Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Rep Methods ; 3(8): 100548, 2023 08 28.
Article in English | MEDLINE | ID: mdl-37671011

ABSTRACT

With a critical need for more complete in vitro models of human development and disease, organoids hold immense potential. Their complex cellular composition makes single-cell sequencing of great utility; however, the limitation of current technologies to a handful of treatment conditions restricts their use in screens or studies of organoid heterogeneity. Here, we apply sci-Plex, a single-cell combinatorial indexing (sci)-based RNA sequencing (RNA-seq) multiplexing method to retinal organoids. We demonstrate that sci-Plex and 10× methods produce highly concordant cell-class compositions and then expand sci-Plex to analyze the cell-class composition of 410 organoids upon modulation of critical developmental pathways. Leveraging individual organoid data, we develop a method to measure organoid heterogeneity, and we identify that activation of Wnt signaling early in retinal organoid cultures increases retinal cell classes up to 6 weeks later. Our data show sci-Plex's potential to dramatically scale up the analysis of treatment conditions on relevant human models.


Subject(s)
Critical Pathways , Organoids , Humans , Cell Differentiation , Neurons , Retina
2.
bioRxiv ; 2023 Jun 03.
Article in English | MEDLINE | ID: mdl-37398481

ABSTRACT

With a critical need for more complete in vitro models of human development and disease, organoids hold immense potential. Their complex cellular composition makes single-cell sequencing of great utility; however, the limitation of current technologies to a handful of treatment conditions restricts their use in screens or studies of organoid heterogeneity. Here, we apply sci-Plex, a single-cell combinatorial indexing (sci)-based RNA-seq multiplexing method to retinal organoids. We demonstrate that sci-Plex and 10x methods produce highly concordant cell class compositions and then expand sci-Plex to analyze the cell class composition of 410 organoids upon modulation of critical developmental pathways. Leveraging individual organoid data, we develop a method to measure organoid heterogeneity, and we identify that activation of Wnt signaling early in retinal organoid cultures increases retinal cell classes up to six weeks later. Our data show sci-Plex's potential to dramatically scale-up the analysis of treatment conditions on relevant human models.

3.
Cell Genom ; 3(6): 100302, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37388919

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of blindness, affecting 200 million people worldwide. To identify genes that could be targeted for treatment, we created a molecular atlas at different stages of AMD. Our resource is comprised of RNA sequencing (RNA-seq) and DNA methylation microarrays from bulk macular retinal pigment epithelium (RPE)/choroid of clinically phenotyped normal and AMD donor eyes (n = 85), single-nucleus RNA-seq (164,399 cells), and single-nucleus assay for transposase-accessible chromatin (ATAC)-seq (125,822 cells) from the retina, RPE, and choroid of 6 AMD and 7 control donors. We identified 23 genome-wide significant loci differentially methylated in AMD, over 1,000 differentially expressed genes across different disease stages, and an AMD Müller state distinct from normal or gliosis. Chromatin accessibility peaks in genome-wide association study (GWAS) loci revealed putative causal genes for AMD, including HTRA1 and C6orf223. Our systems biology approach uncovered molecular mechanisms underlying AMD, including regulators of WNT signaling, FRZB and TLE2, as mechanistic players in disease.

4.
Cell Rep ; 38(4): 110294, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081356

ABSTRACT

We previously used single-cell transcriptomic analysis to characterize human fetal retinal development and assessed the degree to which retinal organoids recapitulate normal development. We now extend the transcriptomic analyses to incorporate single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq), a powerful method used to characterize potential gene regulatory networks through the changes in accessible chromatin that accompany cell-state changes. The combination of scATAC-seq and single-cell RNA sequencing (scRNA-seq) provides a view of developing human retina at an unprecedented resolution. We identify key transcription factors relevant to specific fates and the order of the transcription factor cascades that define each of the major retinal cell types. The changing chromatin landscape is largely recapitulated in retinal organoids; however, there are differences in Notch signaling and amacrine cell gene regulation. The datasets we generated constitute an excellent resource for the continued improvement of retinal organoid technology and have the potential to inform and accelerate regenerative medicine approaches to retinal diseases.


Subject(s)
Cell Differentiation/physiology , Chromatin , Neurogenesis/physiology , Organoids , Retina/embryology , Fetus , Human Embryonic Stem Cells , Humans , RNA-Seq , Single-Cell Analysis
5.
Cell Rep ; 30(5): 1644-1659.e4, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023475

ABSTRACT

To study the development of the human retina, we use single-cell RNA sequencing (RNA-seq) at key fetal stages and follow the development of the major cell types as well as populations of transitional cells. We also analyze stem cell (hPSC)-derived retinal organoids; although organoids have a very similar cellular composition at equivalent ages as the fetal retina, there are some differences in gene expression of particular cell types. Moreover, the inner retinal lamination is disrupted at more advanced stages of organoids compared with fetal retina. To determine whether the disorganization in the inner retina is due to the culture conditions, we analyze retinal development in fetal retina maintained under similar conditions. These retinospheres develop for at least 6 months, displaying better inner retinal lamination than retinal organoids. Our single-cell RNA sequencing (scRNA-seq) comparisons of fetal retina, retinal organoids, and retinospheres provide a resource for developing better in vitro models for retinal disease.


Subject(s)
Fetus/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Retina/cytology , Single-Cell Analysis , Transcriptome/genetics , Cells, Cultured , Humans , Mitosis/genetics , Spheroids, Cellular/cytology , Time Factors
6.
Mol Neurobiol ; 57(2): 668-684, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31435904

ABSTRACT

In the present study, we investigated whether mutant huntingtin (mHTT) impairs mitochondrial functions in human striatal neurons derived from induced pluripotent stem cells (iPSCs). Striatal neurons and astrocytes derived from iPSCs from unaffected individuals (Ctrl) and Huntington's disease (HD) patients with HTT gene containing increased number of CAG repeats were used to assess the effect of mHTT on bioenergetics and mitochondrial superoxide anion production. The human neurons were thoroughly characterized and shown to express MAP2, DARPP32, GABA, synapsin, and PSD95. In human neurons and astrocytes expressing mHTT, the ratio of mHTT to wild-type huntingtin (HTT) was 1:1. The human neurons were excitable and could generate action potentials, confirming successful conversion of iPSCs into functional neurons. The neurons and astrocytes from Ctrl individuals and HD patients had similar levels of ADP and ATP and comparable respiratory and glycolytic activities. The mitochondrial mass, mitochondrial membrane potential, and superoxide anion production in human neurons appeared to be similar regardless of mHTT presence. The present results are in line with the results obtained in our previous studies with isolated brain mitochondria and cultured striatal neurons from YAC128 and R6/2 mice, in which we demonstrated that mutant huntingtin at early stages of HD pathology does not deteriorate mitochondrial functions. Overall, our results argue against bioenergetic deficits as a factor in HD pathogenesis and suggest that other detrimental processes might be more relevant to the development of HD pathology.


Subject(s)
Corpus Striatum/metabolism , Energy Metabolism , Huntingtin Protein/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Mutant Proteins/metabolism , Neurons/metabolism , Superoxides/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Cell Differentiation , Cells, Cultured , Humans , Membrane Potential, Mitochondrial , Trinucleotide Repeat Expansion/genetics
7.
Sci Rep ; 9(1): 3770, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842553

ABSTRACT

Epigenetic changes have been used to estimate chronological age across the lifespan, and some studies suggest that epigenetic "aging" clocks may already operate in developing tissue. To better understand the relationship between developmental stage and epigenetic age, we utilized the highly regular sequence of development found in the mammalian neural retina and a well-established epigenetic aging clock based on DNA methylation. Our results demonstrate that the epigenetic age of fetal retina is highly correlated with chronological age. We further establish that epigenetic aging progresses normally in vitro, suggesting that epigenetic aging is a property of individual tissues. This correlation is also retained in stem cell-derived retinal organoids, but is accelerated in individuals with Down syndrome, a progeroid-like condition. Overall, our results suggest that epigenetic aging begins as early as a few weeks post-conception, in fetal tissues, and the mechanisms underlying the phenomenon of epigenetic aging might be studied in developing organs.


Subject(s)
DNA Methylation , Retina/embryology , Sequence Analysis, RNA/methods , Cells, Cultured , Cellular Senescence , CpG Islands , Epigenesis, Genetic , Humans , Organoids/chemistry , Organoids/cytology , Retina/cytology
8.
Stem Cell Reports ; 12(2): 201-212, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30639213

ABSTRACT

Retinal ganglion cells (RGCs) form the connection between the eye and the brain, with this connectivity disrupted in numerous blinding disorders. Previous studies have demonstrated the ability to derive RGCs from human pluripotent stem cells (hPSCs); however, these cells exhibited some characteristics that indicated a limited state of maturation. Among the many factors known to influence RGC development in the retina, astrocytes are known to play a significant role in their functional maturation. Thus, efforts of the current study examined the functional maturation of hPSC-derived RGCs, including the ability of astrocytes to modulate this developmental timeline. Morphological and functional properties of RGCs were found to increase over time, with astrocytes significantly accelerating the functional maturation of hPSC-derived RGCs. The results of this study clearly demonstrate the functional and morphological maturation of RGCs in vitro, including the effects of astrocytes on the maturation of hPSC-derived RGCs.


Subject(s)
Astrocytes/cytology , Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Retina/cytology , Retinal Ganglion Cells/cytology , Cells, Cultured , Humans
9.
Sci Rep ; 8(1): 14520, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30266927

ABSTRACT

Retinal organoids are three-dimensional structures derived from human pluripotent stem cells (hPSCs) which recapitulate the spatial and temporal differentiation of the retina, serving as effective in vitro models of retinal development. However, a lack of emphasis has been placed upon the development and organization of retinal ganglion cells (RGCs) within retinal organoids. Thus, initial efforts were made to characterize RGC differentiation throughout early stages of organoid development, with a clearly defined RGC layer developing in a temporally-appropriate manner expressing a complement of RGC-associated markers. Beyond studies of RGC development, retinal organoids may also prove useful for cellular replacement in which extensive axonal outgrowth is necessary to reach post-synaptic targets. Organoid-derived RGCs could help to elucidate factors promoting axonal outgrowth, thereby identifying approaches to circumvent a formidable obstacle to RGC replacement. As such, additional efforts demonstrated significant enhancement of neurite outgrowth through modulation of both substrate composition and growth factor signaling. Additionally, organoid-derived RGCs exhibited diverse phenotypes, extending elaborate growth cones and expressing numerous guidance receptors. Collectively, these results establish retinal organoids as a valuable tool for studies of RGC development, and demonstrate the utility of organoid-derived RGCs as an effective platform to study factors influencing neurite outgrowth from organoid-derived RGCs.


Subject(s)
Organoids/cytology , Pluripotent Stem Cells/cytology , Retinal Ganglion Cells/cytology , Culture Media , Genes, Reporter , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Neuronal Outgrowth , Red Fluorescent Protein
10.
Stem Cells Transl Med ; 5(4): 417-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26933039

ABSTRACT

UNLABELLED: The derivation of human induced pluripotent stem cells (hiPSCs) from patient-specific sources has allowed for the development of novel approaches to studies of human development and disease. However, traditional methods of generating hiPSCs involve the risks of genomic integration and potential constitutive expression of pluripotency factors and often exhibit low reprogramming efficiencies. The recent description of cellular reprogramming using synthetic mRNA molecules might eliminate these shortcomings; however, the ability of mRNA-reprogrammed hiPSCs to effectively give rise to retinal cell lineages has yet to be demonstrated. Thus, efforts were undertaken to test the ability and efficiency of mRNA-reprogrammed hiPSCs to yield retinal cell types in a directed, stepwise manner. hiPSCs were generated from human fibroblasts via mRNA reprogramming, with parallel cultures of isogenic human fibroblasts reprogrammed via retroviral delivery of reprogramming factors. New lines of mRNA-reprogrammed hiPSCs were established and were subsequently differentiated into a retinal fate using established protocols in a directed, stepwise fashion. The efficiency of retinal differentiation from these lines was compared with retroviral-derived cell lines at various stages of development. On differentiation, mRNA-reprogrammed hiPSCs were capable of robust differentiation to a retinal fate, including the derivation of photoreceptors and retinal ganglion cells, at efficiencies often equal to or greater than their retroviral-derived hiPSC counterparts. Thus, given that hiPSCs derived through mRNA-based reprogramming strategies offer numerous advantages owing to the lack of genomic integration or constitutive expression of pluripotency genes, such methods likely represent a promising new approach for retinal stem cell research, in particular, those for translational applications. SIGNIFICANCE: In the current report, the ability to derive mRNA-reprogrammed human induced pluripotent stem cells (hiPSCs), followed by the differentiation of these cells toward a retinal lineage, including photoreceptors, retinal ganglion cells, and retinal pigment epithelium, has been demonstrated. The use of mRNA reprogramming to yield pluripotency represents a unique ability to derive pluripotent stem cells without the use of DNA vectors, ensuring the lack of genomic integration and constitutive expression. The studies reported in the present article serve to establish a more reproducible system with which to derive retinal cell types from hiPSCs through the prevention of genomic integration of delivered genes and should also eliminate the risk of constitutive expression of these genes. Such ability has important implications for the study of, and development of potential treatments for, retinal degenerative disorders and the development of novel therapeutic approaches to the treatment of these diseases.


Subject(s)
Cell Differentiation , Cell Lineage , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/physiology , Retina/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Organogenesis/genetics , Organogenesis/physiology , RNA, Messenger/metabolism , Retina/growth & development , Retina/metabolism
11.
Stem Cells ; 34(6): 1553-62, 2016 06.
Article in English | MEDLINE | ID: mdl-26996528

ABSTRACT

Human pluripotent stem cells (hPSCs), including both embryonic and induced pluripotent stem cells, possess the unique ability to readily differentiate into any cell type of the body, including cells of the retina. Although previous studies have demonstrated the ability to differentiate hPSCs to a retinal lineage, the ability to derive retinal ganglion cells (RGCs) from hPSCs has been complicated by the lack of specific markers with which to identify these cells from a pluripotent source. In the current study, the definitive identification of hPSC-derived RGCs was accomplished by their directed, stepwise differentiation through an enriched retinal progenitor intermediary, with resultant RGCs expressing a full complement of associated features and proper functional characteristics. These results served as the basis for the establishment of induced pluripotent stem cells (iPSCs) from a patient with a genetically inherited form of glaucoma, which results in damage and loss of RGCs. Patient-derived RGCs specifically exhibited a dramatic increase in apoptosis, similar to the targeted loss of RGCs in glaucoma, which was significantly rescued by the addition of candidate neuroprotective factors. Thus, the current study serves to establish a method by which to definitively acquire and identify RGCs from hPSCs and demonstrates the ability of hPSCs to serve as an effective in vitro model of disease progression. Moreover, iPSC-derived RGCs can be utilized for future drug screening approaches to identify targets for the treatment of glaucoma and other optic neuropathies. Stem Cells 2016;34:1553-1562.


Subject(s)
Cell Differentiation , Glaucoma/pathology , Induced Pluripotent Stem Cells/cytology , Nerve Degeneration/pathology , Retinal Ganglion Cells/cytology , Cell Line , Glaucoma/complications , Humans , Nerve Degeneration/complications , Optic Nerve Diseases/pathology , Phenotype
12.
Curr Protoc Stem Cell Biol ; 32: 1H.8.1-1H.8.20, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25640818

ABSTRACT

The protocol outlined below is used to differentiate human pluripotent stem cells (hPSCs) into retinal cell types through a process that faithfully recapitulates the stepwise progression observed in vivo. From pluripotency, cells are differentiated to a primitive anterior neural fate, followed by progression into two distinct populations of retinal progenitors and forebrain progenitors, each of which can be manually separated and purified. The hPSC-derived retinal progenitors are found to self-organize into three-dimensional optic vesicle-like structures, with each aggregate possessing the ability to differentiate into all major retinal cell types. The ability to faithfully recapitulate the stepwise in vivo development in a three-dimensional cell culture system allows for the study of mechanisms underlying human retinogenesis. Furthermore, this methodology allows for the study of retinal dysfunction and disease modeling using patient-derived cells, as well as high-throughput pharmacological screening and eventually patient-specific therapies.


Subject(s)
Cell Culture Techniques/methods , Optic Disk/cytology , Pluripotent Stem Cells/cytology , Retina/cytology , Cell Differentiation , Cell Lineage , Cell Proliferation , Embryoid Bodies/cytology , Humans , Neuroepithelial Cells/cytology , Retinal Pigment Epithelium/cytology , Spheroids, Cellular/cytology , Stem Cells/cytology
13.
Stem Cells Transl Med ; 2(4): 255-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23512959

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) possess tremendous potential for the field of regenerative medicine because of their ability to differentiate into any cell type of the body. Such ability has profound implications for translational medicine, because these cells have been implicated for use in cell replacement, disease modeling, and pharmacological screening. However, the translation of established methods for deriving retinal cell types from hiPSCs has been hindered by the use of xenogeneic products for their growth and differentiation. Thus, the ability to derive retinal cell types in the absence of xenogeneic products would represent a significant advancement. The following studies were therefore undertaken to test the ability of hiPSCs to give rise to retinal cells under nonxenogeneic conditions. hiPSCs were maintained in traditional, feeder-free, or xeno-free culture conditions, and their ability to differentiate to a retinal fate was tested. Upon differentiation under all three conditions, cells acquired advancing features of retinal development, eventually yielding cell types of the mature retina. Reverse transcription-polymerase chain reaction and immunocytochemistry confirmed early trends in gene and protein expression patterns in xeno-free derived hiPSCs similar to those in cells derived in mouse embryonic fibroblasts and in feeder-free conditions. Results from this study demonstrate that hiPSCs can be maintained and directed to differentiate into retinal cell types under nonxenogeneic conditions, similar to cells derived using current xenogeneic methodologies. The demonstration of this capability will facilitate future efforts to develop hiPSC-based therapies for retinal disorders and also help to advance in vitro studies of human retinal development.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Retina/cytology , Animals , Cell Line , Cell Proliferation , Embryo, Mammalian/cytology , Feeder Cells/cytology , Fibroblasts/cytology , Humans , Mice , Neurons/cytology , Retinal Pigment Epithelium/cytology , Spheroids, Cellular/cytology
14.
Curr Top Microbiol Immunol ; 367: 163-91, 2013.
Article in English | MEDLINE | ID: mdl-23292211

ABSTRACT

Regeneration of the nervous system requires either the repair or replacement of nerve cells that have been damaged by injury or disease. While lower organisms possess extensive capacity for neural regeneration, evolutionarily higher organisms including humans are limited in their ability to regenerate nerve cells, posing significant issues for the treatment of injury and disease of the nervous system. This chapter focuses on current approaches for neural regeneration, with a discussion of traditional methods to enhance neural regeneration as well as emerging concepts within the field such as stem cells and cellular reprogramming. Stem cells are defined by their ability to self-renew as well as their ability to differentiate into multiple cell types, and hence can serve as a source for cell replacement of damaged neurons. Traditionally, adult stem cells isolated from the hippocampus and subventricular zone have served as a source of neural stem cells for replacement purposes. With the advancement of pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (iPSCs), new and exciting approaches for neural cell replacement are being developed. Furthermore, with increased understanding of the human genome and epigenetics, scientists have been successful in the direct genetic reprogramming of somatic cells to a neuronal fate, bypassing the intermediary pluripotent stage. Such breakthroughs have accelerated the timing of production of mature neuronal cell types from a patient-specific somatic cell source such as skin fibroblasts or mononuclear blood cells. While extensive hurdles remain to the translational application of such stem cell and reprogramming strategies, these approaches have revolutionized the field of regenerative biology and have provided innovative approaches for the potential regeneration of the nervous system.


Subject(s)
Nerve Regeneration/physiology , Animals , Cellular Reprogramming , Humans , Motor Neuron Disease/therapy , Neural Stem Cells/physiology , Parkinson Disease/therapy , Retinal Degeneration/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...