusive due to the requirement for human fetal tissue. Pluripotent stem cell (PSC) derived retinal organoids offer an experimentally accessible solution for investigating the developing human retina. To investigate cellular and molecular changes in developing early retinal organoids, we developed SIX6-GFP and VSX2-tdTomato (or VSX2-h2b-mRuby3) dual fluorescent reporters. When differentiated as 3D organoids these expressed GFP at day 15 and tdTomato (or mRuby3) at day 25, respectively. This enabled us to explore transcriptional and chromatin related changes using RNA-seq and ATAC-seq from pluripotency through early retina specification. Pathway analysis of developing organoids revealed a stepwise loss of pluripotency, while optic vesicle and retina pathways became progressively more prevalent. Correlating gene transcription with chromatin accessibility in early eye field development showed that retinal cells underwent a clear change in chromatin landscape, as well as gene expression profiles. While each dataset alone provided valuable information, considering both in parallel provided an informative glimpse into the molecular nature eye development.
Collapse
Affiliation(s)
- Melissa K. Jones
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Devansh Agarwal
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kevin W. Mazo
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Manan Chopra
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Shawna L. Jurlina
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicholas Dash
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Qianlan Xu
- Center for Translational Vision Research, University of California Irvine, Irvine, CA 92617, USA
| | - Anna R. Ogata
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Melissa Chow
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Alex D. Hill
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Netra K. Kambli
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
- Department of Biotechnology, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Guorong Xu
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Roman Sasik
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Amanda Birmingham
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Kathleen M. Fisch
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
| | - Robert N. Weinreb
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Ray A. Enke
- Department of Biology, James Madison University, Harrisonburg, VA 22807, USA
| | | | - Karl J. Wahlin
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
15
|
Quaranta V, Linkous A. Organoids as a Systems Platform for SCLC Brain Metastasis. Front Oncol 2022; 12:881989. [PMID: 35574308 PMCID: PMC9096159 DOI: 10.3389/fonc.2022.881989] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/04/2022] [Indexed: 12/18/2022] Open
Abstract
Small Cell Lung Cancer (SCLC) is a highly aggressive, neuroendocrine tumor. Traditional reductionist approaches have proven ineffective to ameliorate the uniformly dismal outcomes for SCLC - survival at 5 years remains less than 5%. A major obstacle to improving treatment is that SCLC tumor cells disseminate early, with a strong propensity for metastasizing to the brain. Accumulating evidence indicates that, contrary to previous textbook knowledge, virtually every SCLC tumor is comprised of multiple subtypes. Important questions persist regarding the role that this intra-tumor subtype heterogeneity may play in supporting the invasive properties of SCLC. A recurrent hypothesis in the field is that subtype interactions and/or transition dynamics are major determinants of SCLC metastatic seeding and progression. Here, we review the advantages of cerebral organoids as an experimentally accessible platform for SCLC brain metastasis, amenable to genetic manipulations, drug perturbations, and assessment of subtype interactions when coupled, e.g., to temporal longitudinal monitoring by high-content imaging or high-throughput omics data generation. We then consider systems approaches that can produce mathematical and computational models useful to generalize lessons learned from ex vivo organoid cultures, and integrate them with in vivo observations. In summary, systems approaches combined with ex vivo SCLC cultures in brain organoids may effectively capture both tumor-tumor and host-tumor interactions that underlie general principles of brain metastasis.
Collapse
Affiliation(s)
| | - Amanda Linkous
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
16
|
Gordeeva O, Gordeev A, Erokhov P. Archetypal Architecture Construction, Patterning, and Scaling Invariance in a 3D Embryoid Body Differentiation Model. Front Cell Dev Biol 2022; 10:852071. [PMID: 35573693 PMCID: PMC9091174 DOI: 10.3389/fcell.2022.852071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Self-organized patterning and architecture construction studying is a priority goal for fundamental developmental and stem cell biology. To study the spatiotemporal patterning of pluripotent stem cells of different origins, we developed a three-dimensional embryoid body (EB) differentiation model quantifying volumetric parameters and investigated how the EB architecture formation, patterning, and scaling depend on the proliferation, cavitation, and differentiation dynamics, external environmental factors, and cell numbers. We identified three similar spatiotemporal patterns in the EB architectures, regardless of cell origin, which constitute the EB archetype and mimick the pre-gastrulation embryonic patterns. We found that the EB patterning depends strongly on cellular positional information, culture media factor/morphogen content, and free diffusion from the external environment and between EB cell layers. However, the EB archetype formation is independent of the EB size and initial cell numbers forming EBs; therefore, it is capable of scaling invariance and patterning regulation. Our findings indicate that the underlying principles of reaction-diffusion and positional information concepts can serve as the basis for EB architecture construction, patterning, and scaling. Thus, the 3D EB differentiation model represents a highly reproducible and reliable platform for experimental and theoretical research on developmental and stem cell biology issues.
Collapse
Affiliation(s)
- Olga Gordeeva
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Gordeev
- National Institutes of Health’s National Library of Medicine, Bethesda, MD, United States
| | - Pavel Erokhov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|